Clinical Neurology

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 705

Clinical Neurology

Graeme J. Hankey
MBBS, MD, FRCP (Lond), FRCP (Edin), FRACP
Consultant Neurologist and Head of Stroke Unit
Department of Neurology, Royal Perth Hospital
and Clinical Professor
Department of Medicine, University of Western Australia
Perth, Australia

Joanna M. Wardlaw
MBChB, MD, FRCP, FRCR
Professor and Honorary Consultant Neuroradiologist
Department of Clinical Neurosciences, University of Edinburgh
Western General Hospital
Edinburgh, UK

MANSON
PUBLISHING
Copyright © 2002 Manson Publishing Ltd

ISBN 1–84076–010–9

All rights reserved. No part of this publication may be reproduced, stored in a retrieval system
or transmitted in any form or by any means without the written permission of the copyright
holder or in accordance with the provisions of the Copyright Act 1956 (as amended), or
under the terms of any licence permitting limited copying issued by the Copyright Licensing
Agency, 33–34 Alfred Place, London WC1E 7D, UK.

Any person who does any unauthorized act in relation to this publication may be liable to
criminal prosecution and civil claims for damages.

A CIP catalogue record for this book is available from the British Library.

For full details of all Manson Publishing Ltd titles please write to:
Manson Publishing Ltd
73 Corringham Road
London NW11 7DL, UK

Tel: +44 (0)20 8905 5150


Fax: +44 (0)20 8201 9233

Email: manson@man-pub.demon.co.uk
Website: www.manson-publishing.co.uk

Commissioning editor: Jill Northcott


Project manager: Paul Bennett
Text editor: Ruth C Maxwell
Layout: Top Draw (Tableaux)
Electronic artwork: MTG
Cover design: Patrick Daly
Color reproduction: Tenon & Polert Colour Scanning Ltd, Hong Kong
Printed by: Grafos SA, Barcelona, Spain
Contents
Foreword 6 5 Vertigo 107
Vertigo 107
Preface 7 Benign Paroxysmal Positional Vertigo 110
Vestibular Neuronitis 111
Abbreviations 8 Ménière’s Disease 111
Further Reading 112
1 Neurologic Diagnosis 11
The Neurologic History 11 6 Movement Disorders 113
Neurologic Examination 13 Dystonia 113
Imaging the Brain 23 Essential Tremor 119
Imaging the Cerebral Circulation 26 Chorea 122
Imaging the Spine 29 Myoclonus 124
Lumbar Puncture (LP) and Cerebrospinal Fluid Dyskinesias 129
(CSF) Examination 31 Tourette Syndrome 130
Electroencephalography (EEG) 33 Neuroleptic Malignant Syndrome 133
Nerve Conduction Studies 35 Further Reading 135
Electromyography (EMG) 38
Evoked Potentials 40 7 Developmental Diseases of the
Further Reading 43 Nervous System 136
Arnold–Chiari Malformation 136
2 Disorders of Consciousness 44 Rachischisis (Dysraphism) 139
Coma 44 Tuberous Sclerosis 143
Brain Death 51 Neurofibromatosis 147
Permanent Vegetative State (PVS) 52 Sturge–Weber Disease (Encephalotrigeminal
Locked-in Syndrome 55 Vascular Syndrome) 151
Narcolepsy 56 Hereditary Hemorrhagic Telangiectasia (HHT)
Syncope 59 (Osler–Rendu–Weber Syndrome) 153
Further Reading 64 Further Reading 156

3 Epilepsy 65 8 Inherited Metabolic Diseases of the


Epilepsy 65 Nervous System of Adult Onset 157
Status Epilepticus (SE) 82 Wilson’s Disease (WD) (Hepatolenticular
Juvenile Myoclonic Epilepsy (JME) (Janz Syndrome) 86 Degeneration) 157
Progressive Myoclonic Epilepsies (PME) 87 Hallervorden–Spatz Disease 162
Further Reading 90 Mitochondrial (Oxidative Phosphorylation) Diseases 162
Adrenoleukodystrophy (ALD) 166
4 Headache 91 Metachromatic Leukodystrophy (MLD) 169
Headache 91 Adult-onset Globoid Cell Leukodystrophy (Krabbe
Migraine 95 Disease) 171
Muscle Contraction/Tension-type Headache 101 Late-onset GM2 Gangliosidosis 172
Cluster Headache (Migrainous Neuralgia) 103 Niemann–Pick Disease Type C (NP-C) 174
Further Reading 106 Further Reading 176

9 Traumatic Diseases of the Nervous System 177


Subdural Hematoma 177
Further Reading 180
10 Vascular Diseases of the Nervous System 181 12 Inflammatory Disorders of the
Neurovascular Syndromes 181 Nervous System 337
Transient Ischemic Attacks (TIAs) of the Acute Disseminated Encephalomyelitis (Post
Brain and Eye 186 Infectious Encephalomyelitis) (ADEM) 337
Stroke 192 Multiple Sclerosis (MS) 340
Atherosclerotic Ischemic Stroke 202 Neurosarcoidosis 350
Cardioembolic Stroke 209 Cavernous Sinus Syndrome 354
Infective Endocarditis 215 Further Reading 356
Antiphospholipid Syndrome and other
Prothrombotic States (Thrombophilias) 218 13 Tumors of the Central Nervous System 357
Dissection of the Carotid and Vertebral Arteries 224 Brain Tumors 357
Central Nervous System Vasculitis 227 Gliomas 364
Giant Cell Arteritis (GCA) 234 Optic Nerve Glioma 372
Primary Intracerebral Hemorrhage (PICH) 238 Meningioma 374
Arteriovenous Malformation (AVM) 245 Craniopharyngioma 377
Subarachnoid Hemorrhage (SAH) 249 Pituitary Tumors 379
Cerebral Venous Thrombosis 257 Acoustic Neuroma 383
Vascular Dementia 261 Primary CNS Lymphoma 385
Hypertensive Encephalopathy 265 Germ Cell Tumors of the CNS 388
Pituitary Apoplexy 268 von Hippel–Lindau Disease (VHL) 392
Further Reading 269 Metastases to the CNS 396
Paraneoplastic Syndromes 401
11 Infections of the Nervous System 273 Further Reading 406
Bacterial Infections
Acute Pyogenic (Bacterial) Meningitis 273 14 Degenerative Diseases of the
Subacute and Chronic Meningitis and/or Nervous System 407
Encephalitis 279 Alzheimer’s Disease (AD) 407
Tuberculous Meningo-encephalitis 281 Frontotemporal Dementia Associated with
Intracranial Abscess (Brain Abscess and Subdural Mutation in Tau (Pick’s Disease, Lobar Atrophy) 414
Empyema) 284 Huntington’s Disease (HD) 417
Tetanus 287 Parkinson’s Disease (PD) 420
Whipple’s Disease 289 Diffuse Lewy Body Disease (DLBD) 430
Progressive Supranuclear Palsy (PSP) 432
Viral Infections Multiple Systems Atrophy 435
Acute Aseptic Meningitis 291 Autosomal Dominant Cerebellar Ataxias 437
Viral Encephalitis 292 Friedreich’s Ataxia 441
Herpes Simplex Virus Encephalitis (HSE) 295 Ataxia Telangiectasia (Louis–Bar Syndrome) 444
Varicella-Zoster Virus Encephalomyelitis 299 Further Reading 447
Human Immunodeficiency Virus (HIV)-Associated
Cognitive Motor Complex (HIV-CMC) 301 15 Acquired Metabolic Diseases of the
Subacute Sclerosing Panencephalitis (SSPE) 304 Nervous System 449
Progressive Multifocal Leukoencephalopathy (PML) 307 Hypoxic Encephalopathy 449
Poliomyelitis (Infantile Paralysis) 309 Hepatic Encephalopathy (HE) 452
Central Pontine Myelinolysis 457
Other Infections Further Reading 459
Neurosyphilis 312
Lyme Disease (Neuroborreliosis) 316 16 Nutritional Deficiency and the
Cryptococcal Meningitis 318 Nervous System 460
Mucormycosis of the Nervous System 321 Wernicke–Korsakoff Syndrome 460
Toxoplasmic Encephalitis 323 Vitamin B12 Deficiency 463
Cysticercosis 326 Further Reading 467
Creutzfeldt–Jakob Disease (CJD) 330
Further Reading 335 17 Disorders of the CSF Circulation 468
Hydrocephalus 468
Normal Pressure Hydrocephalus 473
Idiopathic Intracranial Hypertension (IIH)
(Benign Intracranial Hypertension,
Pseudotumor Cerebri) 477
Further Reading 480
18 Cranial Neuropathies 481 Vasculitic Neuropathy 597
Olfactory (Ist Cranial Nerve) Neuropathy 481 Herpes Zoster Infection 599
Optic (IInd Cranial Nerve) Neuropathy 483 Human Immunodeficiency Virus (HIV) Neuropathy 601
Anterior Ischemic Optic Neuropathy (AION) 487 Leprosy 604
Optic Neuritis 489 Paraproteinemic Polyneuropathies 606
Leber’s Hereditary Optic Neuropathy (LHON) 491 Diabetic Neuropathy 610
Papilledema 492 Further Reading 613
Horner’s Syndrome 494
Holmes–Adie Syndrome 497 22 Mononeuropathies 615
IIIrd, IVth, and VIth Cranial Nerve (Ocular Motor) Dorsal Scapular Nerve (to Rhomboids) Neuropathy 615
Neuropathies 499 Long Thoracic Neuropathy 616
Trigeminal (Vth Cranial Nerve) Neuropathy 505 Suprascapular Neuropathy 618
Trigeminal Neuralgia (Tic Douloureux, Paroxysmal Axillary Neuropathy 620
Facial Pain) 509 Musculocutaneous Neuropathy 621
Facial (VIIth Cranial Nerve) Neuropathy 511 Radial Neuropathy 623
Acute Idiopathic Facial Paralysis (Bell’s Palsy) 515 Median Neuropathy 627
Vestibular-cochlear (VIIIth Cranial Nerve) Carpal Tunnel Syndrome (CTS) 630
Neuropathy 517 Ulnar Neuropathy 632
IXth Cranial Nerve (Glossopharyngeal) Neuropathy 520 Lateral Cutaneous Nerve of the Thigh Neuropathy 637
Vagus (Xth Cranial Nerve) Neuropathy 523 Posterior Cutaneous Nerve of the Thigh Neuropathy 639
Spinal Accessory (XIth Cranial Nerve) Neuropathy 525 Femoral Neuropathy 640
Hypoglossal (XIIth Cranial Nerve) Neuropathy 526 Obturator Neuropathy 642
Further Reading 528 Gluteal Neuropathy 643
Sciatic Neuropathy 644
19 Spinal Cord Diseases 529 Tibial Neuropathy 647
Hereditary Spastic Paraparesis 529 Peroneal Neuropathy 650
Spinal Muscular Atrophy (SMA) 532 Sural Neuropathy 654
Motor Neuron Diseases (MND) 534 Pudendal Neuropathy 655
Syringomyelia 541 Further Reading 656
Cervical Spondylotic Myelopathy and
Radiculopathy 545 23 Neuromuscular Junction Disorders 657
Acute ‘Slipped Disc’ 550 Myasthenia Gravis (MG) 657
Spinal Epidural Hematoma 554 Lambert–Eaton Myasthenic Syndrome (LEMS) 664
Spinal Epidural Abscess 555 Further Reading 667
Spinal Arachnoiditis 557
Spinal Cord Infarction 558 24 Muscle Disorders 668
Acute Transverse Myelitis 561 X-lined Dystrophinopathies 668
Human Immunodeficiency Virus (HIV) Myelopathy 564 Facio-scapulo-humeral Muscular Dystrophy (FSHD) 672
Radiation-induced Encephalo-myelo-radiculopathy 565 Limb-girdle Muscular Dystrophies (LGMD) 674
Spinal Cord Tumors 567 Myotonic Dystrophy 678
Further Reading 572 Polymyositis 681
Dermatomyositis 684
20 Autonomic Nervous System Disorders 573 Inclusion Body Myositis (IBM) 687
Autonomic Neuropathy 573 Metabolic and Endocrine Myopathies 688
Further Reading 578 Hypokalemic Paralysis 693
Further Reading 695
21 Diseases of the Peripheral Nerve 579
Peripheral Neuropathy 579 Index 696
Hereditary Neuropathies 584
Hereditary Motor and Sensory Neuropathy
(Charcot–Marie–Tooth Disease) 585
Hereditary Neuropathy with Liability to
Pressure Palsies (HNPP) 587
Guillain–Barré Syndrome (GBS) (Acute
Inflammatory Demyelinating
Polyradiculoneuropathy [AIDP]) 588
Chronic Inflammatory Demyelinating
Polyneuropathy (CIDP) 593
Neuralgic Amyotrophy 595
6

Foreword
Education and learning in clinical neurology involve a the presenting complaint of the patient or according to a
process of integration. The student or trainee in the clinical logical pathophysiologic and anatomic classification scheme.
neurosciences must learn detailed information concerning For each disorder there is a clear definition, discussion of
the clinical presentation, pathology, radiology, electro- etiology, clinical features, and investigations.
diagnosis, and, finally, treatment of neurologic diseases. The authors have extensive experience as clinical
There are many excellent texts where these components are educators. This leads to a clarity of presentation that is ideal
separately reviewed. Clinical Neurology is a work where each for the advanced student or trainee in neurology. Program
area has been integrated into a practical approach to the or course directors responsible for clinical education in
patient. There is a high level of integration that brings neurology will find this text extremely useful. It can be used
together clinical presentation, appropriate diagnostic studies directly as the major source work for a course in advanced
and treatment. The text is concise and, where appropriate, clinical neuroscience.
a bulleted topic format is used. This makes information easy This is an important addition to the textbooks that are
to retrieve. High-quality illustrations present anatomic available in clinical neurology. The overall quality of
drawings, radiographs and pathologic specimens. This production ensures that this will be a durable educational
approach logically leads the reader through different clinical text in neurology.
scenarios from presentation to diagnosis and treatment.
The reader will be introduced to the principles of
neurologic diagnosis at the beginning of the book. This Anthony J Windebank, MD
detailed but concise initiation will be particularly useful to Dean, Mayo Medical School
the senior student or trainee in neurology. This is followed Neuroscience Research – Guggenheim 1501
by an introduction to all of the major types of study used to Mayo Clinic and Mayo Foundation
aid in the diagnosis of patients with neurologic disease. The Rochester
book is organized to be practically useful. Drs Hankey and Minnesota
Wardlaw present material which is organized according to USA
7

Preface
Neurology is an exciting and evolving clinical science. Since The fact that many previously untreatable diseases are
we began our training in neurology (GJH) and neuro- now known to be not only treatable but preventable, has
radiology (JMW) in the 1980s, the understanding and raised new optimism for the probability that treatments will
practice of clinical neurology and neuroradiology have been emerge for other currently incurable neurologic disorders.
transformed. Traditionally, the essence of neurology was the To take full advantage of the breadth of techniques, know-
rigorous application of meticulous clinical skills to localize the ledge and skills now available requires increased collabor-
presence (or absence) and precise location of neurological ation between neurologists, neuroradiologists and other
lesions. Clinical findings were correlated with pathological clinical neuro disciplines.
findings at autopsy, but understanding of disease mechanisms These developments have been one of the prompts to
was poor and therapeutic options were limited. Indeed, most writing this book. Where appropriate and possible, we have
neurological disorders like stroke, epilepsy, Alzheimer’s incorporated them, but only if their clinical effectiveness is
disease, multiple sclerosis, and motor neuron disease were supported by high quality evidence. For therapies, the level
considered untreatable. Neurology was thus regarded by of evidence required is a systematic review of all (published
many outside the specialty as rather erudite and nihilistic. and unpublished) randomized trials, as published and
However, in the past 15 years, advances in neuroimaging, regularly updated in the Cochrane Library and Clinical
basic neuroscience, molecular genetics, and the consequent Evidence. Where such evidence is not available, we have
development and evaluation of therapies have brought new indicated so, and offered empirical recommendations based
meaning and life to the clinical features elicited at the on the best available evidence and our own experience.
bedside. Furthermore, new diseases with potentially Another prompt has been our impression of a void in
devastating consequences have emerged, and traditional rigid middle-sized clinical neurology texts which have some flesh
boundaries between neurology and psychiatry have blurred added to the raw, skeletal content of many handbooks, yet
with increased recognition of the interaction between mind which are not as bulky as comprehensive texts which we find
and body. Thus, clinical neurology has emerged as one of the difficult to carry, read and use. We have therefore taken this
most exciting frontiers in medicine. The array of new and rapidly changing field and focused on the essentials. The
emerging diagnostic and therapeutic options include: book is written and illustrated for students of clinical
neurology, particularly neurologists-in-training and
• Superb, safe, and noninvasive diagnostic imaging of the practicing neurologists, who wish to have ready access to a
vasculature and the structure, metabolism and function comprehensive, up-to-date, and evidence-based guide to the
of the brain and spinal cord by magnetic resonance. This understanding, diagnosis and management of common and
has added a new dimension to clinico-pathologic important neurologic disorders.
correlation, optimized diagnosis, exposed new insights We have included more than 800 illustrations in the text.
into pathophysiology, and facilitated new treatments. Many are images taken from our own patients, whom we
• Advances in catheter technology and interventional would like to thank for allowing us to photograph them or
neuroradiology. The better availability of these less invasive the outcome of their investigations. Furthermore, we would
techniques is changing the focus of neurosurgery, also like to thank Professor John Best, Dr Andrew
neurology and neuroimaging. Chancellor, Professor Byron Kakulas, Dr Robin Sellar,
• The introduction of DNA analysis as a diagnostic and Mr Matthew Wade, and the Department of Medical
prognostic tool. The enormous advances in molecular Illustrations, Royal Perth Hospital, for all providing
biology have revolutionized our understanding of the illustrations, as indicated throughout the book, and Dr
pathogenesis of many inherited and degenerative Peter Silbert for helpful comments on sections of the text.
neurologic conditions and facilitated accurate diagnosis, Finally, we would like to thank our families and colleagues
predictive testing, and genetic counselling. for supporting us in this endeavour. We hope you enjoy the
• Effective treatments have been recognized in large, book and welcome any comments and criticisms.
randomized controlled clinical trials for many neurologic
disorders, such as acute migraine, stroke, limb spasticity Graeme J Hankey
and other movement disorders, partial epilepsy, multiple Joanna M Wardlaw
sclerosis, Parkinson’s disease, Alzheimer’s disease, Guillain-
–Barré syndrome and other immune-mediate peripheral
neuropathies, myopathies, and myasthenia gravis.
However, many treatments are still not very effective and
there is enormous opportunity for improvement.
8

Abbreviations
α-TTP α-tocopherol-transfer protein CHOP cyclophosphamide, doxorubicin, vincristine and
AAG allergic angiitis and granulomatosis of Churg–Strauss prednisone-based therapy
ACA anticardiolipin antibody CIDP chronic inflammatory demyelinating polyneuropathy
ACE angiotensin-converting enzyme CJD Creutzfeldt–Jakob disease
ACh acetylcholine CK creatine kinase
AChR acetylcholine receptor CLAM cholesterol-lowering agentmyopathy
ACTH adrenocorticotropic hormone CMAP compound muscle action potential
AD Alzheimer’s disease CMT Charcot–Marie–Tooth disease
ADCA autosomal dominant cerebellar ataxia CMV cytomegalovirus
ADD attention deficit disorder CNS central nervous system
ADEM acute disseminated encephalomyelitis CO2 carbon dioxide
ADH antidiuretic hormone CoA coenzyme A
ADL activities of daily living COMT catechol-O-methyltransferase
ADP adenosine diphosphate COX cyclo-oxygenase
AF atrial fibrillation CPAP continuous positive airway pressure
AF-MSA autonomic failure with multiple system atrophy CPEO chronic progressive external ophthalmoplegia
AF-PD autonomic failure with Parkinson’s disease CPH chronic paroxysmal hemicrania
AGE advanced glycation end product CPT carnitine palmitoyl transferase
AHC anterior horn cells CREST calcinosis, Raynaud’s phenomenon, esophageal
AIDP acute inflammatory demyelinating dysmotility, sclerodactyly and telangiectasis
polyradiculoneuropathy CSDH chronic subdural hematoma
AIDS acquired immunodeficiency syndrome CSF cerebrospinal fluid
AION anterior ischemic optic neuropathy CT computerized tomography
ALD adrenoleukodystrophy CTS carpal tunnel syndrome
ALS amyotrophic lateral sclerosis CVST cerbral venous sinus thrombosis
AMAN acute motor-sensory axonal neuropathy CXR chest x-ray
AMN adrenomyeloneuropathy DBS deep brain stimulation
AMSAN acute motor-sensory axonal neuropathy DDAVP desmopressin
ANCA antineutrophil cytoplasmic antibody DGC dentate granule cell
ANNA anti-neuronal nuclear antibody DLBD diffuse Lewy body disease
AP antero-posterior DMD Duchenne’s muscular dystrophy
APA antiphospholipid antibodies DML
APCA-1 anti-Yo antibody DNA deoxyribonucleic acid
APLAb antiphospholipid antibody syndrome Dpt dilute prothrombin time
ApoE apolipoprotein E DRG dorsal root ganglia
APP amyloid precursor protein DRPLA dentato-rubro-pallido-luysian atrophy
APTT activated partial thromboplastin time dRVVT dilute Russell’s viper venom time
AR Argyll Robertson DSA digital subtraction angiography
ARR absolute risk reduction DSM IIIr Diagnostic and statistical manual of mental disorders
ASA arylsulfatase A – revised
ATP adenosine triphosphate DVT deep vein thrombosis
AVM arteriovenous malformation DWI diffusion-weighted imaging
anti-AChRAb anti-acetylcholine receptor antibody DZ dizygotic
ßA4 beta amyloid EBV Epstein–Barr virus
BAEP brainstem auditory evoked potential ECA external carotid artery
BAL British anti-lewisite EKG (ECG) electrocardiograph
BBB blood–brain barrier ECOG electrocorticography
BCG bacille Calmette–Guerin ECST
bd twice daily ECT electroconvulsive therapy
BMD Becker’s muscular dystrophy EEG electroencephalogram/electroencephalograph
BP blood pressure EGFR epidermal growth factor receptor
BSE bovine spongiform encephalopathy EITB enzyme-linked immunoelectrotransfer blot
BTX-A/F botulinum toxin A/F ELISA enzyme-linked immunosorbent assay
CADASIL cerebral autosomal dominant arteriopathy with EM electronmicroscopy
subcortical infarction and leukoencephalopathy EMG electromyography/electroencephalogram
CAM computer assisted myelography EPH episodic paroxysmal hemicrania
CANOMAD chronic ataxic neuropathy, ophthalmoplegia, M EPP end-plate potential
protein, agglutination, anti-disialosyl antibodies EPT enhanced physiological tremor
CAVATAS Carotid And Vertebral Artery Transluminal ERG electroretinogram
Angioplasty Study ESR erythrocyte sedimentation rate
CBF cerebral blood flow ET essential tremor
CCA common carotid artery FAME familial adult myoclonic epilepsy
cDNA complementary deoxyribonucleic acid FAST functional assessment staging test
CEA carcino-embryonic antigen FBP full blood picture
CGRP calcitonin gene-related peptide FDG fluorodeoxyglucose
Abbreviations 9

FFI fatal familial insomnia JME juvenile myoclonic epilepsy


FHM familial hemiplegic migraine KCT kaolin clotting time
FPP familial (primary) periodic paralysis K–F Kayser–Fleischer
FSH follicle stimulating hormone KSS Kearns–Sayre syndrome
FSHD facio-scapulo-humeral muscular dystrophy
FTA fluorescent treponemal antibody test LA lupus anticoagulant
FTA-ABS fluorescent treponemal antibody absorption LACI lacunar infarct
LCM lymphocytic choriomeningitis
GABA gamma amino butyric acid LDL low-density lipoprotein
GAP guanosine triphosphatase-activating protein LEMS Lambert–Eaton myasthenic syndrome
GBS Guillain–Barré syndrome LG lymphomatoid granulomatosis
GCA giant cell arteritis LGMD limb-girdle muscular dystrophies
GCI glial cytoplasmic inclusions LH luteinizing hormone
GI gastrointestinal LHON Leber’s hereditary optic neuropathy
GLT glutamate transporter gene LP lumbar puncture
GP general practitioner
GPI glycoprotein I MAG myelin associated glycoprotein
GPl lateral globus pallidus MAO monoamine oxidase
GPm medial/internal globus pallidus MAOI monoamine oxidase inhibitor
GSS Gerstmann Straussler Scheinker syndrome MAP microtubule-associated protein
GTCS generalized tonic-clonic seizures MBP myelin basic protein
GTE glycerol trierucate MCA middle cerebral artery
GTO glycerol trioleate oil MCTD mixed connective tissue disease
GTP guanine triphosphate MELAS mitochondrial encephalomyopathy, lactic acidosis, and
stroke-like episodes syndrome
5-HT 5-hydroxytryptamine MEPP miniature end-plate potential
5-HT3 5-hydroxytryptophan-3 MERRF myoclonic epilepsy with ragged-red fibers
HAART highly active antiretroviral therapy Met methionine
HACEK Haemophilus spp., Actinobacillus MFS Miller Fisher syndrome
actinmycetemcomitans, Cardiobacterium hominis, Eikenella MG myasthenia gravis
corrodens, Kingella spp. MGUS monoclonal gammopathy of uncertain significance
HCG human chorionic gonadotrophin MI myocardial infarction
HCl hydrochloric acid MJD Machado Joseph disease
HD Huntington’s disease MLD metachromatic leukodystrophy
HDL high-density lipoprotein MLF medial longitudinal fasciculus
H&E hematoxylin and eosin MMSE Mini-Mental State exam
HE hepatic encephalopathy MNCV motor nerve conduction velocities
HexA hexosaminidase A MND motor neuron disease
HexB hexosaminidase B MPTP N-methyl-4-phenyl-1,2,3,6,-tetrahydropyridine
hGH human growth hormone MR magnetic resonance
HHT hereditary hemorrhagic telangiectasia MRA magnetic resonance angiography
Hib Haemophilus influenzae type b MRC Medical Research Council
HIV human immunodeficiency virus MRI magnetic resonance imaging
HIV-CMC human immunodeficiency virus-associated mRNA messenger ribonucleic acid
cognitive/motor complex MS multiple sclerosis
HLA human leukocyte antigen MSA multiple system atrophy
HMPAO hexamethylpropylenamine oxime MSU mid-stream urine
HMSN hereditary motor and sensory neuropathy MT metallothionein
HNA hereditary neuralgic amyotrophy mtDNA mitochondrial deoxyribonucleic acid
HNPP hereditary neuropathy with liability to pressure palsies MTS mesial temporal sclerosis
HR heart rate MUP motor unit potentials
HSAN hereditary sensory and autonomic neuropathy MZ monozygotic
HSE herpes simplex virus encephalitis
HSP hereditary spastic paraparesis NAIP neuronal apoptosis inhibitory protein
HS-tk herpes simplex thymidine kinase NASCET North American symptomatic carotid
HSV herpes simplex virus endarterectomy trial
HTI hemorrhagic transformation of the infarct NCV nerve conduction velocities
HTLV human T lymphocyte virus NF neurofibromatosis
NGF nerve growth factor
IA-DSA intra-arterial digital subtraction angiography NMDA N-methyl-D-aspartate
IBM inclusion body myositis NMS neuroleptic malignant syndrome
iC interstitial nucleus of Cajal NNT number needed to treat
ICA internal carotid artery NO nitric oxide
ICH intracerebral hemorrhage NP Niemann–Pick
ICP intracranial pressure NP-C Niemann–Pick disease type C
ICU intensive care unit NPH normal pressure hydrocephalus
IF intrinsic factor NRT nucleus reticularis thalami
IFN interferon NSE neuron-specific enolase
Ig immunoglobulin NTD neural tube defect
IIH idiopathic intracranial hypertension nvCJD new variant Creutzfeldt–Jakob disease
i.m. intramuscular
INR internal normalized ratio OCP oral contraceptive pill
IOH idiopathic orthostatic hypotension OFSM oculo-facial-skeletal myorhythmia
IQ intelligence quotient OMM oculomasticatory myorhythmia
i.v. intravenous OPCA olivopontocerebellar atrophy
IV DSA intravenous digital subtraction angiography OR odds ratio
IVIG intravenous immunoglobulin P100 positive wave at 100 ms
JCV JC virus PA pernicious anemia
10 Abbreviations

PACI partial anterior circulation infarct SLE systemic lupus erythematosus


PaCO2 partial pressure of carbon dioxide SMA spinal muscular atrophy
PAF pure autonomic failure SMN survival motor neuron protein
PAN polyarteritis nodosa SMNT survival motor neuron gene
PAS periodic acid-Schiff SNAP sensory nerve action potential
PCA posterior communicating artery SNc substantia nigra pars compacta
PCA-1 type 1 anti-Purkinje cell antibody SND striatonigral degeneration
PCR polymerase chain reaction SNr substantia nigra reticulata
PCV procarbazine, lomustine, and vincristine SOD Cu/Zn superoxide dismutase
PD proton density SPECT single photon emission computed tomography
PD Parkinson’s disease SS Sjögren’s syndrome
PDE phosphodiesterase SSCP single strand conformation polymorphism
PDW proton density weighted SSEP somatosensory evoked potentials
PDWI proton density weighted image SSPE subacute sclerosing panencephalitis
PE pulmonary embolism SSRI selective serotonin reuptake inhibitor
PED paroxysmal exertion-induced dyskinesia SSS sick sinus syndrome
PEG percutaneous endoscopic gastrostomy STIR short time inversion recovery
PET positron emission tomography STN subthalamic nucleus
PGI2 prostacyclin SUDEP sudden unexplained death in epilepsy
PGL phenolic glycolipid SUNCT short-lasting unilateral neuralgiform headache with
PHD paroxysmal hypnogenic dyskinesia conjunctival injection and tearing
PI protease inhibitor T1W T1 weighted
PICH primary intracerebral hemorrhage T2W T2 weighted
PKD paroxysmal kinesogenic dyskinesia TA Takayasu’s arteritis
PL phospholipid TACI total anterior circulation infarct
PLED periodic lateralized epileptiform discharge TACS total anterior circulation syndrome
PME progressive myoclonic epilepsies TB tuberculosis
PML progressive multifocal leukoencephalopathy TCD transcranial doppler ultrasound
PMR polymyalgia rheumatica TD Tourette disorder
PNKD paroxysmal non-kinesogenic dyskinesia tds three times daily
PNS peripheral nervous system TENS transcutaneous electrical nerve stimulation.
p.o. by mouth (per os) TETA triethylene tetramine dihydrochloride
POCI posterior circulation infarct TF tissue factor
POEMS polyneuropathy, organomegaly, endocrinopathy, TFPI tissue factor pathway inhibitor
monoclonal paraproteinemia, and skin hyperpigmentation TG therapeutic gain
PPRF paramedian pontine reticular formation TIA transient ischemic attacks
PrP prion protein TNF tumor necrosis factor
PROMM proximal myotonic myopathy TOE transesophageal echocardiography
PS-1/2 presenilin-1/2 tPA tissue plasminogen activator
PSA prostatic specific antigen TPHA Treponema pallidum hemagglutination test
PSD periodic synchronous discharge TPI Treponema pallidum immobilization test
PSP progressive supranuclear palsy TPP thyrotoxic periodic paralysis
PSS progressive systemic sclerosis tRNA transfer ribonucleic acid
PTHRP parathyroid hormone-related protein TS Tourette syndrome
PVC procarbazine, lomustine, and vincristine TSH thyroid stimulating hormone
PVS permanent vegetative state TSC tuberous sclerosis complex
PWI perfusion-weighted image TST thermoregulatory sweat test
Q-SART quantitative sudomotor axon reflex test TT thrombin time
TTE transthoracic echocardiography
RA rheumatoid arthritis
TTP thrombotic thrombocytopenic purpura
RBC red blood cells
RCT randomized controlled trial UCH ubiquitin carboxy-terminal hydrolase
REM rapid-eye movement UL upper limb
RFLP restriction fragment length polymorphism UMN upper motor neuron
RIA radioimmunoassay VC vital capacity
riMLF rostral interstitial nucleus of the medial longitudinal VDRL Venereal Disease Research Laboratory slide flocculation
fasciculus test
RNA ribonucleic acid VEP visual evoked potential
ROM rifampicin, ofloxacin and minocycline VGCC voltage-gated calcium channels
RPR rapid plasma reagin VHL von Hippel–Lindau disease
rRNA ribosomal ribonucleic acid Vim ventrointermediate nucleus
RRR relative risk reduction VL ventrolateral thalamus
RT reverse transcriptase VLCFA very long chain saturated fatty acids
rt-PA or r-tPA recombinant tissue plasminogen activator VMA vanillylmandelic acid
SAE subcortical arteriosclerotic encephalopathy VSD ventricular septal defect
SAF scrapie-associated fibrils vWF von Willebrand factor
SAH subarachnoid hemorrhage VZV varicella-zoster virus
SBMA androgen receptor WADA intracarotid amytal test
SC sickle cell WBC white blood cells
SCA spinocerebellar atrophy subtype WD Wilson’s disease
SCLC small cell lung cancer WFNS World Federation of Neurological Surgeons
SDH succinate dehydrogenase WG Wegener’s granulomatosis
SDS Shy–Drager syndrome WH Werdnig–Hoffman
SE status epilepticus WHO World Health Organization
SFEMG single fiber electromyography
SIADH syndrome of inappropriate antidiuretic hormone
Chapter One 11

Neurologic
Diagnosis
THE NEUROLOGIC HISTORY The history must be taken; a relevant, succinct history is
rarely given by the patient. The narrative history should be
emphasized, not a long list of complaints and detailed
The purpose of the assessment of a patient with a suspected systemic enquiry. Brief notes should be recorded as the
neurologic disorder is to answer the following questions: history unfolds, recording what the patient actually said
• Is there a neurologic lesion?/Is this a neurologic rather than your interpretation, to ensure greater reliability.
disorder? Syndromic diagnosis. While taking the history, the clinician must indirectly assess
• What is the level of the neurologic lesion in the neuraxis? the patient’s mental status, and consider the circumstances
Anatomic diagnosis. under which the symptoms occurred (e.g. seizure).
• Where is (are) the neurologic lesion(s)? Anatomic Concurrently, observe the patient for signs of underlying
diagnosis. disease that may be relevant to the presenting complaint
• What is the nature/cause of the neurologic lesion(s)? (1–3). If an adequate history is not available from the
Pathologic diagnosis. patient, or needs verification, other useful sources include
• What are the patient's impairments, disabilities and family members, friends, employers, observers of any events,
handicaps? Functional diagnosis. and previous medical records.
• What is the likely outcome? Prognosis.
• What can be done about it? Treatment.

The clinical history is the most 1 2


important and productive part of the
neurologic assessment. Although the
physical examination is often used to
localize the lesion accurately and elicit
physical signs suggestive of the cause,
the history provides most of the
information about the presence or
absence and nature/cause of the
lesion. Therefore, if time is at a
premium it is preferable to be selective
with the examination rather than with
the history, using the history to select
or target the appropriate examination.

1 This patient, who complained of altered


sensation in the feet and hands, is also
noticeably pale.The cause of her sensory
neuropathy and pallor was pernicious anemia.

3
2 Capillary hemangioma (port wine stain) on the left side
of the face in the distribution of the ophthalmic and
maxillary divisions of the left trigeminal nerve in a patient
with Sturge–Weber syndrome who presented following his
first epileptic seizure.

3 Right Horner’s syndrome (ptosis of the right eyelid and


miosis of the right pupil, arrow) due to right lateral
medullary infarction.
12 Neurologic Diagnosis

TAKING THE HISTORY the patient has to contradict this assumption, which is less
Record the patient’s name, age, address, occupation and likely to produce spurious information.
handedness.
Sleep.
Presenting symptoms and their duration Appetite.
‘What is the problem?’ Nausea and vomiting.
‘Why have you come to see me today?’ Diarrhea.
Weight.
History of the presenting complaint Indigestion.
The mode of onset, evolution and course of the illness Cough.
should be recorded as it unfolds in chronologic sequence Headache.
from the patient’s own account. It can be prompted by
asking the patient: Other systemic symptoms that may be relevant include
fever, sweats, chest pain, shortness of breath, intermittent
‘When did this problem begin and what were you doing claudication, joint aches, and disturbances of sphincter
at the time?’ or function, sexual function and menses.
‘Begin from when you were last perfectly well’.
Sometimes, asking the same question twice in different Smoking and alcohol intake
ways may elicit further information: ‘Have you ever had Record the patient’s smoking, alcohol and other drug habits.
anything like this before?’.
Medications
The clinician’s role is to keep the patient focused on their Current and recent past medications should be recorded.
symptoms and signs, rather than the doctors who were Many patients with a long history of uncontrolled epilepsy or
consulted, the tests ordered, and the opinions of casual migraine state that they have ‘tried everything’ without
observers, unless they are relevant. The onset and evolution success but it is important to ask them to name each drug,
of the symptoms, whether abrupt and gradually improving the dose they took, the length of time it was taken for, and
thereafter, steadily progressive, fluctuating, or recurrent the effect, because commonly an adequate trial of a
often reflects the pathologic nature of the disease process. prophylactic drug has not been tried in an adequate dose for
If the patient or family cannot supply this information, it sufficient duration. Patients often need to be asked specifically
may be possible to ascertain it by what the patient was able if they are taking the oral contraceptive pill, vitamins,
to do at different times (e.g. still walk and work, or not). alternative homeopathic and herbal remedies, or illicit drugs.
The nature of the symptoms and part of the body that is
affected (e.g. loss of power and feeling in the legs together with Past history
urgency of micturition) often reflects the anatomic location of Any past history which could be relevant to the presenting
the lesion (e.g. spinal cord). It is important to ask questions to complaint is very important. For example, in a patient
assist in determining the level and location of the lesion(s), presenting with a first seizure the antenatal, perinatal and
thinking diagnostically while taking the history. Any childhood history is particularly important, as is a history of
precipitating or relieving factors, or associated symptoms should head injury with loss of consciousness.
be recorded, particularly if the patient complains of pain:
Family history
Pain history (acronym: LOTTRADIO) An ever-increasing number of neurologic disorders are being
Location (site) of pain: precise part of the body. recognized as being of genetic origin. Some are caused by a
Onset: when did the pain start and what was the patient mutation in a single gene and show classical Mendelian
doing at the time? Was the onset sudden, slowly inheritance of a characteristic phenotype. All known autosomal
progressive or preceded by other symptoms (e.g. visual dominant, autosomal recessive, and X-linked disorders belong
disturbance). Were there any precipitating and exacer- to this category (e.g. Huntington’s disease). Others are ‘poly-
bating factors (e.g. physical activity, bright light, noise)? genic’ and tend to ‘run in families’, although the inheritance
Type (quality): steady, throbbing, sharp, stabbing, pattern is complex and environmental influences can play a
burning, tight or dull? major role in the final phenotype (e.g. premature vascular
Timing: continuous or intermittent, any habitual pattern: disease, migraine). Determination of the inheritance pattern
on awakening from sleep or at the end of the day? is important because the risk to relatives diminishes more
Radiation: spread of pain to other sites. rapidly as the relationship becomes more distant in polygenic
Associated features (e.g. nausea, vomiting, photophobia, conditions compared with autosomal dominant inheritance.
phonophobia, neurologic symptoms of aura, fever, In addition, when polygenic inheritance is involved, the risk
palpitations, dyspnea). to siblings is increased with increasing number of affected
Duration of pain: seconds, minutes, hours, days. siblings, whereas in autosomal recessive conditions the number
Intensity: severity. of affected siblings does not modify the risk.
Offset (relieving factors): bed-rest, cold, heat, particular The differing versions of the same gene are called alleles.
postures or medications. The term genotype usually refers to the collection of alleles
inherited and carried by an individual. Phenotype refers to
Systems review (acronym: SANDWICH) the appearance of an individual in context (e.g. disease
Since diseases which affect the nervous system may also status, hair color, height, blood group). For twins,
affect other systems, it is important to enquire about other monozygous (MZ) twin pairs are genetically identical, while
systems, phrasing each question to suggest normality so that dizygous (DZ) pairs have no more genetic similarity that
Neurologic Examination 13

other siblings. If both twins in a pair have the same disease NEUROLOGIC EXAMINATION
or trait they are said to be ‘concordant’, while a single
affected twin is one of a ‘discordant’ pair.
The aim of the neurologic examination is usually to confirm
Personal history a diagnostic hypothesis generated from the history.
Details of the domestic, social, and business background Sometimes, the diagnosis is not at all clear from the history,
(e.g. the patient’s personality, relationship with family, in which case it is important to be able to conduct the
friends, employer and peer groups; sexual preference, neurologic examination in a uniform, sequential order so as
attitude to work and life, living arrangements, financial to avoid omission and perhaps also to facilitate record
state) may be relevant to both the genesis of the symptoms keeping. However, in most patients not every neurologic
and the management of the patient. Many neurologic function needs to be assessed. The thoroughness of the
disorders impose some form of handicap on the patient neurologic examination should be governed by the nature
which may compromise their ability to negotiate the of the clinical problem and the physical condition of the
bathroom, other rooms, or stairs in the house, ability to patient (and also the experience of the examiner). For
drive a car, ability to go back to work or hobbies, self- example, a patient presenting with symptoms and signs of a
esteem, and the health and well-being of the partner or median neuropathy at the wrist does not require the same
carer. It is crucial not to ignore the social and emotional intensity of assessment of cognitive and cerebellar function
impact of the illness on the patient and family. as a patient presenting with forgetfulness and clumsiness.

Concluding history GENERAL ASSESSMENT


Failure to take a good history is a common cause of failure to Watch the patient enter the consulting room, noting the
make the correct diagnosis and optimally manage the patient. appearance and behavior of the patient and the gait or form
Before proceeding with the examination, ask: ‘Is there of mobilization (e.g. wheelchair).
anything else you would like to tell me?’ and consider asking Whilst taking the history, listen to the speech and look
at this stage: ‘What do you think the problem is due to?’ closely at the patient’s face, hands (4) and skin (5–7). Also

4 5 6

4, 5 Clubbing of the hands and feet in a young patient with cyanotic 7


congenital heart disease and brain abscess.

6 Dry skin (ichthyosis) of a patient with peripheral neuropathy due to


Refsum’s disease.

7 Purpuric skin rash of meningococcal septicemia.


14 Neurologic Diagnosis

look at the eyes (8–11). Is there evidence of: standardized test of higher mental function should be
• Abnormal facial appearance. undertaken. One of the many available is the Mini Mental
• Ptosis or facial asymmetry. State examination (Table 1).
• Reduced blink frequency, facial expression and voice The Mini Mental State examination is only a screening
volume of Parkinsonism. test however, and fails to assess other higher mental
• Abnormal posture. functions, particularly non-verbal functions. Other aspects
• Wasting or fasciculation. to consider include:
• Tremor or other involuntary movements.
Consciousness (see p.44)
Also determine whether the patient is right or left
handed (i.e. which is likely to be the dominant hemisphere Orientation in time, place and person
for language).
Attention
MENTAL STATE (HIGHER MENTAL FUNCTION) • Concentration: digit span (normal: 6±1 forwards [i.e.
The patient’s alertness, speech, and intelligence can be abnormal <5 digits forward], 4±1 backwards [i.e.
assessed during history-taking; taking the clinical history and abnormal <3 digits backwards]).
evaluating the competence of the patient to give an accurate • Persistence: serial recitation (months of year backwards,
history involves an assessment of higher mental functioning. serial 7s); word list generation (words beginning with a
So, a formal assessment of higher mental function is not letter; normal >12/minute).
necessary in every patient. However, if suspicions are • Resistance to interference: alternating sequences:
aroused or the presenting complaint demands it, a +o+o+o.

8 9

8 The patient’s right eye is affected by ischemic oculopathy (arrow).


Note the congested sclera, cloudy cornea, new vessel formation
(neovascularization) around the limbus of the iris (rubeosis iridis) and
mid-dilated pupil, which indicate chronic anterior segment ocular 9 Close-up view of the right eye in 8 showing rubeosis iridis more
ischemia due to carotid occlusive disease. (Reproduced with clearly.
permission from Hankey GJ and Warlow CP [1994] Transient
Ischaemic Attacks of the Brain and Eye.WB Saunders.)

10 11

10 Fundus of the severely ischemic right eye of the patient in 8 11 Fundus of the normal left eye of the patient in 8.
showing attenuated vessels and pallor of the retina due to retinal
edema caused by severe ischemia.
Neurologic Examination 15

Table 1 Mini-mental state examination


Maximum score Maximum score
Orientation Language
What is the Time? Day? Month? Year? Season? 5 points Name two objects (e.g. a pencil and a watch). 2 points
What is the name of this Ward? Hospital? Town? Repeat a short phrase (e.g.‘No ifs, ands or buts’). 1 point
State? Country? 5 points Execute a three-stage command, scoring 1 point for
each stage (e.g.‘With the index finger of your left hand 3 points
Registration touch the tip of your nose and then your left ear.’).
Name three objects (e.g. car, dog, book) 3 points Read and obey the following ‘Close your eyes’. 1 point
and then ask patient to repeat these. Write a sentence of your own choice 1 point
The number of objects repeated correctly is the score. (the sentence must make sense and contain a subject
Endeavor, by further attempts and prompting, to have all and verb).
three repeated, so as to test recall later. Copy a diagram showing two intersecting pentagons. 1 point

Attention and calculation Total score 30 points


Subtract 7 from 100, and then 7 from the result. 5 points
Repeat this five times, scoring 1 point for each correct Scoring 23 or less denotes cognitive impairment (76% detection
subtraction. If the patient cannot/will not do this: spell rate, 4% false positive). (Adapted from Dick et al., J. Neurol. Neurosurg.
‘world’ backwards. Psychiatry, 1984; 47: 496.)

Recall
Name the three objects repeated in the registration test. 3 points
Score 1 point for each correct answer.

Memory • Dysphasia is a disturbance of language function, causing


• Retrograde: information (e.g. ‘who is the Prime difficulty understanding or expressing spoken or written
Minister/President, how many weeks in a year, dates of language.
World War 1 and II, define an island’). • Dysarthria is a disturbance of articulation of speech.
• Anterograde: verbal: register and recall three objects after
3 minutes; non-verbal: Rey Osterrieth complex figure test. Affect: mood and motivation, response to questions,
attitude to others
Other functions of intellect
• Calculation: serial 7s, or more simple calculations. Perception
• Abstraction: similarities (e.g. orange and banana, dog and • Illusions: misinterpretation of sensory stimuli (e.g.
horse), proverbs (e.g. ‘hunger is the best gravy’). confusional state).
• Praxis: • Delusions: incorrect beliefs that are held with conviction.
– Constructional: draw a clock, cube, flower; block design. • Hallucinations: sensory perceptions unrelated to sensory
– Ideomotor: buccofacial, limbs, body: ‘show how you stimuli.
would blow out a match, use a comb to comb your hair,
use a toothbrush to brush your teeth’. SKULL
• Visual–spatial perception: line bisection, star cancellation. Size and shape.
• Right–left orientation.
SPINE
Communication • Deformity.
• Verbal: • Tuft of hair in lumbosacral region (underlying spina
– Spontaneous speech: ‘describe what you can see in this bifida).
picture’. • Tenderness: spinal, paraspinal.
– Oral comprehension: one, two and three stage • Range of motion.
commands.
– Repetition: ‘If I were here then she could be there’. CRANIAL NERVES
– Naming: objects, body parts, lists of animals, words Nerve I (olfactory nerve)
beginning with a letter (e.g. ‘A’). • Olfactory sensation is seldom impaired in isolation.
– Reading: ‘point to the word ‘X’ on the card’. • As testing is time consuming, it need not be done unless
– Writing: write ‘ocean’, ‘groceries’, ‘some water is not there is a history of head trauma or possible dementia.
good to drink’.
– Articulation: labial sounds (‘pa’), lingual sounds (‘ta’), Nerve II (optic nerve)
palatal sounds (‘ka’). Visual acuity
• Non-verbal: prosody: use of intonation, gesture, • Only relevant if the patient has a history of ocular
appropriate turn-taking. problems or visual symptoms.
• Handedness: language is a function of the dominant • Record visual acuity in right and left eyes at 6 m (20 ft)
hemisphere (the left hemisphere in 96% of right-handers from a Snellen’s chart, with the patient wearing
and more than 50% of left-handers). spectacles if necessary.
16 Neurologic Diagnosis

Color vision senilis and a Kayser–Fleischer ring; the latter may require closer
Color vision can be assessed at the bedside, ideally by testing examination with the ophthalmoscope or slit lamp, but can
each eye with pseudoisochromatic plates. A more simple even be seen by looking at the limbus through an auroscope
convention is to take the bright-red top from a bottle of the from which the attached ear piece has been removed.
mydriatic solutions, place it over a small flashlight, and
present it to each eye. To a patient with optic nerve disease, Fundoscopy
the color red will appear pale or brownish. The degree of Perform fundoscopy in all patients. Ask the patient to fix on
color desaturation can be roughly quantified by asking the an external object with one eye while the other eye is being
patient to express the value of the color as a percentage of examined. The examiner’s head must remain vertical so as
the value of the color perceived by the normal eye. not to obscure the line of vision of the patient’s fixating eye.
Note any abnormalities on the surface of the eye and the
Visual fields lens (e.g. cataract). Then focus on the:
Test first; patients may be dazzled for minutes after pupil • Optic disc: note its color – pale in optic atrophy, pink and
tests and ophthalmoscopy. congested in papilledema (12). N.B. Temporal pallor of
Screen by confrontation, in which the patient is asked to the optic disk is a normal finding in many discs, but it
look at the examiner’s eyes. Check the upper temporal indicates optic atrophy when it is associated with
quadrants by simultaneously wiggling your right and left decreased visual acuity, a field defect, and an abnormality
index fingers in the upper temporal visual fields of the of color vision.
patient and yourself, ensuring that your moving fingers are • Optic cup: diminished in papilledema (12), enlarged in
an equal distance between you and the patient so that you glaucoma.
both have the same visual field. Ask the patient to point (if • Venous pulsation in the optic cup: absent if intracranial
they can) to which fingers are moving: the right, left, or pressure exceeds retinal venous pressure.
both. If any doubt, repeat the test, wiggling one finger at a • Disc margins: blurred in papilledema (12). If the disc
time and then both fingers. Repeat in the lower temporal cannot be seen, look even harder and change focus
quadrants. This technique detects conventional hemianopic because a swollen, red disc of papilledema may not be
and quadrantanopic field defect as well as visual inattention. obvious among the background red retina. In addition,
If there is a visual field defect, the nasal and temporal its surface will not be in focus when focusing on the
fields should be tested individually and the margins of any retina, because it is heaped up, as if looking from above
defect discerned with the use of a 5 mm (0.2 in) white and at a large mountain coming out of the sea.
red pin head. • Retinal vessels: narrow and tortuous with arteriovenous
Central vision and the size of the blind spot should also nipping in hypertension; microaneurysms with diabetes;
be tested by confrontation and using a pin head. cholesterol, platelet or calcific emboli in arterioles
Confrontation testing is however only a screening (13–16), distension or collapse of veins.
technique. Quantitative perimetry (colloquially known as • Retinal surface: exudates, hemorrhages (17), retinitis
‘formal’ fields) is more accurate and precise. pigmentosa, choroiditis.

Pupillary responses
Note the size and shape of each pupil, and the direct and
consensual response of each pupil to stimulation by light
and accommodation.
Argyll Robertson pupils are ‘Accomodation Retaining’
(pneumonic to remember that the accomodation reflex is
preserved in Argyll Robertson pupils, i.e. they both have the
initials ‘AR’); i.e. accommodation reflex present but light 12
reflex absent.
Horner’s syndrome consists of miosis, ptosis and
anhidrosis.
Ask the patient if the light appears equally bright when
shone in both eyes. If there is a difference, look for a relative
afferent pupillary defect. With the patient’s gaze fixed in the
distance, shine the light in one eye and note both pupils
constrict. Move the light to the other eye. During the light
beam’s transit across the nose, both pupils dilate slightly. In
normal eyes, both pupils constrict again when the beam of
light falls onto the other eye. If the patient has a relative
afferent pupillary defect, the pupil (and also the contralateral
pupil) continues to dilate when the light beam falls on it. This
usually indicates retinal or optic nerve dysfunction, but may
be observed with lesions as posterior as the optic tract. The
extreme example of an afferent pupillary defect is the 12 Ocular fundus showing papilledema in a patient with headache due
amaurotic pupil of the completely blind eye. The term Marcus to idiopathic intracranial hypertension. Note the congested swollen
Gunn pupil is often used but what we now call a relative optic disc, loss of the physiological cup, blurred disc margins and
afferent pupillary defect is not what Marcus Gunn described. congested retinal veins. (Courtesy of Mr M Wade, Department of
While the iris is illuminated, look for inflammation, arcus Medical Illustrations, Royal Perth Hospital, Australia.)
Neurologic Examination 17

13 14

13 Fundus examination showing cholesterol emboli (arrows) as bright, 14 Ocular fundus of a patient with inferior temporal branch retinal
orange-yellow, refractile, crystalline, glinting lipid emboli, so-called artery occlusion showing pallor of the inferior half of the retina
Hollenhorst cholesterol plaques.They are usually unilateral, multiple (arrows) due to cloudy swelling of the retinal ganglion cells caused by
and found at the bifurcation of retinal arterioles.They frequently arise retinal infarction. (Courtesy of Mr M Wade, Department of Medical
from ulcerated atherosclerotic plaque in the ipsilateral carotid system. Illustrations, Royal Perth Hospital, Australia.)
(Reproduced with permission from Hankey GJ and Warlow CP [1994]
Transient Ischaemic Attacks of the Brain and Eye.WB Saunders.)

15 16

15 Ocular fundus showing narrow arterioles and veins, pallor of the 16 Ocular fundus showing multiple small retinal infarcts due to fat
retina, and a cherry-red spot over the fovea (arrow) due to emboli (arrows). (Courtesy of Mr M Wade, Department of Medical
accentuation of the normal fovea, which is devoid of ganglion cells, by Illustrations, Royal Perth Hospital, Australia.)
the opalescent halo, in a patient with central retinal artery occlusion.
(Courtesy of Mr M Wade, Department of Medical Illustrations, Royal
Perth Hospital, Australia.)

17 Ocular fundus showing a retinal hemorrhage (arrow) due to 17


thrombocytopenia in a patient who presented with a symptomatic
intracerebral hemorrhage.
18 Neurologic Diagnosis

Nerves III, IV, and VI (oculomotor, trochlear, and Facial sensation


abducens nerves) Not necessary to test rigorously unless the patient complains
Ask the patient to look straight ahead at a target (e.g. the tip of facial sensory disturbance; patients usually know when
of a hat pin or pen; the use of a finger may miss subtle they have a disturbance of spinothalamic or trige-
diplopia) and examine the eyes in the primary position of minothalamic sensation because, for example, when they
gaze, noting the position of each eye (whether the eyes are have a shave or shower they may note the temperature of
conjugate or not), whether there are any involuntary the water feels different. Patients have much greater
movements of the eyes (e.g. nystagmus [vertical, rotational, difficulty in discerning a loss of other sensory modalities.
horizontal], opsoclonus, ocular bobbing, fixational instability:
square wave jerks), the size of the palpebral fissures (whether
there is ptosis or lid retraction on one or both sides) and
whether there is any fatiguability with prolonged gaze. 18
Ask if the patient sees one or two targets. If they have
double vision, assess this with a simple ‘cover-uncover’ test
that is accurate if one extraocular muscle is weak but can be
misleading if two or more muscles are weak:
• Find the position of the target that causes maximum
separation of the images.
• Holding the target steady in your right hand, cover one
of the patient’s eyes with your left thumb (pronating
your wrist and resting the dorsum of your distal left
fingers [flexed at the metacarpophalangeal joints] against
the patient’s lower central forehead), and ask which (if
any) object disappears – the outer (more peripheral)
object is seen by the defective eye and the inner (more
central) object is seen by the ‘good’ eye.
• Swing your left thumb to cover the patient’s other eye
and the other object should disappear. If the same object 18 Eye movements in each of the nine cardinal positions of gaze
disappears, the patient has not understood the test or the showing a left esotropia and left gaze palsy (in each eye, but
diplopia is monocular (i.e. intraocular pathology such as dysconjugate) due to an ischemic stroke involving the abducens nuclear
vitreous hemorrhage; or functional). complex in the left low pons.The patient had non-insulin dependent
diabetes mellitus. (Courtesy of Mr M Wade, Department of Medical
Ask the patient to follow a slowly moving target (no Illustrations, Royal Perth Hospital, Australia.)
faster than 30° per second, otherwise smooth pursuit will
be normally interrupted by saccadic intrusions) to examine
ocular pursuit in each of the nine cardinal positions of gaze 19
(straight ahead, left, right, up, down, up and to the right,
up and to the left, down and to the right and left) and
during convergence and accommodation. Ocular pursuit
should be smooth, maintaining the target fixed on the
macular, but may be interrupted by saccadic intrusions
(jerky, cogwheel or saccadic pursuit) in various disease states
such as extrapyramidal or cerebellar dysfunction. If double
vision occurs in any position of gaze carry out the
‘cover–uncover’ test described above. Nystagmus unrelated
to vestibular disease seldom produces symptoms and should
be closely looked for (see pp.107, 110, 138).
Look voluntarily in each of the cardinal positions of gaze
to test voluntary/saccadic eye movements that are generated
from frontal eyefields in Brodmann’s area 8 of the
contralateral frontal cortex (18, 19). A frontal lobe lesion may
result in a conjugate horizontal gaze palsy to the opposite side
and so the eyes will be deviated to the side of the lesion due
to unopposed action of the contralateral frontal lobe.

Nerve V (trigeminal nerve)


Corneal response 19 Eye movements in each of the nine cardinal positions of gaze
• Need not be tested routinely because it can be showing bilateral ptosis, skew deviation due to hypotropia of the right
unpleasant to the patient, carry a risk of corneal damage, eye, and a supranuclear upward gaze palsy due to an ischemic stroke in
and be unreliable. the left midbrain (causing a nuclear oculomotor palsy) and left
• Perform only if you really want to assess the integrity of thalamus caused by an embolus to the top of the basilar artery,
the trigeminal nerve (e.g. other cranial nerve damage or occluding the P1 segment of the left posterior cerebral artery.
suspected acoustic neuroma or syringobulbia). (Courtesy of Mr M Wade, Department of Medical Illustrations, Royal
• Use cotton wool. Perth Hospital, Australia.)
Neurologic Examination 19

Masticatory muscles head. If it lateralizes to one side this suggests either


• The bulk of the temporalis and masseter should be conductive hearing loss on that side (diminished masking of
examined by inspection and palpation; ask the patient to external sounds) or sensorineural hearing loss on the other
clench their jaw and feel the muscle bulk. side (poor bone conduction to that side). The Rinne and
• To test pterygoid muscle power, ask the patient to open Weber tests are not always reliable. Vestibular nerve function
the mouth fully. A unilateral trigeminal motor neuropathy is tested by eye movement and balance tests.
will manifest with deviation of the lower jaw (chin) to the
paretic side. Bilateral weakness, which may be suspected Nerves IX and X (glossopharyngeal and
if the patient needs to support the jaw with their hand vagus nerves)
during conversation, may be seen in myasthenia gravis Inspect the palate and ask the patient to say ‘Ah’. If the
and myopathies such as dystrophia myotonica. palate moves to one side this indicates a lesion of the vagus
• Less severe weakness may be detected by trying to close nerve (motor to the palate) on the contralateral side; the
the opened jaw with the heel of your palm. contracting palatal muscles pull the palate up and
ipsilaterally. If the palate does not move this may be due to
Jaw jerk bilateral supranuclear, nuclear or infranuclear vagus nerve
Unreliable and unhelpful as a screening test but it has a role lesions, depressed consciousness, or even a disturbance of
in determining the level of bilateral corticospinal tract neuromuscular junction or muscle function.
lesions; if brisk it suggests bilateral lesions above the mid- It is not necessary to test the gag reflex because it is
pons (a trigeminal nerve reflex: stretching and contraction unpleasant to the patient and is rarely abnormal in the
of the masticatory muscles). presence of normal palatal movement (i.e. it is very unusual
to have a palatal sensory deficit due to a IXth nerve palsy
Nerve VII (facial nerve) without a palatal motor deficit due to a Xth nerve palsy).
Facial muscle power will have been assessed informally during
history-taking: facial asymmetry, dysarthria. Ask the patient to: Nerve XI (accessory nerve)
• Raise their eyebrows (or look up to the ceiling): the • Ask the patient if they have neck pain; cervical
frontalis muscle is spared in contralateral upper motor spondylosis is common and neck pain may not only be
neuron (supranuclear) lesions due to bilateral uncomfortable but cause spurious weakness.
hemispheric representation. • Place the palm of your right hand over the patient’s left
• Smile and show their teeth: weak in lower and upper mandible and ask the patient to turn their head to the
motor neuron lesions of VII, and note any asymmetry. left against your resistance to test the power of the right
• Grimace by making a maximal effort to draw the lower sternocleidomastoid. Repeat on the other side.
lip and angle of the mouth downward and outward, at • Place your hand on the patient’s forehead and ask them
the same time tensing the skin over the anterior surface to push forward. This should usually be performed with
of the neck, to test the platysma. The examiner generally the patient lying supine because it may evoke neck pain.
needs to demonstrate the test and instruct the patient to Painless weakness is suggestive of motor neuron disease,
mimic him or her. myasthenia gravis, or a myopathy such as dystrophia
myotonica and polymyositis.
Altered, and even unpleasant taste, may be a symptom of • Ask the patient to elevate (shrug) the shoulders,
facial palsy. observing and palpating any asymmetry of contraction
of the upper trapezius muscles, and test muscle strength
Nerve VIII (auditory and vestibular nerve) by attempting to depress the shoulders by pushing down
Rub your thumb and middle finger together repetitively in on both shoulders against the patient’s resistance. The
both hands, about 2–3 cm (0.8–1.2 in) away from both of the lower portion of the trapezius can be tested by having
patient’s ears. Ask the patient if they hear the sound and if there the patient brace the shoulders backward and down.
is any asymmetry. Alternatively, rub your thumb and middle Unilateral paralysis of the trapezius is evidenced by
finger together repetitively next to one ear (to produce a inability to elevate and retract the shoulder and by
masking sound) and whisper quietly in, or hold a ticking clock difficulty in elevating the arm above the horizontal. The
adjacent to, the other ear (the testing ear) at varying distances. trapezial ridge is depressed, exposing the levator scapulae;
If there is any difficulty hearing, conduct the Rinne and the scapula appears rotated, the upper end laterally and
Weber tuning fork tests and inspect the external auditory down, the lower end up and in.
meatus and tympanic membrane. The Rinne test involves
striking the 256 Hz tuning fork and holding the vibrating Nerve XII (hypoglossal nerve)
tuning fork about 2–3 cm (0.8–1.2 in) from the ear. Ask the Ask the patient to:
patient if they can hear it. Then place the base of a vibrating • Open the mouth: inspect the tongue while it is at rest in
tuning fork behind the ear, over the mastoid process and the floor of the mouth, noting any wasting or fasciculation.
ask the patient if they can hear it and whether the sound is • Protrude the tongue: observe any deviation (toward the
louder ‘in the front’ (outside the ear) or ‘at the back’ (over side of a nuclear/infranuclear lesion, away from the side
the mastoid process). It is normally louder in the front of a supranuclear lesion).
(positive Rinne test), but if not (negative Rinne test), this • Move the tongue quickly from side to side: this is slow
suggests conductive hearing loss, due to blocked ears or a with pyramidal or extrapyramidal disease.
middle ear defect, for example. For the Weber test, the
vibrating tuning fork is placed over the vertex of the head Tongue power can be tested by asking the patient to
in the midline and the patient is asked where they hear the push the tongue into the cheek on each side but there is no
buzzing sound. Normally, it is heard in the middle of the reliable test for tongue power.
20 Neurologic Diagnosis

UPPER LIMBS As the MRC scale lacks sensitivity in discriminating the


Inspection most common degrees of muscular weakness between 4 and
Ask the patient to hold the arms outstretched, palms down, 5, the scale is often subdivided into: 3+, 4–, 4, 4+, 5–, 5 out
with fingers abducted and eyes open. Note any deformity, of 5; or alternatively 4, 4.25, 4.5, 4.75, 5 out of 5.
skin lesions, muscle wasting and fasciculations, and
involuntary movements. Reflexes
• Slight wrist flexion with hyperextension at the meta- • A brisk tap over the muscle tendon or a vibration wave
carpophalangeal joints, particularly if unilateral, is a sign induced by percussion of bone to which the tendon
of hypotonia, most commonly seen in cerebellar disease. attaches, stretches the muscle spindle and Golgi tendon
If bilateral it can be normal, particularly in Asian people. organs, and excites a monosynaptic phasic stretch reflex.
• Slow, unintended movements of the fingers (‘pseudo- • Stretching of the primary spindle endings activates group
athetosis’) suggest a loss of proprioception (‘sensory Ia afferent fibers which synapse directly with alpha motor
ataxia’). This can be confirmed by asking the patient to neurons in the anterior horn of the spinal cord. This
close their eyes and then touch the tip of their nose with results in contraction of the extrafusal muscle fibers
each outstretched index finger, in turn. The patient can innervated by these neurons.
then open their eyes, and carry out the finger-nose test • Stretching of secondary spindle endings activates group
of coordination (see below). II afferent fibers which are mainly inhibitory on extensor
• Tap on each forearm in turn, displacing it down by about motor neurons and facilitatory of flexor motor neurons.
10 cm (4 in) and observe the limb rebound to its former • Stretching of Golgi tendon organs activates group Ib
position; in cerebellar disorders, the rebounding move- afferent fibers.
ment is slow to stop so that the affected limb shoots past • Descending motor pathways (corticospinal, reticulo-
the initial position (rebound phenomenon). spinal, vestibulospinal) regulate alpha and gamma motor
• Ask the patient then to make rapid piano playing move- neuron excitability.
ments, repetitively tap the thumb and index finger, or
perhaps best of all for testing patterned movement, to alter- Deep tendon reflexes Main segment
nately tap two fingers (e.g. the index and middle fingers,
or ring and little fingers) whilst keeping the palm steady (if Jaw jerk Cranial nerve V (motor)
normal, an abnormality of motor function is unlikely).
• Ask the patient to keep the arms outstretched but to Biceps C5,6
supinate the forearms and face the palms up. Observe Brachioradialis/supinator C5,6
over the next 10–15 seconds. Triceps C6,7,8
• Mild flexion of the elbow and gradual pronation and Finger-flexion C8
downward drift of the forearm (‘pronator drift’) on one
side reflects a slight increase in tone of the pronator Reflexes may be depressed or absent if the reflex arc is
muscles compared with supinator muscles that occurs disturbed (e.g. anterior horn cell disease, peripheral
with mild pyramidal dysfunction. neuropathy, muscle disease) or if antagonists contract (e.g.
extrapyramidal rigidity, poor relaxation). However, in many
Tone muscle diseases (excluding inclusion body myositis) the
Muscle tone refers to the activity of the stretch reflexes, reflexes are relatively preserved (i.e. only mildly depressed)
which is usually assessed by the resistance encountered on compared with the degree of weakness. Depressed reflexes
stretching the muscle at increasing velocities. may be enhanced by reinforcement (e.g. clenching the teeth
Passively pronate and supinate the patient’s forearms, and tightly at the time of striking the tendon with the reflex
flex and extend the wrists and elbows, noting a steady hammer).
resistance that is insensitive to the velocity of stretch (‘lead Reflexes may be augmented by an upper motor neuron
pipe’ rigidity) and may be interrupted by tremor (‘cog- (supranuclear) lesion, anxiety, exercise, and thyrotoxicosis.
wheel rigidity’), or an increased resistance in proportion to The reflex may ‘spread’ (e.g. a tap over the distal radius may
the velocity at which the muscle is stretched (‘spasticity’). evoke contraction of the biceps, triceps or finger flexors) if
alpha motor neurons are hyperexcitable or if a wave of
Power vibration spreads to excite muscle spindles in distant
Test in an orderly sequence, proceeding from proximal to muscles, causing them to contract reflexly.
distal muscles, testing abductor groups before adductors,
and flexors before extensors. Coordination
Muscle power may be graded according to the Medical • Finger–nose test: ask the patient to touch the tip of their
Research Council (MRC) scale: nose, then the examiner’s finger which is held at arm’s
0 No movement. length in front of the patient, then their nose again. This
1 A flicker of movement in the muscle. may elicit a tremor (an action tremor accompanies the
2 The muscle can move the limb if the force of gravity is movement throughout its course whereas an intention
eliminated (e.g. support the joint so that it can be tremor increases in amplitude as the target is approached)
rotated around its vertical axis). or dysmetria: the finger falls short of the target or exceeds
3 The muscle can maintain the limb against the force of it (past-pointing) or deviates to one side or the other.
gravity. • Rapid alternating movements: dysdiadochokinesia.
4 The muscle can move the limb against resistance supplied • Rebound phenomenon.
by the examiner.
5 Normal power.
Neurologic Examination 21

Sensation Power
Ask the patient if they have noted altered sensation on any When testing the power of hip flexors, keep one hand under
part of the body, such as altered temperature sensation when the heel of the opposite foot if you suspect the patient is not
having a bath/shower. If so, the area in question can be trying hard to flex the hip (because of pain, malingering or
examined with a pin and a cold object, such as a part of the hysteria) because normal counter pressure will not be felt
metal tuning fork. The pin is advanced from the area of on the opposite side (Hoover’s sign).
apparent sensory impairment toward the normal area until a If lower limb weakness extends up to the trunk, ask the
line of demarcation is reached where sensation becomes patient to try to sit up from the lying position without the
normal. If not, and if there is no ‘pseudoathetosis’ of the out- use of the arms. If this is not possible, the trunk is weak.
stretched fingers (see above), a sensory disturbance is unlikely. The upper limit (the ‘motor level’) can sometimes be
Proprioception can be tested formally by taking hold of ascertained by noting the position of the umbilicus with the
the sides of one of the patient’s distal interphalangeal joints patient supine and then asking the patient to lift their head
between your left thumb and index finger and the sides of off the bed (to contract the rectus abdominis). If the
the distal phalanx between your right thumb and index umbilicus deviates upwards, this indicates that the lower
finger and passively move the patient’s distal phalanx up or abdominal muscles are weaker than the upper abdominal
down, asking the patient (with eyes closed) to state the muscles (Beevor’s sign of a lesion around T10 level).
direction of movement. Only small movements are required
as they should normally be detected. If not, try larger Reflexes
movements and, failing that, proceed proximally to check Deep tendon reflexes
movement at the proximal interphalangeal joints, wrist, and Hip adductors L2,3
elbow if necessary. Knee L3,4
Other sensory modalities that may be tested with the Hamstrings L5
eyes closed include: Ankle S1
• Two-point discrimination (normally >3 mm [>0.1 in] on
the pulps of the fingers). Polysynaptic reflexes that are often relayed through
• Stereognosis (ability to distinguish the nature, size and more than one segment
texture of small objects such as coins placed in the hand). Reflex Segment
• Graphesthesia (figure/number writing).
Corneal Cranial nerve V (sensory) and
The method of testing sensation depends on the VII (motor)
question or hypothesis being explored. If a central lesion is Abdominal
suspected (e.g. the patient also has a hemiparesis) then Upper T9,10
sensation should be tested ‘side to side’, comparing one part Lower T11,12
of the body with the corresponding contralateral side. Cremasteric L1,2
However, if a peripheral lesion is suspected (e.g. the patient Plantar S1,2
has a wrist drop) then the sensation in each of the relevant Bulbocavernosus S2,3,4
dermatomes (e.g. C6,7,8) and peripheral nerve regions (e.g. Anal S3,4,5
radial nerve) should be examined closely.
The plantar reflex
LOWER LIMBS The outer border of the sole of the foot (S1 dermatome) is
Inspection stimulated by a light stroke with a mildly noxious pressure
As for upper limbs, and also check for pes cavus deformity. stimulus such as a key or a spatula (bisected in the longitudinal
plane), after warning the patient of the impending stimulus.
Tone The great toe normally plantar-flexes. Until the age of
With the patient lying on the bed or couch, and the legs 18 months or subsequently, after damage to upper motor
relaxed, grasp the knee with one hand and rotate the leg neuron corticoreticulospinal pathways that inhibit flexor
briskly to and from about the hip. Normally, the foot inverts reflexes, the great toe extends (dorsiflexes) with or without
and everts passively. If tone is decreased, as in cerebellar and abduction (fanning) of the toes (Babinski sign). This is part
lower motor neuron disorders, the passive movement is of a greater uncontrolled protective flexor response which
increased. If tone is increased (as in spasticity and involves all the flexors of the lower limb and which is
extrapyramidal rigidity), the foot excursions are decreased normally inhibited from the brainstem by a reticulospinal
so that the foot moves rigidly with the leg. Because pathway so that flexor pathways may be used for volitional
disorders affecting tone usually have more impact distally, it movement such as walking.
is seldom necessary to test tone at the hip or knee.
Clonus is a rhythmic series of muscle contractions in Bulbocavernosus reflex
response to maintained stretch, that may be elicited at the The examiner flicks or pinches the foreskin of the penis or
ankle by supporting the patient’s knee, flexed about 15°, pricks the foreskin or glans with a pin to evoke a reflex
and then suddenly and forcefully dorsiflexing the patient’s contraction in the bulbocavernosus muscle at the base of the
foot with the palm of the examiner’s right hand. Up to penis. The contraction may be observed, but it is, as a rule,
three beats of clonus is normal. more easily palpated.
22 Neurologic Diagnosis

Anal reflex • Squat down and arise from a squat (to detect proximal
The tip of the finger, covered with a glove, is inserted into hip girdle muscle weakness).
the anal ring, and contraction of the anal ring is felt as the • Stand still with feet together, first with eyes open and
skin around the anus and the perineum is scratched or then shut, and notice any change in stability that may
pricked with a pin; or the contraction simply may be reflect impaired joint position sense (Romberg’s sign). If
observed without being palpated. This is a test specifically uncertain about the result, challenge the patient by
of the external or voluntary anal sphincter. In the presence standing behind them and, when the patient is stable
of a flaccid paralysis of the external anal sphincter, the with eyes open, tell the patient you are going to pull back
normal tonus of the internal sphincter is felt to give way on on the shoulders and ask them to try to remain upright
insertion of the finger. As the finger is withdrawn, the anus (if they cannot, they will fall back into your arms). If they
remains open or patulous. This does not indicate a loss of manage to remain upright, repeat this with the patient’s
internal anal sphincter function. eyes closed.

Coordination N.B. If the patient can walk, and if the history suggests
• Heel–knee–shin test: the patient lifts one leg, places it on a disturbance of gait or lower limb function, it is often
the opposite knee and runs it down the shin. preferable to begin the lower limb examination by asking
• Rapid tapping movement of the feet against the the patient to stand and walk.
examiner’s hand.
• Toe–finger test Functional consequences
It is important to observe the functional consequences of any
N.B. Tests of coordination require reasonable muscle neurologic impairment on the patient’s activities of daily
power. living, such as dressing, feeding, grooming, and walking.

Sensation CONCLUDING THE CONSULTATION


As for the upper limbs. Frequently patients have certain ideas about what their
symptoms are due to (and they may well be right) or they may
Stance and gait be fearful that they have a certain diagnosis (such as one that
• Ask the patient to walk 10 m (33 ft), turn around, and afflicted a friend or family member, such as a brain tumor). By
walk back. asking the patient ‘What do you think the problem is?’ can
sometimes be helpful in management (e.g. if the patient says
Note the speed of initiation of gait, the posture, stride they think it is due to stress and you agree with them), and
length, heel strike, and extent and symmetry of arm swing. sometimes it allows the clinician to be able to reassure the
As the patient turns, pay particular attention to the number
of steps taken to turn and any evidence of slight
overbalancing that may occur with extrapyramidal or
cerebellar lesions. Dystonic movements and tremor are also 20
often enhanced by walking. Pyramidal tract lesions cause the
foot to drag and the toes to scuff (spastic foot drop) because
of involuntary plantar flexion and inversion of the foot to
varying degrees. The knee flexes less than normal (or not at
all) and with unilateral lesions in particular, the pelvis rocks
to the normal side to help raise the dragged foot from the
floor and overcome weakness of the hip abductors (a
Trendelenburg-type gait).
Flaccid foot drop is manifest by the feet having to be
lifted high to avoid catching the toes on the floor. If the
foot is also stamped on the floor, joint position sense is likely
to be impaired. Foot drop may be bilateral (e.g. distal
[sensori-]motor, sciatic or peroneal neuropathies) or
unilateral. A high-stepping gait and stamping is a classical
sign of tabes dorsalis.
Ask the patient to:
• Walk heel-to-toe to test tandem gait; patients with a
lesion of the cerebellar vermis may only manifest truncal
ataxia and have no evidence of limb ataxia on finger–nose
and heel-knee-shin testing.
• Walk on the toes (to detect weakness of the gastrocnemii
due to a sciatic nerve or S1 nerve root lesion, for
example, that may be difficult to elicit otherwise).
• Walk on the heels (to detect weakness of tibialis anterior
due to a peroneal nerve or L4/L5 root, or pyramidal 20 ‘Scout’ view for a CT brain scan.This consists of a lateral view of
tract lesion for example). the skull (usually) with lines representing the axial image positions
superimposed. Note the number at the left-hand end of each line
corresponds with the axial image of that number.
Imaging the Brain 23

patient that they have not got what they a fearing. Alterna- IMAGING THE BRAIN
tively, the patient may be asked: ‘Do you have any concerns
about what the problem/diagnosis might be?’, or ‘Has anyone
suggested what the problem/diagnosis might be?’. COMPUTERIZED TOMOGRAPHY (CT) SCANNING
One of the most frequent complaints patients and Advantages
families make about doctors is that the doctor did not CT (20–24) is a great technique, particularly for acutely ill
explain (or they could not understand) what had happened patients, due to its rapidity, ease of access to the patient, and
and what was likely to happen. So conclude the consultation relatively wide availability. It is useful in acute neurology in
by asking the patient: ‘Do you have any questions or would patients with headache, focal neurologic deficit, loss of
you like to discuss anything else?’. consciousness, and suspected subarachnoid hemorrhage,
hydrocephalus, brain tumor, stroke, brain abscess, prior to
lumbar puncture and for demonstrating cerebrospinal fluid
(CSF) leaks, and for paranasal sinus and mastoid diseases. It
is vital following head and spine trauma, facial fractures,
skull base bone lesions and for post-operative neurosurgical
complications. In the spine it is most useful in trauma, for
lumbar discs, and combined with myelography for thoracic
and cervical lesions.

21–23 Axial views from a 21 22


normal CT brain scan
taken on soft tissue
window settings, at the
level of the fourth ventricle
21), the third ventricle
(2
22) and the bodies of the
(2
23).
lateral ventricles (2

24 Axial view of the skull 23 24


base from a normal CT
brain scan imaged on bone
window settings to display
bone detail optimally (but
at the expense of soft
tissue detail).
24 Neurologic Diagnosis

Disadvantages at the posterior fossa and works up to the vertex. A ‘scout’


CT uses ionizing radiation (an average brain CT scan view (like a lateral skull x-ray) showing where in the patient
exposes the patient to the equivalent of a year of the image slices have been taken, is usually displayed at the
background radiation!). It is not so good for demonstrating start or the end of the sequence of images (20).
white matter diseases (such as multiple sclerosis [MS]), Usually the right side of the patient is on the left side of
encephalitides, small lesions in the temporal lobes and the film, but check! – some scanners display the other way
posterior fossa (because of streak artefact from the adjacent round. A large ‘R’ or ‘L’ should indicate which is which.
bones) and spinal cord lesions. Each image shows the patients name, and often a date of
birth, and the date of the scan – all useful information:
Technique 50 year old females have a different range of disease from
A standard CT brain scan consists of 10 to 15 axial slices 20 year old males.
from the foramen magnum to the vertex at about 1.0 cm To read the images, follow a system much as you would
intervals (0.5 cm slice intervals through the posterior fossa). do to examine the patient, that way you will not miss things.
Modern scanners will produce many more thinner slices Start at the bottom of the head and work up: look at the
very quickly, but there is the danger of increasing radiation orbits, skull base including neural foramina, and petrous
without necessarily increasing diagnostic yield. bones. Then look at the intracranial structures: the IVth
The basic scan can be modified to include thinner slices at ventricle should lie slightly closer to the front of the posterior
narrower intervals to show greater detail (e.g. in the pituitary fossa than the back (21). It should also be in the shape of a
fossa or orbits), or at different scan angles (e.g. coronal), or ‘frown’. If it is nearer the back or starts to ‘smile’, there may
after an injection of intravenous iodinated x-ray contrast (e.g. be a mass distorting it.
iopamidol), or using different scan parameters to demonstrate The brain stem and midbrain and surrounding CSF spaces
bone optimally. A standard scan takes less then 2 minutes to should be symmetrical. The pituitary fossa, IIIrd ventricle,
perform with modern technology (not including the time basal ganglia, lateral ventricles, cerebral white matter and
taken to get the patient into and out of the scanner). cortex should all be symmetrical with similar appearance of
the frontal, parietal, temporal and occipital lobes. It is worth
Precautions ‘eyeballing’ these lobes and the basal ganglia in turn, this
Patients who are, or might be pregnant should not undergo helps to highlight subtle abnormalities (and make obvious
CT unless absolutely essential. If CT is essential, the ones more visible for the learner). If you do all this each time,
abdomen and pelvis should be shielded with lead aprons. you will not miss anything.
Patients with previous allergic reaction to iodinated Finally, if you cannot see anything wrong, double check
contrast should not be given contrast again (although it is the orbits, pituitary fossa, posterior fossa and basal ganglia
worth checking out the patient’s story in some detail as (review areas where people tend to miss things), and possibly
often so-called ‘allergic reactions’ are in fact something else). suggest that the patient be given contrast.
Patients with atopic history are more likely to have a de The review of spine images is on similar principles but the
novo contrast reaction than those without an atopic history. scout view is very useful as it helps you keep your bearings.
Scan techniques these days are complicated with lots of There is nothing worse than getting the disc level wrong.
different possible ways of conducting the investigation. In Count carefully and use the slice number to line up with the
order to get the answer to the patient’s problem with imag- numbering on the scout view. Really helpful scanners display
ing, it is essential to give the radiologist ALL the relevant a mini scout view in the corner of each scan image.
information or you may end up having to repeat the scan
because it was not done in the correct way (to demonstrate MAGNETIC RESONANCE (MR) IMAGING (MRI)
whatever disease condition) the first time, thus exposing the The principles behind how you produce a picture of the brain
patient to the inconvenience of an additional scan plus an by putting the patient into a strong magnetic field are
extra year’s worth of background radiation! The same applies completely beyond the scope of this book and will not be
to all radiologic investigations. There is no such thing as a discussed further. It is more important for the neurology
simple and hazard-free radiologic test. So use them wisely! student to know about the advantages and disadvantages of
MR and a few basics about what tissues look bright or dark.
Appearance of various tissues on CT The principles of reading an MR scan (25–28) are the same
The appearance of a tissue on CT depends on how much as for a CT scan, except that there are usually far more images
radiation it absorbed as the x-ray beam passed through it, to go through (so rather more daunting) including a midline
which in turn is dependent on the atomic number of its sagittal set (which are useful for impressing your tutors by
main components. The higher the atomic number, the looking for herniation of the cerebellar tonsils). Thus apply
greater the absorption. Densely calcified structures like bone the principles outlined above and you will not go far wrong.
absorb most radiation so look white; fresh blood is less
white; gray matter is gray but whiter than white matter; Advantages
white matter is gray but a bit darker; CSF (basically water) The principal advantages of MR are its great ability to
looks black; fat looks even blacker; and air (which absorbs demonstrate soft tissue detail especially in the brain and
virtually no radiation) looks very black. spinal cord, muscles, ligaments and cartilages.
In neurology it is very useful for demonstrating
HOW TO REVIEW A CT SCAN suspected white matter diseases (such as MS, encephalitis,
Most scans, be they CT or MR, are displayed in more or less metabolic disease), cervical and thoracic cord disease,
the same way on the hard copy. The hard copy is printed onto metastatic involvement of the spine, lumbar discs (though
radiographic film and transilluminated to view. There are CT is good for that too), precise delineation of tumors,
usually about 12 images on each film. Usually the scan starts posterior and pituitary fossa diseases.
Imaging the Brain 25

It can be used to show blood vessels (arteries and veins) aneurysm clip may move and tear the artery and the ocular
inside the head and in the neck (MR angiography [MRA]), foreign body may move causing an intraocular hemorrhage).
perfusion and diffusion of fluids in the brain and metabolites Relative contraindications include other metallic
in the brain (using advanced techniques such as MR prostheses or foreign bodies, or tattooed-in eyeliner
spectroscopy). (contains metallic particles which can cause burns). All
Images can be obtained in any plane, at millimeter patients are carefully screened for these factors and x-rayed
intervals, with paramagnetic contrast for demonstration of if necessary to exclude intraocular foreign bodies. Welders
areas of blood–brain barrier breakdown, and using all sorts particularly are prime suspects for intraocular foreign bodies.
of fancy sequences to highlight different features. Deodorants and talcs and eye makeup must be removed as
they may contain metallic bits.
Disadvantages Claustrophobic patients may have difficulty going into
Having a cardiac pacemaker or intracranial aneurysm clip or the very enclosed space of the scanner (though modern
metallic intraocular foreign body are all absolute contra- scanners are becoming more open access).
indications to MR (the pacemaker will stop working, the

25 T1W midline sagittal 25 26


view of the brain. Note
the brain appears as a
rather bland gray texture
and CSF is black.
Subcutaneous fat is
white.

26 T2W axial view of


the brain. Note the CSF
appears white, the gray
matter appears whitish
and white matter dark.
The gray matter is easily
differentiated from the
white.

27 Proton density (PD) 27 28


weighted axial view of the
brain. Note the CSF is
grayish and difficult to
distinguish from brain, gray
matter is whiter than white
matter.

28 Magnified T1W
coronal image of the
pituitary gland (long
arrow), optic chiasm
(arrow head), pituitary
stalk (short arrow), and
internal carotid arteries * *
(asterisks).
26 Neurologic Diagnosis

Appearance of various tissues on MR IMAGING THE CEREBRAL


The two basic sequences which are most commonly used CIRCULATION
are called ‘T1’ and ‘T2’ weighted.
On T1 weighted (T1W) images CSF looks black, gray
matter looks dark gray, white matter looks pale gray, fat CONTRAST ANGIOGRAPHY
looks white, blood varies with the age of the hematoma, This is an invasive technique which is used to obtain images
flowing blood, bone and air look black. of the cerebral blood vessels. It can be done by inserting a
On T2 weighted (T2W) images CSF looks white, gray catheter into the femoral artery under local anesthetic,
matter looks pale gray, white matter looks dark gray, fat wriggling it until the tip lies in one of the arteries to the
looks white (depending on where it is), blood varies with brain, then injecting about 5 ml (a teaspoon) of x-ray
the age of the hematoma, and flowing blood, bone and air contrast through it and taking x-ray pictures rapidly as the
look black. contrast passes through the blood vessels (29–33). In the
old days the images were taken directly onto x-ray film and
How to review an MR scan then the bones had to be subtracted away by hand (very
The method of reviewing an MR scan is the same as for CT tedious). Nowadays most modern angiography machines
(it’s a brain or spine after all!) but there are more images in take the images via a computer so that the bones can be
different planes to look at. The trick is to work through subtracted instantaneously, leaving the picture of the
them systematically. Do not expect the answer to leap out contrast outlining the artery and nothing else (digital
at you and do not feel conscious of how long you are taking subtraction angiography).
to review the film, just take your time and be thorough. A variation of this process is to inject a larger amount of
x-ray dye (up to 100 ml) through a large bore catheter
POSITRON EMISSION TOMOGRAPHY (PET) AND inserted into a peripheral arm vein, waiting until the bolus
SINGLE PHOTON EMISSION COMPUTED of contrast passes through the heart, round the lungs, back
TOMOGRAPHY (SPECT) SCANNING to the heart and up into the head arteries and then taking a
PET and SPECT are isotope techniques whose use is mainly rapid set of pictures of the blood vessels in the head or neck.
confined to research work. Some centers may use either or The bones are then subtracted off instantaneously as for the
both for cerebrovascular disease, diagnosis of dementias, intra-arterial version to leave just the blood vessels outlined
diagnosis of post-radiation necrosis, investigation of with x-ray contrast. This is called intravenous digital
temporal lobe epilepsy, but as neither is widely available, subtraction angiography (IV DSA), but results in poorer
both are expensive and require a source of isotopes, their quality images (than the intra-arterial version) and at some
use is likely to remain mainly experimental for the foresee- risk (due to the greater risk of allergic reactions, cardiac
able future. failure and arrhythmias due to irritation from the large
volume of contrast). The contrast gets diluted as it passes
OTHER MISCELLANEOUS TECHNIQUES
CT cisternography
This technique is used when a CSF leak from the skull base 29
is suspected, to identify the site of the leak prior to surgical 3
repair. About 5–10 ml of x-ray contrast (iopamidol 300 or
equivalent) is introduced into the spinal subarachnoid space 2
through a lumbar puncture and run up into the head by 2
putting the patient in the prone head-down position for a
few seconds. 3
The patient will experience a sudden, severe headache as
soon as the contrast enters the head due to the meningeal
irritation. The patient is then transferred to the CT scanner
in the prone position and scanned in the coronal plane 4
through the paranasal sinuses from the front of the frontal 4
to the back of the sphenoid sinuses. Leakage of contrast is
shown by the high density contrast passing into the sinus
from the cranial cavity and possibly by an associated break
point in the bone of the skull base.
This is an invasive procedure and should not be
undertaken unless a direct therapeutic maneuver is planned
and CSF has been demonstrated in the nasal cavity by litmus 1
paper test. Occasionally it may be justified in patients with
no definite leak but who have recurrent meningitis which is
likely to be due to a dural tear allowing a direct connection
from the nasal to the cranial cavity.

Isotope cisternography 29 Normal arch aortogram done by injecting contrast into the aortic
This technique is used in some centers to diagnose normal arch and then digitally subtracting the bones to leave the image of the
pressure hydrocephalus by demonstrating a slow rate of contrast in the aortic arch (1), common carotid (2), vertebral (3) and
clearance of an isotope from the ventricles. subclavian arteries (4).The origins of all the major head and neck
arteries are visible, though note there is some overlap.
Imaging the Cerebral Circulation 27

through the heart resulting in poorer opacification of the Interpretation of cerebral angiograms
blood vessels plus there is overlap making individual blood The detailed identification of intracranial arteries and veins
vessels difficult to see. IV DSA is not a good way of is a job for the neuroradiologist. The neurologist should
investigating the cranial circulation for all these reasons and know that the carotid arteries supply the front 2/3 of the
it is much better to use intra-arterial angiography. cerebral hemispheres, the vertebral arteries the back 1/3,
Intra-arterial angiography with selective injection of the brainstem and cerebellum, there are about 90 possible
carotid or vertebral arteries is the gold standard for the variations of the anastomotic pathways around the circle of
delineation of the cranial vasculature. It is risky and invasive Willis, and there are lots of potential collateral pathways
and often requires an overnight stay in hospital. In the between the intra- and extracranial arteries which can open
general population it has a 1% risk of stroke and 0.1% risk up if an internal carotid or vertebral artery gets blocked.
of death. In the subsection of older patients with The main abnormalities which one might see on an
symptomatic ischemic cerebrovascular disease who have angiogram include carotid stenosis (focal narrowing of the
been screened with doppler ultrasound and are known to artery), an aneurysm (focal little balloon-like outpouching),
have a carotid stenosis, the risk of intra-arterial angiography an occlusion (abrupt end to the artery), vasculitis (focal
is greater: 4% risk of stroke and 1% risk of death as a direct dilatation and narrowing of the fine branches of the
result of the procedure even in experienced hands. Thus it intracranial arteries), and arteriovenous malformations
should not be undertaken lightly. (irregular networks of wriggly arteries and veins which fill

30 31

3 2
3
2 1

30 Anteroposterior intra-arterial digital subtraction angiogram (DSA) 31 Lateral view of the same arteries as in 30.
showing the internal carotid (1), middle cerebral (2) and anterior
cerebral (3) arteries (arterial phase).

32 Lateral view of 32 33
same arteries as in
30 but in the
capillary phase.
1

33 Anteroposterior 2
2
view (DSA) of the
venous phase of the
intracranial
circulation following
intra-arterial 3
injection.The sagittal
3
(1), both transverse
(2) and sigmoid (3)
sinuses are clearly
seen.
28 Neurologic Diagnosis

quickly before the normal cerebral veins and can occur in in one go has led to the development of CT angiography.
any part of the head) and tumor blood supply (usually a In this technique a large intravenous injection of x-ray
‘blush’ or contrast with displacement or enlargement of contrast is given into an arm vein (around 100 ml) and a
arteries and veins). very rapid scan performed through the length of artery
which is to be imaged. The cross-sectional images thus
MAGNETIC RESONANCE ANGIOGRAPHY (MRA) obtained are processed through the scan computer to
MRA is a recent technique in which the patient lies in the produce 2D or 3D reconstructions of the arteries with the
MR scanner (so all the above contraindications and bones and soft tissues subtracted away.
precautions apply) and the scanner uses the fact that the This technique, like MRA, is very promising for the
moving blood will have different magnetization from the visualization of carotid stenoses, intracranial aneurysms and
static brain to produce an image of the blood vessels (34, arteriovenous malformations, but is still under evaluation and
35). This can be used to look at arteries or veins but there is not yet an alternative to conventional selective intra-arterial
are a number of pitfalls for the unwary: angiography. All the contraindications to CT and x-ray
• The patient must keep very still (i.e. not swallow if the contrast described above apply including the real risk of
examination is on the neck). precipitating cardiac failure or renal failure in elderly patients.
• A tight stenosis in the internal carotid artery may result
in a ‘flow void’ or area where the artery apparently ULTRASOUND
disappears due to the very fast flowing blood. Color doppler ultrasound is the method of choice for
• Although there have been numerous studies comparing screening patients for significant disease of the neck arteries,
MRA with conventional angiography and ultrasound and but only in experienced hands (36–39). There is no place
CT angiography, MRA is still undergoing development, for dabbling in it. It looks easy but is notoriously littered
has problems with observer reliability and should not be with pitfalls for the unwary inexperienced operator. It is
regarded as an alternative to conventional angiography however totally safe and pleasant for the patient and quick
just yet. – an average neck examination takes about 15 minutes.
Ultrasound of the brain in adults is of use in research but
CT ANGIOGRAPHY is not yet appropriate for use in clinical practice with the
The development of very fast CT scanners which can scan exception of intraoperative ultrasound which is increasingly
through a long distance in the body (around 10 cm [4 in]) used in neurosurgery.

34 36 36 Color Doppler
ultrasound. Normal internal
2 3 and external carotid arteries
in systole (top) and diastole
(bottom). Note the blue
flash in the carotid bulb in
1 systole (arrow) which is a
normal finding due to eddy
currents swirling in the
35 1 carotid bulb.

2 3

5
4

34, 35 Anteroposterior (3 34) and axial (3


35) views of an MRA (3D) of
the circle of Willis. Note the internal carotid (1), middle (2), anterior
(3) and posterior (4) cerebral arteries and the basilar tip (5). On the
left there is a dominant posterior communicating artery feeding the
posterior cerebral artery (arrow).
Imaging the Spine 29

37 IMAGING THE SPINE

MYELOGRAPHY
Myelograms were widely used in the past to investigate spinal
disease at all levels (called a radiculogram when confined to
the lumbar region). MRI has largely replaced myelography
although there is still occasionally the need for it in patients
who cannot tolerate MRI or who have pacemakers or other
internal devices which are contraindications to MRI.
About 10–12 ml of x-ray contrast are introduced
through a lumbar puncture (LP) into the spinal subarach-
noid space. The patient’s position is altered to make the
contrast, which is heavier than CSF, roll into the areas of the
spine to be examined. The contrast outlines the nerve roots
and spinal cord and a standard series of x-ray pictures are
taken (see 700, 709–711). The procedure is invasive, so all
37 Color doppler ultrasound. Origin of the vertebral artery (arrow) standard sterile precautions must be taken. The smallest
from the subclavian artery (arrowhead).This is usually visible unless the possible LP needle is used (this reduces post-LP headache).
chest is hyperinflated or the patient is very obese. Myelography is contraindicated in patients with epilepsy
(the contrast agent is epileptogenic) though this is relative,
and pregnant patients (though can be done with lead
shielding if absolutely necessary). Caution is required in
38 patients with asthma, atopic history or history of previous
contrast reaction.
After the myelogram, patients should be encouraged (1)
to drink extra fluid to keep well hydrated and (2) not to lie
flat as that helps stop the contrast entering the head and
causing a bad headache. Note this is contrary to standard
post-neurologic LP instructions. If the patient gets a
headache after the first 24 hours they can lie flat (at that stage
it is usually a low pressure headache due to continuous CSF
leakage through the LP site) and keep drinking extra fluid.

CT MYELOGRAPHY
After the x-ray contrast is put into the spinal CSF, CT scans
can be obtained through an area of interest to help delineate
discs or osteophytes, or whatever.
In the lumbar region, CT without contrast is useful for
examination of patients with suspected disc prolapse, but
38 Color doppler ultrasound.Vertebral artery in the vertebral canal. above the upper lumbar region it is no good for spinal cord
Note the regular shadows (arrows) cast by the vertebrae which or root abnormalities because there is not enough natural
intermittently obscure the view of the artery. contrast between the CSF and cord. Thus x-ray contrast must
be put into the CSF to examine the thoracic or cervical
regions. However, for purely bony problems such as deline-
ation of fracture fragments, plain CT is satisfactory (see 62).
39

39 Color doppler ultrasound. Color ‘power’ doppler ultrasound of the


circle of Willis looking axially across the cranium through the temporal
bone window.The patient’s face is to the left of the image and the back
of the head to the right.The tips of the internal carotid arteries
(arrows), the anterior (arrowhead) and posterior (short arrow)
cerebral arteries are visible.
30 Neurologic Diagnosis

MRI OF THE SPINE coverage), or of localized bits of spine using a spine coil
MR is an excellent tool for delineating all sorts of spinal or (more detail but less coverage). Thus if you are not sure
nerve root disease or disease of the surrounding bones (not where a lesion might be you are better to say that on the
fractures) (40–43). request form because then the radiologist will try to cover
Midline sagittal images can be obtained of the whole a larger area of the spine.
spine using a body coil (not so much detail but good

40 41

40, 41 MRI cervical spine of a 35 year old woman who presented


with a subacute Brown–Sequard syndrome of left hemisensory
disturbance below C5 and right leg weakness in a pyramidal
distribution. 40 Sagittal plane,T2W MR image, shows an ovoid shaped
white area of increased signal (arrow), representing demyelination, in
the upper cervical spinal cord at the level of C3, measuring about
1 cm (0.4 in) in cranio-caudal dimensions, and about 5 mm (0.2 in)
in maximum diameter. 41 Axial plane,T2W MR image at C3 showing
the lesion in the right hemi-cord (arrow).

42 43

42, 43 MRI cervical spine, fast spin echo T2W images, sagittal (4 42)
and axial (443) sections, showing a heterogeneous hypointense and
hyperintense mass in the left side of the upper cervical spinal cord (C1
to C3), and hyperintense edema extending superiorly to the obex and
inferiorly to the lower border of C5, with swelling of the cord.The
lesion is a cavernous hemangioma.
Lumbar Puncture (LP) and Cerebrospinal Fluid (CSF) Examination 31

LUMBAR PUNCTURE (LP) AND • If a lumbar puncture needs to be repeated within 1 or


CEREBROSPINAL FLUID (CSF) 2 weeks, do not use the same interspace because the
EXAMINATION needle may not go into the subarachnoid space but
rather an epidural or subdural collection of CSF that has
formed via a dural leak.
TECHNIQUE
• Ensure the patient is not drowsy and does not have Problems
papilledema or focal neurologic signs that are If the needle hits bone or a nerve root, evoking root pain
unexplained (i.e. that the patient is not at risk of coning down the leg, withdraw the needle to the subcutaneous
with lumbar puncture, see Complications below). tissue, check the alignment of the needle in the sagittal,
• Explain the procedure to the patient and why it is being coronal and axial planes, and re-insert it.
carried out. If CSF does not flow, despite the needle seeming to be
• Lay the patient on their side on the near edge of a firm in the subarachnoid space, do not attempt to aspirate CSF
bed, with the back and legs flexed as much as possible. but try rotating the open needle slightly. Rarely, a ‘dry tap’
Place a pillow between the knees and ensure the is due to a compressive lesion of the cauda equina or chronic
shoulders and pelvis are perpendicular to the floor. adhesive arachnoiditis.
• Ensure the patient is comfortable. If fresh blood emerges from the needle, the needle may
• Identify and mark the L4 and L5 vertebral spinous be in the epidural venous plexus or occasionally a small
processes, the former of which lies between the anterior blood vessel on one of the nerve roots, causing a ‘traumatic
superior iliac spines; the caudal part of the spinal cord tap’, or it may be from a subarachnoid hemorrhage. Try
ends about the level of the interspace between the first again through another disc space (e.g. L3/4).
and second lumbar vertebrae. If the lumbar puncture fails, try again through the L4/5
• Wash your hands and put on a sterile gown and gloves. interspace, and then if necessary the L3/4 interspace. Do not
• Sterilize a large area of skin over the lumbosacral spine go any higher than this because the caudal part of the spinal
and drape the area. cord, which ends about the level of the interspace between
• Draw up 7 ml of 2% lignocaine and inject the skin and the first and second lumbar vertebrae, may be injured by the
subcutaneous tissue down to the supraspinous ligament, needle. Attempts lower than the L4/5 interspace may not
and even the epidural space, between the L4 and L5 be fruitful because the dural sac containing CSF may have
vertebral bodies. ended. Alternatively, try again with the patient sitting up
• Wait a few minutes for the local anesthetic to work, rather than lying on their side, and consider using a standard
during which time check that the stylet tip is smooth and needle in patients with a high body mass index; atraumatic
flush with the outer hollow needle, and assemble the needles have a higher failure rate than standard needles in
three-way tap to the manometer. patients with a high body mass index.
• Insert the lumbar puncture needle (20 gauge atraumatic If lumbar puncture is impossible, a lateral cervical
needle) in the midline of the L4/5 interspace, and advance approach under x-ray control is an alternative. Cisternal
it in the sagittal plane, parallel to the floor and at a slight puncture should not be required.
angle towards the patient’s umbilicus (44). Resistance is
felt as the needle penetrates the supraspinous ligament and
again at the dura. After penetrating the dura, remove the
stylet and, if the needle is in the spinal subarachnoid space,
CSF will flow out of the needle. Attach the three-way tap
and manometer and wait a minute or so for the CSF to
rise up the manometer and stabilize before recording the
CSF pressure (in millimeters of CSF; normal less than
about 200 mm CSF). Check that the CSF moves up and
down a little in the manometer with respiration or with
relaxing the legs (i.e. extending the hips a little).
• Empty the CSF from the manometer into one sterile
bottle for biochemical examination (protein, glucose,
IgG/albumin ratio, oligoclonal bands) and then allow 44
another 3–5 ml of CSF to run from the needle into a Dura mater
sterile bottle for microbiologic examination (cells, Subarachnoid
L4 space
culture) and another bottle for DNA analysis (e.g. Interspinous containing CSF
polymerase chain reaction), spectrophotometry, or ligament
cytology. Label the sterile bottles first, second and third. Vertebral body
L5
• Remove the needle, place a band-aid/plaster over the
skin puncture and advise the patient to lie down in any Supraspinous Intevertebral
comfortable position for an hour or so before getting up. ligament disc
S1
Warn the patient that if they get a headache when sitting Skin
or standing up, to lie down and drink plenty of fluids.
• Take a simultaneous blood glucose test.
• Send the CSF and blood specimens to the appropriate
laboratories immediately. Otherwise refrigerate them at 44 Midline sagittal section through the mid-lumbar spine to show the
4°C (39.2°F). course of a lumbar puncture needle.
32 Neurologic Diagnosis

COMPLICATIONS CONTRAINDICATIONS
• Herniation of the medial temporal lobe through the • Intracranial space-occupying lesion (e.g. abscess, hema-
tentorial opening (transtentorial herniation) or of the toma, tumor), particularly in the posterior fossa.
medulla through the foramen magnum (coning), leading • Obstructive hydrocephalus.
to medullary compression and death (45). This is the most • Generalized brain edema with obliteration of the CSF
serious complication but can be avoided by never doing a cisterns around the upper brainstem.
LP if there is clinical evidence of raised intracranial pressure • If a CT scan is not available, then LP is contraindicated
(see p.477), or focal cerebral or posterior fossa signs. If in patients with clinical features suggestive of the above
there is raised intracranial pressure with shift of the three conditions (e.g. papilledema and/or focal neuro-
intracranial contents, or obstruction to CSF flow, then logic signs and/or coma). If bacterial meningitis is
removing CSF by LP, or continuing leakage of CSF after suspected, it is essential to treat the patient immediately
the LP, may create a lower pressure in the vertebral canal and empirically with broad spectrum antibiotics (after
than in the intracranial compartments, leading to mass collecting blood cultures) rather than risk brain
movements and herniation of the brain. On the other herniation by performing a lumbar puncture in order to
hand, if there is diffusely raised intracranial pressure with obtain CSF and isolate the organism (see p.273).
free flow of CSF through all parts of the intracranial and • Infection of the skin in the lumbar region.
spinal CSF compartments, which can only be discerned • Bleeding diathesis.
with CT or MRI scan, then LP should be safe.
• Spinal nerve root damage, usually caused by inserting the NORMAL VALUES
needle lateral to the midline. • Pressure. Less than 250 mm of CSF/water, and usually
• Low-pressure headache (30% [1–70%] of LPs) due to less than 200 mm, if the patient is relaxed and there is
continuing CSF leakage through the hole in the dura. It is free flow of CSF (the CSF fluctuates in the manometer
present only after sitting or standing for minutes to hours with respiration and coughing). Falsely low CSF pressure
and is relieved by lying down. If the headache comes on may be due to removing too much CSF before
suddenly or is accompanied by drowsiness, then perform a measuring the CSF pressure, a CSF leak around the
CT scan to see if an intracranial subdural or subarachnoid needle, transtentorial herniation and brainstem coning.
hemorrhage is present. Treatment otherwise involves the • Red blood cells: normally the CSF is clear with no red
patient lying flat in bed with the foot of the bed elevated blood cells (RBC). If the CSF is bloody it should be
and drinking plenty of fluids. If the headache persists then centrifuged immediately and the supernatant examined
it can be stopped in some patients by an autologous blood by spectrophotometry (or, if not available, the naked
‘patch’ in the epidural space over the site of the presumed eye). Yellow (xanthochromic) pigmentation is due to the
dural hole or, as a last resort, by open surgery to seal the breakdown of products of hemoglobin (e.g. oxyhemo-
dural leak. Strategies to minimize the incidence of post-LP globin and bilirubin), and is seen in patients with
syndrome are to use a needle of small diameter and subarachnoid hemorrhage (at least 12 hours before),
atraumatic shape, and to reinsert the stylet before removing jaundice or a very high CSF protein. Pinkish pigmen-
the needle. Otherwise a strand of arachnoid that may have tation is due to hemoglobin from disintegrated RBCs.
entered the needle with the outflowing CSF may be
threaded back through the dural defect and facilitate
prolonged CSF leakage along the arachnoid.
• Infection of the CSF, or an epidural abscess, if sterile 45
precautions are not taken or if the needle is inserted
through inflamed or infected skin.
• Transient back stiffness. If persistent, consider a spinal
hemorrhage.
• Spinal hemorrhage (epidural, subdural, or subarachnoid).
This may manifest as severe back and/or nerve root pain,
nerve root compression or spinal cord compression, but
is very rare unless the patient has a bleeding diathesis
(e.g. platelet or coagulation defect).
• Intracranial subdural and subarachnoid hemorrhages are
very rare.

INDICATIONS
• Diagnosis: suspected subarachnoid hemorrhage, menin-
gitis, encephalitis, MS, dementia and idiopathic intra-
cranial hypertension.
• Investigation: previously to introduce contrast media
such as metrizamide for myelography and CT scanning, 45 A potential complication of lumbar puncture if the patient has an
radioisotopes for ventriculo-cisternography, and air for intracranial mass lesion. Photograph of the under-surface of the
pneumoencephalography; MRI has superseded these. cerebellum showing swelling and grooving of the inferior mesial parts
• Treatment: to introduce local spinal anesthetics and occa- of both cerebellar hemispheres (mainly the ventral paraflocculi or
sionally antibiotic or antitumor agents into the subarach- tonsillae) (arrows) which have herniated, with the medulla, through the
noid space, and to remove regularly CSF to lower the CSF foramen magnum, and compressed the medulla. (Courtesy of
pressure in idiopathic intracranial hypertension. Professor BA Kakulas, Royal Perth Hospital, Australia.)
Electroencephalography (EEG) 33

• White blood cells: <5 lymphocytes per mm3 and no ELECTROENCEPHALOGRAPHY (EEG)
polymorphs. If the CSF is contaminated by blood from
a traumatic tap, one alternative is to repeat the CSF after
a few hours (through a different lumbar interspace) or DEFINITION
to calculate the number of white blood cells (WBC) that The EEG is a recording, by means of electrodes attached to
have been introduced into the CSF from the traumatic the scalp, of the electric field generated by the spontaneous
bleed by using the following formula: (WBC in neuronal activity of the underlying brain. The recording
peripheral blood/RBC in peripheral blood) × RBC CSF. reflects the electric currents that flow in the extracellular space
As there are about 1000 times more RBC in the and these in turn reflect the summated effects of innumerable
peripheral blood than WBC, this formula is usually about excitatory and inhibitory synaptic potentials that occur in
0.001 × RBC CSF. An estimate of the true CSF WBC cortical neurons. The spontaneous activity of cortical neurons
count can then be made by subtracting the number of is influenced by subcortical structures, especially the thalamus
WBC introduced from the number observed but, as this and rostral brainstem reticular formation.
tends to be an over-estimate, it is best to consider a definite
excess CSF WBC count as the ratio of observed CSF WBC TECHNIQUE
to calculated number of WBC introduced exceeding 10. Electrodes (usually 21), which are solder or silver–silver
• Glucose: normally more than 50% of the simultaneous chloride discs 0.5 cm (0.2 in) in diameter, are placed
blood glucose. equidistantly over the surface of the scalp according to an
• Protein: 0.15–0.45 g/l (15–45 mg/dl), depending on international convention (10–20 System) (46). They are
the laboratory. A very high CSF protein concentration attached to the scalp by means of adhesive material such as
causes the CSF to clot (Froin’s syndrome). collodion or bentonite (bentoquatam, quaternium-18
• Immunoglobulin G (IgG) is a component of the CSF bentonite), using EKG paste under the electrode to make
protein, normally making up less than 15% of the total contact with the scalp. They are wired up to separate
protein. amplifying units, or ‘channels’. Potential electric differences
• IgG/albumin ratio in the CSF is normally less than between pairs of electrodes or combinations of electrodes
about 0.25. can be recorded, amplified, and displayed on an oscilloscope
• IgG index ([CSF IgG/serum IgG]/[CSF albumin/serum or digital screen or a paper trace moving at a standard speed
albumin]) is a better measure of CSF IgG because it takes of 3 cm/s. The resulting EEG or voltage-versus-time graph
into account the serum level of IgG and albumin; if the appears as a number of parallel wavy lines, each representing
IgG level in the blood is abnormal, it is otherwise difficult one channel.
to interpret the level of IgG in the CSF.
• Oligoclonal bands: perhaps a more sensitive measure of
locally synthesized immunoglobulin in the CSF. Polyacry-
lamide gel electrophoresis separates two or more gamma
globulins (IgG) in the CSF on the basis of their charge
and size, which appear as two or more bands. Need to
check that no similar bands are present in the serum.

Causes of an increase in CSF IgG and


oligoclonal bands
• Increased IgG in the blood and breakdown of the blood-
brain barrier. 46
• Contamination of the CSF by red cells.
• Multiple sclerosis.
• Meningitis.
• Encephalitis.
• Acute post-infectious encephalomyelitis. Fp1 Fp2
• Subacute sclerosing panencephalitis.
• Cerebral infarction.
• Brain tumors. F7 F3 Fz F4 F8
• Neurosyphilis.
• Sarcoidosis.
• Cerebral systemic lupus erythematosus.
• Acute inflammatory demyelinating polyradiculo-
neuropathy (Guillain–Barré syndrome). T3 C3 Cz C4 T4

CSF taken from the cervical spinal canal and the


ventricles contains fewer cells and has a lower protein
T5 P3 Pz P4 76
concentration (up to 0.15 g/l [15 mg/dl]) than lumbar
canal CSF, but the glucose concentrations are similar in the
different compartments. O1 O2

46 The international 10–20 electrode system.


34 Neurologic Diagnosis

Patients are usually examined in the awake state, lying or Abnormal EEG recordings
sitting down in a quiet room with their eyes closed. The • Absent brain waves – generalized ‘electrocerebral silence’:
resting electrocerebral activity is recorded for about the electric activity of the cortical mantle measured at the
30 minutes, during which time a number of ‘activating’ scalp is absent or less than 2 μV. This may be due to
procedures are usually carried out: artefacts (which can be identified as the gains are
increased), hypothermia or acute intoxication with
• Sleep: the patient is allowed to fall asleep, because sleep may anesthetic levels of drugs, such as barbiturates, and severe
enhance potentially epileptogenic or epileptiform activity. diffuse cerebral ischemia and hypoxia.
• Hyperventilation: the patient is asked to breathe deeply • Absent brain waves – localized: indicates a large area of
20 times a minute for 3 minutes, evoking hypocapnia, brain softening, tumor or extra- or subdural hematoma.
alkalosis and cerebral vasoconstriction. The rhythm slows This is rare because most lesions are not large enough to
and a paroxysm of spike wave activity or another abolish brain waves or prevent recording of abnormal
abnormality may be induced. waves arising from the borders of the lesion.
• Photic stimulation: a powerful flashing light (strobo- • Slow waves (delta or theta) – generalized: encountered
scope) is placed about 40 cm (16 in) from the patient’s in coma and any diffuse brain disorder such as a
eyes and flashed at varying frequencies of 1–20 per metabolic/toxic disturbance.
second with the patient’s eyes open and closed. The EEG • Slow waves – localized: recorded over the site of any focal
over the occipital region may show waves corresponding brain lesion such as a hemorrhage, infarct, herpes simplex
to each flash of light (evoked response or photic driving) encephalitis, tumor.
or to abnormal discharges (e.g. a photoparoxysmal • Spikes or sharp waves: transient high-voltage waveforms
response). that have a pointed peak at conventional paper speeds and
a duration of 20–70 ms. Spikes or sharp waves that occur
Other techniques that can be used to identify epileptic interictally are referred to as epileptiform discharges.
foci include: • Spike discharges or sharp waves – localized: indicate a
potentially epileptogenic focus.
• Sleep recordings, which may reveal abnormalities not • Combined spike and slow wave (spike wave) discharges
evident when the subject is awake or in light sleep. – generalized: ‘typical’ if well formed at 3 Hz; ‘atypical’
• Prolonged (6–8 hours) telemetric recordings. if of higher or lower frequency than 3 Hz and if their
• Video EEG. pattern includes multiple spikes (polyspike wave
complexes) (47). Such patterns are commonly associated
RECORDINGS with epilepsy.
Normal EEG recordings • Periodic discharges: may occur regularly in certain
Awake pancortical diseases such as Creutzfeldt–Jakob disease
Amplitude: 50–200 μV. (CJD), subacute sclerosing panencephalitis (SSPE) and
Waveforms/rhythms: metabolic encephalopathies.
• Alpha waves: 8–13 per second (Hz), 50 μV sinusoidal
waves, recorded over the occipital region when the eyes
are closed and are attenuated or suppressed completely
by eye opening or mental activity. The occipital alpha
rhythm depends on synchronous discharge of cells in the
visual cortex.
• Beta waves: 14–22 Hz, low amplitude (10–20 μV) is
recorded maximally and symmetrically from the frontal
regions.
• Theta waves: 4–7 Hz, is prominent in children, gradually
diminishing during adolescence. A small amount of theta 47
activity may be normal over the temporal regions,
somewhat more so in older people (over 60 years of age).
• Delta waves: 1–3 Hz, 50–350 μV is a normal finding
only in early childhood; it is not present in the normal
waking adult.
• Mu rhythm: of alpha frequency but has a characteristic
appearance like the Greek letter μ or the top of a picket
fence, and is distributed asymmetrically in the central
regions of the head.
• Lambda waves: generated in the occipital region by
saccadic eye movements when the eyes are open.

Sleep
The alpha rhythm slows symmetrically and characteristic
waveforms (vertex sharp waves and sleep spindles) appear.
If sleep is induced by benzodiazepine or barbiturate drugs, 47 16 channel electroencephalograph of a patient who was confused
an increase in fast frequencies occurs normally. but able to answer simple questions, showing epileptiform features in a
patient with non-convulsive status epilepticus (arrowheads).
Nerve Conduction Studies 35

ROLE NERVE CONDUCTION STUDIES


• Accurate diagnosis of epilepsy (see p.71).
• Determine the nature of some pancortical disturbances:
SSPE, CJD. These involve percutaneous stimulation of peripheral nerve
• Confirm the clinical localization of some brain lesions fibers and recording of the compound muscle and sensory
(imaging techniques do this better). nerve action potentials.
• Monitor progress (e.g. HSE).
MOTOR NERVE CONDUCTION STUDIES
An EEG should only be requested if there is a clear An accessible motor or mixed sensori-motor nerve (e.g. the
question to be answered and the question is answerable by median nerve at the wrist) is stimulated electrically through
means of EEG. A succinct and relevant clinical history is surface electrodes applied to the skin and the resulting
crucial to accurate interpretation of the recording. compound muscle action potential (CMAP) is recorded by
surface electrodes on the skin over the motor end point of
INTERPRETATION the muscle (e.g. abductor pollicis brevis) (48). The CMAP
Considerable training, skill and caution is required to is filtered, amplified and displayed graphically on a digital
interpret accurately the EEG. There is considerable variation screen. The amplitude of the CMAP is measured in
in the normal EEG and all too often minor deviations can millivolts and reflects the number of depolarized muscle
be erroneously diagnosed as abnormal or, even worse, as fibers. A reduced CMAP amplitude may indicate:
indicative of epilepsy or other brain disorders.
• Fewer motor fibers in the motor nerve (axonal
neuropathy).
• Fewer muscle fibers in a motor unit (denervated muscle).
• Temporal dispersion of conduction velocities
(demyelinating neuropathy).

The delay (latency) between the time that the stimulus


48 was delivered to the time of onset of the CMAP is measured
in milliseconds (e.g. the distal motor latency for median
A nerve stimulation at the wrist may be 3.6 ms in a patient).
If the same nerve is stimulated again, but at a more
B proximal point (e.g. the cubital fossa), the distance
measured between the two stimulus sites (e.g. 200 mm) can
C be divided by the difference in latency for the two responses
(7.6 ms–3.6 ms = 4 ms) to produce a conduction velocity
D in m/s (e.g. 50 m/s).
In newborn infants the motor conduction velocity is
E about one-half that of adults and increases with age to reach
‘adult values’ at about 3–5 years. The conduction velocity
F is a measure of the conduction in the fastest conducting
fibers in the nerve; the large diameter, well myelinated
nerves. Small myelinated and unmyelinated nerve fibers are
not assessed. Nerve conduction velocity is also influenced
A by temperature, decreasing by about 1.9 m/s per °C below
normal temperature. Because proximal nerve fibers have a
B
larger diameter and are a little warmer than distally,
conduction velocities are a little faster in proximal nerve
segments. Limb temperature must be known and controlled
C
before nerve conduction studies can be carried out and
interpreted reliably.
D
Normal distal motor latency to onset of CMAP ms
E Median nerve (wrist to abductor pollicis brevis, 6–8 cm) <4.2
Ulnar nerve (wrist to abductor digiti minimi, 6–8 cm) <3.5
F
Normal motor conduction velocities m/s
Median nerve in forearm >49
48 Upper diagram illustrates the CMAP recorded from the Ulnar nerve in forearm >51
hypothenar eminence after electrically stimulating the ulnar nerve at Common peroneal nerve (extensor digitorum brevis) >41
various points in the upper limb: the supraclavicular fossa (A), axilla (B), Tibial nerve >40
above and below the elbow (C, D, E) and wrist (F).
Lower diagram illustrates the SNAP recorded from various points in SENSORY NERVE CONDUCTION STUDIES
the upper limb after electrically stimulating the ulnar nerve in the little Antidromic conduction in accessible sensory or mixed
finger (orthodromic conduction). Note the different time and voltage sensori-motor nerves can be assessed by electrically
scale compared with motor nerve conduction studies above. stimulating the sensory nerve proximally (e.g. the median or
36 Neurologic Diagnosis

ulnar nerve at the wrist) and recording the resulting sensory Axonal degeneration
nerve action potential (SNAP) distally (e.g. from ring Follows most metabolic, toxic and nutritional diseases of
electrodes around the index or little fingers respectively) peripheral nerves, which result in a disturbance of the
(48). Alternatively, orthodromic sensory conduction can be metabolism of the cell body, or perikaryon, that impedes fast
assessed by electrically stimulating the distal sensory fibers axonal transport and other functions so that the most distal
(e.g. of the median nerve in the index finger) through parts of the axon (which may be up to 1 m (3.3 ft) from the
surface (ring) electrodes applied to the skin and recording cell body) are affected first and die back from the periphery
the SNAP by surface electrodes on the skin over the nerve (‘dying-back neuropathy’).
more proximally (e.g. the median nerve at the wrist).
The SNAP reflects the number of nerve fibers that have Neuropathology
responded to the stimulus and conducted to the recording Axonal degeneration of the distal portion of the nerve fiber,
point. Lesions of the sensory nerves distal to the dorsal root similar to the Wallerian degeneration.
ganglia lead to a reduction or loss of the amplitude of the
SNAP. Sensory nerve action potentials are of very small Neurophysiology
amplitude (10–20 μV) compared with CMAPs (2–20 mV), The intact proximal segments conduct normally but
and consequently sensory nerve conduction velocities are conduction fails over the distal degenerating part of the
more technically demanding and less reliable than motor axon in the same way that it does in Wallerian degeneration.
nerve conduction studies. Sensory nerve conduction
velocity can be calculated by recording the SNAP at two Segmental demyelination
separate points over a nerve. Follows primary damage of the myelin sheath by a
disturbance of Schwann cell metabolism (e.g. some
Normal sensory action potentials hereditary neuropathies), a direct immune-mediated attack
Latency to peak (ms) Amplitude (μV) on the myelin or the Schwann cell (e.g. Guillain–Barré
Median nerve <3.5 >15 syndrome), or toxic damage to the myelin sheath (e.g.
(wrist to index finger, 12–14 cm) diphtheria toxin).
Ulnar nerve <3.1 >10
(wrist to little finger, 10–12 cm) Neuropathology
Sural nerve <4.0 >6 • Demyelination usually begins paranodally and tends to
(point B, 14 cm) affect peripheral nerves proximally (e.g. the roots) and
very distally at the nerve terminal more than (or, as much
These values depend on the age of the patient. The as) intervening regions.
temperature of the limb and the distance between the electric • The axon remains intact unless demyelination is severe,
stimulus and the recording site need to be controlled. causing secondary axonal degeneration.
• Remyelination occurs when Schwann cells divide and
PATHOLOGIES form new internodes of irregular length with thin myelin
Four pathologic processes may affect peripheral nerves. sheaths.
• Repeated episodes of demyelination and remyelination
Wallerian degeneration results in the formation of concentric layers of Schwann
This follows mechanical injury (e.g. transection or crush) or cell cytoplasm around the axon (‘onion-bulb’ formations).
ischemia (e.g. vasculitis) of nerve fibers (49).

Neuropathology
• Within 4–5 days, both the axon and myelin distal to the
site of injury degenerate.
• The degenerating myelin forms linear arrays of ovoids 49
and globules along the degenerating axon.
• The axon of the proximal segment regenerates forming
sprouts which grow distally, and some re-innervate the
surviving distal neurilemmal tubes of the Schwann cell
basement membrane, inside which the Schwann cells
have divided and arranged themselves in line. The
regenerating axons are then myelinated by the Schwann
cells but the nerve fibers are smaller in diameter and have
shorter internodal lengths than originally.

Neurophysiology
• Following nerve transection, the fibers continue to
conduct impulses at normal or near normal conduction
velocities until about 1–4 days later when the fibers
become totally inexcitable and conduction ceases
altogether. 49 Autopsy specimen of the thoracic spinal cord in cross section
• Conduction may be restored following nerve fiber showing evidence of Wallerian degeneration of the contralateral and
regeneration but conduction velocities are rarely as fast ipsilateral corticospinal tracts (arrows) secondary to a more proximal
as previously. corticospinal tract lesion (i.e. internal capsule lacunar infarct).
Nerve Conduction Studies 37

Neurophysiology (H-wave) after a latency that is much longer than that of the
• Demyelination may result in slowing of conduction, direct motor response (M-wave).
conduction block, inconsistent and incomplete trans- Thus, the H-reflex is the electric representation of the
mission of rapid trains of impulses, and increased tendon reflex circuit and provides a means of quantitating
susceptibility to changes in temperature. reflex changes; it is decreased or absent in most peripheral
• In normal myelinated fibers, impulses conduct rapidly from neuropathies and increased in upper motor neuron
one node of Ranvier to the next by saltatory conduction. disorders. It is rarely assessed nowadays in routine neuro-
• In demyelinated nerve fibers conduction between nodes physiologic practice.
is delayed or, as with unmyelinated nerve fibers,
conduction becomes continuous across the demyelinated REPETITIVE STIMULATION STUDIES
segments. The delay in conduction results in slowing of Normally, if repeated supramaximal electric stimuli are
conduction velocity, temporal dispersion, and therefore delivered to a nerve (e.g. the ulnar nerve at the wrist) at
a slightly lower amplitude of the action potential. rates of up to 25 per second (which most patients would not
• Conduction block is present when a proportion of nerve be able to tolerate), each motor response (CMAP) will have
fibers fail to transmit any electric impulses across a the same form and amplitude. If the stimulus continues at
demyelinated segment. When the stimulating electrodes this rate for more than 60 seconds or so, fatigue will occur
are moved proximally along the nerve segment, the and a decrement in successive CMAPs appears.
CMAP attenuates by more than 50% in area and In myasthenia gravis, the initial CMAP produced by
amplitude between a distal and proximal site of electric stimulation is normal (or reduced) but supramaximal
stimulation, indicating failure of conduction in at least stimulation at 2 Hz results in a decrement in amplitude of
some nerve fibers. Conduction block may be caused by the first four responses (50). In the Lambert–Eaton
focal nerve compression and inflammatory neuropathies. myasthenic syndrome, post-exercise facilitation is seen with
a marked increase from initial very low amplitude potentials
Neuronopathy or primary nerve cell degeneration toward normal amplitude motor unit potentials after
• Primary destruction of nerve cell bodies in the anterior 10 seconds of exercising the relevant muscle. This is also a
horn cells (e.g. spinal muscular atrophy, poliomyelitis, feature of botulism.
amyotrophic lateral sclerosis) or dorsal root ganglia (e.g.
hereditary, Friedreich’s ataxia, paraneoplastic). CENTRAL MOTOR CONDUCTION
• The peripheral sensory axons degenerate distally and the Central motor conduction down the corticospinal tract can
ascending sensory tracts in the posterior columns and be measured by electromagnetic stimulation of the motor
other spinal tracts degenerate proximally. As the cell cortex and recording the CMAPs and latencies in distal limb
bodies are destroyed, there is no recovery. muscles.

LATE WAVES
F-waves
F-waves are late, small motor responses that are evoked by
a supramaximal stimulus of a motor nerve. The stimulus to
the nerve (e.g. the median nerve at the wrist) not only
activates motor fibers that travel orthodromically to produce
the initial direct muscle action potential but also activates 50
motor fibers that travel antidromically to the anterior horn
cells. Here, some anterior horn cells may be activated to
produce a later orthodromic response. So, after a latency
longer than that for the direct motor response, a second Normal
small muscle action potential is recorded (F-wave).
The latency of the F-wave is a measure of the time of
conduction in motor fibers from the site of stimulation to
the motor neuron and then back again to the recording site,
and can be a measure of conduction in the proximal
segments of nerves and spinal roots. The consistency of the
response to serial stimuli (e.g. 10 electric shocks) is also
recorded as a measure of conduction but the validity of this
measure remains controversial.
Myasthenia gravis
H-reflex
H-reflex is a monosynaptic reflex evoked by low intensity,
submaximal electric stimulation of a motor nerve (e.g.
posterior tibial nerve), insufficient to produce a direct motor
response, which selectively activates the largest fibers in the
nerve, the afferent Ia fibers originating from the muscle 50 Repetitive nerve stimulation studies. Compound muscle action
spindles. This produces impulses that ascend the Ia sensory potentials evoked by repetitive electrical stimulation of the ulnar nerve
fibers to the spinal cord, where they synapse with the at the wrist at 4 Hz in a normal person (top) and a patient with
anterior horn cells, and are then transmitted down the myasthenia gravis (bottom). Myasthenia gravis is characterized by a
motor fibers to the muscle, producing a muscle contraction decrement in amplitude of the first four responses.
38 Neurologic Diagnosis

ELECTROMYOGRAPHY (EMG) initially positive diphasic potentials of longer duration and


slightly greater amplitude than the biphasic or triphasic
spikes of fibrillation potentials.
The recording through a needle or surface electrode of Fasciculation potentials are single motor unit action
electric activity from muscles. The signals are amplified and potentials produced by spontaneous discharges in
displayed visually on a screen and broadcast audibly through degenerating nerve fibers (52). Such contractions of a
a loudspeaker. motor unit or part of a motor unit may be large enough to
cause a brief visible twitching or dimpling under the skin.
NORMAL MUSCLE ACTIVITY Commonly they are several millivolts in amplitude, last from
Insertional activity 5 to >15 ms and usually fire irregularly. They are evidence
When the needle is moved gently within the muscle, a brief of motor nerve fiber irritability and not necessarily nerve
burst of electric activity occurs (51). fiber destruction or motor unit denervation. They may
occur in normal people in the calves and hands, and may be
At rest induced by low temperature and low serum calcium levels.
No electric activity from muscle fibers at rest (52). However, their occurrence in isolation in a relaxed muscle
is characteristic of motor nerve fiber degeneration, and this
Muscle activation can be confirmed by the presence of associated fibrillation
• With slight voluntary activation of muscle the smallest
motor neuron in the motor neuron pool will begin to
fire slowly, producing a stereotyped recurrent individual
motor unit action potential discharge on the screen and Normal 51
over the loudspeaker.
• With increasing voluntary effort, more and larger motor
units are activated. Initially these are identifiable as
individual potentials on the screen and over the loud- End-plate noise
speaker but as more and more units are recruited they
begin to overlap and produce a ‘recruitment pattern’.
• With maximum effort, a full recruitment pattern is seen. Positive waves

ABNORMAL MUSCLE ACTIVITY


Insertional activity: Myotonic discharge
• Increased in denervation and many forms of muscle
disease.
• Decreased in advanced denervation or myopathy where
muscle fibers have been largely replaced by fat and Bizarre repetitive potential
connective tissue.

At rest (spontaneous)
Fibrillation potentials are characterized by an initial positive 51 Insertional activity. Diagram illustrating the electric activity that may
(downward) deflection followed by a largely negative be evoked by insertion of a needle electrode into muscle.
deflection of 100–300 µV in amplitude and 1–2 ms in The top tracing shows the normal brief discharge of electric activity
duration (52, 53). They represent the spontaneous (insertion potentials) that last little longer than the movement of the
contraction of a single muscle fiber that has lost its nerve needle. Insertional activity may be prolonged in denervated muscle,
supply. If a motor neuron or nerve fiber is destroyed by myotonic disorders, and as a normal variant; and may be reduced in
disease or when its axon is interrupted, the distal part of the periodic paralysis during paralysis and if muscle is replaced by
axon degenerates over several days. Within 10–14 days from connective tissue or fat (e.g. chronic, end-stage myopathy).
the time of denervation, all of the muscle fibers connected ‘End-plate noise’ and associated muscle fiber action potentials is
to the branches of the dead axon (the motor unit) begin to electric activity evoked when the needle is in contact with motor end
generate random spontaneous biphasic or triphasic action plates and irritates small intramuscular nerves.‘Positive waves’ are
potentials (fibrillation potentials) and positive sharp waves evoked in denervated muscle.
(see below). ‘Myotonic discharges’ are the action potentials of muscle fibers firing
Fibrillation potentials may also be recorded in some in a prolonged fashion after external excitation.The potentials take two
primary muscle diseases with muscle fiber splitting, forms (positive waves and brief spikes) depending on the relationship of
inflammation or vacuolation (e.g. Duchenne muscular the recording electrode to the muscle fiber, and wax and wane in
dystrophy, polymyositis, inclusion-body myositis), because amplitude and frequency (40–100 per second) due to an abnormality in
the terminal innervation of some muscle fibers is damaged the muscle fiber membrane, caused by cholesterol-lowering agents
by the disease process. Fibrillation potentials continue until (CLAM), myotonic dystrophy, myotonia congenita, paramyotonia,
the muscle fiber is re-innervated by progressive hyperkalemic periodic paralysis, polymyositis and acid maltase deficiency.
proximal–distal regeneration of the interrupted nerve fiber, ‘Bizarre repetitive potentials’, now referred to as ‘complex repetitive
by the outgrowth of new axons from nearly healthy nerve discharges’ are action potentials of groups of muscle fibers discharging in
fibers (collateral sprouting), or until the atrophied muscle near synchrony at high rates (3–40 per second).They occur in a wide
fibers degenerate and are replaced over many years by range of chronic neuropathies (e.g. poliomyelitis, motor neuron disease,
connective tissue. Fibrillation potentials may take the form spinal muscular atrophy, radiculoneuropathies) and chronic myopathies
of positive sharp waves (51, 53), which are spontaneous, (chronic polymyositis, Duchenne dystrophy, limb-girdle dystrophy).
Electromyography (EMG) 39

potentials and certain changes in the motor unit potentials wave forms. Successive discharges show decrements in
(see below). amplitude. These discharges probably originate in the distal
Myokymia: persistent spontaneous rippling and quivering peripheral nerve, where activity of afferent nerve fibers
of muscles at rest, due to groups of repetitive firing excites distal motor terminals.
potentials, each group firing at its own rate. Often associated
with radiation nerve damage. Voluntary muscle activation
Myotonia: high frequency repetitive discharges which Upper motor neuron lesion: poor drive from the upper motor
generally have a positive sharp waveform and wax and wane neuron results in few motor units being activated and hence
in frequency and amplitude, producing a ‘dive-bomber’ poor recruitment of motor unit potentials. This pattern of
sound over the loudspeaker (51). Elicited during voluntary recruitment may also be seen in patients who do not (e.g.
muscle contraction and mechanically by movement of the hysteria), or cannot (e.g. due to joint pain), make an
needle electrode. After muscle contraction, a prolonged adequate voluntary effort.
afterdischarge occurs for up to several minutes, consisting Lower motor neuron lesion: after denervation of certain
of long trains of fibrillation-like potentials, corresponding muscle fibers and motor units within a muscle, the surviving
to the clinical feature of failure of voluntary relaxation of nerve fibers that innervate other motor units within the
muscle following forceful contraction. muscle begin to sprout new nerve twigs from nodal points
Complex repetitive discharges: spontaneous repetitive and terminals of undamaged axons, and re-innervate some
potentials with a bizarre configuration that start and stop or all of the denervated fibers. This process results in the
abruptly. A sign of chronicity (>6 months) in chronic addition of more muscle fibers to the surviving motor units,
neurogenic processes and some neuromyopathies. which appear on the EMG and large amplitude, long
Continuous muscle fiber activity (neuromyotonia): high duration, complex (polyphasic) potentials, which are
frequency (up to 300 Hz) repetitive discharges of varying characteristic of partial denervation and subsequent

52 Spontaneous electric activity in Normal 52


voluntarily relaxed muscle.
Normal: no electric activity. Fibrillations:
action potentials of single muscle fibers that
are twitching spontaneously and usually Fibrillation
regularly at rates of 0.5–15 per second in
the absence of innervation.They take two
forms (positive waves and brief triphasic or
biphasic spikes).
Fasciculation: action potential of a group
of muscle fibers innervated by an anterior Fasciculation: single discharge
horn cell that discharges in a random,
irregular fashion.They may arise from any
portion of the lower motor neuron, from
the cell body to the nerve terminal.

53 Needle electromyographic recording of a resting muscle showing 53


abnormal spontaneous fibrillation potentials (upward spikes) and
positive sharp waves (downward spikes).
40 Neurologic Diagnosis

re-innervation. With increasing voluntary effort, only one EVOKED POTENTIALS


or a few, large motor units are activated, and they fire
repetitively, resulting in a reduced recruitment pattern (54).
Neuromuscular junction disease: during sustained weak Stimulation of sensory organs or peripheral nerves evokes a
muscle contraction, a single motor unit may vary in small time-locked, event-related response (or potential) in
amplitude. EMG recordings of single muscle fibers the appropriate primary sensory cortex of the brain and a
belonging to the same motor unit may show varying number of subcortical relay stations as well. The larger
intervals between successive discharge of two single muscle random background activity in the EEG or EMG is filtered
fiber action potentials belonging to the same motor unit out by averaging methods. Three types of sensory evoked
(‘jitter’), due to variability in synaptic delay at the potential are commonly performed.
neuromuscular junction. This may increase to the point of
actual block in conduction. VISUAL EVOKED POTENTIALS (VEPs)
Muscle disease (myopathy): muscle fibers are destroyed With recording electrodes placed over the occipital region,
resulting in fewer functional muscle fibers in each motor the patient looks at a black and white checker board pattern,
unit. Activation of these smaller motor units is seen on the which alternates each second, while the overall luminance
EMG as low amplitude, short duration potentials which are of the entire retina remains constant. In a normal person
also commonly polyphasic because the compound motor with synchronous conduction in the optic pathways, this
unit potential is fragmented into its constituent single fiber repetitive visual stimulus evokes a characteristic electric
potentials and because of slowing of the propagated action potential over the occipital region of the head: the pattern-
potentials of affected muscle fibers (54, 55). With voluntary reversal visual evoked potential. By continuously stimulating
muscle activation, a higher proportion of these small motor the retina and recording the signals with electrodes over the
units need to be recruited to generate the force required, occipital region, it is possible to average the signals recorded
resulting in rapid recruitment. after a series of stimuli. Averaging ultimately cancels out
random background EEG activity and clearly defines a
characteristic positive wave at about 100 ms (P100) (56).
Demyelinating lesions along the optic pathway delay
Normal 54 conduction and lead to an increase in the latency (56).
Neurophysiologic evidence of delayed conduction in the
visual pathways is found in about 90% of patients with
clinically definite MS, whether or not they have a past
history of optic neuritis. About one-third of patients with
Myopathy MS who have no history or clinical evidence of optic nerve
involvement have abnormalities of the VEP.
Abnormalities of the P100 may be produced by other
Lower motor neuron disease diseases of the optic pathway such as glaucoma, retinal
diseases, compressive lesions of the optic nerve, toxic and
nutritional optic neuropathies, ischemic optic neuropathy,
and Leber’s hereditary optic neuropathy. Impaired visual
acuity has little effect on the latency but it correlates well
with the amplitude of the evoked response.

54 Motor unit action potentials during weak voluntary contraction of BRAINSTEM AUDITORY EVOKED
the biceps brachii in a normal person (top); a patient with muscular POTENTIALS (BAEPs)
dystrophy (middle): low amplitude, short-duration motor unit The patient wears a set of headphones though which a series
potentials; and a patient with motor neuron disease (bottom): high of between 1000 and 2000 pure tones (clicks) are delivered
amplitude, long-duration motor unit potentials.The time base for the first to one ear and then to the other. Each tone evokes an
action potentials recorded on the left is slower (100 cycles per second) auditory potential. The auditory evoked potential has a
than those on the right (1000 cycles per second). characteristic waveform with seven peaks: the first peak
(wave I) is produced in the cochlea and auditory nerve, the
second in the cochlear nuclei (pons), the third in the
55 superior olive, the fourth in the lateral lemniscus, and the
fifth in the inferior colliculus (midbrain). The generators of
waves VI and VII are uncertain but possibly the sixth is in
the medial geniculate and seventh in the auditory radiation
to the auditory cortex (57).
The BAEPs can be used to assess cochlea and auditory nerve
function and detect lesions of the VIIIth cranial nerve (e.g.
acoustic neuromas) and the auditory pathways of the
brainstem. A lesion that affects one of the relay stations or its

55 Needle electromyographic recording of a contracting muscle


showing a normal motor unit (left), and a small, polyphasic (myopathic)
motor unit (right).
Evoked Potentials 41

56 immediate connections causes a lower amplitude of the wave


or a delay in its latency and an absence or reduction in
amplitude of subsequent waves. These effects are more
pronounced on the side of the stimulated ear than
contralaterally, despite the fact that the majority of the cochlear-
superior olivary-lateral lemniscal-medial geniculate fibers cross
to the opposite side at the level of the superior olivary nuclei.
Almost one half of patients with clinically definite MS
and a lesser number with possible or probable MS have
abnormal BAEPs, even in the absence of clinical symptoms
or signs of brainstem dysfunction (58). However, BAEPs
examine only a relatively short segment of the central
pathways and are less sensitive in detecting demyelination
A than VEPs and MRI imaging of the brain. The latter has
greatly diminished the need for BAEPs.

56 Abnormal pattern-shift visual evoked potentials (VEPs).


A.The upper channel represents a VEP with a vertex reference
derivation and the bottom channel a VEP with an ear reference
derivation.The vertical line represents the upper limit of the latency of
the major positive peak (P100 or P2) for 99% of the normal
population.The positive peak is broad in contour, and its center is
slightly to the right, and therefore exceeds, the upper limit of normal.
B.The upper channel shows a markedly prolonged latency of the
VEP with a latency of the major positive peak being about 150 ms; the
lower channel (ear reference) is contaminated by noise artefact.This
patient has symptomatic visual loss secondary to demyelinating optic
nerve disease. Refractive errors, ocular opacities and visual inattention,
B and lesions posterior to the optic chiasm do not usually prolong the
latency of the VEP but tend to affect the amplitude of the VEP.

2 4 6 ms 57 58 Abnormal brainstem 2 4 6 58
V auditory evoked potentials
I II III IV (BAEP).The upper two
channels (top: A1–CZ, and
below: A2–CZ) recorded
VI after left ear stimulation in
A1–Cz
VII
a patient with MS show
that waves IV and V are
absent, indicating a lesion
in the upper pons,
probably contralateral to
the stimulated left ear.The
solid straight vertical lines
A1–Cz
indicate 2, 4 and 6 ms
respectively.The lower two
channels (top: A1–CZ, and
below: A2–CZ) show a
prolonged III-V interpeak 2 4 6
latency, indicating a lesion
57 Normal far-field brainstem auditory evoked potentials probably in the lateral
(BAEPs). Normally, in response to serial high frequency clicks at lemniscus causing slower
the ear, electrodes at the ear (left: A1, right: A2) and the vertex conduction between the
(CZ) record a total of 7 short-latency waves from within 10 ms of superior olivary complex
the click. By convention, vertex positivity is displayed as an upward (wave III) and inferior
deflection and the individual waves are labelled by their positive colliculus (wave V). As with
peaks.The upper two channels (top: A1–CZ, and below: A2–CZ) other evoked potentials, an
recorded after left ear stimulation in a normal person show abnormality indicates an
duplication of waves I through VII.The solid straight vertical lines anatomic lesion and not a
indicate 2, 4 and 6 ms respectively.The sweep duration is 10.2 ms. specific disease process.
42 Neurologic Diagnosis

SOMATOSENSORY EVOKED POTENTIALS (SSEPs) central pathways though the posterior columns, medial
Consecutive electric stimuli are applied, at frequencies of lemniscus, and thalamus to the parietal cortex are tested.
1.8 Hz, 2.1 Hz or a little faster (but not a multiple of 60, Delay between the stimulus site and Erb’s point or lumbar
to avoid 60 Hz artefact), to sensory or mixed peripheral spine indicates peripheral nerve disease; delay from Erb’s
nerves (e.g. the posterior tibial, median and ulnar nerves) point (or lumbar spine) to C2 implies an abnormality in the
while recording the evoked potentials from electrodes (for appropriate nerve roots or in the posterior columns; delay
the upper limb) over Erb’s point above the clavicle, the of subsequent waves recorded from the parietal cortex infer
dorsal root entry zone, the spinal cord at C2, and the scalp lesions in the medial lemniscus or thalamoparietal
overlying the contralateral parietal cortex, and (for the lower connections.
limb) over the lumbar and cervical spine and the contra- SSEPs are used in cases of suspected MS and cervical
lateral sensory cortex. Characteristic waveforms, designated spondylitic myelopathy and in monitoring the integrity of
by the symbol P (positive) or N (negative) and a number the spinal cord during spinal cord operations such as
indicating the interval of time from stimulus to recording scoliosis surgery. They are abnormal in up to 70% of patients
(e.g. N9), are produced in each of these locations, and with clinically definite MS (61), but are not as sensitive as
averaged by computer (59, 60). VEPs and MRI brain and spinal cord in detecting central
The integrity of the large sensory nerve fibers and their demyelination.

59 60 Fpz’ Cz’

10 20 ms +1.0 μV STIM
N19
C31–Fz

T12S
P22
C31–FII L3S

N12
CII–Fz

N10

Erb2 0 10 20 30 40 50 ms

STIM
59, 60 Normal short-latency somatosensory evoked potentials. 59 Stimulation of the median nerve at the wrist initially evokes a sharp,
predominantly negative potential over Erb’s point at about 10 ms (bottom channel) as the sensory and retrograde motor compound action
potentials pass beneath it in the brachial plexus at midclavicle, behind the sternomastoid.The next response is a surface negative bifid peak (N12)
over the second cervical vertebra (C2) with instrumental phase reversal (channels 2 and 3). Finally a potential is recorded over the contralateral
parietal lobe somatosensory cortex (C3’: 2 cm [0.8 in] behind C3), with a negative peak at about 19 ms (N19) and a positive wave at about 22 ms
(P22). 60 Stimulation of the posterior tibial nerve at the ankle and recording somatosensory evoked potentials at the popliteal fossa, spine and
scalp.

61 10 20 40 61 Abnormal short-latency somatosensory evoked potentials.


Stimulation of the median nerve at the wrist in a patient with multiple
sclerosis and loss of proprioception in the hands evokes potentials
initially over Erb’s point at about 10 ms (bottom channel), but then the
potentials over the second cervical vertebral (N12) and cortex (N19)
are not clearly seen.

N10
Further Reading 43

IMAGING THE BRAIN LUMBAR PUNCTURE AND CSF


FURTHER READING Rudkin TM, Arnold DL (1999) Proton magnetic EXAMINATION
spectroscopy for the diagnosis and manage- Serpell MG, Rawal N (2000) Headaches after di-
ment of cerebral disorders. Arch. Neurol., 56: agnostic dural punctures. BMJ, 321: 973–974.
NEUROLOGIC EXAMINATION 919–926. Steigbigel NH (2001) Computed tomography of
Acierno MD (2001) Ophthalmoscopy for the Costa DC, Pilowsky LS, Ell PJ (1999) Nuclear the head before a lumbar puncture in suspect-
neurologist. The Neurologist 7: 234–251. medicine in neurology and psychiatry. Lancet, ed meningitis – is it helpful? N. Engl. J. Med.,
Blessing WW (2000) Alternating two finger tap- 354: 1107–1111. 345: 1768–1770.
ping as part of the neurological motor exami- Thomas SR, Jamieson DRS, Muir KW (2000)
IMAGING THE CEREBRAL CIRCULATION Randomised controlled trial of atraumatic ver-
nation. Aust. NZ. J. Med., 30: 506–507.
Bendszus M, Koltzenburg M, Burger R, et al., sus standard needles for diagnostic lumbar
Mayo Clinic and Mayo Foundation (1981) Clin-
(1999) Silent embolism in diagnostic cerebral puncture. BMJ, 321: 986–990.
ical Examinations in Neurology 5th edn. W.B.
angiography and neurointerventional proce-
Saunders, Philadelphia.
dures: a prospective study. Lancet, 354: ELECTROENCEPHALOGRAPHY
Pandit RJ, Gales K, Griffiths PG (2001) Effec-
1594–1597. Blume WT (2001) Current trends in electro-
tiveness of testing visual fields by confronta-
Keir SL, Wardlaw JM (2000) Systematic review of encephalography. Curr. Opin. Neurol. 14:
tion. Lancet, 358: 1339–1340.
diffusion and perfusion imaging in acute is- 193–197.
Weaver DF (2000) A clinical examination tech-
chemic stroke. Stroke, 31: 2723–2731. Fish D (2001) Anticipation of epileptic seizures
nique for mild upper motor neuron paresis of
Markus HS (1999) Transcranial Doppler ultra- from standard electro-encephalographic
the arm. Neurology, 54: 531–532.
sound. J. Neurol. Neurosurg. Psychiatry, 67: recordings. Lancet 357: 160–161.
135–137.
44 Chapter Two

Disorders of
Consciousness
COMA

DEFINITION
0 • Occasional eye opening, flexion or extension of muscles
in the limbs, grunting or groaning in response to painful
stimuli, and primitive reflexes are all that may be present.

A state of unresponsiveness in which a person is unaware of PHYSIOLOGY OF CONSCIOUSNESS


themself and the environment and cannot be aroused into Consciousness depends on intact and interacting brainstem
a state of awareness or respond to the environment. reticular formation and cerebral hemipheres. The reticular
formation (from the Latin reticulum, meaning ‘a net’)
CLASSIFICATION OF LEVELS OF CONSCIOUSNESS consists of a net of small and large cells and their connec-
Various stages of clouded or impaired consciousness are tions throughout the brainstem from the medulla to the
recognized and are loosely defined below. However, it is thalamus. All major sensory pathways project to the reticular
more important to describe in detail the clinical state of the formation where they interact before proceeding to the
patient, and postures and responses to various stimuli than sensory cortex. The part of the reticular formation which is
to apply only one of these labels that may be interpreted most concerned with arousal and maintaining wakefulness
differently by different people. is the ascending reticular formation (reticular activating
system) in the midbrain and thalamus, which projects
Normal consciousness diffusely to the cerebral cortex.
• Alert wakefulness.
• Orientated in time and place. PATHOGENESIS
• Responsive immediately and appropriately to stimuli. Coma is caused by:

Confusion • Dysfunction of the ascending reticular formation in the


• Clouding of consciousness. thalamus or brainstem, and/or
• Impaired capacity to think with customary clarity, • Dysfunction of both cerebral hemispheres.
coherence and speed; understand; respond to and
remember stimuli. N.B. Small focal lesions in the brainstem and even larger
• Disorientated in time and place to some degree. lesions of one cerebral hemisphere do not cause coma unless
• Poor attention and concentration; distractible and they encroach on the reticular formation.
forgetful. Supratentorial lesions may cause coma by displacing the
brain laterally or caudally, as a result of their mass effect, and
Delirium compromising the function of the reticular formation in the
• A more profound confusional state. region of the thalamus and midbrain (62, 63). Lateral
• Anxiety, excitement, agitation, motor restlessness, expansion may also compress and disturb the function of
hallucinations, disorientation and sometimes delusions the contralateral cerebral hemisphere.
(e.g. delirium tremens). Infratentorial space-occupying lesions initially compress
adjacent structures, increasing the pressure in this tight,
Obtundation inexpandable compartment. Further expansion leads to
Drowsy and indifferent to the surroundings but responds upward herniation of the brainstem through the tentorial
to verbal stimuli. notch and downward herniation of the cerebellar tonsils and
medulla through the foramen magnum.
Stupor
• Somnolent: patient may appear asleep. Patterns of supratentorial brain shift
• Little or no spontaneous activity, with some kind of Cingulate herniation
voluntary response only to vigorous stimulation or pain, The expanding hemisphere shifts across the intracranial
and then lapsing back into somnolence. cavity, forcing the cingulate gyrus under the falx cerebri,
compressing and displacing the internal cerebral vein and
Coma the ipsilateral anterior cerebral artery (62, 63). Infarction
• Unrousable. in the territory of the anterior cerebral artery may occur.
• Absence of any understandable response to external
stimuli or inner need.
Coma 45

Central or transtentorial herniation of the diencephalon brain shift and herniation. The aqueduct and subarachnoid
Caudal displacement of the hemispheres, basal nuclei, and spaces are also compressed, compromising cerebrospinal
eventually diencephalon and adjoining midbrain through fluid (CSF) circulation, and leading to hydrocephalus and
the tentorial notch (62, 64). The great cerebral vein is further elevation of supratentorial CSF pressure. The medial
compressed, raising the hydrostatic pressure of the deep perforating branches of the basilar artery are stretched (the
territory it drains, causing venous congestion, edema and basilar artery cannot shift downward because it is tethered
even infarction. The resultant cerebral edema further to the circle of Willis), leading to paramedian brainstem
compromises venous flow, leading to more mass effect, ischemia (and hemorrhage if perfusion continues) (65).

62

62 Herniation of the brain. Left: Uncal and transtentorial herniation. A mass such as a cerebral hemorrhage, cerebral infarct or hemorrhagic infarct in one
cerebral hemisphere displaces the diencephalon and mesencephalon horizontally and caudally.The cingulate gyrus on the side of the lesion herniates
under the falx cerebri (top arrow).The uncus of the ipsilateral temporal lobe herniates under the tentorium cerebelli (lower arrows) and becomes
grooved and swollen and may compress the ipsilateral oculomotor (IIIrd cranial) nerve causing pupillary dilatation (Hutchinson's sign).The cerebral
peduncle opposite the supratentorial mass becomes compressed against the edge of the tentorium, leading to grooving (Kernohan's notch) and a
paresis homolateral to the cerebral mass lesion. Central downward displacement also occurs but is less marked than in the adjacent figure on the right.
Right: Central transtentorial herniation. Diffuse or multifocal swelling of the cerebral hemispheres (or bilateral subdural or epidural hematomas)
compress and elongate the diencephalon from above.The mamillary bodies are displaced
63 caudally.The cingulate gyrus is not herniated. (Adapted from Plum F, Posner JB [1985] The
Diagnosis of Stupor and Coma, 3rd edn. FA Davis, Philadelphia, Chapter 2, p92.)

63 Herniation of the brain. Coronal section of the brain of a patient with a massive
right temporal lobe glioblastoma multiforme who died after developing the syndrome of
uncal herniation.

64 Coronal section of the brain of a patient with multiple cerebral metastases causing
brain swelling, raised intracranial pressure, and compression, elongation, and caudal
displacement of the diencephalon and medial temporal lobes.

64 65 65 Axial section through the


mid pons showing Duret
hemorrhages in the central
core of the brainstem of a
patient who died after rapid
expansion of an acute lobar
intracerebral hemorrhage
causing transtentorial
herniation, downward
displacement of the midbrain
and pons, and stretching of the
medial perforating arteries of
the basilar artery (which is
tethered to the circle of Willis
and cannot shift downward).
46 Disorders of Consciousness

Uncal herniation: expanding lesions in the temporal fossa Vascular


shift the inner, basal edge of the uncus and hippocampal Subarachnoid hemorrhage.
gyrus toward the midline so that they bulge over the
incisural edge of the tentorium, and push the adjacent Non-neurologic (psychiatric)
midbrain against the opposite incisural edge (66). The IIIrd Malingering or hysteria.
cranial nerve and the posterior cerebral artery on the side of
the expanding temporal lobe are often caught between the CLINICAL FEATURES
overhanging swollen uncus and the free edge of the Before, or while, undertaking a clinical assessment, the
tentorium or the petroclinoid ligament, leading to a IIIrd patient’s vital functions should be attended to, paying
nerve palsy and occipital and medial temporal lobe particular attention to the airway (with adequate
infarction and swelling due to posterior cerebral artery
territory ischemia, which further compounds the problem.
66
ETIOLOGY
Supratentorial (15%) and infratentorial (15%) lesions
• Brain hemorrhage: intraparenchymal, subdural, subarach-
noid (67).
• Brain infarction.
• Venous-sinus thrombosis.
• Meningo-encephalitis.
• Brain abscess.
• Brain tumor.
• Head injury.

Diffuse brain disturbances (70%)


Metabolic
• Hypoxia (see Hypoxic encephalopathy, p.449): cardiores-
piratory arrest, status asthmaticus, chronic obstructive
airways disease, pneumonia, and prolonged hypotension.
• Hyponatremia: excess water ingestion, decreased water
excretion (e.g. nephrotic syndrome), inappropriate
antidiuretic hormone (ADH) secretion (e.g. central brain
lesions, carbamazepine), or carcinoma (a paraendocrine
phenomenon).
• Hypoglycemia: insulin or oral hypoglycemic agent excess 66 The under-surface of the forebrain at autopsy in a patient who
(pancreatic islet cell tumor, diabetes), prolonged fasting, died after transtentorial herniation showing swelling and grooving of
alcoholism, hypopituitarism and hypothyroidism. the uncus of the left medial temporal lobe (arrows) which has
• Hyperglycemia, hyperosmolality and acidosis: diabetes herniated through the tentorium cerebelli, and compressed the
(N.B. hyperglycemia may not be the cause but may be a midbrain. (Courtesy of Professor BA Kakulas, Royal Perth Hospital,
consequence of other causes of coma such as stroke, head Australia.)
injury and meningitis).
• Renal failure (uremia): predisposes to dehydration, hypoten-
sion, electrolyte disturbances, uremia, metabolic acidosis, 67
infection and impaired excretion of drugs and other toxins.
• Liver failure: predisposes to ammonia and drug toxicity,
and hypoglycemia.
• Hypocalcemia: renal failure, hypoparathyroidism.
• Hypercalcemia: primary hyperparathyroidism, malig-
nancy and prolonged immobilization.
• Hypothermia: hypothyroidism, immersion.
• Hypothyroid.
• Thiamine deficiency: chronic alcoholism.

Toxic
Drugs: overdose of sedative/hypnotic drugs (alcohol,
benzodiazepines, barbiturates, opiates, phenothiazines, and
tricyclic antidepressants alone or in combination).

Inflammatory
Meningo-encephalitis.
67 Section through the pons and cerebellar hemispheres, axial plane,
Epilepsy showing massive fatal pontine hemorrhage extending into the fourth
Generalized seizures (primary or secondary). ventricle. (Courtesy of Professor BA Kakulas, Royal Perth Hospital,
Australia.)
Coma 47

oxygenation), blood pressure and circulation (with fluid and Odor of breath: alcohol, ketones (diabetes), hepatic or renal
electrolyte balance). failure.

HISTORY Clubbing of fingernails: respiratory or gastrointestinal


Contact family and friends to obtain an eyewitness account disease.
of the preceding events, and when the patient was last seen,
how the patient was behaving, whether the patient is a known Heart rate: tachy- or bradyarrhythmia, or atrial fibrillation:
epileptic, alcoholic or drug abuser, and any known psychiatric embolism from the heart to the brain.
and medical conditions, medications or drug habits.
Blood pressure
PHYSICAL EXAMINATION • Hypotension: shock, myocardial infarction, septicemia,
General examination Addison’s disease.
This often yields important clues. • Hypertension: may be secondary or the primary cause
(hypertensive encephalopathy).
Skin and mucous membranes
• Color: blue (cyanosis); pink (carbon monoxide Neck or cardiac bruits: ischemic stroke.
poisoning); pale (anemia); yellow (jaundice).
• Purpura: bleeding diathesis (?intracranial hemorrhage). Tracheal deviation, chest dullness to percussion, bronchial or
• Rash: infection (e.g. meningococcemia) or inflammatory soft breath sounds: respiratory disease.
disease.
• Pigmentation: Addison’s disease. Abdominal organomegaly: liver disease (bleeding diathesis),
• Puncture wounds: diabetic, recreational drug abuse. polycystic kidneys (subarachnoid hemorrhage).

Temperature Neurologic
• Fever: meningitis, encephalitis, brain abscess, systemic Position, posture and spontaneous movements.
infection, recent seizure.
• Hypothermia: exposure to low environmental Level of consciousness: record serially:
temperatures, intoxication with alcohol or other • Glasgow coma scale (Table 2): the most widely used
sedatives, peripheral circulatory failure, or hierarchic grading scale.
hypothyroidism. • Supra-orbital pressure and nail bed pressure to test
response to pain: note nature and (a)symmetry of
Respiration response.
• Deep and rapid: metabolic acidosis, pneumonia,
brainstem lesion. Skull and spine: neck stiffness, Kernig’s sign.
• Shallow and slow: drug intoxication.
• Periodic: brainstem lesion. Brainstem function
• Pupillary reaction:
– Dilatation of one pupil with no response to light: IIIrd
nerve compression by uncal herniation (or other causes:
Table 2 The Glasgow coma scale posterior communicating artery aneurysm).
Score – Non-reactive (fixed) mid-position pupils: mid-brain
Eyes open lesion, anesthesia.
Nil 1 – Small reactive pupils: metabolic/toxic encephalopathy.
To pain 2 – Small (pin-point) pupils: pontine lesion, opiates,
To verbal stimuli 3 pilocarpine eye drops.
Spontaneously 4 – Non-reactive (fixed) dilated pupils: significant brain stem
damage or effect of atropine-like drugs.
Best verbal response – Horner’s syndrome: hypothalamic, brainstem or carotid
No response 1 lesion.
Incomprehensible sounds 2 • Resting position of the eyes and spontaneous eye
Inappropriate words 3 movements:
Disorientated and converses 4 – Minor degrees of divergence of the eyes: normal in
Orientated and converses 5 unconscious patients.
– Roving conjugate or dysconjugate eye movements,
Best motor response similar to those of sleep: common among patients in
No response 1 light coma; indicate an active brainstem.
Extension (decerebrate rigidity) 2 – Conjugate deviation of the eyes to one side: focal
Abnormal flexion of upper limbs (decorticate rigidity) 3 ipsilateral hemispheric or contralateral low pontine lesion,
Flexion – withdrawal to pain 4 or status epilepticus.
Localizes pain 5 – Conjugate depression of the eyes: midbrain tectum
Obeys commands 6 (posterior commissure) lesion or compression (e.g.
hydrocephalus).
Total 15 – Skew deviation: brainstem (pontomedullary junction)
lesion.
48 Disorders of Consciousness

• Resting position of the eyes and spontaneous eye Motor function


movements (continued): • Glasgow coma scale.
– Dysconjugate eyes: nuclear or infranuclear oculomotor • Lateralizing features are important (facial grimace; limbs).
or abducens nerve lesion, or internuclear ophthalmo- • Seizures: focal or generalized.
plegia (medial longitudinal fasciculus). • Multifocal myoclonus: diffuse cortical irritation due to
– Regular conjugate horizontal eye movements (‘ping- anoxia or metabolic disturbance.
pong gaze’): brainstem lesion. • Muscle tone, deep tendon reflexes, and plantar responses:
– Eyes jerk backwards into orbits (retractory nystagmus): abnormal with corticospinal tract lesions and also some-
midbrain lesion. times with hepatic and hypoglycemic encephalopathy.
– Intermittent downward jerking of the eyes (ocular
bobbing): low pons lesion. DIFFERENTIAL DIAGNOSIS
– Spontaneous nystagmus: rare in comatose patients because Coma or not?
the fast phase is a corrective saccade generated by the Confusion
frontal cortex and requires intact and interactive vestibular Confusion is often caused by acute metabolic or toxic
pathways, cerebral cortex and oculomotor system. disturbances, particularly in the elderly (e.g. hypoxia,
• Oculocephalic reflex: rotating the patient’s head from hypercapnia, fluid and electrolyte disturbance, hypogly-
side to side in a comatose patient with an intact cemia, drug intoxication, and drug withdrawal), or sleep
brainstem evokes conjugate eye movements in the deprivation, pain and as a transient phenomenon after
opposite direction to the head movement, so that the seizures. It needs to be differentiated from dysphasia,
eyes move in the orbit but remain ‘focused’, looking amnesia, acute psychosis, the retardation of severe
straight ahead (‘doll’s eye movements’). If the depression and dementia.
oculovestibular reflex is interrupted, due to brainstem
dysfunction, the eyes move dysconjugately, or do not Delirium
move in the orbits, remaining in the mid-position of the Delirium is commonly caused by metabolic and toxic
head, ‘looking’ in the direction of the head movement. disorders but can be mimicked by degenerative brain
• Oculovestibular responses: after examining the intact disease, acute psychosis and hypermania.
tympanic membranes, 50–200 ml of ice cold water is
instilled into one of the external auditory meati. If the Stupor
oculovestibular reflex is intact, the eyes deviate tonically Stupor needs to be differentiated from catatonic
toward the irrigated ear. In a conscious patient (or schizophrenia or severe retarded depression. The EEG is
psychogenic coma) with intact corticopontine fibers, the always abnormal in stupor due to organic disease.
frontal cortex tries to drive the eyes back to mid-position
thus generating the fast phase of nystagmus away from Persistent vegetative state (see p.52)
the side of stimulation. Tonic and conjugate deviation of
the eyes toward the irrigated ear, without corrective Locked-in syndrome (see p.55)
ocular saccades (the fast phase), indicates an intact
brainstem and a supratentorial cause for the coma. If the Brain death (see p.51)
response is dysconjugate, or absent, brainstem
dysfunction is the likely cause of the coma. Both external Etiology of the coma
auditory meati should be irrigated independently; Coma with focal neurologic signs or evidence of head injury
simultaneous irrigation will evoke vertical conjugate eye • Supratentorial or infratentorial mass lesion (vascular,
movement and corrective saccades (nystagmus). inflammatory, neoplastic).
• Corneal response: may be absent in light coma due to • Hypoglycemic encephalopathy.
drug intoxication but otherwise is usually retained until • Hepatic coma.
deep coma and then its absence is a poor prognostic factor.
• Respiration: Coma with meningism but no focal/lateralizing
– Post-hyperventilation apnea: bilateral hemispheric neurologic signs
disturbance. • Subarachnoid hemorrhage.
– Long-cycle Cheyne–Stokes respiration: diencephalic • Meningo-encephalitis.
dysfunction.
– Central neurogenic hyperventilation: low midbrain and Coma without focal/lateralizing neurologic signs or
upper pons lesions. meningismus
– Short-cycle Cheyne–Stokes respiration: medullary Metabolic/toxic encephalopathy:
dysfunction. • Hypoxic encephalopathy.
– Yawning, vomiting, hiccough: brainstem dysfunction. • Hyponatremia. Altered consciousness and seizures,
without focal neurologic signs.
Ophthalmoscopy: look for papilledema, sub-hyaloid hemorr- • Hypoglycemic encephalopathy:
hages, retinal emboli, diabetic or hypertensive retinopathy. – Altered consciousness.
The absence of papilledema or venous pulsation does not – Focal neurologic signs, such as hemiparesis, may be
exclude raised intracranial pressure. present.
– Excess sympathetic adrenergic activity (pallor, sweating,
dilated pupils, tremor, tachycardia).
– Underlying diabetes, alcoholism, hypopituitarism,
hypothyroidism or pancreatic islet cell tumor.
Coma 49

• Hyperglycemia, hyperosmolality and acidosis: • Slow, roving eye movements are not evident.
– Diabetic ketoacidosis: dehydration, hyperosmolality, aci- • Irrigation of the ears with ice-cold water is noxious and
dosis, and deep sighing respirations (Kussmaul breathing). usually evokes nausea, sometimes with vomiting, and
– Hyperosmolar coma, with dehydration but no nystagmus with the fast phase beating away from the side
ketoacidosis: older patients taking oral hypoglycemic of the irrigated ear (which is a cortical response to the
drugs or small doses of insulin tonic deviation of the eyes toward the irrigated ear, that
– Non-ketotic lactic acidosis in diabetic patients taking is mediated by the brainstem reflex).
hypoglycemic agents such as metformin. The blood
sugar may or may not be elevated. INVESTIGATIONS
– Hyperglycemia may not be the cause of the coma; it may Coma with focal neurologic signs or evidence of
be a consequence of other causes of coma such as stroke, head injury
head injury and meningitis. • CT or MRI brain scan: supratentorial or infratentorial
• Renal failure (uremia). mass lesion.
• Liver failure (hepatic encephalopathy, see p.452): • Blood glucose.
– Confusion or delirium. • Liver function tests.
– Asterixis: an inability to maintain the arms outstretched
with the hands and fingers held dorsiflexed because of N.B. Lumbar puncture (LP) is contraindicated in the
periodic inhibition of contraction of the wrist and finger presence of space-occupying lesions that increase the
extensors causing recurrent lapses in posture of the hands pressure in one brain compartment, particularly the
at the wrists, giving rise to the appearance of a ‘liver flap’. infratentorial compartment, because it may lower pressure
• Hypocalcemia: in the spinal canal and increase the pressure differential
– Tetany and seizures: underlying renal failure and across different brain compartments and precipitate
hypoparathyroidism. herniation of brain, particularly through the foramen
• Hypercalcemia: magnum (‘coning’) (see p.31).
– Dehydration, weakness, nausea, anorexia, and vomiting:
Underlying primary hyperparathyroidism or malignancy, Coma with meningism but no focal/lateralizing
or prolonged immobilization. neurologic signs
• Hypothermia and hypothyroidism: • CT brain scan: subarachnoid blood, or focal collections
– History of prolonged immersion in water or exposure to of blood or pus.
the cold. • LP, depending on results of CT scan, to examine CSF for
– Hypothyroidism: coma can be precipitated by cold, infec- blood, inflammatory cells, and micro-organisms.
tion or withdrawal of thyroid medication. Clinical features
include obesity, coarse myxedema facies, low body Coma without focal or lateralizing neurologic signs
temperature and delayed relaxation of the ankle jerks. or meningismus:
• Thiamine deficiency (Wernicke’s encephalopathy [see • Full blood count and ESR.
p.460]): • Urea and electrolytes.
Malnourished chronic alcoholics, recent increased • Plasma and urine osmolality.
glucose intake. • Blood glucose.
– Confusion, ophthalmoplegia (bilateral abducens nerve • Red blood cell transketolase.
palsies), nystagmus and ataxia, but some present in coma. • Thiamine.
Peripheral neuropathy may present. • Liver function tests.
• Calcium and phosphate.
Drug overdose: • Thyroid function tests.
• History and circumstances of discovery. • Arterial blood gases.
• A discrepancy between marked depression of brainstem • Drug and toxin screen in blood and urine.
responses and relatively light coma is present in some • EEG: triphasic waves of hepatic and uremic
patients. encephalopathy, non-convulsive status epilepticus or a
• Pin-point pupils suggest opiate poisoning. post ictal recording.
• CT or MRI brain may be necessary but the chance of
Subarachnoid hemorrhage (see p.249): neck stiffness is finding a focal abnormality is low.
usually, but not always, present. • LP may occasionally be indicated to exclude an
inflammatory or infectious cause.
Meningo-encephalitis (see p.273): fever, neck stiffness may
be absent in a deeply comatose patient.

Generalized epilepsy (nonconvulsive status or post ictal state):


• History of epilepsy.
• Evidence of tongue biting and incontinence.

Non-neurologic (malingering or hysteria):


• Normal rate and depth of respiration, pupillary reactions,
muscle tone, deep tendon and abdominal reflexes, and
plantar responses.
• Attempts to open the patient’s eyes may be resisted.
50 Disorders of Consciousness

TREATMENT Clinical signs


Resuscitation The constellation of brainstem reflexes are the most
• Establish an adequate airway: consider if endotracheal important prognostic clinical signs; there is no single
intubation, assisted respiration and oxygen are required. prognostic sign.
• Establish intravenous access:
– Withdraw blood to measure blood glucose, electrolytes, After 24 hours of coma
metabolites, and drug levels. • Absent pupillary or corneal reflexes, or oculovestibular
– Stabilize the circulation: blood volume expansion or responses, in the absence of sedative drugs: almost
vasodepressor agent may be required. incompatible with recovery of independence, irrespective
of the cause of coma.
Clinical, radiologic and laboratory assessment • Absent corneal reflexes and oculovestibular responses,
(see above) together with extension of the limbs in response to pain:
<3% chance of any useful recovery.
Specific treatment of the underlying cause • Intact pupillary and corneal reflexes, and movements of
For example: localization: 40% chance of achieving independence.
• Naloxone (opiate intoxication).
• Glucose (hypoglycemia). After 48 hours of coma
• Thiamine (Wernicke’s encephalopathy): 100 mg • Nystagmus induced by oculovestibular testing, and
intravenously daily for 7 days and then regular oral vocalization of any recognizable word: 50% chance of
vitamin supplements. good recovery.

Continuing care
• Intensive care unit: protect, monitor and maintain
respiration, circulation, skin, and bladder and bowel
function; correct medical or surgical abnormalities, and
control epileptic seizures.
• Corticosteroids and hyperventilation may be indicated
for cerebral edema but are not of any value in the routine
management of the comatose patient.

PROGNOSIS
Determined by the etiology, depth and duration of coma
and the constellation of clinical signs of brainstem function.

Etiology
A continuing vegetative state is most likely to occur after
head injury or hypoxic ischemic damage. The chances of 68
making a good recovery for patients in coma due to the
following are:

• Stroke, including subarachnoid hemorrhage: <5%.


• Hypoxic-ischemic encephalopathy: 10%.
• Metabolic or infective encephalopathy: nearly 30%.
• Head injury: worse prognosis if intracranial hematoma,
increasing age of patient, and severe systemic injury.

Depth of coma
Poor prognostic factors:
• No response to eye opening (compared with eye opening).
• No vocal response to pain (compared with grunting).
• Poor motor response to pain (compared with flexion of
the limbs).

Duration of coma
• 6 hours: about 12% make a good recovery.
• 24 hours: 10% make a good recovery.
• 1 week: 3% good recovery.
• >1–2 weeks: almost all die or enter a continuing
vegetative state.

68 Ventral surface of the brain of a patient who died from acute


meningo-encephalitis.
Brain Death 51

BRAIN DEATH – Hypothermia.


– Hypotension.
• Permanent vegetative state (see p.52).
DEFINITION • Locked-in syndrome (see p.55).
Death • Psychogenic unresponsiveness.
• Irreversible cessation of all function of the brain or
irreversible cessation of circulation of blood in the body. INVESTIGATIONS
• Irreversible loss of the capacity for consciousness, com- Eliminate remediable or reversible conditions
bined with the irreversible loss of the capacity to breathe. • CT or MRI brain scan: remediable brainstem
compression.
Brain death • Full blood count and ESR.
• Irreversible loss of function of the brain or brainstem. • Urea and electrolytes.
• The state in which brain tissue is damaged to such an • Blood glucose.
extent that vital brain functions are irreversibly impaired, • Liver function tests.
regardless of whether or not the heart is beating. • Thyroid function tests.
• Urine drug samples: convenient but provide retrospective
ETIOLOGY AND PATHOPHYSIOLOGY data on substances eliminated by renal mechanisms and
• Hypoxic-ischemic insults: cardiac arrest, carbon are poor indicators of the amount ingested.
monoxide poisoning, status asthmaticus. • Plasma drug samples: more informative but do not
• Head trauma. measure tissue concentrations at the site of concern, such
• Stroke: ischemic, hemorrhagic. as the brain. There can be wide variability in the rate and
• Brain infections: encephalitis, meningitis (68). extent of drug absorption, with up to fourfold dif-
• Metabolic/endocrine failure. ferences in cerebral effects for a given blood concentra-
• Drug toxicity. tion. The rate of transport of drugs between blood and
brain tissue is a function of several factors: the protein
CLINICAL FEATURES binding and physicochemical properties of the drug, and
The clinical assessment should be carried out by two physi- physiologic factors, such as cerebral blood flow, drug
cians on at least two separate occasions 12–24 hours apart. diffusion, and active transport processes over the
• Unconscious. specialized endothelium of the cerebral capillaries.
• Apnea. • EEG: may be useful in the differential diagnosis of coma
• Absent brainstem function: (e.g. triphasic waves of hepatic encephalopathy) and for
– Pupils: mid position or fully dilated, fixed in diameter determining prognosis.
and non-reactive to sharp changes in the intensity of
instant light. Brain death
– Corneal reflexes: absent. • EEG: electrocerebral silence, but not necessary for the
– Vestibulo-ocular reflexes (oculocephalic [‘doll’s eye’] and diagnosis of brain death if all the clinical criteria are
caloric-induced eye movement): absent. fulfilled (see below).
– Stimulation of any somatic area within the cranial nerve • Cerebral angiography: absent cerebral blood flow.
distribution fails to elicit a motor response: noxious • Radionuclide brain scan: absent cerebral blood flow.
stimulation of the face fails to provoke a facial reflex
response. DIAGNOSIS
– Cough, suck and root reflexes: absent. Lack of clinically observable brain function:
– Gag reflex: absent and there is no response to tugging • The patient must be deeply comatose.
an intratracheal tube or bronchial stimulation by a • All brainstem reflexes must be absent and remain absent
suction catheter passed down the trachea. during repeated testing at intervals up to 24 hours.
– Ventilatory reflexes: absent; there is no spontaneous • The patient has to be maintained on a ventilator because
respiration when the patient’s ventilator is disconnected spontaneous respiration has ceased or is inadequate.
for a time sufficient to ensure that arterial carbon dioxide • The cause of brain death has been fully established,
tension has risen above the threshold for stimulation of having absolutely excluded the effects of hypothermia,
respiration (arterial PaCO2 >8 kPa [60 mmHg] with depressant drugs, and potentially reversible metabolic,
pH <7.30). toxic and endocrine causes of coma.
• Patient not hypothermic or hypotensive. • There is no doubt that the patient’s condition (loss of
• Flaccid tone and absent spontaneous or induced consciousness and loss of brainstem reflex responses and
movement. respiratory center function, or cessation of intracranial
• Consistent examination findings throughout a pre- blood flow) results from irremediable brain damage of
determined period of observation. known etiology and is irreversible.
• Some activity may be detectable (e.g. endocrine function
DIFFERENTIAL DIAGNOSIS or low voltage EEG activity).
• Coma (see p.44):
– Metabolic: hepatic and renal failure; hypo/hyper-
glycemia, hypo/hypernatremia.
– Endocrine failure: hypothyroidism.
– Toxicity: depressant drugs (e.g. sedatives, analgesics,
muscle relaxants, illicit drugs).
52 Disorders of Consciousness

MANAGEMENT PERMANENT VEGETATIVE


Establish brain death with certainty using STATE (PVS)
recognized criteria
• Diagnosis (Wijdicks, 2001).
• Exclusion of potential confounding factors: metabolic, DEFINITION:
endocrine and toxic causes. Wait at least four half-lives of • A condition of ‘wakefulness without awareness’.
elimination of the relevant drug in the absence of factors • The absence of any adaptive response to the external
known to delay excretion. This is usually 3 or 4 days. environment and any evidence of a functioning mind
Evidence of delayed excretion may necessitate confirma- which is either receiving or projecting information, in a
tory cerebral vessel radiocontrast or radionuclide patient who has long periods of wakefulness.
angiography.
• Observation (6–48 hours depending on the age of the The vegetative state
patient). • A clinical condition of unawareness of self and environ-
• Examination: ment in which the patient breathes spontaneously, has a
– Proof of extensive brain damage. stable circulation, and shows cycles of eye closure and eye
– Exclusion of reversible causes of coma. opening which may simulate sleep and waking.
– Clinical examination of brainstem reflexes: pupillary, • May be a transient stage in the recovery from coma or it
corneal, oculocephalic, oculovestibular, gag, cough. may persist until death.
– Assessment of somatic responses.
– Demonstration of apnea despite arterial PaCO2 >8 kPa The continuing or persistent vegetative state
(60 mmHg) with pH <7.30. • The vegetative state has continued for more than 4 weeks
• Repeat examination (more than 2 hours after first and it has become increasingly unlikely that the condition
examination). is part of a recovery phase from coma.
• Certification by two practitioners.
The permanent vegetative state
Prepare and discuss with the family or family • A diagnosis based on a high degree of clinical certainty
spokesperson that the continuing vegetative state is irreversible; usually
• The underlying condition. a continuing vegetative state for more than 12 months
• What constitutes brain death. after head injury and more than 6 months following
• Why and how it has occurred. other causes of brain damage.
• Differences between brain death and permanent • Avoiding this state is one of the most important aspects
vegetative state. of attempting to assess prognosis early in coma.

Provide emotional and practical support EPIDEMIOLOGY


Continual contact between the family and: Prevalence: 10 000 to 25 000 adults and 400–1000 children
• A responsible decision maker. in the USA.
• The intensive care/ward staff providing hour-by-hour
care. PATHOPHYSIOLOGY
• Severe damage to part or all of the cerebral hemispheres,
When counselling with an intact brainstem.
• Sit close enough to be easily seen and heard in an area • Can occur with damage to the more rostral part of the
where you will not be disturbed. brainstem.
• Avoid physical barriers such as tables or desks.
• Achieve and sustain eye contact. PATHOLOGY
• Extend a comforting touch to the relative’s shoulder or Three main patterns:
hand, if appropriate. • Diffuse axonal injury, typically as a sequel to severe closed
head trauma, giving rise to degeneration of the white
Allow time for acceptance of the situation matter throughout the cerebral hemispheres.
• Do not rush or pressurize a decision. • Extensive laminar necrosis of the cerebral cortex,
• Do not become impatient with requests to cover the following global cerebral hypoxia or ischemia (69).
same ground many times. • Thalamic necrosis, occasionally.
• Do not withhold information.
• Do not peripheralize the family in the decision processes. ETIOLOGY
• Do not over-promote the issue of organ donation. • Head injury.
• Hypoxic-ischemic encephalopathy: cardiac arrest, carbon
Involve the family centrally in final decision making monoxide poisoning.
and participation in the ‘switch off’ process if • Stroke: ischemic or hemorrhagic.
requested • Hypoglycemia.
• Intracranial infection.
After brain death is determined, life support • Brain tumor.
is removed • End stage of degenerative brain disorders: Alzheimer’s
• Do not withdraw contact after the ‘switch off’. disease.
Permanent Vegetative State (PVS) 53

CLINICAL FEATURES DIAGNOSIS


• Inattentive and unaware of the surroundings but Two medical practitioners experienced in assessing
breathes spontaneously, without mechanical support, and disturbances of consciousness and awareness should
has a stable circulation. separately assess the patient (this includes discussion with
• At times the eyes are closed and the patient appears other medical and nursing staff, relatives and carers about
asleep, at other times the eyes are open and they seem to the reactions and responses of the patient, and to ensure that
be awake. the patient is not sentient) and document their findings and
• May be aroused by painful or prominent stimuli, opening conclusions in the medical record. If there is any uncertainty
the eyes if they are closed, increasing the respiratory rate, about the diagnosis of the permanent vegetative state, then
or occasionally grimacing or moving the limbs. a re-assessment should be undertaken at a later date.
• When the eyes are open the eyelids may blink in response
to any threat to the eye. DIAGNOSTIC CRITERIA
• A range of spontaneous movements may occur such as Preconditions
roving eye movements, chewing, teeth-grinding, • A cause for the syndrome has been established.
groaning, grunting and even swallowing is possible. • Persisting effects of sedative, anesthetic and neuro-
More distressingly, patients may smile, shed tears, moan muscular blocking drugs have been excluded by the
or scream, without any discernible reason. Sometimes, passage or time or by appropriate analysis of body fluids.
the head and eyes turn fleetingly to follow a moving • Reversible metabolic causes have been corrected or
object or sound. excluded as the cause.
• Body posture may be decorticate or decerebrate.
• Brainstem reflexes (pupillary, oculocephalic [doll’s eye], Clinical criteria
corneal and gag) are usually preserved. All three of:
• Primitive reflexes such as pouting and sucking reflexes, • No evidence of awareness of self or environment at any
grasp reflex, and withdrawal reflexes to pain may be time. No volitional response to visual, auditory, tactile or
present. noxious stimuli, and no evidence of language
• Painful stimuli may provoke an extensor or a flexor comprehension or expression.
response. • Cycles of eye closure and eye opening which may
• Plantar responses are commonly extensor. simulate sleep and waking shall be present.
• Hypothalamic and brainstem function is sufficiently
N.B. Any unambiguous sign of conscious perception or preserved to ensure the maintenance of respiration and
deliberate action (e.g. any evidence of communication, circulation.
including a consistent response to command, or any
purposeful movement) is incompatible with the diagnosis Other clinical features include:
of vegetative state. Careful prolonged observation is • Spontaneous blinking.
required before concluding that a patient’s wakefulness is • Inconsistent, non-purposeful reflex movements in
unaccompanied by awareness. response to external stimuli:
– Apparent smiling.
– Facial ‘grimacing’ to painful stimuli.
– Watering of the eyes.
– Startle myoclonus.
– Occasional movements of the head and eyes towards a
peripheral sound or movement.
– Purposeless movement of the limbs and trunk.
• Retained pupillary and corneal responses (usually).
• Absence of visual fixation and ability to track moving
69 objects with the eyes or show a ‘menace’ response.
• Roving eye movements may be present.
• Conjugate or dysconjugate tonic eye movement, without
corrective saccades (nystagmus) in response to ice water
caloric testing.
• Variable deep tendon reflexes (reduced, normal or brisk)
and plantar responses.
• Clonus and other signs of spasticity may be present.
• Incontinence of bladder and bowel.

69 Microscopic section of a part of a cerebral hemisphere showing


laminar necrosis and demyelination in the white matter due to carbon
monoxide poisoning. (Courtesy of Professor BA Kakulas, Royal Perth
Hospital,Western Australia.)
54 Disorders of Consciousness

DIFFERENTIAL DIAGNOSIS • Single photon emission computed tomography (SPECT)


• Locked-in syndrome (see p.55): a brainstem lesion dis- and positron emission tomography (PET): show a
rupts the voluntary control of movement but arousal and reduction in cerebral metabolism.
the content of awareness are not abolished. Patients are • Neurophysiology studies: EEG, somatosensory evoked
able to communicate by movement of the eyes or eyelids. potentials (SSEPs), and electrodermal techniques; very
• Coma (see p.44): most patients who are in ‘coma’ for variable results. Often these studies provide evidence of
weeks, months or years are in a continuing vegetative some cortical activity, reminding us that an accurate
state. clinical diagnosis of vegetative state does not imply
• Brain death (see p.51): implies the irreversible loss of all cortical silence. They are of little value in predicting
brainstem functions. It is, in a sense, the converse of outcome, other than early EEG evidence of burst suppres-
PVS, in which brainstem function survives while the sion and SSEP evidence of an absent N20 potential.
function of the cerebral hemispheres is lost or gravely
impaired. Brain death is followed, within hours or days, No findings are diagnostic of the permanent vegetative
despite intensive care, by cardiac arrest. state.
• Akinetic mutism: a state of profound apathy with
evidence of preserved awareness and attentive visual PROGNOSIS
pursuit, giving an unfulfilled ‘promise of speech’. The • Determined by the age of the patient, underlying cause
responsible lesions often involve the medial frontal lobes. and the duration of the vegetative state.
• Psychogenic unresponsiveness. • The outlook is better in children, and after traumatic (as
opposed to non-traumatic) brain injury. If the cause is
The differentiation of these conditions from the not head injury, there is very little hope of recovery of
vegetative state is based on clinical findings (Table 3). sentience after 3 months and none after 6 months.
• If the cause is head injury, a longer time should elapse
INVESTIGATIONS (e.g. 6 months) before being confident that the chances
• CT or MRI head scan: may show non-specific focal or of recovery are extremely low (and almost non-existent
diffuse abnormalities, brain atrophy, and hydrocephalus. after 12 months).

Table 3 Differentiation of vegetative state from other conditions

Condition Locked-in syndrome Vegetative state Coma Brain death

Self-awareness Present Absent Absent Absent

Cyclical eye Present Present Absent Absent


opening

Glasgow coma scale


Eye opening 4 4 1–2 1
Motor response 1 1–4 1–4 1–2
Verbal response 1 1 1–2 1

Motor function Eye movement preserved in No purposeful movement No purposeful None or only
the vertical plane and able movement reflex spinal
to blink volitionally movement

Pain sensation Yes No No No

Respiratory Normal Normal Depressed Absent


function or varied

EEG activity Normal or minimally Polymorphic delta or theta Polymorphic Electrocerebral


abnormal –sometimes slow alpha delta or theta silence or theta

Brain Minimal/moderate ≥50% reduction ≥50% reduction Absent or


metabolism reduction great reduction

Prognosis Depends on cause Depends on cause Recovery, No recovery


Recovery unlikely and duration vegetative state,
or death within
2–4 weeks
Locked-in Syndrome 55

MANAGEMENT LOCKED-IN SYNDROME


Establish the diagnosis of a permanent vegetative state by
(1) identifying the clinical state of the patient, (2) the cause
for the syndrome, and (3) the lapse of time. Because many DEFINITION
patients entering a vegetative state emerge from it within a A de-efferented state whereby patients are aware of
few weeks or months, supportive early management is themselves and their environment but are unable to respond
usually appropriate. The diagnosis of a permanent vegetative due to loss of motor and speech function.
state implies that recovery cannot be achieved and further
therapy is futile. It merely prolongs an insentient life for the PATHOGENESIS
patient and a hopeless vigil for relatives and carers. • A supranuclear (upper motor neuon) lesion of the
descending corticospinal tracts, usually in the ventral
Medical care portion of the brainstem (commonly the pons), below
• Appropriate nursing or home care. the level of the IIIrd cranial nerve nuclei, causes paralysis
• Maintain oxygenation, circulation and nutrition. of the muscles innervated by the lower cranial nerves and
• Correct complicating factors such as infection and peripheral nerves.
hypoglycemia. • A widespread nuclear or infranuclear (lower motor
• Sensitive discussion with, and education of, relatives and neuon) disease of motor nerves.
carers about the cause, clinical state, ‘hopeless’ prognosis,
and implications, including the possibility of withdrawing ETIOLOGY
artificial means of administering food and fluid. Ventral brainstem lesion
• Decisions to withdraw nutrition, hydration and other • Infarction or hemorrhage (commonly hypertensive
life-sustaining medication such as insulin for diabetes, patients) (70, 71).
should currently be referred to the court before any • Tumor.
action is taken. • Demyelination (multiple sclerosis).
• Decisions not to intervene with cardio-pulmonary • Central pontine myelinolysis, following profound
resuscitation or prescribe antibiotics are clinical decisions, hyponatremia (see p.457).
but they should take account of, and respect, the views • Head injury.
of the relatives, carers and patient, if known, whether
formally recorded in a written document (or advance Polyneuropathy
directive) or not. • Critical illness polyneuropathy.
• The role of sensory stimulation remains uncertain, • Acute onset post-infectious polyradiculoneuropathy
despite the writings of Hippocrates: ‘the patient in a state (Guillain–Barré syndrome).
of coma should be spoken to in a loud voice, splashed
with cold water and exposed to bright light’.

70 71

71 Autopsy specimen of a cross section of the midpons and


cerebellum in the axial plane of a patient who was ‘locked-in’
showing hemorrhagic infarction in the ventral pons bilaterally.

70 Proton density weighted axial MR scan showing high signal in


the basilar artery (due to thrombus, arrow) and increased signal in
the pons due to pontine infarction (arrowhead) in a patient who
presented ‘locked-in’.
56 Disorders of Consciousness

CLINICAL FEATURES NARCOLEPSY


• Unable to speak.
• Unable to move the limbs.
• Awareness and consciousness are preserved because the DEFINITION
brainstem tegmentum, including the reticular formation A syndrome of excessive sleepiness and abnormally regulated
and oculomotor nerves and pathways are spared. rapid-eye-movement (REM) sleep.
• Able to open the eyes and move them, (particularly in
the vertical plane) and blink, in order to try and EPIDEMIOLOGY
communicate. • Prevalence: 1 in 500 000 (Israel), 1 in 5000 (USA,
Europe), 1 in 600 (Japan).
INVESTIGATIONS: • Age: 5–50 years, usually 15–35 years.
• CT or MRI brain scan: ventral pontine or midbrain lesion. • Gender: M=F.
• Other investigations, as appropriate, to ascertain the
cause (e.g. serum sodium, EEG, EMG). PATHOLOGY
Unknown.
DIAGNOSIS
Diagnosis is clinical, based on the presence of total paralysis ETIOLOGY AND PATHOPHYSIOLOGY
of the limbs and muscles innervated by the lower cranial • Inherited susceptibility: probably dominant with variable
nerves, but with the ability of the patient to open and close penetrance.
the eyes voluntarily and in response to commands, and to • Linked strongly to certain Class II HLA antigens: HLA-
respond to verbal and sensory stimuli by blinking. DR2 and HLA-DQw1 antigens in 98–100% of patients.
Classic narcolepsy (narcolepsy associated with cataplexy)
TREATMENT has almost a 100% association with the DRw15 subtype
General (see Coma, p.44) of the HLA-DR2 antigen, the DQw6 subtype of the
Patients can see, hear and feel everything (including pain HLA-DQw1 antigen, and the HLA-Dw2 subtype of the
and itch) so are sensitive to what staff are saying. They are HLA-D antigen.
also very frustrated that they cannot move. • The ‘narcolepsy-susceptibility’ gene has not been located
• Prevention of complications of immobility: pneumonia, exactly but is likely to be just outside the HLA-DQ and
deep vein thrombosis, contractures, urinary tract infection. HLA-DR subregions on the short arm of chromosome 6.
• Rehabilitation: physiotherapy, swallowing and speech • The occurrence of normal periods of REM sleep at the
therapy, occupational therapy, psychologic support and onset of sleep or within 10 minutes thereafter is the most
therapy. characteristic and striking physiologic abnormality, and
probably indicates impaired sleep–wake regulation rather
Specific than an excessive need for REM sleep.
Correct the underlying cause (e.g. hyponatremia; see • The mechanism may be abnormalities of monoaminergic
Central pontine myelinolysis, p.457). and cholinergic functioning in the brain (e.g. impaired
release of norepinephric neurotransmitters).
PROGNOSIS • Abnormalities in the orexin (hypocretin) neurotrans-
Prognosis is poor. Some patients recover, usually with mitter system are directly involved in the pathogenesis of
residual limb spasticity. narcolepsy in two animal models. Decreased levels of
orexin A in the CSF are found in narcoleptic patients,
and an association of a rare polymorphism in the prepro-
orexin gene with narcolepsy in a cohort of 178 patients
has been reported recently.
• Hypnagogic hallucinations, sleep paralysis and cataplexy
appear to be associated manifestations of REM sleep that
intrude into wakefulness.

CLINICAL FEATURES
Excessive sleepiness (rather than increased sleep)
• Brief (a few to 30 minutes), irresistible episodes of
daytime sleep, sometimes associated with dreaming, and
preceded by increasing drowsiness.
• Most apparent in boring, sedentary situations.
• Partially alleviated by mental stimulation and motor
activity.
• Cannot be fully relieved by any amount of sleep.
• Patients usually feel refreshed after a sleep attack.
• Chronic excessive daytime sleepiness between attacks of
sleep.
• The degree of sleepiness usually remains stable after the
first few months.
• Increased sleepiness after several years suggests the
presence of an additional disorder, such as sleep apnea.
Narcolepsy 57

Cataplexy DIFFERENTIAL DIAGNOSIS


• Attacks, lasting from a few seconds to several minutes, of Epilepsy
sudden reduction or loss of muscle tone, usually precipi- Although a single episode may be clinically indistinguishable
tated by excitement, emotion (such as surprise, anger, from a complex partial or absence seizure, the ‘sleep-like’
laughter) and athletic activities. Severe attacks cause appearance of the patient and the association of the episodes
complete paralysis except for the respiratory muscles. with drowsiness and passive situations helps to distinguish
More common partial episodes cause patients to drop automatic behavior from the repetitive stereotyped behavior
objects, sit down, stop walking, and even lose sphincter that accompanies epileptic seizures.
control (e.g. fecal incontinence).
• Consciousness is almost always maintained. Schizophrenia
• Most attacks last less than 1 minute. Patients with narcolepsy and prominent hallucinations may
• Prolonged episodes may be associated with hallucinations. be misdiagnosed as schizophrenic.
• Rarely, cataplexy that is almost continuous (‘status
cataplecticus’) may occur. Daytime sleepiness
• Sleep paralysis (60% of patients). • Sleep apnea: obstructive sleep apnea is characterized by
• Brief episodes of inability to move during the onset of recurrent episodes of partial or complete upper airway
sleep or on awakening. obstruction during sleep (causing habitual snoring and
• Patient may feel as if he or she is struggling to move. witnessed apneas) which are terminated by arousal. The
• Lasts up to 10 minutes. recurrent arousals have a destructive effect on sleep and
so the common symptoms are excessive daytime sleepi-
Hypnagogic hallucinations (60% of patients) ness, and impairment of mood, memory and concen-
• Hallucinatory experiences, usually visual but can be tration. Consequently, the patients’ work, family and
auditory or tactile, that accompany the onset of sleep or social lives are disrupted and the risk of accidents is
awakening. increased. Obstructive sleep apnea affects about 4% of
• May have a dream-like quality but awareness of sur- middle-aged men and half as many women. Predisposing
roundings is preserved. factors include male gender, increasing age, obesity,
• May accompany sleep paralysis or occur independently. alcohol and sedative abuse, family history, specific upper
airway abnormalities (nasal obstruction, tonsil-
Other symptoms lar/adenoidal hypertrophy, micrognathia, retrognathia,
• Automatic behavior (up to 80% of cases): episodes of Marfan’s syndrome), endocrine diseases (hypothyroid-
semi-purposeful activity, such as writing off the edge of ism, acromegaly) and neuromuscular diseases (decreased
the page, putting clothes in the refrigerator, usually ventilatory drive, decreased upper airway muscle tone).
during monotonous or repetitive behavior, and for which Treatment strategies include modification of predisposing
the patient is amnesic. factors; reduction of weight, smoking, alcohol and
• Brief lapses in speech and concentration. sedatives; modification of sleeping posture; nasal con-
• Disrupted nocturnal sleep. tinuous positive airway pressure (CPAP); dental splints;
• Memory disturbances, due to impaired attention and and surgery (nasal [if obstructed], palatal, mandibu-
fluctuating levels of alertness. lar/maxillary).
• Blurred or double vision (10–14%). • Insufficient sleep.
• Depression (?due to altered central monoamine function). • Medication effects.
• Diencephalic lesions: tumors in the region of the third
ventricle (72).

72 Excessive sleepiness without REM-sleep


abnormalities or cataplexy
• Narcolepsy which progresses with time to include
abnormalities of REM sleep and cataplexy.
• Atypical depression.
• Irregular sleep schedules.
• Idiopathic hypersomnia (a diagnosis of exclusion, having
excluded depression, insufficient sleep, and abnormalities
of REM sleep).

REM sleep at onset of sleep


• Endogenous depression.
• Sleep deprivation: sleep apnea, disturbances of the sleep-
wake schedule.
• Drug and alcohol withdrawal.
• Some structural brain lesions.
72 Macroscopic appearance of the brain, coronal section through the
thalami and upper midbrain showing a pineal germinoma occupying the
third ventricle (arrows) and compressing the rostral midbrain in a
patient who presented with recurrent episodes of daytime sleepiness
and was initially considered to have narcolepsy.
58 Disorders of Consciousness

INVESTIGATIONS CLINICAL COURSE AND PROGNOSIS


• Multiple sleep latency test (73): monitors the time to the • Sleepiness usually precedes cataplexy by several months
onset of sleep and the type of sleep that occurs during but 6–10% of patients experience cataplexy initially, and
attempts to sleep at 2 hourly intervals throughout the 10–15% do not develop cataplexy until 10 years or more
day. More than 80% of patients with narcolepsy have a after the onset of sleepiness.
mean sleep latency of less than 5 minutes and at least two • The degree of sleepiness usually remains stable after the
REM periods at the onset of sleep during the procedure. first few months and rarely lessens. Increased sleepiness
• MRI brain scan is sometimes indicated to exclude the after several years suggests the presence of an additional
most unlikely possibility of a diencephalic structural disorder, such as sleep apnea.
lesion. • Cataplexy, hypnagogic hallucinations, and sleep paralysis
improve or disappear with age in about one-third of
DIAGNOSIS patients.
• Excessive sleepiness and frequent REM periods at the
onset of sleep.
• Confirmation with a formal multiple sleep latency test is
essential in view of the long term implications of the
diagnosis.

TREATMENT
Narcolepsy
• Avoid occupations and situations where nodding off to
sleep is a danger to the patient or to others, such as
driving.
• Take regular 15–20 minute naps.

Stimulants:
• Enhance the synaptic availability of noradrenaline.
• Methylphenidate 10–60 mg/day, or
• Dextroamphetamine 5–50 mg/day. FP1-F3 73
• Higher doses offer little additional benefit and increase
F3-C3
the risk of adverse effects.
• Pemoline or protriptyline may be tried if methyl- C3-P3
phenidate and dextroamphetamine are not successful or P3-O1
not tolerated.
• Modafinil (an alpha1-adrenergic agent) 100 mg bd, FP2-F4
mazindol (an imidazole derivative), and selegiline (a F4-C4
monoamine oxidase-type B inhibitor) improve daytime
C4-P4
alertness and may have fewer adverse effects than
amphetamine. P4-O2
• Narcoleptics are not immune to the problems associated
with the chronic use of stimulants.
LOC
Cataplexy and sleep paralysis ROC
• Tricyclic antidepressants (e.g. imipramine, clomipramine, LUE
protriptyline) in low doses (10–50 mg): act by inhibiting
reuptake of noradrenaline and serotonin rather than LAE
cholinergic blockade.
• Specific serotonin reuptake inhibitors (e.g. fluoxetine):
EMG
may be as effective with fewer anticholinergic adverse
effects.
• Noradrenaline reuptake inhibitors (e.g. viloxazine).
• Gamma-hydroxybutyrate (sodium oxybate): increases 73 Electroencephalography (EEG) of a middle-aged male with
slow-wave sleep and REM sleep; effective in some. episodes of loss of consciousness (sleep attacks) and automatic
• Abrupt discontinuation of tricyclic agents can lead to a behavior (mini-sleeps). Multiple sleep latency test gave evidence of
rebound increase in cataplexy. narcolepsy with short sleep latency (2 minutes) and sleep-onset REM
• Gradual withdrawal of tricyclic antidepressants followed periods (REM latency 1 minute).Typical features during REM sleep
by a drug holiday sometimes helps restore efficacy. include rapid eye movements, absent muscle artefact, and drowsy
EEG pattern.
Disturbed nocturnal sleep LOC: left outer canthus; ROC: right outer canthus; LUE: left under
• Short acting hypnotic (e.g. temazepam) one or twice a eye; LAE: left above eye. Ocular electrodes were referential to A1A2.
week; regular nightly use is rarely beneficial. (Reproduced with permission from the editor (Elaine Wyllie, MD)
and publisher (Williams & Wilkins) of Wylie E (ed) (1997) The
Treatment of Epilepsy: Principles and Practice, 2nd edn.Williams &
Wilkins, Baltimore.)
Syncope 59

SYNCOPE Reduced stroke volume


Acute reduction
Low blood volume:
DEFINITION • Acute fluid loss: burns, vomiting, diarrhea, dehydration.
A transient loss of postural tone and consciousness resulting • Hemorrhage.
from an acute reduction in blood flow to the brain. • Addison’s disease.
• Diuretics.
EPIDEMIOLOGY • Very low salt diet.
• Incidence: The most common non-epileptic cause of loss
of consciousness. Obstruction to venous return to the heart:
• Age and gender: any age and either sex. • Cough/micturition/defecation.
• Cardiac tamponade.
PATHOPHYSIOLOGY
Basic neurophysiology Impaired outflow from the heart: acute myocardial
The conscious state depends on the integrity of the infarction.
brainstem reticular activating system interacting (through
its ascending pathways) with both cerebral hemispheres. Obstruction to outflow from the heart:
Therefore, to disturb consciousness, the function of the • Pulmonary embolus.
brainstem or both cerebral hemispheres needs to be • Aortic dissection.
disturbed (see Coma, p.44).
Chronic reduction
Basic vascular physiology: Impaired outflow from the heart:
• Blood flow to the brain depends on the mean arterial • Cardiomyopathy: dilated.
blood pressure and the intracranial pressure. • Congenital heart disease.
• Blood pressure is the product of the cardiac output
(heart rate × stroke volume) and total peripheral Obstruction to outflow from the heart:
resistance. • Atrial myxoma or ball valve thrombus (impaired
• Blood flow in the brain is maintained by autoregulation ventricular filling).
in response to minor changes in systemic blood pressure. • Hypertrophic obstructive cardiomyopathy.
However, sudden, dramatic reductions in blood pressure • Aortic stenosis (syncope on exertion).
below a mean arterial BP of about 8 kPa (60 mmHg)
result in a parallel fall in brain perfusion, resulting in a Reduced peripheral resistance
gradual loss of consciousness. Central autonomic neuropathy
• Multiple systems atrophy (see p.435).
Altered heart rate • Parkinson’s disease.
Bradyarrhythmia • Primary autonomic failure.
Primary: • Spinal cord injury.
• Complete heart block (Stokes–Adams attack).
• Atrial fibrillation. Peripheral neuropathy
• Sinus arrest. • Diabetic neuropathy.
• Guillain–Barré syndrome.
Secondary: • Tabes dorsalis.
• Carotid sinus hypersensitivity (head turning, neck • Amyloid neuropathy.
massage). • Acute porphyria.
• Swallow syncope: swallowing can activate a vagal reflex. • Autonomic ganglionitis and neuritis.
It may indicate an underlying structural abnormality of
the esophagus, glossopharyngeal neuralgia, digoxin Other
toxicity, or ischemic heart disease. • Vaso-vagal (the most common cause of syncope).
• Stretch syncope. • Drugs: vasodilators (e.g. glyceryl trinitrate, prazosin,
• Ice-cream syncope: cold food held in the mouth can hydralazine, calcium channel blockers) and other anti-
activate a vagal reflex. hypertensives (e.g. diuretics, ganglion blockers), levodopa,
• Prostatic syncope: rectal examination may activate a vagal bromocriptine, antidepressants, phenothiazines, sedatives.
reflex. • Large meal (dilatation of splanchnic vessels).
• Vaso-vagal attack: often precipitated by emotion. • Heat (dilatation of cutaneous vessel).
• Baroreceptor dysfunction (age, hypertension, prolonged • Anemia.
bed rest, autonomic neuropathy). • Hyponatremia.
• Old age and prolonged recumbency.
Tachyarrhythmia
Supraventricular tachycardia:
• Wolff–Parkinson–White syndrome.
• Prolonged QT syndrome.
• Complex partial seizures (very rare).

Ventricular tachycardia.
60 Disorders of Consciousness

ETIOLOGY (see Table 4) • Generalized weakness.


• Symptoms of adrenergic activity such as nausea, hot and
CLINICAL FEATURES cold feelings and sweating.
History • Palpitations and chest pain may indicated a cardiac cause,
The history should be directed firstly toward confirming but palpitations are also common in panic attacks and
that the patient has syncope, and secondly towards finding overbreathing.
the cause. Interview the patient and a witness if available.
Associated features during the attack:
Precipitating/contributory factors • Pale (rather than cyanosed).
• Venesection, acute pain or emotional shock, prolonged • Sweaty/clammy.
standing, overcrowding, heat (vaso-vagal syncope). • Floppy (rather than rigid).
• Valsalva maneuver: deliberate or during weight lifting, • Pulse: absent or difficult to feel (but this cannot be relied
playing a wind or brass musical instrument, or straining upon).
at stool. • Incontinence of urine may occur.
• Coughing. • Motor symptoms, such as a brief tonic contraction of the
• Micturition. trunk, tonic-clonic movements, and multifocal,
• Changes in posture (e.g. standing after sitting or lying in arrhythmic, myoclonic jerks may occur, particularly if an
bed for a prolonged period) and head position. upright or semi-upright posture is maintained.
• Drugs (nitrates, α-adrenoreceptor blockers [veno-
dilators]; diuretics [low blood volume]). Duration
• Hot environment (vasodilatation of cutaneous vessels). Syncope lasts seconds to 1 or 2 minutes, provided the
• Large meal (vasodilatation of splanchnic vessels). patient assumes the recumbent position, and isn't held
upright (by someone or an obstacle).
Pre-syncopal symptoms
• Light headedness. Sequelae
• Faintness. As consciousness is regained quickly there is very little
• ‘Dizziness’ (not vertigo). mental confusion or difficulty recalling the warning
• Dimming or loss of vision in both eyes (not to be symptoms (unless there has been head trauma), in contrast
confused with lone bilateral blindness), sounds seem to to the prolonged confusion, drowsiness and myalgia that
be distant/muffled. may be seen after a generalized seizure. Convulsive syncope

Table 4 Etiology of syncope

Type or cause of syncope Characteristics Severity Population prevalence %


(range)
Reflex-mediated
• Vaso-vagal Warmth, nausea Benign 18 (8–37)
• Situational After daily activity Benign 5 (1–8)
• Other After neck pressure Benign 1 (0–4)
or head turning

Orthostatic hypotension After standing Benign 8 (4–10)

Medications After drug use Benign to severe• 3 (1–7)

Psychiatric Frequent events Benign 2 (1–7)

Neurologic Headache, diplopia Moderate 10 (3–32)


hemiparesis, seizures
Cardiac
• Organic heart disease Chest pain, dyspnea Severe 4 (1–8)
exertional, post operative
• Arrhythmias Sudden syncope, injury 14 (4–38)
– Bradyarrhythmias Moderate
– Tachyarrhythmias Palpitations Severe

Unknown Negative workup Benign to 34 (13–41)


moderate

* Severity ranges from benign (e.g. prazosin-induced syncope) to severe (e.g. quinidine-induced torsades de pointes).
Syncope 61

may occur following prolonged vagal stimulation (causing Cardiovascular


pronounced bradycardia or asystole) with or without a • Pulse: rate, rhythm, character.
prolonged upright posture. The patient becomes pale and • Blood pressure: supine and standing. A fall in diastolic
falls limply, followed by a stiffening of the body or BP of 1.3 kPa (10 mmHg) and systolic BP of 2.7 kPa
opisthtonous, clonic movements, upward deviation of the (20 mmHg) or more in the standing position is defined
eyes, and urinary incontinence. It is common, particularly as postural hypotension.
in children and young adults, affects girls more commonly • Jugular venous pressure: low if hypovolemic, raised if
than boys, and is benign. pulmonary embolism or cardiac failure.
• Heart: auscultation for features of aortic stenosis and
Systemic enquiry other causes of outflow obstruction (e.g. hypertrophic
• Autonomic symptoms: bladder, bowel, sexual, and obstructive cardiomyopathy).
thermoregulatory dysfunction. • Serial pulse and standing BP over 30 minutes. Normally,
• Extrapyramidal symptoms: slowness, stiffness, tremor. the pulse increases to a maximum at the 15th beat after
• Polyneuropathy: altered sensation, weakness. standing but with autonomic failure, loss of
• Medications/drugs: hypotensive. consciousness may occur without a recordable increase
in the peripheral pulse rate or the presence of skin
Physical examination vasoconstriction.
Commonly normal. Skin and mucous membranes are dry
with reduced tissue turgor if dehydrated, pigmented buccal Neurologic
mucosa if Addison’s disease. • Extrapyramidal dysfunction: rigidity, bradykinesia, tremor.
• Brainstem dysfunction.
• Peripheral neuropathy: muscle wasting, weakness,
areflexia, sensory loss.

DIFFERENTIAL DIAGNOSIS
Epilepsy (see Table 5 and p.65)

Vertebrobasilar artery territory ischemia


Table 5 Differential diagnosis: syncope vs Seldom causes isolated loss of consciousness; other
epileptic seizure symptoms of focal brainstem ischemia coexist: vertigo,
Syncope (faints) Seizures (fits)
diplopia, ataxia. Carotid territory ischemia does not cause
loss of consciousness unless bilateral and severe.
Onset Often gradual Sudden
Circumstances Special Non-specific Causes
Relation to upright Common No • Atherothromboembolism.
posture • Embolism from the heart.
Symptoms Light headed/faint Amnesic • Migraine (often children, with a history of severe
Blurred/dimmed vision occipital headache).
Sounds seem distant • Arnold–Chiari malformation (vertebral artery and
Tinnitus brainstem compression).
Weakness • Cervical spondylosis: osteophytes may compress the
Nausea vertebral artery as they traverse the transverse foramen
Hot and cold of the cervical vertebrae and compromise blood flow in
Sweating the posterior circulation but this is a far less common
Skin color Pale Blue or normal cause of ‘dizziness’ and syncope than is commonly
Respiration Shallow Stertorous believed. More frequently, the diagnosis is a peripheral
Aura or premonitory Usually Sometimes labyrinthine disorder such as benign paroxysmal
symptoms positional vertigo (see p.110).
Tone Floppy* Rigid
Convulsion Rare Common
Urinary incontinence Rare** Common
Tongue biting Rare Common
Post ictal confusion Minimal Common and
prominent
Focal neurologic No Occasional
symptoms
Clues to underlying Cardiac arrhythmia
cause Aortic stenosis
Cardiomyopathy
Postural hypotension
* A tonic phase is common in convulsive syncope
** Traditionally not a feature but actually not uncommon.
62 Disorders of Consciousness

Drop attacks determined by the certainty of the clinical diagnosis and nature
• Spontaneous episodic raised intracranial pressure may of the clinical findings.
impair brain perfusion (e.g. colloid cyst of the third
ventricle [74, 75], posterior fossa tumor). Usually Cardiologic
associated headache and visual obscurations Indicated in patients with known or suspected heart disease
• Cryptogenic drop attacks: abrupt episodes of falling to the (e.g. history of congestive heart failure or ventricular
ground without any warning, precipitating factor, or loss arrhythmia), exertional syncope, and recurrent syncope.
of awareness. Usually in middle-aged women and benign.
Non-invasive
Metabolic disorders • EKG: to exclude complete heart block, an aberrant
• Hypoglycemia: usually diabetes but may be caused by conduction pathway, and an acute cardiac event (76, 77).
insulinoma, Addison’s disease, and post-gastrectomy. • 24-hour ambulatory or patient activated EKG recording
Tends to occur after fasting. Associated symptoms may (Holter monitoring): can be useful if the attacks are
include sweating and a sensation of hunger. frequent enough to be captured and the patient is known
• Pheochromocytoma: episodic release of catecholamine to have, or is suspected of having, heart disease.
may lead to transient palpitations, headache, sweating, However, it may fail to detect arrhythmias if they do not
and hypotension. Many patients have postural occur during the monitoring period, and increasing the
hypotension due to chronic excess secretion of period of monitoring only slightly improves sensitivity.
catecholamines, but rarely causes syncope. If an arrhythmia is detected (e.g. frequent ventricular
• Hyperventilation-induced alkalosis. ectopic beats) but it is associated with no symptoms, it
does not prove that the arrhythmia is the cause of the
Sleep disorders syncope. Nevertheless, the arrhythmia may point to an
• Narcolepsy: excessive daytime sleepiness (see p.57). underlying structural heart disease.
• Cataplexy: sudden onset of focal or generalized loss of • Echocardiography: if valvular heart disease, outflow
muscle tone, often precipitated by emotion such as obstruction or cardiomyopathy is suspected.
laughter, and resulting in either episodic limb weakness • Carotid sinus stimulation under EKG control: aims to
or sudden collapse from an upright position (see p.57). detect carotid sinus hypersensitivity but is of limited use.
• Obstructive sleep apnea. Should be performed with the patient lying supine. A
period of more than 3 seconds of asystole on EKG
Psychiatric monitoring is positive, although this response is non-
Pseudoseizures. specific, being present in up to 20% of the asymptomatic
population.
INVESTIGATIONS • Upright tilt table testing: a method of diagnosing vaso-
The history, physical examination and EKG are the core of the vagal syncope in patients with frequent recurrent
diagnostic workup. In most patients, there is an obvious undiagnosed syncopal events but no heart disease and
precipitant and no investigations are required, or at most an patients with recurrent syncope and heart disease but no
EKG and hemoglobin. For example, syncope in the elderly evidence of arrhythmias. The technique involves constant
often results from polypharmacy and abnormal physiologic EKG recording and blood pressure monitoring while the
responses to daily events. The need for further investigations are patient is brought from a supine to an upright position

74 75 74 Contrast-
enhanced CT brain
scan, coronal plane,
showing a colloid
cyst of the third
ventricle as a round
hyperintense mass
lesion (arrow),
causing obstructive
hydrocephalus.

75 T2W MRI brain


scan, axial plane,
showing a colloid
cyst of the third
ventricle (arrow).
Syncope 63

(e.g. 70° head-up tilt) in a series of stages, and Blood


maintained in that position, in the absence of drugs for Basic laboratory tests are not usually helpful and should be
up to 40 minutes. The test is positive if there is syncope minimized:
or presyncope in association with hypotension and/or • Hemoglobin level may reveal anemia.
bradycardia. The sensitivity of this test is up to 70% but • Serum urea and electrolytes and glucose may reveal the
the specificity is uncertain. If there is no abnormal most common metabolic causes.
response, an isoprenaline (isoproterenol) infusion can be • Prolonged fast to exclude hypoglycemia.
given to improve sensitivity further (at the expense of • Short tetracosactrin (cosyntropin) test, if abnormal urea
specificity). It is commenced at 1 µg per minute and the and electrolytes, to exclude Addison’s disease.
rate progressively increased every 5 minutes by a further
microgram per minute at successive tilts. The Urine
isoprenaline (isoproterenol) infusion initially increases 24-hour urine collection for vanillylmandelic acid to exclude
the heart rate and then the rhythm may change from pheochromocytoma.
sinus to another rhythm (e.g. junctional rhythm),
resulting in a fall in blood pressure and symptoms of Neurologic
syncope. The symptoms resolve with resumption of the Neurologic investigation is rarely helpful unless additional
supine posture. The test is again positive if there is neurologic symptoms or signs are present.
syncope or presyncope in association with hypotension • CT or MRI brain scan: if drop attacks or suspected
and/or bradycardia. Arnold–Chiari malformation, hydrocephalus, colloid cyst
of the third ventricle, or subdural hematoma.
Invasive • EEG (combined with simultaneous EKG monitoring): if
• Electrophysiologic studies: directly assess intracardiac suspected seizure disorder. A positive EEG may support
conduction and the presence of inducible supraventricular the diagnosis of epilepsy but a negative study does not
and ventricular arrhythmias by intracardiac stimulation. exclude it.
Although invasive, the morbidity is low (1–2%) and • Nerve conduction studies and EMG: if neuropathy
mortality very low. May be indicated if recurrent and suspected.
disabling syncopal episodes, clinical suspicion of an • Autonomic function tests.
arrhythmia, abnormal resting EKG, arrhythmias on
Holter monitoring, and structural heart disease. Biopsy
Rectal: if amyloidosis suspected (rare).

Psychiatric
Occasionally a psychiatric assessment may be useful in
patients with otherwise unexplained frequent syncopal
76 events and no injury, as patients with anxiety disorders,
panic reaction and somatization disorders may report
syncope as a symptom.

DIAGNOSIS
The key to the diagnosis of syncope is a sound clinical
history from the patient and an eyewitness.

TREATMENT
Aims to identify and correct the specific underlying cause:
• In most patients, whose syncope was due to the
simultaneous occurrence of several predisposing factors
(e.g. dehydrated, tired, hot and stuffy room), an
explanation and reassurance is all that is required.
76 Electrocardiograph (EKG) showing slow atrial fibrillation with a • Attend to any heart disease: e.g. pacemaker insertion may
ventricular rate of about 45 per minute. help patients with complete heart block and carotid sinus
hypersensitivity.
• Correct any metabolic and hormonal deficiencies.
77

77 Electrocardiograph (EKG) showing bifascicular block (a combination


of right bundle branch block and left anterior hemiblock) due to
coronary artery disease in a 70 year old male who presented with three
episodes of collapse due to cardiac syncope related to paroxysmal
bradycardia. Electrophysiologic studies failed to induce clinically significant
tachycardia and the patient responded well to permanent pacemaker
insertion. Bifascicular block can develop into complete conduction block
due to trifascicular block or complete AV block.
64 Disorders of Consciousness

Postural hypotension • Advise patients with recurrent syncope to avoid precipi-


• Stop any drugs that may cause postural hypotension (if tating factors (e.g. large meals) and circumstances that
possible), particularly in the elderly. These include may lead to injury if they do lose consciousness and to
diuretics (particularly in hot weather), α-adrenoreceptor stand slowly. Driving should be restricted until free of
blockers (e.g. prazosin), psychotropic drugs with α- recurrent syncopal episodes for up to 1 year if the cause
adrenoreceptor blocking activity (tricyclic antidepres- of the loss of consciousness has not been elucidated and
sants, antipsychotics, including prochlorperazine treated appropriately.
[Stemetil]); α-methyldopa, nitrovasodilators (e.g.
glyceryl trinitrate, including long-acting formulations), PROGNOSIS
and antiparkinsonian drugs (L-dopa, bromocriptine). Depends upon the underlying cause; benign in many cases,
• Avoid large meals, especially when accompanied by alcohol. a warning sign of sudden death in a minority of others.
• Change posture, from supine to standing, slowly (avoid Sudden death occurs within the next year in about 20% of
sudden changes). patients with syncope due to ischemic or structural heart
• Waist-high supportive garments (e.g. elasticated disease. The spontaneous remission rate is high and the rate
stockings) or antigravity suits may be helpful for venous of sudden death is low in patients with unexplained syncope
pooling but can be uncomfortable. after intensive investigation.
• Elevate the head of the bed at night.
• High salt diet.
• Prophylactic medication such as the mineralocorticoid
fludrocortisone in low dose; sympathomimetics such as
ephedrine; caffeine; non-steroidal anti-inflammatory drugs
such as indomethacin, if there is no contraindication; ergot
alkaloids, theophylline, β-adrenoreceptor antagonists with
intrinsic sympathomimetic activity; disopyramide and
octreotide (somatostatin analogue). Insertion of a
pacemaker may be indicated for cardiogenic syncope if
medical therapy fails.

al., (1999) Brain death diagnosis in mislead- controlled crossover trial of modafinil in the
FURTHER READING ing conditions. Q. J. Med., 92: 407–414. treatment of excessive daytime sleepiness in
Hachinski V (1992) Brain death or loss of human narcolepsy. Neurology, 49: 444–451.
COMA brain life? Arch. Neurol., 49: 572. Douglas NJ (2001) The sleepy patient. J. Neurol.
Bateman DE (2001) Neurological assessment of Inwald D, Jakobovits I, Petros A (2000) Brain Neurosurg. Psychiatry, 71 (suppl 1): i3–i6.
coma. J. Neurol. Neurosurg. Psychiatry, 71 stem death: managing care when accepted Fry JM (1998) Treatment modalities for nar-
(suppl 1): i13–i17. medical guidelines and religious beliefs are in colepsy. Neurology, 50 (Suppl 1): S43–S48.
Bates D (1993) The management of medical conflict. BMJ, 320: 1266–1268. Gencik M, Dahmen N, Wieczorek S, et al (2001)
coma. J. Neurol. Neurosurg. Psychiatry, 56: Lövblad K-O, Basssetti C (2000) Diffusion- A prepro-orexin gene polymorphism is associ-
589–598. weighted magnetic resonance imaging in brain ated with narcolepsy. Neurology, 56: 115–117.
Bates D (2001) The prognosis of medical coma. death. Stroke, 31: 539–542. Guilleminault C, Brooks SN (2001) Excessive day-
J. Neurol. Neurosurg. Psychiatry, 71 (suppl 1): Pugh J, Clarke L, Gray J et al (2000) Presence of time sleepiness. A challenge for the practising
i20–i23. relatives during testing for brain stem death: neurologist. Brain, 124: 1482–1491.
Cartlidge N (2001) States related to or confused Questionnaire study. BMJ, 321: 1505–1506. Krahn LE, Black JL, Silber MH (2001) Narcolep-
with coma. J. Neurol. Neurosurg. Psychiatry, Wijdicks EFM (2001) The diagnosis of brain sy: new understanding of irresistible sleep.
71 (suppl 1): i8–i19. death. N. Engl. J. Med., 344: 1215–1221. Mayo Clin. Proc., 76: 185–194.
Fisher CM (1995) Brain herniation: a revision of Younger SJ (1992) Defining death. Arch. Neurol.,
classical concepts. Can. J. Neurol. Sci., 22: 49: 570–572. SYNCOPE
83–91. Arthur W, Kaye GC (2000) The pathophysiology
Johnson MH (2001) Assessing confused patients. PERMANENT VEGETATIVE STATE of common causes of syncope. Postgrad. Med.
J. Neurol. Neurosurg. Psychiatry, 71 (suppl 1): Andrews K (1999) The vegetative state – clinical J., 76: 750–753.
i7–i12. diagnosis. Postgrad. Med. J., 75: 321–324. Kapoor WN (2000) Syncope. N. Engl. J. Med.,
Meagher DJ (2001) Delirium: optimising man- Kennard C, Illingworth R (1995) Persistent veg- 343: 1856–1862.
agement. BMJ, 322: 144–149. etative state. J. Neurol. Neurosurg. Psychiatry, Linzer M, Yang EH, Estes M III, Wang P, Vor-
Mercer WN, Childs NL (1999) Coma, vegetative 59: 347–348. perian VR, Kapoor WN, for the Clinical Effi-
state, and the minimally conscious state: Di- McLean SAM (2001) Permanent vegetative state cacy Assessment Project of the American Col-
agnosis and management. The Neurologist, 5: and the law. J. Neurol. Neurosurg. Psychiatry, lege of Physicians (1997) Diagnosing syncope.
186–193. 71 (suppl 1): i26–i27. Part 1: Value of history, physical examination,
Overell J, Bone I, Fuller GN (2001) An aid to Review by a working group convened by the and electrocardiography. Ann. Intern. Med.,
predicting prognosis in patients with non-trau- Royal College of Physicians and endorsed by 126: 989–996.
matic coma at one day. J. Neurol. Neurosurg. the conference of Medical Royal Colleges and Linzer M, Yang EH, Estes M III, Wang P, Vor-
Psychiatry, 71 (suppl 1): i24–i25. their Faculties of the United Kingdom (1996) perian VR, Kapoor WN, for the Clinical Effi-
Ropper AH (1986) Lateral displacement of the The Permanent Vegetative State. J. R. Coll. cacy Assessment Project of the American Col-
brain and level of consciousness in patients Physicians Lond., 30: 119–121. lege of Physicians (1997) Diagnosing syncope.
with an acute hemispheral mass. N. Engl. J. Zeman A (1997) Persistent vegetative state. Part 2: Unexplained syncope. Ann. Intern.
Med., 314: 953–958. Lancet, 350: 795–799. Med., 127: 76–86.
Wijdicks EFM, Miller GM (1997) MR imaging of Mathias CJ, Kimber JR (1998) Treatment of pos-
progressive downward herniation of the dien- NARCOLEPSY tural hypotension. J. Neurol. Neursurg. Psy-
cephalon. Neurology, 48: 1456–1459. Aldrich MS (1990) Narcolepsy. N. Engl. J. Med., chiatry, 65: 285–289.
323: 389–393. Parry SW, Kenny RA (1999) Tile table testing in
BRAIN DEATH Aldrich MS (1998) Diagnostic aspects of nar- the diagnosis of unexplained syncope. Q. J.
Bernat JL (1992) Brain death. Arch. Neurol., 49: colepsy. Neurology, 50 (Suppl 1): S2–S7. Med., 92: 623–629.
569–570. Broughton RJ, Fleming JAE, George CFP, et al.
De Tourtchaninoff M, Hantson P, Mahieu P, et (1997) Randomized, double-blind, placebo-
Chapter Three 65

Epilepsy
EPILEPSY • Age: any age. Peak incidence in early childhood and a
second peak in the elderly (over-60 age group).
• Gender: slight excess among males.
DEFINITION
Epileptic seizure CLASSIFICATION
A paroxysmal, stereotyped disturbance of consciousness, Epileptic seizures are now classified as generalized and
behavior, emotion, motor function, perception or sensation partial, based on clinical and electroencephalograph (EEG)
(which may occur singly or in any combination), that results distinctions. This classification replaces such imprecise terms
from cortical neuronal discharge. as ‘grand mal’ and ‘petit mal’.
N.B. Epilepsy may manifest itself in the form of more
Epilepsy than one seizure type in the same patient. In addition,
A condition in which epileptic seizures recur, usually
spontaneously.

EPIDEMIOLOGY Table 7 International classification of epilepsies and


• Incidence: 20–70 per 100 000 per year. epileptic syndromes
• Prevalence: 0.5–1.0% of the population (active epilepsy). Localization-related (local, focal, or partial) epilepsies and syndromes
• Cumulative/lifetime (or total) prevalence: • Idiopathic epilepsy with age-related onset:
– Single seizure (including seizure with acute illness, but – Benign childhood epilepsy with centro-temporal spikes
not febrile seizure): 3%. – Childhood epilepsy with occipital paroxysms
– Active epilepsy and/or on treatment: 4–5 per 1000. • Symptomatic epilepsy
– N.B. These figures exclude febrile convulsions, which
occur in about 5% of children. Generalized epilepsies and syndromes
• Idiopathic epilepsy with age-related onset (listed in order of
age of onset):
Table 6 International classification of seizures – Benign neonatal familial convulsions
– Benign neonatal non-familial convulsions
Prevalence in adults
– Benign myoclonic epilepsy in infancy
Partial seizures (beginning focally) 55%
– Childhood absence epilepsy (formerly known as pyknolepsy)
• Simple partial seizures (without impaired
– Juvenile absence epilepsy
consciousness) 5%
– Juvenile myoclonic epilepsy (formerly known as impulsive
– With motor symptoms
petit mal)
– With somatosensory or special sensory symptoms
– Epilepsy with generalized tonic-clonic seizures (GTCS) on
(e.g. olfactory, auditory, visual hallucinations)
awakening
– With autonomic symptoms
• Other idiopathic epilepsies
– With psychological symptoms
• Idiopathic or symptomatic epilepsy (listed in order of age of
• Complex partial seizures (with impaired consciousness) 15%
onset):
– Simple partial onset followed by impaired consciousness
– West’s syndrome (infantile spasms)
– Impaired consciousness at onset
– Lennox–Gastaut syndrome (childhood epileptic
• Partial seizures evolving into secondary
encephalopathy)
generalized seizures 35%
– Epilepsy with myoclonic-astastic seizures
Generalized seizures (convulsive or non-convulsive) 35% – Epilepsy with myoclonic absence seizures
• Absence seizures • Symptomatic epilepsy
– Typical 1% • Non-specific syndromes:
– Atypical – Early myoclonic encephalopathy
• Myoclonic seizures 1% – Early infantile epileptic encephalopathy with suppression
• Clonic seizures burst.
• Tonic seizures • Specific syndromes:
• Tonic-clonic seizures 30% – Epileptic seizures complicating other disease states (such as
• Atonic seizures cerebral malformations, inborn errors of metabolism, and
• Status epilepticus disorders presenting as progressive myoclonic epilepsy)
Unclassifiable/undifferentiated epileptic seizures 10% Continued on page 66
66 Epilepsy

– Benign familial neonatal convulsions – chromosomes 8q


Table 7 (continued)
and 20q.
Epilepsies and syndromes with both focal and generalized seizures – Benign familial infantile convulsions – chromosome 19.
• Neonatal seizures • Juvenile myoclonic epilepsy – possibly HLA region of
• Severe myoclonic epilepsy of infancy chromosome 6p (controversial: see p.86).
• Epilepsy with continuous spike waves during slow-wave sleep • The ‘spike wave trait’: an autosomal dominantly inherited
• Acquired epileptic aphasia (Landau–Kleffner syndrome) tendency to develop paroxysms of spike wave complexes,
usually at 3 Hz, but infrequently associated with clinical
Special syndromes
seizures.
• Situation-related seizures:
• Some progressive myoclonic epilepsies are inherited as a
– Febrile convulsions
recessive trait (see p.87).
– Seizures related to other identifiable acute metabolic or toxic
events (such as stress, hormonal changes, drugs, alcohol
Partial epilepsy
withdrawal, or sleep deprivation)
Siblings of people with focal epilepsy have a <5% risk of
• Isolated, apparently unprovoked, seizures or isolated status
epilepsy.
epilepticus
• Epilepsies characterized by specific modes of seizure
Simple (single gene) inheritance
precipitation
• Uncommon single gene disorders: tuberous sclerosis,
• Chronic progressive epilepsia partialis continua of childhood
familial cavernous angiomas (chromosome 7q), familial
periventricular heterotopia (chromosome Xq28).
• Autosomal dominant nocturnal frontal lobe epilepsy:
– One family was found to have a missense mutation in a
seizures of one type may be easy to control in some patients gene on chromosome 20 which is closely related to the
but difficult to treat in others. neuronal nicotinic acetylcholine receptor alpha 4 subunit,
Epileptic syndromes are determined by: seizure type(s), which is important for modulating glutamate.
age of onset, EEG findings (interictal and ictal) and – Incomplete penetrance.
associated findings (e.g. family history, neurologic signs). – Onset usually in childhood.
They provide information about the likelihood of – Aura often present on awakening; tonic spasms or
identifying an underlying cause, the prognosis, and the hyperkinetic motor symptoms.
appropriate investigations and management, including • Autosomal dominant partial epilepsy with variable foci.
medications, lifestyle modification and genetic counselling. • Familial temporal lobe epilepsy: dominantly inherited.
• Autosomal dominant rolandic epilepsy with speech
ETIOLOGY dyspraxia.
Epilepsy (like stroke) is a symptom of an underlying
disorder(s) of the brain. The likely causes vary depending Complex (polygenic or multifactorial) inheritance:
on the type of epilepsy and the age of the patient (i.e. more • Idiopathic partial epilepsies: benign partial epilepsy of
likely to be idiopathic in young people, whereas in the childhood with centro-temporal spikes (benign rolandic
elderly about half are idiopathic, a third due to vascular epilepsy).
diseases, and the majority of the remainder due to tumors • Cryptogenic/symptomatic partial epilepsies: temporal
and other structural lesions). lobe epilepsy.
• Situation-related seizures: siblings of people with febrile
Idiopathic seizures have an 8% risk of epilepsy.
In about two-thirds of epilepsies no specific cause is found
(idiopathic epilepsies). Idiopathic generalized absence Perinatal factors
epilepsies: The contribution of perinatal damage to the development
• Age-dependent onset between childhood and adolescence. of partial epilepsy is difficult to assess but is probably less
• Distinct phenotypic expression. important than previously thought. Perinatal causes include
• Frequent familial clustering. birth trauma, intracranial hemorrhage, hypoxia and
• Abnormal EEG in about 10% (but up to 30%) of hypoglycemia.
otherwise healthy family members.
Structural cerebral cortex lesions
Genetic factors Interfere with function of the cortex or lead to cortical gliosis.
Inheritance of epilepsy is predominantly polygenic and
multifactorial. Congenital
• Hamartomas.
Generalized epilepsy • Arteriovenous malformations (cavernous angiomas) (78).
Siblings or children of people with generalized epilepsy have • Tuberous sclerosis (79).
up to a 10% risk of developing epilepsy. • Storage diseases: cerebral lipidosis.
• Primary generalized epilepsies: • Neuronal migration disorders causing cortical malformations.
– 30–50% of cases may have a genetic basis. • Generalized (agyria, pachygyria/lissencephaly and band
– 5–10% of children whose parents or siblings have a heterotopias).
primary generalized epilepsy develop epilepsy. • Unilateral hemispheric (hemimegalencephaly).
– Usually inherited as an autosomal dominant trait, • Focal (cortical dysplasia, polymicrogyria, schizencephaly,
probably polygenic. focal heterotopias).
Epilepsy 67

Trauma • Depolarization waves (paroxysmal depolarization shift)


• Birth injury (trauma or hypoxia). affect dendrites of epileptic neurons.
• Head injury (cortical contusion more than laceration). • Electric current flows from neutral cell bodies in layers 5
and 6 of the cerebral cortex to depolarized dendrites in
Vascular the superficial layer of the cortex, which is recorded by
• Ischemic or hemorrhagic stroke. EEG scalp electrodes as a negative spike discharge.
• Cerebral edema: hypertensive encephalopathy. • The epileptic discharge also activates inhibitory feedback
circuits which repolarize the cell bodies while the
Infection dendrites are recovering from depolarization, resulting
• Meningitis. sometimes in a surface-negative slow wave following the
• Abscess. spike discharge; current flows from the hyperpolarized
• Encephalitis. cell body to the neutral dendrites, causing a surface-
negative slow wave in the EEG.
Tumor • Spike wave complexes may remain localized, spread over
Primary and secondary. the cortex via association fibers, or spread to the opposite
cerebral hemisphere through the corpus callosum causing
Degenerative a ‘mirror focus’ or a generalized epileptic seizure.
Alzheimer’s disease.
Generalized epilepsy
Metabolic/toxic factors • Thalamo-reticular and thalamo-cortical projections
Epileptic seizures may be caused, or exacerbated by, many combine with inhibitory intra-thalamic projections to
disturbances to cellular metabolism, such as hyperthermia, generate a rhythmic discharge.
hypoxia, hypocapnia, hyponatremia, uremia, hypocalcemia, • Absence seizures result from synchronous burst-firing
hypoglycemia, liver failure, porphyria, pyridoxine deficiency, generated in the thalamocortical circuit, which is made up
drugs (e.g. solvents, cytotoxic drugs [e.g. nitrogen mustard of three principal neuronal populations: neocortical
drugs and ifosfamide]), drug and alcohol withdrawal, fatigue, pyramidal cells, thalamic relay neurons, and the exclusively
pregnancy and toxins (e.g. insecticides and heavy metals). GABAergic neurons which form nucleus reticularis thalami
(NRT). The frequency of burst-firing between reciprocally
PATHOPHYSIOLOGY interconnected thalamic relay neurons and neocortical
Focal epilepsy pyramidal neurons is synchronized by NRT neurons.
• Gliosis or a local deficiency of inhibitory neuro- GABAergic neurotransmission is important in the
transmitters (gamma aminobutyric acid [GABA], glycine generation of synchronous burst-firing in the thalamo-
and taurine) lead to a failure of normal modulation of cortical circuit. PET scanning in patients with generalized
dendrites of neurons from surrounding cells. epilepsies shows thalamic activation.
• Shortening of dendrites bring excitatory inputs closer to
the cell body.

78 79

78 Facial photograph of a young male with Sturge–Weber disease and 79 Photograph of the face of a patient with epilepsy due to tuberous
epilepsy due to a left fronto-parietal venous hemangioma of the sclerosis. Note the diffuse small adenoma sebaceum (pink, wart-like
meninges over the left fronto-parietal lobes and atrophy of the angiofibromas) over the cheeks, resembling acne. (Courtesy of Dr AM
underlying subcortex. Note the deep red port wine nevus Chancellor,Tauranga, New Zealand.)
(hemangiomatous malformation) not only within but also outside the
distribution of the left trigeminal nerve.
68 Epilepsy

PATHOLOGY lip smacking


There are many single, and in some patients dual, chewing
pathologies which may cause epilepsy. These are listed in the fiddling, fumbling with clothing
Etiology section (see Structural cerebral cortex lesions, slight jerking of limbs (myoclonus)
above). However, special mention is made below of mesial passing urine
temporal sclerosis because this is the most common falling to the ground (akinetic attack).
pathology in patients with temporal lobe epilepsy. – Suspect in children who:
appear inattentive in class
Mesial temporal sclerosis (MTS) ‘switch on and off’, day dream
• Neuronal loss/gliosis in hippocampus ± amygdala, fall for no obvious reason.
uncus, parahippocampal gyrus is found in 50–75% of – Attacks cease in 80% of children by the age of 20 years.
patients undergoing temporal lobe surgery for temporal • Cause: generated at the thalamic level, involving
lobe epilepsy (80–82). thalamo-cortical projections.
• Synaptic reorganization of dentate granule cell (DGC)
axons (mossy fibers) within the hippocampal formation. Myoclonic seizures
• Pathogenesis is unknown; a history of febrile seizures in • Age group: children and juveniles.
early childhood is more frequent, though not exclusive. • Symptoms: brief jerking of the face, trunk, arms or legs.
• Duration: 1–5 seconds.
CLINICAL AND EEG FEATURES • Ictal EEG: polyspike waves, spike waves, or sharp and
Generalized epileptic seizures slow waves.
Absences (petit mal) • Special features: often occur in series.
• Age group: children and juveniles.
• Symptoms: sudden brief loss of consciousness, staring Atonic or akinetic seizures (astatic)
and blinking; immediately regain consciousness. • Age group: infants and children.
• Duration: few–30 seconds. • Symptoms:
• Ictal EEG: paroxysmal generalized rhythmic regular – Sudden loss of muscle tone causing fall and head injury.
3 (2–4) Hz spike-and-wave discharges. – No loss of consciousness.
• Special features: – May occur together with absence attacks.
– Long absences may be accompanied by: • Duration: a few seconds.
simple actions (automatisms) • Ictal EEG: polyspike waves, flattening, or low-voltage
upward rolling of the eyes fast activity.

80 81

82 80 Microscopic examination of the normal Ammon’s horn showing


the lateral geniculate body in the upper left of the photograph,
Sommer’s sector in the upper right, and the dentate gyrus in the
center.

81 Microscopic examination of sclerosis of Ammon’s horn in a patient


with complex partial seizures.

82 Microscopic examination of infarction of Ammon’s horn in a


patient with complex partial seizures.
Epilepsy 69

• Special features: • Subtypes:


– Suspect in children who fall for no obvious reason. – Focal motor seizures (Jacksonian epilepsy): described by
– May occur together with absence attacks. Hughlings Jackson, English neurologist 1835–1911,
– Absence and akinetic attacks associated with tonic usually start with twitching of face or thumb which
seizures and mental retardation constitute the spreads over several seconds to the ipsilateral arm and leg
Lennox–Gastaut syndrome in which the EEG usually as epileptic activity ‘marches’ over the contralateral motor
shows atypical spike-wave paroxysms of 2–2.5 Hz and cortex.
the prognosis for seizure control is poor. – Focal sensory seizures: arise from primary sensory cortex
• Effect of the seizure: temporary failure of the reticulo- and can take the form of contralateral paresthesia
spinal pathways leading to inhibition of normal postural (somatosensory cortex), unformed visual hallucinations,
mechanisms. e.g. light flashes (visual cortex), transient vertigo and
simple auditory hallucinations such as crackling, buzzing
Infantile spasms (West’s syndrome) (auditory cortex-superior temporal gyrus).
• Age group: first year of life.
• Symptoms: Complex partial seizures
– Forward flexion with arms outstretched (salaam attacks) • Age group: any age.
– Commonly associated with tuberous sclerosis • Symptoms: depend on location of seizure focus (see sub-
• EEG: grossly abnormal (hypsarrhythmia) types, below); impaired consciousness; postictal confusion.
• Special features: • Duration: minutes.
– Mental retardation in most. • Ictal EEG: unilateral epileptiform discharges; may spread
– Seizures may respond to vigabatrin, ACTH or bilaterally.
corticosteroids. • Subtypes:
– Olfactory and gustatory hallucinations: usually
Generalized tonic-clonic seizures (grand mal) unpleasant smells and taste, and dreamy state, arise from
• Age group: any age. uncus of temporal lobe.
• Symptoms: – Visual and auditory hallucinations: formed visual
– Premonitory irritability, headache, elation, depression in hallucinations of objects, animals and people originate in
some; others have no warning. the visual association cortex; complex hallucinations of
– Focal onset (an aura) indicates initial partial seizure. music and conversation may arise in the auditory
– Tonic phase (15–30 seconds): association cortex.
contraction of facial, jaw, limb, chest, trunk muscles – Emotional disturbances: rapid onset of unexplained
initial cry sometimes as air is expelled fear/terror or elation, may arise in the temporal lobe.
loss of muscle tone (fall) – Memory disturbances (transient):
cyanosis; tonic convulsions. amnesia
– Clonic phase: rhythmic jerking/convulsions of limbs. dèja vu (a false sensation of familiarity)
– Post-ictal phase: jamais vu (a false feeling of unfamiliarity and strangeness)
muscle relaxation illusion of time passing more quickly or slowly.
slow deep breathing through clenched jaws – Automatisms:
deep sleep, stupor, confusion for minutes to hours episodes of movements and apparently confused
awareness of headache and muscle soreness later. behaviors, with impaired awareness and consciousness
• Duration: 1–3 minutes. (e.g. lip smacking, chewing movements, picking at
• Ictal EEG: generalized epileptiform activity (focal or clothes, fumbling with objects, and driving a car)
generalized onset). ictal or post-ictal
• Special features: patient cannot remember them.
– Tongue biting and urinary incontinence sometimes.
– A succession of tonic-clonic seizures, without regaining Secondarily generalized seizures
consciousness for more than 30 minutes, is known as • Initial symptoms depend on:
convulsive status epilepticus (see p.82). – Seizure type (simple/complex partial).
– Location of seizure focus.
Partial epileptic seizures (clinical types of seizures) • Subsequent symptoms:
Simple partial seizures (focal) – Generalized tonic-clonic convulsions.
• Age group: any age.
• Symptoms: depend on location of seizure focus (see
subtypes below); no loss of consciousness and no
postictal confusion.
• Duration: seconds to minutes.
• Ictal EEG: contralateral epileptiform discharges.
N.B. scalp recordings may reveal no ictal abnormalities.
• Special features: an ‘aura’ is a simple partial seizure.
70 Epilepsy

Partial epileptic seizures (anatomic origin – Underlying neurologic or developmental abnormality.


of seizures) – Family history of epilepsy in a parent or sibling.
Frontal lobe seizures – If none of these features, the risk of epilepsy is no greater
• Simple partial, complex partial, ± secondary generalization. than for other children. If all of these features, the risk of
• Brief episodes, lasting seconds, of: epilepsy is still only 10%.
– Adversive head movements.
– Prominent motor manifestations (particularly in the DIAGNOSIS OF EPILEPSY
legs). 1. Is it epilepsy?
– Impaired consciousness. • Clinical, based on a detailed description of events
• Recovery is rapid with very little post-ictal confusion. experienced by the patient before, during, and after a
• Recur in clusters several times daily or often overnight. seizure and, as importantly, on an eye-witness account.
• EEG: focal epileptiform activity may be difficult to • May be supported by EEG findings.
identify among movement artefact. • Should be made only when certain, or as certain as one
can be, and communicated to the patient and family when
Temporal lobe seizures plenty of time is available to discuss the issues (see below).
• Complex partial ± secondary generalization: • If there is any doubt about the diagnosis, wait for a
– Epigastric rising sensation. further description of another episode.
– Olfactory and gustatory hallucinations. • The diagnosis is a shock to many people, particularly as
– Dèja vu, jamais vu. they have usually been previously well, and because it fre-
– Oral and other primitive automatisms. quently has major emotional, social, scholastic, vocational
– Visual hallucinations. and economic implications for the patient and family.
– Postictal confusion. • There is more to be lost by prematurely misdiagnosing
a non-epileptic as epileptic than delaying the diagnosis of
Parietal lobe seizures epilepsy by a few days, weeks or even months, provided
• Simple partial ± secondary generalization: safety precautions are attended to in the interim.
– Sensory and/or motor symptoms (Jacksonian march).
– Painful sensations occasionally. 2. What type of epilepsy it is?
• See Classification, above.
Occipital lobe seizures • This can be a clue to the cause, treatment and prognosis.
• Simple partial ± secondary generalization:
– unformed visual phenomena (sparks, flashes, phosphenes). 3. What is the cause of the epilepsy?
See Etiology, above.
SPECIAL FORMS OF EPILEPSY
Febrile convulsions 4. Should the epilepsy be treated?
• 3 months to 6 years of age, usually (2–4% of children This depends on the prognosis for recurrent seizures and
<5 years of age). adverse sequelae, the risks and benefits of antiepileptic
• Boys > girls. treatment, and the patients wishes.
• >one-third have a relative who has had febrile convulsion(s).
• Fever. 5. If so, how should it best be treated?
• Respiratory infections, otitis media and tonsillitis are See Treatment, below.
frequently associated.
• No intracranial infection or metabolic derangement. DIFFERENTIAL DIAGNOSIS
• Benign mostly and not requiring drug treatment. Syncope (faints) (see p.59)
• Diazepam or phenobarbitone (phenobarbital) at times • Gradual onset over several seconds to minutes, and often
of high fever prevents seizures but adverse effects limit in special circumstances (e.g. upright posture).
its use. • Light headedness and nausea followed by loss of
• Rectal diazepam or intranasal midazolam can be used at consciousness.
home to stop prolonged seizures. • Associated pallor and flaccid muscle tone. Incontinence
• Explain to parents, reassure, and watch temperature: the may occur.
mainstay of treatment. • Rapid recovery of consciousness upon restoring the
• Recurrence in 30–40% of children; 75% within the first blood flow to the brain, without prolonged postictal
year, 92% by 2 years. Higher risk of recurrence if confusion or drowsiness.
<18 months of age, family history of febrile convulsions
and lower and less prolonged fever associated with febrile Transient ischemic attacks of the brain (TIA)
convulsion. (see p.186)
• Long term treatment not necessary. • Abrupt onset of negative symptoms of loss of focal
• Epilepsy develops in only 3% of children who have had neurologic or monocular function.
>1 febrile convulsions. • Maximal deficit at onset, without intensifica-
• Features that increase the risk of subsequent epilepsy: tion/spread/‘march’ to other body parts.
– Focal seizure.
– Prolonged seizure (more than 15 minutes). N.B. Partial epileptic seizures usually cause sudden
– Residual neurologic abnormality as a result of the seizure. positive sensory or motor phenomena which spread, albeit
– More than one seizure in the same day, or frequent quickly, to adjacent body parts over a minute or so.
febrile convulsions.
Epilepsy 71

Migraine aura (classical migraine) (see p.95) INVESTIGATIONS


• Past or family history of migraine. Blood tests
• Gradual onset of positive symptoms over 5–20 minutes. • Full blood count and ESR.
• Gradual ‘build-up’/migration/‘march’ from one body • Urea and electrolytes.
part to another, or one modality to another (e.g. from • Blood glucose.
visual symptoms to paresthesia to dysphasia). • Serum calcium and magnesium.
• Headache, nausea and/or photophobia usually follow • Serum prolactin, if doubt as to whether epileptic seizure
the neurologic aura symptoms. or not: serum prolactin is raised two to three times the
baseline level (above 18–23 μg/l [18–23 ng/ml]) at
Movement disorders 15–20 minutes after most generalized tonic-clonic
• Dystonias (see p.113). seizures, and some partial seizures, declining to baseline
• Paroxysmal choreoathetosis: transient unilateral muscle levels by 60 minutes. Prolactin may also be raised after
spasms or movements. syncope however.
• Tremors. • Serum lactate and pyruvate, muscle biopsy and DNA
• Tics. analysis if mitochondrial cytopathy suspected (see p.162).
• Other. • White blood cell (lysosomal) enzyme assay for suspected
storage disorders.
Drop attacks
• Sudden loss of postural tone, often when walking, Electroencephalography (EEG)
causing sudden fall without warning. Role
• Many causes (see p.62). • Aid to the clinical diagnosis of epilepsy:
– A single interictal EEG may show specific epileptiform
Breath-holding attacks discharges in about 35% of epileptics, and may rarely
• Infants or young children. ‘capture’ a seizure.
• Hold their breath, become cyanosed (but can be pale, if – The yield is higher if performed within 24 hours of the
due to head injury), lose consciousness, and may have suspected seizure.
minor clonic limb jerking. – Repeated recordings, often sleep-deprived or during
• Often a form of manipulative behavior when frustrated sleep, increase the yield to up to 85% but the EEG never
and angry. shows epileptiform activity in 15% of epileptics, either
because intermittent epileptic discharges are missed, or
Psychologic disturbances (pseudoseizures) small voltage, epileptic discharges from deep in the brain,
• Hyperventilation attacks. such as mesial temporal cortex, are not detected by scalp
• Panic attacks. electrodes.
• Anxiety attacks. – Abnormalities are more common with idiopathic
• Faints. generalized seizures than partial seizures.
• Tantrums. • Classification of epilepsy into one of the epilepsy
• Deliberate simulation. syndromes.
• Identification of an underlying structural brain lesion
Consider (e.g. focal slow wave, sharp wave or spike wave
• The circumstances and nature of the attack. abnormalities) or neurodegenerative condition (e.g.
• Weeping is common but need to distinguish it from ictal periodic high-amplitude sharp and slow wave complexes
crying. of Creutzfeldt–Jakob disease).
• The emotional context (anxiety, indifference or
ambivalence).
• The history: often vague and inconsistent.
• Evidence of social disruption, personality disturbance.
• History of other functional disorders or unexplained
somatic symptoms.
• Potential for gain.
• A ‘model’ with a real illness (e.g. friend or relative with
epilepsy).
• Ask the patient to hyperventilate and see if the symptoms
can be reproduced.
72 Epilepsy

Patterns – Localized: partial epilepsy, focal brain lesion may be


• Epileptiform activity: spike, sharp wave, or spike-and- present (83–85).
wave discharges (83–85). • Slow wave activity (asymmetric or bilateral), or
– Diffuse: generalized epilepsy (primary or secondary), asymmetric normal rhythms: non-specific.
focal lesion unlikely (86, 87).

83 84

83 EEG of a patient with complex partial seizures due to a right 84 EEG of a 65 year old malewho presented with a large left
medial temporal lobe tumor showing a normal background over the hemisphere cerebral infarct causing a right hemiparesis, dysphasia and a
left hemisphere (top four channels) and random focal polymorphous right homonymous hemianopia that was complicated on day 3 by a
delta slowing and epileptiform sharp wave activity over the right decline in conscious state due to cerebral edema, and concurrent
hemisphere, phase reversing around the right midtemporal area (T4). partial motor seizures involving the right face and limbs.The EEG
shows attenuation of the background rhythm, particularly over the left
hemisphere, and periodic lateralized epileptiform discharges (PLEDs)
85 over the posterior left hemisphere (channels 3 and 4).

85 EEG (16 channel) of a patient with complex partial seizures


showing epileptiform activity (sharp and slow wave) over the right
fronto-temporal region (F8–T8).

87

86

86, 87 Ictal EEGs (86, 8 channel; 87, 16 channel) showing typical 3 Hz spike and wave of absence seizures.
Epilepsy 73

EEG: waking and sleep CT brain scan (usually with intravenous contrast)
• If the waking EEG is normal and epilepsy is suspected, a • Not necessary if a child or young adult with generalized
sleep-deprived recording may unmask epileptiform epilepsy (clinical and EEG).
activity that is enhanced by sleep deprivation and • Indicated if a focal brain lesion is suspected:
entering the sleep state. – Partial seizure.
• EEG prolonged recording + video monitoring over – Focal neurologic signs.
12–24 hours or more: to try and capture an episode. – Lateralized abnormality on EEG.
• May identify a tumor, stroke, AVM, congenital defect or
Electrocorticography (ECOG) area of cerebromalacia which may be responsible for the
Subdural electrodes may be placed via burr holes to monitor patient’s seizures, but fails to identify hippocampal
and localize more accurately a seizure focus, or at operation atrophy and mesial temporal sclerosis.
to guide the extent of the surgical resection. For example,
after conventional temporal lobectomy, residual MRI brain scan
intraoperative epileptiform activity recorded from subdural • Patients with temporal lobe epilepsy who are candidates
electrodes over the posterior medial temporal lobe guides for surgery should have a high definition MRI of the
the surgeon to extend the surgical resection postero- temporal lobes to look for mesial temporal sclerosis
medially. This practice reduces postoperative seizures. (88–90).
(Continued overleaf)

88 89

88 T2W MRI in the coronal plane showing right mesial temporal 90


sclerosis. Note the medial part of the right temporal lobe (arrow) is
atrophied and of slightly brighter signal than the left.

89, 90 MRI brain scans, coronal plane,T2 (89) and T1 (90) weighted
images, in a patient with complex partial seizures, showing enlargement
of the temporal horn of the right lateral ventricle (arrow) due to a loss
of brain substance in the hippocampus and amygdaloid nucleus of the
medial temporal lobe as a result of mesial temporal sclerosis.
74 Epilepsy

MRI brain scan (continued) Single photon emission computed


• Patients with troublesome epilepsy and a normal CT scan tomography (SPECT)
should have an MRI to exclude neuronal migration • Useful for localizing seizure focus, particularly when
disorders, small structural lesions (e.g. hamartomas, clinical features, EEG and anatomic neuroimaging
tumors, or areas of gliosis) in the hippocampus, mesial studies (CT and MRI) are not definitive (97).
temporal sclerosis and hippocampal atrophy which are • Localizes temporal lobe seizure foci more accurately than
missed on CT (88–96). extratemporal foci.
• The diagnosis of mesial temporal sclerosis on MRI can • Ictal SPECT (97% correct) is superior to postictal (71%)
be difficult and may require accurate measurement of the and interictal (48%) SPECT in localizing seizure foci in
temporal lobes to assess size, which with currently unilateral temporal lobe epilepsy. Interictal positron
available methods is very laborious. The MRI results emission tomography (PET) and functional MRI have a
should be interpreted in conjunction with EEG and role here, if available.
clinical findings. Mesial temporal sclerosis appears as a
loss of brain substance in the hippocampus and TREATMENT
amygdaloid nucleus of the medial temporal lobe Aim: to help the patient and family understand the
combined in some cases with increased signal on T2W implications of their epilepsy and to restore normal life by
imaging (88–90). preventing seizures without affecting quality of life.

91 92

91–93 Dysembryoplastic neuroepithelial tumor (arrows).


91 T2W axial and 92 T1W coronal post contrast MRI.
The small size, peripheral site and lack of enhancement are
typical. 93 T1W axial post contrast MRI from a different
93 patient shows a slightly larger tumor with more mixed
signal and one area of enhancement.
Epilepsy 75

General advice • Suggest to the patient that they consider informing their
• Avoid excessive alcohol, fatigue (late nights) and missing close friends and school teacher or immediate work
meals. colleagues of the diagnosis, typical manifestations of
• Contraceptive advice for epileptic women of reproductive seizures, first-aid principles and who to contact in the
age. event of a seizure.

School and work Activities


• Avoid occupations that involve moving heavy machinery, Encourage patients to pursue their interests and participate
electric wiring, truck driving and working at heights (i.e. in all activities as long as they do not put them in a position
roofing); the airlines and armed services do not accept where they could be a danger to themselves or others if they
people who have epilepsy to be a pilot or to handle a gun have a seizure (e.g. driving a car, climbing trees and
respectively. rooftops, swimming alone).

94 95

94, 95 Hamartoma of the tuber cinerarium presenting with gelastic seizures. 94 T1W midline sagittal, and 95 T2W axial MRI
showing a non-enhancing mass (arrow) arising inferior to the hypothalamus. (Courtesy of Dr P Brennan, Consultant
Neuroradiologist, Beaumont Hospital, Dublin, Eire.)

96 97

Ictal Interictal
97 Single photon emission computed tomography (SPECT) scan after
intravenous (i.v.) injection of the radioligand 99m Tc-hexamethylpro-
pylenamine oxime (HMPAO) at the time of a complex partial seizure
showing computer reconstructions of images of regional cerebral
96 MRI brain scan, coronal plane,T2W image, in a patient perfusion obtained with rotating gamma cameras.The ictal scan (left)
with refractory complex partial seizures, showing an shows a red area of increased regional blood flow in the temporal lobe
overall reduction in volume (atrophy) of the entire right on the left (arrow) due to a left temporal lobe seizure focus, which
temporal lobe (left side of the photograph) probably due resolves on the interictal scan (right).
to a previous vascular insult or trauma.The hippocampi
appear normal.
76 Epilepsy

• Most pregnant patients with epilepsy deliver perfectly


Table 8 Driving guidelines (in Australia) healthy babies.
Condition Recommended seizure-free
period before driving Eclampsia
Magnesium sulfate is at least 50% more effective than
Isolated seizure 3–6 months phenytoin or diazepam in preventing recurrent seizures, and
Recently diagnosed epilepsy 3–6 months causes less morbidity in mothers and children.
Chronic epilepsy Up to 2 years Anti-epileptic medication
(history of previously uncontrolled seizures) Indications for treatment
Recurrent seizures due to 3 months • Recurrent unprovoked epileptic seizures that are not
known provocation (e.g. illness) separated by years.
and previously seizure-free • Single seizure in patients with:
– A relevant underlying brain lesion or neurologic
Recurrent seizures on 1 month after resuming effective abnormality, or
withdrawal of medication medication – A specific epileptic syndrome and an unprovoked seizure
2 years if refusing to resume that will eventually need treatment, such as juvenile
medication myoclonic epilepsy.
Seizure causing accident 12 months (minimum) • The risks and benefits of both treatment and no treatment
have been discussed extensively with the patient.
Seizures only in sleep 12 months (if seizure free while • The patient understands that the drug must be taken
awake) regularly and continued for at least 3 years from the time
Temporal lobectomy 12 months of the last seizure, and that about 40% of patients will
need to take anti-epileptic medication lifelong,
Withdrawal of anti-epileptic During the withdrawal and for at depending on the seizure syndrome and other factors.
drug therapy least 3 months thereafter
Principles of anti-epileptic drug therapy
• Aim to control seizures with the lowest effective dose
Driving that causes no adverse effects.
Consider national guidelines: most countries prohibit driving • The goal is not to achieve the quoted ‘therapeutic drug
until the person has been seizure-free for between 6 months level’:
and 2 years, depending on the type of epilepsy, frequency of – A ‘therapeutic’ drug level may not be effective or
seizures, compliance with medication and other issues (Table 8). therapeutic for some patients; higher, so-called ‘toxic’
levels may be required.
Pregnancy – A so-called ‘therapeutic’ drug level may be toxic for
• Seizures increase in about one-third of epileptic women others, and seizures may be controlled with so-called
during pregnancy, often because anti-epileptic drug levels ‘sub-therapeutic’ levels.
have fallen. • Start with low dose and increase slowly until the desired
• Anti-epileptic drug therapy, if indicated, should comprise therapeutic effect (i.e. seizure control) is achieved or until
the most appropriate monotherapy. adverse effects of the drug occur, whichever comes first.
• Fetal malformations such as neural tube defects (e.g. spina • Judge the response primarily on clinical grounds.
bifida), cleft lip and palate, and ventricular septal defect
(VSD) are 25% more common among pregnant women Choice of anti-epileptic drug:
with untreated epilepsy (5–7%) than pregnant women • Select the single most appropriate drug for the seizure
without epilepsy (3.6–5%), and twice as common among type and patient (Tables 9, 10).
pregnant epileptic women taking anti-epileptic drugs • Carbamazepine and valproate are the first line treatments
(7–10%). The risk of a neural tube defect and other skeletal for focal epilepsies.
abnormalities is highest with sodium valproate (1–2%) and • Valproate is the first line treatment for generalized
carbamazepine (0.5–1%), and the risk of cleft lip and palate epilepsies.
and VSD is highest with phenytoin and barbiturates. • Lamotrigine is also very effective for generalized epilepsies.
Supplementing maternal intake of folic acid with 0.4–0.8 • Phenytoin and clonazepam still have a place, but
mg daily reduces the incidence of neural tube defects and barbiturates are now used less.
does not adversely affect epileptic control. The newer • Vigabatrin, lamotrigine, topiramate, tiagabine and
antiepileptic drugs are still under study. gabapentin are of proven benefit in 25–50% of patients
• It is generally best to err on the side of maintaining with refractory partial seizures. It is not clear which is the
seizure control with anti-epileptic drugs than risking most effective drug but vigabatrin is not recommended
seizure recurrence by withholding treatment. because it may cause constriction of the visual fields.
• Pregnant women taking anti-epileptic drugs which enter • Ethosuximide is only used for juvenile abscence epilepsy.
the fetal circulation, induce liver enzymes, and reduce • The cost of newer anti-epileptic drugs (e.g. lamotrigine,
the activity of vitamin K-dependent clotting factors in the topiramate, gabapentin, tiagabine, vigabatrin) is about
fetus, should take vitamin K 20 mg/day during the last 10 times greater than older drugs (phenobarb, phenytoin).
month of pregnancy and be given vitamin K intra-
muscularly during delivery to prevent neonatal hemorr-
hage. Vitamin K is also given to the baby.
Epilepsy 77

Table 9 Drugs of first, second and third choice in the treatment of seizure types
Seizure type First Second Third
Partial Carbamazepine Lamotrigine Phenytoin Clobazam
(Simple/complex)* Sodium valproate Phenobarbitone (phenobarbital)
Topiramate Gabapentin
Tiagabine Vigabatrin
* With, or without, Oxcarbazepine Levetiracetam
secondary generalization Zonisamide
Generalized
Tonic-clonic Sodium valproate Topiramate Phenobarbitone (phenobarbital)
Lamotrigine Lamotrigine Benzodiazepines
Myoclonic Sodium valproate Phenobarbitone (phenobarbital) Clonazepam
Phenytoin Zonisamide

Absence Sodium valproate Lamotrigine Clobazam


Ethosuximide
Undifferentiated Sodium valproate Phenytoin
Lamotrigine

Sodium valproate (Epilim)


• Uncertain mechanism of action. Increases synthesis and Table 10 Adverse effect profiles of anti-epileptic drugs
slows the breakdown of GABA. • Cognitive impairment
• Effective for the primary generalized epilepsies, Marked: phenobarbitone (phenobarbital), clonazepam
particularly tonic-clonic seizures, absences and myoclonus. Mild: topiramate, phenytoin, carbamazepine, clobazam
• As effective as carbamazepine and phenytoin for partial Possible: sodium valproate, oxcarbazepine
seizures, especially if secondary generalization. • Behavioral disturbance
• Available as a 40 mg/ml syrup for pediatric or Marked: phenobarbitone (phenobarbital)
nasogastric tube use, crushable 100 mg tablets, and Mild: clonazepam, clobazam, vigabatrin
200 mg and 500 mg enteric-coated tablets. Possible: topiramate, zonisamide
• Plasma half-life: 6–9 hours, so may be administered as • Ataxia
two or three divided doses daily. Carbamazepine >> phenytoin > sodium valproate
• Starting dose in adults: 200 mg twice daily (bd), given • Tremor
after meals to minimize gastric irritation, and increasing Sodium valproate
to 400 mg bd after 2 days, if tolerated. The full • Visual field defects
pharmacologic action may not occur for some weeks. Vigabatrin
• Dose can be increased slowly to 1 g three times daily • Weight gain
(tds) if necessary and tolerated. Sodium valproate
• Monitoring of blood levels is often not necessary as there • Liver dysfunction
is a poor relation between serum concentrations and anti- Phenytoin, carbamazepine
epileptic efficacy or toxicity. • Osteoporosis
• Drug interactions: increases concentrations of other anti- Phenytoin, phenobarbitone (phenobarbital) > carbamazepine
epileptic drugs such as carbamazepine (and its • Cardiac disturbance
metabolities) and phenobarbitone (phenobarbital), by Phenytoin i.v., carbamazepine oral (bradycardia)
inhibiting their metabolism, and decreases plasma levels • Urinary retention
of phenytoin; it is a minor inhibitor of oxidative Carbamazepine
metabolism but is not a liver enzyme inducer. • Hyponatremia
• Adverse effects: tremor, weight gain due to appetite Carbamazepine
stimulation, and thinning or loss of hair. Cognitive • Hematologic (thrombocytopenia, neutropenia)
impairment is uncommon. Stupor and encephalopathy, Carbamazepine > sodium valproate
although potentially dangerous, are rare idiosyncratic • Skin rash
effects and may be due to accumulation of ammonia. Carbamazepine > lamotrigine
The prothrombin time can be prolonged but not
sufficient to cause bleeding. Liver toxicity, in the form of
a microvesicular steatosis, may occur idiosyncratically,
particularly in young children and when combined with Carbamazepine (Tegretol)
other (?anti-epileptic) drugs. Other adverse effects • First used in 1964.
include anorexia, dyspepsia, nausea, vomiting and rash • Effective for the prophylaxis of generalized tonic-clonic
(predictable), and acute pancreatitis, thrombocytopenia, and partial seizures but not for absence or myoclonic
hyperammonemia, teratogenicity (idiosyncratic). epilepsy (which it may aggravate).
78 Epilepsy

Carbamazepine (Tegretol) (continued) • Starting dose for children: 5 mg/kg daily, for adults
• Available as 100 mg and 200 mg tablets, or 200 and starting and maintenance dose: 300 mg daily, given as a
400 mg controlled-release tablets that are helpful for single dose or in divided doses. (Note, this is not the
avoiding peak-dose adverse effects. loading dose that is required for status epilepticus.)
• Plasma half-life: 24–45 hours initially but after continued • If seizures continue, an increment of 30 mg is
long term use it falls to about 9 (8–24) hours. appropriate, particularly if the serum concentration is
• The drug must be introduced in a low dose to offset mild above 12 mg/l (60 µmol/l).
neurotoxicity (sedation, vertigo, ataxia, diplopia, nausea, • Adverse effects: mental slowing, unsteadiness of gait,
headache). The usual starting dose in adults is 100 mg slurred speech and tremor, and physical examination
three times daily, or preferably, half a 200 mg or 400 mg reveals gaze-evoked nystagmus and tandem gait ataxia.
controlled-release tablet twice daily for 2 days, followed Other predictable adverse effects include nausea,
by half a tablet in the morning and a whole tablet at night, anorexia, vomiting, dyspepsia, cognitive impairment,
and further increases if necessary, aiming to maintain the depression, aggression, drowsiness, headache, paradoxical
plasma level within the therapeutic range and control seizures, megaloblastic anemia, hyperglycemia,
seizures (rather than waiting for another seizure to occur). hypocalcemia, osteomalacia and neonatal hemorrhage.
Even with this cautious approach and the development of Prolonged use is associated with coarsening of facial
tolerance some patients are unable to remain on carbama- features, gum hyperplasia (98), acne and hirsutism.
zepine because of neurotoxicity. In addition a morbilliform • Idiosyncratic effects include blood dyscrasia, lupus-like
skin rash limits its usefulness in 5–8% of patients. syndrome, reduced serum IgA, pseudolymphoma
• Adverse effects: (lymphadenopathy), rash, Stevens–Johnson syndrome,
– Dose-related: neurotoxicity (dizziness, double vision, Dupuytren’s contracture, hepatomegaly and hepato-
unsteadiness), nausea, vomiting, cardiac arrhythmia and toxicity and teratogenicity. Long term use may cause
orofacial dyskinesia. peripheral neuropathy, cerebellar degeneration due to
– Idiosyncratic: skin rash (5–8% of patients), agranulocytosis, Purkinje cell loss and osteomalacia.
aplastic anemia, syndrome of antidiuretic hormone secretion • Drug interactions: an enzyme inducer and may reduce
(leading to fluid retention and hyponatremia) hepatotoxicity, the efficacy of many lipid-soluble drugs such as other
photosensitivity, Stevens–Johnson syndrome, lupus-like anti-epileptic drugs, anticoagulants, corticosteroids,
syndrome, thrombocytopenia and pseudolymphoma. cyclosporine, oral contraceptives, and theophylline. Its
• A major inducer of hepatic cytochrome P450 activity. metabolism may be inhibited, causing neurotoxicity by
Variable autoinduction of metabolism accounts for the enzyme inhibitors such as allopurinol, amiodarone,
wide range of doses and for the substantial interindividual chloramphenicol, cimetidine, imipramine, isoniazid,
variation in concentration found with the same dose. metronidazole, phenothiazines and sulfonamides.
• Drug interactions: carbamazepine accelerates the clearance
of itself (i.e. it induces its own metabolism), ethosuximide, Barbiturates
clonazepam, clobazam, corticosteroids, theophylline, halo- Phenobarbitone (phenobarbital)
peridol, warfarin and hormones. So, most women taking Has been used an anti-epileptic drug since 1912.
the oral contraceptive pill require daily estrogen in a dose of • Inexpensive, widely available and as good as
50 µg. Mutual enzyme induction or inhibition with carbamazepine and phenytoin in controlling generalized
phenobarbitone (phenobarbital), phenytoin, or primidone tonic-clonic and partial seizures.
can result in a small rise or fall in steady-state concentrations • Main drawback: its effect on cognition and behavior:
of either of both drugs. The metabolism of carbamazepine fatigue, listlessness and tiredness in adults and insomnia,
is inhibited, causing neurotoxicity, by sodium valproate, hyperkinesia and aggression in children (and sometimes
cimetidine, danazol, dextropropoxyphene (propoxyphene), in elderly patients). Subtle impairments of mood,
diltiazem, erythromycin, isoniazid, and verapamil. memory and learning can occur in all age groups.
• Usual dose varies from 90 to 300 mg/day.
Phenytoin (Dilantin)
• First used as an antiepileptic drug in 1938.
• Effective for generalized tonic-clonic and partial seizures.
• No longer a drug of first choice, particularly in young
women, because it may cause cosmetic changes (gum 98
hyperplasia, acne, hirsutism, and facial coarsening), as
well as sedation and unfavorable effects on cognitive
function (e.g. attention, memory).
• Available as a 6 mg/ml suspension, as chewable 50 mg
tablets, and as capsules of 30 mg and 100 mg.
• Plasma half-life: about 24 (9–40) hours.
• One of only a few drugs with zero order kinetics at
therapeutic dosage – as the concentration rises, the
capacity of the hepatic cytochrome P-450 enzyme system
to metabolize the drug becomes saturated (usually at
around 300 mg/day), and so a small increment in dose
can produce a large rise in serum level. Conversely, the
circulating concentration may fall precipitously when the 98 Gum hypertrophy in a 20 year old female caused by many years’
dose is modestly reduced. ingestion of phenytoin for epilepsy.
Epilepsy 79

• Long plasma half life of 3–4 days; can therefore be taken • Visual field defects are the most common serious adverse
as a single daily dose. effect, which limits the use of this drug.
• In adults, it should be restricted to patients who cannot
tolerate first-line anti-epileptic drugs or as an adjunct to Lamotrigine (Lamictal)
first line therapy in refractory epilepsy. • Inhibits voltage-gated sodium channels and reduces the
• Withdrawal can lead to a temporary increase in seizure release of glutamate, an excitatory amino acid
frequency. neurotransmitter implicated in the pathophysiology of
epilepsy.
Methylphenobarbitone (Prominal) (mephobarbital) • Effective in both partial and generalized tonic-clonic and
Methylphenobarbitone (mephobarbital) is metabolized to absence seizures in adults and children.
phenobarbitone (phenobarbital), and is given in twice the • A weak inhibitor of dihydrofolate reductase, so long term
dose of phenobarbitone but confers no special advantage. therapy may disturb folate metabolism.
• Does not significantly induce or inhibit hepatic oxidative
Primidone (Mysoline) drug-metabolizing enzymes, nor affect the plasma
Primidone is metabolized to phenobarbitone (phenobarbital) concentrations of concomitant anti-epileptic drugs.
and phenylethylmalonamide, both of which are pharma- • Metabolism induced by anti-epileptic drugs that induce
cologically active. Efficacy is similar to that of phenobarbitone. liver enzymes such as carbamazepine, phenytoin and
phenobarbitone (phenobarbital) (half-life about 15 hours).
Clonazepam (Rivotril) • Metabolism inhibited by sodium valproate (half-life
• A 1,4 benzodiazepine (like diazepam). about 60 hours).
• Effective for generalized tonic-clonic and myoclonic • Plasma half-life: about 29 hours (mean).
seizures, and generalized absence (petit mal) epilepsy and • Eliminated largely as an N-glucuronide conjugate.
partial seizures. • Available as 5 mg dispersible, 25 mg, 50 mg, 100 mg
• Has more sustained and effective anti-epileptic activity and 200 mg standard tablets.
than diazepam (Valium) but tolerance develops. • Starting dose in patients not taking sodium valproate is
• Available as 0.5 mg and 2.0 mg tablets. 25 mg once a day for 2 weeks, followed by 100 mg per
• Dose varies from 0.5–4.0 mg three times daily. day given in two divided doses for 2 weeks. Thereafter,
• Long plasma half life of 20–40 hours. the dose should be increased to achieve the optimal
• Adverse effects include sedation, irritability, and response. The usual maintenance dose is 200–400 mg
aggression. per day in two divided doses.
• Few patients benefit from long term treatment and • Starting dose in patients taking sodium valproate is
nearly half have an exacerbation of seizures when the 25 mg every alternate day for 2 weeks, followed by
drug is withdrawn, particularly if pre-existing brain 25 mg once a day for 2 weeks. The usual maintenance
damage. dose is 100–200 mg a day, given once a day or in two
divided doses.
Clobazam (Frisium) • Adverse effects: a generalized maculopapular skin rash
• A 1,5 benzodiazepine. appears within 4 weeks of gradually starting treatment in
• Less sedative than clonazepam and diazepam but still about 2–5% of patients. The incidence of rash is propor-
commonly cause tiredness as well as depression and tional to the rapidity with which the drug is commenced.
irritability. It usually resolves after immediate withdrawal of lamotri-
• Has a limited role as a long term anti-epileptic drug (like gine, after which the drug can be re-introduced slowly.
clonazepam) but can be effective as a short term Rarely, serious skin rashes such as Stevens-Johnson syn-
treatment to ‘cover’ for special events such as holidays, drome and angioedema have been reported. Some patients
weddings and surgery and for catamenial exacerbations. complain of insomnia which can be minimized by giving
• A single dose of 10–30 mg can also be helpful if taken the second dose in the afternoon. Dose-related adverse
immediately after the first seizure in patients who have effects include dizziness, headache, diplopia, ataxia,
regular clusters of generalized tonic-clonic and partial somnolence, nausea, asthenia, blurred vision and vomiting.
seizures.
Gabapentin (Neurontin)
Vigabatrin (gamma-vinyl GABA) (Sabril) • A GABA-related amino acid.
• A specific, irreversible inhibitor of GABA transaminase, • Mechanism of action: uncertain; it may affect L-type
leading to elevated GABA levels and enhanced inhibitory voltage-dependent calcium channels.
GABAergic transmission. • Effective when used in doses of 1800 mg/day and as an
• Available in 500 mg tablets for use in partial seizures. ‘add-on’ treatment in reducing the frequency of seizures
• Dose: one 500 mg tablet twice daily, increasing weekly by more than half in about 40% of patients with complex
as required to a dose of 2–3 g/day. partial seizures and 60% with secondarily generalized
• Effective as a first line or add-on therapy in reducing tonic-clonic seizures.
partial seizure frequency by more than half in 30–60% of • Not effective for absence seizures.
patients with drug-refractory complex partial seizures. • Available as 300 mg and 400 mg capsules.
• Well tolerated; adverse effects mainly constitute • Usual maintenance dose: 1200–2400 mg/day, but
drowsiness, agitation, irritability, weight gain, depression higher doses may be more effective.
and psychotic behavior. • Does not seem to interact with other anti-epileptic drugs.
• Microvacuolation had been seen in the brains of dogs • Adverse effects on cognitive function may arise with
(but not humans) after vigabatrin treatment. higher doses.
80 Epilepsy

Ethosuximide – Late age at onset (>16 years of age).


• Used only for absence seizures; not effective against – Mental retardation.
generalized tonic–clonic seizures. – Symptomatic epilepsy (i.e. underlying untreated cause).
• Infants require a dose of 20–40 mg/kg per day but – Certain epileptic syndromes, such as juvenile myoclonic
lower weight-related doses are used in adults. In children epilepsy, when unprovoked seizures have occurred.
over 6 years of age, it is started with a dose of 250 mg – Family history of epilepsy.
capsules twice daily, increasing if necessary to three of – Slow wave activity or focal epileptiform activity on EEG
four capsules daily. prior to medication withdrawal.
• Adverse effects include drowsiness and bone marrow – History of atypical febrile seizures.
depression. • There are no definite predictors of recurrence but EEG
findings of epileptiform activity prior to, or during, drug
Topiramate (Topamax) withdrawal are highly predictive of a further seizure if
• A sulfamate derivative structurally unique among anti- anti-epileptic medication is ceased. Relapses may also
epileptic drugs. occur in patients with normal EEGs however.
• Reversibly decreases the number of action potentials in • Withdrawal of any anti-epileptic drug must always be
spontaneous epileptiform bursts and reduces burst gradual in a step-wise fashion, over at least 6 weeks (and
duration in cultured hippocampal neurons, suppressing probably months for barbiturates) because abrupt
intrinsic bursting of proximal subiculum neurons, and withdrawal may provoke rebound seizures.
reducing voltage-gated sodium currents in cultured
cerebellar granule cells. Status epilepticus (see p.82)
• Reduces elevated levels of excitatory amino acids,
glutamate and aspartate. Surgical treatment of epilepsy
• Reversibly enhances post-synaptic GABA receptor Practised for more than a century, but improved under-
currents. standing of the pathogenesis of epilepsy and the recent
• Effective as adjunctive therapy for partial seizures, in advent of high resolution MRI brain imaging and improved
Lennox–Gastaut syndrome and possibly some primary surgical expertise have made it an effective treatment for
generalized epilepsies such as drop attacks. It is not refractory seizures. Once undertaken, it is irreversible.
helpful for, and may aggravate, absence seizures.
• Needs to be commenced gradually, with 25 mg alternate Pre-requisites
days at night or daily at night, otherwise adverse cognitive • The patient has epilepsy and it is resistant to anti-epileptic
effects may be prohibitive. Cognitive disturbances may drugs in maximally tolerated doses.
manifest as slowed speech (and even mimic dysphasia and • No realistic hope of spontaneous remission of the
dysnomia) and a vulnerability to behavioral disturbances epilepsy.
(i.e. cranky, not the same person). Other adverse effects • All diagnostic tests point to a single common epilepto-
include weight loss, renal stones in 1% of patients and a genic focus (see below).
theoretical risk of teratogenicity. • The seizures are causing medical, social and educational
handicap and the patient’s quality of life is likely to
Tiagabine improve after successful surgery.
• A derivative of nipecotic acid that potently and selectively • The risk–benefit ratio for the proposed surgery is
inhibits neuronal and glial GABA uptake. It specifically acceptable.
inhibits the GABA transporter GAT-1 and interacts only • A nationally recognized epilepsy surgery programme
weakly with GABA receptors. with comprehensive pre-and post-surgical evaluation and
• Effective against partial and generalized convulsive seizures. support facilities is available.
• May be contraindicated in generalized absence epilepsy.
Pre-surgical evaluation
Failed monotherapy • Clinical history and seizure pattern.
A single agent (monotherapy) generally achieves satisfactory • Scalp EEG (background, interictal and ictal features).
seizure control without significant adverse effects in about • MRI brain.
half of patients. Although another 15–20% attain better • Neuropsychometry.
control with the addition of a second anti-epileptic drug, it • Visual field examination.
is often better to strive for seizure control with monotherapy • Intracarotid amytal or ‘WADA’ test: determines the
by gradually introducing another agent and then gradually dominant hemisphere for speech and whether memory
withdrawing the initial agent. Overall, about 70% of patients can be sustained without the function of the temporal
can be controlled with monotherapy. lobe that is to be removed. In some centers, this is only
performed in left handed patients.
Withdrawal of antiepileptic medication
• Anti-epileptic drug withdrawal should be considered in If the above tests are concordant and identify a single
patients who have been free of seizures for 2 years or focus and a visible structural lesion (e.g. focal cortical
longer; taking anti-epileptic drugs long term is dysplasia, mesial temporal sclerosis, Sturge–Weber syn-
inconvenient and costly, and associated with adverse drome), then no further investigation may be required.
cognitive and behavioral effects, so it is essential to However, most centers still perform video-EEG to capture
ascertain whether they are still necessary or not. at least a couple of seizures to be absolutely sure that the
• Drug withdrawal is not likely to be successful (i.e. lesion that is to be resected is the lesion responsible for the
seizures recur) if: seizures.
Epilepsy 81

If the scalp EEG is not clearly lateralizing or the MRI several factors such as the epilepsy syndrome and other factors
shows no definite lesion, then further studies are required: listed below.
As stated above (p.80), predictors of an increased risk of
• Video-telemetry with scalp (and, in some centers, recurrence are:
foramen ovale-sphenoidal) EEG recording (video-EEG).
• Interictal and ictal SPECT (may be helpful in some but • Late age at onset.
not all cases), and interictal PET (if available) (99, 100). • Mental retardation.
• Intracranial/subdural electrode recording (usually to • Epilepsy syndrome (e.g. juvenile myoclonic epilepsy).
differentiate between temporal and extratemporal foci, • Etiology (symptomatic epilepsies tend to recur whereas
but again not in all cases). idiopathic and cryptogenic epilepsies do not).
• A family history of epilepsy.
Surgical procedures • Slow wave activity on EEG prior to medication
• Removal of a mass of epileptogenic tissue: withdrawal.
– Amygdalo-hippocampectomy: intractable partial epilepsy • A history of atypical febrile seizures.
and seizure onset in mesial temporal lobe.
N.B. Anterior temporal lobectomy used to be However, there are no definite predictors of recurrence.
undertaken in cases of intractable partial epilepsy with For example, although EEG findings of paroxysmal
seizure onset in temporal lobe and normal memory discharges certainly increase the risk of a relapse after anti-
function in remaining brain, but is no longer done because epileptic drug withdrawal, they don’t indicate a 100% risk,
the seizure focus is often the amygdala/hippocampus, and relapses also occur in patients with normal EEGs. So,
even in the presence of a temporal lobe structural lesion. the EEG findings alone are not a sufficient guide for anti-
• Removal of structurally abnormal tissue: epileptic drug withdrawal. Anti-epileptic drug withdrawal
– Lesionectomy (usually frontal lobe): refractory seizures should be considered in all patients, even those with risk
due to a focal pathology in resectable cortex. factors for recurrence, but other factors can be crucial in
– Hemispherectomy: intractable focal +/– generalized coming to a decision (such as whether the patient is going
seizures with unilateral hemispheric pathology and to drive a motor vehicle or not).
contralateral non-functional hemiparesis.
• Disconnection procedures (separation of epileptogenic
cortex from rest of brain):
– Multiple subpial transections (usually in areas of eloquent 99 100
cortex): intractable partial seizures emanating from
unresectable foci in primary cortices.
– Corpus callosotomy (the anterior two-thirds is sectioned
initially, and the posterior one-third about 6–12 months
later): usually for drop attacks.

Outcome
• Seizure-freedom:
– Anterior temporal lobectomy and amygdalo-
hippocampectomy: >80% of patients. If concordant
video-EEG and MRI brain.
– Lesionectomy: 30–40% of patients (poorer localization
of a single focus, and obvious pathology is less common).
– Hemispherectomy: 75–85% of patients.
– Corpus callosotomy: 30–40% of patients; best response
with atonic and tonic seizures.
• Other favorable outcomes include improved physical
functioning, psychosocial behavior and learning.

PROGNOSIS

Risk of a second seizure after a first seizure


• 31–71%, depending on other risk factors:
Interictal Ictal
– EEG: epileptiform discharges: 80% risk; non-specific
abnormalities: 40% risk; normal: 12% risk. 99, 100 Positron emission tomography (PET) scan of metabolism
of 18-fluorodeoxyglucose (fludeoxyglucose F 18) (a physiologic
Risk of seizure recurrence after >2 years remission radionuclide) that is injected intravenously and its uptake and
About 30% of children with epilepsy who are free of seizures distribution is measured and imaged.The kinetics of brain uptake of
for 2 years or longer will experience a recurrence of seizures 18-FDG reflect the kinetics of glucose brain uptake and therefore
when their medications are withdrawn and about 70% will provide an index of brain metabolism. Interictally (99) there is
remain free of seizures. As epilepsy is a heterogeneous entity evidence of diminished glucose metabolism in the region of the
(like TIA and stroke, see pp.186, 192), the chance of epileptic focus in the left fronto-temporal lobes (a blue or green or
recurrence varies among individual patients and depends on yellow ‘cold spot’) but during the ictus (100) there is increased
metabolism (a white or red ‘hot spot’)
82 Epilepsy

Prediction equation for recurrence of seizures STATUS EPILEPTICUS (SE)


Increased risk
Age >16 years at onset of seizures add 45
Taking >1 anti-epileptic drug add 50 DEFINITION
Seizures occurred after starting anti-epileptic A continuous seizure lasting at least 30 minutes, or two or
drug treatment add 35 more discrete seizures without interictal recovery (i.e.
History of primary or secondarily generalized between which the patient does not recover consciousness)
tonic-clonic seizures add 35 and lasting more that 30 minutes. SE is a medical
History of myoclonic seizures add 50 emergency.
Abnormal EEG in past year add 20
CLASSIFICATION
Decreased risk Convulsive
Increasing time (number of years, t) without seizures add 200/t • Generalized tonic-clonic status epilepticus (SE).
• Partial SE (e.g. somatomotor [epilepsia partialis
subtract 175 continua], somatosensory, aphasic).

Total score T Non-convulsive


Divide total score (T) by 100 and exponentiate z = eT/100 • Generalized SE:
– Absence SE (other names: petit mal SE, epilepsia minor
Probability of recurrence of seizures: continua, spike wave stupor, prolonged petit mal
• Continued treatment automatisms).
By 1 year 1–0.89z – Atypical absence SE.
By 2 years 1–0.79z • Partial SE (confusional state, coma.)
• Slow withdrawal
By 1 year 1–0.69z EPIDEMIOLOGY
By 2 years 1–0.60z • Incidence: 15–30 per 100 000 per year.
• Age: any age.
(Medical Research Council Anti-epileptic Drug Withdrawal Study
Group [1993] Prognostic index for recurrence of seizures after ETIOLOGY
remission of epilepsy. BMJ, 306: 1374–1378.) Epilepsy (two-thirds of convulsive SE, the vast
majority of non-convulsive SE)
Mortality • Poor anti-epileptic drug compliance.
The standardized mortality ratio for epilepsy is high. About • Recent reduction in dose or discontinuation of anti-
25% of deaths may be related directly to seizures: status epileptic drug.
epilepticus, accidental injury and sudden unexplained death • Drug (alcohol, phenobarbitone [phenobarbital]) withdrawal.
in epilepsy (SUDEP).
Symptomatic (one-third of convulsive SE have no
Risk factors in children history of epilepsy)
• Onset of epilepsy in the first 12 months of life. • Traumatic brain injury.
• Severe developmental delay present at the onset of • Stroke: the most frequent cause of simple partial status.
epilepsy. • Hypoxic encephalopathy.
• Symptomatic epilepsy (e.g. brain malformation). • Fever/infection.
• Severe myoclonic epilepsies of infancy and early • Meningoencephalitis.
childhood. • Brain abscess.
• Infantile spasms (deaths mainly due to the treatment of • Brain tumor.
spasms – with steroids and ACTH). • Metabolic disturbance: hyponatremia, uremia, hypogly-
cemia, hepatic encephalopathy, hypocalcemia, hypomag-
Risk factors in adults nesemia, mitochondrial encephalopathy.
• Young age. • Toxins.
• Male sex. • Drug overdose: tricyclic antidepressant, phenothiazine,
• Generalized convulsive seizures. theophylline, cocaine, isoniazid, chemotherapy drugs.
• Underlying structural brain lesions.
• Sleep. PATHOPHYSIOLOGY
• Non-compliance with anti-epileptic medication. Delayed control of SE is associated with prolonged epileptic
activity which may outstrip metabolic capacity and glucose
delivery, and lead to metabolic and hypoxic-ischemic brain
and systemic injury. The seizures compromise cerebral
vascular autoregulation, which in turn compromises
hypothalamic autonomic regulation, and raised intracranial
pressure may supervene.
Complications such as cardiovascular collapse, arrhythmias,
aspiration pneumonia, acute lung injury, and pulmonary
hypertension may compromise cerebral oxygen delivery
further. Metabolic derangement and cerebral and systemic
Status Epilepticus (SE) 83

acidosis with hyperpyrexia, rhabdomyolysis, and disseminated 101


intravascular coagulation may cause multiple organ failure.

CLINICAL FEATURES
The clinical presentation of SE can be categorized into:
• Prolonged seizures.
• Convulsive generalized status characterized by uncon-
sciousness, cyanosis and repeated generalized tonic-clonic
convulsive movements of the limbs with no interictal
recovery.
• Convulsive partial status characterized by repeated partial
seizures manifesting as focal motor convulsion or
neurologic deficits (e.g. somatosensory, visual, auditory)
not associated with altered consciousness or secondary
generalization. The most common forms are:
somatomotor simple partial SE (a succession of simple
partial seizures with Jacksonian march, without persistent
segmental myoclonus) and epilepsia partialis continua 102
(persistent myoclonus in a limited area of the body,
present for weeks or months and sometimes in
combination with somatomotor or tonic-clonic seizures).
• Non-convulsive SE presenting with continuous or
fluctuating clouding of consciousness, confusion,
automatisms, fluttering of the eyelids or other involuntary
orofacial dyskinetic movements and amnesia. Fluctuating
conscious level is more frequent in absence status whereas
focal neurologic signs point to complex partial SE.

History from a witness, examination of the patient, and


investigations must proceed concurrently with instituting
appropriate treatment.

DIFFERENTIAL DIAGNOSIS
Pseudostatus epilepticus should be suspected if there is aberrant
motor activity, an on/off pattern of seizure activity, poor
response to treatment and lack of metabolic consequences.

INVESTIGATIONS
• Electroencephalogram (EEG) (101–103).
• Full blood count and ESR.
• Urea and electrolytes.
• Plasma glucose.
• Liver function tests.
• Plasma calcium, phosphate and magnesium.
• Anti-epileptic drug levels.
• Toxicology screen.
• Arterial blood gases. 103
• EEG.
• CT brain scan: exclude structural intracranial lesion.
• Lumbar puncture: if CNS infection possible.
• Serum lactate: may be useful to help exclude pseudo-
seizures.

101, 102 EEGs (101, 8 channel; 102, 16 channel) of two young


adolescents each in a state of prolonged clouding of consciousness
due to non-convulsive generalized absence status epilepticus.

103 EEG (21 channel) showing generalized non-convulsive status


epilepticus on the left hand side of the recording which is terminated in
the middle of the recording by the i.v. injection of clonazepam 1 mg.
84 Epilepsy

DIAGNOSIS • Correct metabolic abnormalities, such as hypoxia and


• Repetitive epileptic seizures without interictal recovery hypoglycemia, and treat the underlying cause if known:
and epileptiform activity on the EEG. i.e. dexamethasone 16 mg if a brain tumor, arteritis or
• Diagnosis may require EEG monitoring because some parasitic brain infection, and thiamine 100 mg if the
patients have seizure discharge without detectable motor patient is alcoholic. It is not usually necessary, and may
activity and may be difficult to diagnose. be harmful, to give bicarbonate to correct acidosis.
• If the EEG is normal without evidence of even interictal
slowing of the background rhythm then the diagnosis Early SE
should be reconsidered. • Decide promptly whether to use a long term parenteral
• If the diagnosis is in doubt (e.g. ?pseudostatus epilepti- anti-epileptic drug, the aim of which is to prevent seizure
cus), observe one tonic-clonic attack and assess the level recurrence. Most patients are treated with phenytoin,
of consciousness post-ictally. If further doubt, attempt to and some are now treated with fosphenytoin.
capture at least one episode whilst monitoring the patient • Intravenous phenytoin loading dose of 18 mg/kg in
and the EEG. 100–150 ml of 0.9% normal saline given no faster than
50 mg/min by infusion pump, and while monitoring the
TREATMENT pulse, blood pressure, respirations and cardiac rhythm with
All forms of convulsive SE are neurologic emergencies that EKG.
must be treated promptly and aggressively to prevent – Brain concentrations of phenytoin peak at 10 minutes and
adverse sequelae. In non-convulsive status, other medical are three to four times those in plasma after injection.
factors such as the age and respiratory status may need to – If the patient is currently prescribed phenytoin, blood
be taken into consideration. The aims are to stop the levels are usually low due to poor compliance.
seizures as soon as possible and identify and treat the cause. – Never give phenytoin intramuscularly because phenytoin
has a pH of 12.
Immediate treatment for premonitory SE – Phenytoin, when coadministered with diazepam, will
• Ensure protected airway, remove false teeth if present. abolish at least 80% of episodes of convulsive SE.
• Oxygen 10 l/min via high-flow mask. Be prepared for a – Systemic and local reactions to i.v. phenytoin are
rapid sequence induction and endotracheal intubation common. Respiratory and CNS depression, and cardiac
because all anti-epileptic drugs except phenytoin can arrhythmias can be life threatening. Thrombophlebitis
cause neurologic depression and depress respiratory drive necessitates frequent changes of cannulas and makes
even in the face of metabolic acidosis. central administration the preferred route.
• Insert an i.v. line and take 30 ml venous blood for urea • Fosphenytoin is a disodium phosphate ester of phenytoin,
and electrolytes, glucose, liver function tests, calcium, which has been licensed recently because it offers several
magnesium, full blood count, anti-epileptic drug advantages over phenytoin (which has to be given i.v. and
concentrations and toxicology screen. major adverse effects are common). Fosphenytoin is freely
• Administer a benzodiazepine, either: soluble in aqueous solutions and can be administered i.v. or
– i.v. diazepam 2 mg boluses per minute up to 20 mg intramuscularly. Intravenous fosphenytoin is tolerated at
(10 mg if older than 60 years of age or hypotensive), or infusion rates up to three times faster than those for
– i.v. clonazepam 0.2 mg boluses per minute up to 1 mg, or phenytoin, therapeutic levels are established within
– i.v. midazolam up to 5mg, or 10 minutes, it is rapidly metabolized (conversion half-life of
– i.v. lorazepam over 1–2 minutes (or rectal if no venous 8–15 minutes) into phenytoin by endogenous phosphatases
access), or in the body, and it is as effective as phenytoin in treating SE.
– intramuscular midazolam if difficulties with venous access. Intramuscular administration of fosphenytoin is characterized
by rapid and complete absorption, no requirement for
Lorazepam is often preferred as it has a long duration of cardiac monitoring, and a low incidence of adverse effects
anti-epileptic effect and the best parenchymal distribution. which are similar to those of parenteral phenytoin
Any of these measures controls the status in about 80% of (nystagmus, dizziness pruritus, paresthesias, headache,
cases but may take minutes to work. In the 20% of cases who somnolence, and ataxia). It has the promise to be suitable for
do not respond, this regime can be repeated in 5–10 minutes administration in the ambulance because it causes few local
time or another benzodiazepine can be given. Beware of adverse effects (e.g. pain, burning and itching at the injection
adverse events which include respiratory and CNS depression site) and does not cause respiratory and CNS depression.
causing respiratory arrest, hypotension and impaired
consciousness. N.B. Benzodiazepines are not a substitute for N.B. Avoid neuromuscular blockade initially because it
a long term parenteral anti-epileptic drug (see below). masks ongoing ictal activity.

• Dextrostix (glucose test): if low dextrostix reading or Absence status


known diabetic, give glucose 50 ml of 50% i.v., and • Clonazepam i.v. (see above).
thiamine 100 mg i.v. • Valproic acid: via nasogastric or rectal route.
• Arterial puncture (usually femoral artery) for arterial
blood gases. Resistant SE
• Reconsider the diagnosis (particularly if you are saying to
yourself ‘I have never seen anything like this before’: con-
sider pseudostatus and seek EEG confirmation, if possible).
• Review the patient’s ventilatory status and consider
intubation.
Status Epilepticus (SE) 85

• Intravenous phenobarbitone (phenobarbital) • Continue maintenance doses of phenytoin and


15–20 mg/kg, no faster than 100 mg/min (preferably phenobarbital (phenobarbital), and follow levels to
50 mg/min), or until seizures stop (e.g. additional determine optimal doses.
10 mg/kg doses even up to 100 mg/kg; pheno- • Use i.v. fluids and dopamine (up to 10 μg/kg/min) to
barbitone is a very effective drug and is generally better treat hypotension. Decrease the dosage of thiopentone
to pursue than chopping and changing with the (thiopental) or midazolam if any signs of cardiovascular
alternatives below). Monitor for respiratory depression compromise.
and hypotension, or • Discontinue the midazolam infusion at 12 hours while
• Benzodiazepine infusion (e.g. clonazepam 3 mg in monitoring the EEG and observe for further clinical or
500 ml 5% dextrose at 20 ml/hour and titrate). Not a electrographic seizure activity. If seizures recur, reinstate
substitute for phenytoin. Monitor for respiratory the infusion and repeat this step at 12–24 hour intervals
depression, or longer if the patient’s seizures remain refractory.
Or
• Intravenous chlormethiazole 0.8% 40–100 ml over N.B. Although barbiturate coma is extremely effective in
10 minutes. terminating refractory generalized SE, it frequently causes
If seizures stop, continue the infusion at 0.5 to hemodynamic instability due to myocardial depression and
1.0 ml/min and monitor respiration. vasodilatation (and thus requires invasive hemodynamic
Or, monitoring and the use of pressor agents) and patients may
• Intramuscular paraldehyde 5–10 ml, or dilute 10 ml take days to wake up after it has ceased and so they require
paraldehyde in 100 ml 0.9% normal saline and infuse prolonged intubation. Continuous i.v. infusion of
over 10–15 minutes or until seizures stop. midazolam can be as effective and less toxic. Midazolam is
• However, in Australia chlormethiazole is no longer used, a water-soluble benzodiazepine with rapid CNS penetration
and paraldehyde is no longer available. and a short elimination half-life of 1.5–3.5 hours.
• For patients in partial status epilepticus, despite
phenytoin and clonazepam, consider administering Maintenance treatment
vigabatrin, which is very water-soluble, via nasogastric When seizures have been abolished, an appropriate oral
tube in a dose of 1 g stat, which is repeated 8–12 hours regimen is required:
later and then continued 8–12 hourly.
• Regular anti-epileptic medication should be recom-
Refractory SE menced.
Seizure activity for about an hour in which the patient has • For previously untreated patients, continue with
not responded to therapy. phenytoin. An oral loading dose of 300 mg should be
given 6–12 hours after the initial i.v. injection. Thereafter
• Reconsider the diagnosis. the dose can be altered according to clinical response,
• Transfer to an intensive care unit. adverse effects and serum concentrations.
• Monitor vital signs closely and treat hyperthermia. • If the seizure type can be characterized, or if the patient
• Initiate therapy for the underlying etiology as soon as is a young woman, preference may be given to
possible. commencing an oral regimen of carbamazepine or
• Further doses of phenobarbitone (phenobarbital) or a sodium valproate. If the patient is unconscious or cannot
continuous infusion of the older anesthetic agent swallow, both can be given via a nasogastric tube.
thiopentone (thiopental) or the newer agent propofol
will usually control the seizures. (N.B. it is unwise to PROGNOSIS
paralyse the patient, because clinical signs will be lost, Convulsive SE
unless continuous EEG activity is being monitored.) • High mortality (up to 10–12%): more often due to the
• Monitor EEG. underlying cause than direct neurologic damage caused
• If the EEG suggests ongoing seizures or if the patient by the status.
has overt convulsions, and the patient has received full • Morbidity: determined by the underlying cause and
loading doses of phenytoin and phenobarbital (pheno- duration of SE. The longer the duration, the more
barbital), an i.v. infusion of midazolam or clonazepam refractory it becomes to treatment and the higher the
may help. Midazolam has the advantage of a shorter half- morbidity and mortality.
life and so the patient wakes up sooner after the infusion
has ceased. Midazolam is given i.v. with a loading dose Non-convulsive SE
of 200 μg/kg as a slow i.v. bolus, followed by • Generalized absence SE: good outcome if treated
0.75–10 μg/kg/min continuous infusion. Higher doses appropriately.
may be necessary, especially with prolonged infusion, due • Complex partial SE: prognosis depends on the
to possible tachyphylaxis. Adjust the maintenance dose underlying cause and the quality of treatment.
to stop electrographic seizures based on EEG
monitoring. EEG should be recorded continuously for
the first 1–2 hours during and after the initiation of
midazolam loading and infusion, and then monitored
either continuously or for 30–60 minute intervals every
2 hours during the maintenance phase. The primary aim
of therapy is suppression of electrographic seizures.
86 Epilepsy

JUVENILE MYOCLONIC EPILEPSY INVESTIGATIONS


(JME) (JANZ SYNDROME) EEG
Ictal
Rapid (3.5–6 Hz) irregular, generalized spike and polyspike
DEFINITION wave discharge
A clinically distinct form of primary idiopathic generalized
epilepsy characterized by bilateral myoclonic jerks Interictal
experienced with full consciousness. In addition to the jerks, Normal or brief discharges of generalized, symmetric or
most patients also have generalized tonic-clonic seizures, multiple spikes and slow waves at frequencies from
and about one-third also have absence seizures. 3.5–6 Hz in about 95% of untreated patients and about 30%
of patients being treated with valproate, resembling ictal
EPIDEMIOLOGY trace (104, 105). Photosensitivity is common (70–80% of
• Incidence: the most common epilepsy syndrome in cases); a photoconvulsive response is seen in about 33–40%
adolescence. Accounts for 5–10% of all patients with of patients and is not necessarily associated with
epilepsy. photosensitive seizures. Focal EEG abnormalities, either
• Age of onset: 8–18 years (may be later). independent or associated with the generalized paroxysms,
• Gender: M=F. may be seen.

ETIOLOGY AND PATHOPHYSIOLOGY Brain imaging


• Idiopathic, but almost exclusively genetic. CT and MRI brain scan are normal if performed, but are
• Mode of inheritance is complex. Several modes of not necessary in the presence of the above clinical and EEG
inheritance have been proposed: autosomal recessive, features.
polygenic, two loci recessive dominant or recessive
recessive, and single locus dominant. DIFFERENTIAL DIAGNOSIS
• Involvement of more than one gene is implicated by • Familial adult myoclonic epilepsy (FAME): an autosomal
segregation analysis. Linkage of JME to chromosome dominant primary generalized epilepsy, characterized by
15 q has been proposed, as has a locus near the human adult-onset (mean 37 years) of myoclonic jerks in the
leukocyte antigen (HLA) region of the short arm of upper and lower extremities, tremulous finger move-
chromosome 6p. A recent study found no linkage to ments, and rare generalized tonic-clonic seizures. Myo-
chromosome 15 but positive linkage to chromosome 6, clonic episodes can be precipitated by fatigue, insomnia
but it may be that JME is linked to different genes in and photic stimuli. The EEG is similar to JME. The
different ethnic groups. FAME gene has been localized to chromosome 8q24.
FAME is similar to JME in that it is probably a primary
CLINICAL FEATURES disorder of membrane excitability with a benign (non-
• Myoclonic jerks, typical absences and generalized tonic- progressive) course, but is distinguished by an older age
clonic seizures occur in a characteristic age-related of onset (in adulthood), whereas JME has onset in early
sequence. adolescence and myoclonic jerks predominantly on
• Myoclonic jerks, which follow a circadian pattern and awakening.
occur when drowsy or shortly after awakening, are • Progressive myoclonic epilepsy: seizures are more likely
bilateral, single or multiple, and affect the arms secondary to neurodegeneration rather than being a
predominantly. They result in the patient dropping and primary disorder of neuronal excitability. Patients quickly
spilling things first thing in the morning (i.e. at the develop dementia and ataxia, and usually live only 10–20
breakfast table) and settle down over an hour or two. years after diagnosis.
Frequently, they are not recognized by the patient as • ‘Epilepsy with generalized tonic-clonic seizures on
being of any significance and are not reported. awakening’: usually commences in the second decade of
• Juvenile-type absences occur in 10–30% of cases. life. Commonly it may be precipitated by sleep depriva-
• Generalized tonic-clonic seizures occur in about 90% of tion, photosensitivity, and is associated with a positive
patients, usually in the morning. They are almost family history, but there is no myoclonic component.
invariably preceded by a flurry of myoclonus, building
up to a crescendo. DIAGNOSIS
• Seizures are commonly provoked by factors such as sleep • History of myoclonus on awakening, usually early in the
deprivation, alcohol ingestion, and, to a lesser extent, morning; all older children, adolescents and young adults
menstruation and photic stimuli such as flickering lights. who present with a tonic-clonic seizure should be asked
• A family history of epilepsy in close relatives is not about early morning myoclonus because they may not
uncommon. The clinical picture in affected family volunteer this information.
members from the same pedigree is variable and includes • Seizures that occur mainly in the morning.
different syndromes, such as childhood absence epilepsy, • Typical EEG.
juvenile absence epilepsy, or epilepsy with generalized
tonic-clonic seizures on awakening. Asymptomatic family TREATMENT
members may have abnormal EEGs showing typical The aims of treatment are to control absences and GTCS
diffuse polyspike wave or spike wave complexes similar and minimize myoclonus on awakening. Medical manage-
to the interictal discharges seen in affected members. ment is often very successful in achieving these goals.
Progressive Myoclonic Epilepsies (PME) 87

Anti-epileptic drugs PROGRESSIVE MYOCLONIC


• Sodium valproate is the drug of choice because it EPILEPSIES (PME)
achieves complete control in more than 80% of patients.
• Clonazepam is an effective alternative, but is less
accepted by patients because it may have adverse effects DEFINITION
on cognition, particularly with long term use. A heterogeneous group of rare genetic disorders, most of
• Lamotrigine seems to be effective, both as add-on which are familial (autosomal recessive inheritance),
therapy and monotherapy. On occasions, the adverse characterized by onset in childhood or adolescence of
effects of valproate (weight gain, tremor and hair loss) generalized or fragmentary myoclonic seizures, often other
may be such that lamotrigine can be considered as a epileptic seizures, and progressive neurologic deterioration,
suitable alternative. especially cerebellar ataxia and dementia.
• Other anti-epileptic drugs may control the GTCS but
not the myoclonus. Indeed, carbamazepine and EPIDEMIOLOGY
vigabatrin may make it worse. • Incidence: rare.
• Age of onset: childhood or adolescence.
Lifestyle • Gender: M=F.
Lifestyle issues need to be discussed in detail with the
patient and the family, such as avoiding seizure-precipitating
factors like late nights (sleep deprivation), alcohol, and
flickering lights in nightclubs, particularly at the time of
menstruation.

Pregnancy
Due to the early age of onset and the lifelong nature of
JME, many female patients are likely to contemplate
pregnancy while taking the necessary anti-epileptic drug
therapy. If the patient is a sexually active female, the risks of
epilepsy and anti-epileptic drugs associated with pregnancy
need to be carefully discussed. Sodium valproate has a 1–2%
risk of severe spina bifida. The teratogenicity of lamotrigine
is unknown. If pregnancy is a possibility, sodium valproate 105
should be supplemented with prophylactic folic acid (0.5–5
mg daily) for at least 1 month before conception and for the
first 3 months of pregnancy. In some patients with provoked
seizures, it may be possible to cease anti-epileptic medication
before conception and for the first 3 or 4 months of preg-
nancy, if the risk of seizure recurrence and its complications
(including sudden death) are thought to be very low.

PROGNOSIS
A life-long condition for most patients but excellent response
to valproate. Withdrawal of anti-epileptic medication is
associated with a high relapse rate (in 80% of patients).

104

104, 105 Interictal EEGs showing the typical 4–6 Hz bilaterally


symmetric generalized polyspike and wave pattern in an adolescent
with juvenile myoclonic epilepsy.
88 Epilepsy

ETIOLOGY ized (including myoclonic) seizures, and a fixed neurologic


Disorders in which the clinical presentation is deficit with or without slow deterioration.
typically PME • Inherited metabolic disorders (see p.157).
• Unverricht–Lundborg disease (previously termed ‘Baltic • Tuberous sclerosis (see p.143).
myoclonus’): caused by mutations in the human EPM1 • Birth injury.
gene, which encodes cystatin B, an intracellular protein • Encephalitis (see p.273).
that competitively inhibits intralysosomal cystein
proteases known as cathepsins. The function of cystatin The PMEs can usually be distinguished from these
B may be to prevent intracellular damage that could disorders by evidence of normal development until the onset
otherwise occur after ‘leakage’ of cysteine proteases from of the disease, and a relatively rapid decline thereafter.
lysosomes into the cytoplasm.
• Lafora body disease (106): caused by a mutation in one Progressive myoclonic encephalopathies
of the four exons of the EPM2A gene or non-coding Progressive brain disorders characterized by myoclonus with
regions of EPM2A. The EPM2A gene codes for laforin, or without generalized (including myoclonic) seizures:
which is a functional protein tyrosine phosphatase. • Inborn errors of metabolism:
• Sialidosis. – Gaucher’s disease.
– Hexosaminidase deficiency (see p.172).
Disorders in which the clinical presentation is • Infections:
occasionally PME – Subacute sclerosing panencephalitis (see p.304).
• Neuronal ceroid lipofuscinosis. – Creutzfeldt–Jakob disease (see p.330).
• Mitochondrial cytopathies. – Post-encephalitic parkinsonism (see p.420).

Uncommon causes of PME Progressive myoclonic ataxias


• Huntington’s disease. A syndrome comprising myoclonus and progressive
• Hallervorden–Spatz disease. cerebellar ataxia with infrequent or absent epileptic seizures
• Neuroaxonal dystrophy. and little or no cognitive dysfunction. Causes include:
• Wilson’s disease. • Mitochondrial disease.
• Gaucher’s disease (non-infantile neuronopathic form). • Spinocerebellar degenerations.
• GM2 gangliosidosis (late infantile, juvenile forms). • Celiac disease (possibly).
• Biotin-responsive encephalopathy.
INVESTIGATIONS
CLINICAL FEATURES • Full blood count:
• Myoclonic seizures (generalized or fragmentary): sudden, – Lymphocyte vacuolation in sialidosis and certain cases of
brief, lightning-like jerks that may be generalized or neuronal ceroid lipofuscinosis.
limited to one or more muscle groups. They are not – Pancytopenia is common in Gaucher’s disease.
associated with loss of consciousness and are frequently • Blood and CSF lactate: raised in mitochondrial cytopathies.
precipitated by stimuli such as movement, bright light,
or stress.
• Epileptic seizures of other types.
• Progressive neurologic decline: particularly cerebellar
ataxia and cognitive decline (dementia). 106
DIFFERENTIAL DIAGNOSIS
Non-epileptic myoclonus (see Myoclonus, p.124)

Myoclonic epilepsies
Epileptic seizures accompanied by myoclonus:
• Infantile spasms.
• Lennox–Gastaut syndrome.
• Juvenile myoclonic epilepsy (see p.86).

Other, more common, generalized epilepsies


Early in the course of PME the clinical triad of myoclonic
seizures, tonic-clonic seizures and progressive neurologic
decline may not be present and PME can be mistaken for
more benign forms of epilepsy before progressive neurologic
signs or intractable seizures develop:
• Typical absence epilepsy.
• Primary generalized epilepsy combined with anti-
epileptic drug toxicity. 106 Light microscopic examination of the brain of a 20 year old
patient with Lafora body disease, who died after a 6 year history of
Other static or progressive secondary generalized focal occipital seizures and relentless decline in cognitive function,
epileptic encephalopathies showing the presence of a characteristic periodic acid–Schiff (PAS)-
Brain disorders characterized by various forms of general- positive inclusion (Lafora body).
Progressive Myoclonic Epilepsies (PME) 89

• Blood mitochondrial DNA analysis: mutations in some Type II


cases of mitochondrial cytopathy. • Age of onset: 10–30 years.
• Serum tartrate-resistant isoenzymes of acid phosphatase: • Clinical features: cherry red macular spot; dysmorphic
elevated in Gaucher’s disease. features.
• Serum and leukocyte β-hexosaminidase A and B: GM2 • Investigations: storage material in lymphocytes and other
gangliosidosis. cells; excess urine sialic acid-rich oligosaccharides.
• Urinary thin-layer chromatographic oligosaccharide • Diagnosis: severe deficiency of α-N-acetylneuraminidase
screen: sialidosis. in leukocytes; and partial β-galactosidase deficiency in
• Urinary sediment dolichol estimation: neuronal ceroid most cases.
lipofuscinosis.
• Urinary organic acid estimation: biotin-responsive Neuronal ceroid lipofuscinosis
encephalopathy. Late infantile (Jansky–Bielschowsky disease)
• Electroencephalograph: • Age of onset: 2–4 years.
– Typically shows a slowing of background rhythms with • Clinical features: severe seizures, rapid regression, fundal
generalized epileptiform discharges and photic sensitivity. changes of attenuated retinal vessels and macular degeneration.
– More specific findings include occipital discharges in • Investigations: ERG: absent B wave; VEP: enlarged.
Lafora–body disease and photosensitive spikes on low- • Diagnosis: curvilinear profiles on EM of skin, muscle,
frequency stimulation in late infantile neuronal ceroid rectum or brain biopsy; elevated urinary sediment
lipofuscinosis and in certain adult cases. dolichol in 92% of cases.
• Visual evoked potentials (VEP): may be enlarged or
diminished in neuronal ceroid lipofuscinosis. Juvenile (Spielmeyer–Vogt disease)
• Electroretinogram (ERG): absent B wave in neuronal • Age of onset: 4–10 years.
ceroid lipofuscinosis. • Clinical features: visual failure, fundal changes of optic
• Somatosensory evoked potentials: enlarged in many atrophy, attenuated retinal vessels and macular
forms of PME. degeneration.
• Skin + muscle biopsy: • Investigations: ERG: absent B wave; VEP: diminished.
– Eccrine sweat gland duct cells stained for polysaccharide • Diagnosis: fingerprint profiles on EM of skin, muscle,
(Lafora–body disease). rectum or brain biopsy; elevated urinary sediment
– Acid phosphatase stain and electronmicroscopy (EM) dolichol in 85% of cases.
(neuronal ceroid lipofuscinosis).
– Modified Gomori’s trichrome and oxidative enzyme Adult (Kuf’s disease)
reactions for ragged-red fibers (mitochondrial cytopathy). • Age of onset: 12–50 years.
• Clinical features: progressive dementia, myoclonic
DIAGNOSIS epilepsy, spasticity, ataxia, normal optic fundi.
Unverricht–Lundborg disease (previously termed • Investigations: VEP: marked photosensitivity at a low
‘Baltic myoclonus’) frequency stimulation in some cases.
• Age of onset: 8–13 years. • Diagnosis: curvilinear profiles on EM of skin, muscle,
• Pathology: widespread neuronal degeneration in the brain. rectum or brain biopsy; elevated urinary sediment
• Clinical features: severe myoclonus, little or no dementia. dolichol in some cases.
• Diagnosis: clinical and genetic (i.e. DNA analysis:
mutation in the EPM1 gene). Mitochondrial cytopathies (see p.162)
• Age of onset: any age.
Lafora body disease • Clinical features: short stature, optic atrophy, deafness,
• Age of onset: 11–18 years. muscle weakness due to myopathy, stroke-like episodes,
• Clinical features: partial occipital seizures, inexorable lactic acidosis, intrafamily variation in age of onset and
dementia. clinical severity, maternal inheritance.
• Investigations: EEG – focal or multifocal posterior • Investigations: elevated blood or CSF lactate levels;
epileptiform discharges, generalized discharges, and mitochondrial DNA mutations.
progressive slowing of background rhythm. • Diagnosis: mitochondrial DNA mutation, ‘ragged-red’
• Diagnosis: Lafora bodies (periodic acid–Schiff [PAS]- fibers on skeletal muscle biopsy.
positive inclusions polyglucosan bodies]) on light
microscopy of skin, muscle, liver or brain biopsy (106). TREATMENT
DNA analysis: mutation in one of the four exons of the Symptomatic relief of myoclonus and seizures
EPM2A gene or non-coding regions of EPM2A. • Valproic acid.
• Clonazepam.
Sialidosis • 5-Hydroxytryptophan with carbidopa.
Type I • Piracetam as an add-on agent.
• Age of onset: 8–20 years.
• Clinical features: severe myoclonus, gradual visual failure N.B. Patients with the PMEs are particularly prone to
and cherry red macular spot; absence of dementia. neurotoxic side effects, especially with phenytoin (which
• Investigations: storage material in lymphocytes and other should not be used) and the uncritical use of multiple
cells; excess urine sialic acid-rich oligosaccharides. anticonvulsants.
• Diagnosis: severe deficiency of α-N-acetylneuraminidase
in leukocytes.
90 Epilepsy

Specific therapy Genetic counselling


• Mitochondrial encephalomyopathies (see p.162).
• Neuronal ceroid lipofuscinosis: ?antioxidants. PROGNOSIS
• Biotin-responsive encephalopathy. The natural history of many of these disorders is not known
• Gaucher’s disease: bone marrow transplantation. but is usually one of progressive mental and physical
disability.

Kwan P, Brodie MJ (2000) Early identification of STATUS EPILEPTICUS


FURTHER READING refractory epilepsy. N. Engl. J. Med., 342: Allredge BK, Gelb AM, Isaacs M, et al. (2001) A
314–319. comparison of lorazepam, diazepam, and
Kwan P, Brodie MJ (2001) Neuropsychological placebo for the treatment of out-of-hospital
EPILEPSY effects of epilepsy and antiepileptic drugs. status epilepticus. N. Engl. J. Med., 345:
Alovi M, Rogawski MA, Avanzini G (2001) Gen- Lancet, 357: 216–222. 631–637.
eralised epileptic disorders: An update. Epilep- Lhatoo SD, Langan Y, Sander JWAS (1999) Sud- Heafield MTE (2000) Managing status epilepti-
sia, 42: 445–457. den unexpected death in epilepsy. Postgrad. cus. BMJ, 320: 953–954.
Brodie MJ, French JA (2000) Management of Med. J., 75: 706–709. Jallon P, Coeytaux A, Galobardes B, Morabia A
epilepsy in adolescents and adults. Lancet, Lhatoo SD, Johnson AL, Goodridge DM, et al. (1999) Incidence and case-fatality rate of sta-
356: 323–329. (2001) Mortality in epilepsy in the first 11 to tus epilepticus in the Canton of Geneva.
Brown TR, Holmes GL (2001) Epilepsy. N. Engl. 14 years after diagnosis: multivariate analysis Lancet, 353: 1496.
J. Med., 344: 1145–1151. of a long-term, prospective, population-based Lowenstein DH, Alldredge BK (1998) Status
Chadwick D, for the Vigabatrin European cohort. Ann. Neruol., 49: 336–344. epilepticus. N. Engl. J. Med., 338: 970–976.
Monotherapy Study Group (1999) Safety and MacDonald BK, Cockerell OC, Sander JWAS, Treiman DM (2001) Therapy of status epilepticus
efficacy of vigabatrin and carbamazepine in Shorvon SD (2000) The incidence and life- in adults and children. Curr. Opin. Neurol.,
newly diagnosed epilepsy: a multicentre ran- time prevalence of neurological disorders in a 14: 203–210.
domised double-blind study. Lancet, 354: prospective community-based study in the
13–19. UK. Brain, 123: 665–676. JUVENILE MYOCLONIC EPILEPSY
Chadwick D (2000) Seizures and epilepsy after MacDonald BK, Johnson AL, Goodridge DM, et Biraben A, Allain H, Scarabin JM, et al. (2000)
traumatic brain injury. Lancet, 355: 334–336. al. (2000) Factors predicting prognosis of Exacerbation of juvenile myoclonic epilepsy
Dalby NO, Mody I (2001) The process of epilep- epilepsy after presentation with seizures. Ann. with lamotrigine. Neurology , 55: 1758.
togenesis: A pathophysiological approach. Neurol., 48: 833–841. Durner M, Hinnar S, Resor SR, et al. (2000) No
Curr. Opin. Neurol., 14: 187–192. Mani KS, Rangan G, Srinivas HV, et al. (2001) evidence for a major susceptibility locus for ju-
DeToledo JC, Lowe MR (2001) Seizures, lateral Epilepsy control with phenobarbital or pheny- venile myoclonic epilepsy on chromosome
decubitus, aspiration, and shoulder dislocation. toin in rural south India. The Yelandur Study. 15q. Am. J. Med. Genetics, 96: 49–52.
Time to change the guidelines? Neurology, 56: Lancet, 357: 1316–1320. Sander T, Bockenkamp B, Hildmann T, et al.
290–291. Marson AG, Chadwick DW (2001) New drug (1997) Refined mapping of the epilepsy sus-
Devinsky O (1999) Patients with refractory treatments for epilepsy. J. Neurol. Neurosurg. ceptibility locus EJM1 on chromosome 6.
seizures. N. Engl. J. Med., 340: 1565–1570. Psychiatry, 70: 143–148. Neurology, 49: 842–847.
Devinsky O, Cramer J (2000) Safety and efficacy Nilsson L, Farahmand BY, Persson P-G, et al. Serratosa JM, Delgado-Escueta AV, Medine MT,
of standard and new anti epileptic drugs. Neu- (1999) Risk factors for sudden unexpected et al. (1996) Clinical and genetic analysis of a
rology, 55 (Suppl 3): S5–S10. death in epilepsy: a case-control study. Lancet, large pedigree with juvenile myoclonic epilep-
D’Onofrio GD, Rathlev NK, Ulrich AS, et al. 353: 888–893. sy. Ann. Neurol., 39: 187–195.
(1999) Lorazepam for the prevention of re- Parra J, Augustijn PB, Geerts Y, Van Emde Boas
PROGRESSIVE MYOCLONIC EPILEPSIES
current seizures related to alcohol. N. Engl. J. W (2001) Classification of epileptic seizures:
Fedi M, Reutens D, Dubeau F, et al., (2001)
Med., 340: 915–919. A comparison of two systems. Epilepsia, 42:
Long-term efficacy and safety of piracetam in
Durner M, Keddache MA, Tomasini L, et al. 476–482.
the treatment of progressive myoclonic epilep-
(2001) Genome scan of idiopathic generalized Rosenow F, Lüders H (2001) Presurgical evalua-
sy. Arch. Neurol., 58: 781–786.
epilepsy: evidence for major susceptibility gene tion of epilepsy. Brain, 124: 1683–1700.
Marseille Consensus Group (1990) Classification
and modifying genes influencing seizure type. Sachdeo R, Beydoun A, Schachter S, et al. (2001)
of progressive myoclonic epilepsies and relat-
Ann. Neurol., 49: 328–335. Oxcarbazepine (Trileptal) as monotherapy in
ed disorders. Ann. Neurol., 28: 113–116.
Feely M (1999) Drug treatment of epilepsy. BMJ, patients with partial seizures. Neurology, 57:
Minassian BA, Ianzano L, Delgado-Escueta AV,
318: 106–109. 864–871.
Scherer SW (2000) Identification of new and
French JA, Gidal BE (2000) Antiepileptic drug in- Sawle GV, Ramsay MM (1998) The neurology of
common mutations in the EPM2A gene in
teractions. Epilepsia, 41 (Suppl 8), S30–S36. pregnancy. J. Neurol. Neurosurg. Psychiatry,
Lafora disease. Neurology, 54: 488–490.
Gross RA, Theodore WH (1999) ‘The seat of the 64: 711–725.
Minassian BA, Ianzano L, Meloche M, et al.
discharge’ and the molecules that govern Stefan H, Halász P, Gil-Nagel A, et al. (2001) Re-
(2000) Mutation spectrum and predicted
seizures. Neurology, 52: 441–443. cent advances in the diagnosis and treatment
function of laforin in Lafora’s progressive my-
Hart YM, Sander JW, Johnson AL, Shorvon SD of epilepsy. Eur. J. Neurol., 8: 519–539.
oclonic epilepsy. Neurology, 55: 341–346.
(1990) National General Practice study of Stephen LJ, Brodie MK (2000) Epilepsy in elder-
Minassian BA, Andrade DM, Iazano L, et al.
epilepsy: Recurrence after a first seizure. ly people. Lancet, 355: 1441–1446.
(2001) Laforin is a cell membrane and endo-
Lancet, 336: 1271–1274. Temkin NR (2001) Antiepileptogenesis and
plasmic reticulum-associated protein tyrosine
Holmes LB, Harvey EA, Coull BA, et al. (2001) seizure prevention trials with antiepileptic
phosphatase. Ann. Neurol., 49: 271–275.
The teratogenicity of anticonvulsant drugs. N. drugs: Meta-analysis of controlled trials.
Pennacchio LA, Lehesjoki AE, Stone NE, et al.
Engl. J. Med., 344: 1132–1138. Epilepsia, 42: 515–524.
(1996) Mutations in the gene encoding cys-
Johnson MR, Sander JWAS (2001) The clinical Theodore W, Wasterlain CG (1999) Do early
tatin B in progressive myoclonic epilepsy
impact of epilepsy genetics. J. Neurol. Neuro- seizures beget epilepsy? Neurology, 53:
(EPM1). Science, 271: 1731–1734.
surg. Psychiatry, 70: 428–430. 898–899.
Plaster NM, Uyama E, Uchino M, et al. (1999)
King MA, Newton MR, Jackson GD, et al. (1998) Toone BK (2000) The psychoses of epilepsy. J.
Genetic localization of the familial adult my-
Epileptology of the first-seizure presentation: a Neurol. Neurosurg. Psychiatry, 69: 1–4.
oclonic epilepsy (FAME) gene to chromosome
clinical electroencephalographic and magnetic Walczak TS, Leppik IE, D’Amelio M, et al.
8224. Neurology, 53: 1180–1183.
resonance imaging study of 300 consecutive pa- (2001) Incidence and risk factors in sudden
tients. Lancet, 352: 1007–1011. unexpected death in epilepsy. A prospective
Krauss GL, Krumholz A, Carter RC, et al. (1999) cohort study. Neurology, 56: 519–525.
Risk factors for seizure-related motor vehicle Wiebe S, Blume WT, Girvin JP, et al. (2001) A
crashes in patients with epilepsy. Neurology, randomized, controlled trial of surgery for
52: 1324–1329. temporal-lobe epilepsy. N. Engl. J. Med., 345:
311–318.
Chapter Four 91

Headache
HEADACHE ETIOLOGY (see Table 12)

CLINICAL HISTORY
CLINICAL CLASSIFICATION AND PREVALENCE OF • Types of headache: how many types of headache do you
DIFFERENT TYPES (see Table 11) suffer from?
• Length of history of headache: is this a new headache or
not; has it changed in character, severity or frequency?
• Features of the headache itself:
Table 11 Clinical classification and prevalence of – Location/site of headache.
different types of headache in the general population (%) – Onset.
– Type/quality of headache (throbbing, steady).
Primary % – Timing.
Muscle contraction/tension-type headache 69 – Severity.
Idiopathic stabbing headache 33 – Radiation.
(ice cream/ice pick headache) – Associated features (e.g. nausea, photophobia,
Migraine 16 phonophobia, symptoms of aura, fever, neurologic
Exertional headache 1 symptoms such as weakness, diplopia, clumsiness,
Cluster headache 0.1 disturbance of balance, altered cognitive function; altered
consciousness).
Secondary % – Exacerbating factors (e.g. physical activity, bright light,
Systemic infection 63 noise).
Head injury 4 – Relieving factors.
Drug induced headache 3 – Duration of headache.
Vascular disorders 1 – Predisposing factors.
Subarachnoid hemorrhage <1 – Premonitory features.
Brain tumor 0.1 • Family history.
• Medications/drugs.

Table 12 Etiology of headache


Children Adults Elderly
(3–16 years) (17–65 years) (65+ years)
Tension headache ++ +++ +
Migraine ++ +++ +
Idiopathic stabbing headache + + +
Exertional headache + ++ –
Cluster headache + ++ +
Post-traumatic ++ +++ +
Drug-induced headache – ++ ++
Cervicogenic (referred from neck) – + +++
Cranial arteritis – + +++
Subarachnoid hemorrhage + ++ +
Subdural hematoma +/– ++ ++
Brain abscess + + +
Brain tumor + ++ ++
Idiopathic intracranial hypertension +/– ++ –
Glaucoma – + ++
Paget’s disease of the skull – + ++
Cerebral venous sinus thrombosis +/– + +
Arnold–Chiari malformation +/– + +/–
92 Headache

PHYSICAL EXAMINATION • Neck stiffness.


• Fever or other signs of systemic illness (e.g. temporal • Upper cervical spine facet joints.
artery tenderness [107, 108]). • Papilledema (109).
• Conscious state. • Subhyaloid heamorrhage (110).
• Eye movements.
• Facial weakness.
107 • Limb weakness or incoordination.
• Gait disturbance.

Table 13 Differential diagnosis of primary headaches

Muscle Migraine Cluster


contraction/ headache
107 Lateral photograph of the forehead of a patient with giant cell tension headache
arteritis showing a visibly enlarged, tender, temporal artery (arrows).
Gender Male = female Female Male
predominance predominance
(9:1)
108
Pain
• Location Bilateral 2/3 hemicranial Orbit or temple
Orbit or frontal
• Type Steady tightness Throbbing Boring
• Severity Moderate Severe Very severe
• Duration Chronic, Hours to days 15–180 minutes,
usually daily often nocturnal

Aura None Often None

Nausea/vomiting None Often Uncommon

Photophobia, None Often Uncommon


phonophobia

Ipsilateral None Rare Typical


autonomic
features
108 Temporal artery in cross-section showing features of active giant
cell arteritis with fibroblastic proliferation in the intima (on the left of Periodicity Episodic or Episodic Clusters
the photograph), fibrinoid necrosis in the subintima, disruption of the chronic, (weeks to
internal elastic lamina, histiocytic proliferation, multinucleated giant cells, usually daily months)
1–8 attacks/day
and infiltration of adventitia by lymphocytes and plasma cells.

109 110

109 Ocular fundus showing papilledema in a patient with idiopathic 110 Ocular fundus showing subhyaloid hemorrhage in a patient with
intracranial hypertension. (Courtesy Mr M.Wade, Department of subarachnoid hemorrhage. (Courtesy Mr M.Wade, Department of
Medical Illustrations, Royal Perth Hospital, Australia.) Medical Illustrations, Royal Perth Hospital, Australia.)
Headache 93

DIFFERENTIAL DIAGNOSIS • Subarachnoid hemorrhage (110): sudden onset, severe


Primary headaches (see Table 13) headache, stiff neck, subhyaloid hemorrhage (see p.249).
• Cerebral venous sinus thrombosis (111, 112) (see
Secondary headaches p.257).
• Meningitis: fever, meningismus (stiff neck) (see p.273, • Subdural hemorrhage (113) (see p.177).
291). • Cerebral tumor: headache in early morning, neurologic
• Temporal arteritis (108): elderly, scalp tenderness, jaw signs often present, personality change, seizures (see
claudication, high ESR (see p.234). p.357).
• Idiopathic (benign) intracranial hypertension (109): • Arnold–Chiari malformation (114) (see p.136).
young, obese women, papilledema (see p.477).

111, 112 CT brain scan 111 112


with contrast (111) and MRI
brain scan (112),T1W post
contrast axial images showing
a filling defect at the torcula
due to venous sinus
thrombosis (the empty delta
sign, arrows).

113 CT brain scan, axial


image, showing a right
subdural hematoma as slight
hyperdensity over the
convexity of the right
cerebral hemisphere
(arrows).

114 MRI brain scan, sagittal


image, showing an
Arnold–Chiari malformation
(arrow).

113 114
94 Headache

INVESTIGATIONS (see Table 14)

Table 14 Investigation of the patient presenting with headache

Headache onset Abrupt Subacute Subacute Progressive Intermittent


(hours) (hours to days) (days to weeks) (may be abrupt)

Associated features*:
Unilateral headache + +
Bilateral headache + + + + +
Visual symptoms + +
Photophobia + +
Family history +
Personality change +
Seizures + +
Focal neurologic + + +
symptoms and signs
Fever + +
Neck stiffness + +
Papilledema +
Subhyaloid blood +
Coma + + + +
Rash +

Recurrent +

Suspect** Subarachnoid Meningitis Viral Structural Migraine


hemorrahge encephalitis intracranial
lesion

Think Is there a high index of suspicion for bacterial meningitis?


Are immediate blood cultures and empirical antibiotics required?

Imaging (CT or MRI) Subarachnoid Normal May be normal Lesion: Normal;


blood or normal (meninges may subdural hematoma not indicated
enhance) tumor, abscess except to exclude
other causes

Lumbar puncture: May be unsafe if there is evidence of raised intracranial pressure (see p.32), a mass, lesion, a swollen brain, a bleeding
diathesis or sepsis at the site of the lumbar puncture
Needs to be delayed at least 12 hours after onset of suspected subarachnoid hemorrhage, and if CT brain scan is
negative

Cerebrospinal fluid
Appearance Xanthochromia Turbid Clear or turbid Contraindicated Not indicated
RBC + 0 0 0
WBC <3 (normal) Polymorphs Lymphocytes <3 (normal)
Gram stain Negative Organisms Negative Negative
Protein Normal or raised Raised Raised Normal
Glucose Normal Low Normal or low Normal

Management Consult Antibiotics Acyclovir Consult Analgesics


Neurosurgery EEG Neurosurgery Antiemetic
and Neuroradiology HSV PCR Specific treatment:
(?surgery, ?GDC coil) ergotamine
Cerebral angiography sumatriptan

See p.249 See pp.273, 291 See p.292 See pp.177, 357 See p.95

* Not all need to be present


** Muscle contraction/tension-type headache is the most common cause of headache
+ Present
Blank: absent
Migraine 95

Restrict to patients presenting with MIGRAINE


• Atypical symptoms or signs indicative of organic
pathology.
• Certain physical signs, such as papilledema or a red DEFINITION
tender scalp vessel. A symptom complex, or syndrome, that manifests as discrete
• Unremitting course which is unresponsive to episodes of headache associated with other features of
conventional treatment. sensory sensitivity.

If suspicion of secondary headache consider EPIDEMIOLOGY


• Full blood count. Prevalence
• ESR in patients over 50 years old. Lifetime
• Blood biochemistry. • Women: 33% (95% CI: 31–37%).
• CT or MRI brain scan (111–114): depends of the type • Men: 13% (95% CI: 12–16%).
of headache and any associated features. For example,
sudden onset headache associated with loss of 1 year
consciousness suggests probable subarachnoid • Women: 25% (95% CI: 23–29%).
hemorrhage or pituitary apoplexy in which case CT brain • Men: 7.5% (CI: 7–9%).
scanning is the initial investigation of choice. Headache • Higher in Caucasians than African Americans, than
coming on over several weeks which is worse in the Asians.
morning and on coughing suggests raised intracranial
pressure in which case a contrast enhanced CT scan Age
(?brain tumor) or MRI scan (?venous sinus thrombosis), • Onset is nearly always (90%) before age 50 years; 25%
perhaps followed by angiography would be appropriate. begin in childhood.
If an Arnold–Chiari malformation is suspected, then the • Peak incidence at age 10–12 for males and 14–16 years
craniocervical junction and brain needs to be imaged by for females.
high resolution CT or MRI scan. The imaging • Peak prevalence at age 50 years for men and 35 years for
requirements of headache with localizing features females.
requires a little more thought: for example pituitary • Attacks commonly increase in frequency at the
disease (MRI), ear disease (MRI for internal auditory menopause, but may decrease (or become acephalgic).
meatus, CT for middle and external ear problems) or
sinus disease (CT to diagnose plus look at the ostia), but Gender
in any case it is essential to state all the relevant history • Children: M=F.
on the request card or the investigation may not answer • Adolescents and adults: F>M = 2–3:1.
the problem.
ETIOLOGY
DIAGNOSIS Unknown.
Before a primary type of headache such as muscle
contraction/tension headache or migraine is diagnosed, Migraine without aura
secondary headaches should be considered and eliminated A combination of genetic factors (possibly involving calcium
on clinical grounds or by appropriate investigations. ion channel function) and environmental factors (e.g.
stress); first degree relatives of probands with migraine
TREATMENT without aura have a twofold increased risk of migraine
• Explain the problem to the patient. without aura compared with the general population; the
• Identify and avoid any precipitating factors. probandwise concordance rate is higher in monozygotic
• Institute appropriate specific treatment (see Table 14). (MZ) than dizygotic twins (0.43 [95% CI: 0.36–0.49] vs.
0.31 [95% CI: 0.26–0.36]).

Migraine with aura


Largely genetic; first degree relatives of probands with
migraine with aura have a fourfold increased risk of migraine
with aura compared with the general population; the pro-
bandwise concordance rate is higher in monozygotic than
dizygotic twins (0.50 [95% CI: 0.38–0.62] vs. 0.21 [95%
CI: 0.12–0.30]). However, environmental factors are also
important as the pairwise concordance rate is less than 100%
in MZ twin pairs (i.e. it is about 34% [95% CI: 23–45%]).
96 Headache

Familial hemiplegic migraine (FHM) neurons that innervate the cranial circulation. This peptide
A rare autosomal dominant subtype of migraine with aura. is not only a vasodilator but it also mediates a sterile
Genes for FHM map to chromosomes 19p13 and 1q but neurogenic inflammation (vasodilatation and edema) within
some families with FHM do not link to either locus, the dura mater.
indicating genetic heterogeneity of FHM. The CACNa1A
gene at 19p13 encodes the α1A subunit (the ion conducting Migraine aura and headache
part) of a brain specific P/Q type voltage-dependent At the onset of aura, regional cerebral blood flow to the
calcium channel, suggesting that migraine may be a ‘cerebral clinically involved part of the brain is reduced by about 20%,
calcium channelopathy’. and reduced neuronal activity spreads in a wave across the
cerebral cortex, usually beginning in the occipital region and
PATHOPHYSIOLOGY slowly moving forward (spreading depression of Laeo).
Triggered by the action of a multitude of environmental and Migraine headache begins while regional cerebral blood
biochemical factors on the cerebral cortex or hypothalamus flow is reduced. Platelets in the blood release serotonin (5-
(the latter of which, in turn, is modulated by seasonal hydroxytryptamine, 5-HT), and this leads to platelet
patterns, diurnal and biologic clocks, and hormonal factors aggregation. During the headache, the level of a vasodilator
and coitus); the premonitory symptoms of elation, yawning peptide, CGRP increases in the external jugular venous
or a craving for sweet foods, experienced by about 25% of blood, and some intracranial arteries (particularly those of
patients, suggest hypothalamic activation. the dura mater) become dilated and inflamed. Vascular
dilatation and neurogenic inflammation is believed to be
Triggers responsible for the pulsatile nature of the headache.
• Emotional stress and tension. Migraine attacks can be ameliorated by activating 5-
• Relaxation after stress. hydroxytryptamine 1D (5-HT1D) presynaptic receptors
• Fatigue. within the vessel wall, thus blocking release of vasoactive
• Hormonal changes: fall in estradiol levels at menstruation neuropeptides, causing vasoconstriction of certain cerebral
and midcycle. and dural arteries, and inhibiting depolarization of
• High dose estrogen-containing contraceptives. trigeminal axons, functionally blocking activation of
• Strong sensory stimulation: bright or flickering light; trigeminal perivascular nerve terminals.
loud noise; strong smells; occipital nerve compression
(e.g. ‘swim-goggle migraine’). CLINICAL FEATURES
• Head trauma, such as heading the ball in soccer Precipitating factors
(‘footballer’s migraine’). See Triggers, above.
• Food idiosyncrasies/allergies: rich foods (e.g. chocolate,
fatty foods), red wines, specific dietary amines (cheese). Phase one: prodrome
• Missing meals. • Occurs in 25–50% of migraineurs.
• Sleeping late in the morning. • Gradual onset and evolution over up to 24 hours.
• Meteorologic changes. • Lightheadedness, dulled perception, irritability,
• Vasodilators, such as alcohol, monosodium glutamate, withdrawal, cravings for particular foods (particularly
and anti-anginal agents. sweet foods), frequent yawning, elation and speech
• Substance misuse. difficulties.
• Physical activity (footballer’s migraine, coital cephalgia).

Trigeminovascular reflex (115)


The activated cerebral cortex and hypothalamus stimulate 115 Trigger factors
brainstem nuclei, dorsal raphe nuclei (which contain
serotonin) and locus coeruleus (containing noradrenaline) Hypohalamus Cortex
and trigger the trigeminovascular reflex which constitutes
serotonergic and noradrenergic pathways that project from Thalamus
the brainstem to the cortical microcirculation and the spinal Dorsal
trigeminal nucleus and spinal cord. raphe Locus
Dura
Trigeminal nucleus coeruleus
Axons of the first division of the trigeminal nerve, which
ganglion
innervate the pain-sensitive intracranial structures,
depolarize as a result of direct neuronal activation or
vasodilatation of dural and cerebral arteries, or both, leading
to central transmission of nociceptive pain signals to bipolar Blood
neurons in the trigeminal ganglion and on to the trigeminal vessel Medulla
nucleus in its most caudal extent in the caudal medulla and
the dorsal horn of the spinal cord at C1 and C2 (accounting
for the commonly reported neck pain with migraine). C1
Impulses are then transmitted to the ventroposteriomedial
nucleus of the thalamus via the quintothalamic tract, from
where they are relayed to the cortex. C2
Stimulation of the trigeminal ganglion leads to the
release of powerful vasodilator neuropeptides such as 115 Diagram showing how the trigeminovascular pathway may be
calcitonin gene-related peptide (CGRP) from trigeminal activated to produce migraine headache.
Migraine 97

Phase two: aura Associated features


• 15–25% of migraine attacks are associated with aura. • Scalp tenderness on the affected side (about two-thirds
• Visual symptoms most commonly: blurred vision, of cases).
flashing lights (photopsia) or shimmering zigzag lines of • Nausea (90% of patients).
light (fortification spectra), sometimes around an area of • Vomiting (60%).
impaired vision or blindness (scintillating scotoma) in a • Diarrhea (20%).
part of the visual field of one or both eyes (116). • Heightened awareness of sensations such as smell and
• Somatosensory: tingling or pins and needles, or less noise.
commonly numbness, in the face, arm, hand or leg. • Fluid retention at onset, and polyuria as headache
• Dysphasia: difficulty understanding and expressing speech. subsides.
• Gradual onset, symptoms ‘build up’ or progress over
5–10 minutes, then subside within 5–60 minutes. Duration
• Followed within 60 minutes by headache; the aura may • 4–72 hours.
continue into the headache phase. • Commonly 2–6 hours in children, and 6–24 hours in
adults.
Phase three: headache
Present in most, but not all migraine attacks (cf. ‘migraine Phase four: postdrome
without headache’ or ‘acephalgic migraine’). For up to 24 hours after the headache has subsided, most
migraineurs feel tired and ‘drained’ or ‘washed-out’, with
Site aching muscles. Others however, become euphoric for a
• Unilateral in two-thirds of patients, and bilateral in one- period of time.
third.
• Frontotemporal region commonly, spreading to occipital Periodicity with recurrence
region. Migraine is paroxysmal; clearly defined episodes recur as
often as 4–6 times each month.
Quality
• Throbbing/pulsatile. Family history
• Moderate to severe. Family history of migraine is present in more than half of
patients.
Aggravating factors
• Physical activity/movement. SPECIAL FORMS
• Bright light (photophobia, 80%). Migraine variants
• Loud noise (phonophobia). Less than 5% of migraineurs.

Retinal migraine
Monocular, rather than binocular hemianopic visual
disturbance.

116 Illustration of 116 Ophthalmoplegic migraine:


the type of visual • Paralysis of ≥1 of the ocular cranial nerves, usually the
aura a migraineur IIIrd nerve, at the height of a migraine headache.
may describe; in this • The paralysis usually resolves but may persist after
case shimmering recurrent episodes.
zigzag lines of light • This entity does not embrace a transient dilatation or
(fortification constriction (Horner’s syndrome) of one pupil as this is
spectra). quite commonly seen during severe migraine attacks.
• The cause of the cranial neuropathy in ophthalmoplegic
migraine is probably transient ischemia of the cranial
nerve.

Vertebrobasilar migraine
Gradual onset and evolution over several minutes of
brainstem, cerebellar and visual disturbances, often accom-
panied or followed by headache and syncope.

Hemiplegic migraine
• Hemiparesis preceding or occurring with a migraine
headache.
• A family history of hemiplegic migraine is often present
and the gene is located on chromosome 19 or 1.
98 Headache

Migrainous infarction Secondary


• Permanent focal neurologic symptoms persisting beyond • Drug-rebound headache: a periodic daily bilateral
24 hours after the cessation of migraine headache. headache that has gradually increased in frequency, and
Cranial CT or MRI scan shows features consistent with changed in character from the typical migraine
cerebral infarction. headaches, in concurrence with increasing consumption
• The cause is probably arterial thrombosis, provoked by and misuse of analgesic drugs, particularly those which
arterial spasm and a procoagulant state (e.g. cigarette also contain caffeine.
smoking and the oral contraceptive pill). • Systemic infection.
• Giant cell arteritis.
Menstrual migraine • Raised intracranial pressure: idiopathic intracranial
Just before menstruation, plasma estradiol (rather than hypertension, brain tumor.
progesterone) levels fall rapidly below about 20 ng/ml
which sets in motion a series of changes (perhaps through Vertebrobasilar migraine
prostaglandins) that culminate in the onset of migraine in Neurocardiogenic syncope.
about 60% of women migraineurs and exclusively at that
time in about 14%. Migraine is relieved by pregnancy in Migraine aura without headache
about 60% of women, many, but not all, of whom have a (acephalgic migraine)
history of menstrual migraine. • TIA.
• Epileptic seizure.
Migraine in childhood • Arteriovenous malformation.
• Headache and vomiting are common but the child may • Mitochondrial DNA disorders (e.g. MELAS, see p.162).
be unable to describe the symptoms and may simply • Cerebral autosomal dominant arteriopathy with
appear pale, ill, limp, and inert, complaining of poorly subcortical infarction and leukoencephalopathy
localized abdominal pain. (CADASIL) (see p.261).
• Fever up to 38.5°C (101.3°F) may be present so that the
suspicion of appendicitis or mesenteric adenitis often INVESTIGATIONS
arises. • Should only be necessary if headache is suspected to be
• Rather than accept a label of ‘bilious attacks’ or ‘periodic secondary to another disorder.
syndrome’, recurrent headaches or vomiting attacks in • ‘Alarm symptoms’ include:
children which may develop at times of excitement or – Onset above age 50 years.
stress should be considered as possibly migrainous and – Aura without headache.
not psychosomatic. – Aura symptoms of acute onset without spread.
– Aura symptoms that are very brief (<5 mins) or unusually
DIFFERENTIAL DIAGNOSIS long (>60 min).
Abrupt onset of headache (‘thunderclap headache’) – Aura symptoms that are stereotyped (i.e. always at the
Primary same body site).
• Cluster headache. – Sudden increase in migraine frequency or change in
• Benign exertional or sex headache. migraine characteristics.
• Idiopathic stabbing headache. – High fever.
– Abnormal neurologic examination.
Secondary • The role of imaging in patients with suspected migraine is
• Subarachnoid hemorrhage. to exclude structural causes for the headache such as AVMs
• ‘Sentinel headache’ (enlargement of a cerebral aneurysm or tumors. A contrast-enhanced CT scan is satisfactory for
without rupture). this, and is usually normal (unless there is an AVM or
• Intracerebral hemorrhage. tumor). If MRI is performed, the T2W image occasionally
• A precipitous rise in blood pressure: drug induced shows areas of altered signal in the white matter which may
headache. be residual ischemic changes following a recent or
• Head injury. prolonged attack, or may rarely be due to CADASIL. The
• Acute obstruction of the CSF pathways. appearance is non-specific however.

Unilateral headache DIAGNOSIS


• Cluster headache. Clinical
• Temporal arteritis. • At least 5 attacks.
• Glaucoma. • Attacks last 4–72 hours if untreated or unsuccessfully
• Temperomandibular joint disease. treated.
• Internal carotid or vertebral artery dissection. • At least two of:
• Structural intracranial lesion. – Unilateral headache.
– Pulsating headache.
Continuous or daily headache – Moderate or severe headache.
Primary – Headache aggravated by routine physical activity.
• Tension headache: just headache, and sometimes mild • At least one of:
photophobia and phonophobia but no other features of – Nausea, with or without vomiting.
sensory sensitivity. – Photophobia.
• Mixed migraine/tension headache. – Phonophobia.
Migraine 99

TREATMENT Specific antimigraine agents


Avoid precipitating/triggering factors Ergot alkaloids:
Identify these by keeping a diary if necessary. • Alpha adrenergic agonists with potent 5-HT1 receptor
affinity. Also stimulants of dopamine D2 receptors in
Treatment of the acute migraine attack brainstem and gut (vomiting and diarrhea), and vascular
Ancillary measures adrenergic and 5-HT2 receptors (vasoconstriction).
• Rest in a quiet dark room. • Ergotamine exists in several forms:
• Intravenous fluids if severely dehydrated. – 1 mg capsule of ergotamine tartrate, or combined with
caffeine.
Non-specific analgesics and antiemetic/prokinetic compounds – 2 mg suppository of ergotamine tartrate combined with
• Treat as early as possible (e.g. aspirin 600–900 mg or caffeine 100 mg.
ibuprofen 400–800 mg, together with metoclopramide – 2 mg tablet of ergotamine tartrate combined with
10 mg), and wait 40 minutes. If headache persists, try a caffeine and an antiemetic.
specific treatment such as ergotamine 1 mg capsule or • Dihydroergotamine mesylate can also be given orally
sumatriptan 50 mg tablet, and wait >1 hour. If headache (absorbed erratically), or more effectively, by suppository
persists, repeat. (See below.) (1–2 mg), inhalation (0.36 mg), sublingual (1–2 mg),
• Anti-emetic and prokinetic compounds (e.g. intramuscular (0.5–1 mg) and intravenous (0.5–1 mg)
metoclopramide 10 mg or domperidone 10–20 mg) if routes.
nausea and vomiting are a problem. Metoclopramide is • Effective in about half of cases.
preferable because it improves the oral absorption of • If two doses at intervals of 2–6 hours are ineffective, no
other drugs (which is impaired in migraine attacks) and more should be given for that attack.
may have a favorable central effect. If vomiting is severe, • Limitations include poor oral and rectal bioavailability
suppositories of domperidone, prochlorperazine, or (<3%); frequent, long-lasting adverse effects (nausea,
chlorpromazine may be helpful. vomiting, diarrhea, muscle cramps, malaise, cold/ting-
• Simple analgesic drugs: ling fingers and toes); and risk of headache and ergotism
– Aspirin 2 or 3 × 300 mg chewable tablets (600–900 mg) (generalized vasospasm, causing acral cyanosis, digital
orally. necrosis, intermittent claudication, and tissue infarction)
– Paracetamol (acetaminophen) 2 × 500 mg tablets (1g) with chronic recurrent use (>1 day per week).
orally. • The drug of choice in a limited number of migraine
– Compound codeine-containing analgesic (e.g. codeine sufferers who have infrequent or long duration headaches
15–30 mg) but may cause or exacerbate nausea. and are likely to comply with dosing restrictions. For
• Non-steroidal anti-inflammatory drugs: most migraine sufferers requiring specific migraine
– Ibuprofen. treatment, a triptan is generally a better option from both
– Naproxen: oral, rectal. an efficacy and side-effect perspective.
– Diclofenac: oral (potassium salt-rapid absorption),
intramuscular. Triptans:
– Ketorolac: intramuscular. Selective and potent agonists of 5-HT1B, 1D, 1F, and to
• Other non-specific drugs: some extent 5-HT1A receptors. Antimigraine effects are
– Chlorpromazine: intramuscular, but long term considera- mediated by:
tions (e.g. tardive dyskinesia). • Inhibition of firing of cells in trigeminal nuclei (5-HT1B or
– Narcotic analgesic use is highly controversial, not D receptors in the brainstem: second generation triptans).
evidence-based, and is associated with prominent adverse • Inhibition of dural neurogenic inflammation and plasma
effects and a high risk of dependency. Most patients who extravasation (presynaptic 5-HT1D receptor stimulation
require narcotics are misusing analgesics or ergots. on trigeminal neurons).
– Lignocaine infusion: may be indicated for prolonged • Vasoconstriction of meningeal, dural, cerebral or pial vessels
severe migraine unresponsive to other therapy or for (stimulation of presynaptic vascular 5-HT1B receptors).
rebound headache (for the latter, all analgesics are
withheld during the infusion). Procedure: a 12-lead EKG First-generation triptans: sumatriptan:
is obtained and examined before and 30–60 minutes • A specific and selective agonist of 5-HT1D presynaptic
after starting the infusion. Lignocaine is delivered by a receptors on cranial blood vessels, inhibiting trigeminal
pump device at a rate of 2 mg/min (no bolus is given). neuronal firing at the trigeminal nerve ending.
The patient is attached to a bedside cardiac monitor, and • Available as subcutaneous injection (6 mg), oral tablets
a rhythm strip is obtained every 5 minutes for the first (25, 50 and 100 mg), nasal spray (20 mg), and rectal
30 minutes, then every 15 minutes for 3 hours, and preparations.
thereafter every 2 hours. Pulse rate and blood pressure • Subcutaneous sumatriptan injection (6 mg):
are measured every 5 minutes for the first 30 minutes, – Bioavailability: 96%.
then every 15 minutes for 3 hours, and thereafter every – Therapeutic plasma levels: within 10 minutes.
2 hours while the patient is awake. The infusion is – 79% of patients improved at 2 hours after injection
maintained until the patient has been headache free for (‘therapeutic gain’ [TG]: active minus placebo = 52%);
at least 12 hours. The duration of infusion should not 71% (TG: 51%) improved within 1 hour.
exceed 14 days. Contraindications include significant – 60% of patients pain-free at 2 hours after injection (TG:
heart disease (e.g. severe sinoatrial, atrioventricular, or 42%); 43% (TG: 35%) pain-free after 1 hour.
intraventricular heart block), epileptic seizures, or allergic (Continued overleaf)
reaction to lignocaine.
100 Headache

First-generation triptans: sumatriptan (continued): Prevention


• Oral sumatriptan tablets (100 mg): Non-pharmacologic
– Bioavailability: 14%. • Avoid precipitating factors (e.g. dietary-chocolate,
– Therapeutic plasma levels: within 30–90 minutes. oranges, monosodium glutamate).
– 59% (95% CI: 57–60%) of patients improved at 2 hours • Stress reduction through relaxation exercises and tapes,
after tablets(TG: 33%). meditation, yoga, swimming and similar strategies will
– 29% (27–30%) of patients pain-free at 2 hours (TG 26%). reduce migraine frequency in many patients.
• Oral sumatriptan is more effective than conventional • Regular exercise such as swimming.
treatment with aspirin and metoclopramide or oral ergo- • Acupuncture in short courses by an experienced therapist
tamine plus caffeine, particularly in the second and third can be a useful adjunct to other strategies in some
attacks, suggesting greater consistency for sumatriptan. patients.
• Recurrence of headache occurs within 24–48 hours in
about one-third of responders to sumatriptan (and any Pharmacologic
other acute antimigraine drug). Repeated drug adminis- • The indication for prophylactic therapy is when the
tration is usually effective, but the headache may recur patient needs it. This is usually when the migraine attacks
again. are frequently interfering with their life and recurring
• Adverse effects of sumatriptan are common but are every 2 weeks or so and not responding quickly and
usually mild and short-lived. The most frequent are adequately to acute treatment.
tingling, paresthesias, and warm sensations in the head, • Efficacy is limited: at most about half of patients will have
neck, chest, and limbs; less frequent are dizziness, a reduction in attack frequency of half or more.
flushing, and neck pain or stiffness. The risk and intensity • Adverse effects occur commonly.
is greater with the fixed subcutaneous formulation. • The choice of prophylactic agent is primarily determined
‘Chest-related symptoms’ include short-lived heaviness by the patient and which potential adverse effects are
or pressure in the arms and chest, shortness of breath, most acceptable (see Table 15).
chest discomfort, anxiety, palpitations, and, very rarely, • Discuss the adverse effect profile of each drug with the
chest pain. The mechanism is unknown. The risk of patient and determine their preference. Asthma (beta
sumatriptan-induced myocardial ischemia in the absence blockers) and weight gain (pizotifen [pizotyline] and
of coronary artery disease appears to be acceptable (e.g. valproate) are by far the major concerns.
no greater than the risk of exercise-induced myocardial • Establish realistic expectations with the patient before
ischemia in sportsmen). starting: the medication may reduce the frequency of
attacks but uncommonly abolishes attacks, and so
Second-generation triptans: occasional breakthrough attacks requiring acute
• Zolmitriptan 2.5 mg, 5 mg: similar to oral sumatriptan. treatment will occur.
• Naratriptan 2.5 mg: slower action and perhaps fewer and • Start slowly, with dosage increments every 7–10 days to
less severe adverse effects, but lower efficacy. minimize adverse effects.
• Rizatriptan 10 mg, 40 mg: better efficacy and • Encourage patients to persist for at least 3 months to
consistency, and similar tolerability. adequately trial the drug and because most adverse
• Almotriptan 12.5 mg: similar efficacy, better consistency effects become less prominent with time.
and tolerability. • Follow on with a drug free interval to reassess the
• Eletriptan 20 mg, 40 mg and 80 mg: 80 mg orally is frequency and severity of migraine attacks.
more effective than sumatriptan 100 mg orally with
similar consistency but lower tolerability. Menstrual migraine
• Frovatriptan: possibly lower efficacy than oral sumatriptan. • Try standard prophylactic (interval) therapy, as above,
before hormone manipulation.
Advantages over oral sumatriptan: • Continuous bromocriptine therapy, 2.5 mg tds, added
• Higher bioavailability: 45–75%. to the existing prophylactic regime may be beneficial.
• More rapid therapeutic plasma levels: within 30–60 Adverse effects, such as light-headedness and nausea can
minutes. be minimized by gradual introduction of medication,
• Greater potency at 5-HT1B/D receptor sites. beginning with 1.25 mg daily and followed by daily
• Increased lipophilicity and brain penetration (hence, incremental 1.25 mg increases over 1 week to full dosage
direct attenuation of excitability of cells within the (i.e. 25 mg tds).
trigeminal nuclei of the brainstem, as well as vasocon- • Non-steroidal anti-inflammatory drug, such as diclofenac
striction and peripheral inhibition of trigeminal peri- (enteric coated) 50 mg bd commencing 24 hours before
vascular terminals). anticipated menstruation (to attempt to counteract the
• Cheaper alternatives for patients who do not respond to overflow of prostaglandin from the contracting uterus).
oral sumatriptan. • Application of a gel containing 1.5 mg estradiol to the
skin 48 hours before the expected onset of menstruation.
None of these agents is consistently effective in all patients • Subcutaneous implantation of estradiol pellets, starting
and all attacks, and some cause disturbing adverse effects. with 100 mg, inhibits ovulation and maintains estradiol
Rizatriptan 10 mg, eletriptan 80 mg, and almotriptan levels, while regular monthly periods can by induced by
12.5 mg provide the highest likelihood of success. cyclical oral progestogens. Depoprovera, a different oral
Ergotamine and sumatriptan should not be prescribed for contraceptive pill, or 3 monthly cycles of the oral
patients with suspected coronary artery disease, Prinzmetal contraceptive pill are alternative strategies.
variant angina, or uncontrolled hypertension.
Muscle Contraction/Tension-type Headache 101

• Tamoxifen citrate, 10–20 mg daily preceding and during MUSCLE CONTRACTION/TENSION-


menstruation may help; tamoxifen competes at estrogen TYPE HEADACHE
and anti-estrogen binding sites and is a calcium channel
blocker. However, the anti-estrogenic effect in young
women, such as osteoporosis, is an obvious problem with DEFINITION
this strategy. An episodic or chronic continuous headache due to
sustained muscle contraction.
CLINICAL COURSE
• Migraine is paroxysmal. Clearly defined episodes of EPIDEMIOLOGY
migraine recur as often as three or six times each month • Prevalence:
but sufferers remain symptom-free between attacks. – Episodic tension-type headache: 38% 1 year period
• The frequency of migraine attacks may increase until it prevalence.
develops into chronic daily headache (transformed – Chronic tension-type headache: 2.2% 1 year period
migraine), often as the result of stress or the over-use of prevalence.
ergotamine or analgesics. • Age:
• Migraine symptoms frequently change over time. – Any age.
• The severity of the attacks often diminish with time and – Onset before 10 years of age in 15%.
in some patients the attacks cease in latter years, – Peak prevalence 30–39 years of age.
particularly after the menopause in women. • Gender: F > M: 1.2:1.
• In some women however, attacks increase in frequency
at the menopause. ETIOLOGY
• Remission occurs in 70% of pregnancies. Unknown. Perhaps, at least in part, a disorder of the CNS
• For young women, below the age of 25 years, the with probable trigeminal activation and sensitization of
relative risk of ischemic stroke among migraineurs is second-order trigeminal neurons and some peripheral
increased (compared with age-matched controls), component. Generation of nitric oxide may have a role in
particularly among those taking the oral contraceptive central sensitization.
pill, but the absolute risk is extremely small (17–50 per
100 000 per year).

Table 15 Adverse effects of propyhlactic agents in migraine

Agent Dose Adverse effects


β-adrenoreceptor blockers
Propranolol 40–240 mg/day Tiredness, exercise intolerance, postural hypotension, vivid dreams,
Metoprolol 100–200 mg/day probably contraindicated in asthmatic patients
Anti-epileptic drugs
Sodium valproate 400–800 mg/day Drowsiness, tremor, weight gain, abnormal liver enzymes, risk of
teratogenicity in pregnant women
5-HT receptor antagonists
Pizotifen (pizotyline) 0.5–3 mg nocte Drowsiness, sedation, weight gain
Methysergide 1–4 mg/day Drowsiness, leg cramps, small (0.5%) risk of retroperitoneal or pleural
fibrosis if treatment is not stopped for 1 month every 4–6 months and
screened with physical examination, CXR, and renal function
Non-selective calcium channel blockers
Flunarizine Constipation; rare extrapyramidal adverse effects with flunarizine
Verapamil 40–120 mg tds Effective in preventing cluster headache
Vitamins
Riboflavin (vitamin B2) 400 mg/day Diarrhea, polyuria
Herbal medicines
Feverfew
Non-steroidal anti-inflammatory drugs
Naproxen Small risk of peptic ulceration
Hormonal manipulation
Antidepressants
Amitriptyline 50–150 mg/day Drowsiness, dry mouth, blurred vision
Dothiepin Useful for concurrent tension-type headaches
102 Headache

Predisposing factors • Associated symptoms: nausea may be present, but not


Genetic factors vomiting or sensory sensitivity to head movement, light or
First degree relatives of probands with chronic tension sound (although one of the latter pair is acceptable). Patients
headache have about three times the risk of chronic tension are commonly polysymptomatic and may have a fear of an
headache than the general population, suggesting the underlying brain tumor or other medical ailment.
importance of genetic factors in chronic tension headache.
DIFFERENTIAL DIAGNOSIS
Physical abnormalities: • Migraine: muscle contraction/tension-type headaches
• Imbalance of bite: some patients have an uneven bite that occur in about half of migraine sufferers, often as a
throws strain on one temperomandibular joint and leads to background pain, but are not usually unilateral and are
the development of a clicking noise in the joint and pain not accompanied by sensitivity to light, noise or physical
radiating from the affected joint over the face and head motion, nor vomiting or visual disturbance (as migraines
(Costen’s syndrome). Dental attention is required to correct are). Sometimes migraine is gradually transformed into
the bite and it is necessary to reduce excessive jaw clenching chronic muscle contraction/tension-type headache, but
(bruxism) and the underlying anxiety state. more frequently it is episodic muscle contraction/ten-
• Cervical spondylosis: degenerative changes in the cervical sion-type headache which becomes chronic. In both
spine may lead to spasm of cervical muscles which may re- instances overuse of drugs frequently plays a role in
spond to manipulative or other therapy for the neck problem. aggravating the disorder. Discontinuation of daily drug
• Eye strain: refractive errors or ocular imbalance may be a source intake often results in improvement.
of tension and should be corrected. At school or at work, the • Cervicogenic pain: muscle contraction/tension-type
patient should sit in a comfortable chair which is adjusted to headache may also occur as a result of pain referred from
the height appropriate for the desk (to ensure good posture) the neck.
with light adjusted to the correct angle for comfort. • Chronic drug-induced (analgesics or ergotamine)
• Prolonged fasting, as occurs for the traditional Jewish Day of headache (often in a migraineur).
Atonement fast (Yom Kippur), is a strong precipitator of a • Raised intracranial pressure.
nonpulsating, bilateral, frontal headache of mild to moderate
intensity, particularly among chronic headache sufferers. The INVESTIGATIONS
mechanism may include metabolic changes, such as Not necessary unless a secondary headache is suspected.
hypoglycemia or the accumulation of certain metabolites but
the headache tends to occur within 18 hours of fasting. DIAGNOSIS
Based on the history.
Psychologic factors
• Stress may be clearly associated with exacerbations of head- Episodic tension-type headache:
ache in some patients and may be helped by readjustment of A At least 10 previous headaches episodes fulfilling criteria
stresses, alteration of life-style or psychologic counselling. B–D listed below. Number of days with such headache
• Sleep disturbance is a major perpetuating factor in <180/year (<15/month).
patients with intermittent muscle contraction headache, B Headache lasting from 30 minutes to 7 days.
which contributes to it becoming chronic. C At least 2 of the following pain characteristics:
• Chronic tension-type headache is usually, but not always, – Pressing/tightening (non-pulsating) quality.
related to or exacerbated by anxiety or depression (as – Mild or moderate intensity (may inhibit, but does not
well as sleep disturbance). prohibit activities).
– Bilateral location.
PATHOPHYSIOLOGY – No aggravation by walking stairs or similar routine
• Constant involuntary tightening or overcontraction of the physical activity.
frontal, temporal and occipital muscles, induced mentally or D Both of the following:
physically, may be the cause in some patients who experience – No nausea or vomiting (anorexia may occur).
some relief by learning to relax the appropriate muscles. – Photophobia and phonophobia are absent, or one but
• Psychogenic mechanisms are relevant in other patients. not the other is present.
• Some form of sensitization with second-order trigeminal E At least one of the following:
neurons is likely to be involved. – History, physical and neurologic examinations, and
appropriate investigations do not suggest headache
CLINICAL FEATURES associated with head trauma, vascular disorders, non-
• Site: usually bilateral, and diffuse or at the vertex of the vascular intracranial disorder, substances or their
head, around the head, or in the neck or occiput. withdrawal, non-cephalic infection, metabolic disorder,
Occasionally unilateral. or disorders of the cranium, neck, eyes, ears, nose,
• Nature: non-pulsatile, tight, pressing, heavy or band-like sinuses, teeth, mouth, or other facial or cranial structures.
sensation. – One of the disorders mentioned in E above is present,
• Onset: may occur during times of fatigue, stress, or anxiety. but tension-type headache does not occur for the first
• Timing: episodic, but may become constant, occurring time in close temporal relation to the disorder.
all day every day, for months and even years.
• Exacerbating and relieving factors: episodic headache Chronic tension-type headache
may be relieved by relaxation or analgesics. Chronic daily A Average headache frequency >15 days/month
headache is not relieved by analgesics and excessive (180 days/year) for >6 months fulfilling criteria B–D
medication may itself induce or exacerbate headache. listed below.
Cluster Headache (Migrainous Neuralgia) 103

B At least 2 of the following pain characteristics: CLUSTER HEADACHE


– Pressing/tightening (non-pulsating) quality. (MIGRAINOUS NEURALGIA)
– Mild or moderate severity (may inhibit, but does not
prohibit activities).
– Bilateral location. DEFINITION
– No aggravation by walking stairs or similar routine A rare form of primary headache marked by recurrent
physical activity. episodes, lasting 15–180 minutes, of excruciating unilateral
C Both of the following: periorbital pain and associated autonomic features, that tend
– No vomiting. to occur once or twice a day in bouts or clusters, lasting
– No more than one of the following: nausea, photophobia from weeks to months at a time, separated by remission
or phonophobia. periods of months or 1–2 years.
D As for E above.
EPIDEMIOLOGY
TREATMENT • Incidence: 6 (3–10)/100 000/year.
Reassurance • Lifetime prevalence: 0.3 (95% CI: 0.2–0.6)/1000.
Prompt and convincing. • Age: any age, but unusual in children and most common
20–50 years of age.
Consider discontinuing or minimizing heavy • Gender: mainly men (ratio 10:1).
analgesic intake
For patients in whom episodic tension-type headache or, less ETIOLOGY AND PATHOPHYSIOLOGY
frequently, migraine become transformed into chronic • A neurovascular disorder, which is hypothesized to be
tension-type headache, overuse of analgesic drugs frequently generated in the CNS in pacemaker or circadian regions
plays a role in aggravating the disorder, and discontinuation of the hypothalamic gray matter. The trigeminal/cervical
of daily drug intake often results in improvement. nuclear overlap is also central to the pathogenesis (see
Migraine, p.95). Activation of the trigeminovascular
Relaxation exercises, psychotherapy and system (i.e. release of calcitonin gene-related peptide
biofeedback training [CGRP] from peripheral terminals of trigeminal
A course in relaxation training may be highly beneficial in a nociceptive neurons, which supply cephalic blood vessels)
well motivated patient. They are now conducted by underlies symptoms of cluster headache. Increases in
psychologists, physiotherapists, and many hospitals and neuropeptide markers of this system rapidly return to
community health centers. Various forms of biofeedback normal after treatment with sumatriptan.
may assist relaxation but do not make a substantial impact. • The mechanism by which nitroglycerine can induce a
headache attack in cluster headache patients which is
Pharmacotherapy indistinguishable from a spontaneous attack is at least
• Tricyclic antidepressants: amitriptyline, in particular, may be partly due to activation of the trigeminovascular system.
very helpful, commencing with one-half of a 25 mg tablet
at night and gradually increasing to three tablets (75 mg) as CLINICAL FEATURES
a single nocturnal dose, provided there are no adverse effects • Severe, boring, unilateral periorbital pain that may radiate
such as drowsiness and confusion. If effective, an improve- upwards over the frontotemporal region and downwards
ment will usually be noticed within 2 weeks. Treatment to the face, jaw, neck and shoulder.
should be continued for about 6 months and then gradually • Edema of the ipsilateral eyelid.
phased out to see whether it is still necessary or whether • Redness of the ipsilateral eye (conjunctival injection).
improvement can be maintained with relaxation alone. • Watering of the ipsilateral eye (lacrimation).
• Anxiolytics, such as the benzodiazepines, have a limited • Miosis with or without ptosis ipsilaterally in about 20%
role. They should be used for short periods only, and of cases (117); permanent partial Horner’s syndrome
under supervision, because of the risk of habituation, (ptosis and miosis) ipsilaterally in about 5% of patients.
dependency and drug-induced headache. • Swelling, dilatation, and tenderness of the superficial
• Sodium valproate, 200–400 mg three times daily, may temporal vessels occasionally.
help control chronic tension-type headache. • Rhinorrhea and a blocked nostril ipsilaterally.
• Temporal spacing of attacks: the pain usually lasts 15
minutes to 2 hours and recurs once or twice a day, often
at night at the same time, for several weeks (active
117 periods), followed by an attack-free period (remission
period).
• Vasodilators, notably alcohol, nitrates and calcium
channel blockers, may precipitate attacks during the
active period, but not in remission periods.

117 Facial photograph during an attack of cluster headache showing


unilateral eyelid edema, ptosis, miosis and conjunctival injection of the
right eye.
104 Headache

DIFFERENTIAL DIAGNOSIS Cluster-tic syndrome


Primary short-lasting headaches with prominent • Cluster headache.
autonomic features • Additional symptoms of trigeminal neuralgia (see p.509),
Chronic paroxysmal hemicrania (CPH) which are always on the same side, in the same division,
• Frequent brief attacks of excruciating unilateral orbital, and can be provoked by the same stimuli.
supra-orbital or temporal pain (distribution of
ophthalmic division of trigeminal nerve) that last 2–45 Primary short-lasting headaches with few or no
minutes and recur five or more times daily (periods with autonomic features
lower daily frequency may occur). The pain is usually Trigeminal neuralgia (see p.509)
stabbing or boring, but may be throbbing as it builds up. • Gender: F>M.
• At least one associated autonomic symptom, such as • Pain: V2/V3>V1, in the territory of supply of the
conjunctival injection, lacrimation, nasal congestion, maxillary (V2) and mandibular (V3) branch of the
rhinorrhea, ptosis or eyelid edema. trigeminal nerve (i.e. cheek and jaw) more commonly
• Attacks usually (but not always) resolve rapidly within than the ophthalmic branch (V1) (i.e. forehead),
days of initiating treatment with indomethacin in an stabbing, and very severe.
adequate dose of up to 150 mg/day orally. • Attack duration: less than 1 second.
• Age of onset: usually in the twenties, but may occur in • Attack frequency: few to many/day.
children.
• Gender distribution: F>M (3:1). Idiopathic stabbing headache
• Pathophysiology: unknown. • Gender: F>M.
• Secondary causes include cavernous sinus meningioma, • Pain: any part of the head, stabbing, and severe.
gangliocytoma of the sella turcica, frontal lobe tumor, • Attack duration: less than 1 second.
arteriovenous malformation, intracranial hypertension, • Attack frequency: few to many/day.
connective tissue disease, and pancoast tumor. • Responds to indomethacin.
• Distinguished from cluster headache by the shorter dur-
ation and higher frequency of attacks, the female pre- Hypnic headache
ponderance, and the selective response to indomethacin. • Rare.
• Age: elderly (67–84 years).
Episodic paroxysmal hemicrania (EPH) • Gender: M>F (5:3).
• Extremely rare. • Pain: generalized, throbbing, and moderately severe.
• Frequent brief attacks of very severe, unilateral orbital • Attack duration: 15–30 minutes.
and/or temporal headache lasting from 1–30 minutes, • Attack frequency: 1–3 per night, often awakening patient
and recurring three of more times a day. from sleep.
• Accompanying ipsilateral autonomic features. • Responds to lithium carbonate 600 mg at bedtime.
• Age of onset: 12–51 years.
• Gender distribution: F=M. Cough headache
• Absolute response to indomethacin. A bilateral headache of sudden onset, lasting less than 1
• May evolve from a disorder with distinct intervals into a minute, and precipitated by coughing, in the absence of any
chronic unremitting form identical to CPH. intracranial disorder, such as a Chiari type I malformation.

Short-lasting unilateral neuralgiform headache with Benign exertional headache


conjunctival injection and tearing (SUNCT) syndrome A bilateral throbbing headache, lasting from 5 minutes to
• Rare. 24 hours, specifically provoked by physical exercise and
• Males predominate: M>F (17:2). unassociated with any systemic or intracranial disorder, such
• Paroxysms of unilateral moderately severe orbital or as subarachnoid hemorrhage, sinusitis and brain metastases.
temporal stabbing or throbbing pain lasting between 5
and 250 seconds (i.e. <5 minutes). Headache associated with sexual activity
• Average about 28 attacks a day (range 3–100). • Bilateral headaches precipitated by masturbation or
• Conjunctival injection is often present and prominent; coitus, in the absence of any systemic or intracranial
other autonomic features may occur such as lacrimation, disorder, such as subarachnoid hemorrhage.
sweating of the forehead and rhinorrhea. • Three types are recognized:
• Precipitating factors, such as mechanical movements of – A dull ache in the head and neck that intensifies as sexual
the neck, are common. excitement increases.
• Pathogenesis: most likely central activation of the trige- – A sudden, severe, explosive headache occurring at orgasm.
minovascular reflex, rather than a peripheral vasculitis. – A postural headache, resembling that of low CSF
• SUNCT syndrome may be secondary to posterior fossa pressure, developing after coitus.
lesions such as homolateral cerebellopontine angle
arteriovenous malformations. Other secondary or longer-lasting unilateral
• Refractory to all treatments so far described, including headaches
indomethacin. Migraine (see p.95)
• Women are affected more commonly than men.
• Headache often lasts a lot longer, up to 72 hours.
Cluster Headache (Migrainous Neuralgia) 105

• Migraine causes people to want to remain still in a quiet C Headache associated with at least one of the following
dark room whereas cluster headache is so severe that signs which have to be present on the painful side:
patients often pace the floor restlessly or wander conjunctival injection, lacrimation, nasal congestion,
outdoors to seek relief in the cold night air. rhinorrhea, forehead and facial sweating, miosis, ptosis,
eyelid edema.
Giant cell (cranial) arteritis (see p.234) D Incidence of attacks ranging from one attack on alternate
• Age >50 years at onset. days to 8 attacks/day.
• New onset of localized headache, that may be unilateral, E History and/or physical and neurologic examinations do
bilateral or occipital. not suggest other disorders associated with head trauma.
• Claudication of the jaw or tongue during eating.
• Symptoms of systemic upset (fever, sweats, malaise, Chronic
weight loss, polymyalgia). Attacks occur for more than 1 year without remission, or
• Scalp and temporal artery tenderness or decreased with remission lasting less than 14 days. The attacks are
temporal artery pulse. clinically indistinguishable from episodic cluster headache.
• Elevated ESR >50 mm/hour.
• Biopsy showing necrotizing arteritis. TREATMENT
• Responds within 24 hours of commencing prednisolone Acute attack
60 mg/day. • Oxygen 100% at 6–8 l/min often affords relief within
10 minutes, or
Glaucoma • Sumatriptan 100 mg po or 0.6 mg subcutaneous
• Recurrent attacks of pain in the eye and forehead, that injection (a 5-hydroxytryptamine (5HT1D) receptor
may be precipitated by sitting in the dark, mydriatics or agonist) aborts the pain in most cases.
emotional upset.
• Accompanying features include vomiting, visual Prophylactic
impairment, cloudiness of the cornea, discoloration of During the susceptible period, regular medication can be
the iris, a dilated pupil and circumcorneal injection. taken in anticipation of attacks:
• An arcuate scotoma and pallid cupped disc are charac- • Ergotamine tartrate 1–2 mg (e.g. one-half of a 2 mg
teristic of narrow-angle glaucoma, which is often familial. suppository) daily, or as a nocturnal dose.
• Tonometry is necessary to confirm elevated intra-ocular • Methysergide 1–2 mg three times daily, as for migraine,
pressure. if ergotamine is ineffective.
• Verapamil 40–120 mg tds for the duration of the cluster
Acute sinusitis (frontal, ethmoidal, maxillary) (e.g. 14 days).
• A dull, aching, throbbing pain over the affected sinus, • Prednisone 50 mg daily, for 3 days and then reduced
worse on bending or with the head in a certain position rapidly to a dose (usually about 25 mg a day) that is just
in bed. sufficient to prevent the attacks. This is effective in about
• The pain is temporarily eased by decongestant nasal drops 80% of cases in whom it should be maintained for the
and antibiotics and seldom lasts more than 1–2 weeks. duration of the bout before weaning off slowly.
• Usually associated with coryza and purulent nasal
discharge. Chronic cluster headache
• Lithium carbonate, 250 mg two or three times daily, the
Cervicogenic headache (see p.101) dose being adjusted to maintain a blood level of
Facial trauma with soft tissue injury 0.5–1.2 mmol/l. Adverse effects include confusion and
Pituitary adenoma (see p.379) tremor.
Pseudoaneurysm within the cavernous sinus (see pp.354, 508) • Verapamil in large doses.
Ipsilateral vertebral artery aneurysm (see p.249)
Occipital lobe arteriovenous malformation (see p.245) Chronic paroxysmal hemicrania
Upper cervical meningioma (see p.375) • Indomethacin, 25 mg three times daily, increasing to 50
mg tds after 1 week if there is no response. Occasional
INVESTIGATIONS patients require higher doses or slow-release
Nil usually, unless secondary causes of headache need to be indomethacin preparations at night to treat break
excluded: blood count (thrombocythemia), ESR (cranial through headaches.
arteritis), chest x-ray (pancoast tumor), vasculitic • Other agents that may be effective include naproxen,
investigations, and CT or MRI brain scan. Should the pain calcium channel blockers such as verapamil,
become bilateral, a lumbar puncture may be indicated to acetazolamide 250 mg tds, and possibly oxygen for
exclude intracranial hypertension. longer attacks.

DIAGNOSIS CLINICAL COURSE


Diagnostic criteria The headaches usually recur once or twice a day, often at
Episodic night, for a period of 4 weeks to 4 months. Remission is
A At least 5 attacks fulfilling criteria B–D. then usual, until the next cluster ensues, months or
B Severe, unilateral orbital, supraorbital and/or temporal 1–2 years later.
pain lasting 15–180 minutes if untreated.
106 Headache

Hand PJ, Stark RJ (2000) Intravenous lignocaine Schwartz BS, Stewart WF, Simon D, Lipton RB
FURTHER READING infusions for severe chronic daily headache. (1998) Epidemiology of tension-type
Med. J. Aust., 172: 157–159. headache. JAMA, 279: 381–383.
Hoffman EP (2001) Hemiplegic migraine – Tomkins GE, Jackson JL, O’Malley PG, et al.
HEADACHE downstream of a single-base change. N. Engl. (2001) Treatment of chronic headache with
Goadsby PJ (1999) Short-lasting primary J. Med., 345: 57–59. antidepressants: a meta-analysis. Am. J. Med.,
headaches: focus on trigeminal autonomic Launer LJ, Terwindt GM, Ferrari MD (1999) The III: 54–63.
cephalgias and indomethacin-sensitive prevalence and characteristics of migraine in a Welch KMA (2001) A 47-year-old woman with
headaches. Curr. Opin. Neurol., 12: 273–277. population-based cohort. The GEM study. tension-type headaches. JAMA, 286:
Lance JW (2000) Headache and face pain. Med. J. Neurology, 53: 537–542. 960–966.
Aust., 172: 450–455. Montagna P (2000) Molecular genetics of mi-
Van Gijn J (1999) Pitfalls in the diagnosis of sud- graine headaches: A review. Cephalgia, 20: CLUSTER HEADACHE
den headache. Proc. R. Coll. Physicians. Edin. 3–14. Ekbom K (1991) Treatment of acute cluster
29: 21–31. Silberstein SD, for the US Headache Consortium headache with sumatriptan. N. Engl. J. Med.,
(2000) Practice parameter: Evidence-based 325: 322–326.
MIGRAINE guidelines for migraine headache (and evi- Goadsby PJ, Edvinsson L (1994) Human in vivo
Bahra, Matharu MS, Buchel C et al. (2001) Brain- dence-based review). Neurology, 55: 754–763. evidence for trigeminovascular activation in
stem activation specific to migraine headache. Tfelt-Hansen P, Saxena PR, Dahlof C, et al. cluster headache. Brain, 117: 427–434.
Lancet, 357: 1016–1017. (2000) Ergotamine in the acute treatment of Goadsby PJ, Lipton RB (1997) A review of parox-
Bateman DN (2000) Triptans and migraine. migraine. A review and European consensus. ysmal hemicranias, SUNCT syndrome and
Lancet, 355: 860–861. Brain, 123: 9–18. other short-lasting headaches with autonomic
Boyle CAJ (1999) Management of menstrual mi- Ulrich V, Gervil M, Kyvik KO, et al. (1999) Evi- feature, including new cases. Brain, 120:
graine. Neurology, 53 (Suppl 1): S14–S18. dence of a genetic factor in migraine with aura: 193–209.
Ducros A, Denier C, Joutel A, et al. (2001) The A population-based Danish twin study. Ann. Leone M, D’Amico D, Frediani F, et al. (2000)
clinical spectrum of familial hemiplegic mi- Neurol., 45: 242–246. Verapamil in the prophylaxis of episodic clus-
graine associated with mutations in a neuronal ter headache: A double-blind study versus
calcium channel. N. Engl. J. Med., 345: MUSCLE CONTRACTION/TENSION-TYPE placebo. Neurology, 54: 1382–1385.
17–24. HEADACHE May A, Bahra A, Büchel C, Frackowiak RSJ,
Ferrari MD (1998) Migraine. Lancet, 351: Goadsby PJ (1999) Chronic tension-type Goadsby PJ (1998) Hypothalamic activation
1043–1051. headache: where are we? Brain, 122: in cluster headache attacks. Lancet, 352:
Ferrari MD, Roon KI, Lipton RB, Goadsby PJ 1611–1612. 275–278.
(2001) Oral triptans (serotonin 5-HT1B/1D Hand PJ, Stark RJ (2000) Intravenous lignocaine
agonists) in acute migraine treatment: a meta- infusions for severe chronic daily headache.
analysis of 53 trials. Lancet, 358: 1668–1675. Med. J. Aust., 172: 157–159.
Gervil M, Ulrich V, Kyvik KO, et al. (1999) Mi- Holroyd KA, O’Donnell FJ, Stensland M, et al.
graine without aura: a population-based twin (2001) Management of chronic tension-type
study. Ann. Neurol., 46: 606–611. headache with tricyclic antidepressant medica-
Goadsby PJ, Ferrari MD, Olesen J, et al. for the tion, stress management therapy and their
Eletriptan Steering Committee (2000) Eletrip- combination. A randomized controlled trial.
tan in acute migraine: A double-blind, place- JAMA, 285: 2208–2215.
bo-controlled comparison to sumatriptan. Ostergaard S, Russell MB, Bendtsen L, Olesen J
Neurology, 54: 156–163. (1997) Comparison of first degree relatives
Goldstein DJ, Roon KI, Offen WW, et al. (2001) and spouses of people with chronic tension
Selective seratonin 1F (5-HT 1F) receptor ag- headache. BMJ, 314: 1092–1093.
onist LY334370 for acute migraine: a ran- Russell MB, Ostergaard S, Bendsten L, Olesen J
domized controlled trial. Lancet, 358: (1999) Familial occurrence of chronic tension-
1230–1234. type headache. Cephalgia, 19: 207–210.
Chapter Five 107

Vertigo
VERTIGO of an underlying vestibular pathology. A sensation of
spinning is the most commonly described vertiginous
symptom, and is attributed to semicircular canal involve-
DEFINITION ment. Linear sensations of rocking, tilting and sudden
An illusion of movement. dropping are also valid descriptors of vertigo and probably
reflect involvement of the otolith organs (utricle and
EPIDEMIOLOGY saccule) which sense linear motion.
Incidence: very common. • Vertigo due to a vestibular disorder tends to be episodic,
and triggered or aggravated by head movements, rather
ETIOLOGY AND PATHOPHYSIOLOGY than postural changes alone. Conversely, constant dizziness,
Peripheral (most common) persisting for months and years, that is not associated with
Inner ear (semicircular canals, utricle, saccule) or aggravated by head movements does not usually have a
• Benign paroxysmal positional vertigo (25% of cases). primary vestibular cause; more commonly it has a
• Vestibular neuronitis (‘viral’ labyrinthitis). psychiatric basis. Moving or large field visual stimuli, such
• Ménière’s disease. as large cinema screens and supermarket aisles, may induce
• Benign recurrent vertigo. visual vertigo. Sometimes this leads to secondary
• Trauma (including perilymph fistula): head injury. agoraphobia (e.g. avoidance of large shopping complexes)
• Infection: otitis media, syphilis. and a misdiagnosis of a primary psychiatric disorder.
• Vascular lesions. • Associated symptoms include nausea, vomiting and
ataxia, due to the vertigo. The risk of psychologic
Vestibular nerve complications, such as panic disorder and depression
• Meningitis. increases in proportion to the chronicity of vestibular
• Acoustic neuroma and other cerebello-pontine angle symptoms. This makes assessment difficult as an
tumors (usually cause unsteady gait and rarely cause underlying vestibular disorder may be overlooked in
vertigo, particularly if there is no deafness). patients with prominent psychiatric complaints.
• Ototoxins: aminoglycosides, frusemide (furosemide) • If doubt remains after taking the history, reproduce in
(cause imbalance rather than vertigo). the patient the sensations of physiologic vertigo (rotation
on the spot with eyes closed) and lightheadedness
Central (hyperventilation) which can be compared with the
• Tumors (usually posterior fossa). patient’s presenting symptoms.
• Vertebro-basilar ischemia (but may also cause infarction
of the labyrinth): cerebellar or brainstem infarction. Is the vertigo due to a peripheral (labyrinthine) or
• Vascular malformation in brainstem (VIII nucleus) central (CNS) lesion?
• Multiple sclerosis involving brainstem. A neurologic examination targeted to an assessment of
• Trauma to brainstem. standing and walking balance, eye movements, hearing,
• Basilar migraine (may also cause end organ or peripheral otoscopy, and the Dix–Hallpike maneuver is important to
involvement). elicit focal neurologic signs and exclude central pathology.
• Arnold–Chiari malformation.
• Syringobulbia. Labyrinthine
• Drugs (alcohol, anti-epileptic drugs, barbiturates). • Vertigo generally more prominent than ataxia and
• Complex partial seizures can cause vertigo but almost nystagmus (exception: bilateral vestibular failure).
always with other more typical symptoms. • Auditory symptoms common.
• Vertigo is rarely, if ever, due to cervical spondylosis. • Nystagmus, typically a mixed torsional (ocular motion in
the frontal plane, sometimes incorrectly referred to as
CLINICAL FEATURES AND DIAGNOSIS rotatory) and horizontal nystagmus that beats away from
‘Doctor, I’m dizzy’ is one of the most common symptoms the side of the lesion, is readily suppressed by visual
in neurologic practice. fixation and abates quickly as the acute vertigo settles.
• No focal neurologic features.
Is it vertigo or another cause of dizziness such as • Positive Dix–Hallpike maneuver in patients with benign
pre-syncope? positional vertigo (see p.110): mixed torsional and
• Encourage the patient to describe the dizzy sensation in upbeating nystagmus begins after a short latent period,
their own words. is accompanied by moderately severe vertigo, fatigues
• A symptom of relative movement, whether it is one of self after up to 30 seconds, and is often less marked with
or of the environment, is usually indicative of vertigo and repeated maneuvers (habituation).
108 Vertigo

CNS Multiple sclerosis


• Ataxia and nystagmus are often more prominent than • Demyelination in the brainstem, involving the VIII
vertigo. Vertigo may occasionally be severe, as in lateral nucleus, may cause an acute vestibulopathy that may
medullary infarcts. superficially resemble vestibular neuronitis.
• Auditory symptoms rare. • A careful history and examination will usually detect
• Focal neurologic features may be present (e.g. diplopia, other CNS features of demyelination (see p.340).
dysarthria, dysphagia, visual field loss, hemisensory or
hemimotor disturbance). Acoustic neuromas
• Nystagmus may be unidirectional, bidirectional, or • Arise from the vestibular nerve (hence are more correctly
vertical (upbeating or downbeating). termed vestibular schwannomas) but do not usually
• Other central eye movement abnormalities are often present with vestibular symptoms because of the very
present (e.g. gaze palsy, skew deviation [vertical misalign- gradual loss of vestibular function that allows central
ment of the eyes]). compensation.
• The most common presentation is with unilateral tinnitus
What is the cause of the vestibular lesion? and hearing loss (see p.383).
Duration of vertigo
• Attacks lasting seconds only, particularly if they are Multi-sensory dizziness or disequilibrium
associated with head movements or postural changes, are • A syndrome which tends to occur in the elderly and
highly suggestive of either an uncompensated peripheral manifests with disequilibrium and vague, non-specific
vestibular lesion or benign positional vertigo (see p.110). dizziness when walking.
• Episodes lasting minutes may occur in migraine and, • It is a deafferentation syndrome caused by multiple
rarely, vertebro-basilar ischemia. sensory deficits such as visual impairment, peripheral
• Vertigo of hours duration occurs in conditions such as neuropathy, vestibular deficits and cervical spondylosis.
Ménière’s disease (see p.111). • Patients often have lower limb orthopedic impairments
• Vertigo that develops over several hours and gradually that will accentuate the disability.
resolves over a period of days to weeks is typical of • An important syndrome to recognize because sedative
vestibular neuronitis (see p.111). and vestibular suppressant drugs have the potential to
exacerbate the problem.
Associated features
Other clinical features associated with the vertigo are often Bilateral vestibular failure
a clue to the cause (see below). • Most commonly caused by aminoglycoside antibiotics
such as gentamicin ototoxicity, which may have occurred
Migraine in the setting of non-toxic blood levels and normal renal
• A common cause of vertigo, particularly in younger age function.
groups. • Ataxia and motion-induced oscillopsia are the usual
• May occur as an aura preceding a migraine headache, or presenting symptoms.
even as an isolated phenomenon (benign recurrent • Oscillopsia is a to-and-fro movement of the visual
vertigo). environment which, in this instance, is due to failure of
• Rarely, patients can present with chronic fluctuating the vestibulo-ocular reflex. These symptoms do not
spontaneous and positional vertigo, punctuated by usually become apparent until the patient starts to
intermittent headaches. mobilize and for this reason the diagnosis is often not
made until after the patient is discharged from hospital.
Vertebrobasilar ischemia • Vertigo is not a common or pronounced feature because
• Rarely presents with isolated vertigo. the vestibular loss is almost always bilateral and symmetric.
• Very occasionally a patient presents with intermittent,
isolated vertigo of minutes duration but this does not Perilymph fistula
usually persist as the sole manifestation for prolonged • An abnormal communication between the perilymph
periods. Generally, there are other focal neurologic compartment of the inner ear and middle ear caused by
symptoms present. a labyrinthine rupture in the region of the round or oval
• Likewise, exceptional patients with cerebellar infarcts may windows.
present with apparently isolated vertigo, however the • May be associated with head injury, barotrauma or
degree of gait ataxia is usually more severe and prolonged middle ear surgery.
than would be expected from the nature of the vertigo. • Presenting symptoms include fluctuating hearing loss,
• These patients are usually much older and often have ataxia, episodic vertigo (often triggered by exertion or
multiple vascular risk factors compared with the typical Valsalva maneuver), tinnitus and aural pressure.
patient with vestibular neuronitis. • The diagnosis is often difficult to make. A positive fistula
• Contrary to previous claims, vertigo that is triggered by test accompanied by vertigo and nystagmus in response
head extension is almost never due to vertebral artery to pressure insufflation of the ear canal may suggest the
occlusion by cervical osteophytes. Most patients with diagnosis but is neither specific nor sensitive.
head-extension vertigo have peripheral vestibular • Surgical exploration is often required when the index of
disorders, particularly benign positional vertigo. suspicion is high, particularly if there is significant hearing
loss.
Vertigo 109

DIFFERENTIAL DIAGNOSIS • Vestibular suppressant drugs should be reserved for the


Lightheadedness or presyncope (see p.59) treatment of acute vertigo that is accompanied by severe
• Postural hypotension. autonomic or vegetative symptoms. Suitable drugs
• Cardiac arrhythmia. include prochlorperazine, promethazine and chlorpro-
• Impaired cardiac output. mazine. Chronic treatment with vestibular suppressant
• Vaso-vagal attacks. drugs should be avoided if possible because of the risk of
• Hyperventilation and anxiety. development of drug-induced Parkinsonism and tardive
• Anemia. dyskinesia (see p.129). There is also some anecdotal
• Hypoglycemia. evidence that prolonged drug therapy may retard the
process of CNS compensation.
Other • Physical treatment programmes are often helpful for
• Migraine (see p.95). patients with persistent motion-induced vertigo following
• Complex partial seizure (see p.69). an acute vestibular insult who have failed to compensate
• Vertebrobasilar ischemia (see p.181). fully. Customized vestibular rehabilitation programmes are
often more effective than generic techniques such as the
INVESTIGATIONS Cawthorne–Cooksey exercises. Exercise programmes are
• CT brain scan. If central pathology is suspected, CT is designed to induce the symptoms and, by a process of
used, but it may miss some pathologies, such as habituation, are able to promote central compensation and
demyelination and ischemia. Specific contrast imagery thereby reduce the persistent symptoms.
should only be used if there are auditory features to • Treatment of psychiatric complications is important and,
suggest VIIIth nerve pathology. for this reason, a psychologist is a useful member of the
• MRI brain scan. If CT is negative and central pathology vestibular rehabilitation treatment team.
still suspected, MRI can be used. • Urgent referral to an otorhinolaryngologist is indicated if
• Audiometry. To document the presence and patterns of auditory or vestibular symptoms are triggered by pressure
hearing loss. changes (barotrauma or Valsalva maneuver), a feature
• Brainstem auditory evoked potentials (see p.42). These which suggests the possibility of a perilymph fistula.
have traditionally been used to exclude VIIIth nerve and
brainstem pathology, however MRI is now the gold Migraine (see p.95)
standard. • Anti-migraine therapy can lead to a dramatic relief in
• Other tests of vestibular function, such as caloric and some cases. In otherwise unexplained cases of recurrent
rotational chair testing, may be indicated. vertigo, an empirical trial of prophylactic migraine therapy
• Otorhinolaryngologic (ear, nose and throat) assessment. (e.g. pizotifen (pizotyline), propanolol and verapamil) is
This should be considered if there are significant auditory often indicated, particularly if there is a history of
signs (hearing loss, tinnitus pressure or aural fullness, headache.
particularly if asymmetric), or signs of suppurative middle • The vertiginous aura of migraine does not respond to
disease. interval therapy with drugs such as ergotamine or the
triptans.
CLINICAL COURSE
Sudden unilateral loss of vestibular function usually causes Ménière’s disease (see p.111)
acute, severe vertigo that persists for hours to days. Even • Can be problematic to treat.
without recovery of the underlying vestibular deficit, • Salt restriction and diuretic therapy are the major medical
resolution of the severe disabling symptoms occurs as a treatment modalities.
result of equilibration of the tonus in the brainstem • Betahistine, a vasodilator, can also be helpful. It has been
vestibular nuclei via the CNS process known as compensation. proposed that this medication improves blood flow to
During this recovery period the spontaneous symptoms the inner ear though the exact mechanism of action
resolve, but patients may continue to complain of short- remains unclear.
lived episodes of vertigo induced by head or body motion. • Surgery is sometimes indicated when medical therapy
Persistent motion-induced symptoms following an acute fails.
vestibular insult reflect incomplete central compensation. In – The rationale for endolymphatic sac decompression is to
contrast, recurrent spontaneous episodes of vertigo indicate reduce the pressure in the endolymph compartment.
an unstable vestibular lesion resulting from active underlying – Labyrinthectomy or vestibular nerve section is often
pathology. Such conditions demand specific medical or performed when sac surgery fails, but is reserved for
surgical treatment. unilateral cases.
– More recently, local gentamicin ablation via a series of
TREATMENT injections into the middle ear cavity, has proved to be a
General useful and less invasive management option.
• Reassure the patient that, although the symptoms are
severe, the underlying cause is often not dangerous. Multi-sensory dizziness or disequilibrium
• Avoid sedative and vestibular suppressant drugs which
may exacerbate the problem.
• Balance rehabilitation programs may improve function.
110 Vertigo

BENIGN PAROXYSMAL DIFFERENTIAL DIAGNOSIS


POSITIONAL VERTIGO • Post head trauma vertigo.
• Ménière’s disease.
• Vestibular neuronitis.
DEFINITION • Migraine.
Episodic rotational vertigo of brief duration induced by • Complex partial seizure.
head movement. • Vertebrobasilar ischemia.

EPIDEMIOLOGY INVESTIGATIONS
• Incidence: at least 10 per 100 000 per year. One of the Electronystagmography shows a reduced vestibular response
most common vestibular conditions, accounting for on the affected side.
more than 25% of presentations in patients with
peripheral vestibular disorders. DIAGNOSIS
• Age: occurs predominantly in older age groups with a This is usually a clinical diagnosis. The Dix–Hallpike maneuver
mean age of onset of 55 years. will induce, after a latent interval of 1–2 seconds, vertigo and
• Gender: men and women are affected equally. mixed torsional and vertical nystagmus will be observed. The
nystagmus and vertigo increase and then decline over a period
ETIOLOGY AND PATHOPHYSIOLOGY of 10–20 seconds. If the Dix–Hallpike test is repeated the
Most cases in the elderly are due to cupulolithiasis of the vertigo and nystagmus are reduced (i.e. fatiguability). Tests such
posterior semicircular canal. Degenerated otoconial deposits or as electronystagmography are used only occasionally.
crystals, which are thought to arise from the utricular matrix,
form a heavy mass of sediment in the posterior semicircular canal TREATMENT
and, in response to a provocative head movement, result in Acute episodes
excessive displacement of the sensory organ, or cupula, via a • Meclizine 25–50 mg 3 times/day.
plunger effect. A predisposing cause is identified in a small num- • Diazepam 5–10 mg 1–3 times/day.
ber of patients: head trauma: 15%; vestibular neuronitis: 18%. • Anti-emetics such as prochlorperazine 25 mg suppository.

CLINICAL FEATURES Recurrent episodes


• Brief episodes of rotational vertigo occur associated with Physical exercise therapy: The principle of this treatment is to
specific head positions such as head extension, lying have the patient repeat head movement maneuvers in order to
down or turning over in bed. induce adaptation of the symptoms. The repeated head
• There is a latency of up to 45 seconds before symptoms movement is thought to induce loosening and dispersion of the
begin. sediment in the cupula of the posterior semicircular canal. One
• Vertigo lasts for less than a minute. form of physical therapy described in 1988 (the liberatory
• Symptom fatiguability occurs with repeated head movement. maneuver) involves moving the seated patient quickly from
• Vertical-torsional nystagmus accompanies the vertigo. sitting to lying with the affected ear down, then quickly over so
• Dix–Hallpike maneuver. the other ear is down, and then back to the sitting position.
Another form of therapy (the canalith repositioning
In the Dix–Hallpike maneuver the head of the sitting patient procedure) involves a 5-position cycle in which the patient’s
is tipped back and rotated 45° to one side, and the patient is head is moved about in such a way as to displace theoretically
moved briskly to the supine position so that the head remains any loose material in the posterior semicircular canal into the
at about 45° below the horizontal. It has been traditionally utricle of the vestibular labyrinth. The first step is to move
taught that the head is positioned over the edge of the couch, the patient from a sitting to a reclining position, and extend
but it is easier to perform and more comfortable for the patient the patient’s head over the end of the table at a 45° angle.
if the head is positioned over the back of a pillow. The patient is The second step is to turn the patient’s head to the opposite
instructed to report any vertigo and to keep their eyes open. side. The next step is to roll the patient over onto that side.
After a short latent period of up to 5 seconds or more, the The head is slightly angled while the patient is looking down
patient with a positive test complains of vertigo and manifests at the floor. The fourth step is to return the patient to a
nystagmus which is a mixed upbeating and torsional nystagmus sitting position. The final step is to tilt the patient’s chin
that fatigues up to 30 seconds later. The patient is then moved down. The time spent in each position is equal to the latency
quickly to the sitting position with the head kept in the same plus the duration of the positional nystagmus observed
plane. The nystagmus may reverse in this position. The during the initial diagnostic Dix–Hallpike maneuver. The 5-
maneuver is then repeated with the head turned 45° in the position cycle is repeated until no positional nystagmus is
opposite direction. The vertigo and nystagmus is often less elicited during any of the position changed or until a total of
marked with repeated maneuvers (habituation). five cycles has been performed.
Nystagmus that does not have all of these characteristics A surgical procedure to plug, and effectively paralyse, the
may be seen in unusual variants of benign positional vertigo offending canal is available but is rarely required.
involving semicircular canals other than the commonly
involved posterior canal, but can also be a rare presenting CLINICAL COURSE AND PROGNOSIS
feature of central lesions in the posterior fossa (e.g. non- Spontaneous resolution occurs in most cases. Episodes may
fatiguing downbeating nystagmus). A positive Dix–Hallpike last only a few days, but some patients can experience
maneuver requires the presence of a typical nystagmus recurrent episodes for 2 months or more. In some cases,
response. The complaint of dizziness without associated episodes occur recurrently for more than a year, and in
nystagmus during positional testing is non-specific in nature. others it can persist chronically.
Ménière’s Disease 111

VESTIBULAR NEURONITIS MÉNIÈRE’S DISEASE

DEFINITION DEFINITION
Acute onset of a usually singular episode of vertigo without • Recurrent episodes of severe vertigo associated with
associated deafness or tinnitus. It is also known as acute unilateral hearing loss and tinnitus, with spontaneous
peripheral vestibulopathy, and vestibular neuritis. recovery within hours or days.
• Also known as endolymphatic hydrops.
EPIDEMIOLOGY
• Age: usually occurs in the fourth to sixth decades of life. EPIDEMIOLOGY
• Gender: M=F. • Age: most frequent in the fifth decade of life.
• Gender: M=F..
ETIOLOGY AND PATHOPHYSIOLOGY
• The aetiology is unknown, but is assumed to be viral in ETIOLOGY AND PATHOPHYSIOLOGY
most cases. An increase in the volume of the endolymphatic fluid in the
• A reactivation of herpes simplex virus type 1 (HSV-1) semicircular canals. Most cases are idiopathic, but some
infection is likely in some cases, based on recent studies cases are attributed to acoustic trauma, congenital inner ear
which have detected HSV-1 DNA in about 60% of abnormalities, syphilis, or vascular disorders associated with
human vestibular ganglia and nuclei. The various patterns hypertension or diabetes.
of HSV-1 infection of vestibular structures are compatible
with virus migration from the vestibular ganglia to the CLINICAL FEATURES
vestibular nuclei and from the ipsilateral to the • Sudden onset of intense vertigo which may take many
contralateral vestibular nucleus via commissural fibers. hours to resolve.
• Unilateral tinnitus and decreased hearing associated with
CLINICAL FEATURES a sensation of fullness and increased pressure in the ear.
• Vertigo that develops suddenly or over several hours • Horizontal-torsional nystagmus beating toward the
associated with nausea, vomiting and ataxia. Vertigo is affected ear is observed.
aggravated by any head movement. • There may be a previous history of recurrent vertigo
• Torsional nystagmus to the side opposite the lesion is accompanied by fluctuating auditory symptoms (tinnitus,
observed. hearing loss and aural fullness). If so, the hearing may
• Hearing is preserved. have recovered between attacks but there is usually a
stepwise or gradual loss of hearing, classically involving
DIFFERENTIAL DIAGNOSIS the low frequencies in the early stages.
• Ramsay Hunt syndrome (herpes zoster infection of the • Drop attacks (otolithic crises of Tumarkin) can occur in
VIIIth cranial nerve). uncontrolled cases.
• Post head trauma vertigo. • Vestibular hydrops (recurrent vertigo without auditory
• Drug-induced vertigo, e.g. aminoglycosides. accompaniments) has been described, but this rarely
• Internal auditory artery ischemia: may also cause a similar occurs as an isolated phenomenon for prolonged periods.
syndrome, particularly in older age groups. Many chronic cases of so called ‘vestibular hydrops’ have
• Acute labyrinthitis: suspect when vertigo is accompanied other causes such as migraine.
by prominent auditory symptoms. This can occur in its
bacterial form as a complication of suppurative middle DIFFERENTIAL DIAGNOSIS
ear disease or as a result of viral infection. • Post head trauma vertigo.
• Benign paroxysmal positional vertigo.
INVESTIGATIONS • Migraine.
Electronystagmography shows a reduced vestibular response • Complex partial seizure.
on the affected side. • Vertebrobasilar ischemia.
• Cogan’s syndrome.
DIAGNOSIS
• Usually a clinical diagnosis. The Dix–Hallpike maneuver INVESTIGATIONS
will induce vertigo and torsional nystagmus will be • Audiometry shows low frequency hearing loss initially,
observed. with high frequency hearing loss occurring later.
• Tests such as electronystagmography are used only • Electronystagmography shows a reduced vestibular
occasionally. response on the affected side.

TREATMENT DIAGNOSIS
• Meclizine 25–50 mg 3 times/day. The diagnosis is primarily clinical with supportive evidence
• Diazepam 5–10 mg 1–3 times/day. from audiometry and electronystagmography.
• Anti-emetics such as prochlorperazine 25 mg suppository.

CLINICAL COURSE AND PROGNOSIS


Gradually resolves over a period of days to weeks in most
cases. Less than 10% of cases experience recurrent episodes.
112 Vertigo

TREATMENT • Medical labyrinthectomy using aminoglycoside drugs


Acute episodes given either systemically or intratympanically, can be used
• Meclizine 25–50 mg 3 times/day. in patients with either unilateral or bilateral disease.
• Diazepam 5–10 mg 1–3 times/day. • Sectioning of the VIIIth nerve on the affected side.
• Anti-emetics such as prochlorperazine 25 mg supposi- • Shunting procedure from the semicircular canals to the
tory. mastoid region and subarachnoid space.
• Salt restriction and diuretic therapy have been used to try
and prevent attacks, but there is no conclusive evidence CLINICAL COURSE AND PROGNOSIS
of their efficacy. • Recurrence of attacks is a hallmark of the disease.
• Many patients experience decremental hearing loss with
For severe cases recurrent episodes. Initially hearing loss in the low
For severe cases with recurrent disabling episodes unrelieved frequencies is observed. As the disease progresses, high
by medical therapy there are a number of options: frequency hearing loss is seen.
• Surgical labyrinthectomy is usually reserved for strictly • Contralateral involvement occurs in 20–30% of cases.
unilateral cases.

FURTHER READING BENIGN PAROXYSMAL POSITIONAL MÉNIÈRE’S DISEASE


VERTIGO Saeed SR (1998) Diagnosis and treatment of
Froehling DA, Bowen JM, Mohr DN, et al. Ménière’s disease. BMJ, 316: 368–72.
VERTIGO (2000) The canalith repositioning procedure Vesterager V (1997) Tinnitus – investigation and
Brandt T, Bronstein AM (2001) Cervical vertigo. for the treatment of benign paroxysmal posi- management. BMJ, 314: 728–731.
J. Neurol. Neurosurg. Psychiatry, 71: 8–12. tional vertigo: a randomised controlled trial.
Halmagyi GM, Cremer PD (2000) Assessment Mayo Clin. Proc., 75: 695–700.
and treatment of dizziness. J. Neurol. Neuro- Furman JM, Cass SP (1999) Benign paroxysmal
surg. Psychiatry, 68: 129–136. positional vertigo. N. Engl. J. Med., 341:
Neuhauser H, Leopold M, von Brevern M, et al. 1590–1596.
(2001) The interrelations of migraine, verti-
go, and migrainous vertigo. Neurology, 56: VESTIBULAR NEURONITIS
436–444. Arbusow V, Strupp M, Wasicky R, et al. (2000)
Sloan PD, Coeytaux RR, Beck RS, Dallara J Detection of herpes simplex virus type 1 in
(2001) Dizziness: state of the science. Ann. human vestibular nuclei. Neurology, 55:
Intern. Med., 134: 823–832. 80–882.
Chapter Six 113

Movement Disorders
DYSTONIA ETIOLOGY
Dystonia is a symptom or sign of an inherited or acquired
brain disease, that usually involves the basal ganglia; it is not
DEFINITION a psychologic disturbance.
A syndrome of intermittent or continuous sustained muscle
contractions, frequently causing twisting and repetitive Inherited primary or idiopathic dystonias
movements or abnormal postures of virtually any part of the • Early-onset primary torsion dystonia (generalized):
body. – Autosomal dominant inheritance.
– Sporadic.
EPIDEMIOLOGY – X-linked.
Common, but the correct diagnosis is frequently delayed. • Adult-onset primary dystonia (focal).
• Incidence – focal dystonia: 1 (0.1–4)/100 000/year. • Dopa-responsive dystonia (Segawa disease).
• Prevalence (per million population): 391.
– Generalized dystonia: 62. Acquired secondary or symptomatic dystonias
Idiopathic: 34 (USA); 50 (Israel – Ashkenazi Jews). Drugs
Symptomatic: 28. • Dopamine receptor-blocking (antipsychotic/neuroleptic)
– Focal dystonia: 329. drugs: acute dystonic reactions and tardive dystonias.
Idiopathic: 296. • Dopaminergic (antiparkinsonian) drugs: dystonic dyskinesias.
Cranial dystonia (86). • Anti-epileptic drugs.
Spasmodic dysphonia (52). • Antidepressants.
Spasmodic torticollis (89).
Writer’s cramp* (69). Metabolic disorders
Symptomatic 33. • Wilson’s disease.
• Mitochondrial cytopathy.
* Writer’s cramp appears the most common in clinical • Leigh disease.
neurologic practice, and may have been underdiagnosed in • Glutaric aciduria.
prevalence studies. • Methylmalonic acidemia.
• Homocystinuria.
• Age: any age. • Metachromatic leukodystrophy.
• Gender: M=F. • Pelizaeus–Merzbacher disease.
• GM1 and GM2 gangliosidosis.
PATHOLOGY • Neuronal ceroid lipofuscinosis.
No consistent pathological changes have been identified in • Juvenile dystonic lipidosis.
primary dystonia, but when focal brain lesions are identified, • Lesch–Nyhan syndrome.
the basal ganglia (particularly the putamen) or rostral
midbrain are usually involved. Other neurodegenerative disorders
• Huntington’s disease.
PATHOPHYSIOLOGY • Parkinson’s disease.
One of the main neurophysiologic mechanisms of dystonia • Progressive supranuclear palsy.
is defective control of inhibitory interneurones in the • Multiple system atrophy.
cerebral cortex, brainstem and spinal cord. Functional • Hallevorden–Spatz disease.
imaging and animal model studies suggest reduced • Progressive pallidal degenerations.
inhibition of cortical motor output leads to the generation • Neuroacanthocytosis.
of excess motor activity during voluntary movement. Other • Ataxia telangiectasia.
studies have identified abnormal activation of group 1a • Familial basal ganglia calcification.
inhibitory interneurones in the spinal cord, with loss of
normal reciprocal inhibition, co-contraction of agonists and Cerebral palsy
antagonists, and tonic or phasic co-contraction. Genetic • Kernicterus.
studies have reaffirmed that dystonia is mostly inherited as • Perinatal brain injury.
an autosomal dominant condition with reduced penetrance. • Brain malformations.
A number of families with generalized and focal dystonia
have been described, and at least 12 different genotypes Trauma
identified to date. • Head trauma.
• Peripheral (limb and spine) trauma.
114 Movement Disorders

Infections • Adult-onset dystonia: usually sporadic; an underlying


• Viral encephalitis. cause in about 10% of patients; dystonia usually starts in
• Cerebral toxoplasmosis. body parts other than the legs and frequently remains
focal or segmental; rapid progression.
Focal basal ganglia lesions
• Stroke. Distribution of dystonia
• Brain tumor. • Typically, dystonia begins in one part of the body as a
• Arteriovenous malformation. focal action dystonia. The legs are commonly affected
• Toxins (exposure to manganese, carbon disulfide). first in children (onset before 13 years of age), less
commonly in adolescents (onset 13–20 years of age), and
Toxins very rarely in adults (onset after age 20 years). The
• Manganese. dystonia progresses to a multifocal or generalized
• Carbon disulfide. dystonia in about 60% of children, 35% of adolescents,
• Wasp sting. and 3% of adults.
• Hemidystonia: any age; an underlying pathologic cause
The chances of identifying an underlying pathologic can be identified in over 80% of cases (usually a
cause for dystonia depends on the age of onset and the structural basal ganglia lesion such as stroke, tumor or
distribution of the dystonia (see below). AVM).
• Segmental, multifocal or generalized dystonia: commonly
CLINICAL FEATURES inherited; an underlying cause in about 45% of patients.
Dystonia • Focal dystonia: underlying cause found in less than 10%.
Muscle contractions that are prolonged (often twisting a
part of the body into characteristic postures), and Etiology of dystonia
commonly intermittent (causing repetitive, often rhythmic Drug-induced dystonia
jerks into dystonic postures) but are sometimes continuous, Neuroleptic antipsychotics and anti-emetics produce two
causing constant twisted postures. types of dystonic reaction: acute dystonic reactions and
Initially, movements and postures appear only during tardive dystonias.
specific movements (action dystonia) and subside with rest. • Acute dystonic reactions or oculogyric crises (see below)
Voluntary movements are accompanied by ‘overflow’ of follow exposure to D2 receptor antagonists (half of cases
muscle spasms and dystonic postures spreading to adjacent occurring within 48 hours, and 90% within 5 days of
muscle groups not normally recruited in the task. If the exposure) and most commonly in young men. The
syndrome progresses (as is common with children but rare in dystonia particularly affects the eyes, jaws, neck and
adults), the initial action dystonia becomes evident at rest and trunk. Pre-treatment with anticholinergics reduces the
then spreads to affect other parts of the body. Dystonia is risk of oculogyric crises. Oculogyric crises resolve with
therefore usually considered a ‘mobile’ movement disorder anticholinergics and withdrawal or reduction of the
because fixed postures and persistence of dystonia at rest occur offending drug.
only in severe, advanced dystonia and hemiplegic dystonia. • Tardive (‘delayed’) dystonia occurs in 1.5–2% of patients
Associated rhythmic postural tremors and myoclonus receiving long term neuroleptic therapy. Less frequently,
(jerky movements), at about 3 Hz or at faster rates, similar levodopa, dopamine antagonists and selective serotonin
to those of benign essential tremor, are superimposed on reuptake inhibitors may also produce dystonia. The latency
the dystonic postures in many patients. Combinations of between drug exposure and onset of dystonia ranges from
these movements frequently lead to misdiagnosis. 3 weeks to 40 years, with 20% occurring in the first year.
Spasms are relieved by ‘sensory tricks’ or ‘gestes Tardive dystonia most commonly affects the cranial, neck
antagonistique’, in which tactile stimuli applied to or near and trunk muscles. Paradoxical dystonia (worse at rest and
the affected body part relieve the muscle spasm. Movements relieved by movement) may occur. Treatment is based on
also disappear during rapid eye movement sleep and deep withdrawal of the offending drug if possible. Remission
(stage three and four) sleep. rates are low (10–20%) and may take up to 5 years after
drug withdrawal. Treatment with tetrabenazine and
HISTORY anticholinergics is helpful in about half of cases.
The clinical features (and course) are determined by the
patient’s age, anatomic distribution of the dystonia and its Past history of relevance
cause. • Abnormal perinatal history.
• Delayed milestones.
Age of onset • Precipitating illness.
• Childhood-onset: commonly inherited and usually • Seizures.
autosomal dominant; an underlying pathologic cause can
be identified in up to about 40% of cases; dystonia usually Family history of dystonia
starts in the feet and legs, with a disorder of gait, and
progresses to become generalized or multifocal; speech PHYSICAL EXAMINATION
tends to be involved early. It progresses slowly over about Physical signs which may be a clue to an underlying cause
10 years from a focal action dystonia to a generalized of secondary dystonia:
dystonia. • Intellectual impairment.
• Adolescent-onset: an underlying cause in 30% of patients; • Defects of vision, hearing, or sensation.
intermediate progression. • Kayser–Fleischer rings (118).
Dystonia 115

• Abnormalities of optic fundi or eye movements. • Abductor spasm of vocal cords: whispering dysphonia
• Loss of postural reflexes. (rare).
• Pyramidal, cerebellar, or sensory signs. • Spasm false vocal cords: inspiratory breathing dystonia.
• Hepato-splenomegaly.
Writer’s cramp (arm dystonia)
SPECIAL SYNDROMES • Writing provokes stiffness, spasm or tremor of digits and
Early onset primary torsion dystonia (generalized) hand, with overflow of muscle activity to proximal arm
• Autosomal dominant inheritance. muscles.
• GAG deletion in DYT1 gene on chromosome 9 (9q34) • Resolves completely with cessation of writing.
which codes for the ATP-binding protein torsin A. • May affect other fine manual tasks in 25% of cases.
• Penetrance: about 30-40%.
• Expression of dystonia in relatives: highly variable. Dopa-responsive dystonia with diurnal variation
• Age of onset: 12.5 years (mean), 4–44 years (range). (Segawa’s disease)
• Onset: in one limb in 90% of patients; it is rare to begin • Autosomal dominant inheritance (5–10% of inherited
in the neck or larynx unless the onset is in adulthood. childhood dystonias) with reduced penetrance.
The dystonia spreads to the trunk but rarely affects • Gene defects: several autosomal dominant mutations in-
cranial muscles. A past or family history of an action volving the gene GCH1 coding for the enzyme guanine
tremor is common. triphosphate (GTP) cyclohydrolase I on chromosome
14q. The enzyme is involved in the synthesis of tetrahy-
Adult-onset focal dystonia syndromes drobiopterin (BH4) in the metabolic pathway manu-
Spasmodic torticollis (cervical dystonia) facturing dopamine and serotonin. The clinical picture
• Torsion or tilting of the head due to sustained results from dopamine deficiency. Other genetic defects
contraction of neck muscles (119). include autosomal recessive mutations in the gene for
• Associated tremulous or jerky head movements are tyrosine hydroxylase, which also interrupts dopamine
common. synthesis.
• Neck pain is common, affected muscles may • Children and adolescents affected.
hypertrophy. • F>M: 3:1.
• Arm or oromandibular dystonia and blepharospasm • Lower limb dystonia: usually starts in the legs as an
occur in a small percentage of patients. equinovarus deformity causing walking difficulties.
• Fluctuates diurnally, being less in the morning and be-
Cranial dystonia comes worse as the day goes on, or becomes noticeable
Blepharospasm: in the afternoon and evening, particularly with exercise.
• Bilateral orbicularis oculi muscle spasm results in inability • Other features: postural instability; later progression to
to maintain eye opening. a parkinsonian syndrome.
• Sometimes associated with cervical or oromandibular • Responds to small doses of levodopa.
dystonia. • Many cases are misdiagnosed as ‘cerebral palsy’.

Oromandibulfacial dystonia (Meige syndrome)


• Forceful opening, closure or lateral deviation of the jaw.
• Closure, pursing or retraction of lips.
• Protrusion of tongue.

Spasmodic dysphonia (laryngeal dystonia) 119


• Adductor spasm or vocal cords: strained speech, glottal
stops and variation of voice pitch.

118

118 Typical appearance of a corneal Kayser–Fleischer ring in Wilson’s 119 Photograph of a patient with focal cervical dystonia (spasmodic
disease (arrows). torticollis). (Courtesy of Dr AM Chancellor,Tauranga, New Zealand.)
116 Movement Disorders

Paroxysmal kinesigenic dystonia • Laryngeal muscles: laryngeal dystonia (spasmodic


• Autosomal dominant inheritance. dysphonia): vocal cord adduction, which gives rise to a
• Childhood-onset. strangled speech, or vocal cord abduction, causing a
• Brief (seconds to minutes) attacks of dystonia (or chorea) whispering dysphonia.
precipitated by sudden movement, that occur several • Sternocleidomastoid, posterior neck, and indeed all
times a day. cervical muscles: cervical dystonia (spasmodic torticollis).
• Consciousness is preserved. • Hand, forearm and arm: brachial dystonia (writer’s
• Anti-epileptic drugs, such as carbamazepine and cramp and other occupational cramps).
phenytoin, are effective. • Back and trunk: axial dystonia (causing scoliosis, lordosis,
kyphosis, or tortipelvis).
Paroxysmal non-kinesigenic dystonia • Foot, leg and thigh: foot and leg dystonia.
• Autosomal dominant inheritance.
• Childhood-onset. Axial dystonia
• Infrequent episodes of dystonia without loss of Confined to neck or back.
consciousness, which are not precipitated by movement,
and which last many minutes to hours. Segmental dystonia
• Clonazepam is sometimes effective. Involves ≥2 adjacent body parts. For example:
• Cranial dystonia (Meige or Brueghel’s syndrome):
Myoclonic dystonia combination of blepharospasm and oromandibular and
• Autosomal dominant inheritance. lingual dystonia.
• Childhood-onset. • Craniocervical dystonia (cranial dystonia and spasmodic
• Dystonia combined with fast myoclonic jerks. torticollis).
• Alcohol relieves the myoclonus but has less effect on the • Craniocervical dystonia with dystonic arms.
dystonia. • Oromandibular dystonia.

Acute dystonic reactions Hemidystonia


• Uncommon. Confined to >1 body part (i.e. arm and leg) on the same
• Tends to affect children and adolescents more than adults. side; uncommon, caused by a structural lesion of the
• Usually drug-induced: occurs within a few hours (or less contralateral basal ganglia.
often, days) after one or a few normal doses of:
phenothiazines (e.g. prochlorperazine, chlorpromazine); Multifocal dystonia
butyrophenones (e.g. haloperidol); metoclopramide; ≥2 non-contiguous parts (e.g. eyes, neck, and one leg):
antihistamines; tetrabenazine; chloroquine; amodiaquine. • Dystonia responsive to levodopa.
• Dystonia of head and neck muscles causing neck • Myoclonic dystonia responsive to alcohol.
retraction, opisthotonos, trismus, tongue protrusion,
spasmodic torticollis, choking and oculogyric crisis. Generalized dystonia
• Can be painful and is frightening. Whole body, or most of it including all limbs and axial muscles.
• A family history of similar reactions may be present.
• Fever and tachycardia may be present. Paroxysmal dystonia
• Differential diagnosis includes tetanus, but acute dystonia • Paroxysmal kinesigenic dystonia responsive to anti-
often resolves in 1 or 2 hours or following i.v. epileptic drugs.
anticholinergic (e.g. benztropine 1–2 mg [lower dose in • Paroxysmal non-kinesigenic dystonia responsive to
children] or diazepam [5–10 mg i.v.]). clonazepam.
• A second exposure to the offending drug may be well
tolerated. According to etiology (see Etiology, p.113)

CLASSIFICATION DIFFERENTIAL DIAGNOSIS


Dystonia may be classified according to age at onset, (see Etiology, p.113)
anatomic distribution, or cause (primary, inherited, or Other movement disorders
secondary, symptomatic dystonia). • Wilson’s disease (see p.157).
• Huntington’s disease (see p.417).
According to age of onset • Parkinson’s disease (see p.420).
• Early onset primary torsion dystonia • Gilles de la Tourette’s syndrome (see p.130).
• Adult onset focal dystonia • Essential tremor (see p.119).
• Tardive dyskinesia (see p.129):
According to anatomic classification – Continuous involuntary choreiform movements of the
Focal dystonia orobuccolingual and masticatory muscles.
Confined to only one part of the body. Examples include: – Present in about 20% of patients on long term
• Orbicularis oculi and neighboring facial muscles: antipsychotic medication.
blepharospasm (involuntary spasms of eye closure). – Dopamine receptor hypersensitivity is probably responsible.
• Jaw and mouth: oromandibular dystonia. – Difficult to treat; withdrawal of the responsible
• Tongue: lingual dystonia. antipsychotic drug may induce a remission, but this can
• Pharyngeal muscles: dystonic dysphagia. take months or even years, and may be impractical if the
patient has a serious underlying psychiatric disorder.
Dystonia 117

– Increased doses of dopamine antagonists (e.g. INVESTIGATIONS (see Table 16)


tetrabenazine) paradoxically may lead to improvement, • Exclude Wilson’s disease (see p.157) if onset is before
presumably by depleting presynaptic dopamine, but may age 50 years: screen with slit lamp examination of the
also cause depression. eyes and serum ceruloplasmin. If any doubt, perform a
– Baclofen, benzodiazepines, and calcium-channel blockers liver biopsy to measure the copper content.
may be effective; anticholinergic make tardive dyskinesia • MRI of the brain if suspect basal ganglia structural lesions
worse. (i.e. stroke, post-traumatic hemorrhage, tumor, AVM, or
• Psychological movement disorders. metabolic or degenerative disorders) (120, 121).
• Blood or genomic DNA: PCR-RFLP of GAG deletion
in DYT1 dystonia gene on 9q34 (e.g. 128 bp fragment).

Table 16 Investigation of patients with dystonia 120


Idiopathic dystonia Symptomatic
dystonia
Onset in Onset in Onset at
childhood or adult life any age
adolescence
Blood
Full blood count and erythrocyte
sedimentation rate + + +
Urea and electrolytes, glucose, liver
function, calcium, phosphate + + +
Ceruloplasmin concentration + + +
Syphilis serology + + +
Antinuclear antibodies + +
Acanthocytes (neuroacanthocytosis) + +
Creatine kinase +
Uric acid +
Hypoxanthine-guanine
phosphoribosyltransferase activity +
Amino acid concentration +
White blood cell lysosomal enzymes +
Alpha-fetoprotein + 121
DNA analysis + + +
Urine
Amino acids (concentration) +
Organic acids +
Oligosaccharides +
Mucopolysaccharides +
24 hour copper excretion +
Cerebrospinal fluid + +
Biopsy
Skin +
Muscle (ragged red fibers of
mitochondrial cytopathy) +
Bone marrow +
Other
Slit lamp examination of the eyes
(Kayser–Fleischer rings) + + +
CT or MRI scan of the brain + + +
Nerve conduction studies and EMG + +
120, 121 Cranial CT scan (120) and MRI T2W
EEG + +
image (121) of a patient with right hemidystonia
Electroretinography +
showing a slit-like area in the left putamen (low
Evoked potentials + +
density on CT and high density on MRI, arrows)
* Wilson’s disease should be excluded in all patients with symptom onset representing necrosis due to previous hypertensive
before the age of 50 years hemorrhage.
118 Movement Disorders

DIAGNOSIS Adult-onset focal dystonia


• Is it dystonia? Clinical (see above). • Anticholinergic agents: effective in about one in five
• Is there a cause for the dystonia? Etiology, clinical and people. Large doses (40–120 mg benzhexol [trihexy-
laboratory investigations (see Table 16, above). phenidyl] daily) may be required and a long latent
interval of weeks to months may elapse before improve-
TREATMENT ment occurs.
Most dystonias can be controlled to some extent, often after • Botulinum toxin A (BTX-A), injected in small doses into
a trial of several medications (as with the treatment of the dystonic muscle(s), with or without EMG guidance,
migraine), but their underlying cause cannot often be cured. is effective within 7–10 days in 80–90% of patients with
focal dystonias such as blepharospasm, spasmodic
General torticollis, and spasmodic dysphonia. It is taken up by
• Physiotherapy: helps prevent dystonic muscles developing peripheral nerve terminals and prevents calcium-
contractures. dependent release of acetylcholine, resulting in chemical
• Speech and occupational therapy: provide aids to denervation and paralysis of the injected muscle, and
communication and mobility. Mechanical braces are reduction or abolition of the dystonic spasms. The effect
usually not helpful and may lead to problems such as skin lasts for about 2–4 months when terminal sprouting
ulceration as the dystonic spasms fight the constraint. restores muscle end plate neurotransmission. Up to 5%
• Psychotherapy (supportive): often useful for this of initially responsive patients may subsequently not
distressing condition. respond, due to incorrect storage of the toxin,
• Psychiatric assessment and intervention: if secondary underdosing, injection of inappropriate muscles, a
depression occurs. worsening or change in the pattern of dystonia (possibly
with involvement of deep, inaccessible muscles),
Specific development of contractures, altered perception of
Childhood- and adolescent-onset segmental, multifocal and response, and the development of immunity. Immunity
generalized dystonia is associated with very frequent injections, ‘booster’
• Levodopa 100 mg and carbidopa 25 mg, one tablet injections, and higher doses. Higher doses of BTX-F may
twice daily. Build up to two tablets three times daily for be effective with few adverse effects.
3 months. If responsive, continue long term. N.B. • Facial nerve section is a possible treatment of last resort
Adverse effects of long term levodopa that are seen in for blepharospasm in those in whom anticholinergic
Parkinson’s disease do not occur. drugs and botulinum toxin are ineffective and who are
• Anticholinergic agents: if no response to levodopa, start prepared to have a permanent facial palsy, but it is
benzhexol (trihexyphenidyl) in low dose (2.5 mg twice fortunately hardly ever done. Chemical myotomy is
daily) and increase slowly (by 2.5 mg a day every 7–14 another alternative.
days) up to a dose that is effective or causes intolerable • Peripheral nerve or root section for torticollis is rarely
adverse effects such as dry mouth, blurred vision, urinary indicated.
hesitancy, anorexia, weight loss, forgetfulness, confusion, • Thalamotomy for stable hemidystonia can be useful but
behavioral change, hallucinations and chorea. Young bilateral thalamotomy for generalized dystonia is rarely
people may tolerate doses up to 180 mg a day. About contemplated, mainly because of the risk of
half of patients benefit from anticholinergics. compromising speech.
• Benzodiazepines (e.g. diazepam), often in high dose, • Pallidotomy and pallidal stimulation are currently being
seem to relax dystonic spasms. evaluated in the treatment of dystonia with encouraging
• Baclofen, orally in high dose, or intrathecally in much results.
lower doses.
• Carbamazepine. Curative
• Dopamine receptor antagonists and neuropletics such as Identify, treat and eliminate the underlying cause.
phenothiazines, haloperidol (2–6 mg/day), tetrabenazine
and pimozide (alone or in combination with NATURAL HISTORY
anticholinergic drugs or benzodiazepines) may dampen • Primary (idiopathic) dystonia of childhood- or
down excessive movements by causing a degree of drug- adolescent-onset tends to progress in the first 5 years of
induced parkinsonism but they may also lead to tardive the illness, more slowly over the next 5 years and very
dyskinesia. Tetrabenazine, which depletes the brain of slightly, if at all, in adulthood.
dopamine, carries less risk of inducing tardive dyskinesia • Idiopathic adult-onset focal dystonia does not tend to
but it may cause depression. progress.
• Risperidone (1.5–3 mg/day), a neuroleptic with high • About 1 in 20 (5%) patients with any form of idiopathic
affinity for 5-HT2 receptors, may be effective in patients dystonia may experience a spontaneous improvement or
with idiopathic segmental dystonia partly insensitive to even resolution. This is most likely in the first 5 years of
haloperidol. the illness. Although subsequent relapses are common,
• Combination of low dose tetrabenazine (25 mg three some patients experience remissions lasting years or
times daily), pimozide (gradually increasing the dose up decades.
to 12 mg daily) and benzhexol (trihexyphenidyl) • The estimated risk for children or siblings of familial cases
(gradually increasing to the maximum tolerable dose). developing clinical dystonia is about 20%; the risk for
sporadic cases is lower at about 8–14%.
Essential Tremor (ET) 119

ESSENTIAL TREMOR (ET) • It is thought that the cerebellum introduces an error in


the timing of muscle bursts during voluntary movement,
and repeated corrective movements lead to tremor.
DEFINITION
A low frequency (4–9 Hz) postural tremor which is absent CLINICAL FEATURES
at rest and not associated with the clinical signs of Tremor
parkinsonism or other neurologic deficits. • Postural or action tremor of the finger, hands and
forearms when they are held outstretched against gravity,
EPIDEMIOLOGY or used for specific, usually visually-guided, manual tasks
• Incidence: 8 (95% CI: 4–14)/100 000/year. (‘position-specific postural tremor’).
• Prevalence (lifetime): 0.3–1.7%. • Variable kinetic component (tremor during any form of
• Age of onset: bimodal: young adults (median about 15 movement).
years) and elderly. • 4–12 cycles per second.
• Gender: M=F. • Bilateral, but may be asymmetric.
• Other body parts involved in about half of cases: the
PATHOLOGY head (40%), tongue, lips, voice (15–20%), face, and
No characteristic pathological or biochemical findings. trunk; and postural tremor of lower limbs (15%).
• Visible.
ETIOLOGY • Persistent, but the amplitude may fluctuate.
• Hereditary: autosomal dominant inheritance (50% of • Exacerbated by emotional upset such as excitement and
patients) via a penetrant autosomal dominant gene. The anger.
responsible gene(s) remain unknown. • Improved temporarily by alcohol, in small quantities, in
• Sporadic: probably the same entity as hereditary ET. about half of patients.
• Relatively longstanding (i.e. longer than 5 years).
PATHOPHYSIOLOGY • Froment’s sign (a rhythmic resistance to passive
• Unknown. movements of a limb about a joint when there is a
• A central source of oscillation that is influenced by voluntary action of another body part).
somatosensory reflex pathways.
• Enhanced olivocerebellar oscillation and activation of DIFFERENTIAL DIAGNOSIS (see Tables 17, 18)
cerebellothalamocortical pathways probably have an Intention or ‘terminal’ tremor due to
important role in the generation and transmission of the cerebellar disease
tremor because: • Slower (3–5 cycles per second).
– Functional imaging studies (PET and functional MRI) • Principally in a horizontal plane.
reveal increased olivary glucose metabolism and increased • Not present at rest.
blood flow bilaterally in the cerebellum and red nuclei • Increases with action and progressively increases during
and contralaterally in the globus pallidus, thalamus and a voluntary movement (terminal or intention tremor).
primary sensorimotor cortex of patients with essential • Unlike kinetic tremor, which is tremor during any form
tremor. of movement; ‘intention’ or ‘terminal’ tremor is the
– Lesions of the ipsilateral cerebellum diminish essential pronounced exacerbation of kinetic tremor toward the
tremor. end of goal-directed movement.
– Ventrointermediate thalamotomy or microstimulation • The head may be affected, but usually as titubation
reduce tremor. (repetitive nodding or ‘yes-yes’).
• May be incapacitating.
• Responds to propranolol and diazepam.

Table 17 Common tremors classified according to frequency and behavior


Frequency (Hz) Behavioral characteristics Disease locus Disease processes
2.5–3.5 Postural/kinetic Cerebellar/brainstem Alcoholic degeneration, multiple sclerosis,
trauma
4–5 Rest Substantia nigra/striatum Parkinson’s disease, drugs (neuroleptics,
?MPTP)
Rest/postural/movement/kinetic Red nucleus
Postural/kinetic Cerebellum
5.5–7.5 Postural/kinetic ?Cerebellum Essential tremor, Parkinson’s disease,
?Cerebrum–cerebellum drugs (valproate, lithium, antidepressants)
8–12 Postural/kinetic ?Cerebellum Enhanced physiologic tremor,
essential tremor, drug intoxications
120 Movement Disorders

Table 18 Common tremors classified according to clinical characteristics and distribution


Clinical characteristics Distribution Causes
Rest tremor Limbs, head, jaw Parkinson’s disease
Midbrain disease
Postural tremor Arms (when outstretched) Physiologic tremor
Essential tremor
Dystonic tremor
Cerebellar tremor
Neuropathic tremor
Task-specific action tremor During particular tasks Isolated voice tremor
Jaw tremor
Writing tremor
Intention tremor Approaching target Cerebellar disease
During purposeful movements Midbrain disease
Standing tremor Legs, trunk Orthostatic tremor

Parkinsonian tremor • Drug withdrawal:


• Typically 6 (3–7) cycles per second. Alcohol. Barbiturates.
• Present at rest. Benzodiazepines. Opiates.
• Not increased with posture or action. • Metabolic:
• Involves the limbs and head. Hypoglycemia. Metabolic encephalopathies.
• ‘Pill rolling’ character. Pheochromocytoma. Thyrotoxicosis.
• Exacerbated by emotion. • Exaggerated physiologic response:
• Responds to levodopa and anticholinergic medications. Anxiety. Fatigue.
Fright. Strenuous exertion.
Alcohol withdrawal
• 6–10 cycles per second. Management is directed to correction of the underlying
• Head and limbs involved. medical illness or cessation of any contributing drug. If
• Periodic: appears 8–12 hours after last drink. idiopathic EPT becomes inconvenient or socially
• Responds to diazepam and chlordiazepoxide. embarrassing, propranol may be helpful.

Metabolic derangements, such as thyrotoxicosis Primary orthostatic tremor


• 10–20 cycles per second. • Isolated, high frequency (14 Hz) bilaterally synchronous
• Head and upper limbs involved. tremor of legs and trunk when standing still.
• Symptoms and signs of metabolic disturbance. • May not be detected easily without palpation of the leg
• Responds to correcting metabolic disturbance. muscles.
• Patients may only complain of unsteadiness when
Enhanced physiologic tremor (EPT) standing.
A rapid 8–12 Hz small amplitude, barely visible, postural • Responds to clonazepam and, in some cases,
tremor, typically of the upper limbs, which occurs as a pramipexole; ET does not respond to clonazepam.
normal phenomenon during muscle contraction. EPT
manifests as a postural and kinetic hand tremor. It can be Isolated position-specific or task-specific tremors
difficult to distinguish between EPT and early stages of (including occupational tremors and primary
hereditary ET in a young person. There is no current writing tremor)
reliable way of making this distinction but the family history • Have the appearance of localized ET but the task
can be a clue. specificity of dystonia.
• May respond to anticholinergics.
Causes
• Drugs: Rubral tremor
Adrenergic drugs. Amiodarone. • A severe tremor with features of a parkinsonian rest
Amphetamines. Antipsychotic drugs. tremor and marked exacerbation during movement.
Caffeine. Cimetidine. • Not always associated with lesions of the red nucleus but
Corticosteroids. Cyclosporine A. may result from lesions of the ipsilateral cerebellar
Lithium. Oral hypoglycemics. dentate nucleus or superior cerebellar peduncle in the
Serotonin reuptake inhibitors. midbrain (dentato-rubral tracts).
Sodium valproate. Theophylline. • Most commonly seen in multiple sclerosis.
Thyroxine. Tricyclic antidepressants.
Essential Tremor (ET) 121

Dystonia TREATMENT
• Highly asymmetric postural tremor is probably a form of Not all patients require treatment; only those severely
dystonia (see p.113). affected.
• Isolated voice tremor may be due to laryngeal dystonia
and other dytonias of the vocal apparatus. Medical
Propranolol and primidone have been shown in controlled
Psychogenic trials to afford a partial reduction in tremor amplitude in
• Unphysiologic variations in tremor frequency (>1 Hz). about two-thirds of cases, and are the mainstay of treatment.
• Unusual and inconsistent behavioral characteristics. • Propranolol 10–40 mg mane or bd (up to about
• Spontaneous remissions. 240–320 mg/day), or metoprolol 25 mg twice daily
• Psychiatric or social factors (multiple somatizations, with 25 mg increments to effect or until a maximum of
secondary gain, litigation or compensation pending). 50 mg three times daily, leads to variable improvement.
About half of patients experience a reduction in tremor
DIAGNOSIS amplitude of up to 50%. The effect is greatest on postural
A clinical diagnosis based on the long duration of limb tremor. However, not all patients are helped and
symptoms; positive family history; lack of rigidity, the tremor is seldom abolished. There is little point in
bradykinesia or other neurologic signs; symmetry; and prescribing a nocte dose. Adverse effects and relative
alcohol responsiveness. contraindications for propranolol include heart failure,
bradycardia, hypotension and asthma. The mechanism of
Diagnostic criteria (predominantly for action of propranolol is thought to be blockade of
research purposes) peripheral skeletal muscle β2 receptors. Selective β1
Definite ET blockade is less effective than propranolol.
• Characteristic bilateral tremor on maintaining posture, • Primidone (Mysoline) 50–250 mg daily if necessary, if
typically of the outstretched upper limbs of more than 5 propranolol is ineffective or contraindicated. May be as
years duration. It is absent at rest, not made strikingly worse effective as beta blockers but adverse effects are common.
with movement and is not associated with extrapyramidal Care is required when introducing primidone because of
or cerebellar signs (see Clinical features, above). nausea, sedation and unsteadiness, which may be
• No definite evidence of sudden onset. sufficiently severe to warrant stopping the drug. A low
• No direct or indirect trauma to the brain, spinal cord or starting dose minimizes adverse effects, particularly if taken
relevant part of the peripheral nervous system in the in the evening before retiring. There appears to be no
preceding 3 months. added benefit to increasing the daily dose beyond 250 mg.
• No recent exposure to tremorgenic drugs. • Clonidine (an alpha adrenergic agent) in doses of
• Not in a state of drug withdrawal. 0.1–0.9 mg daily.
• Normal neurologic examination other than tremor. • Patients with alcohol responsive tremors may find
• No history or clinical evidence suggestive of psychogenic judicious use of alcohol to be helpful before social
origins of tremor. engagements.
• No evidence of stepwise deterioration. • Other drugs that may be helpful but also have adverse
effects include clonazepam (see Table 19), alprazolam,
Probable ET flunarizine, clozapine, nicardipine and carbonic
• Tremor: same as above (for definite ET). anhydrase inhibitors such as methisnozole and
• Duration more than 3 years. acetazolamide. Gabapentin is currently undergoing
• No evidence of isolated or localized tremors such as evaluation for the treatment of tremor.
primary orthostatic tremor, isolated voice tremor,
isolated position-specific or task-specific tremor, or
isolated tongue or chin tremor.

Possible ET
Type 1 Table 19 Effective tremorlytic drugs
Patients satisfy the criteria for definite or probable tremor Disease/disorder Effective drugs
but exhibit other neurologic disorders (such as
parkinsonism, dystonia, myoclonus, peripheral neuropathy, Enhanced physiologic β-blockers, alcohol
or restless legs syndrome) or other neurologic signs of tremor
uncertain significance (i.e. hypomimia, decreased arm swing, Essential tremor β-blockers, alcohol, primidone,
mild bradykinesia in isolation). phenobarbitone (phenobarbital)
‘Kinetic-predominant’ Clonazepam
Type II essential tremor
Monosymptomatic and isolated tremors of uncertain Primary orthostatic tremor Clonazepam
relation to ET. Parkinson’s disease Levodopa, dopamine agonists,
(rest tremor) anticholinergics, amantadine,
Task-specific tremors and isolated tremors of the voice, selegiline
tongue, head and legs have all been considered part of the Parkinson’s disease ?β-blockers, ?alcohol
spectrum of ET but it may be that the label ‘ET’ is not (postural tremor)
appropriate for these diverse disorders whose etiologies are Multiple sclerosis Isoniazid, clonazepam
not yet known.
122 Movement Disorders

Botulinum toxin CHOREA


Botulinum toxin can be injected locally into the splenius
capitus to reduce ‘no–no’ tremor without adverse effects
(e.g. weakness), but it does not work well when injected DEFINITION
into forearm muscles for wrist tremor (it causes wrist Involuntary, abrupt, irregular, arrhythmic and purposeless
weakness). movements of variable amplitude and usually involving the
face, hands and feet, which flow randomly from one body
Surgical treatment part to another.
• Stereotactic thalamotomy is usually employed only for
the most severe tremors because of the significant risks PATHOLOGY
of adverse effects (dysarthria, weakness, dysequilibrium, Basal ganglia lesions, such as infarction, hemorrhage, tumor,
and persistent paresthesia), particularly with bilateral and calcification, and caudate nucleus atrophy (Hunting-
operations. About 70–90% of patients experience some ton’s disease, 122).
relief from tremor.
• Thalamic stimulation with chronically implanted ETIOLOGY AND PATHOPHYSIOLOGY
electrodes, positioned with the use of intraoperative Hereditary neurologic disorders
recordings in the nucleus ventralis intermedius of the • Huntington’s disease (see p.417).
thalamus, confers lower risk. High frequency (100–200 • Benign familial chorea.
Hz) stimulation, controlled by a subcutaneously • Familial paroxysmal choreoathetosis.
implanted box on the chest wall, is thought to produce • Focal paroxysmal kinesigenic choreoathetosis.
its beneficial effect by inducing depolarization block. • Dentato-rubro-pallido-luysian atrophy (DRPLA).
Good results have been achieved in the vast majority of • Wilson’s disease.
patients (up to 88% with parkinsonian tremor and 68% • Cerebellar degenerations.
with ET). It is hoped that the benefits will be maintained • Neuroacanthocytosis.
with longer term follow-up.
A randomized comparison of thalamotomy and Vascular
thalamic stimulation found that both were effective in • Stroke: basal ganglia infarction or hemorrhage (hemi-
relieving drug resistant tremor but thalamic stimulation chorea, hemiballism).
produced greater functional improvement. Bilateral • Polycythemia rubra vera.
thalamic stimulation may have a lower complication rate • Essential thrombocythemia.
than bilateral thalamotomy. Dysarthria and aphonia • Vasculitis.
complicate up to 20–25% of bilateral thalamic lesions (or
stimulators), though with stimulation speech improves Immune-mediated
when the stimulator is switched off. • Sydenham’s (rheumatic) chorea: a complication of beta
hemolytic streptococcal infection.
PROGNOSIS • Systemic lupus erythematosus.
• ET is slowly progressive but seldom becomes severe. • Antiphospholipid antibody syndrome.
• Other neurologic impairments do not occur. • Encephalitis lethargica.

Metabolic/hormonal
• Hormone replacement therapy.
• Oral contraceptives (rare and reversible).
• Pregnancy (chorea is rare and stops within weeks of
delivery but may recur in subsequent pregnancies).
• Thyrotoxicosis.
• Hypocalcemia/hypoparathyroidism.
• Hyperglycemia.
• Fahr’s syndrome.
• Hallervorden–Spatz disease.
• Lesch–Nyhan syndrome.
• Ataxia telangiectasia.
• Kernicterus (neonatal hyperbilirubinemia).

Toxic
• Drugs:
Amiodarone. Amphetamines.
Antihistamines. Bromocriptine.
Butyrophenones. Carbamazepine.
Dopamine receptor-blocking drugs
(e.g. metoclopramide, phenothiazines).
Flunarizine. Flupenthixol.
L-dopa. Nomifensine.
Oral contraceptives (rare and reversible).
Phenobarbitone (phenobarbital). Phenytoin.
• Carbon monoxide poisoning.
Chorea 123

Structural: brain tumor (basal ganglia) DIFFERENTIAL DIAGNOSIS


Other hyperkinetic movement disorders:
Other • Ballism (perhaps a severe form of chorea): abrupt,
• Brain anoxia. irregular, large amplitude, random movements, usually
• Cerebral palsy. of the arm, due to a lesion of the subthalamic nucleus of
Luys, that is usually vascular.
CLINICAL FEATURES • Myoclonus (see p.124): brief, shock-like muscle jerks of
History abrupt onset and offset.
• Age: onset in children and adolescents (e.g. Sydenham’s • Tics (see p.130): repetitive simple and complex stereo-
chorea, benign familial chorea, kernicterus); adulthood typed movements, particularly affecting the face and
(e.g. Huntington’s disease). upper body, which can be suppressed by an effort of will.
• Chorea: • Tremor (see p.119): a rhythmic sinusoidal movement,
– Precipitants: sudden movement or startle (e.g. focal which may be present at rest (rest tremor), during the
paroxysmal kinesigenic choreoathetosis); alcohol, caffeine, maintenance of certain positions (postural tremor), during
emotion, stress (e.g. familial paroxysmal choreoathetosis). any active movement (kinetic or action tremor), or during
– Duration: brief paroxysms of chorea, tonic, dystonic or specific movements (task-specific action tremor).
athetoid movements lasting only a minute or two and • Dystonia (see p.113): a syndrome of sustained muscle
may be unilateral or bilateral (e.g. focal paroxysmal contractions and spasms resulting in twisting or repetitive
kinesigenic choreoathetosis). movements superimposed on abnormal postures.
– Course: progressive (e.g. Huntington’s disease, kernicterus). • Athetosis (literally ‘no fixed position’): twisting and
• Other history: writhing movements of distal limbs and fingers, generally
– Past history: kernicterus; cerebral palsy; stroke; streptococ- regarded as a distal expression of dystonia.
cal infection; previous episodes of chorea with pregnancy; • Idiopathic orofacial dyskinesia (see p.129).
use of the oral contraceptive pill or in other circumstances. • Cerebellar ataxia (see pp.437, 441, 444).
– Family history: chorea; dementia; suicide.
– Medications: oral contraceptives, other drugs and INVESTIGATIONS
duration of exposure. Blood
– Systemic enquiry: pregnant or not? • Full blood count (polycythemia rubra vera).
• Peripheral blood smear (acanthocytes).
Physical examination • ESR (vasculitis).
Chorea • Antinuclear antibody (systemic lupus erythematosus).
• Abrupt, unpredictable, fleeting, irregular, purposeless • Anticardiolipin antibody.
movements make the patient appear restless and fidgety. • Activated partial thromboplastin time (lupus anticoagulant).
The patient may try to disguise the involuntary • Creatine phosphokinase.
movements by terminating them with a semipurposeful • Thyroid function tests (thyrotoxicosis).
or purposeful voluntary movement. • Serum calcium (hypocalcemia).
• The movements may affect any or many parts of the • Copper and ceruloplasmin (Wilson’s disease).
body. Hemichorea suggests a unilateral basal ganglia or • Antistreptolysin O and anti-DNAase B titers (Syden-
subthalamic lesion. Exceptionally wild, flinging ham’s rheumatic chorea).
movements of an arm or leg on one side is called • DNA analysis for Huntington’s disease (expanded CAG
hemiballismus and is usually caused by a lesion, trinucleotide repeat on chromosome 4) and the DRPLA
commonly vascular, in the subthalamic nucleus of Luys. mutation.
• Speech may become slurred, swallowing impaired, the
hands clumsy, the gait unsteady, and falls occur. Imaging
• Ask the patient to hold their tongue out and arms out CT or MRI brain scan (basal ganglia lesion such as an
and observe for choreiform movements. infarct, hemorrhage, tumor, calcification; caudate nucleus
• Ask the patient to grip your right index finger with their atrophy [Huntington’s disease]).
left hand and your left index finger with their right hand
tightly and constantly. Observe for a ‘milk-maid’ grip
characterized by intermittent irregular flexion and
relaxation (chorea) of the patient’s fingers. 122
Other features
• Subcortical dementia (Huntington’s disease).
• Depression/behavioral, affective or psychotic disorder
(Huntington’s disease).
• Other movements: dystonia, tics, dyskinesia (neuro-
acanthocytosis), athetosis (kernicterus).
• Depressed tendon reflexes (subclinical motor neuropathy:
neuro-acanthocytosis).

122 Pathology specimen of a patient with Huntington’s disease


showing atrophy of the caudate nuclei and frontal lobes, and
secondary dilatation of the frontal horns of the lateral ventricles.
124 Movement Disorders

TREATMENT MYOCLONUS
Identify and remove the underlying cause if possible.

Symptomatic control of chorea DEFINITION


Dopamine receptor antagonists Sudden, very brief, shock-like involuntary movements
• Tetrabenazine, oral, 12.5 mg twice daily, increasing caused by muscular contractions (positive myoclonus) or
gradually if necessary and if tolerated, to 50 mg three inhibitions (negative myoclonus). Myoclonus is a primarily
times daily. descriptive term; it is not a diagnosis. (see Tables 20, 21).
• Haloperidol, oral, 0.5 mg three times daily, increasing
gradually if necessary and if tolerated to 10 mg three EPIDEMIOLOGY
times daily or more. • Incidence: 1.3 per 100 000 person years.
• Thiopropazate, oral, 5 mg three times daily, increasing • Prevalence: 8.6 per 100 000 population (lifetime).
as required and tolerated to 30 mg three times daily. • Age: any age; incidence and prevalence increase with
• Pimozide, oral, 2 mg daily, increasing as required and increasing age.
tolerated to 20 mg daily. • Gender: M>F.

Adverse effects include depression, drowsiness, PATHOPHYSIOLOGY


parkinsonism and tardive syndromes. An abnormal electric discharge in the neuraxis (commonly
Tetrabenazine is a synthetic benzoquinolone that depletes the cerebral cortex or brainstem reticular formation) leads
presynaptic storage of monoamines and blocks postsynaptic to a loss of normal inhibitory influences, an abnormally
dopamine receptors. It has the advantage over conventional rapid and hypersynchronous recruitment of multiple motor
neuroleptic dopamine antagonists that it rarely causes a units, and brief (<100 ms) ‘shock-like’ myoclonic jerks.
dystonic reaction and has not been reported to caused
tardive dyskinesia. The most common adverse effects are Epileptic myoclonus (see Table 21)
drowsiness (about one-third of cases), parkinsonism (about • Cortical reflex myoclonus: a fragment of focal or
one-quarter), depression (one-sixth), and insomnia, nervous- localization-related epilepsy, due to the paroxysmal
ness/anxiety, or akathisia (each about one-tenth of cases), all depolarization shift, an excessively long excitatory post-
of which can be controlled with reduction in dosage. synaptic potential that induces a rapid burst of action
potentials in the neurone. The EEG signal is high ampli-
Sodium valproate tude multiple and rapid spikes followed by a slow wave.

Phenytoin
Focal paroxysmal kinesigenic choreoathetosis.

Clonazepam
Familial paroxysmal choreoathetosis.

Prednisolone Table 20 Anatomic classification of myoclonus


Antiphospholipid antibody syndrome. Prednisolone may Myoclonus Anatomic origin Etiology
help, but is unproven. type

PROGNOSIS Focal Cerebral Vascular (AVM)


The prognosis depends on the cause. cortex Inflammatory (encephalitis)
Hemichorea and hemiballismus due to stroke usually resolve Tumor
within a few weeks or months. Brain stem Vascular, demyelination, tumor
(‘palatal
myoclonus’)
Spinal cord Vascular (ischemia)
Inflammatory (myelitis)
Tumor
Compression
Generalized/ Cortical or Anoxia (post hypoxic myoclonus)
multifocal brainstem Encephalitis
Creutzfeldt–Jakob disease
Metabolic encephalopathy
Spinocerebellar degenerations
Tay–Sachs disease
Lafora body disease
Whipple’s disease
Celiac disease
Drugs and toxins
Idiopathic (‘benign essential
myoclonus’)
Myoclonus 125

• Reticular reflex myoclonus: a fragment of a type of Essential myoclonus (no known causes and
generalized epilepsy, which originates in a hyperexcitable no other neurologic deficit)
caudal brainstem reticular formation. A generalized spike • Hereditary:
discharge, frequently associated with, but not time- – Autosomal dominant inheritance with variable
locked to, the jerk, usually follows the first EMG sign of penetrance.
myoclonus. – Onset before 30 years, usually in first or second decade.
• Primary generalized epileptic myoclonus: a fragment of – Men and women are affected equally.
primary generalized epilepsy, originating diffusely from – Non-disabling myoclonus, generalized or focal, with a
the cerebral cortex, which is driven diffusely and syn- predilection for the face, trunk, and proximal limb
chronously by subcortical inputs that trigger paroxysmal muscles.
events. – Myoclonus is reduced or absent at rest, absent in sleep,
and exacerbated by stress and movement.
Non-epileptic myoclonus – Sometimes associated with ET.
Segmental myoclonus: hypothesized to originate from the • Sporadic: similar to above but no family history.
spinal cord or brainstem from hyperactive alpha motor • Nocturnal myoclonus (periodic movements of sleep).
neurones that have lost normal inhibitory inputs.
Epileptic myoclonus (epileptic seizures
ETIOLOGY accompanied by myoclonus)
• Genetic: various forms of familial myoclonic epilepsy. • Generalized epileptic myoclonus:
• Acquired: diffuse brain damage: hypoxia, encephalitis, – More common in children and adolescents than adults.
metabolic disturbances. – Infantile spasms.
– Lennox–Gastaut syndrome.
Physiologic myoclonus (in normal people) – Juvenile myoclonic epilepsy (see p.86).
• Benign hypnagogic myoclonus (hypnic jerks): sudden, • Myoclonus associated with isolated spike discharges in
singular jerks on falling asleep. the motor cortex:
• With anxiety or exercise. – Idiopathic stimulus-sensitive myoclonus.
• Hiccoughs (singultus). – Isolated epileptic myoclonic jerks.

Symptomatic myoclonus
Static myoclonic encephalopathies
Myoclonus following an acute brain insult.

Table 21 Descriptive classification of myoclonus


Stimulus sensitive Non-stimulus sensitive
Cortical reflex Reticular reflex Periodic Rhythmic Irregular
Site Face, distal limb Face, proximal limb Face, limb Eyes, palate, limb Face, limb
Laterality Unilateral Bilaterally Bilateral usually Symmetric usually Unilateral
or bilateral synchronous or bilateral
Rhythmicity Irregular Irregular Periodic Rhythmic Irregular
Provoking factors Action, various Action, various None None Indefinite
stimuli stimuli
Duration of Short Short Relatively long Long Indefinite
EMG discharge (100–400 msec)
Spread Rostral to caudal Bulbar to rostral None None None
EEG Spike, contralateral Spike, no relation Can be associated No correlate No correlate
central, time with myoclonus with PSDs
locked to myoclonus
Cortical SEP Giant Not enhanced Not enhanced Not enhanced Not enhanced
Long latency reflex Enhanced Enhanced Not enhanced Not enhanced Not enhanced
Main causes PME Post anoxic CJD Pontine lesion PME
Post anoxic PME SSPE Essential
CJD Spinal Spinal
Alzheimer’s disease
PME: Progressive myoclonic epilepsy and allied diseases; CJD: Creutzfeldt–Jakob disease; SSPE: Subacute sclerosing panencephalitis;
PSD: Periodic synchronous discharge. Spinal myoclonus is sometimes stimulus sensitive
126 Movement Disorders

Post-hypoxic (chronic) action myoclonus: CLINICAL FEATURES


• Myoclonic jerks in isolation or repetitively. History
• Induced by attempts at voluntary movements (action or • Exposure to drugs, toxins or anoxia.
intention myoclonus) or by some types of sensory • Current and past medical problems: epileptic seizures.
stimuli. • Family history.
• Associated with spikes in the EEG.
Examination
Post-hypoxic (acute) myoclonus: Myoclonus
• Generalized, rhythmic, and stimulus-sensitive myoclonus. Involuntary, sudden, very brief, shock-like movements of
• Occurs as an acute effect of hypoxia while the patient is the face, limbs or trunk which tend to be isolated,
in, or recovering from, coma. asymmetric, repetitive and irregular.
• Frequently transient.
• Causes includes: head injury, thalamotomy and stroke. Anatomic site:
• A poor prognostic sign when it occurs in the recovery • Generalized (involving the whole body).
phase of hypoxic coma. • Regional/multifocal (confined to two or more
contiguous regions).
Progressive myoclonic encephalopathies • Focal/segmental (confined to one region).
A progressive brain disorder characterized by myoclonus
with or without seizures. Activation: myoclonus may occur at rest (‘spontaneous
myoclonus’) or be induced by a stimulus such as a sudden
Inborn errors of metabolism: noise, flash of light, or touch (reflex myoclonus) or
• Gaucher’s disease. voluntary movement (action myoclonus), or some
• Hexosaminidase deficiency (GM2 gangliosidosis) (see combination of these factors.
p.172).
Temporal pattern: sporadic or repetitive/periodic, rhythmic
Infectious encephalopathies: or irregular: examples of rhythmic myoclonus include palatal
• Subacute sclerosing panencephalitis (see p.304). myoclonus, or symmetric flexion of the limbs or trunk; the
• Herpes simplex encephalitis (see p.295). latter typically occurs in progressive brain disorders such as
• Arbovirus encephalitis (see p.293). SSPE and infantile spasms (infants with brain damage and
• Creutzfeldt–Jakob disease (see p.330). violent movements).
• Post-encephalitic parkinsonism (see p.420).
Associations:
Progressive myoclonic epilepsies (see p.87). • Other forms of generalized epilepsy; a series of myoclonic
jerks may precede tonic-clonic seizures.
Progressive myoclonic ataxias: a syndrome comprising • Lapses in contraction of muscles (negative myoclonus),
myoclonus, progressive cerebellar ataxia with infrequent or which may cause falls (akinetic attacks) or asterixis: the
absent epileptic seizures and little or no cognitive ‘flapping tremor’ of various encephalopathic states.
dysfunction due to diseases such as mitochondrial disease,
spinocerebellar degenerations, and celiac disease. Other neurologic signs
• Cognition.
Metabolic encephalopathies: • Cerebellar function.
• Dialysis dementia. • Extrapyramidal function.
• Hepatic encephalopathy (see p.452).
• Hypoglycemia. SUBTYPES (see Table 20, p.124)
• Non-ketotic hyperglycemia. Focal myoclonus
• Hypocalcemia. Myoclonus restricted to one muscle group or body part.
• Hyponatremia.
• Renal failure. Cortical myoclonus
• Respiratory failure. • A form of focal motor seizure due to an abnormal focal
electric discharge in or near the motor cortex.
Toxic encephalopathies: • May be spontaneous or precipitated by action, muscle
• Heavy metal poisons. stretch and particularly touch and photic stimulation
• Bismuth. (stimulus-sensitive).
• Methyl bromide. • Manifests as one or more myoclonic jerks of a limb or the
• Strychnine. face on the side contralateral to the cortical discharge.
• Lithium. • If repetitive and regular, it is synonymous with epilepsia
• Beta-lactams. partialis continua.

Degenerative disorders:
• Alzheimer’s disease (see p.407).
• Wilson’s disease (see p.157).
• Huntington’s disease (see p.417).
• Progressive supranuclear palsy (see p.432).
Myoclonus 127

Brainstem (palatal myoclonus/nystagmus) Generalized myoclonus


• Regular, slow, continuous, rhythmic (1–3 times per • Usually arise from the brainstem.
second) contraction of the soft palate which may give rise • May be stimulus-sensitive, especially to sound.
to a rhythmic clicking in the ear and difficulty with • Treatment may be difficult and is based on the use of
speaking and swallowing. anticonvulsants (e.g. clonazepam, sodium valproate,
• Sometimes the ocular, facial and diaphragmatic muscles primidone), often in combination.
may be involved.
• Movements do not cease during sleep. DIFFERENTIAL DIAGNOSIS
• Caused by a lesion in the brainstem that may involve the Myoclonus or not?
triangular pathway between the red nucleus in the mid- • Clonus: a series of rhythmic, uniphasic or monophasic
brain, the ipsilateral inferior olivary nucleus in the lower (i.e. unidirectional) contractions and relaxations of a
medulla, and the contralateral dentate nucleus in the group of muscles, thus differing from myoclonus, which
cerebellum (Guillain–Mollaret’s triangle). are shock-like contractions of a group of muscles that are
• Onset usually 10–12 months after the precipitating cause irregular in rhythm and amplitude and, with few
and remains permanently. exceptions, asynchronous and asymmetric in distribution;
and also differing from tremors, which are always
Hemifacial spasm diphasic or bidirectional.
• Involuntary painless muscle twitching around one eye. • Asterixis: sudden pauses of muscular activity (a form of
• Exacerbated by eye closure. negative myoclonus): the basis of the ‘flapping tremor’
• May gradually spread to involve the other facial muscles of various encephalopathic states.
ipsilaterally and lead to additional twitching of the corner • Tics (see p.130): simple motor tics are rapid brief jerks
of the mouth and platysma. involving a single group of muscles, usually of the head
• Mild weakness around the corner of the mouth may be and arms, which appear very much like myoclonus.
present but no other signs. However, unlike myoclonus, they can be suppressed for
• Causes include facial nerve compression by a cerebello- short periods at the expense of increasing tension and
pontine angle mass which needs to be excluded by CT distress to the patient. Complex motor tics involve
or MRI scan, particularly if there is an absent ipsilateral sequences of movements that persist far too long to be
corneal reflex or sensorineural deafness. confused with myoclonus.
• Although not a classical subtype of myoclonus, it is in- • Startle reflexes: an exaggerated startle response is a quick
cluded here for completeness and to highlight the over- jerk involving the face, head, neck, and sometimes the
lap between some involuntary movement syndromes. proximal limbs. Although myoclonus may occasionally
be triggered by startle, usually it is spontaneous or
Segmental myoclonus elicited by other stimuli.
• Restricted to one part of the body innervated by the • Chorea (see p.122): choreiform movements are abrupt,
brainstem or spinal cord. unsustained, irregular and non-stereotyped movements
• May be caused by brainstem or spinal cord trauma, of variably changing speed that seemingly flow at random
arteriovenous malformation, tumor and transverse from one muscle group to another, whereas myoclonic
myelitis. movements are repetitive and stereotyped in both
• Distinguish from cortical epileptic discharges causing direction and degree.
simple partial motor seizures, epilepsia partialis continua • Dystonia (see p.113): a syndrome of excessive muscle
and cortical myoclonus, which may be evident on the contractions, which frequently cause twisting and
EEG. repetitive movements, or abnormal postures. Quick
movements can occur in dystonia, but their association
Nocturnal ‘myoclonus’ (periodic leg movements of sleep) with slower movements or postures distinguishes them
• Abrupt, rather stereotyped, withdrawal movements, from myoclonus.
usually of one leg but sometimes one arm, synchronously • Tremor (see p.119): a rhythmic and sinusoidal oscillation
or asynchronously, followed by a brief tonic contraction of one body part in relationship to another. Although
which can occur intermittently throughout the night, myoclonus can be rhythmic, the movement form
during non-REM sleep, every 30 seconds for up to resembles the appearance of a ‘square wave’ temporal
several hours. pattern, with a distinguishable interval between each
• Most common in the elderly. movement.
• Not epileptic. • Fasciculations: appear as muscle twitches which do not
• Quite benign but can disrupt sleep. result in the movement of a whole muscle (as does
• May be associated with restless legs syndrome. myoclonus), and result from spontaneous firing of motor
units or bundles of muscle fibers, usually in the setting
Multifocal myoclonus of peripheral nerve disease.
Usually caused by widespread cortical pathology. • Cerebellar ataxia: in patients with moderate or severe
action myoclonus, it is difficult to determine if cerebellar
signs are present as well, unless the myoclonus can be
controlled with drug therapy.
128 Movement Disorders

INVESTIGATIONS Specific medication for myoclonus


First line • Sodium valproate, benzodiazepines (clonazepam) and
• Electrolytes. lamotrigine are the most effective conventional drugs.
• Glucose. Valproate use may be limited by gastrointestinal effects,
• Renal function. sedation and liver toxicity. Clonazepam can be limited by
• Hepatic function. sedation, imbalance, behavioral changes, and tolerance.
• Drug and toxin screen. • Piracetam may be useful.
• Brain imaging: CT or MRI scan. • Serotonin precursors may be used for post-hypoxic
• EEG (123): generalized spike wave discharges, photo- myoclonus or progressive myoclonic epilepsy.
sensitivity, focal (particularly posterior) epileptiform • Phenytoin has a deleterious effect.
discharges, vertex spikes in REM sleep, may be found in • In contrast to the general principle of monotherapy in
most of the disorders that cause the progressive epilepsy, polytherapy may produce additional benefit in
myoclonic epilepsy syndrome. Slowing of background the treatment of severe myoclonus, although drug
activity occur in all forms of PME but is usually much toxicity needs to be avoided.
more prominent and early in those diseases with diffuse
neuronal damage and storage compared with those Physical therapy
diseases with restricted neuronal degeneration.
Social rehabilitation
Second line
• Polygraphic EEG–EMG monitoring and back-averaging: PROGNOSIS
the most essential and useful electrophysiologic Depends on the cause; myoclonus may occur as a transient
technique to try and show a relationship between a or persistent phenomenon in many conditions such as viral
cortical spike and a muscle contraction. EEG-EMG encephalitis, suppurative meningitis, intoxications with
backaveraging techniques can be used to detect EEG strychnine and tetanus, and metabolic disorders such as
potentials that are not present in routine EEG uremia and anoxic encephalopathy.
recordings. This technique increases the signal-to-noise
ratio and records preceding EEG waveforms that are
time-locked to the myoclonus.
• Somatosensory-evoked potentials (SEP): useful for
further investigating the pathophysiology of myoclonus.
Giant somatosensory-evoked potentials may be found in
stimulus-sensitive, cortical reflex myoclonus caused by
most of the disorders that cause the progressive
myoclonic epilepsy syndrome.
• Copper studies: Wilson’s disease.
• Enzyme activities: storage diseases.
• Genetic testing: mitochondrial disease, Huntington’s
disease, spinocerebellar ataxias.
• Chromosomal fragility: ataxia-telangiectasia.
• Spinal imaging: segmental pathologic condition.
• Tissue biopsy: storage disease, mitochondrial disease.

DIAGNOSIS
Clinical and electrophysiologic.
123
TREATMENT
The myoclonus may not be severe enough to warrant
symptomatic treatment. Treatment of the underlying cause
(e.g. renal failure) may obviate the need for specific
treatment. Anti-epileptic treatment for seizures may reduce
the myoclonus.

123 EEG, 8 channel, sleep recording, in a young adult with myoclonic


seizures, showing trains of fast spikes followed by a single slow wave
(multispike and wave).
Dyskinesias 129

DYSKINESIAS Paroxysmal kinesigenic dyskinesia


• Decreased inhibitory influences of the medial globus
pallidus and substantia nigra on the thalamus, or
DEFINITION • Damage to the thalamic reticular nucleus itself, thus
Abnormal dystonic and choreoathetoid involuntary making the thalamus more sensitive to superficial or deep
movements that may occur spontaneously, after a sudden sensory input.
voluntary movement, after prolonged physical exertion, or
during sleep, which occur during full consciousness, and are Paroxysmal hypnogenic dyskinesia (PHD)
of variable duration. May be associated with frontal epilepsy.

EPIDEMIOLOGY CLINICAL FEATURES


Prevalence Tardive dyskinesia
• Tardive dyskinesia: about 20% of patients on long-term Continuous involuntary movements of the orobuccolingual
antipsychotic medication. and masticatory muscles of a choreiform nature.
• Paroxysmal dyskinesia: rare, except when associated with
late stage Parkinson’s disease. Paroxysmal dyskinesis
Precipitating factors
Age of onset Paroxysmal kinesigenic dyskinesia (PKD):
• 21 years (mean); range 1–60 years (paroxysmal • Sudden voluntary movement of a limb; orofacial
kinesogenic dyskinesia [PKD]). movements such as yawning and talking; movements
• 30 years (mean); range 1–77 years (paroxysmal non- associated with startle or hyperventilation.
kinesogenic dyskinesia [PNKD]).
• 13 years (mean); range 1–29 years (paroxysmal exertion- Paroxysmal exertion-induced dyskinesia (PED):
induced dyskinesia [PED]). • Walking, running for 5–15 minutes.
• Idiopathic paroxysmal dyskinesias have a younger age of
onset compared with secondary dyskinesias. Paroxysmal hypnogenic dyskinesia (PHD):
• Gender: F<M = 3:5 (i.e. about 60% as common in • Sleep.
women than men).
Prodromal symptoms immediately prior to onset of attack
CLASSIFICATION Muscle tension; tingling; numbness; dizziness.
Continuous dyskinesias
Tardive dyskinesia. Site of involvement
Limbs, often unilaterally; oromandibular and facial muscles:
Paroxysmal dyskinesias dysarthria or anarthria.
Kinesigenic (Paroxysmal kinesigenic dyskinesia)
Abrupt onset of episodes of abnormal involuntary move- Nature of involvement
ments after a sudden voluntary movement. Dystonia and/or choreoathetosis or athetosis.

Non-kinesigenic (Paroxysmal non-kinesigenic dyskinesia) Associated symptoms


Spontaneous onset of attacks of abnormal involuntary Pain in the affected body part sometimes; perhaps sleep
movements. disorders (e.g. sleep apnea, narcolepsy).

Exertion-induced dyskinesia Exacerbating factors


Attacks of abnormal involuntary movements precipitated by Paroxysmal kinesigenic dyskinesia (PKD):
prolonged physical exertion. • Stress; fatigue; menses; alcohol; cold; heat.

Hypnogenic dyskinesia Duration of attacks


Attacks of abnormal involuntary movements occurring only From seconds up to several hours to days. Attacks may
during sleep. become shorter or longer with time.

ETIOLOGY SPECIAL FORMS


Tardive dyskinesia Superior oblique myokymia, causing paroxysmal diplopia or
Long term antipsychotic medication. ‘shimmering of vision’.

Paroxysmal kinesigenic dyskinesia DIFFERENTIAL DIAGNOSIS


Familial: 65% of cases, usually autosomal dominant • Epileptic seizures (movement-induced seizures, reflex
inheritance with complete or incomplete penetrance. Linked epilepsy or subcortical epilepsy): paroxysmal dyskinesias
to the pericentromeric region of chromosome 16. are dystonic or choreoathetoid with preserved conscious-
ness, a normal EEG during attacks, and absence of a post
PATHOPHYSIOLOGY ictal state.
Tardive dyskinesia • Psychogenic: attacks associated with or triggered by
Dopamine receptor hypersensitivity. hyperventilation and odors.
130 Movement Disorders

INVESTIGATIONS TOURETTE SYNDROME (TS)


Cranial CT and MRI scan: often normal.

TREATMENT DEFINITION
The precipitating circumstance is the most important factor A genetically determined chronic neuropsychiatric disorder
in determining the underlying mechanism of the paroxysmal that develops in children and adolescents and is
movement disorder, the natural history and response to characterized by multiple motor and vocal tics (quick,
treatment. Long lasting attacks suggest a good response to involuntary movements that occur repeatedly and non-
medication. rhythmically in the same way) which last more than 1 year
and cause considerable social, vocational and functional
Tardive dyskinesia impairment. It is commonly associated with obsessive–com-
• Difficult to treat. pulsive behavior, attentional and executive dysfunction, and
• Withdrawal of the responsible antipsychotic drug may aggressive behavior. The syndrome was first described in
induce a remission, but this can take months or even 1825 by Dr Georges Gilles de la Tourette.
years, and may be impractical if the patient has a serious
underlying psychiatric disorder. EPIDEMIOLOGY
• Increased doses of dopamine antagonists (e.g. tetra- • Prevalence: 3–10/1000; much more common than
benazine) paradoxically may lead to improvement, previously appreciated; many cases are mild and do not
presumably by depleting presynaptic dopamine, but may come to medical attention or are not recognized.
also cause depression. • Tics: common during childhood development (4–12%
• Baclofen, benzodiazepines, and calcium-channel blockers of children aged 6–12 years), and possibly more common
may be effective; anticholinergics make tardive dyskinesia in children with learning problems. The relationship
worse. between such tics and the disorder of TS remains unclear.
Commonly, tics are transient and disappear.
Paroxysmal dyskinesias • Age of onset: before age 21, usually 2–15 years.
Almost all patients with PKD improve with anti-epileptic drugs • Gender: M>F (3:1); as yet unexplained.
in contrast to only one-quarter of patients with PNKD. Short-
lasting (and non-kinesigenic) attacks generally fail to PATHOLOGY
improve with medication. Carbamazepine and phenytoin Limited histopathologic data have failed to identify any
seem more effective than other anti-epileptic drugs in PKD definitive pathologic changes in the striatum (124).
and clonazepam in PNKD. Drugs that enhance GABAergic
neurotransmission, such as benzodiazepines, valproic acid, and ETIOLOGY AND PATHOPHYSIOLOGY
phenobarbitone (phenobarbital), increase the latency to the • Commonly familial and inherited as an autosomal
onset of attacks and reduce the severity, while drugs that dominant trait with reduced penetrance and variable
impair GABAergic transmission decrease the latency. expression.
• Clonazepam (PNKD). • A linkage has been found to chromosome 18 in some
• Tetrabenazine (PKD). families.
• Lorazepam (PHD). • No evidence for genetic linkage at the dopamine D2 or
• Chronic stimulation of the ventro intermediate (Vim) D3 receptor loci has been found.
thalamus may impove dystonic paroxysmal non- • It is hypothesized by some that the TS genetic locus is
kinesigenic dyskinesia. common and is involved in normal brain development,
and that the occurrence of the clinical disorder TS
represents an excessive expression or abnormal
persistence of normal developmental characteristics. The
appearance of the typically associated clinical features of
TS (i.e. tics, obsessive–compulsive behavior, inattention,
and hyperactivity) commonly occurs during normal
childhood development (as a form of ‘developmental’ or
‘physiological’ TS) and reflects normal synaptogenesis in
basal ganglia and limbic regions. The medical disorder
TS differs only quantitatively from this process.
• Other hypotheses are that:
– TS is a basal ganglia disorder characterized by striatal
dopamine receptor hypersensitivity.
– TS is a cingulate and orbitofrontal cortex disorder
characterized by dysfunction of the central endogenous
opioid system in limbic and orbitofrontal basal
ganglia/thalamic loops.
• Reports of TS following head trauma, carbon monoxide
poisoning, neuroleptic drugs, other toxic and metabolic
encephalopathies, Creutzfeldt–Jakob disease, Hunting-
ton’s disease, and encephalitis lethargica are more likely
to be coincidental than causal.
Tourette Syndrome (TS) 131

CLINICAL FEATURES are periodic changes in the type, number, frequency, and
Chronic, multiple motor and vocal tics location of the tics, and in the waxing (increasing) and
Tics involving the face or neck (e.g. repeated eye blinking waning (decreasing) of their severity. Symptoms will some-
and throat clearing) is the most common initial feature. times mysteriously disappear for weeks or months at a time.
Other common initial symptoms include complex move- Although tics are described as being quick, involuntary
ments such as touching, striking or hitting, and jumping. movements that occur repeatedly and non-rhythmically in
Tics are brief stereotyped movements (motor tics) or the same way, they are not strictly ‘involuntary’. Most
sounds produced by moving air through the nose, mouth people with TS have some control over their symptoms.
or throat (vocal tics). They are classified as motor and vocal, However, this control, which can be exerted from seconds
and simple and complex, although the boundaries of these to hours at a time, tends to delay more severe tics. They
are not well defined: become irresistible and must eventually be executed; some
• Simple motor tics: eye blinking, facial grimacing, neck people wait until they are in private before doing so.
jerking, shoulder shrugging.
• Complex motor tics: facial gestures, smelling, groaning Associated disorders, behaviors and consequences
behaviors, touching, stamping, hitting or biting oneself, Coexist in about half of all patients.
jumping.
• Simple vocal tics: sniffing, coughing, throat clearing, Obsessive–compulsive disorder (about 50% of cases)
grunting, snorting, barking. The presence of obsessions or compulsions causing marked
• Complex vocal tics: repeating words or phrases out of distress or interfering with normal functioning. Obsessions
context, palilalia (increasingly rapid repetition of a word are recurrent persistent ideas, thoughts, impulses or images
or phrase), echolalia (repetition of words), and coprolalia that are intrusive and inappropriate and cause anxiety or
(foul language). distress. They are a product of the person’s own mind and
are not simply excessive worries about real-life problems.
With time, the tics become more prominent and involve Examples include fear of contamination, need for symmetry,
the shoulders and arms, followed by the legs. Involvement and pathologic doubt. Compulsions are repetitive behaviors
of the muscles of respiration causes vocal tics such as or mental acts that a person feels driven to perform, often in
grunting, barking or coughing noises which interrupt response to an obsession. The aim of them is to prevent or
conversation. The expulsion of air imparts an explosive reduce anxiety or distress, not to provide pleasure or grati-
quality to the speech and occasional associated hissing fication. Examples include checking, washing, counting,
sounds. needing to ask or confess, and symmetrizing or being precise.
Some patients develop repetitive involuntary explosive
stereotyped verbal utterances during compulsive expiration, Attention deficit disorder (ADD)
which may consist of obscene expletives (coprolalia). Characterized by five or more of the following: failure to
Patients may also repeat the last word or sentence of others’ give attention to details or making careless mistakes in
(echolalia) or their own speech (palilalia), repeat motor acts schoolwork, work, or other activities; frequent difficulty
(echopraxia) and sometimes obscene (copropraxia) and organizing tasks and activities; easy distractibility by
other gestures. extraneous stimuli; failure to follow through on instructions
The tics occur many times a day (usually in bouts) nearly and finish work or duties; often leaving seat in classroom or
every day, or on again, off again for more than a year. There in other situations in which remaining seated is expected;
difficulty playing or engaging in leisure activities quietly;
talking excessively.

Sleep disorders
Sleep talking, insomnia, nightmares, enuresis, and
124 somnambulism.

Other types of behavior


• Learning difficulties.
• Hyperactivity.
• Self-injury.
• Antisocial behavior.
• Aggressive behavior.
• Inappropriate sexual activity.
• Lack of discipline.

These disorders and behaviors may wax and wane and


may persist even after the tics have largely disappeared. Many
patients are left-handed or ambidextrous and have minor
neurologic abnormalities. A family history of tics, obses-
sive–compulsive behavior, or ADD is commonly present.

124 Brain of a drug addict showing basal ganglia (globus pallidus)


disease bilaterally due to the use of illicit drugs.Tics are occasionally
due to organic basal ganglia disease.
132 Movement Disorders

DIFFERENTIAL DIAGNOSIS Tourette disorder (TD)


Normal variants (i.e. physiologic) 1 Criteria 1–5 for Tourette syndrome are satisfied.
• Tics are common in children between the ages of 2 Tics cause distress or significant impairment in social,
2–6 years but most are transient and disappear. academic, occupational, or other important areas of
• ‘Colds’ or ‘allergies’: some of the most common tics, functioning.
such as eye blinking, sniffing, throat clearing, and cough-
ing are commonly ascribed to ‘colds’ and ‘allergies’. Clinical features increasing confidence for the
• ‘Nervous habits’: tics such as facial grimaces or head jerks diagnosis of TS or TD
are often considered to be ‘nervous habits’. 1 Multiple tic types that vary over time.
2 Tics wax and wane in severity.
Developmental basal ganglia syndrome 3 Coprolalia.
Primary (hereditary) 4 A positive family history of TS or TD.
Primary tic disorders:
• Tourette syndrome. N.B. The type or severity of tic is not considered in the
• Chronic motor tic disorder. diagnosis of TS. For example, coprolalia is not required for
• Chronic vocal tic disorder. the diagnosis; it occurs in less than one-third of clinic
• Transient tic disorder. populations and in less than 5% of affected members of
studied TS pedigrees.
Related primary neuropsychiatric disorders:
• Primary obsessive–compulsive disorder. TREATMENT
• Primary ADD. Education and counselling of the patient and family.
• Developmental stuttering.
Psychologic support
Inherited neurologic disorders Promote the child’s self-esteem and competency and
• Huntington disease. provide support in the challenges of school or work and in
• Neuroacanthocytosis. social relationships.
• Torsion dystonia.
Pharmacologic intervention
Secondary (symptomatic) Symptomatic treatment of TS with drugs can be helpful but
• Autisms, pervasive developmental disorder. improvements are usually incomplete and short-lasting.
• Mental retardation.
• Fetal alcohol syndrome. Tics
• Intrauterine infection or exposure to illicit drugs (124). Neuroleptics with dopamine-blocking activity are most
• Perinatal asphyxia. widely used. Those with a greater selectivity for the
• Carbon monoxide toxicity. dopamine D2 receptor subtype, such as haloperidol,
• Encephalitis. pimozide and sulpiride can be effective in reducing the
• Head trauma. frequency and severity of tics for patients with severe
• Stroke. symptoms. Medication needs to be administered carefully
• Post-infectious (immune-mediated). and in small doses to obtain the maximum benefit and the
• Sydenham’s chorea. minimum number and degree of adverse effects; tardive
dyskinesia often develops with prolonged usage. Other
Tic-inducing drugs effective treatments include tetrabenazine, a dopamine-
• Neuroleptic agents. depleting agent, and clonidine, which is a central α2
• Stimulants. adrenergic receptor antagonist and is thought to reduce
presynaptic release of noradrenaline.
INVESTIGATIONS • Pimozide, beginning with 0.5 mg daily and increasing
• Blood: DNA analysis. by 0.5 mg increments to as high as 10 mg daily if
• EEG: abnormal in many cases although non-specifically. necessary, is effective in up to 80% of cases. Adverse
effects include sedation, weight gain, depression, and
DIAGNOSIS restless legs.
Tourette syndrome (TS) • Haloperidol, for patients who fail to respond to pimozide
1 Multiple (two or more) motor and/or vocal tics. because the risk of long term adverse effects such as
2 Onset in the first two decades of life. tardive dyskinesia is greater with haloperidol than
3 Tics present for more than 1 year. pimozide. Starting dose 0.5 mg three times daily by
4 Tics are not caused by tic-inducing drugs (e.g. stimulants mouth, increasing to as much as 10 mg three times daily
or neuroleptic agents), other environmental insults to the if necessary.
brain (e.g. perinatal hypoxia, intrauterine infection, or • Trifluoperazine or thiothixene can be effective in
exposure to alcohol), or to hereditary neurologic refractory cases.
disorders that may produce tics (e.g. neuroacanthocytosis • Clonidine may help a few: increasing up to 6 micrograms
or Huntington’s disease). per kg daily, oral, in three divided doses, but sedation can
5 There is no evidence of significant brain dysfunction (e.g. occur. Withdrawal must be slow to avoid rebound
mental retardation, pyramidal tract signs, or seizures) hypertension.
other than tics. • A combination of neuroleptics may be required.
Neuroleptic Malignant Syndrome 133

• Naltrexone, an opiate antagonist, has improved attention NEUROLEPTIC MALIGNANT


and reduced the number of tics in a double-blind SYNDROME (NMS)
randomized trial.
• Botulinum toxin reduces urge and tic frequency.
DEFINITION
Obsessive–compulsive disorder A rare but potentially life-threatening complication of
Obsessive–compulsive disorder can be controlled with anti- neuroleptic drug treatment.
depressant medication, and drugs with serotonin reuptake
blocking activity, such as clomipramine and fluvoxamine, EPIDEMIOLOGY
appear to be the most effective. • Incidence: 0.2% of patients treated with neuroleptics.
• Age: all ages, particularly young and middle-aged adults;
Attention deficit disorder mean age 40 years.
ADD can be treated with psychostimulant medications, but • Gender: M=F.
when patients have combined features of ADD and TS,
stimulant drugs tend to worsen the tics. Desipramine has ETIOLOGY
been shown in a recent double-blind controlled trial to be D2 dopamine receptor antagonists
useful in the treatment of ADD in TS without a Neuroleptic drugs
deterioration in tic severity. • Phenothiazines: chlorpromazine;
• Butyrophenones: haloperidol – most common.
PROGNOSIS
• TS is a lifelong illness. Non-neuroleptic antiemetic, anesthetic and sedative drugs
• Many people experience remissions of symptoms • Metoclopramide.
(particularly tics), commonly in the late teens and early • Prochlorperazine.
twenties, which may be complete, or incomplete and • Droperidol.
followed by subsequent relapses. • Promethazine.
• Lifelong medical treatment with pimozide or haloperidol
is often required but many patients can be restored to a Withdrawal of dopamine agonists
full active life in the community, without any (anti-parkinsonian medication)
compromise to life expectancy. Infrequent.
• A person with TS has about a 50% chance of passing the
gene to his or her children. However, only about 10% of Risk factors
children who inherit the TS gene will have symptoms • Previous episodes (one-third develop subsequent
severe enough to seek medical attention. episodes on re-challenge).
• Dehydration.
• Agitation.
• Rapid rate and parenteral route of neuroleptic
administration.
• Pre-existing organic brain disease or mood disorders,
particularly if taking lithium.

PATHOPHYSIOLOGY
• An acute reduction in brain dopamine activity; probably
dopamine receptor blockade in the hypothalamus,
caudate and pallidum primarily.
• Commonly due to an increase in dose of neuroleptic
drugs within the therapeutic range, rather than a toxic
manifestation.
• Infrequently, withdrawal of anti-parkinsonian medication
precipitates NMS.

CLINICAL FEATURES
Rapid onset, usually within a few days of starting treatment
with a dopamine receptor antagonist (16% within 24 hours,
66% within 1 week, 96% within 30 days), but sometimes
after stopping treatment.

History
Often there is a history of psychiatric illness, requiring
treatment with a neuroleptic. Occasionally, recent with-
drawal of anti-parkinsonian medication has taken place.
134 Movement Disorders

Physical examination INVESTIGATIONS


• Altered level of consciousness with confusion and • Full blood count (leukocytosis).
agitation (often precedes rigidity and fever). • Serum creatine kinase: elevated (but may be normal).
• Muscle rigidity in all muscle groups ± opisthotonos, • Urinary myoglobin: detected (myoglobinuria).
tremor and dyskinesia. • Arterial blood gases (hypoxia and ?respiratory ?metabolic
• Autonomic dysfunction (hypertension or hypotension). acidosis).
• Hyperthermia (>38°C [100.4°F], fever, diaphoresis). • Culture urine and blood to exclude a septic focus.
• Tachycardia. • EEG: non-focal, generalized slow wave activity in half of
• Tachypnea. cases (125).
• Dehydration is common.
• Ocular abnormalities (oculogyric crises, internuclear DIAGNOSTIC CRITERIA
ophthalmoplegia, and conjugate gaze paresis) occur All five items required concurrently:
infrequently. 1 Treatment with neuroleptics within 7 days of onset
• Extensor plantar responses may be present. (2–4 weeks for depot neuroleptics).
2 Hyperthermia (>38°C [100.4°F]).
ATYPICAL FORMS 3 Muscle rigidity.
NMS may follow cessation of prolonged therapy (6 weeks) 4 Five of the following:
with metoclopramide. – Change in mental status.
– Tachycardia.
DIFFERENTIAL DIAGNOSIS – Blood pressure instability (hypertension or hypotension).
Encephalopathy – Tachypnea or hypoxia.
Primary central nervous system disorders – Diaphoresis or sialorrhea.
• Infection (viral encephalitis, HIV, post-infectious – Tremor.
encephalomyelitis). – Incontinence.
• Tumor. – Creatine phosphokinase elevation or myoglobinuria.
• Stroke. – Leukocytosis.
• Trauma. – Metabolic acidosis.
• Seizures. 5 Exclusion of other drug-induced, systemic, or neuropsy-
• Severe dystonic reaction. chiatric illnesses.
• Akinetic mutism.
TREATMENT
Systemic disorders Early recognition
• Infection (septicemia, tetanus, rabies).
• Metabolic derangement. Discontinue offending drug
• Endocrinopathy (thyrotoxicosis, pheochromocytoma).
• Autoimmune disease (SLE, polymyositis). Supportive
• Heatstroke (history of physical exertion, and absence of • Intravenous fluids for rehydration.
diaphoresis and rigidity). • Temperature reduction.
• Malignant hyperthermia. • Nasogastric feeds.
• Toxins (carbon monoxide, phenols, strychnine, lithium, • Monitor for complications.
cocaine, designer drugs [e.g. ecstasy (methylenedioxy- • Consider early transfer to an intensive care unit.
amphetamine)]).
• Food-related allergic reactions.
• Substance abuse (salicylates, dopamine inhibitors and
antagonists, stimulants, monoamine oxidase inhibitors,
anesthetic agents, anticholinergics, alcohol or sedative 125
withdrawal).
• Serotonin syndrome: this is a potentially fatal hypersero-
toninergic state commonly precipitated by coadministra-
tion of either tryptophan or a selective presynaptic
serotonin reuptake inhibitor (drugs known to increase
serotonin, e.g. clomipramine) with a monoamine oxidase
inhibitor (MAOI, e.g. tranylcypromine). Typical features
include confusion, agitation, restlessness, hypomania,
fever, diaphoresis, shivering, tremor, myoclonus, hyper-
reflexia and incoordination, which coincide with the
addition of, or an increase in the dose of, these drugs.
Spontaneous resolution often occurs within 24 hours of
removing the added agent.

Psychiatric disorder
Catatonia.

125 EEG showing non-specific generalized slow wave activity.


Further Reading 135

Specific • Mortality has declined in recent years although fatalities


• May be required in more severe cases. still occur.
• Benzodiazepines intravenously (e.g. midazolam; • Neuroleptics may be reintroduced safely in the
diazepam 30 mg in divided doses over 24 hours) may management of most patients who have recovered from
help with muscle rigidity. an episode of NMS but a significant risk of recurrence
• Intravenous dantrolene (0.5–3 mg/kg bodyweight does exist, dependent in part on the time elapsed since
i.v./6–8 h or 100–200 mg/day po). recovery and the dose and potency of the neuroleptic.
• Dopamine agonists: amantadine; bromocriptine
(2.5–20 mg/8 h po); levodopa/carbidopa. PREVENTION
• Electroconvulsive therapy may be successful in some • A careful drug history, physical examination, and
cases during and after NMS. laboratory evaluation should be obtained prior to
neuroleptic administration.
PROGNOSIS • Indications for neuroleptics should be clear.
• The syndrome lasts 7–10 days in uncomplicated cases • Increases in dose should be made judiciously based on
receiving oral neuroleptics, recovering spontaneously symptom response, with alternative methods (e.g.
without the need for specific drugs. interpersonal interventions, benzodiazepines) used to
• It may develop earlier and take longer to resolve in optimize behavioral control and sedation.
patients with schizophrenic disorders. • Patients should be sedated and monitored to prevent
• Residual cerebellar damage may occur as a sequelae of agitation, exhaustion and dehydration.
the hyperpyrexia.

McAuley JH (2001) Does essential tremor origi- Matsuo H, Kamakura K, Saito M, et al. (1999)
FURTHER READING nate in the cerebral cortex? Lancet, 357: Familial paroxysmal dystonic choreoathetosis:
492–493. clinical findings in a large Japanese family and
Rehman H (2000) Diagnosis and management of genetic linkage to 2q. Arch. Neurol., 56:
DYSTONIA tremor. Arch. Neurol., 160: 2438–2444. 721–726.
Bressman SB (1998) Dystonia. Curr. Opin. Neu- Schrag A, Muenchau A, Bhatia KP, et al. (1999) Sethi KD (2000) Paroxysmal dyskinesias. The Neu-
rology, 11: 363–372. Overdiagnosis of essential tremor. Lancet, rologist, 6: 177–185.
Britton TC (1998) Torticollis – what is straight 353: 1498–1499. Vidailhet M (2000) Paroxysmal dyskinesias as a
ahead? Lancet, 351: 1223–1224. Schurrman PR, Bosch DA, Bossuyt PMM, et al. paradigm of paroxysmal movement disorders.
Hallett M (1998) The neurophysiology of dysto- (2000) A comparison of continuous thalamic Curr. Opin. Neurol. 13: 457–462.
nia. Arch. Neurol., 55: 601–603. stimulation and thalamotomy for suppression
Janavs JL, Aminoff MJ (1998) Dystonia and of severe tremor. N. Engl. J. Med., 342: TOURETTE SYNDROME
chorea in acquired systemic disorders. J. Neu- 461–468. Jankovic J (2001) Tourette’s syndrome. N. Engl.
rol. Neurosurg. Psychiatry, 65: 436–445. J. Med., 345: 1184–1192.
Krack P, Vercueil L (2001) Review of the func- CHOREA Kurlan R (1997) Diagnostic criteria for genetic
tional surgical treatment of dystonia. Eur. J. Janavs JL, Aminoff MJ (1998) Dystonia and studies of Tourette syndrome. Arch. Neurol.,
Neurol., 8: 398–399. chorea in acquired systemic disorders. J. Neu- 54: 517–518.
Marsden CD, Quinn NP (1990) The dystonias. rol. Neurosurg. Psychiatry, 65: 436–445. Kurlan R (2001) New treatment for tics? Neurol-
BMJ, 300: 139–144. ogy, 56: 580–581.
Misbahuddin A, Warner TT (2001) Dystonia: an MYOCLONUS Marras C, Andrews D, Sime E, Lang AE (2001)
update on genetics and treatment. Curr. Opin. Caviness JN (1996) Myoclonus. Mayo Clin. Proc., Botulinum toxin for simple motor tics. A ran-
Neurol., 14: 471–475. 71: 679–688. domized, double-blind, controlled clinical
Steinberger D, Korinthenberg R, Topka H, et al. Caviness JN, Alving LI, Maraganore DM, Black trial. Neurology, 56: 605–610.
(2000). Dopa-responsive dystonia: Mutation RA, McDonnell SK, Rocca WA (1999) The Palumbo D, Maughan A, Kurlan R (1997) Hy-
analysis of GCH1 and analysis of therapeutic incidence and prevalence of myoclonus in pothesis III. Tourette syndrome is only one of
doses of L–DOPA. Neurology, 55: 1735–1737. Olmsted County, Minnesota. Mayo Clin. several causes of a developmental basal ganglia
Warner TT, Jarman P (1998) The molecular ge- Proc., 74: 565–569. syndrome. Arch. Neurol., 54: 475–483.
netics of the dystonias. J. Neurol. Neurosurg. Singer HS, Wendlandt BS, Krieger M, Giuliano J
Psychiatry, 64: 427–429. DYSKINESIAS (2001) Baclofen treatment in Tourette syn-
Bennett LB, Roach ES, Bowcock AM (2000) A drome. A double-blind, placebo-controlled,
ESSENTIAL TREMOR locus for paroxysmal kinesigenic dyskinesia crossover trial. Neurology, 56: 599–604.
Deuschl G (2000) New treatment options for maps to human chromosome 16. Neurology,
tremors. N. Engl. J. Med., 342: 505–506. 54: 125–130. NEUROLEPTIC MALIGNANT SYNDROME
Deuschl G, Wenzelburger R, Raethjen J (2000) Bhatia KP (1999) The paroxysmal dyskinesias. J. Balzan MV (1998) The neurolept malignant syn-
Tremor. Curr. Opin. Neurol., 13: 437–443. Neurol., 246: 149–155. drome: a logical approach to the patient with
Elble RJ (2000) Origins of tremor. Lancet, 355: Demirkiran M, Jankovic J (1995) Paroxysmal temperature and rigidity. Postgrad. Med. J., 74:
1112–1113. dyskinesias: Clinical features and classification. 72–76.
Finkel MF (2000) Pramipexole is a possible effec- Ann. Neurol., 38: 571–579. Birbeck GL, Kaplan PW (1999) Serotonin syn-
tive treatment for primary orthostatic tremor Filion M (2000) Physiologic basis of dyskinesia. drome: a frequently missed diagnosis? Let the
(shaky leg syndrome). Arch. Neurol., 57: Ann. Neurol., 47 (Suppl 1): S35–S41. neurologist beware. The Neurologist, 5:
1519–1520. Loher TJ, Krauss JK, Burgunder J–M, et al. 279–285.
Louis ED, Ford B, Barnes LF (2000) Clinical sub- (2001) Chronic thalamic stimulation for treat- Hernandez JL, Palacious-Araus L, Echevarria S, et
types of essential tremor. Arch. Neurol., 57: ment of dystonic paroxysmal nonkinesigenic al. (1997) Neuroleptic malignant syndrome in
1194–1198. dyskinesia. Neurology, 56: 268–270. acquired immunodeficiency syndrome. Post-
Hellwig B, Häubler S, Schelter B, et al. (2001) grad. Med. J., 73: 779–784.
Tremor-correlated cortical activity in essential
tremor. Lancet, 357: 519–523.
136 Chapter Seven

Developmental
Diseases of the
Nervous System
ARNOLD–CHIARI MALFORMATION Type II
Cerebello-medullary malformation with meningomyelocele
Cerebellum and medulla are displaced inferiorly and are
DEFINITION deformed (131). The medulla is elongated and often folded
A number of developmental anomalies of the hindbrain, upon itself in an S-shaped pattern or overlies the cervical
base of skull, and upper cervical canal characterized by cord, which may be small with upward directed roots.
caudal displacement of the cerebellar tonsils, and sometimes Meningomyelocele (protrusion of spinal cord/cauda equina,
more of the cerebellum and lower brainstem, through the dura and arachnoid through a defect in the vertebral lamina,
foramen magnum into the cervical spinal canal. forming a cystic swelling) is invariably present (see p.140).

CLASSIFICATION OF CHIARI MALFORMATIONS Type III


Type I Cerebellar herniation with a high cervical or occipito-cervical
Cerebello-medullary malformation meningomyelocele
Herniation of the cerebellar tonsils below the foramen Includes cervical spina bifida, cerebellar herniation through
magnum into the cervical spinal canal with elongation of the defect, and an open, dystrophic posterior fossa. Rarely
the medulla (126, 127). Meningomyelocele is rare. compatible with post-natal life.
Hydrocephalus (see p.468) and syringomyelia (see p.541)
may develop, the latter in up to 50% of cases (128–130).

126 127

126 MRI brain sagittal T1W image of a 40 year old lady with altered 127 Chiari type I malformation: midline sagittal MR T1W imaging
sensation to pain and temperature over the right chest from T4 to T9, (TR=500 ms,TE=15 ms) showing caudal displacement of cerebellar
showing Chiari malformation with herniated cerebellar tonsils (arrow) tissue, with herniation of the cerebellar tonsils below the foramen
within the foramen magnum and reaching the midlevel of the posterior magnum (arrow).
arch of C1, cervical syrinx (arrowheads), and syringobulbia (short
arrow) with the tract extending upwards in the midline of the ventral
medulla to the pontomedullary junction.
Arnold–Chiari Malformation 137

128 129 128, 129 MRI cervical and upper thoracic


spine, sagittal T1W images, showing herniated
cerebellar tonsils (arrow) and a large syringo-
hydro-myelia (arrowheads) extending down
to the upper end plate of T9.

130 MRI thoracic spine, axial gradient echo


image, showing large hyperintense (white,
arrows) central syringo-hydro-myelia
occupying most of the thoracic spinal cord
in cross-section.

Type IV 130
Cerebellar hypoplasia only
May be related to or equivalent to Dandy–Walker
malformation.

EPIDEMIOLOGY
• Incidence: uncommon.
• Age of onset of symptoms: type II: infancy, type I: teens
or young adulthood.
• Gender: M=F.

PATHOLOGY
• Caudal descent of the cerebellar tonsils.
• Medulla and pons elongated.
• Medulla and cerebellum occlude foramen magnum.
• Remainder of cerebellum, which is small, is also
displaced, obliterating the cisterna magna.
• Fibrosis of arachnoid tissue around herniated brainstem
and cerebellum.
• A kink or spur in the upper cervical spinal cord, pushed
posteriorly by the lower end of the fourth ventricle. In this
type of malformation, a meningomyelocele is nearly always
found, and hydromyelia of the cervical cord is common. • Aqueduct stenosis.
• Hydrocephalus (due to aqueduct stenosis or CSF
Associations outflow obstruction at base of the brain).
Bony abnormalities • Syringomyelia and syringobulbia (126, 128–130).
• Posterior fossa: small. • Lumbosacral meningocele or meningomyelocele (see
• Foramen magnum: enlarged and grooved posteriorly. p.140).
• Base of skull: flattened or infolded by the cervical spine. • Filum terminale: lower spinal cord may extend to the
• Fused cervical vertebrae (Klippel–Feil syndrome). sacrum.
• Spinal dysraphism (see p.139). • Others, such as fusion of the corpora quadrigemina, cysts
of the foramen of Magendie, upward herniation of the
CNS abnormalities cerebellum through an abnormally large tentorial notch,
• Developmental abnormalities of the cerebrum: poly- enlargement of the mass intermedia, fusion of the
microgyria. thalami, and hydromyelia.
138 Developmental Diseases of the Nervous System

ETIOLOGY AND PATHOPHYSIOLOGY • Cerebellar ataxia (predominantly affecting the legs and
• Unknown. later the arms).
• Probably a failure of coordinated development of the • Downbeat nystagmus.
brainstem, cerebellum and upper cervical spinal cord. • Weakness and wasting of the tongue, sternomastoid and
• Arnold–Chiari malformations are traditionally viewed as trapezius muscles due to lower cranial nerve palsies (XI
a congenital anomaly but may be acquired. One and XII).
hypothesis is that the CSF pressure difference between • Recurrent vocal cord paralysis, laryngeal stridor, or
the spinal and cranial compartments causes tonsillar respiratory obstruction during physical activity, due to
herniation. Others suggest that a mismatch between the periodic venous congestion in the medulla.
volume of the posterior fossa and its tissue contents may
produce downward herniation of the cerebellar tonsils. Some remain asymptomatic well into adult life and
present later with pain in the back of the head, neck,
CLINICAL FEATURES shoulders and upper limbs, exacerbated by head movement
Chiari type 1 malformation and cough, sneeze or strain (‘hindbrain hernia headache’).
May be asymptomatic or cause symptoms and signs of This may be followed by the evolution of weakness and
progressive dysfunction of the: spasticity in the limbs, gait ataxia and difficulty swallowing.
• Cerebellum (progressive cerebellar ataxia), Clinical features of syringomyelia (see p.541) may also
• Medulla (spastic quadriparesis), or coexist.
• Lower cranial nerves (dysphagia),
at any time during life, sometimes being delayed until adult DIFFERENTIAL DIAGNOSIS
life. • Multiple sclerosis.
• Tumor of the high cervical cord or lower medulla
Other progressive symptoms include those of: (foramen magnum).
• Raised intracranial pressure (e.g. headache) due to • Motor neuron disease (Chiari I may present with
hydrocephalus (see p.468), and progressive severe bulbar palsy and generalized
• Syringomyelia (see p.541). hyperreflexia due to severe stretch injury to lower cranial
nerves [caused by caudal displacement of medulla]
Symptoms occasionally arise acutely, such as headache or and/or direct brainstem compression).
vertigo and ataxia following neck extension (e.g. dental • Autosomal dominant cerebellar ataxia of late onset.
extraction and chiropractic manipulation), coughing, • Dandy–Walker syndrome: a developmental anomaly with
sneezing and Valsalva maneuver (inducing hindbrain cystic dilatation of, or near, the fourth ventricle. It causes
herniation); and syncope on exertion. obstructive hydrocephalus, cerebellar and/or brainstem
signs.
Examination findings include:
• A short ‘bull’ neck in about 25% of cases. INVESTIGATIONS
• Low hairline. MR scan of the brain and cervical spinal cord is the
• Torsional or downbeating nystagmus with neck extension. investigation of choice
• Permanent signs of cerebellar, medulla, high cervical
cord or lower cranial nerve dysfunction. Chiari I malformation
• T1W midline sagittal MRI (or myelography) shows the
Downbeat nystagmus is usually in the primary position of inferior displacement of the cerebellar tonsils, the tips
gaze, and increases in gaze slightly below horizontal, lying in the C1–C2 region (126, 127).
especially in gaze down and laterally. It is due to a disturbance • The tonsils may lie up to 5 mm below the foramen
of the vestibulo-cerebellar pathways and is highly suggestive magnum in normal individuals – this is a fairly frequent
of a craniocervical junction disorder such as Arnold–Chiari finding.
malformation, basilar invagination or ankylosing spondylitis. • The tonsils usually appear ‘pointed’ rather than rounded
However, it may also be seen in cerebellar disease (when as in the normal.
other cerebellar eye signs are usually present), drug intoxi- • The position of the brainstem and fourth ventricle and
cation (phenytoin, carbamazepine, lithium), brainstem the rest of the brain should appear normal in
encephalitis, magnesium depletion, communicating hydro- uncomplicated type 1.
cephalus, Wernicke’s encephalopathy, multiple sclerosis, • Syringomyelia may be present in 20–73% of cases (126,
vascular disease and as a paraneoplastic phenomenon. It may 128–130).
coexist with periodic alternating nystagmus. • May be associated with C2 and C3 fusion (Klippel–Feil
syndrome), short clivus or odontoid or C1 abnormalities.
Chiari type II malformation
Usually presents in infancy with: Chiari II (the Arnold–Chiari malformation)
• Progressive enlargement of the head, due to Midline sagittal and axial images of the brain and spinal cord
hydrocephalus, and are required.
• Spinal meningomyelocele.
Brain:
Diagnosis may be delayed until the spinal cord is • The cerebellar tonsils, vermis and brainstem are herniated
compressed at the cranio-cervical junction causing: through the foramen magnum and the fourth ventricle
• Spastic quadriparesis (limb spasticity, hyperreflexia and outlets are obstructed causing hydrocephalus (131).
extensor plantar responses). • The cervicomedullary junction may be kinked.
Rachischisis (Dysraphism) 139

• The frontal horns of the lateral ventricles are squared-off. RACHISCHISIS (DYSRAPHISM)
• The fourth ventricle is compressed.
• The aqueduct is stretched.
• The superior aspect of the cerebellum is herniated DEFINITION
superiorly through the tentorial hiatus (which is Defective closure of the neural groove (dysraphism).
enlarged), i.e. the whole posterior fossa is too small.
• Corpus callosum agenesis or partial abnormalities are EPIDEMIOLOGY
frequent. • Incidence: the incidence varies widely from one country
• Heterotopias and abnormal gyral patterns are common. to another and is about 1 per 1000. The disorder is more
likely to occur in a second child if one child has already
Spinal cord: been affected (the incidence rises from 1 per 1000 to
• A lumbosacral myelomeningocele is nearly always present 40–50 per 1000). Abnormalities of closure of the
(see p.140). cranium are more frequent than defects of closure of the
• The cord is nearly always tethered. vertebral arches. Even so, spina bifida occulta is found in
• A lipoma of the filum terminale may be present. 5% of the normal population. Meningomyelocele is 10
times more frequent than meningocele.
Less optimal imaging techniques: • Age: infants and children predominantly.
• Plain skull and cervical spine x-rays may show cranial
enlargement, a small posterior fossa with low tentorial ETIOLOGY AND PATHOPHYSIOLOGY
insertion and dural sinuses, basilar impression (platy- Some degree of failure occurs during the first 3 weeks of
basia), enlargement of the foramen magnum, elongation fetal development of the folding and fusion of the dorsal
of cervical arches, widened cervical canal, fusion of midline structures of the primitive neural tube with its
cervical vertebrae, and Klippel–Feil anomaly. normal covering of meninges, bone and skin. Genetic and
• Myelography followed by CT scanning of the cranio- environmental factors are thought to interact. A genetically-
cervical junction (the contrast material in the subarach- determined variant of the 5,10- methylenetetrahydrofolate
noid space helps to reveal the abnormality). reductase gene that specifies a product with reduced enzyme
• Vertebral angiography. activity is associated with NTDs. Folic acid reduces the risk
of NTDs, not by correcting a simple nutritional deficiency
TREATMENT but by overcoming the reduced activity in the conversion of
• The meningomyelocele and hydrocephalus should be 5,10-methylenetetrahydrofolate to 5 methyltetrahydrofolate.
treated promptly in an affected infant.
• When signs of progressive neurologic deficit appear in PATHOLOGY
later life, hydrocephalus (if present) should be treated Brain
surgically by shunting and suboccipital craniectomy and Anencephaly
upper cervical (C1) decompression laminectomy may Absence of cranial vault and its contents (132). The
help decompress the region. undeveloped brain lies in the base of the skull as a small
• If clinical progression is slight or uncertain, a conserva- vascular mass without recognizable nervous tissue. It is the
tive approach is recommended, with ongoing obser- most frequent of the rachischises. It has many associations
vation and reassessment. with other conditions in which the vertebral laminae fail to
fuse. It is incompatible with life.

131 132

131 Chiari II malformation.T1W midline sagittal view of the brain and 132 Pathologic specimen, sagittal section, of a dead fetus with
upper cervical cord shows marked abnormality of the brainstem and anencephaly, showing absence of the cranial vault and its contents.
cerebellum with downwards displacement of the cerebellum and no
true fourth ventricle.
140 Developmental Diseases of the Nervous System

Cranial meningocele dilated fourth ventricle, expands in the midline, causing the
Meninges protrude through a defect in the skull. occipital bone to bulge posteriorly and to displace the
tentorium and torcula upward. In addition, the corpus
Meningoencephalocele callosum may be deficient or absent, and there is dilatation
Meninges and brain parenchyma protrude through a defect of the aqueduct, third and lateral ventricles.
in the skull.
Spinal cord
Dandy–Walker syndrome In the spine, dysraphism is most common in the lumbo-
A failure of development of the midline portion of the sacral area.
cerebellum. A cyst-like structure, representing the greatly
Spina bifida occulta
• Closure defect, due to failure of fusion, of one or more
133 vertebral arches.
• The spinal cord remains inside the vertebral canal and
there is no external sac.
• A congenital dural sinus tract occasionally communicates
the skin with the lumbar CSF sac via the incompletely
closed vertebral arch, and predisposes to recurrent
bacterial meningitis.
• Lipomas or dermoids in the cauda equina region may be
present and may not become symptomatic until later life.
• A skin dimple, vascular anomaly or tuft of hair may be
present over the site of the lesion in the low back in the
midline (133).
• Tethering of the spinal cord: may become symptomatic
in children and sometimes adolescents (see p.142).

133 Photograph of the low back of a patient with spina bifida occulta
showing a tuft of hair over the site of the spina bifida occulta, and a
vertical midline scar following surgical resection of a cauda equina
lipoma that was causing neurologic symptoms and signs.

134 135 136

134, 135 MRI lumbosacral spine, sagittal T1 (134) and dual echo T2W (135) images showing a
lumbosacral meningomyelocele and narrowing of the elongated lumbar cord with cord
tethering.There is no lipomatous or other soft tissue mass related to the spinal dysraphism. Disc
degeneration is noted incidentally at L2/3 and L3/4.

136 MRI cervical spine, sagittal T1W image, of the same patient in 134, 135, showing inferior displacement of the medulla and cerebellum and
elongation of the medulla (Chiari II malformation).
Rachischisis (Dysraphism) 141

Meningocele • Congenital dislocation of the odontoid process and atlas.


Meninges (dura and arachnoid) protrude through a defect • Platybasia and basilar impression.
in the vertebral arch as a cystic CSF-filled dural sac. The
cord remains in the canal. Seldom are there any neurologic Complications
consequences. • Recurrent ascending bacterial meningitis due to the
presence of a sinus tract from skin to lumbar CSF.
Meningomyelocele • Progressive hydrocephalus from an often associated
Meninges and spinal cord or (more commonly the cauda Chiari malformation.
equina) roots protrude through the defect in the neural arch
as a dural sac containing spinal cord or roots closely applied CLINICAL FEATURES
to the fundus of the cystic swelling (134, 135) . It may be Cranial meningoencephalocele
associated with overlying dermal defects, Arnold–Chiari Small encephaloceles protruding into the nasal cavity may
malformation (136), or hydrocephalus. Motor, sensory and cause no neurologic signs unless innocently snipped off,
bladder disturbances are common. when CSF rhinorrhea may result. Larger occipital
encephaloceles may be associated with blindness, ataxia, and
Spina bifida aperta (myeloaraphia/rachischisis) mental retardation.
Exposed neural tube without overlying leptomeninges,
vertebral arch or dermis. The dural sac is open and neural Lumbosacral meningomyelocele
tissue is exposed on the back. Severe neurologic deficits are Lumbosacral polyradiculopathy
always present. The child is typically born with a lumbosacral meningo-
myelocele covered by delicate, weeping skin; sometimes it
Diastematomyelia may have ruptured in utero or during birth. If the sac
A septum (bony spicule or fibrous band) protrudes into the contains elements of spinal cord or cauda equina, there is
spinal canal from the body of one of the thoracic or upper neurologic dysfunction appropriate to the level of the lesion.
lumbar vertebrae and through the spinal cord in an If lumbosacral, the legs do not move unless the sac is
anterior–posterior direction, thus dividing the spinal cord into stroked, which may elicit involuntary movements of the
two for a variable vertical extent (137, 138). The division of legs. The deep tendon reflexes are absent, there is no
the cord may be complete, each half with its own dural sac and response to pinprick over the lumbosacral dermatomes and
set of nerve roots. This longitudinal fissuring and doubleness urine constantly dribbles. If entirely sacral, bladder and
of the cord is referred to as diplomyelia. With growth this leads bowel sphincters are affected but the legs are not.
to a traction myelopathy. Often associated with spina bifida.
Progressive spastic weakness of the leg muscles
Associations Stretching of the spinal cord, which is securely attached to
• Hydrocephalus as a result of aqueduct stenosis. the lumbar vertebrae, during the period of rapid
• Posterior fossa abnormalities. lengthening of the vertebral column (see Tethered cord
• Syringomyelia. syndrome, p.142).
• Klippel–Feil syndrome (fusion of cervical vertebrae or of
atlas and occiput). Acute or progressive cauda equina syndrome
Sudden or repeated stretching of the implicated sensory and
motor nerve roots.
137, 138 Diastematomyelia. 137
T2W midline sagittal (137), Syringomyelia (see p.541)
and T1W axial (138) MRI of
the lumbar spine show a Spina bifida occulta
bony bar (arrow) across the No clinical symptoms. May have overlying tuft of hair
center of the spinal canal (133), discoloration, dimple or dermal sinus.
with neural tissue
(arrowheads) passing round
it. Note also the spinal canal
is abnormally wide and the 138
visible vertebral bodies and
posterior elements are
misshapen.
142 Developmental Diseases of the Nervous System

Tethered cord syndrome DIAGNOSIS


• Derives from adhesion of the conus to one of the lower lum- Pre-natal
bar vertebrae resulting in malascent of the conus medullaris. • Amniotic fluid alpha-fetoprotein and acetylcholinesterase
• Causes: immunoassay (removed at 15–16 weeks of pregnancy);
– Intradural fibrous adhesions. blood contamination is a source of error.
– Diastematomyelia. • Uterine ultrasound.
– Intradural lipomas.
– Dermal sinus tracts. Post-natal
– Tight filum terminale. Clinical ± radiologic (see Features, above).
• Clinical features:
– Onset: may be delayed until adolescence or even later if TREATMENT
low traction on conus; may require additional mechanical Prevention
triggers if low traction on conus. • Genetic screening could identify women who will require
– Progressive sensorimotor (segmental) deficits in the folic acid supplements to reduce their risk of having a
lower limbs (in about three-quarters of cases). child with a NTD.
– Severe pain: perineogluteal region and non-dermatomal • Folic acid supplementation during pregnancy.
leg pain (three-quarters). • Termination of pregnancy if the diagnosis is established
– Cutaneous stigmata of spinal dysraphism, such as at 16 weeks pregnancy and parents agree to a termination.
subcutaneous lipoma, midline hypertrichosis, sacral nevus
(two-thirds). Conservative
– Bowel and bladder dysfunction (two-thirds). If high spinal lesions and total paraplegia, kyphosis,
– Mild upper motor neuron signs coexisting with hydrocephalus, and other major congenital anomalies,
symptoms of conus dysfunction (one-sixth). because of poor prognosis (see below).

Associated hydrocephalus Meningomyelocele


• Rapid increase in head size. Excise in the first few days of life if the objective is to
• Downward deviation of the eyes (‘setting sun’ eyes) due prevent fatal meningitis.
to upgaze paresis as a result of pressure on the posterior
commissure in the rostral midbrain. Hydrocephalus
Ventriculoperitoneal or ventriculoatrial shunt is used if
INVESTIGATIONS hydrocephalus needs to be decompressed.
Meningoceles/meningomyeloceles
• Plain x-rays of the relevant part of the spine are useful to PROGNOSIS
delineate the vertebral abnormalities (e.g. lumbosacral spine Less than 30% of patients with high spinal lesions and total
x-rays may show failure of fusion of one or more vertebral paraplegia, kyphosis, hydrocephalus and other major
arches, hemivertebrae, block vertebrae, diastematomyelia). congenital anomalies survive beyond 1 year. 80–90% of the
• CT brain scan (+/– intrathecal contrast) may further survivors are totally dependent on others for their care because
delineate bony elements and soft tissues (ideally with 2D of some degree of intellectual handicap and paraplegia.
and 3D reconstructions) and may reveal enlarged
ventricles due to hydrocephalus.
• MRI spine (midline sagittal T1 and T2W plus axials of
the relevant part) is the investigation of choice for
imaging the cord, meningocele contents (cord elements
or just CSF) and associated abnormalities such as
tethering of the cord, lipomas of the filum terminale, or
dermoids in the cauda equina region (134, 135).
• Beware of the dangers of lumbar puncture: iatrogenic
damage to the spinal cord by a needle inserted into a
spinal cord that is tethered to the margin of one of the
low lumbar or sacral vertebral bodies.

Diastematomyelia
• Plain x-rays may demonstrate the bony spur plus
associated anomalies.
• CT with intrathecal contrast will demonstrate the
relationship of the bony spur to the cord, nerve roots
and thecal sac.
• MRI is really the investigation of choice (see
Meningocele). It will demonstrate the position of the
cord-tethering and splitting (137, 138).
Tuberous Sclerosis 143

TUBEROUS SCLEROSIS Skin


• Hypomelanotic macules are the first skin lesions to
appear in about 90% of cases of TSC. They vary from a
DEFINITION few millimeters to several centimeters in size, have an
Tuberous sclerosis complex (TSC) is a neurocutaneous oval ash leaf shape with one end round and the other
syndrome characterized by intellectual handicap, epileptic pointed, are arranged in a linear fashion over the trunk
seizures, and hamartomas affecting multiple organs systems, or limbs, contain sweat glands, and become pink when
including skin, kidney, brain and heart. rubbed. Because of the reduced number and function of
melanoblasts, which normally absorb light in the
EPIDEMIOLOGY ultraviolet range, these lesions are more readily seen with
• Prevalence: 3.7 per 100 000 population in Scotland, but a Wood’s lamp, which transmits only ultraviolet rays.
may be as high as 8–9 per 100 000. • Adenoma sebaceum are small, red-pinkish-yellow, wart-
• Age: infancy and childhood. like skin lesions with a smooth glistening surface, about
• The disease may be present clinically and radiologically 2 mm in size, which tend to be limited to the nasolabial
at birth but is usually not recognized until 2–3 years of folds, cheeks, and chin; sometimes also involving the
age when delay in reaching milestones of natural forehead and scalp. They are angiofibromas characterized
maturation, mental retardation, or epileptic seizures by hyperplasia of connective and vascular tissue.
become evident. Adenoma sebaceum appear later, usually • Shagreen patches are thick, slightly elevated, flesh-
between 4–10 years of age, and progress thereafter. colored yellowish skin, 1–10 cm (0.4–4 in) in diameter,
• Gender: M=F. with a ‘pigskin’, ‘orange peel’, or ‘elephant hide’ appear-
ance, over the lower trunk, most often in the lumbosacral
PATHOLOGY region. These are plaques of subepidermal fibrosis.
The TSC affects all tissues, including the lymphatic system, • Subungual fibromas are fibromatous involvement of the
with the possible exceptions of the peripheral nervous nail bed, which usually appear at puberty and continue
system, meninges, skeletal muscle and pineal gland. to develop with age. Other skin changes include café-au-
lait spots, fibroepithelial tags (soft fibromas), and port
Brain wine hemangiomas.
Cortical tubers and subependymal nodules
Macroscopically, the brain is normal in size but may show Other
areas (5 mm–3 cm [0.2–1.2 in]) of broadening, unnatural There is an increased incidence of:
whiteness, and firmness on the surface of the cerebral cortex • Glial tumors.
(‘cortical tubers’). Their cut surface reveals a lack of demarc- • Hydrocephalus.
ation of cortex from white matter and the presence of white • Spina bifida.
flecks of calcium (‘brain stones’). The surface of the lateral • Rhabdomyomas of the skeletal muscle and heart.
ventricles may be encrusted with white or pink-white masses • Endocrine tumors.
(‘subependymal nodules’) resembling candle gutterings. • Cyst formation in the kidneys, pancreas, and liver.
Rarely, nodules of abnormal tissue are seen in the basal • A honeycomb appearance of the lung.
ganglia, thalamus, brainstem, cerebellum and spinal cord. • Phakomas of the retina: composed mainly of neuronal
Microscopically, the tubers are composed of interlacing and glial components.
rows of plump fibrous astrocytes (like an astrocytoma but
lacking in glial fibrillar protein). The structure of the ETIOLOGY AND PATHOPHYSIOLOGY
cerebral cortex and ganglionic structures is diffusely • Familial (i.e. inherited as an autosomal dominant or
disturbed with gliosis and the presence of atypical recessive trait) in about 40% of cases, and sporadic (i.e.
monstrous neurons and giant glial cell forms. Nodules new mutations) in at least 60% of cases.
composed of masses of subependymal glial cells intermixed • Genetic heterogeneity: mutations in two genes. One
with distorted neurons or giant glial cells protrude into the gene, TSC1, is located on the long arm of chromosome
ventricles. These gliomatous deposits may obstruct the 9 (9q34), and a second gene, TSC2, on chromosome
foramina of Monro or the aqueduct or floor of the fourth 16p13.3.
ventricle, causing hydrocephalus. • About 50% of TSC families show genetic linkage to
The cortical tubers and subependymal nodules both TSC1 and 50% to TSC2.
show histologic evidence of abnormal growth and • Among sporadic cases, mutations in TSC2 are more
migration, and occasionally they give rise to subependymal frequent and often accompanied by more severe
giant-cell astrocytomas. Neoplastic transformation of neurologic deficits.
abnormal glial cells usually takes the form of a large-cell • Multiple mutational subtypes have been identified in the
astrocytoma, but may, less commonly, give rise to a TSC1 and TSC2 genes.
glioblastoma or a meningioma. Calcification may occur. • The TSC1 (chromosome 9) and TSC2 (chromosome
16) genes encode distinct proteins, hamartin and tuberin,
respectively, which are widely expressed in the brain and
may interact as part of a cascade pathway that modulates
cellular differentiation, tumor suppression, and intra-
cellular signalling. Tuberin has a GTPase activating
protein-related domain that may contribute to a role in
cell cycle passage and intracellular vesicular trafficking.
144 Developmental Diseases of the Nervous System

CLINICAL FEATURES DIFFERENTIAL DIAGNOSIS


Clinically extremely variable with manifestations ranging from • Mental retardation:
very severe illness to absence of symptoms (i.e. the forme – Acquired destructive lesions: obstructive hydrocephalus.
fruste). Considerable variation is also observed within families. – Chromosomal abnormalities.
• The usual presenting features in childhood are delay in – Multiple congenital anomalies: rubella.
reaching milestones, progressive intellectual handicap and – Developmental abnormality of the brain.
epileptic seizures that are difficult to control, but only – Metabolic and endocrine disease: cretinism.
one aspect may be prominent. Over half of affected – Progressive neurodegenerative disease: lipidoses.
individuals have normal intelligence and a quarter do not – Psychosis.
have seizures. • Epilepsy (see p.65).
• The seizures initially take the form of salaam spasms (i.e. • Adenoma sebaceum: acne vulgaris.
flexion myoclonus with hypsarrhythmia [irregular • Other neurocutaneous syndromes:
dysrhythmic bursts of high-amplitude spikes and slow – Sturge–Weber syndrome (see p.151).
waves in the EEG]), later becoming atypical absence or – Von Hippel–Lindau disease (see p.392).
more typical generalized seizures. – Neurofibromatosis (see p.147).
• Focal neurologic deficits and seizures may arise as a result – Ataxia telangiectasia (see p.444)
of brain tumors.
• Dystonia and athetosis may also occur. INVESTIGATIONS
• Behavioral and affective disorders are present in about CT scan or MRI scan
half of patients. CT scan (141–144) or, preferably, MRI scan (145) of the
• Hypomelanotic macules may be visible on the skin using brain may reveal:
a Wood’s light. • Cortical and subcortical tubers (50% calcified on CT, 3%
• Adenoma sebaceum typically occurs in childhood over the enhance on MR).
cheeks and bridge of the nose in a butterfly distribution • Subependymal nodules (80% calcified on CT, 30%
(139). In some patients it may be more subtle and enhance on MR).
restricted to a few lesions in the nasolabial folds. • White matter lesions: seen on MRI as curvilinear or
• Gingival and subungual (beneath the fingernails) wedge-shaped bands radiating from the ventricles; 12%
fibromas begin to appear in adolescence (140). enhance.
• Phakomata (white nodules) appear in the retina on • Atrophy and enlarged ventricles.
ophthalmoscopy. • Giant cell astrocytoma: usually near the foramen of
• Other congenital abnormalities such as spina bifida and Monro (145). These are larger than the other subepen-
hydrocephalus may be present. dymal nodules and grow and obstruct the foramina. On
• Family history may be revealing. CT or MR a prominent enhancing nodule near the
foramen of Monro is likely to be a giant cell astrocytoma.

139 140

140 Ungual fibroma adjacent to the lateral border of the nail bed
of the left great toe and third toe in a patient with tuberous
sclerosis. (Courtesy of Dr AM Chancellor,Tauranga, New Zealand.)

139 Photograph of the face of a patient with tuberous sclerosis


showing diffuse small adenoma sebaceum (pink, wart-like
angiofibromas) throughout the cheeks, chin and forehead, and larger
lesions in the nasolabial fold on the left. (Courtesy of Dr AM
Chancellor,Tauranga, New Zealand.)
Tuberous Sclerosis 145

141 142 143

141–143 CT scan of the brain, non-contrast, axial slices, showing small white calcified subependymal nodules in the temporal horns bilaterally
(141), frontal horn of the right lateral ventricle and posterior horn of the left lateral ventricle (142), and the lateral ventricles bilaterally (143)
in a patient with tuberous sclerosis.

144 145

145 T1W axial MRI post-contrast showing a


mass at the foramen of Monro (arrow) with
marked contrast enhancement and mixed
signal features in a patient with tuberous
sclerosis.There is secondary hydrocephalus
144 CT scan of the brain, non-contrast, axial slices, showing small white calcified subependymal (the lateral ventricles are very large).
nodules in the frontal horn of the right lateral ventricle, and the lateral ventricles bilaterally
(arrows), not to be confused with the calcified pineal gland centrally (short arrow) and calcified
choroid plexus laterally (arrowheads). (Courtesy of Dr AM Chancellor,Tauranga, New Zealand.)
146 Developmental Diseases of the Nervous System

EKG PREVENTION
Counselling of affected individuals against childbearing.
Echocardiograph
?Cardiac rhabdomyoma. PROGNOSIS
• Slow progression, mainly in cognitive function,
Abdominal ultrasound ultimately leading to death in adolescence or early adult
Liver, kidneys. life.
• Status epilepticus is now a less common cause of death,
Blood with better control of epilepsy.
DNA analysis for mutations in the TS genes located on • Malignant gliomas cause a significant minority of deaths.
chromosomes 9 (TSC1) and 16 (TSC2). • Incomplete forms of TS may have a better prognosis.

DIAGNOSIS
Definite TSC
Two major diagnostic features or one major plus two minor
features.

Probable TSC
One major plus one minor feature.

Possible TSC
One major or two or more minor features.

Major diagnostic features


• Facial angiofibromas (adenoma sebaceum).
• Ungual fibromas.
• Retinal hamartomas.
• Hypomelanotic macules (three or more).
• Shagreen patch.
• Renal angiomyolipoma.
• Cardiac rhabdomyoma.
• Pulmonary lymphangiomatosis.
• Subependymal nodules.
• Subependymal giant cell astrocytomas.
• Tubers.

Minor diagnostic features


• Dental enamel pits in deciduous or permanent teeth.
• Bone cysts.
• Renal cysts.
• Gingival fibromas.
• Hamartomatous rectal polyps.

The diagnosis of people with minimal disease expression


(the forme fruste) can be very difficult. In children the
phenotype is often incomplete or not fully assessable. Hence
mildly affected subjects, at risk for severely affected
offspring, may remain undiagnosed. The detection of
(small) mutations in the TS genes located on chromosomes
9 (TSC1) and 16 (TSC2) has recently become possible and
may be helpful in the diagnosis of ambiguous cases.

TREATMENT
Symptomatic
• Epilepsy: anti-epileptic medication. It is rarely helpful to
attempt tumor excision, particularly in severely affected
individuals.
• Raised intracranial pressure: partial tumor excision or
ventricular decompression.
• Adenoma sebaceum: dermabrasion of facial lesions may
benefit patients who are not mentally impaired but they
regrow slowly.
Neurofibromatosis 147

NEUROFIBROMATOSIS ETIOLOGY AND PATHOPHYSIOLOGY


Autosomal dominant inheritance or spontaneous mutation.
About 50% of cases have no family history.
DEFINITION
The neurofibromatoses are a group of neurocutaneous NF-1
syndromes primarily affecting tissues derived from the neural NF-1 is caused by a mutation of the NF-1 gene located on
crest. They consist primarily of two distinct chromosome 17q11.2; 50–70% of patients inherit the
neuroectodermal disorders (phakomatoses) which are mutation as an autosomal dominant trait and 30–50% (i.e.
inherited as autosomal dominant traits and characterized by the remainder) of patients represent a new mutation. Full
localized overgrowths of ectodermal and mesodermal penetrance is exhibited.
elements in the nervous system and skin. The NF-1 gene codes for a large cytoplasmic protein of
2818 acids. The NF-1 gene product, neurofibromin,
CLASSIFICATION contains a small region in the central part of the protein that
• Neurofibromatosis 1 (NF-1): the more common type, bears sequence similarity with a family of proteins that
also previously called peripheral and von Reckling- regulate the proto-oncogene p21-ras. These p21-ras regu-
hausen’s neurofibromatosis. The responsible gene is latory proteins are collectively termed guanosine triphos-
located on chromosome 17q11.2, and is believed to phatase (GTPase)-activating proteins (GAPs). Since p21-ras
function as a tumor suppressor gene. The protein can transform cells, neurofibromin (which downregu-
product of the NF1 gene is called neurofibromin, a lates/inhibits p21-ras activity) suppresses the growth of cells.
protein that negatively regulates signals transduced by The identification of somatic mutations in NF-1 from tumor
Ras proteins. tissue suggests that NF-1 is probably a tumor suppressor
• Neurofibromatosis 2 (NF-2): the less common type, also gene. The mutation probably results in a loss of NF-1 gene
previously called central neurofibromatosis. The gene is expression, which leads to a loss of neurofibromin.
located on chromosome 22q12. Neurofibromin is hypothesized to function as a tumor
suppressor (negative growth regulator) by:
EPIDEMIOLOGY • Inactivating the p21-ras proto-oncogene, thus reducing
• Incidence: cell proliferation in some cell types, such as Schwann cells
– NF-1: 1 in 2500 live births. and astrocytes (in astrocytomas).
– NF-2: 1 in 33 000 live births. • Regulating microtubule signal transduction in the
• Prevalence: cytoskeleton.
– NF-1: 1 in 5000. • Influencing progression through the cell cycle and
– NF-2: 1 in 210 000. promoting growth arrest at the ‘commitment’ stage of
• Age: the cell cycle, causing cells to exit from the cell cycle and
– NF-1: pigmentary lesions are nearly always present at remain in G0/G1.
birth but neurofibromas are infrequent then. Both
lesions increase in number during late childhood and The loss of neurofibromin causes a loss of suppression of
adolescence. cell growth, predisposing to particular benign and malignant
– NF-2: symptoms usually begin in the ‘teens or early neoplasms, which arise primarily from cells of neural crest
twenties’, but occasionally as early as the first and as late origin (e.g. neurofibromas, neurofibrosarcomas, optic
as the seventh decade of life. gliomas, and pheochromocytoma) and also immature
• Gender: M=F. myeloid cells (e.g. juvenile myelomonocytic leukemia, and
the monosomy 7 syndrome, a childhood variant of myelo-
PATHOLOGY dysplasia). The exact mechanism remains uncertain however.
Skin
• Hyperpigmented lesions (café-au-lait patches) which are NF-2
characterized by an excess of melanosomes in the Inactivating mutations of both alleles of the NF-2 gene on
malpighian cells, which accounts for the dark color (there chromosome 22q12.1, consistent with the tumor suppressor
is no excess of melanocytes) and abnormally large gene hypothesis. The gene product is merlin.
melanosomes in some of the basal cells of the epidermis.
• Skin tumors: the collagen and elastin of the dermis is CLINICAL FEATURES
replaced by a loose arrangement of elongated connective Clinical expression is very variable.
tissue cells.
NF-1 type
Nervous system Skin pigmentation
• Nerve tumors: composed of a mixture of fibroblasts and Multiple (more than 5) brown skin macules appear shortly
Schwann cells, except optic gliomas which contain a after birth, usually on the trunk, but can occur any place on
combination of astrocytes and fibroblasts. the body, and vary in size from a millimeter or two to
• There is an increased incidence of brain tumors such as several centimeters, and in color from a light to a dark
gliomas, meningiomas, and acoustic neuromas. brown (café-au-lait spots). Axillary and inguinal freckles or
• Malignant degeneration of tumors occurs in 2–5% of diffuse pigmentation also occur.
cases, peripherally they become sarcomas and centrally,
astrocytomas or glioblastomas.
148 Developmental Diseases of the Nervous System

Skin neurofibromas • Headache.


These appear in late childhood or early adolescence. They • Epileptic seizures.
are situated in the dermis and form discrete, soft or firm • Short stature.
papules which are flesh-colored or violaceous, vary in size • Cranial bone defects with pulsating exophthalmos.
from a few millimeters to 1 or more centimeters, and come • Bone cysts.
in all shapes: flattened, sessile, pedunculated, conical and • Bone hypertrophy.
lobulated. When pressed, the soft tumors tend to invaginate • Pathologic fractures (pseudoarthrosis).
through a small opening in the skin: ‘buttonholing’. • Bony malformations of the spine.
• Kyphoscoliosis.
Plexiform neuromas • Syringomyelia.
Overgrowth of subcutaneous tissue, often in the face, scalp, • Stroke (ischemic).
neck and chest, which feel like a bag of worms or strings to
palpation. The underlying bone may enlarge. If there is Patients with NF-1 may have Schwann cell tumors on
overlying hyperpigmentation which extends to the midline, any nerve, but bilateral acoustic neuromas are virtually non-
suspect an intraspinal tumor at that level. existent in families with this disorder.

More deeply located neurofibromas NF-2 type


Firm discrete nodules may attach to a nerve which may Bilateral or multiple acoustic neurilemmomas
compress the spinal cord, nerve roots, brachial or • The hallmark.
lumbosacral plexus, and peripheral nerves and cause pain • The first symptom is usually unilateral loss of hearing,
and muscle weakness in the distribution of innervation of which is often first noticed when using a telephone.
the relevant nerves. For example, neurofibromas developing There may be a history of intermittent ringing or roaring
on spinal nerve roots may extend through the intervertebral in one or both ears or some unsteadiness, particularly
foramen in dumb bell fashion and compress the spinal cord when walking at night on uneven ground (when visual
(146–149). If not, they may attain considerable size in the cues are eliminated).
posterior mediastinum or retroperitoneal space. • Other symptoms include vertigo, facial weakness, sensory
change, headache, seizures, or a change in vision.
Vestibulocochlear (VIIIth cranial) nerve sheath neurofibroma • Symptoms are usually caused by pressure on the
Nerve deafness, dizziness, headache, staggering. vestibulocochlear and facial nerve complex.

Meningioma (see p.374) Other features


• Multiple CNS tumors:
Glioma (see p.364) – Neurilemmomas on cranial and spinal nerves.
– Intracranial and intraspinal meningiomas.
Optic nerve glioma (see p.372) – Astrocytoma of the optic nerve, brainstem, spinal cord.
Progressive monocular blindness, optic atrophy, nystagmus, • One or more café-au-lait spots; skin changes are less
enlargement of the optic foramen, abnormal contour of sella common than in type 1.
turcica, and failure to thrive (if the hypothalamus is • Subcutaneous neurofibromas.
invaded). • Lens opacity or cataracts.

Lisch nodules TERMINOLOGY


Nodules appear as small whitish pigmented iris hamartomas. Because NF-1 and NF-2 may have both central and
peripheral nervous system manifestations, the terms used
Choroidal abnormalities previously – ‘peripheral neurofibromatosis’ (for von
Bright, multiple, patchy regions at and around the entire Recklinghausen’s form) and ‘central neurofibromatosis’ (for
posterior pole of most, if not all, eyes when viewed by bilateral acoustic neuroma) – have been discarded as
infrared monochromatic light examination (but are not seen misleading and confusing.
under conventional ophthalmoscopic examination or
fluorescein angiography). These regions correspond to DIFFERENTIAL DIAGNOSIS
hypofluorescent areas on indocyanine-green angiography. • Normal patches of skin pigmentation: 10% of the
population have one or more café-au-lait spots, but not
Other tumors more than six spots, particularly exceeding 1.5 cm
• Ganglioneuromas. (0.6 in) in diameter.
• Pheochromocytomas. • Multiple lipomas.
• Carcinoid tumor of the intestine. • Sporadically occurring tumors of the nervous system:
meningiomas, schwannomas, neurofibromas and gliomas.
Other features or associations
• Macrocephaly.
• Cortical dysplasias.
• Heterotopias.
• Aqueduct stenosis.
• Learning disabilities.
• Intellectual decline.
• Speech impediments.
Neurofibromatosis 149

146 147

146 Cervical spine x-ray, oblique lateral view, showing enlargement of 147 MRI cervical spine, sagittal T1W image showing a well
the C2/3 intervertebral foramen (arrow) caused by a neurofibroma circumscribed, low-density, neurofibroma (arrow) measuring 2 cm
developing on the C3 spinal nerve root and extending through the (0.8 in) in diameter in the anterior-posterior plane and 2.5 cm (1 in) in
intervertebral foramen. the rostral-caudal plane which is compressing the upper cervical spinal
cord at C2.

148 149

149 MRI cervical spine, axial T1W image at the level of C2/3, after
gadolinium contrast injection, showing the same lesion as in 147, 148
as a well circumscribed, high intensity, contrast-enhancing neurofibroma
(arrows) extending from the upper cervical spinal cord at C2 (which is
148 MRI cervical spine, coronal T1W image, after gadolinium contrast displaced to the left [to the right in the image]) through the C2/3
injection, showing the same lesion in 147 as a well circumscribed, dumb intervertebral foramen.
bell-shaped, high intensity, contrast-enhancing neurofibroma (arrow)
measuring 4 cm (1.6 in) laterally in the coronal plane, extending from
the upper cervical spinal cord at C2 through the C2/3 intervertebral
foramen.The upper cervical spinal cord is displaced to the left (to the
right in the image) and compressed to a crescentic shape.
150 Developmental Diseases of the Nervous System

INVESTIGATIONS • Freckling in the axillary or inguinal regions.


NF-1 • Optic glioma (see p.372).
• Slit lamp examination of irides. • Two or more Lisch nodules (iris hamartomas).
• Neuropsychologic assessment of IQ. • A distinctive osseous lesion such as sphenoid dysplasia or thin-
• Visual evoked potentials. ning of long-bone cortex with or without pseudoarthrosis.
• Molecular DNA analysis. • A first-degree relative (parent, sibling, or child) with NF-1
• 24-hour urinary catecholamines if pheochromocytoma by the above criteria.
suspected.
• Imaging: Molecular DNA analysis
– Bone dysplasias and pseudarthroses of the long bones are
best shown on plain radiography occasionally combined NF-2
with CT. Clinical
– MRI is the investigation of choice for brain and optic nerve One of the following clinical criteria:
abnormalities though CT can be used (contrast enhanced). • Bilateral vestibulocochlear (VIIIth cranial) nerve tumors
– Optic gliomas (pilocystic astrocytomas pathologically) imaged by CT or gadolinium-enhanced MRI scan.
are seen in 15–40% of patients with NF-1. MRI (or CT) • A first-degree relative (parent sibling or child) with NF-2
shows thickening and increased signal (on T2W image) and either a unilateral VIIIth nerve tumor, or any two of the
of the optic nerves (usually bilaterally) or chiasm with following: neurofibroma, meningioma, glioma, schwan-
variable enhancement. The signal alterations may extend noma, or early- (juvenile) onset posterior subcapsular
posteriorly along the optic pathways (see p.372). lenticular opacity.
– Areas of increased signal on T2W image are often seen
in the basal ganglia, cerebral peduncles, and hemispheric Molecular DNA analysis
white matter. The exact cause of these is debated, but
growth or new enhancement after adolescence may Mild and severe phenotypic subtypes of NF-2 can be defined
indicate malignant gliomatous change and requires short using age at onset of symptoms (≥20 years vs. <20 years),
time interval follow-up scans. number of associated intracranial tumors (<2 tumors vs. ≥2
– De novo cerebellar, brain stem and cerebral astrocytomas tumors), and spinal tumors (absent vs. present).
are also seen in NF-1. It is important to distinguish between NF-1 and NF-2
– Plexiform neurofibromas favor the Vth nerve and orbit, because the genetic basis and natural history are distinct. It
are soft and elastic, and appear as enlarged nerves. These is also important to differentiate between NF-2 and the
may undergo sarcomatous change. sporadic unilateral acoustic neuroma, the latter of which is
– Astrocytomas of the spinal cord also occur (MRI not inherited, it tends to develop later in life, and raises far
demonstrates these best). fewer problems in management (see p.383).

NF-2 TREATMENT
• Audiometry. • Most people do not experience any functionally disabling
• Molecular DNA analysis. complications.
• Imaging: • Surgical resection or decompression of tumors is indicated
– MRI is the investigation of choice for brain and spinal for gliomas and meningiomas and when neurofibromas
lesions. compress cranial and peripheral nerves or spinal cord. The
– Enhanced T1W MRI is the best technique to demon- skin tumors should not be excised unless they are
strate bilateral acoustic schwannomas. Any coincidental cosmetically objectionable or increase in size, suggesting
meningiomas or schwannomas elsewhere in the brain malignant change. Plexiform neuromas of the face should
(Vth nerve is the next most common site) will be seen as be dealt with by an experienced plastic surgeon because
strongly enhancing (white) masses, and gliomas (e.g. they may involve cranial nerves superficially or affect the
ependymomas) may also be seen. underlying bone by eroding it (a pressure effect) or
– Other brain abnormalities include aqueduct stenosis, causing it to hypertrophy from increased blood supply.
polymicrogyria and pachygyria. • Treatment of acoustic neuroma (see p.383).
– In the spine, multiple nerve sheath tumors occur on the • Genetic counselling for patient and family, including
nerve roots usually in the cauda equina. These may be presymptomatic diagnosis of family members.
intra or extra dural.
– Other spinal abnormalities include dural ectasia (lateral PROGNOSIS
thoracic outpouchings of dura and scalloping of the NF-1
posterior surfaces of the vertebral bodies) and syringomyelia. A progressive disease but the prognosis varies with the grade of
severity, being more favorable in those with only a few lesions.
DIAGNOSIS
NF-1 NF-2
Clinical • Bilateral acoustic neuromas are inherited in an autosomal
Two or more of the following clinical criteria: dominant pattern with penetrance of over 95%, so that for
• Six or more café-au-lait macules whose greatest diameter is: any offspring of an affected parent the risk that these tumors
– >5 mm (>0.2 in) in prepubertal individuals, and will develop is about 50%. There are marked inter-family
– >15 mm (>0.6 in) in postpubertal individuals. differences in tumor susceptibility and disease severity.
• Two or more neurofibromas of any type or one plexi- • The mean age at death is about 35 years. Almost all deaths
form neurofibroma. are a result of a complication of neurofibromatosis.
Sturge–Weber Diseases (Encephalotrigeminal Vascular Syndrome) 151

STURGE–WEBER DISEASE CLINICAL FEATURES


(ENCEPHALOTRIGEMINAL • Cutaneous angiomatosis (vascular nevus) on one side of
VASCULAR SYNDROME) the face and scalp involving the first (ophthalmic)
division of the trigeminal nerve.
– Present at birth.
DEFINITION – Deep red (port wine nevus).
A rare, congenital and sporadic neuroectodermal – Varies in its extent (150): it may be limited to only the
degeneration (phakomatosis) characterized by: upper eyelid and forehead or may extensively involve the
• A cavernous or capillary cutaneous hemangioma (port entire head and even other parts of the body.
wine stain) on one side of the face, but sometimes – Involvement of the upper eyelid nearly always indicates
asymmetrically bilateral, in the distribution of the an associated brain lesion. Nevi lying entirely below the
ophthalmic division of the trigeminal nerve, together upper eyelid or high on the scalp are not usually
with, associated with a brain lesion, but can occasionally be
• A venous hemangioma of the meninges, usually associated with a vascular malformation of the meninges
ipsilateral to the skin lesion, and overlying the brainstem and cerebellum.
• Atrophy, gliosis and calcification of the underlying – Margins may be flat or raised.
cerebral cortex. – Surface may elevated or irregular due to soft or firm
papules, composed of vessels.
In 1879, W Allen Sturge described a child with sensori- • Epileptic seizures (partial and secondary generalized),
motor seizures contralateral to a facial ‘port wine mark’, and which usually begin in infancy or early childhood, some-
in 1922 and 1929, Parkes Weber radiographically demon- times followed by transient postictal (Todd’s) or permanent
strated the atrophy and calcification of the cerebral hemi- paralysis, are usually the first neurologic symptom.
sphere homolateral to the skin lesion. • Intellectual handicap in some cases.
• Hemiparesis, hemisensory defect, and homonymous
EPIDEMIOLOGY hemianopia contralateral to facial nevus in severe cases,
• Incidence: uncommon. Isolated facial port wine stains may arise abruptly or insidiously. The arm and leg may
without neurologic complications occur in about 1 in be small.
5000 births. • Angiomatosis of choroid of eye with abnormal distension
• Age: present from birth. and enlargement of the eyeball (buphthalmos) ipsilateral
• Gender: M=F. to the lesion, sometimes with later glaucoma, causing
blindness.
PATHOLOGY • Megalencephaly due to impaired cerebral venous return
• Capillary or cavernous hemangiomatous malformation has been reported.
within but not always limited to the ophthalmic division • Hypertrophy of the face in some cases: increased
of the trigeminal nerve; in some cases, additional divi- cutaneous vascularity may result in an overgrowth of
sions of the trigeminal nerve and other body parts are connective tissue and underlying bone.
involved (150).
• Venous hemangioma of the leptomeninges in the
parieto-occipital region. When the skin lesion involves
the ophthalmic division of the trigeminal nerve, the
venous hemangioma is usually present in the occipital 150
lobes, whereas a facial nevus is more often associated with
involvement of the parietal and frontal lobes.
• Atrophy of the subcortex of the brain on the side of the
facial lesion, beneath the large number of abnormal
blood vessels in the meninges, probably due to stagna-
tion of blood flow and consequent hypoxia. In some
cases, a band of calcification develops within the lesion
in the second and third layers of the cerebral cortex.

ETIOLOGY AND PATHOPHYSIOLOGY


• Unknown.
• Sporadic usually; familial occurrence is exceptional.
• Intrauterine developmental malformation.
• There seems to be a close correlation between the mal-
development of the embryonic vasculature of the eyelid
and forehead and that of the parieto-occipital parts of the
brain.

150 Facial photograph showing a cutaneous capillary hemangioma


(port wine stain) on one side of the face in the distribution of the
ophthalmic and maxillary divisions of the trigeminal nerve.
152 Developmental Diseases of the Nervous System

DIFFERENTIAL DIAGNOSIS TREATMENT


• Facial nevi, especially the flat midline ones: of no • Symptomatic control of complications such as epileptic
neurologic significance. seizures (with anti-epileptic medications), and glaucoma
• Elevated strawberry nevi: of no neurologic significance. if they arise.
• Epidermal nevus syndrome: an epidermal nevus or linear • Laser therapy may reduce the size and intensity of the
sebaceous nevus is associated with a variety of hemicranial skin lesion.
and neurologic abnormalities, such as thickening of the • The brain lesions are usually too extensive to be treated
skull bones, unilateral cerebral atrophy, porencephalic surgically, though resection may be considered for
cyst, leptomeningeal hemangioma and arteriovenous intractable epilepsy.
malformation ipsilateral to the nevus.
• Hemangioma of the trunk or upper or lower limb PROGNOSIS
associated with a spinal cord vascular malformation • Most patients survive for many years, often with epileptic
(Klippel–Trenaunay syndrome). seizures and some residual neuropsychologic deficit and
• Hemangioblastoma of the retina and cerebellum (von hemiparesis.
Hippel-Lindau disease, see p.392). • Meningeal nevi do not enlarge to form a mass lesion and
• Familial telangiectasia (Osler–Rendu–Weber disease, see they are rarely the source of subarachnoid or brain
p.153). hemorrhage.

INVESTIGATIONS
Skull x-ray
Uusually negative just after birth but when it is taken at the
end of the second year reveals characteristic parieto-occipital
cortical calcification, often as parallel lines, ‘railroad tracks’
or ‘tramline calcification’.

Cranial CT
Cranial CT scan at an earlier age shows a ‘tramline’ pattern
of intracranial, usually parieto-occipital, hyperdensity (due to
calcification) of the cerebral cortex on the side of the brain
beneath meningeal angiomatosis (151); atrophy of that side
of the brain and abnormal deep cerebral venous drainage.

MR scanning
MRI may show more abnormalities including:
• Accelerated myelination in the affected hemisphere in the
early stages of the condition. 151
• Abnormal low signal in the white matter on the side of
the port wine stain.
• Enhancement of the pia mater overlying the affected
cortex probably due to ischemia (which is the underlying
lesion).
• Abnormal draining deep cerebral veins.
• Abnormalities on both sides of the head though more
pronounced on the side of the cutaneous abnormality.

Cerebral angiography
The abnormal meningeal vessels, which are predominantly
veins, are not well seen (in contrast to arteriovenous
malformations) because the drainage of the hemisphere
ipsilateral to the port wine stain is all to the deep cerebral
veins, there being no cortical drainage (152, 153).
Prominent focal dilatation of the deep veins and apparent
strictures of the venous sinuses may be present.

151 CT brain scan (with contrast) showing an area of serpiginous


calcification in the right parietal lobe at the junction of cortex with
white matter (arrows), typical of Sturge–Weber syndrome.
Hereditary Hemorrhagic Telangiectasia (HHT) (Osler–Rendu–Weber Syndrome) 153

152 HEREDITARY HEMORRHAGIC


TELANGIECTASIA (HHT)
(OSLER–RENDU–WEBER SYNDROME)

DEFINITION
A group of autosomal dominant inherited disorders
characterized by multiple telangiectases involving the skin,
mucous membranes, viscera (particularly the gastrointestinal
[nose, pharynx, gut] and genito-urinary tracts) and occasion-
ally the nervous system. Rendu first recognized the combin-
ation of hereditary epistaxis and telangiectases in 1896 as a
specific distinct entity from hemophilia. Osler and Weber
produced prominent case reports in the following decade.

EPIDEMIOLOGY
• Prevalence: 1 in 2350 (France) to 1 in 40 000 (England).
• Age: children and adults.
• Gender: M=F.

PATHOLOGY
Telangiectases
• Range from small focal dilatations of postcapillary venules
to large, markedly dilated and convoluted venules which
extend through the entire dermis, have excessive layers of
smooth muscle without elastic fibers, and often connect
directly to dilated arterioles. Perivascular lymphocytes.
• Bright red or violaceous.
152 Lateral view of the venous phase of a right carotid angiogram • Blanch under pressure.
(same patient as in 151) demonstrating the grossly abnormal venous • Range in size from that of a pinhead to >3 mm (0.1 in).
drainage of the right hemisphere – the anterior half of the sagittal sinus • Tendency to bleed.
is absent (arrowheads indicate missing sinus) as is the deep venous • Skin, gastrointestinal tract.
drainage, consequently the blood drains via enlarged cortical and peri-
cavernous veins (arrows). Arteriovenous malformation
• Direct connections between arteries and veins, lacking
capillaries.
153 • Brain (5–10%).
• Spinal cord.
• Pulmonary (5–15%): predilection for lower lobes.
• Gastrointestinal tract.

Aneurysm
Intracranial.

ETIOLOGY AND PATHOPHYSIOLOGY


• Autosomal dominant inheritance.
• Gene locus: chromosome 9q33-q34 in some families,
and chromosome 12q in other families.
• The gene for HHT at chromosome 9q3 is endoglin.
Endoglin encodes an integral membrane glycoprotein
that is the most abundant protein on endothelial cells to
bind transforming growth factor β. Transforming growth
factor β modulates several processes of endothelial cells,
including migration, proliferation, and adhesion and the
composition and organization of the extracellular matrix.
• The fundamental lesion is probably a dysplasia of the
vessel wall.
• Bleeding is thought to be due to the mechanical fragility
of the vessel.
• Pulmonary arteriovenous malformations create a right-to-
153 Lateral view of the venous phase of a left carotid angiogram left shunt that may lead to cerebral hypoxia and polycythemia
(same patient) showing the grossly abnormal venous drainage of the and may allow the passage of paradoxical emboli
left hemisphere – all via the deep veins, there being no communication (thrombotic, septic) from the systemic venous circulation or
to the superficial system.The port wine stain was on the left. right heart to the brain causing stroke and cerebral abscess.
154 Developmental Diseases of the Nervous System

CLINICAL FEATURES Gastrointestinal arteriovenous malformations,


• Severe or recurrent episodes of: telangiectases, angiodysplasias
– Epistaxis. • Endoscopy.
– Upper or lower gastrointestinal hemorrhage. • Angiography.
– Hematuria. • CT for liver lesions.
– Sudden focal neurologic dysfunction: stroke syndrome
due to intracranial or intraspinal hemorrhage, or arterial DIAGNOSIS
obstruction by paradoxical embolism of thrombus from Clinical
the systemic venous circulation or right heart to the brain. Any two of the following:
• Iron deficiency anemia. • Recurrent epistaxis.
• Progressive focal neurologic dysfunction: enlargement of • Telangiectases elsewhere than in the nasal mucosa.
the intracranial or intraspinal vascular lesions, or • Evidence of autosomal dominant inheritance (the disease
occurrence of brain abscess due to embolism of septic is found in heterozygotes but penetrance may be
material to brain via pulmonary fistulae. incomplete).
• Epileptic seizures: arteriovenous malformation. • Visceral involvement.
• Family history.
• Dyspnea, fatigue, cyanosis, polycythemia, clubbing, chest Molecular DNA
bruit. Mutation of the endoglin gene on chromosome 9q3, in
• Telangiectases on lips, tongue, palate, nasal mucosa, face, some cases.
conjunctivae, trunk, nail beds, finger pulps (154, 155).
• Cerebral and spinal telangiectases are usually TREATMENT
asymptomatic. Nasal telangiectases
• Humidification.
INVESTIGATIONS • Packing.
• Full blood count. • Transfusion.
• Iron studies if microcytic, hypochromic anemia. • Estrogen therapy.
• Septal dermoplasty.
Brain arteriovenous malformation • Laser ablation.
• CT brain scan. • Cautery eradicates a bleeding lesion, but satellite ones
• MRI brain scan. tend to form.
• MR angiography.
Skin telangiectases
Pulmonary arteriovenous malformation • Topical agents: oxidized cellulose (Oxycel or Gelfoam)
• Chest x-ray (156–158). applied to the lesion.
• Arterial blood gases and finger oximetry. • Laser ablation.
• High resolution helical CT (159, 160).
• Pulmonary angiography.

154 155

154, 155 Telangiectases of the upper and lower lips (154) and
tongue (155) in a patient with hereditary hemorrhagic telangiectasia.
Hereditary Hemorrhagic Telangiectasia (HHT) (Osler–Rendu–Weber Syndrome) 155

156 157

156, 157 Chest x-ray. Posterior–anterior (156) and lateral (157)


views showing a pulmonary arteriovenous malformation in the left
anterior mid zone (arrow, 156) and the anterior aspect of the left
lower lobe (arrow, 157).

158 159

160

158 Chest x-ray posterior–anterior showing a pulmonary


arteriovenous fistula (arrows). (Reproduced with permission from
Hankey GJ,Warlow CP [1994] Transient Ischaemic Attacks of the Brain 159, 160 Chest CT scan, images in the axial plane, at the level of the
and Eye. WB Saunders, London.) mid thorax (159) and the right hemidiaphragm (160) showing a
pulmonary arteriovenous malformation in the left anterior mid zone
(arrow, 159) and the anterior aspect of the left lower lobe (arrow, 160).
156 Developmental Diseases of the Nervous System

Pulmonary arteriovenous malformation Gastrointestinal arteriovenous malformation


• Embolotherapy. • Transfusion.
• Surgical resection. • Photocoagulation.
• Ligation of arterial supply. • Estrogen-progesterone therapy.
• Antibacterial prophylaxis at the time of a dental or
surgical procedure. PROGNOSIS
The telangiectases first appear during childhood, enlarge
Central nervous system arteriovenous malformation during adolescence, and may assume spidery forms,
(see p.245) resembling the cutaneous telangiectases seen with liver
• Neurovascular surgery. cirrhosis, in late adult life.
• Embolotherapy.
• Stereotactic radiosurgery.

Santamarta D, Kusak ME, De Campos JM, Sierra Yasunari T, Shiraki K, Hattori H, Miki T (2000)
FURTHER READING JM (1999) Increased cerebrospinal fluid flow Frequency of choroidal abnormalities in neu-
through the foramen of Magendie after de- rofibromatosis type 1. Lancet, 356: 988–992.
compression for Chiari I malformation. J.
Neurol. Neurosurg. Psychiatry, 66: 799. HEREDITARY HEMORRHAGIC
ARNOLD–CHIARI MALFORMATION TELANGIECTASIA
Ikusaka M, Iwata M, Sasaki S, Uchiyama S (1996) TUBEROUS SCLEROSIS Guttmacher AE, Marchuk DA, White RI Jr
Progressive dysphagia due to adult Chiari I Crino PB, Henske EP (1999) New developments (1995) Hereditary haemorrhagic telangiecta-
malformation mimicking amyotrophic lateral in the neurobiology of the tuberous sclerosis sia. N. Engl. J. Med., 333: 918–924.
sclerosis. J. Neurol. Neurosurg. Psychiatry, complex. Neurology, 53: 1384–1390. Maher CO, Piepgras DG, Brown RD Jr., et al.
357–358. O’Callaghan FJK (1999) Tuberous sclerosis. BMJ, (2001) Cerebrovascular manifestations in 321
Koehler PJ (1991) Chiari’s description of cerebel- 318: 1019–1020. cases of hereditary hemorrhagic telangiectasia.
lar ectopy (1891). J. Neurosurg., 75: Stroke, 32: 877–882.
823–826. NEUROFIBROMATOSIS
Sahuquillo J, Rubio E, Poca MA, et al. (1994) Créange A, Zeller J, Rostaing-Rigattieri S, et al.
Posterior fossa reconstruction: a surgical tech- (1999) Neurological complications of neu-
nique for the treatment of Chiari I malforma- rofibromatosis type 1 in adulthood. Brain,
tion and Chiari I/syringomyelia complex: pre- 122: 473–481.
liminary results and magnetic resonance imag- Huson SM (1999) What level of care for the neu-
ing quantitative assessment of hindbrain mi- rofibromatoses? Lancet, 353: 1114–1116.
gration. Neurosurgery, 35: 874–885.
Chapter Eight 157

Inherited Metabolic
Diseases of the
Nervous System of
Adult Onset
WILSON’S DISEASE (WD) Microscopic
(HEPATOLENTICULAR • Marked hyperplasia of protoplasmic astrocytes
DEGENERATION) (Alzheimer type II cells) in the cerebral cortex, basal
ganglia, brainstem nuclei, and cerebellum.
‘ • Nerve cell loss and some degree of degeneration of
When trouble is sensed well in advance, it can be easily remedied; if myelinated fibers in lenticular nuclei, substantia nigra,
you wait for it to show itself, any medicine will be too late because and dentate nuclei are usually apparent.
the disease will have become incurable. As the doctors say of a
wasting disease, to start with it is easy to cure but difficult to ETIOLOGY AND PATHOPHYSIOLOGY
diagnose; after a time, unless it has been diagnosed and treated at Genetic
the outset, it becomes easy to diagnose but difficult to cure. Autosomal recessive inheritance. Therefore, it is not sex-
Machiavelli 1513 linked (it occurs equally in men and women), and in order
to inherit it, both parents must carry a Wilson’s disease
DEFINITION gene, which each passes to the affected child. If both parents
A rare autosomal recessive disease caused by a gene defect have the WD gene, then the chance of the child getting WD
on chromosome 13 that results in a disorder of (i.e. receiving an abnormal gene from each parent) is 25%.
hepatobiliary copper excretion that leads to the deposition All children of a person with WD will receive a WD gene
of free copper in nearly all the body's tissues, particularly the from the affected person. 50% of children of a carrier will
liver, brain, kidneys and cornea. receive a WD gene, since the carrier has one normal and one
abnormal gene.
EPIDEMIOLOGY The gene defect, designated ATP7B, spans an 80 kb
• Prevalence: 1:30 000 (30 per million) are affected; 1:100 region of chromosome 13q14.3 and encodes a copper
individuals in the general population carry the Wilson’s transporting ATPase.
disease gene (i.e. have one normal and one abnormal More than 50 unique mutations in ATP7B, a relatively
gene). large gene, have been found to cause Wilson’s disease to date.
• Age: onset: 8 to >50 years. Most are spontaneous mutations in the gene and are single
– Neurologic and psychiatric presentations somewhat later base transversions or deletions. A substantial number of other
than hepatic. cases are simply transmitted from generation to generation.
• Gender: M=F. Although only one gene is involved in Wilson’s disease,
many different mutations cause various defects in its
PATHOLOGY product, a copper-transporting ATPase. The existence of
Varies with rate of progress of the disease. different alleles and combinations of two alleles (intralocus
genetic heterogeneity) explains much of the variability in
Macroscopic Wilson’s disease among different families. For example,
Lenticular (putaminal and pallidal) nuclei: mutations that result in complete absence of the gene
• Frank cavitation (rapidly and advancing fatal form). product are associated with development of liver disease at
• Shrinkage and light-brown discoloration (more chronic a particularly early age.
form). Because most patients are compound heterozygotes,
containing different mutations of the WD gene on each
allele, genetic screening for this disease is complicated.
However, in a particular family, if the precise mutation is
identified, a genetic diagnosis is possible. Individuals
heterozygous for the WD gene (i.e. only one abnormal
158 Inherited Metabolic Diseases of the Nervous System of Adult Onset

gene; carriers) may manifest mild abnormalities in copper • Parkinsonian, with rigidity and bradykinesia of the
metabolism but do not develop the disease; one normal tongue, lips, pharynx, larynx and jaw; dysarthria,
allele is adequate for disease prevention. dysphagia, hoarseness and drooling.
• Choreic movements of the limbs.
Hepatic copper metabolism and pathogenesis of WD • Dystonic postures of the limbs, with hypertonia and
Disruption of one of the genes controlling copper rigidity; the latter two subgroups being more common
metabolism, located on chromosome 13, leads to a in children.
reduction in the rate of biliary excretion of copper, and an
accumulation of excess copper in the body (e.g. in the liver, Other features
and extrahepatic sites such as Descemet’s membrane in the • Kayser–Fleischer (K–F) rings (161, 162):
cornea and the basal ganglia of the brain). The role of – A golden brownish coloration on the outer margin
ceruloplasmin is unclear. (limbus) of the cornea present in virtually all patients
with neurologic and 80% with hepatic WD.
CLINICAL FEATURES – Almost diagnostic of WD.
• Variable: asymptomatic or a range of hepatic, neurologic – Gonioscopic examination provides earliest detection, and
and psychiatric symptoms. later may be seen through an auroscope or ophthalmo-
• No two patients are ever quite the same, even in a sibship. scope or with the unaided eye.
– Slit lamp examination is necessary to demonstrate
Neurologic and psychiatric disease definitely copper granules in Descemet’s membrane.
Insidious onset and progressive course. • Loss of motor control: bizarre spontaneous movements.
• Change in personality and behavior (argumentative,
The four main clinical categories excessively emotional).
• Pseudosclerotic, with tremor of the limbs (postural and • Depression.
intention) that closely resembles that seen in multiple • Cognitive decline (dementia).
sclerosis and which can be severe enough to be described • Seizures (rare).
as ‘wing-beating’, titubation of the head, incoordination, • Occasional autonomic dysfunction, causing hyperhidrosis
limb ataxia, and dysarthria (seen most commonly in and exophthalmos.
adults).
Ultimately, the ‘classic syndrome’ develops: dysarthria,
dysphagia and drooling, rigidity and slowness of movements
of the limbs, fixed flexed postures, fixity of facial muscles
161 with mouth constantly agape, giving an appearance of
grinning or a ‘vacuous smile’; slow saccadic eye movements,
a virtual anarthria (bulbar extrapyramidal syndrome); and
tremor in repose which increases when the limbs are
outstretched (coarse, ‘wing-beating’ tremor).
In most neurologic cases, the liver lesion plays a relatively
minor role.

161, 162 Kayser–Fleischer ring. A 1–3 mm thick brown ring (but


may be green, yellow, blue, ruby red, or a mixture of these colors)
of sulfur–copper complexes in Descemet’s membrane at the
corneal margin.

163 Sunflower cataract.

162 163
Wilson’s Disease (WD) (Hepatolenticular Degeneration) 159

Liver disease • Hereditary ceruloplasmin deficiency: mutations of the


May take many forms: ceruloplasmin gene on chromosome 3q; autosomal
• Asymptomatic. recessive phenotype; dementia, involuntary movements
• Chronic active hepatitis. and diabetes; iron deposition in the brain and liver
• Cirrhosis. instead of copper.
• Fulminant hepatitis with massive necrosis; there does not • Pseudo-K–F rings have been described in:
appear to be a significant increased risk of hepatocellular – Primary biliary cirrhosis.
carcinoma. – Chronic active hepatitis.
• Pallor of mucous membranes (hemolytic anemia). – Progressive intrahepatic cholestasis of childhood.
• Jaundice. – Intraocular copper foreign body.
• Sunflower cataracts (163). – Multiple myeloma.
• Azure lunulae of the nailbeds. – Infestation with Schistosoma japonicum and African
• Hepatocellular carcinoma is rare, in contrast to hemo- trypanosomiasis.
chromatosis. – Copper therapy.

DIFFERENTIAL DIAGNOSIS INVESTIGATIONS


More than half of patients are initially suspected to have a Patient
disease other than WD. • Slit lamp examination for K–F rings: may be absent early
• Parkinson’s disease: WD mainly affects the bulbar in disease, invariably present in patients with neurologic
musculature and spreads caudally. or psychiatric symptoms.
• Psychiatric disease: anxiety; depression; schizophrenia. • Full blood count: hemolytic anemia, thrombocytopenia.
• Multiple sclerosis. • Liver function tests: abnormal.
• Encephalitis. • Serum ceruloplasmin: <200 mg/l (<20 mg/dl) (in 95%
• Myasthenia gravis. of patients); sensitivity 95% (normal levels
• Thyrotoxicosis. [200–300 mg/l; 20–30 mg/dl] in 5% of patients);
• Hypothyroidism. specificity high but level also decreased in 20% of
heterozygote carriers.
• Serum copper: low, <15.7 μmol/l (<100 μg/dl);
normally decreased in proportion to serum cerulo-
plasmin; markedly elevated in fulminant Wilsonian
hepatitis.
• DNA analysis: gene defect on chromosome 13.
• 24-hour urinary copper excretion: >1.6 μmol (>100 μg)
in 24 hours (normal <0.8 μmol [<50 μg]); completeness
164 of collection is important.
• 24-hour urinary copper excretion post-penicillamine: high,
>25.1 μmol (>1600 μg); may also be increased in
cholestatic disorders.
• Cranial CT scan: normal or may show atrophy of the
caudate nuclei and brainstem, with hypodense areas in
the basal ganglia, dentate nuclei and thalami.
• MRI brain: normal or increased signal intensity on T2W
image in the above areas (164), atrophy of the caudate
nuclei and scattered areas of increased signal in the brain-
stem, midbrain, red nucleus, periaqueductal gray matter
and external capsules. Generalized atrophy may occur.
• Liver biopsy: copper concentration: high, >3.9 μmol/g
(>250 μg/g) dry weight (normal <0.8 μmol/g
[<50 μg/g]); can be intermediate in heterozygotes,
cholestatic disorders; can be elevated in primary biliary
cirrhosis, childhood cirrhosis.
– Histology: steatosis, glycogen nuclei, fibrosis, chronic
active hepatitis, cirrhosis.
– Rhodanine histochemistry: positive, present in some but
not all nodules; may be negative early in disease
– Electron microscopy: stage specific mitochondrial and
lysosomal alterations.
• Failure to incorporate 64Cu into ceruloplasmin.
• Aminoaciduria: present and persistent in most but not all
cases.

164 T2W MR brain scan, axial section through the basal ganglia Relatives
showing high signal in the lateral parts of the basal ganglia (arrows) Screening by clinical, biochemical and genetic assessment.
due to Wilson’s disease.
160 Inherited Metabolic Diseases of the Nervous System of Adult Onset

DIAGNOSIS Grandchildren of WD patients have a 1/400 chance of


Clinical and laboratory evidence of: having the disease: all children of the patient are carriers;
• K–F rings. from the patient’s child, grandchildren have a 50% chance
• Low serum ceruloplasmin and copper. of inheriting a gene (1/2); from the other parent, a normal
• High urine copper excretion over 24 hours. person, they have a 1/200 chance of inheriting the gene
• High hepatic copper content on liver biopsy. (1/2 × 100).

The abundance of specific mutations of the gene on Presymptomatic screening


chromosome 13 and their location at multiple sites across All siblings, aunts, uncles, children, nieces, nephews and
the genome have limited molecular genetic diagnosis to cousins should be screened for WD, because those with
kindreds of known patients. The DNA-based diagnostic test even mild or non-apparent WD will ultimately become
can be done only in siblings of an index patient whose seriously ill if not treated.
diagnosis was made according to phenotypic criteria and The most appropriate screening test is 24-hour urine
only if DNA from both patients is available. measurement of copper, which should be performed in
asymptomatic individuals at about age 5 years and again at
Presymptomatic risk age 15 years. Blood ceruloplasmin is helpful but is normal
Siblings of WD patients have a 25% chance of having the in 5–10% of WD patients. Screening can begin for low
disease. Since both of a sibling’s parents are carriers, 1/4 ceruloplasmin and abnormal liver function tests at age
have the disease, 2/4 will be carriers, and 1/4 will have no 2 years. Ophthalmologic assessment for K–F rings can be
WD gene. diagnostic but they do not have to be present to have WD.
Children of WD patients have a 1/200 chance of having
the disease: from the patient, their children will definitely TREATMENT (see Table 22)
inherit the abnormal gene; for the patient’s spouse, a normal WD is a manifestation of copper toxicosis and, in most
person, the chance of carrying the gene is 1/100 and the cases, symptoms can be prevented or reversed by achieving
chance they will pass it on is 1/2. and maintaining a negative copper balance.

Table 22 Recommended treatment of, and pharmacologic agents for,Wilson’s disease


Recommended treatment
Clinical status Recommended treatment
Asymptomatic Maintenance therapy
Penicillamine
Trientine
Zn salts
Neurologic/psychiatric symptoms Penicillamine
Trientine
BAL (can use in conjunction with oral therapy)
Tetrathiomolybdate (experimental)
Chronic active hepatitis, or Penicillamine
Hepatic insufficiency Trientine
Fulminant hepatic failure Orthotopic liver transplantation
Chelation therapy and plasmapheresis pendingtransplantation
Pharmacologic agents
Agent Daily adult dose Mode of action
Penicillamine* 1–2 g orally in 2–4 divided doses Chelator
Possible inducer of metallothionein (MT)
Trientine hydrochloride 1.2–2.4 g orally in 2–4 divided doses Chelator
Zinc salts 600 mg of metallic zinc orally Prevents copper absorption by inducing intestinal MT;
in three divided doses may also induce hepatic MT
British antilewisite (BAL) (dimercaprol) 3 ml of 10% BAL in peanut oil i.m. Chelator
Tetrathiomolybdate** Up to 2 mg/kg orally in divided doses Chelator
* Administered with supplemental pyridoxine 25 mg orally daily **Experimental
Wilson’s Disease (WD) (Hepatolenticular Degeneration) 161

Presymptomatic • Monitor clinical response, full blood count, proteinuria,


• Reduction of dietary copper to less than 1 mg/day: and urine copper excretion.
avoid copper-rich foods (liver, mushrooms, cocoa, • A further 8% or so show no response to treatment.
chocolate, nuts and shellfish). • The appropriate drug needs to be continued for the
• Zinc sulfate, 200 mg orally, three times daily, blocks patient’s lifetime.
copper absorption from the gut. Adverse effects:
epigastric pain, nausea, vomiting, and sideroblastic In about 20% of patients, chelation treatment results in
anemia. an initial worsening of neurologic symptoms before
improvement begins, and a few (1–2%) may deteriorate
Symptomatic rapidly once treatment is started. This is thought to reflect
Medical the mobilization of copper from tissues, e.g. liver, and its
Limit dietary copper to less than 1 mg/day (see above). redistribution in the brain, and for the redistribution of
copper within neurons themselves.
Chelating agents: Chelating agents have been used during pregnancy in
Penicillamine (a copper chelator) can be taken orally. Begin WD patients without teratogenic effects.
with 250 mg orally twice a day before food and increase
slowly over a few weeks to 1.0–2.0 g daily in 2–4 divided Surgical
doses. Add pyridoxine 25 mg daily, particularly during Liver transplantation: cures WD by removing the site of the
pregnancy, a growth spurt, malnutrition or prolonged metabolic lesion; chelation therapy need not be continued.
intercurrent illness. Pyridoxine deficiency, due to d- If there is irreversible liver damage due to acute and chronic
penicillamine, can be detected early by the presence of liver failure its place in the management of advanced
abnormal tryptophan metabolites in the urine. About 10% neurologic disease is less clear.
develop penicillamine sensitivity (rash, arthralgia, fever,
leukopenia): temporarily reduce the dose or try a course of CLINICAL COURSE AND PROGNOSIS
cortisone. Reinstitute in low dose (250 mg daily) with small, Course and prognosis vary greatly. Untreated, the course is
widely spaced increases. If the penicillamine sensitivity invariably progressive and fatal. Most patients reach a
persists or if severe immune-mediated reactions (lupus-like terminal stage with severe dystonia and contractures, though
or nephrotic syndromes) occur, the drug should be a few may become akinetic. The disease may last from a few
discontinued and zinc acetate (50 mg elemental zinc 5 times months (it tends to run a more acute course in younger
daily) or trientene substituted. patients) to many years with occasional periods of relative
Triethylene tetramine dihydrochloride (trientine, TETA) remission. With treatment, most patients improve or recover
is as effective as penicillamine and has fewer adverse effects, completely, but some do not. Some get worse before they
but is expensive. It is taken orally, 1.2–2.4 g daily in 2–4 get better (about 25%), some get worse and do not get
divided doses before food. It is poorly absorbed from the better, remaining permanently disabled, and some die,
gut. TETA reduces intestinal copper absorption and fortunately very few. Pseudosclerotic patients have a better
increases urinary copper excretion. Add zinc acetate 50–150 prognosis than the dystonic.
mg as a single dose at night (separate from the ingestion of
trientene tablets) to prevent zinc deficiency. Toxic reactions PREVENTION
are rare: sideroblastic anemia, reactivation of penicillamine- Younger siblings can now be identified by means of
induced lupus, colitis and duodenitis. molecular DNA techniques as normal, heterozygous or
presymptomatic carriers of the WD gene. For the latter and
Metal to metal antagonists: relatives detected by screening (see above), prophylactic
Zinc sulfate or acetate can be used if penicillamine or treatment can be started. Experience to date shows that
trientene toxicity occurs. Doses are taken orally, 200 mg such patients remain in good health so long as they take
three times daily. It blocks copper absorption from the gut. their medication.
Toxic reactions are rare: nausea, epigastric pain, vomiting,
sideroblastic anemia. It is useful for maintenance therapy,
treatment of the pregnant patient, and the initial as well as
maintenance treatment of the presymptomatic patient. It
does not seem to be optimal for initial treatment because it
is somewhat slow acting and may take several months to
control copper toxicity.
Ammonium tetrathiomolybdate blocks intestinal
absorption of copper and binds copper present in tissues.
Early studies suggested it may depress bone marrow and
cause serious epiphyseal deformities in growing animals (and
presumably children). A safe and effective initial treatment
in a recent open study of 33 patients.

Metal-binding agent:
British antilewisite (BAL) (dimercaprol): if advanced
neurologic lesions which have failed to respond to other
treatments. Crosses the blood–brain barrier better than
chelating agents.
162 Inherited Metabolic Diseases of the Nervous System of Adult Onset

HALLERVORDEN–SPATZ DISEASE MITOCHONDRIAL (OXIDATIVE


PHOSPHORYLATION) DISEASES

DEFINITION
An autosomal recessive inherited condition characterized by DEFINITION
iron deposition in the basal ganglia and progressive pyramidal A clinically, biochemically and genetically diverse group of
and extrapyramidal signs. Also known as pigmentary degener- disorders that result from structural, biochemical, or genetic
ation of the globus pallidus, substantia nigra and red nucleus. derangement of mitochondria.

EPIDEMIOLOGY EPIDEMIOLOGY
• Incidence: rare. • Incidence: not uncommon; being recognized with
• Age: late childhood or early adolescence. increasing frequency.
• Gender: M=F. • Prevalence:at least 1 in 50 000.
• Age: any age.
ETIOLOGY AND PATHOPHYSIOLOGY • Gender: either sex.
• Autosomal recessive inheritance.
• No known biochemical defect. STRUCTURE AND FUNCTION OF MITOCHONDRIA
AND THEIR DNA
PATHOLOGY Every mammalian cell contains multiple (about 800)
Deposits of iron in the basal ganglia: mitochondria, the main function of which is to generate
• Macroscopic: intense brown pigmentation of the globus energy for cellular processes by producing ATP through
pallidus, substantia nigra (particularly the anteromedial oxidative phosphorylation. For this task, the extensively
parts) and red nucleus. folded inner membrane of the mitochondrion is equipped
• Microscopic: granules and larger amorphous deposits of iron with the respiratory chain, a redox system whose com-
mixed with calcium in the walls of small blood vessels or free ponents (complexes I–V) are encoded by two separate
in the tissue; loss of neurons and medullated fibers in the genetic systems; 13 subunits are encoded on mitochondrial
most affected regions; and swollen axon fragments. DNA and more than 67 on nuclear DNA. Each mito-
chondrion contains 2–10 molecules of its own, extra-
CLINICAL FEATURES chromosomal mitochondrial DNA.
• Spastic quadriparesis, with hyperreflexia and Babinski signs.
• Rigidity. Mitochondrial DNA (mtDNA)
• Dysphagia and dysarthria. • Distinct from DNA in the cell nucleus, it is 16 569 base-
• Dystonia. pairs long, a closed circular and double-stranded
• Choreoathetosis. molecule that encodes 13 protein subunits of four
• Cognitive decline. biochemical complexes and the 24 structural RNAs and
• Ataxia and myoclonus in some patients. 22 transfer RNAs that are required for the
intramitochondrial translation of the protein-coding unit.
DIFFERENTIAL DIAGNOSIS • Almost exclusively inherited maternally because the
• Wilson’s disease. spermatocyte contributes virtually no mitochondria to
• Other dystonias. the zygote at fertilization.
• Mitochondrial disease: Leigh disease. • Does not recombine; mutations accumulate sequentially
• Neuroaxonal dystrophy. through maternal lineages.
• Has a very high mutation rate (10 times as frequent as
INVESTIGATIONS nuclear DNA) and has no introns, so that a random
• Serum iron studies: normal. mutation will usually strike a coding DNA sequence.
• CT brain scan: normal or symmetric non-enhancing areas • Mutations may occur in each type of mitochondrial gene.
of reduced density in the lenticular nuclei. • Different populations (normal and mutant mtDNA) can
• MRI brain: reported to show low signal in the basal coexist in the same cell (heteroplasmy) because each cell
ganglia (lentiform nucleus) on T2W image possibly due contains multiple mitochondria, and because each
to the iron deposition, and increased signal in the mitochondrion contains 2–10 DNA molecules. This
periventricular white matter. However, this is rather non- condition, allows an otherwise lethal mutation to persist.
specific as these features also occur in Wilson’s disease Homoplasmy is the presence of either completely normal
and a number of other more common conditions. or completely mutant mitochondrial DNA.
• When cells replicate, the proportions of mutant and
DIAGNOSIS normal molecules can shift as mitochondrial DNA is
Clinical and radiologic. partitioned into daughter cells because mitochondria
segregate randomly to daughter cells at each mitotic
TREATMENT division (mitotic segregation). The resulting cell
• Supportive. generation might inherit different amounts of coexisting
• L-dopa (levodopa) may provide temporary symptomatic mtDNA populations (mosaicism).
relief.
• Iron chelating agents are not helpful.

PROGNOSIS
Slowly progressive over 10–20 years.
Mitochondrial (Oxidative Phosphorylation) Diseases 163

• The phenotypic expression of a pathogenic mutation • Histochemistry: cytochrome c oxidase (complex IV)
exhibits a threshold effect, depending on the relative negative fibers.
proportion of the mutant to the complementing wild • Electron microscopy: structurally abnormal mitochondria
type genome. The proportion of mutant mitochondrial containing paracrystalline inclusions (condensations of
DNA required for the occurrence of a deleterious mitochondrial creatine kinase between mitochondrial
phenotype (clinical disease), known as the threshold inner and outer membranes).
effect, varies among people, organ systems and tissues, • Biochemical studies: defective oxidative phosphorylation.
and depends on the delicate balance between oxidative • mtDNA analysis usually positive for one of the
demand and supply. recognized mutations.

ETIOLOGY AND PATHOPHYSIOLOGY CLINICAL FEATURES


• Gross structural rearrangements (single deletions, Mitochondrial DNA mutations cause an extensive array of
multiple deletions, or duplications) or point mutations clinical disorders because virtually all tissues in the body
in mitochondrial DNA. depend to some extent on oxidative metabolism and are
• Mitochondrial mutations can be inherited maternally and prone to be affected by mitochondrial DNA mutations.
by mendelian patterns. A minority of patients fit neatly into well defined syndromes
• An autosomal dominant locus on chromosome 10 can such as MELAS, MERRF, CPEO, or the KSS (see below).
predispose to deletions of mitochondrial DNA.
• Mutations with the potential to cause a lethal impairment Neurologic
of oxidative phosphorylation (gross structural defects or N.B. Common and typical features are shown in italics.
point mutations in critical regions) are viable only if they
are heteroplasmic. Brain:
• The majority of the milder, missense mutations in • Generalized (and partial) seizures.
protein-coding genes are homoplasmic. • Myoclonus: spontaneous and stimulus-sensitive.
• Ataxia.
It can be difficult to link a mutation to a clinical disease • Dementia.
for several reasons: • Stroke-like episodes in a young person.
• Mitochondrial DNA is highly polymorphic. • Dystonia.
• There is a dissociation between the genotype and the • Psychiatric disorder (particularly depression).
phenotype.
• Different mutations can be associated with the same Cranial nerves:
phenotype. • Optic neuropathy: optic atrophy, peripheral pigmentary
• The same mutation can be associated with different retinopathy, cataracts.
phenotypes. • Sensorineural deafness.
• Epigenetic factors (e.g. alcohol and tobacco use) can • Aminoglycoside deafness.
affect clinical manifestations.
Spinal cord: myelopathy.
Even so:
• Mitochondrial encephalomyopathy phenotype: tRNA Peripheral nerves: peripheral neuropathy; usually
mutations predominate. asymptomatic.
• Leber’s hereditary optic neuropathy phenotype: protein-
coding gene mutations predominate. Muscle:
• Myopathy: mild and proximal; gross wasting and
The mechanism of stroke-like episodes in MELAS weakness are rare.
syndrome (see below) is probably a combination of defects • Hypotonia.
in neuronal metabolism as well as in cerebral vasculature. • Chronic progressive external ophthalmoplegia: evolves
slowly, hence diplopia is uncommon.
PATHOLOGY • Fatiguability and exercise intolerance.
MELAS syndrome (see p.164) • Recurrent myoglobinuria.
Brain
• Light microscopy: diffuse fibrillary gliosis with abundant Systemic
gemistocytes, focal evidence of ischemic neuronal injury Stature: short.
and edema.
• Electron microscopy: bizarre enlarged mitochondria and Eyes:
changes consistent with cellular edema. Strongly reactive • Cataracts.
succinate dehydrogenase staining in cerebral blood • Pigmentary retinopathy.
vessels.
• Mitochondrial DNA analysis: point mutation at nucleotide Ears: hearing loss.
position 3243 of the mitochondrial tRNA Leu(UUR)
gene in most (82%) of brain mitochondria in one study. Heart:
• Cardiac conduction defects and heart block.
Muscle • Cardiomyopathy.
• Modified Gomori trichrome stain: proliferation of
subsarcolemmal mitochondria (ragged-red fibers).
164 Inherited Metabolic Diseases of the Nervous System of Adult Onset

Endocrine: • Ptosis (165, 166).


• Diabetes mellitus. • Progressive external ophthalmoplegia.
• Hypoparathyroidism. • Atypical pigmentary retinopathy.
• Growth/multiple hormone deficiency. • Deafness (166).
• Cerebellar ataxia.
Gastrointestinal: • Heart block.
• Episodic nausea and vomiting. • Diabetes mellitus.
• Hepatopathy. • CSF protein elevated above 1 g/l (100 mg/dl).
• Intestinal pseudo-obstruction.
• Exocrine pancreatic dysfunction. Myoclonus, cortical blindness and hemiparesis are not
• Villous atrophy. features of KSS.

Renal: Etiology:
• Glomerulopathy. • Spontaneous (i.e. usually not maternally inherited),
• Proximal nephron dysfunction, with aminoaciduria, heteroplasmic deletions of the mitochondrial genome at
phosphaturia and glycosuria. high levels are found in 80% of cases.
• The most frequent deletion (30–50% of cases) is a 4.9 kb
Hematologic: sideroblastic anemia/pancytopenia. deletion (‘common deletion’) that spares the origins of
replication.
Other: lactic acidosis.
Prognosis:
Family history for evidence of mendelian or maternal • Many patients die in the third and fourth decade of life.
inheritance.
B. Transfer RNA mutation
CLASSIC PHENOTYPES Mitochondrial encephalomyopathy, lactic acidosis, and stroke-
I. PRIMARY MITOCHONDRIAL DNA MUTATIONS like episodes (MELAS) syndrome
A. Mitochondrial DNA rearrangements Clinical and biochemical features:
Chronic progressive external ophthalmoplegia (CPEO) • Onset at any age.
• Clinical features: • Stroke-like events causing subacute focal brain dysfunction.
– Ptosis (165, 166). • Seizures and/or migraine headache.
– Ophthalmoplegia. • Lactic acidosis.
– Limb myopathy. • Other clinical and laboratory features: retinitis
– Variable constellation of other clinical and laboratory pigmentosa, cerebellar ataxia, myopathy, cardiomyopathy,
features. diabetes, proximal renal tubule defects, and lactic
• Etiology: most commonly sporadic, large, single acidemia and hyperalaninemia.
deletions in mitochondrial DNA.
Etiology:
Kearns–Sayre syndrome (KSS) • Inherited maternally.
A form of CPEO. • A point mutation at nucleotide position 3243 in the tRNA
Clinical features: Leu(UUR) gene in 80% of cases but this mutation is not
• Onset before age 20 years. specific for MELAS syndrome; it has multiple phenotypic
effects.

165 Myoclonic epilepsy with ragged-red fibers (MERRF) syndrome


Clinical and pathologic features:
• Myoclonus.
• Epileptic seizures: myoclonic epilepsy, generalized
seizures, or focal seizures.
• Cerebellar ataxia.
• Mitochondrial myopathy with ragged-red fibers.
• Variable constellation of other clinical and laboratory
features: dementia, optic neuropathy, deafness, corticospinal
tract degeneration, peripheral neuropathy, myopathy,
proximal renal tubule dysfunction, cardiomyopathy and
166 lactic acidemia plus hyperalaninemia.
• Maternal relatives may be asymptomatic or have partial
clinical syndromes, including lipomas in a characteristic
‘horse collar’ distribution and cardiovascular disease.

Etiology: mutations at nucleotide positions 8344 and 8356


in the tRNA Lys gene.

165, 166 Ptosis, progressive external ophthalmoplegia and


sensorineural deafness in a patient with Kearns–Sayre syndrome.
Mitochondrial (Oxidative Phosphorylation) Diseases 165

C. Missense mutations • CSF protein: elevated.


Neuropathy, ataxia, and retinitis pigmentosa and • CSF organic and amino acids: elevated lactate.
maternally inherited Leigh disease
Clinical features: Neurophysiology
• Proximal muscle weakness. • Nerve conduction studies: may demonstrate a peripheral
• Sensory neuropathy. (predominantly axonal) neuropathy.
• Ataxia. • EMG: normal or mildly myopathic.
• Retinal pigmentary degeneration.
• Developmental delay. Imaging
• Dementia. • Brain CT or MRI: basal ganglia calcification or focal
• Epileptic seizures. signal abnormalities.
– MELAS syndrome causes cortical infarct-like lesions (but
Etiology: in an atypical distribution for normal arteriovascular
• Inheritance: maternal. territories) usually affecting the posterior parts of the
• Mutation: heteroplasmic missense point mutations at cerebrum. These lesions may disappear in time leaving only
nucleotide position 8993 in the ATPase 6 gene. mild sulcal dilatation. Leigh’s disease causes periaqueductal
gray and putaminal abnormal signal intensity, swollen
Leber’s hereditary optic neuropathy (LHON) (see p.491) caudates on MRI, and low density in the putamina on CT.
• Phosphorus-31 nuclear magnetic resonance spectros-
II. NUCLEAR DNA MUTATIONS copy: lactate may be detected in the abnormal areas.
Nuclear DNA mutations causing multiple mtDNA
rearrangements Histology
Autosomal dominant chronic progressive external • Open skeletal muscle biopsy is diagnostic, although occa-
ophthalmoplegia: (chromosome 10q23.3-24.3 locus) sional patients with mitochondrial myopathy due to
mtDNA mutations and those with LHON may have
Autosomal recessive chronic progressive external normal biopsies.
ophthalmoplegia • Histochemical analysis typically reveals ragged-red fibers
on Gomori trichrome staining, which reflect mitochondrial
INVESTIGATIONS proliferation (167). These fibers stain strongly for
Laboratory succinate dehydrogenase (SDH, complex II), and often
• Full blood count: may reveal pancytopenia. stain negatively for COX (complex IV) in CPEO, KSS, or
• Urea, electrolytes and creatinine: renal tubular dysfunc- MERFF but positively in MELAS. COX-negative fibers
tion may be seen as part of the Fanconi syndrome. are normal over age 40 years and the proportion increases
• Plasma glucose: commonly elevated (diabetes). with age, but does not normally exceed 5%. Ragged-red
• Creatine kinase: normal or mildly increased. fiber-positive and SDH-positive and COX-negative fibers
• Blood organic acid (lactate and pyruvate), amino acid, may also be seen in inflammatory myopathies.
and carnitine concentrations: lactic acidosis, raised • Ultrastructural analysis may show intramitochondrial
lactate : pyruvate ratio. paracrystalline inclusions (168).
• Urine (24-hour collection) organic acid, amino acid, and • Biochemical analysis of isolated mitochondria or muscle
carnitine concentrations: increased lactate, pyruvate and tissue by enzyme studies, polarography, or spectroscopy
alanine or a generalized aminoaciduria. may identify the site(s) of the defect within the
• EKG: cardiac conduction defects. respiratory chain, and this can help to direct molecular
• Audiography: sensori-neural hearing loss. genetic analysis.

167 168

167 Skeletal muscle biopsy, stained with modified Gomori trichrome, 168 Electron microscopy of skeletal muscle showing a
showing a ragged-red fiber with large pink subsarcolemmal aggregates subsarcolemmal collection of mitochondria with paracrystalline
of mitochondria (arrow). inclusions.
166 Inherited Metabolic Diseases of the Nervous System of Adult Onset

Molecular genetic analysis ADRENOLEUKODYSTROPHY (ALD)


Molecular genetic studies of DNA derived from blood
(platelets, white cells), hair follicles, urine or muscle are
analysed for mitochondrial DNA mutations: virtually all DEFINITION
point mutations can be detected readily in available tissues, An X-linked recessive peroxisomal disorder characterized by
such as leukocytes, but major structural mutations, such as an excessive accumulation and deposition of unbranched, very
deletions, require the analysis of skeletal muscle. Analysis is long chain saturated fatty acids (VLCFAs) with more than 22
necessary for genetic counselling. carbon atoms in many tissues and body fluids, leading to
adrenal failure and demyelination in the brain and spinal cord.
DIAGNOSIS The disease ranges from the rapidly progressive childhood
• Mitochondrial disease should be considered in any cerebral form, which is characterized by diffuse demyelination
patients with an unexplained multi-system neurologic in the cerebral hemispheres and leads to total disability during
disorder, particularly if there are eye signs and deafness. the first decade, to the milder adrenomyeloneuropathy
• The diagnosis is straightforward when the clinical (AMN), which is a distal axonopathy that most severely
findings indicate a well defined mitochondrial cytopathy involves the distal aspects of the long tracts in the spinal cord
syndrome. In less clear cut cases diagnosis can be difficult and which is compatible with survival to the eighth decade.
and, to be certain, the diagnosis should be supported by
at least two laboratory investigations, including serum EPIDEMIOLOGY
lactate analysis, muscle biopsy biochemistry and • Prevalence: 1/20 000 males.
histochemistry, and mitochondrial DNA studies on • Age: children, adolescents and young adults.
muscle or other tissues. – ALD affects children; AMN which is a milder form of
• There is no overall screening test that is sensitive and ALD, affects adults, with onset at 20–30 years of age.
specific for the presence of mtDNA mutations. • Gender: males and heterozygous females (sex-linked
Sequencing the entire mtDNA region is impractical. recessive).

TREATMENT PATHOLOGY
• Therapeutic trials have been limited with no adequate ALD
controlled studies. • Massive and diffuse demyelination, often asymmetrically,
• Various agents that naturally act as cofactors in the in various parts of the cerebral hemipheres, beginning
electron transport, or are designed to bypass decreased most commonly in the parieto-occipital region, and later
respiratory chain complex activity by providing an involving the brainstem, optic nerves and spinal cord
additional redox system have been assessed; these include (169).
coenzyme Q10 (ubidecarenone), succinate, vitamin K3 • Perivascular infiltration with lymphocytes and macro-
(menadione), vitamin C, thiamine, vitamin B2 phages, resembling that seen in multiple sclerosis.
(riboflavin), dichloroacetate, and idebenone 1, a novel • Degradation products of myelin are visible in macro-
quinone, among others. phages in recent lesions, namely sudanophilic demye-
• Corticosteroids have been associated anecdotally with lination. Axis cylinders are damaged but to a lesser degree.
improvement and with fatal metabolic acidosis. • Adrenal gland cortex atrophy.
• Gene therapy is a future hope. • Abnormally large amounts of VLCFAs in histiocytes in
brain and adrenals.
PROGNOSIS • Testes show marked interstitial fibrosis and atrophy of
Variable. the seminiferous tubules.
• Electron microscopy shows characteristic lamellar
cytoplasmic inclusions in the macrophages of the brain
and adrenals and the Leydig cells of the testes.

AMN
• A distal axonopathy most severely involving the distal
aspects of the long tracts in the spinal cord.
• The inflammatory reaction is mild or absent.

ETIOLOGY AND PATHOPHYSIOLOGY


• X-linked recessive inheritance.
• The disease maps to chromosome Xq28.
• Mutations have been described spread over most of the
X-ALD gene, and have been of different types: missense,
nonsense, deletions and insertions.
• The gene product ALD-P encodes a 70 kDa peroxisomal
membrane protein PMP70.
• The gene mutation is believed to result in an abnormality
in a peroxisomal VLCFA coenzyme A (CoA) synthetase
or ligase, ultimately resulting in impaired β-oxidation in
the peroxisomes and accumulation of VLCFAs in organs
such as the adrenals, brain and spinal cord.
Adrenoleukodystrophy (ALD) 167

• The same mutation can cause ALD or AMN within the AMN
same family but more commonly, the great majority of • Adult males.
X-ALD kindreds have different X-ALD gene mutations. • Chronic, progressive spinal cord syndrome (myelopathy)
Additional factors presumably play a part in determining characterized by spastic paraparesis, sphincter dysfunc-
the final phenotype. tion, and sometimes mild sensory loss.
• Adrenal insufficiency (70%): generalized weakness,
CLINICAL FEATURES weight loss, skin pigmentation, attacks of nausea and
The phenotypic presentation is extremely variable but vomiting; usually precedes the neurologic symptoms.
follows two broad patterns: a predominantly cerebral form • Peripheral neuropathy: difficult to detect in presence of
(ALD) occurs in about 35–50% of cases, and a spinal cord prominent pyramidal signs.
form (AMN) in 28–40%. In addition, another 8% manifest • Uncommonly, dementia and cerebellar ataxia may occur.
only as idiopathic Addison’s disease, and another 10% are • Heterozygote adult females (at least 20%) may develop,
asymptomatic at diagnosis, but many of whom develop in the fourth or fifth decade of life, a very slowly
some form of the disease later in life. progressive spastic paraparesis with mild sphincter and
Heterozygous women may also develop symptoms that sensory disturbance.
resemble those of AMN, but they tend to develop later in • A positive family history of slowly progressive spastic
life (up to 10 years later) and the lower limb spasticity and paraparesis may be present.
sensory deficits are milder.
The cause of the variable phenotypic expression is unclear DIFFERENTIAL DIAGNOSIS
and no correlation between genotype and phenotype has ALD
been found. • Multiple sclerosis.
• Diffuse cerebral sclerosis of Schilder (Schilder disease,
ALD encephalitis periaxalis diffusa): non-familial; children and
• Boys, less commonly adolescent and adult men. young adults; large, sharply outlined, asymmetric focus
• Pseudobulbar palsy: dysarthria; dysphagia; quadriparesis; of myelin destruction, often involving an entire lobe or
emotionalism. hemisphere of the brain, typically extending across the
• Dementia (i.e. progressive learning difficulties and corpus callosum; dementia, pseudobulbar palsy, varying
decline in scholastic performance). degrees of hemiparesis and paraparesis, homonymous
• Personality change. hemianopia, cortical blindness, deafness; chronic
• Cortical blindness. progressive or relapsing remitting course; no evidence of
• Deafness. adrenal atrophy or sex-linked male inheritance.
• Ataxia. • Other leukodystrophies (globoid cell leukodystrophy of
• Seizures occasionally. Krabbe [see p.171], metachromatic leukodystrophy, see
• Bronzing pigmentation of oral mucosa and skin around p.169) which have a specific inherited biochemical defect
nipples and over elbows, knees, and scrotum, before in the metabolism of myelin proteolipids and are
becoming more diffuse. characterized clinically by progressive visual failure,
• Rapidly progressive clinical course. mental deterioration, and spastic paralysis; and patho-
• Heterozygotes of ALD or AMN may be clinically logically by massive and more or less symmetrical
symptomatic. destruction of the white matter of the cerebral hemi-
• There may be a positive family history. spheres. Each may involve peripheral nerves as well as
CNS tissue.

AMN
• Hereditary spastic paraparesis.
• Multiple sclerosis.
169 • Spinal cord arteriovenous malformation.
• Spinal cord neoplasm.
• Dopa-responsive dystonia.

169 Brain, coronal section, showing massive, asymmetric demyelination


adjacent to the posterior horns of the lateral ventricles (arrows).
168 Inherited Metabolic Diseases of the Nervous System of Adult Onset

INVESTIGATIONS TREATMENT
• Urea and electrolytes: most patients have clinical or Adrenal insufficiency
biochemical evidence of impaired adrenal function and Adrenal steroid replacement therapy: may prolong life,
reserves: low sodium and chloride levels and elevated increase general strength, and improve school performance
potassium levels. but does not alter neurologic disability.
• Adrenal function tests: serum cortisol levels decreased,
urinary excretion of corticosteroids reduced, plasma Neurologic disability
ACTH increased, ACTH stimulation test negative: lack Three therapeutic approaches are under current
of rise in 17-hydroxyketosteroids after ACTH stimulation. investigation.
• Plasma VLCFAs (C26:0): increased several fold. The
plasma VLCFA ratio C26:0/C22:0 is also increased Dietary therapy
several-fold compared with values for control. Attempts to lower the levels of saturated VLCFAs have
• Skin fibroblast concentrations of VLCFAs: increased. included diets enriched in monounsaturated fatty acids
• CT brain scan: shows symmetric areas of low density in (oleic acid), devoid of VLCFAs, supplemented with glycerol
the parieto-occipital white matter, with later involvement (glyceryl) trioleate oil (GTO) and glycerol (glyceryl)
of the temporal, parietal and frontal lobes. This can trierucate (GTE) (the 4:1 mixture of GTO and GTE oil is
extend across the splenium of the corpus callosum and popularly referred to as Lorenzo’s oil), and containing
into the cerebellum. Contrast enhancement may occur erucic acid (an omega 9 mono-unsaturated 22-carbon fatty
around the periphery of the lesions. Occasionally there acid), which competes with saturated fatty acids for the
may be calcification and mass effect in the white matter microsomal fatty acid elongating enzyme system. Although
lesions. marked reduction of plasma levels of lignoceric (C24:0) and
• MRI brain scan (170) in ALD shows increased signal on hexacosanoic acid (C26:0) have been achieved, there has
T2W image, decreased on T1W image in the areas been little or no effect on neurologic progression in patients
outlined above, and loss of gray/white matter who are already neurologically affected. One explanation is
differentiation. MRI is more sensitive than CT and may that little erucic acid crosses the blood–brain barrier and
also show extension into the long motor tracts and visual enters the brain. At present, dietary therapy is of limited
pathways. Enhancement may occur at the edges of the value in correcting the accumulation of saturated VLCFAs
white matter lesions. Magnetic resonance spectroscopy in the brains of patients with ADL.
shows a diminution in the N-acetylaspartate peak and an
increase in the choline peak. In AMN, MRI brain is often Bone marrow transplantation
normal, but shows involvement of the spinal cord and When undertaken at an early stage of the disease in child-
peripheral nerves. hood, long term benefits have been described in small series.
• CSF protein may be elevated.
• Nerve conduction studies: often abnormal in AMN and Immunosuppression
suggest a mixture of axonal loss and multifocal Experimental.
demyelination.
• Molecular genetic analysis: for possible sequence PROGNOSIS
variations in the ALD gene by polymerase chain reaction • ADL: rapidly progressive and fatal disorder within 3–5
(PCR) amplification and single strand conformation years after clinical symptoms are detected.
polymorphism (SSCP) analysis. • AMN: slowly progressive course over years. About 35%
of patients will have substantial neurologic progression
DIAGNOSIS during a 3 year period, and cerebral involvement may
• Demonstration of excessive VLCFAs in plasma or skin develop in up to half at some point. The presence and
fibroblasts (a ketogenic diet can cause raised levels in severity of adrenal insufficiency has no bearing on the
normals) in a patient with appropriate clinical and severity of the neurologic disease.
neuroimaging findings and family history. • Male siblings of the patient have a 50% risk of receiving
• The progressive childhood form of X-ALD may be the ALD gene from their mother but because the
accompanied by ‘non-diagnostic’ concentrations of disorder is so clinically heterogeneous with various
plasma VLCFAs. phenotypes, the risk of a male sibling developing AMN
is less than 50%.
Presymptomatic screening
DNA analysis is the most reliable method for establishing the
carrier status in X-ALD kindred; about 10% of heterozygous
women have normal plasma VLCFA levels using current
assays. However, the distribution of mutations over the
whole coding region complicates such detection. Study of
the ALP-P expression in white blood cells or fibroblasts may
help circumvent this problem and identify heterozygous
women, particularly when the mutation is not identified.
Metachromatic Leukodystrophy (MLD) 169

METACHROMATIC – Caused by a mutation affecting the polyadenylation of


LEUKODYSTROPHY (MLD) the arylsulfatase mRNA.
– Low arylsulfatase is not accompanied by accumulation of
sulfatides in the organs, because the residual activity is sup-
DEFINITION posedly sufficient to ensure normal sulfatide metabolism.
An autosomal recessive lysosomal disorder characterized by – Memory disturbances, ataxia, fatigue, tremor or loss of
demyelination of the white matter in the CNS and the vision are present.
peripheral nerves. – Blood DNA analysis reveals homozygosity for the
pseudodeficiency allele.
EPIDEMIOLOGY • Multiple sclerosis.
• Incidence: rare (only 50 or so cases of adult form • Huntington’s disease.
described in the literature). • Parkinson’s disease.
• Age: late infantile form (age at onset, 1–2 years); juvenile
form (age at onset, 3–15 years); adult form (age at onset,
older than 16 years): mean age of onset 23 years; range
16–62 years. 170
• Gender: M=F.

PATHOLOGY
• Accumulation of sulfatides in the brain, peripheral nerves,
and non-neural organs.
• Demyelination of the periventricular white matter (171)
and peripheral nerves, with reduction of myelin sheath
thickness in peripheral nerve.

ETIOLOGY AND PATHOPHYSIOLOGY


• Deficiency of the enzyme arylsulfatase A (ASA), which
hydrolyses various sulfatides, including galactosyl
sulfatide and lactosyl sulfatide, the major sulfate-
containing lipids of the nervous system.
• Autosomal recessive inheritance.
• The gene is located on chromosome 22q13.
• Eight different MLD alleles:
– Type 0 alleles without residual activity of arylsulfatase;.
– Type R alleles with some residual activity of arylsulfatase.
• Late infantile form (age at onset, 1–2 years): homo-
zygosity for type 0 alleles.
• Juvenile form (age at onset, 3–15 years): compound
heterozygosity for type 0/type R alleles. 170 T2W axial MRI showing diffuse bilateral increased signal mainly in
• Adult form (age at onset, older than 16 years): the parieto-occipital white matter in adrenoleukodystrophy.The more
homozygosity for type R alleles. anterior white matter is beginning to look affected.

CLINICAL FEATURES
Late infantile form (age at onset, 1–2 years)
Gait and behavioral disturbance.
171
Juvenile form (age at onset, 3–15 years)
Gait and behavioral disturbance.

Adult form (age at onset, older than 16 years)


Progressive neurologic or psychiatric symptoms and signs,
which include any of:
– Dementia.
– Behavioral abnormalities: aggressiveness, irritability,
impaired social awareness, and inappropriate behavior.
– Ataxia.
– Paraparesis.
– Polyneuropathy.

DIFFERENTIAL DIAGNOSIS
• Pseudodeficiency of arylsulfatase:
– A common genetic polymorphism, with an estimated
gene frequency of 7.3%. 171 Brain, coronal section, showing extensive demyelination of the
periventricular white matter (arrows).
170 Inherited Metabolic Diseases of the Nervous System of Adult Onset

INVESTIGATIONS • Reduced myelinated fiber density and myelin sheath


Blood thickness.
• Arylsulfatase activity in leukocytes: decreased (less than • Ultrastructural examination reveals various types of
35–110 nmol/h per mg of protein). inclusion, mainly lamellar zebra-like bodies and tuffstone
• DNA analysis. bodies.

Urine DIAGNOSIS
Accumulation of sulfatides in urinary sediment. • Progressive symptoms of mental deterioration, behavioral
abnormalities, or ataxia in combination with hypo-
EMG myelination of the CNS and slowing of NCV.
Slowing of nerve conduction velocities (NCV): mean • Decreased arylsulfatase activity in leukocytes or
peroneal NCV: 25 m/s (range: 0–30 m/s [late infantile], fibroblasts (not specific).
10–28 m/s [juvenile], 15–39 m/s [adult]; normal • Increased amounts of sulfatides in urinary sediment.
44–57 m/s). • Morphologic demonstration of accumulation of
sulfatides in various tissues, e.g. sural nerve or brain.
CT scan of the brain • DNA analysis: to confirm or exclude a pseudodeficiency
Extensive areas of hypodensity in the white matter, especially state of arylsulfatase activity.
in the frontal lobes, which does not enhance (172, left).
TREATMENT
MRI brain Late infantile
Extensive increased signal in the white matter on T2W Bone marrow transplantation: conflicting results.
image (172, right) due to diffuse demyelination of the peri-
ventricular white matter. The imaging features on CT and CLINICAL COURSE AND PROGNOSIS
MRI are non-specific. Late infantile form (age at onset 1–2 years)
• Rapid course.
CSF • Fatal outcome.
Normal or slightly elevated protein concentration.
Juvenile form (age at onset 3–15 years)
Skin biopsy More protracted course.
Decreased arylsulfatase activity in fibroblasts
(<270–770 nmol/h per mg of protein). Adult form (age at onset, older than 16 years)
• Slowly progressive.
Peripheral (e.g. sural) nerve biopsy with • The interval between the onset of one symptom (e.g.
acidic cresyl violet staining behavioral abnormality or ataxia) and the occurrence of
• Accumulation of sulfatides as brown metachromatic the second symptom is about 3.7 years (range
deposits in Schwann cells and in large perivascular 1–20 years). The third symptom occurs, on average,
macrophages. 2 years (range 0–7 years) later.
• Segmental demyelination and remyelination with slight • In the final stages , most are demented, severely ataxic,
onion bulb formation (less active in adult than late and have behavioral abnormalities. Psychosis is very rare.
infantile and juvenile forms). • Death occurs 10 years (range 3–24 years) after onset of
symptoms.

172 172 CT brain scan (left);T2W MRI


(right) shows increased white
matter signal.
Adult-onset Globoid Cell Leukodystrophy (Krabbe Disease) 171

ADULT-ONSET GLOBOID CELL PATHOLOGY


LEUKODYSTROPHY (KRABBE Macroscopic
DISEASE) A marked reduction in the cerebral white matter, which feels
firm and rubbery.

DEFINITION Microscopic
A rare autosomal recessive disorder caused by deficiency of • Widespread demyelination and astrocytic gliosis in brain,
the lysosomal enzyme galactosylceramide β-galactosidase (β- spinal cord, and nerves.
galactocerebrosidase), resulting in accumulation of galacto- • Perivascular infiltration of large multinucleated macro-
cerebroside in the central and peripheral nervous systems, phages containing accumulated galactocerebroside
particularly the white matter of the brain and causing (globoid cells) (173).
demyelination. • Tubular or crystalloid inclusions in Schwann cell
One of the leukodystrophies, a group of inheritable cytoplasm seen on electron microscopy.
diseases in which the abnormal metabolism of myelin • Large-fiber, demyelinating, sensorimotor polyneuropathy.
components leads to progressive demyelination.
CLINICAL FEATURES
EPIDEMIOLOGY • Asymmetric spastic quadriparesis or hemiparesis, with
• Incidence: rare. brisk or absent/depressed reflexes.
• Age: adult-onset: a rare variant of the more common • Visual failure with optic atrophy.
infantile-onset form of Krabbe disease. • Cognitive decline/dementia in some patients.
• Gender: M=F. • Cerebellar ataxia.
• Peripheral neuropathy.
ETIOLOGY AND PATHOPHYSIOLOGY • Developmental delay, deafness, rigidity and seizures with
• Autosomal recessive inheritance. infantile onset.
• A variety of mutations of the human galactocerebrosidase
gene on chromosome 14q24.3-q32.1 have been DIFFERENTIAL DIAGNOSIS
identified. • Other lysosomal storage diseases: MLD, ALD, GM2
• Deficiency of the lysosomal enzyme galactosylceramide gangliosidosis.
β-galactosidase (β-galactocerebrosidase), which catalyses • Multiple sclerosis.
lysosomal hydrolysis of myelin-specific galactolipids • LHON.
including galactosylceramide (galactocerebroside) and • Wilson’s disease.
galactosylshingosine (psychosine), leads to the accumu- • Mitochondrial disease: Leigh disease.
lation of galactosylsphingosine (galactocerebroside) or
psychosine, which is neurotoxic to both the central and
peripheral nervous systems, resulting in demyelination of
the white matter of the brain and peripheral nerves.

173 Microscopic section, high power, of brain 173


white matter showing large histiocytes
containing galactocerebroside (globoid cells)
in a patient with Krabbe disease. (Courtesy
of Professor BA Kakulas, Royal Perth Hospital,
Australia.)
172 Inherited Metabolic Diseases of the Nervous System of Adult Onset

INVESTIGATIONS LATE-ONSET GM2 GANGLIOSIDOSIS


• EEG: normal initially, eventually non-specific slow,
disorganized background. Occasional multifocal
paroxysmal or epileptiform discharges are seen but DEFINITION
generally there are no spikes. The GM2 gangliosidoses comprise a spectrum of disorders of
• EMG: normal, or decrease in sensory and motor nerve nervous system function, each characterized by a deficiency
conduction velocity and evidence of denervation, consis- of either β-hexosaminidase A (Hex A) alone or in com-
tent with a large-fiber sensori-motor polyneuropathy. bination with a deficiency of β-hexosaminidase B (Hex B).
• CSF: normal 0.15–0.45 g/l (15–45 mg/dl)or elevated The classic infantile forms, Tay–Sachs disease and
protein (0.7–4.5 g/l). Sandhoff disease, result in megalencephaly, blindness,
• CT brain scan: normal or symmetric non-enhancing areas seizures, quadriparesis, and death in early childhood.
of reduced density in the internal capsule, basal ganglia Late-onset GM2 gangliosidosis is a variant form of
or deep white matter. Tay–Sachs disease characterized by onset of multisystem
• MRI brain: T2W images, show abnormal high signal, neurologic symptoms and signs in adolescence or in early
consistent with demyelination, in the periventricular adult life, and slower progression with survival into adult life.
white matter of the frontal, parietal and occipital lobes
and cerebellar white matter. EPIDEMIOLOGY
• Nerve biopsy: large-fiber sensori-motor demyelinating • Incidence: rare.
polyneuropathy may be seen. Electron microscopy • Age: onset in adolescence or early adult life.
reveals clusters of electron-dense filamentous inclusions • Gender: M=F.
surrounded by a plasma membrane within the Schwann
cell cytoplasm. PATHOLOGY
• White blood cell galactocerebrosidase activity: reduced. Macroscopic
• Cultured skin fibroblasts: markedly reduced galacto- Enlargement of the brain.
cerebrosidase activity.
• Molecular analysis of cloned galactocerebrosidase Microscopic
complementary DNA (cDNA) may reveal mutations • Loss of neurons and a reactive gliosis.
within the gene for galactocerebrosidase. For example, • Distension of remaining nerve cells throughout the CNS
there may be a G283 → A transition (Gly95 → Ser), which with glycolipid (174).
results in an inactive galactocerebrosidase enzyme, and a
G147 → C transversion located at the last base of exon 1 ETIOLOGY AND PATHOPHYSIOLOGY
(-1 position of the 5´ splicing sequences), which results • Autosomal recessive inheritance.
mostly in abnormally spliced mRNA. • Deficiency of Hex A, which normally cleaves the N-acetyl
galactosylamine from gangliosides, resulting in
DIAGNOSIS accumulation of GM2 gangliosides.
• Markedly reduced galactocerebrosidase activity in • A single point mutation in exon 7 of the Hex A gene,
leukocytes or fibroblasts. whereby a G → A substitution at nucleotide 805 causes
• Typical inclusions in Schwann cell cytoplasm. the substitution of glycine at position 269 of the Hex A
• Symmetric demyelinating lesions on MRI brain scan. α-chain with a serine (i.e. Gly269 → Ser substitution in
• Mutation in the galactocerebrosidase gene. the α-chain). The effect of this mutation is that it results
in an α-chain with reduced stability, indirectly affecting
TREATMENT its association with the β-chain of hexosaminidase so that
• Supportive: there is no effective medical treatment. it is not processed to mature Hex A.
• Bone marrow transplantation hasn’t proved effective but Or:
may have a role if undertaken early in the disease. • Compound heterozygosity for the functionally silent 4-
• Genetic replacement therapy may be the way of the bp insertion in exon 11 (which leads to deficiency of the
future: reversal of the galactocerebrosidase deficiency in α-chain mRNA and the absence of α-chain protein,
cultured cells has been accomplished by SL3-3 recom- typical of the infantile form of the disease) and for a
binant retrovirus harboring a human galactocere- mutation, T538 → C, resulting in the missense
brosidase cDNA. Tyr180 → His. The effect of this mutation is to decrease
the stability of the α-chain and indirectly affect the
PROGNOSIS formation of mature Hex A.
Progressive: can be slow or rapid.
CLINICAL FEATURES
Considerable clinical heterogeneity; features include:
• Progressive ataxia, dysarthria, incoordination and
irregular postural tremor: poor handwriting.
• Progressive proximal limb neurogenic muscle weakness:
may climb stairs by mounting one at a time rather than
by alternating the feet.
• Spasticity with or without Babinski signs bilaterally.
• Dystonia.
• Intellectual impairment.
• Seizures.
Late-onset GM2 Gangliosidosis 173

• Myoclonus. Skin
• Intermittent psychosis in some patients. Cultured fibroblasts: preparation of poly A+ mRNA.
• Restricted vertical and horizontal pursuit (supranuclear
gaze paresis). MRI brain
• Slow or hypometric horizontal and vertical saccades. MRI may show white matter disease, and brain atrophy with
• Ocular dysmetria. widening of cerebellar sulci and atrophy of brainstem.
• Internuclear ophthalmoplegia (unilateral).
Nerve conduction studies and EMG
DIFFERENTIAL DIAGNOSIS • Absent or reduced amplitude sensory nerve action
• Multiple sclerosis. potentials.
• Friedreich’s ataxia. • Normal or near-normal motor nerve conduction
• Vitamin E deficiency. velocities.
• Neuroacanthocytosis. • Widespread chronic partial denervation.
• Syphilis.
Sural nerve biopsy
INVESTIGATIONS Chronic neuropathy with predominant axonal degeneration
Blood and regeneration.
• Full blood count and film, including acanthocyte screen.
• Urea and electrolytes. Rectal mucosal biopsy
• Glucose. Glycolipid distension of ganglion cells.
• Liver function tests.
• Vitamin E, B12 and folate. DIAGNOSIS
• Creatine kinase. • Plasma, leukocyte and fibroblast hexosaminidase activity
• Lipid and protein electrophoresis. assay.
• Lysosomal enzyme analysis: hexosaminidase A activity in • DNA analysis.
plasma and leukocytes: reduced; isolation of genomic
DNA from leukocytes. TREATMENT
Supportive and symptomatic.
Chest x-ray
PROGNOSIS
Cerebrospinal fluid Slow progression with survival into adult life.

174

174 Distension of nerve cells with glycolipid.


174 Inherited Metabolic Diseases of the Nervous System of Adult Onset

NIEMANN–PICK DISEASE ETIOLOGY AND PATHOPHYSIOLOGY


TYPE C (NP-C) • Impaired intracellular homeostatic responses to
exogenous low-density lipoprotein (LDL)-derived
cholesterol due to a defect of intracellular cholesterol
DEFINITION esterification. The primary defect is still unknown.
A panethnic autosomal recessive neurovisceral storage • Abnormal egress of free cholesterol from lysosomes to
disease characterized by a unique error in cellular trafficking other cellular organelles such as plasma membrane and
of exogenous cholesterol, and resulting in a variable degree the Golgi apparatus.
of cognitive and behavioral decline, vertical supranuclear • Autosomal recessive inheritance.
ophthalmoplegia and ataxia. Previously termed atypical • Primary genetic defect still unknown.
juvenile lipidosis and juvenile dystonic lipidosis. • Gene locus: linked to chromosome 18q11-12 in most
patients.
CLASSIFICATION • Genetic heterogeneity.
• Type A: early infantile NP who die in infancy; primary
sphingomyelinase deficiency. CLINICAL FEATURES
• Type B: hepatosplenomegaly without nervous system Pre-school-onset
involvement; primary sphingomyelinase deficiency. • History of prolonged neonatal jaundice.
• Type C: slowly progressive neurologic disease. • Hepatosplenomegaly: non-progressive.
• Type D: slowly progressive neurologic disease but • Dystonia: initially focal in hand or foot and may later
appears restricted geographically to Nova Scotia. generalize.
• Ataxia, dysarthria and dysmetria.
EPIDEMIOLOGY • Vertical supranuclear gaze palsy:
• Incidence: underestimated, but rare. The adult variant – Impaired voluntary vertical saccades.
occurs in only 5% of patients with Niemann–Pick disease. – Normal horizontal saccades and oculocephalic reflexes.
• Age: childhood, adolescence or early adulthood. • Cognitive decline causing difficulty at school.
• Gender: M=F.
Adult-onset
PATHOLOGY • Progressive dementia and psychosis.
Brain (175) • Vertical supranuclear ophthalmoplegia (may not be
• Accumulation of unesterified cholesterol in lysosomes. present in adult disease).
• Light microscopy reveals storage material in ballooned • Ataxia, dysarthria and dysmetria.
cortical neurons. • Dystonia.
• Electron microscopy reveals storage material as • Cataplexy may occur.
membranous and osmiophilic granules in lysosomes. • Seizures can occur later in course.
• Axonal spheroids.
• Neurofibrillary tangles similar to those in Alzheimer’s DIFFERENTIAL DIAGNOSIS
disease: stain positive with Alz50 antibody and consist of School difficulty
paired helical filaments (in adults). • Attention deficit disorder.
• No senile plaques. • Other learning disabilities.
• Absence seizures.
Other tissues • Early dementias (see immediately below).
Foamy histiocytes (lipid-laden macrophages) in bone
marrow, liver, spleen, skin, skeletal muscle and eye.

175 175 Coronal


sections of the brain
at autopsy of a
patient with
Niemann–Pick
disease.
Niemann–Pick Disease Type C (NP-C) 175

Dementia • Skin fibroblast cultures with LDL-derived tritiated oleate:


• Neuronal ceroid lipofuscinosis. – Severely delayed rate of cholesterol esterification.
• Leukodystrophies. – Filipin staining reveals accumulation of unesterified free
• Subacute sclerosing panencephalitis. cholesterol in lysosomes.
• HIV infection. – Reduced acid sphingomyelinase activity in some patients:
• Multiple sclerosis. a secondary phenomenon due to excessive cholesterol
• Psychosis. sequestration.
• Fluorescence microscopy may be useful to detect cells
Dystonia from patients with sphingolipidoses by the color and
• Idiopathic torsion dystonia. subcellular distribution of the fluorescence, by the
• Dopa-responsive dystonia. presence of red lysosomes.
• Wilson’s disease.
• Amino- and organic acidopathies (glutaric aciduria, mito- DIAGNOSIS
chondrial diseases). • Vertical supranuclear gaze palsy associated with various
• GM2 gangliosidosis. degrees of splenomegaly and other neurologic signs.
• Characteristic inclusions in lipid-laden cells in tissues such
Cataplexy as skin or liver.
• Other sleep disorders.
• Epilepsy (akinetic/atonic seizures). Definitive diagnostic test
• Syncope. • Skin fibroblast cultures with LDL-derived tritiated oleate:
• Periodic paralysis. severely reduced rate of cholesterol esterification. Filipin
staining reveals accumulation of free cholesterol in
Vertical supranuclear gaze palsy lysosomes.
• Dorsal midbrain syndrome (upgaze: posterior commis- • There is no correlation between the degree of the
sure, downgaze: rostral interstitial nucleus of the medial cholesterol esterification defect and the clinical severity
longitudinal fasciculus). of the disease.
• Tumors: pineal, tectal.
• Hydrocephalus. TREATMENT
• GM2 gangliosidosis. Specific
• Mitochondrial disease. • Nil available to date that is effective.
• Non-ketotic hyperglycinemia. • Neither low cholesterol diet nor cholesterol-lowering
• Maple syrup urine disease. medication has been shown to be effective to date.

Isolated splenomegaly Supportive


• Leukemia. • Physical therapy to maximize mobility and minimize
• Lymphoma. contractures.
• Histiocytosis. • Speech and swallowing therapy to avoid aspiration
• Storage diseases (Gaucher, Niemann–Pick types A and B). pneumonia.
• Infections. • Psychologic support.
• Anticholinergic medication for dystonia.
Severe neonatal jaundice • Protriptyline hydrochloride or clomipramine hydro-
• Biliary atresia. chloride for cataplexy.
• Congenital infections. • Anti-epileptic medication for epilepsy.
• Alpha1-antitrypsin deficiency. • Haloperidol or other antipsychotic drugs for psychotic
• Tyrosinemia. manifestations.
• Gastrostomy in late stages if swallowing no longer
INVESTIGATIONS possible.
• Blood: normal chemistry, hypercholesterolemia is not a
feature. Genetic counselling
• PCR-based DNA linkage study: linkage to the major • Parents who are both carriers of the disease gene have a
18q11-12 Niemann–Pick type C locus. 25% chance of having an affected child in each pregnancy.
• CT / MRI brain: may be normal or show multiple areas • Pre-natal diagnosis: cultured chorionic villus biopsy
of demyelination (high signal on T2W image), gliosis specimens or cultured amniotic cells with LDL-derived
and moderate atrophy. tritiated oleate for rate of cholesterol esterification.
• Proton magnetic resonance spectroscopy: decreased N- • Obligatory carriers of the gene that causes NP-C cannot
acetyl aspartate and increased choline in various parts of be detected by these methods because carriers have
brain of moderately to severely affected patients. cholesterol esterification and free cholesterol accumu-
• Brainstem auditory evoked potentials: absent acoustic lation in lysosomes that range between normal and half
reflex early in disease. the values of affected homozygotes.
• CSF: normal.
• Abdominal ultrasound: may show hepatosplenomegaly. PROGNOSIS
• Bone marrow aspirate: lipid-laden phagocytic foam cells • Pre-school onset: inexorable progression to death,
with numerous small cytoplasmic vacuoles (sea-blue usually in the teenage years.
histiocytes). • Progression is slower in patients with later onset.
176 Inherited Metabolic Diseases of the Nervous System of Adult Onset

ADRENOLEUKODYSTROPHY ADULT-ONSET GLOBOID CELL


FURTHER READING Bezman L, Moser AB, Raymond GV, et al. (2001) LEUKODYSTROPHY
Adrenoleukodystrophy: incidence, new muta- Bernardini GL, Herrera DG, Carson D, et al.
tion rate, and results of extended family (1997) Adult-onset Krabbe’s disease in sib-
GENERAL screening. Ann. Neurol., 49: 512–517. lings with novel mutations in the galactocere-
Gray RGF, Preece MA, Green SH, et al. (2000) Ito R, Melhem ER, Mori S, et al. (2001) Diffu- brosidase gene. Ann. Neurol., 41: 111–114.
Inborn errors of metabolism as a cause of neu- sion tensor brain MR imaging in X-linked Satoh J-I, Tokumoto H, Kurohara K, et al. (1997)
rological disease in adults. An approach to in- cerebral adrenoleuco dystrophy. Neurology, 56: Adult-onset Krabbe disease with homozygous
vestigation. J. Neurol. Neurosurg. Psychiatry, 544–547 T1853C mutation in the galactocerebrosidase
69: 5–12. Moser HW (1997) Adrenoleukodystrophy: phe- gene. Unusual MRI findings of corticospinal
notype, genetics, pathogenesis and therapy. tract demyelination. Neurology, 49:
WILSON’S DISEASE Brain, 120: 1485–1508. 1392–1399.
Brewer GJ, Johnson V, Dick RD, et al. (1996) Moser HW (1999) Treatment of X-linked
Treatment of Wilson disease with ammonium adrenoleukodystrophy with Lorenzo’s oil. J. LATE-ONSET GM2 GANGLIOSIDOSIS
tetrathiomolybdate. Arch. Neurol., 53: Neurol. Neurosurg. Psychiatry, 67: 279–280. De Gasperi R, Gama Sosa MA, Battistini S, et al.
1017–1025. Sawaya RA (2000) Adrenoleukodystrophy: a re- (1996) Late-onset GM2 gangliosidosis: Ashke-
Fink JK, Hedera P, Brewer GJ (1999) Hepatolen- view. The Neurologist, 6: 214–219. nazi Jewish family with an exon 5 mutation
ticular degeneration (Wilson’s disease). The Shapiro E, Krivit W, Lockman L, et al. (2000) (Tyr180ÆHis) in the Hex A a-chain gene.
Neurologist, 5: 171–185. Long-term effect of bone-barrow transplanta- Neurology, 47: 547–552.
Pyeritz RE (1997) Genetic heterogeneity in Wil- tion for childhood-onset cerebral X-linked
son disease: Lessons from rare alleles. Ann. In- NIEMANN-PICK DISEASE TYPE C
adrenoleukodystrophy. Lancet, 356: 713–718. Chen C-S, Patterson MC, Wheatley CL, O’Brien
tern. Med., 127: 70–72.
METACHROMATIC LEUKODYSTROPHY JF, Pagano RE (1999) Broad screening test
MITOCHONDRIAL (OXIDATIVE Chen C-S, Patterson MC, Wheatley CL, O’Brien for sphingolipid-storage diseases. Lancet, 354:
PHOSPHORYLATION) DISEASES JF, Pagano RE (1999) Broad screening test 901–905.
Leonard JV, Schapira AHV (2000) Mitochondri- for sphingolipid-storage diseases. Lancet, 354: Lossos A, Schlesinger I, Okon E, et al. (1997)
al respiratory chain disorders I: mitochondrial 901–905. Adult-onset Niemann-Pick type C disease.
DNA defects. Lancet, 355: 299–304. Hageman ATM, Gabreels FJM, de Jong JGN, et Arch. Neurol., 54: 1536–1541.
Leonard JV, Schapira AHV (2000) Mitochondri- al. (1995) Clinical symptoms of adult Schiffmann R (1996) Niemann-Pick disease type
al respiratory chain disorders II: neuro- metachromatic leucodystrophy and arylsul- C. From bench to bedside. JAMA, 276:
degenerative disorders and nuclear gene de- phatase A pseudodeficiency. Arch. Neurol., 52: 561–564.
fects. Lancet, 355: 389–394. 408–413.
Chapter Nine 177

Traumatic Diseases of
the Nervous System
SUBDURAL HEMATOMA ETIOLOGY
• Head trauma causing rupture of small pial vessels:
particularly relevant in acute subdural hematoma; may
DEFINITION be minor in chronic subdural hematoma among elderly
Hemorrhage into the subdural space, usually caused by alcoholics and patients on anticoagulants.
rupture of bridging veins which pass from the pia-arachnoid • Rupture of:
over the brain to a dural sinus. – A saccular or mycotic aneurysm of a major intracerebral
artery.
EPIDEMIOLOGY – An intracavernous aneurysm of the carotid artery.
• Incidence: 6 (95% CI: 3–12) per 100 000 per year. – An aneurysm of the middle cerebral artery.
• Lifetime prevalence: 0.3 (95% CI: 0.2–0.6) per 1000. – An arteriovenous malformation; a vascular tumor.
• Age: all age groups but more frequent among the elderly. • Moyamoya syndrome.
The average age is about 65 years. • Ventricular decompression for hydrocephalus.
• Gender: M=F. • Dural metastases.
• Lumbar puncture.
PATHOLOGY
Acute subdural hematoma (<3 days old) Risk factors
A collection of fresh clotted blood in the subdural space, • Older age: presumably cerebral atrophy makes the
frequently combined with epidural hemorrhage, cerebral bridging veins to the venous sinuses more vulnerable to
contusion and laceration (176, 177). an acceleration/deceleration injury.
• Alcohol abuse.
Subacute subdural hematoma (3 days–3 weeks old) • Bleeding diathesis: anticoagulant drug use, the most
A mixture of subdural clot and liquid. common precipitant, accounting for about 25% of all
subdural hematomas, and 50% of those without obvious
Chronic subdural hematoma (>3 weeks old) trauma; thrombolytic treatment.
• Almost entirely liquid (and some altered blood) in the • CSF shunt.
subdural space, encysted by fibrous membranes (pseudo- • Epilepsy.
membranes) which have grown from the dura.
• Bilateral in 20–25% of cases.

176 Autopsy specimen of 176 177


a fatal subdural hematoma
over the right hemisphere
(arrows).

177 Autopsy specimen of


a fatal subdural hematoma
over the parasagittal region
of one hemisphere,
beneath the dura, which
has been retracted.
178 Traumatic Diseases of the Nervous System

PATHOPHYSIOLOGY – Confusion, apathy, drowsiness and stupor.


Acute subdural hematoma – Decline in cognitive function and speed of thinking
Caused by traumatic tearing of subdural cortical surface (subacute dementia).
vessels or bridging veins to the venous sinuses. Symptoms are – Mild focal neurologic symptoms and signs (e.g.
caused by an expanding fresh blood clot, compressing incomplete hemiparesis, aphasia); all of which may
underlying brain (causing focal neurologic signs) and raising fluctuate, and may be transient.
intracranial pressure (causing drowsiness and confusion), • Patients presenting late may be in coma or have a hemiplegia.
leading to transtentorial herniation if not arrested. • Seizures occur rarely, mostly in alcoholics or patients with
Unlike arterial hemorrhage into the epidural space, cerebral contusions.
venous hemorrhage into the subdural space is usually
arrested by rising intracranial pressure. SPECIAL FORMS
Subdural hygroma
Chronic subdural hematoma • A collection of blood and CSF in the subdural space.
Injury leads to hemorrhage from a subdural bridging vein • Pathophysiology: thought to be due to a tear in the arach-
or cortical surface vessel into the subdural space over one noid which permits CSF to pass from the subarachnoid
cerebral hemisphere and occasionally over both hemi- into the subdural space, where it may accumulate, presum-
spheres, interhemispheric (usually aneurysmal) or subcere- ably due to a ball-valve effect of the arachnoidal tear.
bellar. The hemorrhage is under low pressure and may be • Etiologic factors:
small and not of any clinical consequence. – Head injury.
Over the next few days, fibroblasts invade the clot, and – Meningitis as an infant or young child.
fine connective tissue membranes form over the inner – Ventriculoperitoneal shunting of a hydrocephalic brain.
(cortical) and outer (dural) surfaces, so that at about – Pneumoencephalography (in the past).
3 weeks, the hematoma becomes completely enclosed by • Clinical features: asymptomatic or confusion, drowsiness,
membrane and attached to the dura. During this time, the irritability and fever.
clot progressively breaks up and liquefies. • CT brain scan shows a subdural fluid collection of the
The liquefied hematoma is resorbed in many cases but in same density as CSF. No contrast enhancement is seen
others it gradually enlarges, probably because of repeated because there are no vascular membranes.
small hemorrhages from the fine, friable vessels in the • Treatment: as for chronic subdural hematoma: aspirate
connective tissue membranes that have formed. Hemorr- or drain the subdural fluid if symptomatic.
hage may be promoted by accumulation of fibrin degrada-
tion products in the liquefied hematoma, and depletion of DIFFERENTIAL DIAGNOSIS
coagulation cascade components. Acute epidural hemorrhage
It is likely that there is a balance between resorption and • Usually a history or evidence of head trauma, or a
enlargement of the hematoma: if production exceeds resorp- temporal or parietal skull fracture.
tion, the hematoma enlarges and acts as a space-occupying • Consciousness may not have been disturbed, or may have
mass, compressing the underlying brain (causing focal been lost momentarily, followed a few hours or a day later (the
neurologic signs) and eventually producing distortion and interval may be longer with venous bleeding) by headache of
herniation of the brain. increasing severity, with vomiting, drowsiness, confusion, and
Chronic subdural hematomas tend to expand slowly, seizures or signs of hemispheric dysfunction such as dysphasia
allowing the brain to compensate to some extent, so that at and hemiparesis. As coma develops, the pupil on the side of
times the hematoma may be quite large with considerable the hematoma dilates (because the IIIrd nerve is compressed
brain distortion before causing symptoms. in the tentorial notch by downward displacement of the
temporal lobe), bilateral corticospinal tracts signs may develop,
CLINICAL FEATURES and signs of transtentorial herniation ensue (see p.44).
Depend on the site and size of the hematoma and maximum • X-ray of the skull may show a fracture line.
brain compression, and the rate of hematoma enlargement. • CT or MRI scan of the brain establishes the diagnosis by
revealing a lens-shaped clot with a smooth inner margin (180).
Acute subdural hematoma • Lumbar puncture is contraindicated.
Supratentorial • Surgical evacuation of the clot and ligation of the bleeding
Usually initial loss of consciousness (from the head trauma), vessel (usually the middle meningeal artery) can be life-saving.
which may persist or be followed by a short lucid interval of Alternatively, if the condition is advancing too rapidly for any
several hours where consciousness is regained, followed by investigation to be done, the placement of burr holes on the
progressive headache, confusion and deterioration of conscious same side of the skull as the dilated pupil can also be life-saving.
state. Lateralizing neurologic signs may be present.
Subarachnoid hemorrhage
Infratentorial Some chronic subdural hematomas may present with acute
Headache, vomiting, dysphagia, pupillary inequality, cranial headache, mimicking SAH (see p.249).
nerve palsies, truncal and gait ataxia, and, rarely, a stiff neck.
Transient ischemic attack or stroke
Chronic subdural hematoma Subdural hematoma may rarely present with the abrupt onset
• Most patients are elderly. of focal neurologic signs, and they may be initially transient.
• About half recall a head injury of varying severity.
• Subacute onset, over several days to weeks, of: Subacute delirious reaction
– Headache (present in about 80% of cases). Drug intoxication.
Subdural Hematoma 179

Depression the inner table of the skull and the cortex. The mass
effect may cause midline shift and compression of the
Dementia ipsilateral lateral ventricle (181).
Alzheimer’s disease (see p.407). • Subacute subdurals (i.e. around 10–21 days old) may be
isodense with brain and difficult to identify unless
Brain tumor (see p.357) secondary features such as compression of the ipsilateral
ventricle, midline shift and loss of cortical sulci are
INVESTIGATIONS sought. Contrast may help to highlight subacute
Cranial CT scan subdurals (the vascular membrane will enhance making
• The imaging investigation of subdural hematomas is with the cortical margin more visible).
CT – this will exclude most acute and chronic subdurals • Chronic subdurals are hypodense (black) so are easier to
(178, 179), and acute extradurals (180), if the scan is see (179). They may also have mass effect depending on
carefully examined. their size.
• Acute subdural hematomas are hyperdense (white) • In cases of doubt a repeat CT scan 1 week or so later or
compared with normal brain, crescentic, lying between an MRI will sort it out.

178 Acute on chronic 178 179


subdural hematoma over
the left hemisphere. Note
the layer of higher density
sediment posteriorly
indicating some recent
hemorrhage
(arrowheads) and the
considerable midline shift.

179 Bilateral chronic


subdural hematomas (or
hygromas as sometimes
called [arrowheads]).
Note the absence of
cortical sulci and the
compressed lateral
ventricles indicating that
these are not just large
CSF spaces due to
atrophy.

180 Acute extradural 180 181


hematoma over the
right parietal lobe for
comparison. Note the
whiteness (indicating
fresh hemorrhage) and
the localized position
(subdurals tend to spread
more extensively over the
hemisphere).

181 Plain cranial CT scan


showing a recent subdural
hematoma (arrowheads)
as a high density (white)
over the right hemisphere
causing mass effect with
compression of the right
lateral ventricle and
midline shift.
180 Traumatic Diseases of the Nervous System

MRI • If the CT brain scan suggests that a significant amount


• MRI is more sensitive for detecting hematomas; the of actual clot is present or that the membranes have
properties of hemoglobin breakdown products give created a loculated chronic subdural hematoma (CSDH),
signal changes that make even small subdurals stand out a craniotomy may the most appropriate choice.
quite clearly no matter what their age. • A reduction of 20% of the volume of the subdural fluid
• The problem with using MR in the first instance is that is all that is required to reduce the subdural pressure to
these patients are often confused and unwell, so they close to zero, which usually leads to clinical improve-
move in the MR scanner and the end result may not be ment. In addition to reducing any pressure effects,
diagnostic. It is better to go for the quick technique that drainage allows washing out of the high concentration
suits ill patients, which is CT. of fibrin degradation products which have contributed
to the CSDH enlargement, thus shifting the balance
EEG back in favor of resorption. It is uncommon for the brain
Usually abnormal bilaterally, sometimes with reduced to immediately re-expand following its period of
voltage or electric silence over the hematoma (due to compression. Post-operative CT brain scan usually
dampening effects) and high-voltage slow waves over the demonstrates some residual subdural fluid and air, and it
contralateral hemisphere, because of the displacement of the may take up to 6 months for the CT to show complete
brain to that side. Unilateral focal delta activity is also quite resolution of the hematoma. The patient’s clinical course
non-specific. is therefore more important for detecting complications
than the post-operative CT scan.
CSF
Not indicated, but if done, the pressure may be normal or Complications
elevated; the appearance may be clear and acellular, bloody • Post-operative infection, particularly subdural empyema,
or xanthochromic, depending on the presence or absence occurs in <1% of patients but can be fatal.
of recent or old contusions and subarachnoid hemorrhage; • Reaccumulation, presumably caused by bleeding from
and the protein may be relatively low. the vascular outer membrane, occurs in 10–20% of
patients, and may lead to deterioration or failure to
Cerebral angiography improve post operatively. CT scan usually shows a new
Shows displacement of cortical branches of the middle collection of hyperdense or mixed density hematoma,
cerebral arteries inward from the inner surface of the skull, and redrainage is generally required.
and contralateral displacement of the anterior cerebral artery. • Epilepsy may occur in up to 10% of patients.

DIAGNOSIS NATURAL HISTORY


• Suspect in any patient with recent onset of headache or Usually progressive or fluctuating clinical course, sometimes
drowsiness, progressive impairment of memory or with spontaneous resolution. If the level of consciousness
intellect, or other symptoms suggestive of a space- remains normal, spontaneous resolution may occur, even
occupying lesion. with an acute onset. However, sudden deterioration may
• Confirm by cranial CT scan, but beware that, after some occur days or weeks after onset.
days, the density of the hematoma approaches that of
surrounding tissue and may be difficult to detect. PROGNOSIS
• If the CT appears normal and the index of suspicion is 85–90% of patients make a good functional recovery with
high, MRI is the definitive investigation; bilateral carotid appropriate treatment. The outcome correlates with the
angiography used to be. patient’s neurologic condition at the time of diagnosis and
treatment: most patients with poor outcomes are
TREATMENT significantly impaired (e.g. comatose) before treatment.
• Conservative, with careful observation, if there is no
definite neurologic compromise or significant radiologic
mass effect (e.g. if the maximum thickness is less than
1 cm [0.4 in]).
• Rapid correction of the coagulation status, if appropriate
(e.g. anticoagulant-associated subdural).
• Active neurosurgic intervention is indicated if neurologic
symptoms and signs are severe and progressing, and if
the clot is large enough to explain the clinical features.
Medical treatments such as mannitol and corticosteroids FURTHER READING
are ineffective.
• The surgical options include:
– Making a simple twist drill hole that allows the subdural Maurice-Williams RS (1999) Chronic subdural haematoma: an everyday
problem for the neurosurgeon. Br. J. Neurosurg., 13: 547–590.
fluid to escape Maurice-Williams RS (2001) Bedside treatment of chronic subdural
– Making one or two burr holes and washing out the haematoma? Lancet, 357: 1308–1309.
subdural space. Reinges MHT, Hasselberg I, Rohde V, et al. (2000) Prospective analysis of
– Performing a formal craniotomy, which may be accom- bedside percutaneous tapping for the treatment of chronic subdural
haematomas in adults. J. Neurol. Neurosurg. Psychiatry, 69: 40–47.
panied by excision of some of the subdural membrane. Van Havenbergh T, Van Calenbergh F, Griffin J, Plets C (1996) Outcome
• A burr hole or twist drill hole is a lesser undertaking than of chronic subdural haematomata, and analysis of prognostic factors. Br.
a formal craniotomy. J. Neurosurg., 10: 35–39.
Chapter Ten 181

Vascular Diseases of
the Nervous System
NEUROVASCULAR SYNDROMES: ANATOMIC (see Table 23) AND
CLINICAL (see Table 24) CLASSIFICATION

Table 23 Anatomic classification


Arterial territory Area supplied Typical syndrome if total territory involved

Internal carotid artery*


Ophthalmic artery Retina and optic nerve Monocular blindness or altitudinal field defect

Anterior choroidal artery Globus pallidus (internal) Contralateral hemiparesis, hemisensory loss, and homonymous hemianopia
Internal capsule(posterior limb)
Choroid plexus

Middle cerebral artery (MCA) Frontal lobe Contralateral central facial weakness, hemiparesis and hemisensory loss (arm>leg),
Parietal lobe homonymous hemianopia, and global aphasia (dominant hemisphere) or visual-spatial-
Superior temporal lobe perceptual dysfunction (non-dominant hemisphere)

Medial lenticulostriate Internal capsule Contralateral pure motor hemiparesis

Lateral lenticulostriate Putamen Contralateral hemiparesis, dysphasia (dominant hemisphere) or visual-spatial-


Globus pallidus (external) perceptual dysfunction (non-dominant hemisphere)
Caudate nucleus
Internal capsule
Corona radiata

Superior division of MCA Frontal and anterior parietal lobes Contralateral central facial weakness, hemiparesis and hemisensory loss, ipsilateral
deviation of head and eyes, and global or motor aphasia (dominant hemisphere)
Pre-rolandic branch Contralateral face and arm weakness, and motor aphasia (dominant hemisphere)
Rolandic branch Contralateral central facial weakness, hemiparesis and hemisensory loss; and
dysarthria (resembling lacunar syndrome)
Anterior parietal branch Conduction aphasia and bilateral ideomotor apraxia

Inferior division of MCA Inferior parietal and Homonymous hemianopia,Wernicke’s aphasia or agitated confusional state
lateral temporal lobes (dominant hemisphere), left visual neglect (right-sided lesion)
Posterior parietal branch
Angular branch
Posterior temporal branch
Anterior temporal branch
Temporal polar branch

Anterior cerebral artery Anterior and superior Contralateral foot and leg weakness or hemiparesis (leg>arm), abulia, incontinence,
medial frontal lobe grasp reflexes

Vertebrobasilar territory
Vertebral and basilar artery** Brainstem and cerebellum Various syndromes including diplopia, ophthalmoplegia, or gaze palsies;
vertigo, nausea and nystagmus; dysarthria, dysphagia, and bulbar weakness;
ipsilateral facial sensory loss and weakness (nuclear or infranuclear);
hiccoughs and respiratory failure; contralateral hemiparesis or tetraparesis,
contralateral or bilateral sensory loss, coma
Continued on page 182

* Complete internal carotid artery occlusion, if symptomatic, usually produces symptoms in the MCA territory, but the ophthalmic and anterior cerebral
artery territories can also be involved alone, or in combination with the MCA, depending on collateral supply.
** Basilar artery occlusion may also involve the territory of one or both posterior cerebral arteries.
N.B: Incomplete syndromes are common, and the symptoms of infarction and hemorrhage are similar.
182 Vascular Diseases of the Nervous System

Table 23 (continued)

Arterial territory Area supplied Typical syndrome if total territory involved

Basilar artery
Top of basilar artery Rostral midbrain Variable pupillary abnormalities
Part of thalamus Ptosis or lid retraction
Inferior temporal, Supranuclear vertical gaze paresis
occipital lobes Somnolence
Hemiballismus
Amnesia
Cortical blindness

Superior cerebellar artery Midbrain (dorso-lateral), Ipsilateral Horner’s syndrome


superior cerebellar peduncle, Ipsilateral limb ataxia and tremor
superior cerebellum Contralateral spinothalamic sensory loss
Contralateral central facial weakness
Contralateral IVth nerve palsy sometimes

Anterior inferior Base of pons Ipsilateral Horner’s syndrome


cerebellar artery Rostral medulla Ipsilateral facial sensory loss (pain, temperature)
Rostral cerebellum Ipsilateral nuclear facial and abducens palsy
Cochlea Ipsilateral deafness and tinnitus
Vestibule Vertigo, nausea, vomiting and nystagmus
Ipsilateral ataxia of limbs and dysarthia

Posterior inferior Lateral medulla, Ipsilateral Horner’s syndrome


cerebellar artery inferior cerebellum Ipsilateral facial sensory loss (pain, temperature)
Vertigo, nausea, vomiting and nystagmus
Ipsilateral paralysis of palate (dysphagia)
Ipsilateral paralysis of larynx (dysphonia)
Ipsilateral ataxia of limbs
Contralateral hemisensory loss below neck

Paramedian branches Paramedian pons Any of the lacunar syndromes:


Pure motor hemiparesis
Ataxic hemiparesis
Pure hemisensory loss
Hemiparesis-hemisensory loss
Internuclear ophthalmoplegia
Locked-in syndrome, if bilateral

Posterior cerebral artery Occipital lobe Contralateral homonymous hemianopia


Cortical blindness if bilateral
Inferior temporal lobe Amnesia (especially if bilateral)

Paramedian Rostral medial midbrain Hemisensory-motor loss


mesencephalic arteries

Thalamic-subthalamic Postero-medial Hemisensory loss, amnesia


(thalamoperforating) thalamus inferiorly

Posterior communicating artery


Polar arteries Anterior lateral thalamus Hemisensory loss, amnesia
(tuberothalamic)

Thalamo-geniculate Ventrolateral thalamus Pure hemisensory loss

Posterior choroidal Anterior and posterior Hemisensory loss, amnesia


arteries thalamus

* Complete internal carotid artery occlusion, if symptomatic, usually produces symptoms in the MCA territory, but the ophthalmic and
anterior cerebral artery territories can also be involved alone, or in combination with the MCA, depending on collateral supply.
** Basilar artery occlusion may also involve the territory of one or both posterior cerebral arteries.
N.B. Incomplete syndromes are common, and the symptoms of infarction and hemorrhage are similar.

182 T2W axial MRI of an infarct in the right hemisphere (arrow),


obtained at 12 hours after symptom onset, in a patient with a left
hemiparesis, visual-spatial-perceptual dysfunction and a left homonymous
hemianopia (a total anterior circulation syndrome). Note the altered
signal in gray and white matter and the mass effect.
Neurovascular Syndromes 183

Table 24 Clinical features, anatomy, pathology, etiology, and prognosis of the four clinical stroke syndromes

Total anterior circulation Partial anterior circulation Lacunar syndrome Posterior circulation syndrome
syndrome (TACS) (182, 183) syndrome (PACS) (184–186) (LACS) (187) (POCS) (188–194)

Clinical features 1 Hemiparesis and Any combination of two of 1 Hemiparesis or Brainstem symptoms
hemisensory loss and preceding three in TACS 2 Hemisensory loss or and signs (e.g. diplopia,
2 Homonymous hemianopia and or (3) alone, or monoparesis 3 Hemisensorimotor vertigo, dysphagia, ataxia,
3 Cortical dysfunction loss or bilateral limb deficits,
(dysphasia or visual-spatial- 4 Ataxic hemiparesis hemianopia or cortical
perceptual dysfunction) blindness)
No hemianopia or
cortical dysfunction

Anatomy Fronto-temporal-parietal lobes Lobar Small deep lesion in Brainstem and/or cerebellum
or either corona radiata,
Thalamus/internal internal capsule,
capsule/occipital lobe thalamus or ventral
pons

Pathology Infarction (85%) or Infarction (85%) or Infarction (95–98%) or Infarction (85%) or


Hemorrhage (15%) Hemorrhage (15%) Hemorrhage (2–5%) Hemorrhage (15%)

Etiology Infarction: occlusion of ipsilateral Infarction: occlusion of Infarction: usually Infarction: occlusion of
ICA or MCA, and occasionally branch of MCA or PCA; perforating artery VBA or PCA, or branches;
PCA; by embolism from heart, by embolism from heart, microatheroma/ by in situ thrombosis or
aortic arch or carotid or aortic arch, or carotid or lipohyalinosis or embolism from heart,
vertebrobasilar arteries, vertebrobasilar arteries rarely arteritis or aortic arch or VBA
or in situ thrombosis embolism

Hemorrhage: any of possible Hemorrhage: any of Hemorrhage: any, Hemorrhage: any


causes (see p.238) possible causes (see p.238). but usually of possible causes (see p.238)
hypertensive small
vessel disease

Recurrence rates Low High in first 3 months Low but steady over High in first 2 months
12 months and steady over 12 months

Prognosis at Poor Fair Fair Fair


1 year
Dead 60% 15% 10% 20%
Dependent 35% 30% 30% 20%
Independent 5% 55% 60% 60%

ICA: internal carotid artery; MCA: middle cerebral artery; PCA: posterior cerebral artery;VBA: vertebrobasilar artery

182 183

183 Brain, coronal section showing necrosis of the right parietal and
temporal lobes, basal ganglia and internal capsule (arrow) due to an old
infarct in the right middle cerebral artery territory causing a total anterior
circulation syndrome. (Courtesy of Professor BA Kakulas, Department of
Neuropathology, Royal Perth Hospital, Australia.)
184 Vascular Diseases of the Nervous System

184 185 184,185 Plain cranial CT


scan showing hyperdensity
(due to blood clot) in the
origin of the left middle
cerebral artery (184,
arrow), and left
striatocapsular infarction in a
patient who presented with
a partial anterior circulation
syndrome (right hemiparesis
and dysphasia/cognitive
deficit) (185, arrow).

186 187

187 Brain at autopsy showing a lacunar infarct (arrow) in the internal


capsule of a patient with a previous lacunar syndrome. (Courtesy of
Professor BA Kakulas, Department of Neuropathology, Royal Perth
Hospital, Australia.)

186 Plain cranial CT scan showing a 188


homogenous area of high density due to
hemorrhage in the right frontal lobe of an elderly
man with amyloid angiopathy and a partial
anterior circulation syndrome (left hemiparesis
and cognitive/visual-spatial-perceptual
dysfunction).

188, 189 Eye signs of a patient with a posterior


circulation syndrome due to an embolus to the
top of the basilar artery causing midbrain
infarction (188). Note the lid retraction, skew
deviation, right internuclear ophthalmoplegia, and
upgaze palsy (189).
Neurovascular Syndromes 185

189 190

189 Lid retraction, skew deviation, right internuclear ophthalmoplegia,


and upgaze palsy in a patient with a posterior circulation syndrome due
to an embolus to the top of the basilar artery causing midbrain
infarction (see also 188).

190 MRI brain scan,T2W image, showing an area of high attenuation,


consistent with recent infarction in the right lateral medulla (arrow).

191

192

191 MR angiogram showing patchy signal in the right vertebral artery


consistent with dissection of the right vertebral artery (arrow).

193
192 Autopsy specimen of brain, horizontal slice through the medulla
at the level of the olives, showing pallor in the right lateral medulla
(arrow).

193, 194 MRI scan,T2W image (193), and ventral surface of the
cerebellum and medulla at autopsy (194) showing infarction in the
right posterior inferior cerebellum (arrows) due to occlusion of the
posterior inferior cerebellar artery.

194
186 Vascular Diseases of the Nervous System

TRANSIENT ISCHEMIC ATTACKS (TIA) Somatosensory


OF THE BRAIN AND EYE Altered feeling on one side of the body, in whole or in part.

Visual
DEFINITION • Loss of vision in one eye, in whole or in part.
A clinical syndrome characterized by: • Loss of vision in the left or the right half of the visual field.
• Sudden onset of loss of focal brain or monocular function. • Bilateral blindness.
• Symptoms are thought to be due to inadequate brain or • Double vision*.
ocular blood supply as a result of arterial thrombosis or em-
bolism associated with disease of the arteries, heart or blood. Vestibular
• Symptoms last less than 24 hours. The 24 hour time A spinning sensation*.
limit for the duration of symptoms is purely arbitrary,
having more to do with the earth's rotation than biology; * In isolation these symptoms do not necessarily indicate
there is no qualitative difference between patients with transient focal cerebral ischemia.
TIA and mild ischemic stroke in terms of etiology and
prognosis, only a quantitative difference in terms of Non-focal neurologic symptoms
duration of symptoms. Non-focal neurologic symptoms are not neuroanatomically
localizing and are not due to TIAs unless accompanied by
EPIDEMIOLOGY additional focal neurologic symptoms.
• Incidence: about 50 per 100 000 per year (crude incidence). • Generalized weakness and/or sensory disturbance.
• Age: middle-aged and elderly; incidence increases with • Faintness and/or imbalance.
increasing age. • Altered consciousness or fainting, in isolation or with
• Gender: M=F (age <55 years), M>F (age 55–75 years), impaired vision in both eyes.
F>M (age >75 years). • Incontinence of urine or feces.
• Confusion or memory disturbance.
ETIOLOGY • A spinning sensation*.
A TIA is not a disease but a symptom of disease of the • Difficulty swallowing*.
arteries, heart or blood. • Slurred speech*.
• Double vision*.
Arterial disease (embolism/low flow) 75–80% • Loss of balance*.
• Extracranial large artery (aorta, carotid, vertebral)
atherothromboembolism 40–45%. * If these symptoms occur in combination, or with focal
• Intracranial large (e.g. middle cerebral, vertebrobasilar) neurologic symptoms, they may indicate transient focal
artery atheroma 5–10%. cerebral ischemia.
• Intracranial small (perforating) artery lipohyalinosis/
microatheroma 25% (‘lacunar’ TIA). Where is the TIA? (see Table 25)
• Non-atheromatous arterial disease (e.g. congenital,
arteritis, dissection) <5%. What is the cause of the TIA? (see Etiology [above]
and Stroke [p.192])
Cardiac disease (embolism) (see p.209) 20%
Table 25 Where is the TIA?
Hematologic disease (thrombo-embolism)
(see p.218) <5% Arterial Territory
Symptom Carotid Either Vertebro-
CLINICAL FEATURES basilar
Is it a TIA? Dysphasia +
• Sudden onset. Monocular visual loss +
• Loss of focal neurologic or monocular function. Unilateral weakness* +
• Symptoms maximal at onset: they do not spread or intensify. Unilateral sensory disturbance* +
• Symptoms resolve within 24 hours. Dysarthria** +
Homonymous hemianopia +
Focal neurologic symptoms and signs Unsteadiness/ataxia** +
Motor symptoms Dysphagia** +
• Weakness or clumsiness of one side of the body, in whole Diplopia** +
or in part. Vertigo** +
• Simultaneous bilateral weakness*. Bilateral simultaneous visual loss +
• Difficulty swallowing*. Bilateral simultaneous weakness +
Bilateral simultaneous sensory disturbance +
Speech/language disturbances Crossed sensory / motor loss +
• Difficulty understanding or expressing spoken language.
• Difficulty reading or writing. * Usually regarded as carotid distribution
• Slurred speech*. ** Not necessarily a TIA if an isolated symptom, only if associated
• Difficulty calculating. with >1 other symptom on the list
Transient Ischemic Attacks (TIA) of the Brain and Eye 187

DIFFERENTIAL DIAGNOSIS consciousness, loss of personal identity, focal neurologic


TIA of the brain symptoms or epileptic features.
• Migraine aura (with or without headache): younger patients, • Labyrinthine disorders: benign recurrent vertigo, benign
positive symptoms (visual scintillations, tingling), spread or paroxysmal positional vertigo, acute labyrinthitis, Ménière’s
march of symptoms over minutes. disease.
• Partial (focal) epileptic seizures: symptoms are positive • Metabolic disorders: hypoglycemia, hyperglycemia,
(jerking, tingling), march over seconds, stereotyped hypercalcemia, (hyponatremia).
recurrences, usually respond to anti-epileptic drugs. • Hyperventilation or panic attacks, somatization disorder.
• Transient global amnesia: abrupt onset of loss of • Intracranial structural lesion: meningioma, tumor, giant
anterograde episodic memory for verbal and non-verbal aneurysm, arteriovenous malformation, chronic subdural
material, usually accompanied by repetitive questioning, hematoma.
which resolves within 24 hours. There is no clouding of • Acute demyelination: multiple sclerosis.
• Syncope.
• Drop attacks.
195 Ocular fundus 195 • Mononeuropathy/radiculopathy.
showing golden-orange • Myasthenia gravis.
cholesterol crystals • Cataplexy.
(Hollenhorst plaques)
in the cilioretinal artery TIA of the eye
(arrows). (Reproduced Retina
from Warlow, Dennis, Vascular:
van Gijn, Hankey, • Low retinal artery perfusion :
Sandercock, Bamford, – Internal carotid artery atherothromboembolism
Wardlaw [2000] Stroke: or other arterial disorders (195–197).
A Practical Guide to – Embolism from the heart.
Management, 2nd edn. – Retinal migraine.
Blackwell Scientific, • High resistance to retinal perfusion:
Oxford, UK.) – Intracranial arteriovenous malformation.
– Central or branch retinal vein thrombosis.
– Raised intraocular pressure (glaucoma).
– Raised intracranial pressure (blowing the nose).
196 Ocular fundus 196 – Increased blood viscosity.
showing an embolus in – Retinal hemorrhage (198).
a peripheral branch of
the inferior temporal Non-vascular:
arteriole (arrow). • Paraneoplastic retinopathy.
• Phosphenes.
• Lightning streaks of Moore.
• Chorioretinitis.
197 Ocular fundus
photograph of a patient Optic nerve
with inferior temporal Vascular (anterior ischemic optic neuropathy):
branch retinal artery • Systemic hypotension.
occlusion showing pallor • Arteritis (e.g. giant cell).
of the inferior half of • Malignant arterial hypertension.
the retina due to cloudy
swelling of the retinal
ganglion cells caused by
retinal infarction.The 197 198
inferior temporal
branch arteriole is
attenuated and contains
embolic material.

198 Ocular fundus


showing a small retinal
hemorrhage in a patient
complaining of transient
monocular visual
disturbance.
188 Vascular Diseases of the Nervous System

Non-vascular: Magnetic resonance angiography (MRA) or intra-arterial


• Papilledema. digital subtraction angiography (IA-DSA) (202–207)
• Optic neuritis and Uhthoff’s phenomenon. Used if duplex ultrasound evidence of >70% stenosis of
• Dysplastic coloboma. symptomatic carotid artery (perhaps lower threshold to >50%
stenosis if the duplex is unreliable and a true 70% stenosis
Eye/orbit could be underestimated as 50% stenosis), and the patient is
• Vitreous hemorrhage. fit and willing for carotid surgery/stent (205–207).
• Reversible diabetic cataract. Dynamic CT angiography is also under evaluation (208).
• Lens subluxation. N.B. Practice varies with some surgeons performing carotid
• Orbital tumor (e.g. optic nerve sheath meningioma): endarterectomy on the result of ultrasound alone.
gaze-evoked loss of vision.
Transthoracic echocardiography (TTE)
INVESTIGATIONS Used if there is any abnormality of the heart clinically, on
All patients EKG or chest x-ray, followed by transesophageal
• Full blood count: polycythemia, anemia, thrombocy- echocardiography (TOE) if necessary.
tosis/cytopenia.
• ESR: arteritis, infective endocarditis, myxoma, infections.
• Plasma glucose: diabetes, hypoglycemia.
• Plasma cholesterol: hypercholesterolemia. 199
• EKG: atrial fibrillation, left ventricular hypertrophy, silent
MI.
• Urinalysis: diabetes, renal disease, infective endocarditis,
arteritis.

Selected patients
Depending on patient’s symptoms, age, general physical
condition and willingness to be investigated and treated.

CT brain scan
CT is used to exclude a non-vascular cause of symptoms of
TIA of the brain (e.g. meningioma, arteriovenous malforma-
tion) that may present like a TIA about 1% of the time
(199). Not all patients with TIA need a CT brain scan, such
as those with TIAs of the eye and perhaps single carotid
territory TIAs. Patients with posterior circulation TIA or
multiple carotid territory TIAs, particularly if stereotyped,
are more likely to have a symptomatic intracranial structural
lesion and should undergo CT brain scan.

Duplex ultrasound of the neck vessels (200, 201)


If a carotid territory TIA is present and the patient is fit and
willing for carotid artery surgery or stenting (should imaging 199 Cranial CT scan of a patient presenting with symptoms
reveal a severe carotid stenosis on symptomatic side). suggestive, but atypical of a transient ischemic attack which
were due to this right frontal arteriovenous malformation.

200 201

200 Color doppler ultrasound of a tight internal carotid artery 201 Color doppler ultrasound of an internal carotid artery occlusion.
stenosis. Note the black outline of the arterial wall (arrowheads) The outline of the internal carotid artery is visible but there is no color
with a thin color jet in the center indicating the stenosis (arrow). signal within it, whereas the external carotid is clearly patent.
Transient Ischemic Attacks (TIA) of the Brain and Eye 189

202 203 204

202, 203 Intra-arterial angiograms of fibromuscular 204 Angiogram of a patient with painful arm and
hyperplasia. Note the narrowed and beaded hand and intermittent blue fingers. It shows a cervical
appearance of the internal carotid artery (arrow). rib and subclavian artery compression (arrow).

205 206 207

205 Intra-arterial DSA of a tight internal carotid artery stenosis 208


(arrow).

206, 207 2D and 3D time of flight MRA of an internal carotid artery


stenosis (arrow). Note the ‘flow void’ at the point of stenosis which
may make it difficult to measure the exact degree of narrowing.

208 Spiral CT angiogram of an internal carotid artery stenosis


(arrow). Note the anatomic detail is good, but lots of calcification
(there is only a little in this example) may obscure the detail of the
lesion.
190 Vascular Diseases of the Nervous System

Transesophageal echocardiography (TOE) • Absolute benefit is greatest in those who have a high rate
Used for young patients (less than about 50 years) without a of vascular events, such as the elderly.
history of cardiac disease and with unexplained TIA (or • Risks of hemorrhage are low and far exceeded by the
ischemic stroke) to exclude left atrial myxoma or thrombus, benefits.
and ‘congenital’ cardiac defects such as patent foramen ovale • Aspirin is safe and effective in preventing subsequent
and atrial septal aneurysm. Also indicated in unexplained TIA vascular events by about 13–25%; low dose aspirin
(and ischemic stroke) to identify complex plaque of the (75–150 mg) is as effective as higher doses and causes less
ascending aorta and aortic arch. adverse effects such as dyspepsia and GI hemorrhage.
• Clopidogrel 75 mg/day is safe and marginally, but
Other tests significantly, more effective than aspirin (reduces vascular
• Urea and electrolytes: if hypertensive, on antihypertensive events by about 10% more than aspirin, from an average
agents, or diabetic. of about 6% to 5.4% per year) but is more costly. It is
• Hemoglobin A1C: if elevated fasting plasma glucose. associated with less gastrointestinal upset and bleeding
• VDRL/RPR and TPHA: if increased pre-test probability than aspirin in a dose of 325 mg per day, but has a slight
of meningovascular syphilis. excess (about 1%) of diarrhea and skin rash than aspirin.
• Thyroid function tests: if atrial fibrillation or • Ticlopidine is also slightly (about 10% [CI: 2–18%]) more
hypercholesterolemia. effective than aspirin but is more costly and has a small but
• Antinuclear antibody assay: if vasculitis suspected (e.g. important risk of thrombotic thrombocytopenic purpura
elevated ESR). and neutropenia. It is also associated with a higher rate of
• Autoimmune screen. diarrhea and skin rash than clopidogrel. Clopidogrel has
• Blood cultures (infective endocarditis). superseded ticlopidine in many countries because it is of
• HIV serology. similar efficacy (i.e. marginally but significantly better than
• Lipoprotein fractionation. aspirin by about 10%) but is safer and in most countries
• 24-hour EKG. (e.g. Malaysia is an exception) less costly.
• Transcranial doppler ultrasound (intracranial arterial • Aspirin in combination with dipyridamole may be more
stenosis). effective than aspirin alone, by about 15% (CI: 4–26%), but
• Alpha 1-antitrypsin (arterial dissection/aneurysm). further trials (e.g. European/Australasian Stroke Prevention
• Temporal artery biopsy and leptomeningeal/cortical in Reversible Ichaemia Trial [ESPRIT]) are in progress.
biopsy (arteritis). • Anticoagulation with warfarin/coumadin is probably no
• Thrombophilia screen (see p.220): if unexplained TIA, more effective and more hazardous than antiplatelet therapy
particularly if younger than 50 years of age, positive past in secondary prevention of vascular events among patients
or family history of premature arterial or venous occlusive with TIA due to atherothromboembolism. This comparison
disease, recurrent miscarriages, or an abnormality on the is also being studied in the ESPIRIT trial.
blood film such as thrombocytopenia. • Treatment should continue for at least 2 years after the
TIA, probably longer and probably for life if well-
DIAGNOSIS tolerated.
Based on the clinical history; there are usually no
confirmatory physical signs or investigations. There is Carotid endarterectomy
considerable inter-observer variation in the diagnosis of TIA, Carotid endarterectomy is effective in patients with recent
even among experienced neurologists, particularly if the carotid territory TIA or mild ischemic stroke, who have severe
patient has forgotten the symptoms, finds them difficult to carotid stenosis (>80% European Carotid Surgery Trial
describe, or was frightened by the attack and more [ECST] method, >70% NASCET method of measurement)
preoccupied with the immediate outcome. and who are fit and willing for surgery. It reduces the 3 year
risk of stroke from 26.5% (8.8% per year) to 14.9% (5.0% per
TREATMENT year), a relative risk reduction of 44% (95% CI: 21–60%), and
Treat the underlying cause. absolute risk reduction of 11.6% (3.8% per year). The number
needed to treat (NNT) to prevent one stroke at 3 years is 9,
Giant cell arteritis (see p.234) and to prevent one stroke each year is 26. Only up to about
Steroids. 16% of patients with recent carotid ischemia and severe
carotid stenosis on the symptomatic side benefit from carotid
Infective endocarditis (see p.215) endarterectomy. These are probably patients who score four
Antibiotics. or more in the prognostic model below (see Table 26).
The net effectiveness of carotid endarterectomy is also
Atherothromboembolism (see p.202) determined by the surgical perioperative stroke and death
Vascular risk factor control rate. Referring doctors (and patients) should have access to
• High blood pressure. the perioperative stroke and death rate of their prospective
• Cigarette smoking. surgeon(s), derived from independent and rigorous audit.
• High serum cholesterol. However, interpretation of unusually high or low operative
• Diabetes mellitus. risks must take into account the effects of chance and case-
mix. Otherwise, over-simplistic interpretation of crude results
Antiplatelet therapy may lead to unjustified criticism of individual surgeons, and
• Reduces the risk of stroke, MI or vascular death by about not to improvements in patient care. The decision to operate
a quarter, irrespective of age, sex, and blood pressure; from should consider all the important prognostic factors for stroke
an average of about 8% to 6% per year. and perioperative stroke and death.
Transient Ischemic Attacks (TIA) of the Brain and Eye 191

carotid angioplasty/stenting was stopped after only 17


Table 26 Prediction model for benefit from carotid patients had been randomized because of five peroperative
endarterectomy among patients with recent carotid strokes among the seven patients treated with carotid stenting
ischemia and severe symptomatic carotid stenosis (compared with no strokes among the 10 patients treated
Independent risk factors for stroke with best medical therapy with carotid endarterectomy). The only other completed trial
Ischemic events of the brain (as opposed to the eye) 1 point has been the Carotid And Vertebral Artery Transluminal
Recent ischemic events (in the past 2 months) 1 point Angioplasty Study (CAVATAS) which randomized 504
Severe degree of carotid stenosis: patients with carotid artery stenosis (97% symptomatic) to
80–89% stenosis 1 point carotid endarterectomy (253 patients) or carotid angioplasty
90–99% stenosis 2 points (251 patients; stents were used in 26%). Safety data indicate
Carotid plaque irregularity 1 point that the perioperative (within 30 days) rate of all stroke or
death was 9.9% among patients allocated carotid endarterec-
Independent risk factors for perioperative risk of stroke and death tomy and 10.0% among patients allocated carotid angioplasty.
• Female gender -0.5 points The perioperative (within 30 days) rate of disabling stroke or
• Systolic BP>24 kPa (180 mmHg) -0.5 points death was 5.9% among patients allocated carotid endarterec-
• Peripheral vascular disease -0.5 points tomy and 6.4% among patients allocated carotid angioplasty.
There was a significant excess of cranial nerve palsy
amongst patients allocated carotid endarterectomy (8.7%)
compared with carotid angioplasty (0%, p<0.001) and a
significant excess of wound hematoma amongst patients
Percutaneous transluminal carotid and vertebral allocated carotid endarterectomy (6.7%) compared with
angioplasty/stenting carotid angioplasty (1.2%, p<0.0015). Efficacy data, to date,
Carotid angioplasty with stenting is a promising alternative show that carotid restenosis is more common after carotid
to carotid endarterectomy that is still being evaluated in angioplasty but is rarely symptomatic, within the first few
clinical trials. It is however also being practised increasingly, years. Most importantly, there is no difference in rate of stroke
in the absence of robust evidence of safety and long term during the first 3 years after carotid angioplasty or end-
efficacy, because it is thought to be associated with fewer arterectomy, but further follow-up is required and is ongoing.
perioperative complications than carotid endarterectomy and
equal efficacy in preventing stroke (209–211). However, true Extracranial to intracranial bypass surgery
modest differences in the outcome of patients treated by No longer practised outside research settings following con-
carotid angioplasty/stenting and carotid endarterectomy will clusive evidence from a large randomized trial that the
only be detected reliably by large randomized trials. balance of surgical risk and eventual benefit did not favor
One trial in a single center without previous experience in surgery.

209 210 211

209–211 Selective carotid angiogram, subtraction views, showing a tight stenosis of the origin of the internal carotid artery (209, arrow), which
has been dilated by means of balloon catheter angioplasty (210) and dilated further with the deployment of a stent (211).
192 Vascular Diseases of the Nervous System

Cardiogenic embolism (see p.209) STROKE


Anticoagulation: warfarin (target INR 2.0–3.0), if major
cardiac source of embolism:
• Atrial fibrillation (non-rheumatic): reduces annual stroke DEFINITION
risk by two-thirds from about 12% to 4%. A generic term describing a clinical syndrome characterized
• Recent (<3 months) myocardial infarction: treat for at by the sudden onset of a focal neurologic deficit (or coma)
least 3 months, and continue longer if chronic AF or due to infarction of, or hemorrhage into or over, a part of
other risk factor: large anterior MI or apex involvement; the brain. The deficit persists for longer than 24 hours or
mural thrombus; high risk (AF, heart failure, previous leads to earlier death.
embolic event).
• Dilated cardiomyopathy. EPIDEMIOLOGY
• Cardiac failure. • Incidence: 2 per 1000 per year.
• Valvular heart disease: mitral stenosis or regurgitation; • Age: any age but the incidence of stroke increases steeply
mechanical prosthetic aortic or mitral valve. with age.
• Complicated patent foramen ovale not closed surgically • Gender: M>F in middle age, F>M in old age.
(associated with atrial septal aneurysm or mitral valve • Burden: stroke is the third most common cause of death
prolapse) – anticoagulation is controversial/unproven. (10–12% of all deaths) in developed countries and a
major cause of adult physical disability.
PROGNOSIS
As a group, TIA patients have an increased risk of stroke and PATHOGENESIS
other serious vascular events of about 8–10% per year. The Cerebral infarction (75–80%)
risk of stroke is about 4–5% in the first month, 12% in the • Large artery atherothromboembolism (50%):
first year, 29% over 5 years. The risk of a coronary event is – Extracranial (aorta, carotid, vertebral arteries) (40–45%).
about 3% per year. – Intracranial (ICA, MCA, ACA, Vertebral, Basilar, PCA)
As individuals, TIA patients have a variable prognosis. (5–10%).
Independent predictors of an increased risk of stroke and • Small artery microatheroma/lipohyalinosis (lacunar
other cardiovascular events are: syndromes) (25%).
• TIAs of the brain (compared with TIAs of the eye). • Embolism from the heart (20%).
• Increasing age. • Blood disease (thrombophilia) (<5%).
• Increasing number of TIAs in the previous 3 months (i.e. • Non-atheromatous arterial disease (e.g. dissection,
multiple attacks). arteritis) (<5%).
• Peripheral vascular disease.
• Carotid stenosis >80%. Intracerebral hemorrhage (10–15%) (212)
• Hypertensive lipohyalinosis and microaneurysms (40%).
Other adverse prognostic factors may include: past history • Bleeding diatheses (e.g. antithrombotic drugs, thrombo-
of any TIA, left ventricular hypertrophy, carotid plaque cytopenia) (10%).
morphology (e.g. ulceration), impaired cerebrovascular reserve. • Arteriovenous malformation (10%).
• Amyloid angiopathy (10%).
• Hemorrhagic transformation of cerebral infarct (10%).
212 • Aneurysm (8%).
• Intracerebral tumor (5%).
• Arteritis (<5%).
• Drugs: sympathomimetics (e.g. amphetamines, cocaine)
(<5%).
• Arterial dissection (<5%).
• Intracranial venous thrombosis (<5%).

Subarachnoid hemorrhage (5%)


• Aneurysm.
• Arteriovenous malformation.

Unknown (5%)

Vascular risk factors and diseases in first-ever


ischemic stroke*
• Hypertension (BP >21.3/12 kPa [160/90 mmHg] on
two occasions pre-stroke): 52%.
• Angina and/or past myocardial infarction: 38%.

212 Axial slice of the brain at autopsy showing bilateral basal ganglia
hemorrhage with rupture into the ventricles in a patient with severe
hypertension.
Stroke 193

• Current smoker: 27%. – Self-audible bruits: internal carotid artery stenosis


• Claudication and/or absent foot pulses: 25%. (distal); dural arteriovenous fistula; glomus tumor;
• Major cardiac embolic source: 20%. carotico-cavernous fistula.
• Transient ischemic attack (TIA): 14%. • Medications/drugs:
• Cervical arterial bruit: 14%. – Blood pressure-lowering/vasodilators.
• Diabetes mellitus: 10%. – Hypoglycemic drugs.
• Any of the above: 80%. – Cocaine, amphetamines, ephedrine, phenylpropano-
lamine, ecstasy (methylenedioxyamphetamine).
* Sandercock PAG, Warlow CP, Jones LN, Starkey IR – Oral contraceptives; estrogens in men.
(1989) Predisposing factors for cerebral infarction: The – Allopurinol.
Oxfordshire Community Stroke Project. BMJ, 298: 75–80. – Interleukin 2.
– Deoxycoformycin (pentostatin).
CLINICAL FEATURES – L-asparaginase (asparaginase).
Is it a stroke? • Past medical history:
• Sudden onset. – Previous stroke or TIA.
• Loss of focal neurologic function (see TIA, p.186). – Vascular risk factors (see above).
• Symptoms and signs are maximal within a few seconds or – Recent myocardial infarction.
minutes, but sometimes evolve over a few hours or – Rheumatic fever.
progress in a stepwise fashion due to propagation of – Inflammatory bowel disease.
thrombus, recurrent embolization, or hemorrhagic – Celiac disease.
transformation of the infarct. Progression over more than – Irradiation of the head or neck.
a few hours or failure to improve after a few days, suggests – Recurrent deep venous thrombosis.
complications (e.g. brain edema, hydrocephalus) and an – Recurrent miscarriages.
alternative diagnosis (e.g. tumor). • Family history: (gene/chromosome location, if known)
• Symptoms persist beyond 24 hours (by arbitrary definition). – Connective tissue disorders:
Ehlers–Danlos syndrome type IV: (collagen 3A1).
Where is the stroke? (see Neurovascular Pseudoxanthoma elasticum.
syndromes, p.181) Marfan’s syndrome: (fibrillin).
Fibromuscular dysplasia.
What is the cause of the stroke? Familial mitral valve prolapse.
Infarction or hemorrhage? – Hematologic disorders:
There are no clinical features that reliably distinguish Sickle cell (SC) disease/trait.
intracerebral hemorrhage from infarction. Headache, coma Antithrombin III deficiency.
at onset, vomiting and rapid deterioration are more Protein C deficiency.
common with hemorrhage but may also occur with Protein S deficiency.
infarction. The sudden onset of severe headache associated Plasminogen abnormality/deficiency.
with neck stiffness or coma distinguishes subarachnoid Dysfibrinogenemia.
hemorrhage (SAH) from other causes (see SAH, p.249). Hemophilia and other inherited coagulation factor
deficiencies.
Clues to the cause of the ischemic and hemorrhagic – Others:
stroke, or alternative diagnosis Familial hypercholesterolemia.
Clinical history: Neurofibromatosis.
• Age, sex and race. Homocystinuria.
• Presenting complaint: Fabry’s disease.
– Onset, if gradual: consider low flow (hemodynamic) Hereditary cerebral amyloid angiopathy: Dutch type,
ischemic stroke; migraine; structural intracranial lesion; (amyloid precursor protein).
multiple sclerosis. Hereditary cerebral amyloid angiopathy: Icelandic type,
– Precipitating factors: standing up quickly, heavy meal, hot (cystatin C).
weather, hot bath, warming the face, exercise, valsalva Migraine.
maneuver, chest symptoms such as pain or palpitations, Familial cardiac myxoma.
starting or changing blood pressure-lowering drugs, head Familial cardiomyopathies.
turning – all suggest low flow; hypoglycemia. Mitochondrial cytopathy: (mitochondrial tRNA-leu).
– Injury to head or neck: vertebral/carotid artery dissec- CADASIL (cerebral autosomal dominant arteriopathy
tion, chronic subdural hematoma. with subcortical infarcts and leukoencephalopathy):
• Associated symptoms: (notch3 mutations, 19q12).
– Recent headache: carotid/vertebral dissection; giant cell Sneddon’s syndrome.
arteritis (or other vasculitis); migrainous stroke/TIA; Arteriovenous malformations.
intracranial venous thrombosis; structural intracranial lesion. Cerebral cavernous malformation: (7q22; genetic
– Epileptic seizures: cortical infarction or hemorrhage; heterogeneity).
intracranial venous thrombosis; mitochondrial cytopathy; Intracranial saccular aneurysms.
non-vascular intracranial lesion. Polycystic kidney disease: (polycystin).
– Malaise: inflammatory arterial disorders; infective Hereditary hemorrhagic telangiectasia-1: (endoglin).
endocarditis; cardiac myxoma; cancer; thrombotic Hereditary hemorrhagic telangiectasia-2: (activin
thrombocytopenic purpura; sarcoidosis. receptor-like kinase 1).
194 Vascular Diseases of the Nervous System

Examination: DIFFERENTIAL DIAGNOSIS OF STROKE


• Body weight. (IN ORDER OF FREQUENCY OF OCCURRENCE
• Pulses: IN GENERAL PRACTICE)
– Present or absent (peripheral vascular disease). • Metabolic/toxic encephalopathy (hypoglycemia, non-
– Tenderness (giant cell arteritis). ketotic hyperglycemia, hyponatremia, Wernicke–Korsa-
– Rhythm (atrial fibrillation). koff syndrome, hepatic encephalopathy, alcohol and drug
• Blood pressure: intoxication).
– Supine and erect (hypertension, postural hypotension). • Functional/non-neurologic (e.g. hysteria).
– Both arms (subclavian steal or aortic dissection). • Epileptic seizure (postictal Todd’s paresis).
• Neck bruits*. • Structural intracranial lesions (subdural hematoma,
• Neck stiffness. tumor, arteriovenous malformation).
• Heart: murmurs (valvular heart disease). • Encephalitis (e.g. herpes simplex virus)/brain abscess.
• Eye: • Head injury.
– Conjunctival injection and rubeosis iridis (ischemic • Peripheral nerve lesion(s).
oculopathy) (8, 9). • Hypertensive encephalopathy.
– Horner’s syndrome (carotid dissection). • Multiple sclerosis.
• Ocular fundi: retinal emboli; hypertensive, diabetic or • Creutzfeldt–Jakob disease.
ischemic retinopathy.
• Neurologic examination: impairments (see Neurovascular INVESTIGATIONS
syndromes, p.181), disabilities and handicaps. Observe All patients
the patient attempting functional tasks such as those Immediate CT brain scan(213–217)
required for everyday activities of daily living. These are • To exclude non-stroke pathology (213–215):
vital in assessing and monitoring the patient’s prognosis – Unclear history of sudden onset of focal neurologic
and structuring an appropriate rehabilitation program. symptoms (i.e. because of coma, dysphasia, confusion,
no witness);
*N.B. Listening for carotid bruits is not particularly helpful – Atypical clinical features (i.e. gradual onset, seizures, no
as their absence does not exclude tight carotid stenosis and clear focal neurologic signs); young patient (age
their presence does not mean that the patient has tight <50 years) with no vascular risk factors.
common or internal carotid artery stenosis; a carotid bruit • To distinguish intracranial hemorrhage (216) from
can be a normal finding in young adults; it can be due to cerebral infarction (CT must be done within about
external carotid stenosis (which is irrelevant to the patient's 10 days of stroke).
symptoms); it can be transmitted from the heart and great
vessels in patients with aortic or mitral valve disease, a patent Intracranial hemorrhage is seen immediately as a
ductus arteriosus or coarctation of the aorta; and it can be homogeneous area of high density; infarction, if seen, as an
part of a diffuse neck bruit in patients with thyrotoxicosis area of low density. Within the first few hours of infarction,
or a hyperdynamic circulation associated with pregnancy, there is loss of gray/white matter differentiation, slight
anemia, fever and hemodialysis. hypodensity and a little swelling (loss of visibility of the
sulci). The lesion is wedge-shaped if cortical and rounded if
Skin signs of underlying causes of TIA or stroke subcortical. The occluded artery may appear hyperdense
• Finger clubbing(14): cancer; right to left intracardiac (whiter) due to the presence of thrombus (this sign is
shunt; pulmonary arterial venous malformation; infective reliable if present, but its absence means little as far as
endocarditis; inflammatory bowel disease. whether or not the artery is patent).
• Splinter hemorrhages: infective endocarditis; cholesterol Within the first few days the infarct margins become clearly
embolization syndrome; vasculitis. demarcated, the infarct more hypodense (darker), and the
• Scleroderma: systemic sclerosis. swelling increases (large infarcts may produce considerable
• Livedo reticularis: Sneddon’s syndrome; systemic lupus mass effect with midline shift and uncal herniation). After the
erythematosus; polyarteritis nodosa; cholesterol emboliz- first week, the swelling subsides and the infarct becomes of
ation syndrome. similar density to normal brain making it difficult to see
• Lax skin: Ehlers–Danlos syndrome; pseudoxanthoma (fogging). Fogging lasts for a week or so. After several
elasticum. months, the infarct is a shrunken black area (‘hole’).
• Skin color: anemia; polycythemia; cyanosis (right to left Up to 50% of all infarcts will not be positively identified
intracardiac or pulmonary shunt). by CT; larger and older infarcts are more likely to be visible
• Porcelain white papules/scars: Kohlmeier–Degos’ disease. (but see above, fogging). Hemorrhagic transformation of
• Skin scars: Ehlers–Danlos syndrome. the infarct (HTI) can occur any time within the first few
• Petechiae/purpura/bruising: thrombotic thrombocyto- weeks, but is most frequent within the first week and in
penic purpura; fat embolism; cholesterol embolization patients with large infarcts, high blood pressure, or who are
syndrome; Ehlers–Danlos syndrome. given antithrombotic and thrombolytic drugs. HTI may be
• Oro-genital ulceration: Behçet’s disease. massive or minor (petechial); massive HTI may be
• Rash: Fabry’s disease. indistinguishable from a de novo primary intracerebral
• Epidermal nevi: epidermal nevus syndrome. hemorrhage (PICH) (217–219).
• Café-au-lait patches: neurofibromatosis.
• Thrombosed veins, needle marks: i.v. drug abuse. • To ascertain the likely cause of ischemic or hemorrhagic
stroke.
• Otherwise as for TIA patients (see p.186).
Stroke 195

213 214 215

213 Plain cranial CT scan showing low density in the cortex and subcortex of the left frontal 216
lobe (arrows) and also in the periventricular regions of a patient who presented with sudden
onset of dysphasia and right hemiparesis and was diagnosed as having suffered an ischemic
stroke (but see 214, 215).

214, 215 Post contrast CT scan in the same patient as 213 showing ring enhancement of the
left frontal cortical/subcortical lesion (arrow) (214) and another ring enhancing lesion in the left
anterior temporal lobe (arrow) due to metastatic carcinoma (215).

216 Non-contrast cranial CT scan showing a focal area of high density in the frontal lobe on
the right due to hemorrhage secondary to amyloid angiopathy.

217, 218 Hemorrhagic transformation of an arterial infarct. CT scan (217) within 4 hours of
the stroke shows a high density (white) area in the left temporal lobe. Cerebral angiogram (AP
view) (218) 1 hour later shows occlusion of the left middle cerebral artery (arrow) and dilated
lenticulostriate arteries.These images are typical of early hemorrhagic transformation of an
arterial infarct, which can be difficult to differentiate from primary intracerebral hemorrhage.

219 Axial section of the brain at post mortem showing a large hemorrhagic cortical/subcortical
infarct in the right frontal lobe due to embolic occlusion of the right anterior cerebral artery and
pre-rolandic branch of the upper division of the right middle cerebral artery by an embolus
from the origin of the right internal carotid artery.

217 218 219


196 Vascular Diseases of the Nervous System

Selected patients dependency at 1 year after stroke from about 62.0% to


As for TIA patients (see p.188). 56.4%. This is an odds reduction of about 29% (odds ratio
(OR) 0.71), a relative risk reduction (RRR) of 9% (95% CI:
MRI brain scan 4% –14%) and absolute risk reduction (ARR) of 5.6%.
With or without diffusion and perfusion weighted imaging, if: Therefore, treating 1000 patients in a stroke unit prevents
• A CT scan performed >10 days after stroke shows a low- about 56 patients from dying or becoming dependent
density area that could be infarction or resolving compared to treating 1000 in a general ward. The number
hemorrhage and it is essential to distinguish them. of patients needed to treat (NNT) in a stroke unit to
• CT scan is negative and it is crucial to be able to localize prevent one from death or dependency is about 18.
the infarct. MRI is more sensitive in detecting infarcts Although preferable, a geographically dedicated stroke unit
than CT (particularly brainstem and small subcortical is not necessary. There are insufficient data on the cost of
infarcts), but can be difficult to use in very sick, confused stroke unit care but it does not increase length of stay.
patients and may show so many ‘holes’ in some patients Patients with TIA and mild non-disabling stroke do not
that it is just confusing. The evolution of the appearance need admission if facilities are available for rapid, appropriate
of infarction is similar to CT (including fogging). A investigation and follow-up as an outpatient.
recent infarct is bright on T2W imaging (T2WI) and
dark on T1WI. An occluded artery shows up as an absent General
‘flow void’, sometimes with a bright blob in it on T2WI Assess, manage and monitor:
(the clot). Special sequences can show very early changes • Airway: care must be taken to maintain a patent airway
of infarction (within minutes of the interruption of blood in patients with depressed consciousness or bulbar
flow) such as diffusion- or perfusion-weighted imaging dysfunction.
or spectroscopic imaging, but these are still being • Oxygen therapy: if the patient is hypoxic (oxygen
evaluated. saturation <95%); of no benefit if hemoglobin is fully
• Arterial dissection suspected (N.B. catheter angiography saturated.
remains the gold standard). • Vital systemic signs: temperature, heart rate, blood
• Trying to identify the response to thrombolytic therapy pressure, respiration, fluid input and output. Maintaining
(e.g. perhaps the greatest benefit is obtained with a large fluid balance and preventing dehydration is essential.
diffusion/perfusion mismatch). • Vital neurologic signs: Glasgow coma scale, pupils, body
postures.
CSF examination • Fever: should be investigated (MSU, CXR, blood
Useful if SAH is still suspected despite a normal CT scan cultures, sputum culture and so on) and treated
(must wait until at least 12 hours have elapsed since the appropriately; elevated temperature is an adverse
onset of stroke symptoms before doing the CSF prognostic factor.
examination) or if positive blood VDRL or TPHA. • Blood pressure (BP) rises transiently after stroke, especially
hemorrhagic stroke, and falls during the first 7 days.
DIAGNOSIS
A clinical diagnosis, based on the sudden onset of a focal Current empirical policy is not to initiate new
neurologic deficit, which is correct about 95% of the time. antihypertensive therapy for the first 7 days after stroke
Factors that may reduce the reliability of the clinical because a reduction in blood pressure may substantially
diagnosis of stroke include: lower cerebral blood flow in the ischemic penumbra
• Unclear history: impaired conscious level; dysphasia; surrounding an infarct (because of loss of autoregulation in
dementia. ischemic brain) and lead to further ischemic brain damage,
• Unusual symptoms: persistent headache; personality particularly if there is a tight stenosis of the feeding artery.
change. Blood pressure should only be lowered, and cautiously at
• Unusual signs: fever; papilledema. that (by about 15% over 24 hours), in the presence of
• Young age: less likely to have degenerative vascular hypertensive crises such as hypertensive encephalopathy, left
disease. ventricular failure, aortic dissection, or intracerebral
• Progressing neurologic signs. bleeding, or if the blood pressure is very high and poses a
real risk of cerebral hemorrhage. We don’t know what level
The diagnosis of stroke can sometimes be confirmed by of blood pressure is ‘too high’ but treatment is recom-
CT or MRI brain scan, or at autopsy. The CT scan can be mended if: systolic BP >26.7–29.3 kPa (200–220 mmHg)
normal in stroke, provided other causes of an apparent focal (ischemic stroke) or >24–26.7 kPa (180–200 mmHg)
neurologic deficit, such as multiple sclerosis and psychogenic (hemorrhagic stroke); or diastolic BP >16–17.3 kPa
behavior, have been excluded. (120–130 mmHg) (ischemic stroke) or >13.3–14.7 kPa
(100–110 mmHg) (hemorrhagic stroke).
TREATMENT Oral antihypertensive agents are recommended, such as
a diuretic, beta-blocker, ACE inhibitor (low dose) or
Organized and coordinated multidisciplinary calcium-channel blocker; nifedipine capsules, and parenteral
care (Stroke Unit) medications should be avoided because of the rapid and
Compared with conventional care in a general ward, sometimes precipitous fall in blood pressure.
organized and coordinated care by an interested and trained
multidisciplinary team in a stroke unit reduces death and
Stroke 197

• Low blood pressure may reflect hypovolemia due to Specific


excessive losses by sweating and insufficient intake by Acute ischemic stroke
mouth or tube, and should be corrected by raising the • Aspirin 160–300 mg daily for acute ischemic stroke
foot of the bed and by fluid replacement via a safe route. reduces death and dependency from 47.0% to 45.8%, a
• Swallowing function: maintain fluid and electrolyte RRR of 3% (95% CI: 1–5%) and an ARR of 1.2%.
balance and nutrition initially with i.v. saline and/or Therefore, treating 1000 patients prevents about 12
nasogastric tube until swallowing function is assessed by dying or becoming dependent. The NNT is 83. All
a trained nurse or speech pathologist and dietary patients with acute ischemic stroke should be given
requirements by a dietician. Percutaneous endoscopic aspirin immediately (unless allergic/intolerant of aspirin
gastrostomy tube is preferred for patients who require or entering a trial of thrombolysis).
feeding by tube for more than about 2 weeks. • Intravenous thrombolysis with streptokinase or tissue
• Bladder function: after attempted voiding, residual plasminogen activator (tPA) within 6 hours of ischemic
bladder volume of urine is estimated by a nurse trained stroke reduces death and dependency from 59.6% to
in the use of bladder ultrasound. If the residual urine 55.2%, an odds reduction of 17% (95% CI: 6–27%), a
volume is more than 100 ml, the bladder should be RRR of 9% (95% CI: 4–14%) and ARR of 4.4%. Treating
emptied by catheter every 6–8 hours. 1000 patients prevents 44 from death or dependency.
• Immobile patients: should be turned frequently (e.g. The NNT is 23. For patients presenting within 3 hours,
every 2 hours) to prevent pressure sores. Proper i.v. tPA reduces death and dependency from 67.8% to
positioning and early mobilization by nurses and 55.2%, an odds reduction of 42% (95% CI: 26–54%) and
physiotherapists help to prevent complications of ARR of 12.6%. Treating 1000 patients prevents 126
immobility such as joint contractures, pneumonia and from death or dependency. The NNT is 8. However,
painful ‘frozen’ shoulder. Deep venous thrombosis and thrombolysis is also hazardous. The risk of early death
pulmonary embolism can be prevented by regular passive (within 14 days of stroke) is increased from 15.4%
and active joint movement, early mobilization, graduated (control) to 19.0% (thrombolysis), a 3.6% absolute excess
compression support stockings, aspirin and subcutaneous (OR: 1.3, 95% CI: 1.13–1.5). This is because of an
heparin. excess of early symptomatic intracranial hemorrhage
• Blood glucose should be maintained within normal (2.5% [control] vs. 9.4% [thrombolysis]; OR: 3.5, 95%
limits; elevated blood glucose is an adverse prognostic CI: 2.8–4.4), and fatal intracranial hemorrhage (1.0%
factor and may be harmful. [control] vs. 5.4% [thrombolysis]; OR: 4.15, 95% CI:
• Depression should be treated with psychosocial support 3.0–5.8) (see Table 27, p.199).
and antidepressants if necessary. Early socialization
may help. Thrombolytic therapy for acute ischemic stroke is
• Brain edema: cytotoxic edema, which does not cause therefore very much like carotid endarterectomy for
much brain swelling (i.e. no net change in brain volume, symptomatic severe carotid stenosis (see TIA, p.186); both
just a shift of fluid from extracellular to intracellular are associated with an early hazard yet a greater longer term
space) is followed by vasogenic edema (which does cause net benefit. The burning question is no longer whether
brain swelling). Specific treatments for brain edema (see thrombolysis (and carotid endarterectomy) is effective, but
below) do not reduce the edema but shrink the in whom it is effective, in whom it is ineffective, and in
surrounding normal brain. The onset, degree and whom it is dangerous. At present it is not known exactly
duration of the effect of each agent on intracranial which combination of clinical and imaging features reliably
pressure (ICP) is quite variable. identify patients who will benefit or be harmed by
– General: restrict fluids, elevate head of bed, treat fever; thrombolysis. Nor is it clear what is the optimal thrombo-
avoid hypoxia and hypercapnia; avoid hypo-osmolar fluids. lytic agent, dose, half-life, and route of administration.
– Specific: osmotic diuretics, providing BBB intact. A bolus Furthermore, the most effective concomitant neuro-
of mannitol (1g/kg i.v. as a 20% solution over 1 hour), protective, antithrombotic and antihypertensive regime, if
usually decreases ICP by about 50% after about 90 any, remains to be established. What it known from current
minutes but returns to the pre-treatment levels after data is that the clinical features of the patient and the
about 210 minutes. findings of early brain imaging have considerable potential
– Steroids: should be effective for vasogenic edema, but for refining patient selection for thrombolysis.
have not been shown to be effective in acute stroke. Plain CT brain scan signs of major infarction which
– Hyperventilation if patient is deteriorating: quick onset involves more than one-third of the middle cerebral artery
of effect on ICP (within 2–30 minutes) but it lasts only territory predicts an increased risk of symptomatic
for about 30 minutes. intracranial hemorrhage after thrombolysis. However, it is
– Barbiturates: useful because they reduce ICP more than neither sensitive nor specific, and the interrater reliability
they reduce BP, so cerebral perfusion pressure increases. among physicians involved in acute stroke care is less than
– CSF drainage, e.g. ventricular drain. ideal. A new CT scoring system (ASPECTS) is reported to
– Surgical decompression (hemicraniectomy): when the be a simple and reliable method of quantifying early
ICP is rising despite the above measures, and before ischemic changes on CT scan, like an ‘ECG of the brain’,
signs of herniation (ipsilateral pupillary loss of reactivity and which accurately predicts risk of symptomatic
followed by dilatation, periodic breathing [even in intracranial hemorrhage and functional outcome in ischemic
conscious patient], contralateral Babinski sign). stroke patients treated with intravenous thrombolytic
therapy (Barber et al., 2000). However, its external validity
and reliability remains to be established.
198 Vascular Diseases of the Nervous System

Magnetic resonance (MR) imaging can provide a Primary intracerebral hemorrhage (PICH) (see p.238)
perfusion-weighted image (PWI) which reveals the region Surgery for PICH is associated with a non-significant increase
of brain which is underperfused and ‘at risk’, and a in odds of death and dependency at 6 months (OR: 1.23, 95%
diffusion-weighted image (DWI) which identifies the core CI: 0.77–1.98). Further evidence from ongoing trials is awaited.
of early infarction. A mismatch between the acute PWI
lesion and the (smaller) DWI lesion may represent the Subarachnoid hemorrhage (see p.249)
ischemic penumbra of potentially salvageable brain. Clinical Early aneurysm surgery is now usual practice for patients in
trials are evaluating whether patients with a PWI/DWI good clinical condition but this has not been supported by a
mismatch are likely to benefit clinically from early randomized controlled trial. Antifibrinolytic drugs prevent re-
reperfusion. If confirmed, a further challenge will be to bleeding but increase the risk of cerebral ischemia and have no
optimize the availability and feasibility of undertaking MRI net effect on overall outcome. Oral nimodipine helps to prevent
brain scans within the first few hours of stroke onset in sick, delayed cerebral ischemia and significantly reduces the risk of a
dysphasic, disorientated and claustrophobic patients with poor outcome by about 31% (RR: 0.69, 95% CI: 0.58–0.84).
acute stroke. Finally, patient selection for thrombolysis may
also be improved by proof of arterial occlusion by means of Minimizing complications
non-invasive imaging (e.g. magnetic resonance angiography, Complications after stroke (see Table 29, p.200) are common
transcranial doppler). yet preventable. The key is to anticipate them in high risk
Clearly, further studies are required, and are in progress patients, implement appropriate prevention strategies, and
(e.g. International Stroke Trial – 3) to refine patient regularly (at least daily) assess patients. Patients at particular
selection, and establish the balance of risks and benefits of risk are the elderly, those with pre-existing handicap or
thrombolysis in a broader range of patients presenting at diabetes, a total anterior circulation syndrome (TACS),
different stages, with differing types and severities of stroke, urinary incontinence and hospitalized for more than 30 days.
different risk factors, and different brain imaging findings.
Whilst awaiting the results of these studies, rt-PA has been Rehabilitation
licenced for use in the United States (1997), Canada (1999) Rehabilitation begins on day 1 as an integral part of acute
and Germany (August 2000) for patients presenting within treatment and aims to maximize survival free of handicap
3 hours of acute ischemic stroke who are similar to the by facilitating normal recovery and minimizing compli-
patients included in the trials and provided there is a stroke cations. It involves an organized, coordinated multi-
service which can ensure its safe administration. disciplinary team undertaking regular patient assessment,
rt-PA remains to be licenced for use in other parts of short and long term goal setting with the patient and
Europe and America, and in Australasia and Asia. One family/carer (including discharge planning), intervention,
lesson learnt from the North American experience is that re-assessment, and re-intervention. Failure to achieve pre-
treating patients who violate the guidelines outlined in Table specified goals usually reflects inaccurate assessment,
28 is associated with excess risk and poor patient outcome. unrealistic expectations, progression of comorbid conditions
If rt-PA is to be licenced in other countries, it is essential (e.g. angina, arthritis), or the intercurrence of a
that its use is restricted, at least initially, to dedicated stroke complication (see Table 29, p.200) or another stroke.
units which are appopriately equipped, prepared to respond
to demand and adhere strictly to current guidelines (see
Table 28), and prepared to undertake prospective, systematic
and rigorous audit as part of a national register (if not as
part of an approved randomized trial). This will ensure that
stroke patients have access to this effective (yet risky)
treatment, that stroke physicians gain experience and
expertise in its use, and that quality control and patient
selection continues to be optimized by correlation of
baseline demographic, clinical, imaging and treatment data
with early and long term patient outcome.
• Heparin and heparinoid in acute ischemic stroke have no
net effect on death or dependency (RRR: 1%, 95% CI:
–5 to 6% or (–5 – +6), despite significantly reducing the
odds of deep vein thrombosis by about 79% (OR: 0.21,
95% CI: 0.15–0.39) and pulmonary embolism by about
39% (OR: 0.61, 95% CI: 0.45–0.83).
• Trials of neuroprotective agents (e.g. selfotel, aptiganel,
chlormethiazole, tirilazad, lubeluzole) have failed to
identify a favorable treatment effect, and some have
revealed dose-limiting intolerance and systemic adverse
effects, such as excessive sedation and hypertension.
Trials of other agents (e.g. magnesium) are in progress.
• Other medical treatments such as hemodilution, corti-
costeroids, and glycerol (glycerin) have not been proven
effective.
Stroke 199

Table 27 Risks and benefits of thrombolytic therapy for acute ischemic stroke
Outcome events Thrombolysis (%) Control (%) OR 95% CI of OR Absolute risk per 1000
patients treated (95% CI)
Within 6 hours
Early (<10 days)
• Death 16.6% 9.8% 1.85 1.48–2.32 +68 (44–93)
• Fatal
Intracranial 5.4% 1.0% 4.15 2.96–5.84 +44
hemorrhage
• Symptomatic
Intracranial 9.4% 2.5% 3.53 2.79–4.45 +70 (58–83)
hemorrhage
At final follow-up (3–6 months)*
• Death 19.0% 15.4% 1.31 1.13–1.52 +36 (17–56)
• Death or
dependency 55.2% 59.6% 0.83 0.73–0.94 - -44 (-15 to -73)
Within 3 hours
At final follow-up (3–6 months)*
• Death 22.0% 20.7% 1.11 0.84–1.47 +17 (-28 to +62)
• Death or
dependency 55.2% 67.8% 0.58 0.46–0.74 -126 (-71 to -181)
* The final follow up varied among the different studies – some completed the study after 3 months, some after 6 months

Table 28 Suggested guidelines for the use of intravenous rt-PA in ischemic stroke
• Thrombolysis with intravenous r-tPA should be considered in all patients with a definite ischemic stroke who present within 3 hours of onset.
• Thrombolytic therapy should only be administered by physicians with expertise in stroke medicine, who have access to a suitable stroke
service, with facilities for immediately identifying and managing hemorrhagic complications.
• Thrombolysis should be avoided in cases where the CT brain scan suggests early changes of major infarction (e.g. edema, sulcal effacement,
mass effect).
Other exclusion criteria
Past history:
• Any intracranial hemorrhage.
• Stroke, serious head injury or myocardial infarction in previous 3 months.
• Gastrointestinal or urinary bleeding within previous 21 days.
• Major surgery within previous 14 days.
• Arterial puncture or noncompressible site within previous 7 days.
• Heparin exposure in preceding 48 hours and partial thromboplastin time not normal.
Current (i.e. pre-treatment):
• Seizure at stroke onset.
• Neurologic deficits are mild.
• Neurologic condition is improving rapidly.
• Systolic blood pressure >24.7 kPa (185 mmHg) or diastolic blood pressure >14.7 kPa (110 mmHg).
• Oral anticoagulant use or INR >1.7.
• Platelet count <100 × 109/l.
• Prolonged partial thromboplastin time.
• Blood glucose <2.8 mmol/l (50 mg/dl) or >22 mmol/l (400 mg/dl).
• Caution in patients with severe stroke (NIH stroke scale score >22).
• Discuss potential benefits and adverse effects of treatment with patient and family before treatment.
• Recommended dose of r-tPA is 0.9 mg/kg up to a maximum of 90 mg, the first 10% of the dose as a bolus over 1 minute, the rest as an
infusion over 60 minutes.
• Perform neurologic assessments every 15 minutes during infusion of rt-PA, every 30 min for the next 6 hours, and every 60 minutes for the
next 16 hours. If severe headache, acute hypertension, or nausea and vomiting occur, discontinue the infusion and obtain an emergency CT
brain scan.
• Measure blood pressure every 15 min for 2 hours, every 30 minutes for 6 hours, and every 60 minutes for 16 hours; repeat measurements
more frequently if systolic blood pressure is >24 kPa (180 mmHg) or diastolic blood pressure is >14 kPa (105 mmHg), and administer
antihypertensive drugs as needed to maintain blood pressure at or below those levels.
200 Vascular Diseases of the Nervous System

Prevention of recurrent stroke and Cigarette smoking


other vascular events There are no randomized controlled trials (RCTs), but
Secondary prevention strategies should be targeted at the observational studies suggest that stopping smoking
cause of the stroke (e.g. steroids for arteritis, antibiotics for decreases the risk of stroke by at least 1.5 times, from 7% to
infective endocarditis) and begun immediately, because the 4.7%, a RRR of 33% (95% CI: 29–38%) and ARR of 2.3%.
risk of recurrent stroke is greatest immediately after stroke. The NNT is about 43.
In most cases, the cause is atherothromboembolism or
cardiogenic embolism (see Pathogenesis, p.192). Cholesterol reduction
Observational studies do not support an association between
Blood pressure reduction increasing plasma cholesterol and all types of fatal stroke but
Lowering the BP of hypertensive TIA/stroke patients by this may represent a positive association between increasing
0.7–0.8 kPa (5–6 mmHg) diastolic and 1.3–1.6 kPa cholesterol and ischemic stroke due to large artery athero-
(10–12 mmHg) systolic for 2–3 years, or by 1.2 kPa thrombosis being diluted by a weaker association with
(9 mmHg) systolic and 0.5 kPa (4 mmHg) diastolic for 4 ischemic stroke due to intracranial small vessel disease, and
years reduces their RR of stroke by 28% (95% CI: 17–38%). a possible negative association with hemorrhagic stroke. A
If their average annual risk of stroke is 7.0%, antihyper- recent large (as yet unpublished) RCT of cholesterol-lower-
tensive therapy reduces this risk to 4.8%; a RRR of 28% and ing therapy in TIA/stroke patients, and other high vascular
ARR of 2.2%. Treating 1000 patients prevents about 22 risk patients (The Heart Protection Study, www.hpsinfo.org),
strokes each year. The NNT is 45. showed that lowering serum cholesterol by about 1 mmol/l
More intensive blood-pressure lowering (e.g. by 1.6 kPa (37 mg/dl) over 5 years with simvastatin 40 mg/day was
[12 mmHG] systolic and 0.7 kPa [5 mmHG] diastolic) for associated with a reduced risk of stroke and other serious
more prolonged periods (e.g. 4 years) yields even greater vascular events by about 24% (SE 2.6) (p<0.0001),
benefits (e.g. 43% [95% CI: 30–54%] reduction in relative risk irrespective of the patient’s baseline serum cholesterol. If the
of stroke). These results irrespective of the patient’s baseline annual stroke rate among TIA/stroke patients is 7% per year,
blood pressure, type of stroke, geographic region of residence simvastatin should reduce the annual stroke rate from 7% to
and ethnicity, and the time since the previous stroke. 5.3%, a RRR of 24%, ARR of 1.7%, and NNT of 59.

Table 29 Complications after stroke


Complication Prevention strategy
Any time after stroke
Epileptic seizures Anticipate after cortical stroke; treat after first seizure (e.g. carbamazepine, valproate, lamotrigine)
Pneumonia Assess swallowing function immediately and regularly; if risk of aspiration, feed by tube
(nasogastric, PEG, i.v.); chest physiotherapy
Fever Exclude infection (urine, chest, i.v. line, heart), infarction (chest, heart, bowel, limb), DVT, PE, drug allergy
After first few days
Urinary problems Post-void bladder ultrasound; if high residual urine volume, empty bladder by catheter, and repeat
6–8 hourly; avoid indwelling catheters
Useful appliances: absorbent pads and pants, urinals + non-spill valve for men, bedside commode,
penile sheath
Bowel problems Monitor frequency of bowel movements; maintain fluid and fiber intake
Dehydration Monitor and ensure adequate fluid intake
Pressure sores Good nursing (regular turning); natural sheepskin fleeces and bootees, padded/foam mattresses,
Roho cushions, low air loss beds
Venous thromboembolism Anticipate if immobile; mobilize early; full length graduated compression stockings; aspirin or
s/c heparin 5000 U bd if ischemic stroke
Spasticity and contractures Regular physiotherapy, and exercise at home; baclofen, diazepam; botulinum toxin (focal spasticity)
Pain Exclude treatable cause; amitriptyline or lamotrigine 200 mg/day for central post-stroke pain
Painful shoulder Recognize high risk patient (low tone, weakness, neglect, proprioceptive loss, visual field defect)
and avoid pulling on shoulder when nursing and treating the patient; physiotherapy (positioning,
mobilization, exercises); support: orthoses, sling, cuff (but may promote spasticity); medication:
analgesics (systemic, local); other physical: ultrasound, acupuncture, biofeedback,TENS; surgery:
sympathectomy, contracture relief, humeral head suspension
Falls and fractures Recognize high risk patient and environment; regularly and systematically look for signs of fracture
as patients with communication problems, sensory loss or neglect may not report injury
Low mood and anxiety Recognize high risk patient (previous depression, language problems, poor functional status, social
isolation); explain to patient nature, sequelae and prognosis of stroke; psychotherapy, antidepressants
Emotionalism Amitriptyline 25 mg nocte, or fluoxetine
PEG: percutaneous endoscopic gastrostomy; i.v.: intravenous; DVT: deep vein thrombosis; PE: pulmonary embolism;
TENS: transcutaneous electric nerve stimulation.
Stroke 201

Antiplatelet drugs Prognosis


Aspirin, given to TIA/ischemic stroke patients, reduces the Death and disability
RR of stroke and other important vascular events by about The outcome of stroke depends on the location, size and
13% (95% CI: 4–21%), from 7.0% to 6.0% per year; an ARR pathology of the lesion. Within the first 30 days after stroke,
of 1.0%. The NNT is 100. about 20–25% of patients die (10% for patients with cerebral
Compared with aspirin, clopidogrel reduces the risk of infarction but 50% intracerebral and subarachnoid
stroke and other important vascular events from about 6.0% hemorrhage).
(aspirin) to 5.4% (clopidogrel) per year, a RRR of 10% (95% The prognostic clinical indicators of poor survival include:
CI: 3–17%), and ARR of 0.6%. The NNT is 166 compared • Depression of consciousness.
with aspirin. Clopidogrel is also as safe as aspirin and, unlike • Urinary incontinence.
ticlopidine, does not cause an excess of neutropenia and • Pupillary abnormalities.
thrombocytopenia. Compared with aspirin, the combination • Gaze paresis.
of aspirin and modified-release dipyridamole reduces the risk • Severe limb weakness.
of stroke from about 6.0% (aspirin) to 5.1% per year, a RRR • Bilateral extensor plantar responses.
of 15% (95% CI: 4–26%), and ARR of 0.9%. The NNT is • Cardiac failure.
111 compared with aspirin. • Atrial fibrillation.

Anticoagulants At 1 year after stroke, about one-third of all stroke patients


Long term oral anticoagulation (target INR 2.0–3.0) is have died, about one-third are physically or cognitively
indicated for TIA/ischemic stroke patients who have a high disabled and are dependent on others for help with activities
risk source of embolism in the heart such as recent trans- of daily living, and about one-third are independent.
mural anterior myocardial infarction, dilated cardio- The prognostic indicators for poor functional recovery
myopathy, valvular heart disease, and AF. Anticoagulating include:
TIA/ischemic stroke patients in AF reduces the annual risk • Urinary incontinence.
of stroke from 12.0% to 4.0%, a RRR of 67% (95% CI: • Poor postural control.
43–80%) and ARR of 8.0%. The NNT is 12. The benefit of • Cognitive dysfunction.
anticoagulation is even greater in AF patients who have a • Poor motivation.
higher absolute risk of stroke, such as the elderly (age >75 • Visual-spatial-perceptual dysfunction.
years), and those with a history of hypertension, diabetes, • Proprioceptive loss.
or previous TIA or stroke, and echocardiographic evidence • Severe motor loss.
of impaired left ventricular function. • Initially complete dependence in activities of daily living.
Anticoagulation is no more effective than aspirin in
preventing recurrent stroke among patients with ischemic The patient’s chances of staying at home or returning
stroke due to atherothrombosis (Mohr et al. [2001]). home from hospital depend not only on the severity of their
stroke but also on other factors such as premorbid disability,
Carotid endarterectomy for symptomatic carotid stenosis whether they were living alone previously and their social
See TIA (see p.186). support and access to community services.

Carotid endarterectomy for asymptomatic carotid stenosis Stroke recurrence


Carotid endarterectomy is rarely indicated for asymptomatic The average risk of recurrent stroke in patients with a first-
severe carotid stenosis because it only reduces the 3 year risk ever stroke is about:
of stroke from 9.2% (3.1% per year) to 7.4% (2.5% per year), • 13% in the first year (15 times the risk in the general
an RRR of 20% (odds reduction 32%, 95% CI: 10–49%), and population).
ARR of 1.8% (0.6% per year). The NNT is 55 to prevent one • 4% per year for subsequent years, so that by 5 years,
stroke at 3 years, or 166 to prevent one stroke each year. about 30% will have suffered a recurrent stroke.

Carotid stenting and angioplasty Epileptic seizures


Experimental and being evaluated in clinical trials (see TIA, • 2% of patients with a first-ever stroke have a seizure at
p.186). stroke onset.
• 11% have a later seizure in the first 5 years of follow-up,
Estrogen-replacement therapy but nearly half of these patients have only one seizure.
A recent RCT showed that estrogen therapy (1 mg of • The risk of seizures is increased in survivors of intra-
estradiol-17β per day) for post-menopausal women with cerebral and subarachnoid hemorrhage, and total
recurrent TIA/ischemic stroke did not reduce mortality or anterior circulation infarction.
the recurrence of stroke, and therefore should not be • Stroke survivors who are independent at 1 month after
prescribed primarily for the secondary prevention of stroke have a very low risk of future seizures. Hence,
cerebrovascular disease (Viscoli et al. [2001]). stroke patients who are functionally competent may
return to driving after 30 days.
CLINICAL COURSE AND PROGNOSIS
Clinical course
Among stroke survivors, improvement begins within the
first few days and continues most rapidly in the first 3
months and more slowly over the next 6–12 months.
202 Vascular Diseases of the Nervous System

ATHEROSCLEROTIC Sites
ISCHEMIC STROKE • Wedge-shaped cortical/subcortical infarcts: usually due
to large or medium-sized cerebral artery occlusion (e.g.
MCA or its branches) by in situ thrombosis or embolus.
DEFINITION • Elongated sickle-shaped strips of infarction of variable
Arteriosclerosis is a generic term embracing all varieties of width from the frontal to the occipital lobes due to
structural changes that result in hardening (and thickening) infarction in the borderzone between the most distal
of the wall of large and small, and elastic and muscular parts of the anterior and middle cerebral arteries, often
arteries and arterioles (‘arteriolosclerosis’). due to severe carotid occlusive disease.
Atherosclerosis is one form of arteriosclerosis, • Small, deep lacunar infarcts, commonly in the internal
characterized by an intimal pool of necrotic, proteinaceous capsule, thalamus and ventral pons due to disease
and fatty substances in the hardened arterial wall (athere = affecting the small (40–800 microns diameter)
porridge or gruel in Greek). perforating arteries of the brain (222); i.e. the
Atherosclerotic ischemic stroke is cerebral infarction lenticulostriate perforating branches of the middle
caused by low blood flow to a part of the brain as a result cerebral artery (MCA), the thalamoperforating branches
of the complications of atherosclerosis: acute in situ of the proximal posterior cerebral artery, and the
thrombotic arterial occlusion, low flow distal to a severely perforating branches of the basilar artery to the
narrowed or occluded artery, or embolism of atherosclerotic brainstem. These usually manifest as a ‘lacunar’ clinical
plaque or thrombus to the brain. syndrome and make up about 25% of ischemic strokes
and TIAs.
EPIDEMIOLOGY
• Atherosclerosis is the most common, but not the only, Large and medium artery atherosclerosis
cause of cerebral ischemia and infarction, accounting for • Fatty streaks: focal accumulations of subendothelial
up to 75% of cases. smooth muscle cells and macrophages containing lipid
• Age: atherosclerosis begins in children and young adults (cholesterol and cholesterol ester).
and is almost universal in the elderly. • Fibrous plaque: a central core of lipid and cell debris
• Gender: both sexes, slight excess in men during middle age. surrounded by smooth muscle cells, collagen, elastic
fibers and proteoglycans. A band of fibrous tissue (the
PATHOLOGY fibrous cap) separates the lipid core (the atheroma) from
Cerebral infarction the lumen of the vessel.
Ischemic necrosis of brain tissue • Mature atheroma: three main constituents: proliferated
Pale infarction cells, predominantly smooth muscle cells; lipids
Devoid of blood. After 8–48 hours of ischemia, the infarcted (cholesterol esters) and lipid-laden macrophages (‘foam
gray and white matter swells, the line of demarcation cells’); connective tissue elements such as elastins and
between gray and white matter becomes indistinct, the white glycosaminoglycans.
matter loses its smooth ‘grain’, becoming uneven and • Complicated lesions: mature fibrous plaque with various
granular, and the swollen tissue feels softer. Histologically, types of degenerative changes such as calcification (due
there is evidence of necrosis: a hematoxylin and eosin stain to precipitation of calcium salts in the tissues),
reveals neurons with a brightly eosinophilic cytoplasm and intraplaque hemorrhage, intimal ulceration, rupture and
a darker nucleus, oligodendrocytes and glial cells with mural thrombosis.
homogeneously pale or dark nuclei that have serrated
borders and a collapsed or retracted nuclear membrane, Sites
astrocytes which are swollen with fragmented processes, axis • Large- and medium-sized arteries (e.g. aortic arch),
cylinders which are fragmented and myelin sheaths which particularly at places of arterial branching (e.g. the
swell and disintegrate after 8 hours (220). carotid bifurcation [223, 224]), tortuosity (e.g. the
After 2 days, the infarcted brain becomes mushy and carotid siphon), and confluence (e.g. the basilar artery
friable. Polymorphonuclear neutrophilic leukocytes, macro- [225, 226]).
phages and other phagocytic cells infiltrate the necrotic • Individuals with atheroma affecting one artery almost
tissue, and are particularly prominent around small vessels, always have atheroma affecting several other arteries,
where they phagocytose degradation products and become either with or without clinical manifestations.
transformed into fatty macrophages. After 10 days, the • Some sites are remarkably free of atheroma (e.g. ICA
swelling subsides, the necrotic tissue becomes liquefied and, between its origin and the siphon, and the main cerebral
if the infarct is small, early cavitation takes place as early as arteries distal to the circle of Willis). Consequently,
3 weeks (221). occlusion of a branch of the MCA is more likely to be
due to embolism than local thrombosis on an
Hemorrhagic infarction atheromatous plaque.
Extravasation of blood from many small vessels in the
infarcted area. It is usually due to embolic occlusion of a
cerebral artery, which causes ischemic necrosis of the brain
tissue supplied by the artery and necrosis of the artery itself
(due to lack of blood supply to the artery via the vasa
vasorum). If the embolus lyses or fragments distally, blood
flows into the necrotic artery and penetrates that arterial
wall, resulting in hemorrhagic infarction.
Atherosclerotic Ischemic Stroke 203

220 221

220 Early acute ischemic necrosis of brain, hematoxylin and eosin 222
stain, showing hypoxic neurons with brightly pink (eosinophilic)
cytoplasm and darker nuclei, and oligodendrocytes (which are sensitive
to ischemia) which have homogeneously dark nuclei.

221 Microscopic section of the brain at low power showing


thrombotic occlusion of a branch of a cerebral artery and surrounding
brain infarction in the stage of organization and scar formation.

222 Microscopic section of an organized, cavitated old lacunar infarct.

223 Longitudinal section of the 223 224


common carotid artery and proximal
internal carotid artery at autopsy
showing atheroma of the origin of
the internal carotid artery.
(Reproduced with permission from
Hankey GJ,Warlow CP [1994]
Transient Ischaemic Attacks of the Brain
and Eye,WB Saunders.)

224 Autopsy specimen of a cross-


section of the internal and external
carotid arteries in the axial plane
showing a patent external carotid
artery and complete occlusion of the 225 226
internal carotid artery due to in situ
atherothrombosis.

225 CT brain scan with contrast


showing a very ectatic basilar artery
(arrows).

226 Photograph of the base of the


brain at post mortem showing
atheromatous aneurysmal dilatation
and tortuosity of the basilar artery.
(Reproduced with permission from
Hankey GJ,Warlow CP [1994]
Transient Ischaemic Attacks of the Brain
and Eye,WB Saunders.)
204 Vascular Diseases of the Nervous System

Intracranial small vessel PATHOGENESIS


lipohyalinosis/microatheroma Atherogenesis
• Replacement of muscle and elastin in the arterial wall Atheroma is initiated by endothelial injury and begins as
with collagen. intimal fatty streaks in children. In genetically susceptible
• Subintimal hyalinization. individuals, it is then amplified and accelerated by factors such
• Tortuosity of the vessel. as hypertension, cigarette smoking, diabetes and hyper-
cholesterolemia. Its distribution is determined by racial factors
Lipohyalinosis/microatheroma may also lead to arterial (black and Oriental races tend to have more intracranial and
rupture and intracerebral hemorrhage, perhaps due to less extracranial atheroma) and arterial anatomy; atheroma
microaneurysm formation (so-called Charcot–Bouchard occurs at sites of hemodynamic shear stress and endothelial
aneurysms), but these may be artifacts of the pathologic trauma; boundary zone flow separation and blood stagnation
specimens. or turbulence, all of which might promote thrombosis which
itself may be involved in the progression of atheroma. Over
ETIOLOGY AND PATHOPHYSIOLOGY many years, arterial smooth muscle cells proliferate and the
Genetic predisposition intima is invaded by macrophages, fibrosis occurs, and intra-
Genetically predisposed individuals (e.g. strong family and extracellular cholesterol and other lipids are deposited to
history of atherosclerotic disease due to presumed polygenic form fibrolipid plaques. These plaques invade the media,
inheritance) are likely to develop atheroma prematurely, or spread around and along the arterial wall, and so the wall
to have particularly extensive or severe atheroma, when thickens and the lumen narrows. Later the plaques become
exposed to causal risk factors. necrotic and calcified.

Environmental risk factors Atherosclerotic ischemic stroke


The major risk factors for ischemic stroke and TIA are age Acute cerebral ischemia begins with the occlusion of a
(an 80 year old has about 30 times the risk of ischemic stroke cerebral blood vessel, usually by in situ thrombus or
as a 50 year old, probably due to more prolonged exposure embolism of thrombus (or other material) from a more
to causal risk factors), raised BP (definitely causal), atrial proximal source. Modern ideas about the pathogenesis of
fibrillation (AF), diabetes mellitus and cigarette smoking. vessel thrombosis began with Rudolph Virchow’s
dissertation in 1845 in which he enunciated his famous triad
Risk factors for cerebral infarction that thrombosis was due to changes in the vessel wall,
Definite changes in the pattern of blood flow and changes in the
• Age. constituents of the blood. It has since been established that
• Male sex. the most critical event in the formation of an arterial
• High blood pressure. thrombus is injury to the vascular endothelium.
• Cigarette smoking.
• Diabetes mellitus. Normal endothelium
• AF. Under resting physiologic conditions, the endothelium
• Ischemic heart disease. prevents thrombus formation. It acts as a physical barrier
• Heart failure. separating hemostatic from reactive subendothelial
• High plasma fibrinogen. components, and its negative surface charge may help to
• Carotid bruit/stenosis. repel platelets. It possesses anticoagulant properties
• TIA. attributable to constitutional expression of thrombomodulin
• Peripheral vascular disease. and heparin sulfate, endogenous synthesis of ectoenzymes
which degrade platelet agonists such as ADP, and
Possible endogenous synthesis of high local concentrations of the
• Hyperlipidemia. vasodilators prostacyclin (PGI2) and nitric oxide (NO)
• Hyperhomocystinemia. which inhibit platelet aggregation.
• High plasma factor VII coagulant activity.
• Low blood fibrinolytic activity. Injured endothelium: atherosclerosis
• Raised hematocrit. The most common cause of injury to the vascular
• Raised tissue plasminogen activity antigen. endothelium is atherosclerosis (but there are other causes
• Plasma viscosity (largely determined by plasma fibrinogen). such as arterial dissection [see p.224] and trauma). Acute
• Physical activity. coronary syndromes, and presumably many acute ischemic
• Obesity. stroke syndromes caused by arterial disease, are precipitated
• Snoring and sleep apnea. by thrombus which develops on an atherosclerotic plaque
• Recent infection. in which the overlying endothelium is eroded or, perhaps
• Family history of stroke. more commonly, the atherosclerotic plaque has ruptured.
• Diet (salt, fat). It remains uncertain exactly what triggers a ‘dormant’
• Alcohol (none, heavy drinking). atherosclerotic plaque to rupture, and become active and
• Race. symptomatic, but inflammation in the vessel wall is thought
• Social deprivation. to be a major factor (see below).
Atherosclerotic Ischemic Stroke 205

The vulnerable, unstable plaque Under conditions of low shear stress, platelets adhere to
Ruptured plaques contain a large core of eccentrically located subendothelial collagen and fibronectin through the binding
lipid-laden macrophages (foam cells) engorged with oxidized of platelet glycoprotein Ia–IIa receptors. Under conditions
low density lipoprotein (LDL) cholesterol, and a thin friable of high shear stress, platelets adhere to subendothelial vWF
overlying fibrous cap devoid of smooth muscle cells. The by means of platelet glycoprotein Ib–V-IX (Ib/IX).
oxidized LDL within the lipid core stimulates plaque Platelets become activated when specific platelet
inflammation, which undermines the structural integrity of the receptors bind to various agonists such as collagen,
plaque and activates the endothelium to a pro-inflammatory thrombin, thromboxane A2, adenosine diphosphate (ADP),
and procoagulant state. The oxidized LDL and other adrenaline and arachidonic acid. A series of intracellular
inflammatory stimuli initiate recruitment of inflammatory cells reactions takes place. The final common pathway of platelet
into the lesion, and serve as a second messenger to enhance activation is the assembly of the Gp IIb/IIIa receptor on
the synthesis of other vascular inflammatory products such as the surface of activated platelets. Under resting conditions,
adhesion molecules and cytokines. Cytokines and metallo- the surface of a platelet contains 50 000 to 80 000 copies
proteinases weaken the fibrous cap. The plaque is believed to of the Gp IIb-IIIa (aIIbb3) receptor. Upon platelet
rupture because the large lipid core redistributes the shear activation, the platelet GPIIb-IIIa receptor undergoes a
stress on the thin fibrous cap and very high loads are imparted conformational change, enabling it to bind both vWF and
upon localized areas of the weakened cap. fibrinogen, resulting in irreversible platelet adhesion and
aggregation, respectively. The dimeric nature of the
Triggers of plaque rupture fibrinogen molecule allows it to bind to Gp IIb-IIIa
Recent epidemiologic studies suggest that plaque receptors on two separate platelets, resulting in interplatelet
inflammation (and rupture) may be triggered by exposure bridging, platelet aggregation, and growth of the primary
(acute or chronic) to an exogenous infectious antigen. platelet clot.
There is a consistent significant relationship between
symptomatic coronary artery disease and moderately Platelet recruitment into the thrombus
elevated markers of inflammation (e.g. fibrinogen, C- Adherent, activated platelets recruit additional platelets into
reactive protein, albumin, serum amyloid A and leukocyte the growing thrombus by three mechanisms which are
count), particularly C-reactive protein. Some studies have coordinated around the central role of thrombin.
identified a higher than expected incidence of chronic Firstly, activated platelets release ADP from storage
infections of the teeth, gums and lungs, and with Chlamydia granules, and the ADP binds to the ADP receptor of
pneumoniae, Heliobacter pylori, and cytomegalovirus. adjacent platelets, which activates them.
Although there is emerging evidence that these associations Secondly, activated platelets generate and release
with chronic infections may be coincidental rather than arachidonic acid, which is metabolized by the enzyme cyclo-
causal, a similar, yet less robust, body of evidence is also oxygenase to prostaglandin endoperoxides, which in turn is
mounting for the role of a systemic, low grade, inflam- converted by thromboxane synthetase to thromboxane A2,
matory response being an integral part of the pathogenesis a potent vasoconstrictor and platelet agonist. The platelets
of acute ischemic stroke due to atherosclerosis. also release other eicosanoids, such as prostaglandin F2a and
serotonin, which induce further vasoconstriction and
Sequelae of plaque rupture: atherothrombosis platelet aggregation.
Following plaque rupture or endothelial erosion, blood is Thirdly, the modified membrane of activated platelets
exposed to the endothelial basement membrane and promotes assembly of clotting factors on the platelet surface
extracellular matrix. Von Willebrand factor (vWF), which is (V, X, VIII), thereby amplifying thrombin generation (see
a large, multimeric protein synthesized by the endothelium below). Thrombin is central to platelet aggregation.
and secreted into the subendothelium, binds to extracellular
collagen, primarily through its A3 domain. Platelets adhere Coagulation
to the subendothelial collagen (particularly types I and III), Coagulation is initiated by exposure of blood to tissue
vWF and fibronectin by means of platelet-membrane factors located in the necrotic core of ruptured
glycoprotein receptors, which are receptors for adhesive atherosclerotic plaques, in the subendothelium of injured
proteins. The largest glycoprotein (Gp) is designated I, the vessels, and on the surface of activated leukocytes attracted
smallest IX. Letters a and b were added when better to the damaged vessel. The original cascade/waterfall
techniques allowed resolution of single protein bands on hypothesis of blood coagulation is that there are two
electrophoresis into two separate bands (e.g. Gp I became activating pathways: (1) the tissue factor or extrinsic
Ia and Ib). The most abundant receptor is the integrin pathway; and (2) the contact or intrinsic pathway. A revised
family which are heterodimic molecules composed of a and hypothesis of blood coagulation maintains that there is a
b subunits. single coagulation pathway, triggered by vessel injury and
tissue factor (TF) (the factor VIIa/TF complex). Tissue
factor binds factor VIIa and the resulting factor VIIa tissue
factor complex activates both factors IX and X (i.e. the
intrinsic and extrinsic pathways are integrated in vivo).
Factor IXa assembles on the surface of activated platelets as
part of the intrinsic tenase complex which comprises factor
IXa, factor VIIIa, and calcium.
206 Vascular Diseases of the Nervous System

Factor Xa, generated through the extrinsic (factor Thromboembolism


VIIa/tissue factor) and the intrinsic tenase complex, The plaque and thrombus may also embolize, in whole or
assembles on the surface of activated platelets as part of the in part, to obstruct a smaller distal artery, perhaps the same
prothrombin-activating (prothrombinase) complex, which one on several occasions. Therefore, emboli may consist of
consists of factor Xa, factor Va, and calcium. When any combination of cholesterol debris, platelet aggregates
assembled in this way, the prothrombinase complex and fibrin. Emboli are transmitted to the brain or eye via
generates a burst of thrombin activity. Thrombin (IIa) their normal arterial supply which itself varies in distribution
activates platelets and factors V and VIII and also converts between individuals. So, an embolus from the origin of the
fibrinogen to fibrin; thrombin then binds to fibrin where it ICA usually goes to the eye or anterior two-thirds of the
remains active. cerebral hemisphere but on occasion it may go to the
As a blood clot forms at the site of vessel injury, occipital cortex if the blood in the posterior communicating
plasminogen, an inert circulating protein closely bound to artery (PCA) is flowing from the ICA to the PCA.
the deposited fibrin, is slowly activated (by tissue However, if an artery is occluded, such as the ICA,
plasminogen activator which has been activated by ipsilateral MCA distribution cerebral infarction may still
kallikrein) to form plasmin, which digests the fibrin clot to occur due to an embolus travelling from the contralateral
give fibrin degradation products. The net result is that ICA origin via the anterior communicating artery; from the
platelets and later fibrin accumulate at, and are limited to, blind stump of the proximal ICA or from disease of the
the site of vascular injury. The rest of the vasculature ipsilateral external carotid artery (ECA) via orbital
remains free of platelet and fibrin deposits because, in collaterals; or from the tail of thrombus in the ICA distal to
circulating blood, the tendency of the coagulation the occlusion. Ischemic stroke ipsilateral to an ICA
mechanism to be activated is counterbalanced by inhibitory occlusion may also be due to low flow in its normal territory
factors in the blood such as antithrombin III (which of supply, or within a boundary zone, particularly if the
inactivates factors IX, X, XI and XII). However, at the point collateral blood supply is poor. Emboli from the neck
of vessel injury, the activation of the coagulation mechanism arteries (or from the heart) seldom seem to enter the small
is so powerful that the inhibitors are overwhelmed. perforating arteries of the brain to cause lacunar infarction,
The three major inhibitory systems which modulate the perhaps because the emboli are too large.
coagulation pathway are the protein C anticoagulant path- Dilatation and ectasia of atheromatous cerebral arteries
way, tissue factor pathway inhibitor (TFPI), and anti- is unusual; such vessels often contain thrombus which may
thrombin. Protein C is activated by the thrombin/throm- embolize, or occlude small branch arteries of the ectatic
bomodulin complex on the endothelial cell surface. When vessel. Cranial nerve and brainstem compression are other
thrombin binds to thrombomodulin (an endothelial occasional complications.
membrane protein) it undergoes a conformational change
at its active site that converts it from a procoagulant enzyme CLINICAL FEATURES
to a potent activator of protein C. Activated protein C acts Atherothromboembolism is an acute on chronic disease.
as an anticoagulant in the presence of protein S by Although the formation of atherothrombotic plaque is a
proteolytic degradation and inactivation of factors Va and long and chronic process over many years, the clinical
VIIIa. TFPI binds and inactivates factors Xa and the manifestations usually occur acutely (e.g. an ischemic stroke)
TFPI/factor Xa complex and then inactivates factor VIIa and tend to cluster in time (e.g. stroke tends to occur
within the factor VIIa/tissue factor complex. Antithrombin sooner rather than later after a TIA) as a result of the
inactivates free thrombin and factor Xa, but these clotting sudden breakdown and ‘activation’ of an atherothrombotic
enzymes are protected from inactivation by antithrombin plaque. The clinical features depend on the site of arterial
when they are bound to fibrin and activated platelets, occlusion, and the degree and duration of focal brain
respectively. ischemia; see Stroke (p.192) and Neurovascular syndromes
(p.181).
Sequelae of atherothrombosis
Arterial occlusion INVESTIGATIONS
The atherothrombotic plaque and thrombus may grow to As for TIA (see p.188) and Stroke (see p.194).
obstruct the arterial lumen (224), the intraluminal
thrombus may propagate proximally or distally, or the
thrombus may be lysed by fibrinolytic mechanisms in the
vessel wall and blood.
Atherosclerotic Ischemic Stroke 207

227 228 229

227–229 Typical evolution of cerebral infarction on CT. Left parietal cortical infarct at 3 hours after symptom onset showing loss of the normal
basal ganglia outlines on the left (arrows) (227); same patient at 3 days after the stroke showing a well defined low density area with mass effect
(the left lateral ventricle is effaced) (228); same patient at 3 months after the stroke showing a shrunken, well defined area of CSF density (229).
This appearance will persist indefinitely.

DIAGNOSIS 230
Extracranial large artery atherothromboembolism
More likely to be the cause of ischemic stroke or TIA if
• Older patient: age >60.
• Total or partial anterior circulation infarction/ischemia
(clinical syndrome ± CT/MRI evidence) (227–229).
• Posterior cerebral artery territory infarction/ischemia.
• Border zone infarction/ischemia (clinical syndrome ±
CT/MRI evidence) (230).
• Cerebellar infarction/ischemia.
• Carotid, subclavian, or vertebral bruit; absent carotid
pulses; unequal radial pulses.
• Ultrasound or angiography shows >50% stenosis of the
symptomatic artery.
• Other clinical complications of atherothrombosis: angina,
past myocardial infarction, claudication, femoral bruits,
absent foot pulses, or vascular risk factors.

230 CT scan showing a border zone infarct in the left cerebral


hemisphere (arrows). Note the low density area (infarct) runs along
the border of the middle with anterior cerebral and middle with
posterior cerebral artery territories.
208 Vascular Diseases of the Nervous System

Less likely if 231


• Young patient: less than 40 years of age.
• Lacunar infarct/TIA (clinical syndrome ± CT/MRI
evidence) (231).
• Definite cardiac embolic source (see p.209).
• Clear evidence of alternative mechanism (eg. migrainous
stroke, high ESR and giant cell arteritis, arterial
dissection and so on).
• No vascular bruits, normal pulses.
• No arterial stenosis on duplex sonography or
angiography.
• No evidence of atherothrombosis elsewhere.

Intracranial small vessel disease


More likely to be the cause of ischemic stroke or TIA if
• Lacunar syndrome (clinically defined).
• CT/MRI shows a small deep and relevant infarct in
internal capsule/basal ganglia area, cerebral peduncle or
pons (231–234), or is normal.
• No clinical (cervical bruits) or ultrasonographic/angio-
graphic evidence of arterial stenosis >50%, or occlusion
of the symptomatic artery in the neck.
• No evidence of a cardiac embolic source.
• Vascular risk factors present, particularly hypertension,
smoking and diabetes.

TREATMENT
See Stroke (p.196). 231 T2W MRI of a lacunar infarct in the centrum semiovale (arrow).

PROGNOSIS
10% die within the first 30 days after cerebral infarction, and
death occurs usually between days 3 and 5, when mass effect 232
due to cerebral edema associated with massive cerebral
infarction is maximal.

232 Plain cranial CT scan, axial plane, showing high density (whiteness)
due to acute thrombus in the basilar artery and a wedge-shaped area
of low density in the left paramedian pons representing infarction due
to occlusion of a paramedian branch of the basilar artery.
Cardioembolic Stroke 209

233 CARDIOEMBOLIC STROKE

DEFINITION
Embolism of material from the heart to the brain (235)
causing ischemia or infarction of a part of the brain or eye,
with or without hemorrhagic transformation of the infarct.

EPIDEMIOLOGY
Embolism from the heart probably accounts for about 20%
of ischemic stroke and TIAs.

ETIOLOGY AND PATHOPHYSIOLOGY


Cardiac sources of embolism in anatomic sequence
Right to left shunt (paradoxical emboli from the venous
system) via
• Patent foramen ovale.
• Atrial septal defect.
• Ventricular septal defect.
• Pulmonary arteriovenous malformation.

Left atrium
• Thrombus: AF*; sinoatrial disease (sick sinus syndrome);
atrial septal aneurysm.
• Myxoma and other tumors*.

*Substantial risk of embolism.

235

234

233, 234 MRI brain (233) and autopsy specimen of brainstem (234)
showing an axial section through the mid pons and floor of the fourth
ventricle, and a small area of cavitation, due to organized lacunar
infarction, in the ventral pons on the left due to occlusion of a
paramedian perforating branch of the basilar artery.The basilar artery,
containing thrombus, can be seen ventral to the pons.The patient had
a history of a previous ataxic hemiparesis.

235 Diagrammatic illustration of the pathway a left ventricular


thrombus takes as it embolizes to the brain.
210 Vascular Diseases of the Nervous System

Mitral valve Atrial fibrillation


• Rheumatic endocarditis (stenosis* or regurgitation). The most common cause of cardioembolic stroke,
• Infective endocarditis*. accounting for up to 12% of all ischemic strokes, and an
• Mitral annulus calcification. even greater proportion of ischemic strokes in the very
• Mitral valve prolapse. elderly where its frequency in the population is highest.
• Non-bacterial thrombotic (marantic) endocarditis. Atrial fibrillation is the cause of stroke in many of these
• Libmann–Sacks endocarditis. patients but it is not always the cause because:
• Antiphospholipid antibody syndrome. • Other possible causes of stroke, which may also be the cause
• Prosthetic heart valve*. of the AF, such as ischemic heart disease and hypertension
• Papillary fibroelastoma. (e.g. carotid atheroma, intracerebral hemorrhage), are
present in about 20% of fibrillating stroke patients.
Left ventricle • Some AF patients have lacunar (presumed non-embolic)
• Mural thrombus: syndromes.
– Acute myocardial infarction (within previous few weeks)* • ‘Only’ about 13% of non-rheumatic fibrillating patients have
(236, 237). detectable (by TOE) thrombus in the left atrium (although
– Left ventricular aneurysm or akinetic segment. some thrombi may have embolized or be too small to be
– Dilated cardiomyopathy*. detected) and it is unknown if these patients have a higher
– Mechanical ‘artificial’ heart*. stroke risk than those without detectable thrombi.
– Blunt chest injury (myocardial contusion). • In a few cases the AF is caused by the stroke.
• Myxoma and other tumors*.
• Hydatid cyst. The average absolute risk of stroke in unanticoagulated non-
• Primary oxalosis. rheumatic AF patients is about 5% per annum (six times greater
than in those in sinus rhythm) and about 12% per annum in
Aortic valve
• Rheumatic endocarditis (stenosis or regurgitation).
• Infective endocarditis*. 236
• Syphilis.
• Non-infective thrombotic (marantic) endocarditis.
• Libman–Sacks endocarditis.
• Antiphospholipid antibody syndrome.
• Prosthetic heart valve*.
• Calcific stenosis/sclerosis/calcification.

Congenital heart disease (particularly with right to left shunt)

Cardiac manipulation/surgery/catheterization/valvulo-
plasty/angioplasty

*Substantial risk of embolism.

Prevalence of potential cardiac sources of embolism 236 Electrocardiograph of a patient with a recent acute anteroseptal
in patients with first-ever ischemic stroke* myocardial infarction showing sinus tachycardia, and Q waves and ST
• Any AF (238): 13%. segment elevation in leads V1–V3.
– Without rheumatic heart disease: 12%.
– With rheumatic heart disease: 1%. 237
• Mitral regurgitation: 6%.
• Recent (<6 weeks) myocardial infarction: 5%.
• Prosthetic valve: 1%.
• Mitral stenosis: 1%.
• Paradoxical embolism: 1%.
• Any of the above: 20%.
• Other sources of uncertain significance: aortic
stenosis/sclerosis; mitral annulus calcification, mitral
valve prolapse and so on: 11%.

*Sandercock PAG, Warlow CP, Jones LN, Starkey IR


(1989) Predisposing factors for cerebral infarction: The
Oxfordshire Community Stroke Project. BMJ, 298: 75–80.

Note:
• Not all cardiac sources of embolism pose equal threats
(see Diagnosis).
• Not all emboli are of the same size or the same material (fibrin, 237 Cross-section of the left ventricle showing mural thrombus
platelets, calcium, infected vegetations, tumor and so on). adjacent to an area of myocardial infarction.
Cardioembolic Stroke 211

unanticoagulated fibrillating TIA/stroke patients. The risk of • Mechanical valves have a higher risk of embolism than
stroke among patients in AF is variable; some are at particularly tissue valves, but there is no difference in stroke risk
high risk and others at particularly low risk of embolization. between the different types of mechanical valve.
• Some Bjork–Shiley tilting disc valves have disintegrated
Risk factors for embolization in AF patients and embolized pieces to the brain.
Low risk: no other detectable heart disease (so-called lone AF). • Prosthetic mitral valves are more prone to thrombosis
High risk: than aortic valves.
• Rheumatic mitral valve disease. • Infective endocarditis is a potential risk for any type of
• Previous cerebral or systemic embolic event. prosthetic valve.
• Increasing age.
• Hypertension. Rheumatic valvular disease
• Diabetes. • Rheumatic mitral stenosis/regurgitation is a well
• Left ventricular systolic dysfunction. recognized cause of left atrial dilatation causing
• Enlarged left atrium defined by echocardiography. thrombus formation and embolism to the brain.
• Spontaneous echo contrast in the left atrium, probably a • The valves also degenerate so even patients in sinus
consequence of blood stasis. rhythm who have no thrombus in the left atrium are at
• Left atrial thrombi, left atrial appendage size and risk of embolism of degenerate and sometimes calcific
dysfunction, and various hemostatic variables are perhaps fragments of valve into the circulation.
adverse risk factors also. • Stroke may also occur as a result of infective endocarditis
Uncertain risk: (see p.215) and intracerebral hemorrhage due to
• Recent onset AF. anticoagulation in these patients.
• Paroxysmal AF: probably depends on frequency and
duration of episodes of AF. Non-rheumatic sclerosis/calcification of the aortic
• Thyrotoxic AF. and mitral valves
These may also be a source of embolism in some patients but
Coronary heart disease unless calcific emboli are seen in the retina or on CT it is diffi-
Coronary heart disease is common in patients with TIA and cult to attribute confidently the TIA or ischemic stroke to this
ischemic stroke: about 20–40% have a past history of MI or condition, which is very common in normal elderly people.
current angina. Stroke may occur in up to 5% of patients
with recent acute myocardial infarction, due to: Mitral valve (or leaflet) prolapse
• Embolism of left ventricular mural thrombus. Uncomplicated mitral valve prolapse is not a cause of embolism
• Systemic hypotension. from the heart to the brain. It is only likely to be relevant to
• Intracerebral hemorrhage secondary to thrombolysis, the etiology of an ischemic stroke or TIA if it is complicating
anticoagulants or aspirin. infective endocarditis, AF, gross mitral regurgitation, or
• Embolism of catheter thrombus during coronary thrombus in the left atrium. Prolapse may be familial and
angioplasty/stenting. associated with various inherited disorders of connective tissue.
• Concurrent non-cardiac cause of stroke.
After the acute period, the risk of stroke is much lower, about Infective endocarditis (see p.215)
1% in the first year, perhaps higher if there is persisting left
ventricular thrombus. Chronic left ventricular aneurysm after Non-bacterial thrombotic (marantic) endocarditis
MI often contains thrombus but embolization is uncommon. (see Infective endocarditis, p.215)

Prosthetic heart valves Non-ischemic ‘primary’ cardiomyopathies


• The risk of embolism is about 2% per annum for all Primary cardiomyopathies are well recognized causes of
prosthetic valves, provided patients with mechanical intracardiac thrombus, particularly the dilated type rather
valves are on anticoagulants. than hypertrophic subaortic stenosis. Many are familial.

Sinoatrial disease (sick sinus syndrome)


238 • May be associated with intracardiac thrombus and
embolism, particularly if bradycardia alternates with
tachycardia, or the patient is in AF.
• May be familial.

Atrial septal aneurysm


• Uncommon.
• May be complicated by thrombus and embolism to the brain.
• Often associated with a patent foramen ovale and so has the
potential for paradoxical embolism from the venous system.
The presence of both atrial septal aneurysm and patent
foramen ovale increases the risk of recurrent stroke
(hazard ratio 4.2, 95% CI: 1.5–12).

238 Electrocardiograph showing atrial fibrillation.


212 Vascular Diseases of the Nervous System

Paradoxical embolism from the venous system, or • Posterior circulation infarction occurs when moderate or
right atrium small sized emboli impact in the top of the basilar artery
Patent foramen ovale or one of its branches.
• Common; present in about 10% of normal people. • TIA of the brain or eye.
• An uncommon cause of ischemic stroke; although • Asymptomatic: some emboli do not occlude arteries
bubbles can be shown to move from the right to the left completely or, if they do, there is sufficient collateral
side of the heart frequently, it must be rare for venous circulation to maintain tissue integrity.
thrombus to do so without also going to the lungs (and
causing symptoms), or at least to be able to make a The following strongly suggest embolism
certain diagnosis of such an event during life. from the heart
• A recent study of 581 patients, aged 18–55 years, with • Non-lacunar infarcts (i.e. TACI, PACI, POCI syndromes).
ischemic stroke of unknown origin in the preceding 3 • AF.
months (Mas et al., 2001) has shown that after 4 years • Recent acute MI.
the risk of recurrent stroke was 2.3% (95% CI: 0.3–4.3) • Valvular heart disease.
among patients with patent foramen ovale alone, 15.2% • Emboli, particularly calcific emboli, visible in the retina.
(95% CI: 1.8–28.6) among patients with both a patent • Embolic infarction in other organs (e.g. kidney: hematuria).
foramen ovale and atrial septal aneurysm, and 4.2% (95%
CI: 1.8–6.6) among patients with neither of these cardiac INVESTIGATIONS
abnormalities. There were no recurrences among patients CT brain scan
with atrial septal aneurysm alone. • Single or multiple wedge-shaped cortical/subcortical
infarcts, with or without hemorrhagic transformation.
Atrial septal defect • High density in the middle or basilar artery on the non-
contrast CT suggestive of blood clot or calcium.
Ventriculoseptal defect (rarely) • Infarction in the territory of the top of the basilar artery
or superior cerebellar artery.
Pulmonary arterial venous malformation
• Occurs in isolation or as part of hereditary hemorrhagic EKG (electrocardiograph): AF, IHD
telangiectasia (see p.153).
• Clues are clubbing, cyanosis, hemoptysis, bruit over the Chest x-ray
chest and a ‘coin lesion’ on the chest x-ray.
Echocardiography
Intracardiac tumors Indications
• Rare. • Multiple cortical/subcortical cerebral infarcts in different
• Myxomas are the most common heart tumor and most arterial territories.
occur in the left atrium. • Wedge-shaped cortical/subcortical infarct or hemorr-
• Some are familial. hage infarct, or striatocapsular infarct and no carotid
• Myxomas may cause: lesion on carotid ultrasound.
– Constitutional upset: malaise, weight loss, anemia, raised • Clinical, EKG or CXR evidence of heart disease.
ESR and hypergammaglobulinemia. • Age <45 years and no cause found for TIA or stroke.
– Cardiac symptoms: shortness of breath, palpitations, syncope.
– TIA or ischemic stroke: embolism of tumor or Preferred echocardiographic technique for detecting
complicating thrombus. various cardiac disorders
– Primary intracerebral or subarachnoid hemorrhage: myxo- Transthoracic echocardiography
matous emboli to sites of earlier symptomatic or even • Left ventricular thrombus (239, 240).
asymptomatic embolic occlusions can cause fusiform and • Left ventricular dyskinesis.
irregular aneurysmal dilatations at these sites which can • Mitral stenosis.
rupture. • Mitral annulus calcification.
• Myocardial hydatid cysts and intracardiac calcification • Aortic stenosis.
due to primary oxalosis are even rarer causes of embolism
to the brain, the former with the subsequent develop- Transesophageal echocardiography
ment of intracranial cysts. • Atrial thrombus.
• Atrial appendage thrombus.
Myocardial contusion • Spontaneous echo contrast (241).
• Blunt chest injury. • Intracardiac tumors.
• May be associated with left ventricular thrombus and • Atrial septal defect*.
embolization. • Atrial septal aneurysm.
• Patent foramen ovale* (242, 243).
CLINICAL FEATURES • Mitral and aortic valve vegetations.
• Major stroke (i.e. total anterior circulation infarction) • Prosthetic heart valve malfunction.
occurs when large emboli impact permanently in the • Aortic arch atherothrombosis/dissection.
internal carotid artery or trunk of the MCA. • Mitral leaflet prolapse?
• Partial anterior circulation infarction occurs when smaller *Transcranial doppler. Detection of intravenously injected
emboli impact in a distal branch of the anterior or MCA. air bubbles is less invasive, more specific, but not quite so
sensitive; galactose particle suspension increases sensitivity.
Cardioembolic Stroke 213

239 240

2
4

3
1

239 Transthoracic two-dimensional echocardiograph, apical four 240 Higher magnification of the left ventricular apical thrombus
chamber view, showing thrombus in the apex of the left ventricle (arrow). (239, 240 reproduced with permission from Hankey GJ,
(arrow). 1: left atrium; 2: left ventricle; 3: right atrium; 4: right ventricle. Warlow CP [1994] Transient Ischaemic Attacks of the Brain and Eye,
WB Saunders, London.)

241 242

1 3 4
2
2

3
4 1

241 Biplane transesophageal echocardiograph, longitudinal view, in a


patient with atrial fibrillation showing left atrial appendage, an enlarged
left atrium and spontaneous echo contrast. In the absence of regular
atrial contractions and the presence of a dilated left atrium, blood flow
in the left atrium is slower than normal and is seen on the real time 243
echocardiography study to be swirling around slowly in the left atrium;
slowly flowing or static blood has increased echogenicity and is called
spontaneous echo contrast (as opposed to echo contrast that is
introduced into the circulation such as agitated hemaccel). 1: mitral
valve; 2: left atrial appendage; 3: left atrium; 4: left ventricle. (Reproduced 4
with permission from Hankey GJ,Warlow CP [1994] Transient 2
Ischaemic Attacks of the Brain and Eye,WB Saunders, London.)

242, 243 Transthoracic two-dimensional echocardiograph, apical four


chamber view, using contrast (agitated hemaccel) in a patient with a
right-to-left shunt due to a patent foramen ovale. Pre-valsalva (242): 3
1
after intravenous injection of agitated hemaccel, contrast appears in
right atrium and right ventricle without passage across the patent
foramen ovale because left atrial pressure is slightly greater than right
atrial pressure. N.B. Contrast does not traverse the pulmonary
circulation to appear in the left atrium unless there is a right-to-left
shunt such as a pulmonary arteriovenous malformation.Valsalva (243):
after intravenous injection of agitated hemaccel and during valsalva maneuver (which increases right atrial pressure), contrast appears in right
atrium and simultaneously moves into the right ventricle (across the tricuspid valve) and left atrium (through the patent foramen ovale) where it is
seen here crossing the mitral valve and entering the left ventricle (arrow). 1; left atrium; 2: left ventricle; 3: right atrium; 4: right ventricle.
(Reproduced with permission from Hankey GJ,Warlow CP [1994] Transient Ischaemic Attacks of the Brain and Eye,WB Saunders, London.)
214 Vascular Diseases of the Nervous System

DIAGNOSIS Immediate anticoagulants probably worthwhile


Patients may have two or more competing causes of cerebral Best time to start anticoagulants unclear
ischemia, such as carotid stenosis and atrial fibrillation, so • Acute MI within past few weeks and confirmed ischemic
one cannot always be sure which is the cause in an individual stroke or TIA.
patient. • Disabling ischemic stroke and AF.

Embolism from the heart to the brain or eye Aspirin or no antithrombotic therapy; anticoagulants
More likely to be the cause of ischemic stroke or TIA if not worthwhile or contraindicated
• An identified cardiac source of embolism, particularly one Low risk of recurrent cardioembolic stroke without
with a substantial embolic risk (see Prognosis, below). anticoagulants
• Ischemic events in more than one arterial territory, • Heart disease with a low risk of embolism.
particularly if more than one organ is involved. • Other, non-cardiac lesion more likely cause of cerebral
• No evidence clinically (bruits, palpation), on ultrasound infarct: severe ipsilateral carotid stenosis; likely disease of
or by angiography of arterial disease (>50% stenosis) in intracranial small vessels (i.e. lacunar infarct).
the neck.
• Calcific emboli in the retina (very rare). Little to gain from long term anticoagulation
• Calcific emboli on brain CT (even rarer). • Suspected cholesterol embolization syndrome (i.e.
• No vascular risk factors. ischemic stroke during cardiac catheterization, after
• Age <50 years. cerebral angiography or other vascular procedure).
• No other explanation for the stroke. • Already severely disabled before the stroke.
• Moribund or predicted to be severely disabled in the
Less likely if long term.
• Lacunar syndrome (clinical syndrome + CT/MRI evidence).
• Low flow infarction/ischemia (clinical syndrome + High risk of cerebral hemorrhage on anticoagulants
possibly CT/MRI evidence [e.g. borderzone infarction]). • Infective endocarditis.
• Large cerebral infarct with midline shift and/or evidence
Uncertain if of major hemorrhagic transformation on brain CT.
• Hemorrhagic transformation of the infarct. • Likely to comply poorly with anticoagulation therapy and
• Past TIA. its monitoring after discharge.
• Severe, uncontrolled hypertension.
High risk of embolism
• Non-rheumatic or rheumatic AF. Contraindication to anticoagulants
• Infective endocarditis. These depend on individual circumstances and are seldom
• Prosthetic heart valve. absolute.
• Recent MI. • History of bleeding disorder:
• Dilated cardiomyopathy. – Hemophilia.
• Intracardiac tumors. • Uncorrected major bleeding disorder:
• Rheumatic mitral stenosis. – Thrombocytopenia.
– Hemophilia.
Low risk of embolism – Liver failure.
• Mitral valve prolapse (uncomplicated). – Renal failure.
• Mitral annulus calcification. • Uncontrolled severe hypertension:
• Patent foramen ovale with no evidence of deep venous – Systolic pressure over 26.7 kPa (200 mmHg).
thrombosis. – Diastolic pressure over 16 kPa (120 mmHg).
• Atrial septal aneurysm. • Potential bleeding lesions:
• Aortic sclerosis. – Active peptic ulcer.
– Esophageal varices.
TREATMENT – Intracranial aneurysm.
Of acute ischemic stroke of suspected cardioembolic origin. – Proliferative retinopathy.
– Recent organ biopsy.
Immediate anticoagulants very likely to be – Recent trauma or surgery to head, orbit, spine.
worthwhile – Recent stroke, but patient has not had a brain CT scan
Start as soon as possible or MRI.
TIA or ischemic stroke with complete recovery within – Confirmed intracranial or intraspinal bleeding.
1–2 days + high risk of embolism (e.g. AF). – History of heparin-induced thrombocytopenia or
thrombosis (if heparin planned).
Best time to start unclear • If warfarin planned:
Non-disabling ischemic stroke, no hemorrhagic transform- – Homozygous protein C deficiency (risk of skin necrosis).
ation of the infarction, and AF (?start within 3–7 days of – History of warfarin-related skin necrosis.
stroke, earlier if small infarct and normal blood pressure; – Uncooperative/or unreliable patients (long term therapy).
later if large infarct or high blood pressure). – Risk of falling.
– Unable to monitor INR.
Infective Endocarditis 215

Adverse effects of heparin INFECTIVE ENDOCARDITIS


Local minor complications of subcutaneous heparin at
injection site
• Discomfort. DEFINITION
• Bruising. Direct infection of the endocardial surface of the heart
(heart valves and mural surface) by any of a wide range of
Local complications of intravenous heparin at cannula site micro-organisms.
(or elsewhere)
• Pain at cannula site. EPIDEMIOLOGY
• Infection at cannula (sometimes with severe systemic • Incidence: 2 cases per 10 000 population per year. Little
infection). change over the past 30 years.
• Reduced patient mobility because of infusion lines and • Age: adults: 50% >50 years of age; uncommon in childhood.
pump. • Gender: M=F.

Major systemic complications PATHOLOGY


• Intracranial bleeding: • Platelets and fibrin deposited on an endocardial surface
– Hemorrhagic transformation of cerebral infarct damaged by turbulent flow or instrumentation, followed
(potentially disabling or fatal). by colonization with organisms possessing appropriate
– Intracerebral hematoma. adherence properties.
– Subarachnoid hemorrhage. • Direct binding to endothelium of normal hearts by
– Subdural hematoma. organisms such as Staphylococcus aureus.
• Extracranial hemorrhages: • Mature vegetations, due to further platelet and fibrin
– Subcutaneous (can be massive). deposition, within which microbial multiplication may
– Visceral (hematemesis, melena, hematuria). proceed uninhibited.
• Thrombocytopenia:
– Type I: dose and duration related, reversible, mild, ETIOLOGY AND PATHOPHYSIOLOGY
usually asymptomatic, not serious and often resolves • Introduction of pathogens into the circulation, exposing
spontaneously. a susceptible intracardiac or intravascular lesion to
– Type II: idiosyncratic, allergic, severe (may be attachment and colonization.
complicated by arterial and venous thrombosis). Affects • The coincidence of tissue damage from turbulent blood
3–11% of patients treated with intravenous heparin and flow or a foreign body, and lesions or procedures that
less than 1% of patients treated with subcutaneous introduce pathogens.
heparin.
• Osteoporosis. Organisms
• Skin necrosis. • Streptococci: 60–80% of cases.
• Alopecia. • Staphylococci: <30%.
• Gram-negative bacteria (Salmonella, other Entero-
bacteriaceae, Pseudomonas spp.): <10% .
• Fungi (Candida albicans, Aspergillus): <4%.
• Coxiella burnetii (Q-fever).
• Chlamydia.
• Mycoplasma.
• Non-toxigenic Corynebacterium diphtheriae biovar gravis.

Risk factors
Heart abnormalities
• Rheumatic heart disease (especially mitral valve defects).
• Congenital heart disease (especially bicuspid aortic valve
in the elderly).
• Mitral valve prolapse: eightfold excess risk.
• Degenerative heart disease: calcified mitral valve annulus;
post infarct mural thrombosis.

Iatrogenic and lifestyle-associated factors


• Pre-existing dental caries.
• Intravenous drug misuse.
• Prosthetic heart valves.

Procedures involving mucosal surfaces associated with


bacteremia
• Upper airway: oral/dental procedures (15–40%).
• Gastrointestinal (10%).
• Urologic (10–30%)
• Obstetric (<10%).
216 Vascular Diseases of the Nervous System

Pediatric risk factors – Vegetations may embolize to cause ischemic stroke (and
• Previous cardiac surgery. sometimes global encephalopathy due to multiple emboli),
• Congenital heart defects, particularly those involving the ischemia in other organs, and pulmonary embolism.
septum. – Similar vegetations are found in systemic lupus erythe-
• Intravascular catheterization. matosus and the antiphospholipid antibody syndrome.
• Malignancy.
CLINICAL FEATURES • Tuberculosis.
Infection • Connective tissue disease: systemic lupus erythematosus
• Fever (90% of cases) and chills (40%): may be absent in associated with Osler’s nodes and Roth’s spots.
the elderly or following antibiotic treatment.
• Myalgia and arthralgia (40%). INVESTIGATIONS
• Systemic upset: anorexia, weight loss, cough, dyspnea, No specific diagnostic tests:
hemoptysis, chest/abdominal pain (10–25%). • Full blood count: mild normochromic normocytic
• Clubbing of fingers in long-standing or prolonged anemia (80% of cases); neutrophil leukocytosis in acute
infective endocarditis. infective endocarditis but not in chronic infective
endocarditis.
Systemic embolism • ESR and serum C-reactive protein: raised in 90–100% of
• Linear (splinter) hemorrhages may be found under the cases.
nails (244). • Urinalysis: often abnormal, with proteinuria, hematuria,
• Conjunctival and oral small, red, petechial hemorrhagic and occasionally casts.
lesions with a pale center. • Blood cultures to detect constant bacteremia: three sets
• Non-tender subcutaneous erythematous maculopapular in first 24 hours: positive in 90% of cases; negative blood
lesions (Janeway’s spots), on pulp of the fingers (245), cultures (10% of cases) suggest Legionella, ‘HACEK’
that may ulcerate: suggest staphylococcal infection. (Haemophilus spp., Actinobacillus actinmycetemcomitans,
• Erythematous or purple, painful, tender nodules (Osler’s Cardiobacterium hominis, Eikenella corrodens, Kingella
nodes) on the palms of the hands, soles of the feet, pulp spp.) organisms and non-bacterial infective endocarditis.
of the fingers, or other sites. • Serology: antistaphylococcal antibodies, antistreptolysin
• Gangrene of fingers, toes, or larger portions of the O titer.
extremities. • Serial EKGs: may detect conduction defects (aortic root
• Retinal hemorrhages: small, occasionally flame-shaped abscess), pericarditis and MI (emboli).
hemorrhages that may have pale centers (Roth spots). • 2D echocardiography: valve vegetations (246), abscess
• Pulmonary emboli (tricuspid valve). formation, and valve destruction, but can be normal.
• Ischemic stroke or TIA (about 20% of patients): • Doppler flow analysis and cardiac catheterization with
sometimes as the initial presentation but more often in angiography have a role in pre-operative evaluation.
the context of someone who is already ill, perhaps in • CSF: can be normal but more than 100 poly-
hospital, and before the infection has been controlled; morphs/mm3 in patients with stroke is said to suggest
caused by embolism of infected vegetations. endocarditis; however, as high or higher counts have
• Hemorrhagic transformation of a brain infarct: quite been described in primary intracerebral hemorrhage and
common, sometimes due to the (unwise) use of in hemorrhagic transformation of an infarct, but not in
anticoagulation. cerebral infarction.
• Primary intracerebral hemorrhage, and rarely sub- • Cerebral angiography to detect unruptured (mycotic)
arachnoid hemorrhage: due to pyogenic vasculitis or aneurysms with a view to surgery is unnecessary, as these
ruptured mycotic aneurysms, which can be single or aneurysms do not always rupture and tend to resolve
multiple and most often affect the distal branches of the with time.
MCA.
• Meningitis. DIAGNOSIS
• Diffuse encephalopathy, perhaps due to showers of small Fever, cardiac murmur, positive blood cultures and
emboli. vegetations seen on echocardiography, but these features are
• Discitis. not invariable. For the Duke criteria, see Heiro et al.,
• Renal emboli (hematuria). (1998), Table 3, Mylonakis and Calderwood (2001),
Further reading, p.271.
Cardiac dysfunction
• Heart murmur: (85%); ‘changing’ or ‘new’ murmurs: TREATMENT
<10% of cases, usually staphylococcal infection. Medical
• Congestive heart failure. • Question the patient closely for a history of penicillin
‘allergy’.
DIFFERENTIAL DIAGNOSIS • Antimicrobial chemotherapy with sustained levels of
• Non-bacterial thrombotic (marantic) endocarditis: bactericidal drugs (preferably synergistic) administered
– Cachectic and debilitated, often elderly, patients with via a central intravenous line (see Table 30).
cancer (adenocarcinomas usually) and sometimes dis- • Because antibiotics may penetrate vegetations poorly,
seminated intravascular coagulation, burns and septi- prolonged treatment is required to ensure eradication
cemia. and prevent relapse.
– Small, sterile, friable valve vegetations, consisting of fibrin
and platelets.
Infective Endocarditis 217

Table 30 Therapy for infective encarditis (also see Table 4, Mylonakis and Calderwood [2001])

Organism First line therapy Duration Alternative therapy


Streptococcus Penicillin G, 10–20 mU/day (benzylpenicillin 1.8 g 4 hourly) 4 weeks Ceftriaxone, vancomycin
viridans plus gentamicin, 3 mg/kg/day (1 mg/kg 8 hourly) 2 weeks Streptomycin
(maximum 240 mg)
Enterococci Penicillin G, 20–30 mU/day (benzylpenicillin 1.8–2.4 g 4 hourly) 6 weeks Ampicillin, vancomycin
plus gentamicin, 3 mg/kg/day (1 mg/kg 8 hourly) 4–6 weeks
(maximum 240 mg)
Staphylococcus Flucloxacillin, 12 g/day i.v. (2 g 4 hourly) 6 weeks Nafcillin, vancomycin (1 g 12 hourly)
aureus plus gentamicin, 3 mg/kg/day 3–5 days Fusidic acid
(maximum 240 mg)
Organism not identified Empirical therapy Duration Alternative therapy
Native valve Penicillin G (1 g 4 hourly) plus gentamicin (4–6 mg/kg daily) Until organism identified Ampicillin plus gentamicin
plus flucloxacillin (2 g 4 hourly)
Prosthetic valve Vancomycin (1 g 12 hourly) plus gentamicin (4–6 mg/kg daily)
Culture negative Penicillin G 6 weeks
plus gentamicin 2 weeks
(i.e. as for enterococci unless Q fever or fungus suspected)

244 245

244 Linear splinter hemorrhages under the nails, which can be difficult 245 Pulps of the fingers showing erythematous maculopapular lesions
to differentiate from traumatic lesions. (Janeway’s spots) which were non-tender.

• Close collaboration with the microbiology laboratory to 246


assess antibiotic sensitivities, including minimum inhibitory
concentrations and minimum bactericidal concentrations.
• Peak and serum levels of aminoglycosides must be
monitored.
• Anticoagulation is not advised for native valve infective
endocarditis because of the increased risk of intracranial 2
hemorrhage. If anticoagulation is required during
prosthetic valve infective endocarditis, close monitoring
of coagulation times is required.
• Fungal infection seldom responds to antifungal agents
(e.g. amphotericin B), and early surgery is often required. 1

246 Transthoracic 2D echocardiograph, parasternal long axis view,


showing vegetations (arrow) on the anterior leaflet of the mitral valve,
which is situated behind the aortic valve.This image is taken during
diastole when the mitral valve is open and the aortic valve closed. 1: left atrium; 2: left ventricle. (Reproduced with permission from Hankey GJ,
Warlow CP [1994] Transient Ischaemic Attacks of the Brain and Eye, WB Saunders, London.)
218 Vascular Diseases of the Nervous System

Surgery ANTIPHOSPHOLIPID SYNDROME


Required in up to one-quarter of patients (247, 248), AND OTHER PROTHROMBOTIC
particularly prosthetic valve infective endocarditis. STATES (THROMBOPHILIAS)
Indications
• Fever persisting after 2 weeks of antimicrobial therapy: Disorders of the blood that predispose to recurrent venous
suggests antimicrobial failure, intracardiac abscess and possibly arterial thrombosis.
formation, embolization, or other complications.
• Valve dysfunction.
• Arrhythmias. ANTIPHOSPHOLIPID SYNDROME
• Refractory heart failure.

Surgical repair of any asymptomatic mycotic aneurysm, DEFINITION


which may be found after a ruptured aneurysm has been A heterogeneous disorder, both in terms of its clinical
repaired, is unnecessary because these aneurysms do not manifestations and range of autoantibodies, which is
always rupture and tend to resolve with time. characterized by thrombosis (venous or arterial), recurrent
miscarriage, or both, in association with persistent positive
Prevention of bacterial infective endocarditis laboratory tests for antiphospholipid antibody: lupus
Antibiotic prophylaxis before undergoing procedures anticoagulant (LA), anticardiolipin antibody (ACA), or
associated with bacteremia is indicated for adults with both, on repeated studies. Thrombocytopenia is an
conditions predisposing to infective endocarditis (see Risk occasional feature.
factors, p215, and Table 31).
Antiphospholipid antibodies (APAs)
PROGNOSIS • APA are a family of antibodies which are specific for
• Untreated: recovery is rare. several plasma proteins, such as human prothrombin and
• Appropriate antibiotic treatment: >70% survival for β2 glycoprotein (β2 GPI), which may bind to
endogenous valve infection and >50% survival for phospholipid (PL) surfaces.
prosthetic valve infection. • LA and ACA are different APAs but occur together in
• Poor prognostic factors: congestive heart failure, extreme about 60% of patients with the phospholipid antibody
age, aortic valve or multiple valve involvement, prosthetic syndrome; in the remaining 40%, only one is present.
valve involvement, Gram negative bacillus, fungal or • LAs are immunoglobulins (IgG, IgM, IgA, or mixtures)
polymicrobial bacteremia, failure to identify the which interfere with one or more of the in vitro
infectious agent due to negative blood cultures, phospholipid-dependent tests of coagulation (e.g.
antimicrobial resistance to antibiotics, and delay in activated partial thromboplastin time [APTT], dilute
initiating therapy. Russell viper venom time [dRVVT], dilute prothrombin
time [dPT], and kaolin clotting time [KCT]). They
therefore slow the rate of thrombin generation and
therefore clot formation in vitro.
• ACAs are detected by immunoassay, most commonly
enzyme-linked immunosorbent assay.

247 248

247, 248 Vegetations consisting of platelets, fibrin and colonizing micro-organisms removed surgically from an infected heart valve.
Antiphospholipid Syndrome and Other Prothrombotic States (Thrombophilias) 219

EPIDEMIOLOGY ETIOLOGY
• Prevalence: 1–40% or so of ischemic stroke/TIA patients Primary
have raised circulating IgG or IgM anticardiolipin No evidence of other underlying disease.
antibodies and/or the lupus anticoagulant, depending
on the selection of patients, the timing of the blood Secondary
sample after the onset of cerebral ischemia, the • Associated with rheumatic and connective tissue
laboratory methods, and what level is deemed ‘normal’. disorders: systemic lupus erythematosus (SLE): about
However, only a few of these patients have some or all of 30–40% of patients with SLE have LA; rheumatoid
the constellation of features known as the APA arthritis; systemic sclerosis; temporal arteritis; Sjögren’s
syndrome. syndrome; psoriatic arthropathy; Behçet’s syndrome;
• Age: any age. others.
• Gender: F>M. • Associated with other conditions:
– Infections: viral (e.g. HIV-1, varicella, hepatitis C);
PATHOLOGY bacterial (e.g. syphilis); parasitic (e.g. malaria).
Thrombi are typically ‘bland’ and may be found in vessels – Drug exposure: chlorpromazine; hydralazine; quinidine;
of any size and on heart valves, such as the mitral valve quinine; antibiotics; phenytoin; valproate; procainamide.
leaflets. There is no evidence of inflammation of the vascular – Lymphoproliferative diseases; malignant lymphoma;
wall. paraproteinemia.
– Miscellaneous conditions: autoimmune thrombo-
PATHOPHYSIOLOGY OF THROMBOSIS (see cytopenia; autoimmune hemolytic anemia; sickle-cell
Atherosclerotic ischemic stroke, p.204) disease; intravenous drug abuse; livedo reticularis;
The paradoxical association between the presence of auto- Guillain–Barré syndrome.
antibodies with in vitro anticoagulant effects and the
occurrence of a prothrombotic state is not fully understood. CLINICAL FEATURES
Patients with antiphospholipid syndrome have evidence of Any vascular site can be affected so there is a wide range of
persistent coagulation activation (increased plasma concen- clinical manifestations:
tration of markers of thrombin generation such as
fibrinopeptide A) and, as stated above, vascular occlusion is Dermatologic
due to thromboembolism or embolization from sterile • Sneddon’s syndrome: livedo reticularis (249), stroke-like
vegetations on heart valves, and not vasculitis. episodes and hypertension.
It is unlikely that a single prothrombotic mechanism • Non-healing ulceration of the ankles and skin necrosis.
operates. Possible mechanisms include:
• Perturbation of the protein C system: the most likely
cause of venous thrombosis: APAs interfere with activated
protein C down-regulation of factors Va and VIIIa,
resulting in an acquired activated protein C resistance.
• Alteration of the antithrombin III heparin sulfate down- 249
regulation of serine proteases: some APAs have specificity
for heparin sulfate found on the surface of endothelial
cells.
• Up-regulation of tissue factor expression by endothelial
cells.

In some cases the APAs may represent an


epiphenomenon.

249 Livedo reticularis in a patient with antiphospholipid antibody


syndrome.

Table 31 Prophylaxis for infective endocarditis


Procedure Standard prophylaxis Penicillin allergy or penicillin given in last month
Dental (local anesthetic) Amoxycillin (amoxicillin), 3 g p.o. 1 hour Clindamycin or erythromycin
before, then amoxycillin, 1.5 g p.o. 6 hours later
Adults with: prosthetic valve, Amoxycillin (amoxicillin), 1 g i.v. 30 min before, Vancomycin 1g i.v. 100 min before,
past infective endocarditis, plus gentamicin, 120 mg iv 30 min before plus gentamicin 120 mg i.v. 30 min before
high-risk procedure then amoxycillin, 500 mg p.o. 6 hours later
(e.g. genito-urinary)
220 Vascular Diseases of the Nervous System

Neurologic These are indirect coagulation assays sensitive to the


• TIA, stroke, or multifocal encephalopathy due to arterial phospholipid-dependent steps of blood coagulation. At least
or venous thrombosis in any sized vessel (250–254). two assays, with sensitive reagents and techniques must be
Venous thromboembolic events account for about 70% used, most commonly the APTT with another test.
of cases and arterial events the remaining 30%. The most
common site for arterial thrombi is the cerebral Immunologic assays
circulation. • Anticardiolipin antibody (ACA): detected by means of
• Migraine-like headaches. solid-phase immunoassay, such as enzyme-linked
immunosorbent assay (ELISA) or radio immunoassay
Obstetric (RIA), employing cardiolipin or other negatively charged
• Recurrent spontaneous miscarriage/fetal loss due to phospholipids as the antigen to measure antibody
intrauterine death in the latter part of the first trimester concentration and binding avidity.
or early second trimester. • Other phospholipids, e.g. phosphatidyl serine.
• Intrauterine foetal growth retardation. • Antibodies to β2 GPI .
• Early-onset pre-eclampsia.
• Prematurity. Other thrombophilia diagnostic blood tests
• Antithrombin activity. (N.B. heparin reduces
Pediatric antithrombin antigen and activity by 10–15%.)
Post-infectious (e.g. varicella) thromboembolic events. • Protein C activity. (N.B. Warfarin reduces protein C and
protein S levels by 30%. These tests must be conducted
INVESTIGATIONS FOR ANTIPHOSPHOLIPID prior to, or at least 2 weeks after, cessation of warfarin.)
SYNDROME OR ANY OTHER SUSPECTED • Protein S antigen level.
PROCOAGULANT STATE (THROMBOPHILIA) • Factor V Leiden genetic analysis or activated protein C
Indications resistance functional analysis.
• Young (<50 years) and no other cause found for TIA or • Prothrombin G20210A mutation.
ischemic stroke. • Antinuclear antibodies.
• Past history or family history of premature arterial or • Serum protein levels, serum protein electrophoresis and
venous thrombosis, especially if unusual sites (cerebral, plasma viscosity: indicated in patients with elevated ESR
mesenteric, hepatic veins). or suspected hyperviscosity syndrome.
• Past history of recurrent miscarriages. • Hemoglobin electrophoresis/sickle test: indicated in
• Abnormal full blood picture or blood film (e.g. appropriate racial groups (i.e. black patients) to detect
thrombocytopenia). sickle cell trait or disease.
• VDRL/RPR positive: may be a false positive in the • Fibrinogen.
presence of ACAs because cardiolipin is the antigen used • Platelet aggregation.
in the VDRL assay. However, the VDRL is positive in • Fasting plasma homocysteine (before and after a
only about 25% of patients with APLAb. methionine load if possible).

Thrombophilia diagnostic tests


• Full blood count and film.
• ESR.
• Prothrombin time.

Coagulation assays for lupus anticoagulant (LA)


• Activated partial thromboplastin time (APTT): lacks
sensitivity but remains the most appropriate screening 250
test for the LA. A prolonged APTT that fails to correct
when affected plasma is mixed with normal plasma
implies inhibition of the clotting system rather than
deficiency of a component, and is the laboratory hallmark
of LA.
• Dilute Russell’s viper venom time (dRVVT):
prolongation of the dRVVT will not correct with the
addition of normal plasma in the presence of LA (in
contrast to the clotting factor deficiency). For
confirmation, a platelet neutralization procedure, to
show the dependence of phospholipid inhibitors, should
be performed.
• Kaolin clotting time test: also detects LA, but it is not as
sensitive as the dRVVT, and sensitivity is highly reagent
dependent.
• Tissue thromboplastin inhibition test. 250 Ocular fundus photograph showing engorged retinal veins and
multiple retinal hemorrhages due to a central retinal vein occlusion.
(Courtesy of Mr M Wade, Department of Medical Illustrations, Royal
Perth Hospital, Australia.)
Antiphospholipid Syndrome and Other Prothrombotic States (Thrombophilias) 221

DIAGNOSIS Lupus anticoagulant (LA)


Thrombophilia Four sequential steps are necessary to establish the diagnosis
The diagnosis is usually established from routine first line of LA:
investigations (full blood count, platelet count and ESR) 1 Demonstration of an abnormal phosoholipid- (PL)
and standard thrombophilia screening and diagnostic tests. dependent coagulation test (e.g. APTT, dRVVT).
2 Proof that the abnormality in step 1 is due to an inhibitor
Antiphospholipid antibody syndrome (synonym: anticoagulant).
Cannot be diagnosed on the basis of a single raised titer of 3 Establishing the PL-dependence of the inhibitor.
ACA in the serum. The titer must be substantially raised on 4 Ruling out other coagulopathies.
several occasions and associated with not just cerebral
ischemia but also with some combination of deep venous Ischemic stroke due to a procoagulant state
thrombosis, recurrent miscarriage, livedo reticularis, cardiac It can be very difficult to attribute confidently the cause of
valvular vegetations, thrombocytopenia and migraine. the ischemic stroke to the hematologic disorder if the
patient has other disease that could have caused the stroke.

251 CT brain scan showing 251 252


a triangular filling defect in the
venous sinus at the torcula
(arrow) after i.v. contrast
(‘empty delta’ sign) due to
venous sinus thrombosis.

252 MRI brain scan, proton


density weighted image,
showing hypointense (black)
flow voids in the middle and
posterior cerebral arteries
(arrowheads), and a
hyperintense (white) filling
defect (arrow) representing
loss of the flow void in the
venous sinus at the torcula
(due to the thrombus).

253 Proton density 253 254


weighted MRI of a right
thalamic infarct (arrow) at 24
hours in a young patient with
the antiphospholipid antibody
syndrome.

254 Same patient as in 253,


adjacent image shows high
signal in the right posterior
cerebral artery indicating that
it is occluded by fresh
thrombus (arrow).
222 Vascular Diseases of the Nervous System

More often than not, the hematologic disorder is one of Platelets


several factors predisposing to thrombus formation, such as • Essential thrombocythemia (idiopathic primary
coexistent activated protein C resistance, or coexistent athero- thrombocytosis):
thrombosis, trauma or dehydration. Irrespective, the hema- – Platelet count >500 × 109/l, causes arterial and venous
tologic disorder frequently needs treating in its own right. thrombosis.
– Headache with transient focal and non-focal neurologic
TREATMENT disturbances are the most common neurologic
Uncertain; no controlled trials of aspirin, anticoagulants, or symptoms.
immunosuppressive therapy have been undertaken. – Occasionally, platelet function is defective predisposing
to bleeding.
Thromboembolic events – Other causes of thrombocytosis should be excluded:
Long term anticoagulant therapy, INR: 2.5 (empirical, malignancy, splenectomy or hyposplenism, surgery and
controlled trials are needed), if risks considered to be other trauma, hemorrhage, iron deficiency, infections,
outweighed by benefits. Otherwise antiplatelet therapy. polycythemia rubra vera, myelofibrosis and leukemia.

Pregnancy Qualitative abnormalities of formed blood elements


Heparin 24 000 units per day s.c. and aspirin 75 mg per day. Erythrocytes
Low molecular weight heparin may cause less osteoporosis, • Sickle cell (SC) disease:
heparin-induced thrombocytopenia, and bleeding events – Ischemic stroke is common in homozygotic children and
than standard heparin and does not require blood young adults; intracranial hemorrhage may also occur
monitoring. sometimes.
– Stroke is rare in heterozygotic adults, and usually in the
PROGNOSIS context of a hypoxia-provoked sickle-cell crisis.
Uncertain, but limited studies suggest a high rate of – Small and large arteries, and veins, are occluded by
recurrent stroke and other vascular events in patients with thrombi as a result of the rigid red blood cells, raised
ischemic stroke and the antiphospholipid syndrome (hence whole blood viscosity, thrombocytosis, impaired
the empirical recommendation for long term anticoagu- fibrinolytic activity, and arterial stenosis due to fibrous
lation therapy). proliferation of the intima; the last can be detected in the
MCA with transcranial doppler and is predictive of stroke.
OTHER PROCOAGULANT STATES • Hemoglobin SC disease:
Quantitative abnormalities of formed blood – may also be complicated by stroke.
elements • Paroxysmal nocturnal hemoglobinuria:
Erythrocytes – Very rare.
• Polycythemia rubra vera (primary polycythemia): – Intracranial venous thrombosis is more common than
– Hematocrit >0.50 in males and >0.47 in females, arterial thrombosis.
provided the patient is rested, normally hydrated, and the – Anemia, abdominal pain, recurrent deep venous
blood has been taken without venous occlusion. thrombosis, dark urine, hemolysis and a low platelet and
– Raised red cell mass. granulocyte count are recognized features.
– Increased whole blood viscosity, raised platelet count,
and enhanced platelet activity, may cause TIAs, cerebral Hyperviscosity
infarction and intracranial venous thrombosis. Paraproteinemias
– Platelet function may also be defective, predisposing to • Waldenstrom’s macroglobulinemia, multiple myeloma
intracranial hemorrhage. and perhaps cryoglobulinemia:
• Anemia (iron deficiency and presumably other types): – Most commonly cause the ‘hyperviscosity syndrome’, a
– If severe, usually causes non-specific neurologic global encephalopathy characterized by headache, ataxia,
symptoms such as generalized weakness, poor diplopia, dysarthria, lethargy, poor concentration,
concentration and faintness but in the presence of severe drowsiness and coma, visual blurring, and deafness. The
arterial disease, it may provoke focal cerebral ischemia. retina shows dilatation and tortuosity of the veins, venous
occlusions, papilledema and hemorrhages; similar
Leukocytes symptoms may also be due to uremia, hypercalcemia or
• Leukemia: lymphoma complicating the paraproteinemia.
– May predispose to cerebral arterial or venous occlusion – Arterial or venous cerebral infarction may occur if vessels
because of increased whole blood viscosity. become occluded by acidophilic material which are
– More commonly is associated with intracranial hemorr- probably precipitants of the abnormal plasma proteins.
hage because of the hemostatic defect or CNS leukemic – Intracranial hemorrhage also occurs due to the reduced
infiltration. L-asparaginase (asparaginase) treatment for number and impaired reactivity of platelets, perhaps as a
leukemia can cause both cerebral ischemia and result of uremia.
hemorrhage.
• Malignant angioendotheliosis (intravascular lymphoma): Coagulation disorders
– Can present like a stroke but the neurologic involvement Antithrombin III
soon becomes diffuse and progressive. • Inactivates thrombin and activated factors X, IX, XI, and
XII (but not VII).
Antiphospholipid Syndrome and Other Prothrombotic States (Thrombophilias) 223

• An important mediator of the anticoagulant effect of patient must be properly assessed and other potential
heparin; heparin increases the activity of antithrombin- causes of ischemic stroke (e.g. arterial dissection,
III by 100-fold. Indeed, heparin resistance is a clue to paradoxical embolism from the venous system) must be
low antithrombin-III activity. considered and excluded if possible.
• Deficiency may be inherited (as an autosomal dominant – Low levels of these coagulation factors may be caused by
trait with variable penetrance in about one in every the acute stroke itself or its treatment (anticoagulants),
2000–5000 people), or acquired as a result of severe liver so the results must be confirmed by repeated testing on
disease (reduced synthesis), intravascular thrombosis several later occasions.
(consumption), the nephrotic syndrome (renal loss), or – Acute stroke is associated with activation of procoagulant
use of medications such as L-asparaginase or the oral and fibrinolytic pathways, as manifest by a significant
contraceptive pill. decrease in functional antithrombin III and plasminogen
and an increase in thrombin–antithrombin complex, total
Protein C protein S, tissue plasminogen activator, plasminogen
• A vitamin K-dependent plasma protein that is synthesized activator inhibitor-1, and D-dimer.
in the liver in an inactive form.
• When activated by thrombin in the presence of calcium, Factor II (prothrombin) mutation (G20210A)
it inhibits the procoagulant activity of factor Va and A mutation in the prothrombin gene (G20210A) results in
factor VIIIa and inactivates plasminogen activator elevated prothrombin levels and carries a nearly threefold
inhibitor 1. increased risk of venous thrombosis, but does not appear to
• Deficiency may be inherited as an autosomal dominant be associated with an increased risk of arterial thrombosis
trait, or acquired as a result of warfarin therapy or severe and ischemic stroke.
liver disease (reduced synthesis), disseminated intra-
vascular coagulation and acute thrombosis. Accounts for Immunologic disorders
about 10% of cases of venous thrombosis. Antiphospholipid syndrome (see p.218).
• Resistance to activated protein C is commonly caused by
an autosomal dominant inherited mutation in the factor Prothrombotic states of uncertain cause
V gene (substitution of glutamine for arginine-506), Thrombotic thrombocytopenic purpura (TTP)
which was discovered in Leiden, the Netherlands, in • Rare.
1994; hence the inherited defect is called the Leiden • Microangiopathic hemolytic anemia, thrombocytopenia,
factor V mutation. Accounts for 15–50% of cases of fever, and renal failure are characteristic.
venous thrombosis. • Platelet microthrombi cause infarcts in many organs,
including the brain.
Protein S • Neurologic symptoms in 90% of cases, and are the
• A cofactor of protein C; free protein S increases the presenting symptoms in 60%.
affinity of protein C for phospholipid and enhances the • Fluctuating encephalopathy (confusion, disorientation,
inactivation of factors Va and VIIIa by activated protein epileptic seizures, with or without focal features), rather
C. than a stroke syndrome, is the usual presentation.
• Deficiency may be inherited as an autosomal dominant • The patient is unwell with malaise, fever, skin purpura,
trait, or acquired as a result of warfarin therapy, severe renal failure, proteinuria, hematuria, thrombocytopenia,
liver disease (reduced synthesis), and the nephrotic hemolytic anemia and fragmented red blood cells.
syndrome. Accounts for about 5% of cases of venous • Brain CT may be normal, or show infarcts, or
thrombosis. occasionally intracerebral hemorrhage, more commonly
due to heparin treatment than the disease itself.
Hereditary deficiency of coagulation inhibitors (antithrombin • Non-convulsive status epilepticus is another treatable
III, protein S, protein C), activated protein C resistance cause of altered mental status in these patients.
(factor V Leiden mutation), and hereditary abnormalities
of fibrinolysis: plasminogen deficiency and/or abnormality Cancer
• Conditions in which spontaneous and recurrent venous Laboratory abnormalities of coagulation or fibrinolysis are
thrombosis (often in legs, occasionally in brain) and commonly found, particularly in patients with metastases.
rarely, arterial thrombosis are presenting or complicating About 2% of patients with cancer have a TIA or stroke at
features. some stage.
• Usually familial (to make the diagnosis of familial Possible causes of ischemic stroke or TIA include:
deficiency, family members must be tested). • A coagulopathy mediated by intravascular mucinosis,
• The relevance of low levels of these natural low-grade disseminated intravascular coagulation, or the
anticoagulants in the etiology and prognosis (risk of patient’s chemotherapy.
recurrent stroke) for many patients with ischemic stroke • Embolism of non-infected heart valve vegetations (non-
of ‘no other cause’ is uncertain so the potential benefits bacterial thrombotic, or marantic, endocarditis).
of any treatment are uncertain. • Tumor or septic emboli, sometimes with aneurysm
• Difficulties arise attributing the cause of the stroke to one formation.
of these conditions: • Sepsis (fungi, herpes zoster, infective endocarditis).
– Familial deficiency of antithrombin III, protein C and • Hemostatic failure (leukemia, thrombocytopenia, etc).
protein S are not uncommon in the general population • Hyperviscosity syndrome (myeloma).
so the hypercoagulable state may be coincident and not • Neoplastic compression or invasion of neck arteries.
a sole or contributing cause of the stroke. Therefore, the • Irradiation of neck arteries.
224 Vascular Diseases of the Nervous System

Disseminated intravascular coagulation DISSECTION OF THE CAROTID AND


• Manifests as an acute or subacute global encephalopathy, VERTEBRAL ARTERIES
rather than stroke-like episodes, in a very sick patient.
• CT brain scan reveals widespread hemorrhagic infarcts
and hemorrhages. DEFINITION
• Low platelet count, low plasma fibrinogen, raised fibrin Tearing of the intima and/or media of arteries.
degradation products and raised D-dimer point to the
diagnosis. EPIDEMIOLOGY
• Incidence: symptomatic ICA dissection: >2.6 per 100 000;
Pregnancy and the puerperium probably higher as may cause transient minor symptoms
The relative risk of stroke in the pregnant woman is 13 times (or be asymptomatic). Vertebral artery dissection: 1–1.5
the risk in the non-pregnant woman of the same age, but the per 100 000 per year. One of the commonest causes of
absolute risk of stroke in the last trimester of pregnancy and stroke in young people, and 2% of all ischemic strokes.
the puerperium is no more than 30 per 100 000 deliveries • Age: any age; can occur in the very young.
(i.e. about 1 in every 3000 deliveries). About three-quarters • Gender: M=F.
of ischemic strokes are due to arterial occlusion and one-
quarter venous occlusion. Causes include: PATHOLOGY
• Paradoxical embolism from the venous system of the Tear in the intima or media, leading to bleeding within the
pelvis or legs. arterial wall, which tracks or dissects longitudinally and
• Valvular heart disease. circumferentially between the intima and media, or media
• Cardiomyopathy of pregnancy. and adventitia of the arterial wall. The dissection can tear
• Arterial dissection during labor. through the intima allowing the partially coagulated
• Hematologic disorders. intramural blood to enter the lumen of the artery.

Less commonly: Site


• Amniotic fluid embolism. Predominantly extracranial:
• Air or fat embolism. • Carotid dissections: the vast majority occur just above
• Metastatic choriocarcinoma. the bifurcation of the common carotid artery into the
internal and external carotid arteries; common carotid
Estrogens/oral contraceptives artery and intracranial carotid dissections are rare.
• High estrogen dose oral contraceptive use is associated Multiple dissections in 25% of patients.
with a threefold increased risk of TIAs and stroke. But • Vertebrobasilar dissections: at the C2 level in more than
the absolute risk is very small. 80% of cases, possibly reflecting increased susceptibility to
• The risk is greater in women who are older, who smoke mechanical torsion and stretch at this location. Extracranial
and who have other vascular risk factors such as dissections are commonly bilateral (60% of cases).
hypertension.
• Exogenous high doses of estrogens given to elderly men Complications
for treatment of prostatic cancer and male survivors of Pseudoaneurysms may form if the dissection spreads through
MI increase their risk of vascular death. the media of the artery; intracranial dissections may rarely
present as a mass lesion or may rupture and present with sub-
Heparin-induced thrombocytopenia arachnoid and intracerebral hemorrhage; extracranial pseudo-
Heparin may paradoxically lead to thrombus formation and aneurysms rarely rupture because the artery wall is much thicker.
thrombocytopenia by two mechanisms:
• Type I consists of a transient decrease in platelet count ETIOLOGY AND PATHOPHYSIOLOGY
1–5 days after heparin is started and is thought to be due Spontaneous
to reversible clumping of platelets. Predisposing conditions
• Type II is a persistent depression of the platelet count • Point mutation in one allele of the COL1A1 gene that
beginning 3–22 days after the introduction of heparin encodes the proα1(I) chains of type I procollagen,
which is thought to be mediated by IgG antibodies resulting in substitution of alanine for glycine (G13A) in
against a heparin–platelet membrane complex. about half of the α1(I) chains of type I collagen.
• Family history: 5% of cases.
Nephrotic syndrome • Genetic disorders of collagen.
Can be complicated by ischemic stroke, perhaps due to • Marfan’s syndrome.
‘hypercoagulability’: loss of antithrombotic proteins in the • Ehlers-Danlos syndrome types IV and VI.
urine. • Cystic medial necrosis.
• Osteogenesis imperfecta.
Desmopressin and intravenous immunoglobulin • Pseudoxanthoma elasticum.
May cause hypercoagulability and ischemic stroke, perhaps • Polycystic kidney disease.
by altering blood viscosity, hemorrheology, platelet • Fibromuscular dysplasia.
aggregability or clotting factor levels. • Reticular fiber deficiency.
• Accumulation of mucopolysaccharides.
Snake bite • Possibly atheromatous risk factors (hypertension,
May cause ischemic stroke but is more likely to cause diabetes, smoking, high cholesterol).
defibrination and bleeding. • Possibly hyperhomocystinemia.
Dissection of the Carotid and Vertebral Arteries 225

Trauma Carotid dissection


• Sports. • Pain around the eye or frontal region, sometimes in the
• Whiplash injury. neck, and sometimes generalized and non-specific, is
• Neck manipulation. common and may be the only feature.
• Activities such as reversing the car or painting the ceiling. • Acute or delayed focal monocular or carotid territory
• Iatrogenic: cerebral angiography. ischemic symptoms: ipsilateral visual loss; contralateral
hemisensori-motor deficit; difficulty speaking; onset of
Mechanisms of brain or ocular ischemia ischemic symptoms usually within a few days of
• Occlusion of the dissected artery by the false lumen or dissection but can be as long as several weeks and even a
by superimposed thrombus. few months.
• Embolism of thrombus that may have formed on the • Symptoms of single or multiple cranial nerve palsy: III,
intimal flap or in the wall. IV, V,VI, VII,IX, X, XI, XII.
• Pulsatile tinnitus may occur.
Mechanisms of cranial nerve involvement • Dysgeusia (impaired taste sensation due to VII nerve
in ICA dissection [chorda tympani] palsy) may occur.
• Compression or stretching of the cranial nerve by the
expanded artery. This is likely for lower cranial nerves IX Vertebrobasilar dissection
to XII which lie close to the ICA below the jugular • Pain.
foramen in the retrostyloid and posterior retroparotid • Focal vertebrobasilar ischemic symptoms:
space and may be compressed or stretched by an occipital/temporal lobe, brainstem, cerebellum; most
expanded or aneurysmal ICA. This is particularly so for commonly a lateral medullary or cerebellar infarct.
cranial nerves X and XII which have the longest anatomic • Upper limb peripheral motor deficits: bilateral distal
relationship to the ICA. upper limb amyotrophy.
• Interruption of the blood supply to the cranial nerve via
the nutrient vessels, which are small (200–300 microns Examination
in diameter) branches of the ICA. Mechanisms include Carotid dissection
mechanical compromise by dissection, distal • Focal monocular or carotid territory ischemic symptoms.
embolization, or pressure gradient changes in collateral • Oculosympathetic palsy (Horner’s syndrome) ipsilaterally
supply (hemodynamic). There are three vascular systems (255).
which play a significant role in nutrient supply to most • Cranial nerve palsy (single or multiple): III, IV, V, VI,
of the cranial nerves. Accepting that anatomic variations VII, IX, X, XI, XII, in at least 10% of patients with
are common, they basically involve: extracranial ICA dissection; may occur without
– The inferolateral trunk: often arises from the ICA, and symptoms of carotid ischemia.
supplies cranial nerves III, IV, VI, and the first division • Neck bruit.
of V.
– The middle meningeal system: derives from the external Vertebrobasilar dissection
carotid artery, and supplies the second and third division • Focal vertebrobasilar territory ischemic symptoms.
of cranial nerve V, and also VII. • Cervical radiculomyelopathy.
– The ascending pharyngeal system derives from the
external carotid artery and supplies cranial nerves IX DIFFERENTIAL DIAGNOSIS
through to XII. Carotid dissection
Brainstem stroke: the combination of unilateral Horner’s
Mechanism of cervical spinal cord or root syndrome and lower cranial nerve palsies with contralateral
disturbance in vertebrobasilar dissection hemisensori-motor deficit may be due to brainstem
Dissection of the vertebral arteries, which lie very close to dysfunction and draw attention to the vertebrobasilar
the spinal roots, and from which arise the anterior spinal circulation but may also be a ‘false localizing sign’ of a
artery, may lead to symptoms of a cervical radiculo- unilateral extracranial carotid artery dissection. A history of
myelopathy due to nerve ischemia or compression by neck trauma, and pain and a bruit over the carotid artery
hematoma in vessel wall. may be a clue to the diagnosis of carotid dissection.

CLINICAL FEATURES
May be asymptomatic; a history of preceding trauma is
often present, although it may be trivial and may not be 255
relevant.

History
Any combination of the following symptoms, which may be
minor and transient or more persistent.

255 Horner’s syndrome (left, arrow) due to interruption of the


ascending postganglionic pupillodilator oculosympathetic fibers in the
wall of the left internal carotid artery by dissecting blood.
226 Vascular Diseases of the Nervous System

Vertebrobasilar dissection 256


• Migraine: visual symptoms and headache due to
vertebrobasilar dissection.
• Transient global amnesia: may be presenting symptom
of dissection.
• Cervical radiculopathy.

INVESTIGATIONS
Doppler/duplex ultrasound scan (2 256)
Scans most commonly show very poor flow in the artery
giving a ‘to and fro’ high resistance signal. The appearance
may suggest a stenosed or occluded ICA or CCA with a
smooth outline. Occasionally the line of the dissection and
a double lumen can be imaged. Vertebrobasilar ultrasound
is more difficult technically and not as reliable.

MRI brain and neck


T1 images through the narrowed segment of the artery may
show a narrowed arterial lumen (as a narrower flow void) 256 Color doppler ultrasound of a localized internal carotid dissection
with a rim of high signal which is the thrombus in the with a small intimal flap (arrows).
arterial wall (257). Intramural hemorrhage is almost
pathognomonic of dissection and differentiates dissection
from vasospasm in patients with subarachnoid hemorrhage. 257
Magnetic resonance angiography (MRA)
MRA may show a stenosed or occluded ICA or CCA, or
tapering of the lumen at the dissection.

CT scan
CT may image the dissected artery but experience is limited.

Contrast intra-arterial angiography


(the gold standard)
This is often not required if duplex and MR scanning are
available.
Carotid dissection may show as a smoothly stenosed ICA
or CCA with a double lumen or intimal flap (258), or a
smooth tapering occlusion. The ‘string sign’ is due to
hematoma in the wall of the artery compressing the normal
lumen to a ‘fine thread’. Sometimes the artery is completely
occluded but the occlusive stump often has a tapered shape,
suggestive of dissection. Other angiographic findings
include intraluminal clot, intimal flaps, pseudoaneurysm
formation (usually at the base of the skull), and evidence of
distal emboli obstructing smaller intracranial arteries (259). 258 257 PDW axial MRI of the
Vertebrobasilar dissection has more variable findings; the skull base showing the carotid
string sign is less consistent and there may just be an area of canals.There is a ring of high
localized narrowing, which may be difficult to differentiate signal around the left internal
from ‘spasm’ in patients who have presented with carotid artery (arrow) which is
subarachnoid hemorrhage. Other findings include suggestive of dissection (the
outpouchings, double lumens, and irregular areas of high signal is due to the
narrowing and dilatation. thrombus within the arterial
wall), though is not specific.
Skin biopsy and collagen analysis
• Point mutation in the α1(I) chain of type I collagen.
• Fibrillin defect that produces Marfan syndrome. 258 Intra-arterial angiogram
• COL3A1 gene defect (produces disorders of type III of a localized internal carotid
procollagen in Ehlers–Danlos syndrome type I, and dissection with a small intimal
associated with multiple arterial aneurysms). flap (arrows).
• Gene for lysyl hydroxylase (results in Ehlers–Danlos
syndrome type IV).
• PKD1 gene (results in polycystic kidney disease).
• Alpha1-antitrypsin.
Central Nervous System Vasculitis 227

DIAGNOSIS CENTRAL NERVOUS


Appropriate clinical findings confirmed radiologically (with SYSTEM VASCULITIS
non-invasive doppler/duplex ultrasound and MRI/MRA,
or catheter angiography) or pathologically. Consider as a
diagnosis in all cases of young stroke, particularly if there is DEFINITION
no evidence of a source of embolism in the heart, and in the A heterogeneous group of disorders characterized by
elderly if there is no evidence of atheroma or its risk factors. histologic evidence of inflammation, and often necrosis, of
blood vessels and clinico-pathologic evidence of brain
TREATMENT ischemia or, less commonly, hemorrhage.
Uncertain as there have been no large, randomized trials:
• If symptomatic extracranial carotid dissection causing EPIDEMIOLOGY
carotid territory transient ischemic attack or mild • Incidence: rare.
ischemic stroke: immediate antithrombotic therapy with • Age: any age.
antiplatelet agent or anticoagulation (INR: 2.0–3.0) for • Gender: either sex.
4 weeks–6 months.
• If major ischemic stroke or intracranial carotid or CLASSIFICATION
vertebrobasilar dissection: no antithrombotic treatment Large arteries (aorta and its primary branches)
in view of the risk of secondary intracranial hemorrhage Takayasu’s arteritis.
and the patient has little to lose from a further ischemic
event in that arterial territory. Large and medium-sized arteries
• Surgery: uncertain role; perhaps for intracranial dis- Giant cell (temporal) arteritis.
sections which present with subarachnoid hemorrhage.
• The management of pseudoaneurysm at the base of the Medium-sized and small muscular arteries
skull remains uncertain. Primary systemic necrotizing angiitides
• Polyarteritis nodosa.
PROGNOSIS • Allergic angiitis and granulomatosis of Churg–Strauss.
• Good recovery usually if the patient presents with • Polyangiitis overlap syndrome.
headache or Horner’s syndrome. • Wegener’s granulomatosis.
• The prognosis after focal ischemic events depends on the • Lymphomatoid granulomatosis.
severity of the neurologic deficit.
• Dissections may heal in days to weeks; arterial stenosis Angiitis associated with other systemic diseases
often resolves and arterial occlusion sometimes • Sarcoidosis.
recanalizes. • Behçet’s disease.
• Pseudoaneurysms usually remain. • Relapsing polychondritis.
• Recurrent arterial dissection rate: about 1% per annum; • Inflammatory bowel disease.
about six (2–18) times greater if there is a family history • Kohlmeier–Degos disease.
of arterial dissection.
Hypersensitivity angiitis associated with connective tissue
disease
• Systemic lupus erythematosus.
259 • Mixed connective tissue disease.
• Sneddon’s syndrome.
• Rheumatoid arthritis.
• Sjögren’s syndrome.
• Scleroderma.

Primary isolated angiitis of the CNS


Isolated granulomatous angiitis of the CNS.

Other angiitis syndromes


• Eales’ disease.
• Radiation angiitis.

259 Intra-arterial DSA of an internal carotid dissection (arrow)


following a road accident. Note the occluded middle cerebral artery
(arrowhead) due to distal embolization.
228 Vascular Diseases of the Nervous System

Small vessels (arterioles, capillaries, venules) Non-immunopathogenic


Hypersensitivity angiitis • Infiltration of blood vessel wall or surrounding tissue by
• Exogenous stimuli proved or suspected: microbiologic agents.
– Drug-induced angiitides. • Direct invasion of blood vessel by neoplastic cells.
– Henoch–Schönlein purpura. • Unidentified mechanisms.
– Serum sickness and serum sickness-like reactions.
– Angiitis associated with infectious diseases. Mechanisms of tissue dysfunction
• Endogenous antigens likely to be involved: • Angiitis (causing ischemia or hemorrhage).
– Angiitis associated with neoplasms (particularly lymphoid • Coagulopathy (cf. antiphospholipid syndrome, p.218).
malignancies). • Emboli (cf. non-bacterial thrombotic endocarditis, p.216).
– Angiitis associated with other underlying diseases. • Compression from granulomas.
– Angiitis associated with congenital deficiencies of the • Antineuronal antibody effects.
complement system (hypocomplementemic angiitis).
• Mixed cryoglobulinemia. CLINICAL FEATURES
• Cutaneous angiitides. • Variable in onset, nature and duration.
• Features are determined partly by the size and location
PATHOLOGY of the involved vessel(s).
Acute, subacute or chronic inflammation in the arterial • Most commonly vasculitis presents as an acute or
and/or venous wall with or without granuloma formation subacute focal or diffuse encephalopathy or meningo-
and necrosis (260). encephalopathy with headache, altered mentation,
seizures and cognitive and behavioral abnormalities, with
Vascular lesions multifocal neurologic signs.
Granulomatous angiitis • Less commonly, patients present with a multiple sclerosis-
A distinctive chronic inflammatory reaction of blood vessels like picture (i.e. relapsing and spontaneously remitting focal
characterized by a predominance of modified macrophages neurologic dysfunction), features of a rapidly progressive
(i.e. epithelioid cells) which are aggregated into nodular space-occupying lesion, multiple cranial neuropathies (e.g.
clumps referred to as granulomas and which respond to Wegener’s granulomatosis) and rarely, with a spinal cord
foreign bodies by coalescing to form giant cells that often syndrome, extrapyramidal syndrome or stroke syndrome.
conglomerate around the foreign body. • Systemic symptoms and signs may be present such as
fever, headache, malaise, weight loss, joint aches and
Necrotizing angiitis pains, facial rash, livido reticularis.
Inflammation and necrosis (usually fibrinoid necrosis) of
vessel walls. CLINICAL HISTORY
Demographic data
Brain lesions • Age: young: Takayasu’s arteritis, SLE; older: GCA.
• Focal or multi-focal cerebral infarction. • Gender: F: Takayasu’s arteritis.
• Intracerebral hemorrhage. • Race: oriental: Takayasu’s arteritis.
• Subarachnoid hemorrhage.
Symptoms
ETIOLOGY • Headache: GCA.
A primary manifestation of disease or a secondary • Scalp, face, temporal pain: GCA.
component of another disorder such as connective tissue • Blindness: GCA, WG.
disease, drug abuse, neoplasia or infection (see • Diplopia: WG.
Classification, above). • Syncope: Takayasu’s arteritis.
• Jaw/tongue claudication: GCA, Takayasu’s arteritis.
PATHOGENESIS • Arm claudication: Takayasu’s arteritis.
Immunopathogenic • Sinus pain and drainage, nasal discharge: WG.
• In situ formation or deposition of immune complexes in • Oral ulcers: SLE, Behçet’s disease.
blood vessel wall leading to activation of the • Dry eyes, dry mouth: SS.
complement-mediated inflammatory response. • Anxiety, depression: SLE.
• Direct antibody-mediated damage via antibodies directed • Fever, headache, malaise, fatigue, weight loss, arthralgia,
at endothelial cells or other tissue components. sweats: non-specific.
• Antibody-dependent cellular cytotoxicity directed against • Skin rash: PAN, AAG, LG, sarcoid, Behçet’s disease,
blood vessels. SLE, Sneddon’s, hypersensitivity.
• Cytotoxic T lymphocytes directed at blood vessel • Malar rash: SLE.
components. • Photosensitivity: SLE.
• Granuloma formation in blood vessel wall or adjacent to • Joint aches and pains (arthritis): SLE, RA.
blood vessel. • Stiffness/aches/pains in neck, shoulders, lower back,
• Cytokine-induced (i.e. interleukin 1, TNF-alpha) hips and legs (polymyalgia): GCA.
expression of adhesion vehicles for leukocytes on • Chest pain (pleurisy, pericarditis): SLE, LG, Behçet’s.
endothelial cells. • Asthma: AAG.
• Cough, shortness of breath: LG, sarcoid, Behçet’s.
• Abdominal pain: PAN, inflammatory bowel disease,
Kohlmeier–Degos disease.
Central Nervous System Vasculitis 229

Past history • Chondritis (auricular, nasal, laryngo-tracheal): relapsing


• Allergy: AAG. polychondritis.
• Deep vein thrombosis/pulmonary emboli: SLE, APLAb, • Altered mental state, dementia, psychosis: SLE.
Behçet’s. • Blindness or altitudinal visual field defect: GCA.
• Recurrent spontaneous abortions: SLE, APLAb. • Uveitis: sarcoid, Behçet’s disease.
• Epileptic seizures: SLE. • Optic nerve head swelling (262)/pallor/hemorrhage:
• Anemia: non-specific. GCA.
• Thrombocytopenia: SLE, APLAb. • Retinal periphlebitis, hard exudates: sarcoid.
• (False) positive VDRL: APLAb. • Retinal vein occlusion: Behçet’s disease.
• Infection with toxoplasma, aspergillus, varicella-zoster, • Mononeuritis multiplex: PAN, AAG,WG, LG.
cytomegalovirus, herpes simplex virus, HIV: infectious • Temporal artery tenderness, thickening, nodularity: GCA.
arteritis. • Absent peripheral pulses: TA.
• Illicit drug abuse: amphetamine, cocaine: drug-induced • Carotid/chest bruit: TA.
arteritis. • Hypertension: TA, PAN.
• Blood pressure difficult to record or different between
Family history the arms: TA.
• Collagen vascular diseases or angiitides. • Aortic regurgitation: TA.
• Multiple spontaneous abortions: APLAb. • Pleuritic or pericardial friction rub: SLE.
• Deep venous thrombosis or pulmonary emboli: APLAb. • Chest crackles/wheeze: AAG, LG, SLE.
• Neonatal heart block in patient’s child (woman). • Arthritis: sarcoid, SLE, RA.

PHYSICAL EXAMINATION
• Malar rash: SLE.
• Skin nodules, purpura: AAG, LG, sarcoid, Abbreviations: TA:Takayasu’s arteritis; GCA: giant cell arteritis;
Kohlmeier–Degos disease, hypersensitivity. PAN: polyarteritis nodosa;AAG: allergic angiitis and
• Skin macules, papules, plaque: LG, Sneddon’s syndrome. granulomatosis of Churg–Strauss;WG:Wegener’s
• Skin infarcts (261). granulomatosis; LG: lymphomatoid granulomatosis;
• Skin fibrosis: scleroderma. SLE: systemic lupus erythematosus; RA: rheumatoid arthritis;
• Oral ulcers: SLE, Behçet’s disease. APLAb: antiphospholipid antibody syndrome; MCTD: mixed
• Ischemic necrosis of lips, palate, nasal septum: TA, WG. connective tissue disease; SS: Sjögrens syndrome
• Sinus inflammation/nasal mucosal ulceration: WG.

260 261

260 Histologic section of basal meninges showing infiltration with 262


mononuclear cells and multinucleate giant cells (arrows) in a patient
with infective arteritis of the CNS due to tuberculosis.

261 Vasculitic infarcts of the skin in a patient with polyarteritis nodosa.

262 Ocular fundus of a patient with a left optic neuropathy due to


sarcoidosis showing a swollen, congested optic disc with peripapillary
hemorrhages (papillitis).
230 Vascular Diseases of the Nervous System

DIFFERENTIAL DIAGNOSIS Causes of segmental narrowing of cerebral arteries on


Non-vascular disorders cerebral angiography
• Dementias. • Arteritis (non-infective, infective).
• Meningo-encephalitides. • Arterial dissection.
• Multiple sclerosis. • Vasospasm (drugs, subarachnoid hemorrhage, severe
hypertension, ?migraine).
Cardiac disorders • Arteriosclerosis.
• Non-bacterial thrombotic endocarditis. • Fibromuscular dysplasia.
• Cardiac tumor. • Moyamoya disease.
• Radiation.
Hematologic disorders • Leptomeningitis (infective, carcinomatous, chemical).
• Coagulopathies (paraproteinemia, thrombotic thrombo- • Multiple emboli/recanalizing embolism.
cytopenic purpura). • Intracerebral hematoma.
• Antiphospholipid antibody syndrome. • Sickle cell disease.
• Neoplasia (vascular, malignant angioendotheliosis,
Angiopathies (non-inflammatory) meningeal, glial, metastatic: atrial myxoma).
• Isolated angiopathy of the CNS. • Trauma: closed head injury.
• Arterial dissection. • Surgical manipulation.
• Vasoconstriction or due to drug use (methamphetamine • Neuroectodermal dysplasia.
or cocaine).
• Cholesterol embolization syndrome. Systemic inflammation
• Malignant angioendotheliomatosis (intravascular • Full blood count*:
lymphomatosis). – Anemia: TA, GCA, PAN, WG, SLE.
• Neurofibromatosis. – Neutrophilia: PAN, WG.
• Atherosclerosis. – Lymphocytopenia: LG, SLE.
• Fibromuscular dysplasia. – Eosinophilia: AAG, hypersensitivity.
• Moyamoya disease. – Thrombocytosis: GCA, PAN.
• Angiitis (see Classification above). – Thrombocytopenia: SLE, APLAb.
• ESR*: TA, GCA, PAN, WG, hypersensitivity.
INVESTIGATIONS • C-reactive protein*.
MRI brain* • Immunoglobulins A, G, M, E*:
For imaging brain parenchymal lesions. It may show: – Hypergammaglobulinemia: TA, WG, SLE, hypersensitivity.
• Normal brain. – Hypogammaglobulinemia: PAN.
• Areas of increased signal on T2 and PDW images typically • Complement C3, C4, C9*:
in peripheral white matter and gray matter (unlike MS – Hypocomplementemia: SLE, hypersensitivity.
where the corpus callosum is typically affected [263]). • Circulating immunocomplexes (Raji test).
There may be evidence of hemorrhage in these lesions.
Underlying connective tissue disease
Cranial CT scan may be normal or show single or Nucleic acids
multiple non-enhancing areas of low density, which may • Antinuclear antibodies*: SLE (sensitivity 95%, specificity
involve both cerebral gray and white matter. low).

Intra-arterial angiography* DNA


For imaging arterial lesions: • ds DNA: SLE (sensitivity 50–70%, specific).
• May show areas of alternate narrowing and dilatation of
intracranial arterial branches (‘beading’) (264), or areas Ribonucleoproteins
of extracranial arterial occlusion (e.g. Takayasu’s arteritis) • U1 ribonucleoprotein (RNP)*: MCTD, SLE, SS
(265). (sensitivity high, specificity low).
• In primary angiitis of the brain the small intracranial • Sm*: SLE (sensitivity 5–30%, specific).
arteries and arterioles are involved, whereas in vasculitis • Ro (SS-A)*: SS/SLE overlap (high sensitivity and
complicating meningitis, tumors or other causes the specificity for SS).
major basal intracranial arteries may be involved. • La (SS-B)*: SS/SLE overlap (high sensitivity and
However, the angiogram may be normal even in biopsy- specificity for SS).
proven vasculitis, particularly if the affected vessels are
<500 microns in diameter and too small to be seen, so DNA binding proteins
biopsy is strongly recommended (see below). • Histones: drug induced SLE.
• Magnetic resonance angiography (MRA) is not reliable • Ku: sclerodactyly.
enough to confirm or exclude vasculitis; it is too prone • PCNA: severe SLE.
to flow-related artefacts and shows insufficient detail to
image the peripheral small arteries satisfactorily. Spiral CT Cell membrane antigens
has not been widely evaluated. • Cardiolipin: APLAb, SLE (sensitivity 20–30%, specificity
low).
Central Nervous System Vasculitis 231

Other 263
• IgM rheumatoid factor-latex fixation test*: RA
(sensitivity 80%, specificity low).
• Antineutrophil cytoplasm antibodies*: WG (sensitivity
and specificity 90%).

*Important tests.

Abbreviations: TA:Takayasu’s arteritis; GCA: giant cell arteritis;


PAN: polyarteritis nodosa;AAG: allergic angiitis and
granulomatosis of Churg–Strauss;WG:Wegener’s
granulomatosis; LG: lymphomatoid granulomatosis;
SLE: systemic lupus erythematosus; RA: rheumatoid arthritis;
APLAb: antiphospholipid antibody syndrome; MCTD: mixed
connective tissue disease; SS: Sjögrens syndrome 263 MRI (T2W image) shows areas of high signal at the gray-white
matter junction (arrows) in a patient with biopsy-proven primary
cerebral angiitis.

264 265

264 Intra-arterial angiogram shows dilatation and strictures of the


small peripheral branches of the middle and anterior cerebral arteries
(arrows). (Courtesy of Dr E Teasdale, Consultant Neuroradiologist,
Institute of Neurological Sciences, Southern General Hospital,
Glasgow, UK.)

265 An arch aortogram from a patient with Takayasu’s arteritis – note


the stricture of the left subclavian (arrow) and occlusion of the right
subclavian (arrowhead) arteries. (Courtesy of Dr A Reid, Consultant
Radiologist, Royal Infirmary, Glasgow, UK.)
232 Vascular Diseases of the Nervous System

Extent of visceral and other organ involvement • The risk of serious morbidity from brain biopsy is about
• Urine: 0.5–2.0%.
– Glomerular red cells: PAN, WG, LG, SLE. • The decision to biopsy is made on an individual basis; if
– Casts*. an extended period of treatment with potentially
– Protein (if positive, 24-hour urine protein)*. hazardous immunosuppressant drugs is being
– Eosinophils. considered, then the need to establish an unequivocal
• Creatinine: PAN, WG, LG, SLE. tissue diagnosis becomes of primary importance.
• Liver function tests: transaminases*: GCA.
• Hepatitis B surface antigenemia: PAN. The results of blood and CSF laboratory tests, EEG,
• Chest x-ray* (infiltrates, nodules, cavities) (266, 267): brain imaging with CT and MRI, and cerebral angiography
WG, LG, SLE, sarcoid. are neither sensitive nor specific but are usually essential to
• Gallium scan: sarcoid. rule out infectious or malignant disease, which can mimic
• Ophthalmologic examination using low dose fluorescein CNS angiitis clinically.
angiography with slit-lamp video microscopy of the
anterior segment: slowing of flow, multifocal attenuation DIAGNOSIS
of arterioles, erythrocyte aggregates, areas of small vessel The diagnosis is made histologically. A combined
infarction, and multifocal segments of intense leakage leptomeningeal and wedge cortical tissue biopsy, preferably
from post-capillary and collecting venules. of the temporal tip of the non-dominant hemisphere and
including a longitudinally-orientated surface vessel is
Underlying infection required. If organs other than the brain are affected, biopsy
• Antibodies against borreliosis*, salmonellosis, yersiniosis, specimens should be obtained.
toxoplasmosis, aspergillus.
• Viral antibodies (including varicella-zoster virus, TREATMENT
cytomegalovirus, HIV, herpes simplex virus). The decision to treat and how will depend on the clinical
• Hepatitis screening. condition and course of the patient and the philosophies of
• VDRL, TPHA. the attending clinician. If the patient is well then time is
• Cryoglobulins*. available to allow a period of observation of the natural
• Paraproteins (serum protein electrophoresis)*. history of the disease. If the patient is very sick, however,
• CSF*: empirical immunosuppressive therapy, may be commenced
– Cell count, total protein, glucose, quantitative analysis of whilst awaiting biopsy confirmation (see below).
immunoglobulins, oligoclonal band analysis. Once the histologic diagnosis is confirmed then anti-
– Moderate mononuclear CSF pleocytosis (usually inflammatory and immunosuppressive therapy (with corti-
50–80 cells/mm3; but can be up to 800 cells/mm3). costeroids and/or cyclophosphamide) should be considered,
– CSF protein usually elevated to around 1–1.5 g/l bearing in mind that the treatment of CNS angiitis is
(10–15 g/dl), but may be up to 5.8 g/l (58 g/dl). inferred from clinical experience with systemic angiitis or
– Xanthochromia, hypoglycorrhachia and an elevated CSF anecdotal, lacking any controlled study. Start with oral or
gammaglobulin with oligoclonal banding of IgG have intravenous glucocorticoid (e.g. prednisolone 1–2 mg/kg
been reported. per day) in divided doses every 8–12 hours. After the disease
– The primary value of CSF studies is to exclude other is controlled, reduce to one morning dose, and thereafter,
causes of a meningitis. taper the daily dose as rapidly as clinical disease permits.
Ideally, patients should be slowly converted to alternate-day
Associated coagulation abnormalities therapy with a single morning dosage of short acting
• PT, APTT, dRVVT*: APLAb, SLE (35%). glucocorticoid (prednisone, prednisolone, methyl-
• Anticardiolipin antibodies*: APLAb, SLE (35%). prednisolone) so as to minimize adverse effects; prednisone
doses of 15 mg daily (or less) given before noon usually do
Others not suppress the hypothalamic pituitary axis. However, the
• Angiotensin converting enzyme: sarcoidosis. disease may flare on the day off steroids, in which case use
• Lysozyme and beta2-microglobulin: sarcoidosis. the lowest single daily dosage that suppresses disease.
• Cell-mediated immunity (anergy): sarcoidosis, LG. Strategies to minimize adverse effects of steroid include:
• Sinus x-ray/CT: WG. • Calcium (1000 mg daily) for most patients to minimize
• Echocardiography: atrial myxoma. osteoporosis.
• EEG: frequently abnormal, showing generalized non- • Vitamin D 50 000 units one to three times weekly if 24-h
specific slow wave activity of variable degree and location. urinary calcium excretion <120 mg, (monitor for
• Indium-labelled white-cell brain imaging: increased uptake hypercalcemia).
and accumulation in the brain of indium-labelled leukocytes. • Estrogen replacement therapy if post-menopausal.
• Calcitonin and biphosphonates may also be useful.
Brain biopsy • Hyperglycemia, hypertension, edema and hypokalemia
• Cerebral or spinal cord biopsy establishes the diagnosis should be treated.
in about 70% of cases. • Infections should be identified and treated early.
• False-negative biopsies occur in about 30% of autopsy- • Immunizations with influenza and pneumococcal vaccines
proven patients. are safe and should be given if the disease is stable.
• The relatively low diagnostic yield from cortical biopsy
may be related to the segmental nature of the lesions and The addition of cyclophosphamine (1.5–2.5 mg/kg per day
the lack of leptomeningeal tissue in the biopsy. orally) to prednisolone (1 mg/kg per day) may be effective.
Central Nervous System Vasculitis 233

Strategies to minimize adverse effects of cyclopho- 6 If a syndrome is recognized and if it is associated with an
sphamide include: underlying disease or an offending antigen, treat the
• Monitor the leukocyte count closely, keeping it above underlying disease or remove the offending antigen
3000 per cubic millimeter and the neutrophil count where possible.
above 1500 per microlitre. 7 Establish the histologic diagnosis of angiitis by obtaining
• If severe neutropenia or bladder toxicity occurs despite a tissue biopsy before committing the patient to a long
low doses of cyclophosphamide and a fluid intake of course of immunosuppressive medication.
>3 liters/day, azathioprine 1–2 mg/kg per day or 8 Determine the extent of disease activity.
methotrexate can be used successfully with prednisone. 9 Start treatment with appropriate agents in disorders in
• A pulsed regimen of 10–15 mg/kg intravenously once which treatment is of proven benefit and is essential (i.e.
every 4 weeks has less bladder toxicity that daily oral prednisone in temporal arteritis; cyclophosphamide and
doses (1.5–2.5 mg/kg per day) but bone marrow prednisone in Wegener’s granulomatosis).
suppression can be severe. 10 In patients with systemic angiitis, start with glucocor-
ticoid therapy. Add a cytotoxic agent such as methotrexate
When the disease has been controlled for a few months, or cyclophosphamide if an adequate response does not
taper immunosuppressive agents and attempt to discontinue result of if the disorder is likely to respond only to
them. The role of antiplatelet agents and anticoagulants is cytotoxic agents, such as Wegener’s granulomatosis.
uncertain. Campath-1H humanized monoclonal antibody 11 Avoid immunosuppressive therapy (glucocorticoids or
treatment remains experimental. cytotoxic agents) in disorders which rarely result in
irreversible organ system dysfunction and which usually
Clinical approach to the patient with do not respond to such agents.
suspected CNS vasculitis 12 Closely follow patients for development of toxic adverse
1 Is it cerebrovascular (i.e. TIA, stroke, vascular meningo- effects of treatments.
encephalopathy) or not? 13 Continually attempt to taper glucocorticoids to an
2 Where is (are) the lesion(s) neuroanatomically? alternate-day regimen and discontinuation when possible,
3 What is the pathologic nature of the lesion(s): ischemic and to taper and discontinue cytotoxic drugs as soon as
or hemorrhagic? is feasible upon induction of remission.
4 What is the etiology of the lesion(s): have more common 14 In the event of unacceptable adverse effects or lack of
disorders of the arteries (atheroma, dissection), heart and efficacy, consider alternative agents such as azathioprine.
blood been excluded?
5 Are there other clinical features or investigation results PROGNOSIS
to indicate that this is part of a specific vasculitic Variable.
syndrome (i.e. sinus disease: Wegener’s granulomatosis;
jaw claudication: temporal arteritis)?

266 267

266 Chest x-ray, posterio-anterior view, showing bilateral hilar 267 Chest x-ray, posterio-anterior view, of a patient with Wegener’s
lymphadenopathy in the patient in 262 with sarcoidosis. granulomatosis showing a loss of volume in the upper zones of the
lungs bilaterally and coarse interstitial infiltrate with confluence and
cavitation in the upper zones bilaterally.
234 Vascular Diseases of the Nervous System

GIANT CELL ARTERITIS (GCA) • Immunopathogenic mechanisms, especially cell-mediated


immunity, are probably involved. An antigen-driven
immune response is likely to be the primary event, and
DEFINITION arterial damage a secondary effect. There is no direct
A systemic angiitis that involves a wide variety of medium relationship with other connective tissue diseases.
and large arteries, and tends to affect older people (over 50
years) causing two main clinical syndromes: temporal CLINICAL FEATURES
(cranial) arteritis and polymyalgia rheumatica, which History
respond rapidly to corticosteroid therapy. Systemic
Consitutional symptoms: fever, malaise, fatigue, anorexia,
EPIDEMIOLOGY weight loss, night sweats, depression, and arthralgias.
• Incidence:
– Temporal arteritis: 18 per 100 000 per annum among Myalgic
people >50 years; higher incidence in northern than Proximal, symmetric muscle pain and stiffness of
southern Europe. polymyalgia rheumatica:
– Polymyalgia rheumatica: 13–68 per 100 000 population • Intense pain and stiffness in the neck, shoulders and
aged over 50 years per annum. buttocks.
• Prevalence: 223 per 100 000 (1 in 500) among people • Worse in the morning: patients ‘roll out of bed like a log’.
over age of 50 years.
• Race: mainly white people. Arteritic
• Gender: F>M; 2–3:1. • Headache (92% of cases):
• Age: elderly; seldom under age 60 years, extraordinarily – Due to partial occlusion of an artery.
rarely under 50 years (one case reported at age 35 years). – The site varies: may be temporal, occipital, or
generalized; severe and persistent; patients may sit up in
PATHOLOGY a chair all night.
Macroscopic – Often described as a new type of pain.
Any medium or large artery in the body (aorta, carotid, • Pain, swelling, erythema and tenderness over the affected
vertebral, coronary, femoral and so on) may be affected, but arteries (e.g. superficial temporal arteries may stand out
most commonly branches of the external carotid (superficial and be tender on brushing the hair).
temporal, facial, occipital arteries), ophthalmic, vertebral and • ‘Claudication-like’ symptoms of partial arterial occlusion:
posterior ciliary arteries, as well as the aorta and its branches. – Pain on chewing (jaw and tongue claudication due to
These are all vessels with substantial quantities of elastin in maxillary and lingual artery occlusion): up to 65% of
their walls. Curiously, other vessels with lesser amounts of cases.
elastin, such as the proximal central retinal artery, and the – Pain and blanching of the tongue (lingual artery).
petrous and cavernous portions of the internal carotid – Intermittent claudication and Raynaud’s phenomenon,
arteries, and their branches, may also be involved, but the more common in arms (‘aortic arch syndrome’).
cervical segment of the carotid artery is minimally involved • Ischemic symptoms of total arterial occlusion:
and there is a striking lack of arteritic involvement of the – Visual impairment (25–50% of cases): usually sudden,
intracranial arteries except in rare cases. Particularly painless deterioration of vision in one eye, often on
common sites of arterial stenosis in the cerebral circulation waking in the morning. It may persist.
are the internal carotid artery just before dural penetration – Neurologic problems (31% of cases): TIA of the brain or
and the extracranial vertebral artery just before entering the stroke (7%); TIA of the eye (amaurosis fugax): 10%;
skull. deafness; peripheral neuropathy.

Microscopic Examination
A panarteritis characterized by intimal proliferation and • Thickening, tenderness and nodularity of the temporal
thickening, destruction of the internal elastic lamina, and arteries, sometimes with reduced or absent pulsation
infiltration of the media by mononuclear cells (271).
(predominantly T lymphocytes of the helper/inducer • Bruits over large arteries; tenderness of arteries to
subset), giant cells, and occasional eosinophils with palpation occasionally.
granuloma formation (268–270). The granulomatous • Visual acuity varies from 6/6 to no light perception;.
changes are considered classical of GCA although the • Visual field defects; altitudinal visual field defects (loss of
presence of giant cells is not required for the diagnosis. either the upper or more commonly the lower half of the
field in one eye) are particularly common (in contrast to
ETIOLOGY AND PATHOGENESIS the central scotoma usually occurring in optic neuritis
• Unknown. due to demyelination) and are due to occlusion of the
• Genetic predisposition: upper or lower division of posterior ciliary artery, which
– Increased prevalence in Northern Europeans. supplies the optic nerve; inferior nasal sectorial defect;
– Reports of multiple family cases. central scotoma.
– Frequency of human leukocyte antigen HLA-DR4 is • Ophthalmoscopy: distended veins, a swollen optic disc
twice normal. (may be segmental), and, occasionally, cotton-wool spots
– Association with DRB1-04 variants, particularly the (272) and splinter or flame-shaped hemorrhages at or
second hypervariable region of DRB1-04. near the disc margin.
Giant Cell Arteritis (GCA) 235

268 269

268–270 Cross-section of a temporal artery biopsy. Low power 270


magnification (268) showing the lumen is occluded by extensive
granulomatous inflammatory reaction involving the total thickness of the
vessel. Higher power (269) showing the intimal surface is replaced by a
fibroblastic proliferation, occluding the lumen; and the sub-intimal zone
has extensive fibrinoid necrosis with marked disruption of the internal
elastic lamina.There is histiocytic proliferation, and nodules of eosinophil
cells and occasional multinucleated giant cells are present. In the outer
muscle and adventitial coats there is a light infiltrate of lymphocytes and
plasma cells. High power section (270) of a temporal artery biopsy
from a patient with active giant cell arteritis showing (in the center of
the field) a multinucleate giant cell with the nuclei arranged around the
periphery of the cell in a horseshoe pattern (arrow).

271 272

272 Fundus photograph of anterior ischemic optic neuropathy due to


giant cell arteritis of the posterior ciliary artery showing a swollen
optic disc, cotton-wool spots at the disc margin, and distended veins.
(271, 272 courtesy of Mr M Wade, Department of Medical
Illustrations, Royal Perth Hospital, Australia.)

271 Lateral photograph of the forehead of a patient with giant cell


arteritis showing a visibly enlarged, tender, temporal artery.
236 Vascular Diseases of the Nervous System

50–90% of patients with the clinical syndrome of INVESTIGATIONS


temporal arteritis (constitutional symptoms, headache, Blood
temporal artery tenderness, ± jaw or tongue claudication, • ESR or plasma viscosity as measures of plasma protein
anterior ischemic optic neuropathy, and TIA or ischemic changes: the most useful supporting investigation for the
stroke) present with, or develop, the syndrome of clinical diagnosis of GCA or PMR. The ESR is elevated
polymyalgia rheumatica (constitutional symptoms and above 40 mm per hour in two-thirds of patients, and is
proximal muscle pain and stiffness). often over 100 mm per hour, but can occasionally be
normal, as can the plasma viscosity. Plasma viscosity,
DIFFERENTIAL DIAGNOSIS unlike ESR, is independent of red cell morphology and
Giant cell arteritis packed cell volume, and is only slightly influenced by age
Systemic upset and sex. Other acute phase reactants, such as C-reactive
• Infection. protein, are also raised but are no more helpful than the
• Malignancy. ESR in assessing the disease or its activity.
• Myeloma. • Full blood count: a mild to moderate normochromic,
• Depression. normocytic anemia is usually present. Thrombocytosis,
due to increased production of platelets in GCA, has
Headache been associated with the occurrence of ocular and
• Brain tumor. cerebral ischemic complications of GCA but the reason
• Cervical spondylosis. is unclear. Thrombocytosis in GCA does not necessarily
reflect a more severe degree of angiitis, nor does it
Visual loss contribute to ischemic complications unless it predisposes
• Other types of arteritis (see CNS vasculitis, p.227): to thrombosis in narrowed inflamed arteries.
immune-mediated and infectious (e.g. Lyme disease). • Liver function tests: mild dysfunction, particularly
Usually different age (younger) and distribution of increased alkaline phosphatase levels.
vascular lesions. • Thyroid function tests: if fever and weight loss.
• Non-arteritic cerebrovascular disease of the brain or eye: • Creatine kinase: to rule out inflammatory muscle disease.
atheroma, dissection. • Antinuclear antibodies, C3 complement factor,
• Leber’s hereditary optic neuropathy (mitochondrial antineutrophil cytoplasmic antibody (ANCA), immune
cytopathy) (see pp.162, 491). complexes, rheumatoid factor to exclude SLE and RA.
• Multiple sclerosis (see p.340). • Blood and urine cultures: to exclude infectious arteritis.
• Serologic tests for Borrelia burgdorferi, Q-fever,
Jaw pain salmonellosis, brucellosis, syphilis, viral hepatitis.
Dental conditions.
Radiology
Facial pain Color duplex ultrasound of the superficial temporal artery:
Trigeminal neuralgia or sinus disease. the most specific sign is a dark halo around the arterial
lumen, which may be due to edema in the artery wall.
Ear pain with or without vertigo
Otologic conditions. Surgery
Temporal artery biopsy
Polymyalgia rheumatica (PMR) • This is not necessary in a straightforward case of
Systemic upset and muscle pain/stiffness polymyalgia rheumatica with no sign of temporal arteritis
• Malignancy: myeloma, leukemia or lymphoma, because blindness is uncommon in these patients.
metastatic cancer. However, always consider, and in most cases do it, in
• Arthritis: osteoarthritis of the cervical spine, RA, patients with suspected temporal arteritis. It is
connective tissue disease. particularly important to do in patients with an atypical
• Muscle disease: polymyositis, dermatomyositis: weakness presentation to avoid later doubts about the diagnosis.
is the predominant feature rather than intense muscle • Do a biopsy as soon as possible, but do not delay steroid
pain. treatment; although treatment will reduce the chance of
• Infection: viral, bacterial; osteomyelitis; miliary a positive biopsy result, positive results after starting
tuberculosis. steroids have been reported up to 1 week, and even 3–6
• Metabolic disease: hypothyroidism, metabolic bone months in some cases. A positive biopsy gives the clinician
disease. confidence to prescribe and continue long-term steroid
• Miscellaneous: fibromyalgia syndrome; Parkinson’s treatment which may have adverse effects (and avoid
disease. possible litigation).
• Take the biopsy from the clinically abnormal side. The
biopsy is positive in 60–80% of patients with TA and
15–20% of those with PMR. The sensitivity of biopsy
depends on the quality of the biopsy, particularly biopsy
length and preparation for histologic examination. If
temporal arteries on both sides appear normal, remove a
4–5 cm (1.6–2 in) length of artery from one side and
examine histologically by serial sections because arterial
lesions can be segmental.
Giant Cell Arteritis (GCA) 237

• If the biopsy is negative, and the diagnosis of GCA is still most physicians believe steroids to be necessary to achieve
suspected, the other superficial temporal artery should complete control of symptoms.
be biopsied. If both biopsies are negative, the diagnosis
of GCA still cannot be ruled out. In these cases, the Initial dose
diagnosis is clinical. • Prednisolone 40 mg (25–50 mg)/day for TA, and
• The biopsied artery with GCA shows severe intimal 15 mg/day for PMR.
thickening and reduction in vessel lumen. The internal • If neurologic or neuro-ophthalmologic symptoms such
elastic lamina is disrupted with fragmentation and as stroke or acute visual failure are present, initial dose is
sometimes destruction. There is pronounced infiltration 50–80 mg/day of prednisone.
by histiocytes, lymphocytes, epithelioid cells, and giant • Maintain for 4 weeks.
cells in the artery wall, particularly the media and intima • Reduce dose by 5 mg/day every 1–2 weeks until dose is
adjacent to the internal elastic lamina (see Pathology 10 mg/day.
above, 268–270) • Careful, frequent monitoring of symptoms remains the
• Occasional complications of temporal artery biopsy best guide to management; the ESR and C-reactive
include damage to the facial nerve, skin necrosis, protein are not always reliable markers of disease activity
drooping of the eyebrow, and stroke due to an although they correlate closely with symptoms in most
interruption of a collateral circulation. patients.
• Alternatively, methotrexate 10 mg/week (for 24
Other months) plus corticosteroid in high initial dose, tapering
• MRI brain if indicated (e.g. oculomotor paresis). quickly to cease after 4 months, is as safe as corti-
• CSF if indicated (e.g. suspected infectious arteritis). costeroid therapy alone and more effective in controlling
disease, in one recent trial (Jover et al., 2001).
DIAGNOSIS
The diagnosis of temporal arteritis and polymyalgia Maintenance dose
rheumatica is largely clinical, being based heavily on the • Prednisolone 5–7.5 mg/day for about 6–12 months.
history (there are few if any clinical signs) and the clinical • Then, if asymptomatic and ESR is normal, reduce the
response to steroids, and is largely one of exclusion. dose gradually (e.g. 1 mg/day every 2–3 months). There
Diagnosis can only be confirmed histologically, by biopsy is a fine line between too rapidly reducing the dose of
of an affected artery, usually the superficial temporal artery steroids, which leads to relapse, and too slowly reducing
(or occipital artery), and preferably one which is tender. the dose, which leads to adverse effects such as hyper-
tension, osteoporosis, skin fragility and diabetes mellitus.
Temporal arteritis (TA) • Adverse effects of steroids are related to the initial dose,
American Rheumatism Association 1990 diagnostic criteria. the total cumulative dose, and maintenance doses above
At least three of the five criteria: 5 mg of prednisone a day. Withdrawing steroids often
• Age at disease onset ≥50 years. proves difficult and judicious use of simple analgesia and
• New onset of localized headache. non-steroidal anti-inflammatory drugs may be helpful.
• Abnormal temporal artery clinically: tenderness or Despite this, most patients are still taking glucocorticoids
decreased pulse. after 2 years and up to half of them at 4 years.
• Elevated ESR (≥50 mm/hour). • In patients with biopsy-proven GCA in whom reduction
• Abnormal temporal artery biopsy. in corticosteroid dose is difficult, methotrexate
(7.5–20 mg per week) or azathioprine may allow a
The triad of temporal headache, blindness, and jaw reduction in steroid dose. Intermittent cyclical etidronate
claudication (aching of the jaw on repeated chewing caused may prevent some of the corticosteroid-induced bone
by involvement of the branches of the external carotid loss, if given when steroid treatment is begun.
artery), if present in an elderly person, is almost
pathognomonic for the diagnosis of temporal arteritis. PROGNOSIS
• Ophthalmic GCA starts as a unilateral condition but may
Polymyalgia rheumatica (PMR) become bilateral after days, months, or years.
A clinical syndrome of middle-aged and elderly patients • Between one-third and one-half of patients can stop
characterized by: steroids after 2 years.
• Pain and stiffness of the neck, shoulders and pelvic girdle. • Relapses are most likely during the initial 18 months of
• Constitutional symptoms: weight loss, fever, fatigue. treatment and within 1 year of withdrawal of steroids.
• Elevated ESR. Patients should be urged to report back immediately if
• Rapid response to small doses of corticosteroids. arteritic symptoms occur.
• Reliable predictors of relapse have yet to be identified,
Many do not have symptoms or signs of temporal although it is unusual for patients who presented with
arteritis; up to one-third have a positive temporal artery PMR to experience an arteritic relapse, unless they have
biopsy for GCA. clinical features of TA such as recent headache, jaw
claudication and abnormal temporal arteries.
TREATMENT AND PROGNOSIS
Prompt treatment with corticosteroids is necessary to relieve
symptoms of TA and PMR, and to prevent blindness in
patients with TA. The use of non-steroidal anti-
inflammatory drugs has been advocated in some cases, but
238 Vascular Diseases of the Nervous System

PRIMARY INTRACEREBRAL mutations in the amyloid precursor protein (APP) gene


HEMORRHAGE (PICH) cause the Dutch variant of hereditary cerebral
hemorrhage with amyloidosis; mutations in exon 2 of
cystatin C cause the Icelandic variant of hereditary
DEFINITION cerebral hemorrhage with amyloidosis, but patients may
Bleeding into brain parenchyma with formation of a focal not be of Icelandic origin; apolipoprotein E (Apo E) ε4
hematoma. allele may be a risk factor: present in 40% of patients.
• Vascular malformations (arteriovenous and cavernous
EPIDEMIOLOGY angiomas):
• Incidence: about 20 per 100 000 population per year; – Dural or brain, often abutting ependymal or pial surfaces
accounting for about 10% of all strokes. Slightly more or involving choroid plexus.
common among US blacks and individuals of Chinese – Most common cause in young normotensive people.
and Japanese ancestry. – Seizures and headaches commonly antedate hemorrhage.
• Age: any age, incidence increases with increasing age. – Cavernous angiomas tend to be multiple and familial.
• Gender: M=F. • Carotico-cavernous fistula.
• Hereditary hemorrhagic telangiectasia.
PATHOLOGY • Saccular aneurysms: cause 1 in 13 intracerebral hemorr-
Site hages (2 in 13 of those <65 years old), usually in
• Putamen or internal capsule (273, 274): 30%. conjunction with subarachnoid hemorrhage.
• Caudate nucleus: 5%. • Atheromatous aneurysm.
• Entire basal ganglia region: 5%. • Septic arteritis and mycotic aneurysms: most are
• Lobar (275): 30%. superficial in the cortex/subcortex.
• Thalamus: 15%. • Necrotizing angiitis of the CNS*.
• Cerebellum: 10%. • Arterial dissection.
• Pons or midbrain: 5%. • Intracerebral tumors:
– Primary (glioblastoma, oligodendroglioma, medullo-
ETIOLOGY blastoma, hemangioblastoma).
The likely cause depends on the age of the patient: – Metastases (melanoma, bronchial carcinoma, renal
arteriovenous malformations are the most common cause carcinoma, choriocarcinoma, endometrial carcinoma).
in the young, degenerative small vessel disease in middle and • Intracranial venous thrombosis*.
old age, and amyloid angiopathy in old age. • Moyamoya syndrome.
• Occult head injury*.
Raised blood pressure • Trauma:
• Acute arterial hypertension: – Usually multiple and abut on the basal brain surfaces,
– Alcohol (also antiplatelet action, and coexistent liver particularly the orbital frontal and medial and inferior
disease). temporal lobes.
– Amphetamines (may also cause a vasculitis). – A hematoma may only become visible with CT days after
– Cocaine and other sympathomimetic drugs. the injury (‘spät apoplexy’) due to delayed bleeding into
– Monoamine oxidase A inhibitors. an area of injured blood vessels.
– Exposure to extreme cold. • Hemorrhagic brain infarction: reperfusion of infarcted
– Trigeminal nerve stimulation. blood vessels and brain.
– Post carotid endarterectomy.
– Post heart transplantation. Bleeding diathesis*
– Post correction of congenital heart lesions. • Anticoagulants:
• Chronic arterial hypertension, causing lipohyalinosis and – Risk of ICH is about 1% per year.
microaneurysms (see below). – Increased risk if elderly, previous stroke, hypertensive and
if INR >4.0.
Arterial disease – Usually slowly evolving lobar or cerebellar hematomas
• Lipohyalinosis, fibrinoid necrosis and, possibly, micro- with high fatality rates.
aneurysms in small, penetrating vessels: • Antiplatelet drugs: probably a relatively minor
– The most common cause in middle and old age. contributory factor.
– Hemorrhages often occur deep in putamen (40%), caudate • Thrombolytic treatment: 0.75% of patients with MI (>2%
nucleus (8%), thalamus (15%), cerebral hemispheres (lobar) risk if elderly >65 years, low body weight <70 kg,
(20%), cerebellum (8%) and brainstem (8%). hypertensive, and given alteplase as opposed to
• Amyloid (congophilic) angiopathy* (276): streptokinase; 0.3% risk if none of these risk factors).
– The most common cause in old age, accounting for up • Thrombocytopenia.
to a third of hematomas in the elderly. • Hemophilia.
– If young or middle aged, suspect hereditary amyloid • Hereditary factor V deficiency: usually autosomal recessive.
angiopathy (Icelandic or Dutch). • Leukemia.
– Tend to be lobar, multiple and recurrent. • Diffuse intravascular coagulation.
– May be preceded by episodes of transient neurologic • Occult head injury.
defects or subarachnoid bleeding.
– May be associated with dementia, due to multiple *Causes of multiple hemorrhages in the brain parenchyma.
microinfarcts or hemorrhages or Alzheimer’s disease;
Primary Intracerebral Hemorrhage (PICH) 239

PATHOGENESIS CLINICAL HISTORY


Rupture of capillaries, arterioles, and small arteries leads to Preceding circumstances
extravasation of blood into brain parenchyma resulting in a • Neck trauma (arterial dissection).
hematoma. As the local pressure increases, surrounding • Physical activity: heavy exertion, defecation, lifting, sexual
capillaries are stretched and rupture so that the hematoma intercourse.
enlarges on its outer circumference like a rolling snowball, • Administration of recreational drugs (e.g. amphetamines).
dissecting along fibers tract pathways, until it is ultimately • Ischemic stroke (hemorrhagic transformation of an
contained by increased pressure in the surrounding tissue, infarct).
it decompresses itself by rupturing into a ventricle or the • Puerperium (choriocarcinoma, intracranial venous
subarachnoid space, or the bleeding stops due to activation thrombosis).
of the coagulation system.

273 274

273 Brain, coronal section, showing a fresh putaminal hemorrhage in a 274 Brain, coronal section, showing an old putaminal hemorrhage as a
patient with severe hypertensive small vessel disease. (Courtesy of slit-like cavity, with surrounding brown hemosiderin pigmentation.
Professor BA Kakulas, Royal Perth Hospital,Western Australia.) (Courtesy of Professor BA Kakulas, Royal Perth Hospital,Western
Australia.)

275 276

275 Brain coronal section, at autopsy showing a large lobar


intracerebral hematoma that was fatal.

276 CT brain scan from a patient with pathologically-proven amyloid


angiopathy. Note multiple hemorrhages of different ages in different
parts of the brain (arrows).
240 Vascular Diseases of the Nervous System

Onset PHYSICAL EXAMINATION


• Sudden, with symptoms and signs gradually increasing General
over seconds or minutes, or rarely hours. • Petechiae or bruising (generalized hemostatic disorder).
• Often whilst physically active. • Signs of malignant disease (clubbing [277], cutaneous
melanoma, hepatosplenomegaly, lung collapse).
Symptoms • Needle marks (drug addict).
• Altered consciousness and/or a focal neurologic deficit • Hypertension and hypertensive end organ disease
reflecting a loss of function of the site of bleeding in the (retinopathy, cardiomegaly).
brain. • Subhyaloid hemorrhages.
• Headache: localized (but may be generalized) at first, • Heart murmur (infective endocarditis).
often in occipital area, and may later become generalized,
spreading down the back of the neck. Neurologic
• Seizures are common in patients with cortical/sub- • Focal neurologic deficits: determined by site and size of
cortical hematomas. hematoma. If the hematoma remains small, no other
signs develop. If the ICH becomes large, with raised
Past history intracranial pressure, headache, vomiting and decreased
• Hemophilia. level of consciousness occur.
• Hemorrhage in other sites of the body (hemostatic • Depressed conscious level: about one-third of patients;
disorder). determined by site and size of hematoma.
• Hypertension (lipohyalinosis or microaneurysms): past • Dementia.
history of hypertension in about 50% of patients with
spontaneous ICH. See Table 32 Clinical features of primary intracerebral
• Cancer (particularly melanoma, bronchial or renal hemorrhage.
carcinoma).
• Epileptic seizures (cortical AVM, tumor, amyloid INVESTIGATIONS
angiopathy). Imaging
• Headache (AVM). The role of imaging is to (1) demonstrate the symptomatic
• Valvular heart disease (septic embolism). lesion (i.e. the intracerebral hemorrhage) and (2) identify
any underlying cause such as an AVM or tumor.
Medications/drugs
• Oral anticoagulant drugs.
• Recreational drugs such as cocaine and amphetamines.

Family history
Intracranial hemorrhage
• Hemophilia and other inherited coagulation and platelet
disorders.
• Sickle cell disease/trait. 277
• Vascular malformations sometimes.
• Saccular aneurysm sometimes.
• Hereditary hemorrhagic telangiectasia.
• Amyloid (congophilic) angiopathy: autosomal dominant
inheritance (Iceland and Holland).
• von Hippel–Lindau syndrome.
• Ehlers–Danlos syndrome.
• Pseudoxanthoma elasticum.

Dementia
• Alzheimer’s disease.
• Vascular dementia: amyloid angiopathy.

277 Finger clubbing in a patient with hemorrhage into brain


metastases from the lung (carcinoma of the bronchus).
Primary Intracerebral Hemorrhage (PICH) 241

Table 32 Clinical features of PICH


Clinical features Site of hemorrhage Likely causes
Hemiparesis (contralateral) Putamen and globus pallidus Hypertension
Hemisensory loss (contralateral) AVM
Conjugate eye deviation (ipsilateral) Tumor
Dysphasia (Dominant posterior putamen) Amphetamines/
Visual-spatial neglect (Non-dominant posterior putamen) cocaine
Moyamoya
Apathy Caudate nucleus Hypertension
Abulia AVM
Agitation Tumor
Forgetfulness Drugs
Conjugate deviation toward side of lesion Anticoagulants
Hemiparesis (Internal capsule) Moyamoya
Hemisensory loss (contralateral) Thalamus: ventrolateral Hypertension
Ataxia (contralateral) AVM
Small, poorly reactive pupils Thalamus: medial Tumor
Downward and inward deviation of the eyes Drugs
Upgaze paresis (supranuclear) Anticoagulants
Pseudo-VIth nerve palsies Moyamoya
Somnolence
Apathy
Anterograde amnesia
Dysphasia (dominant)
Visual-spatial neglect (non-dominant)
Behavioral and motor signs Lobar: frontal Amyloid angiopathy
Conjugate eye deviation toward side of lesion AVM
Hemisensory loss (contralateral)
Contralateral hemisensory loss, visual-spatial neglect, Lobar: parietal Hypertension
cognitive, and behavioral disturbance Anticoagulants
Dysphasia, agitation, hemianopia Lobar: temporal Trauma
Homonymous hemianopia Lobar: occipital Aneurysms
Endocarditis
Venous thrombosis
Coma Pons: large, central Hypertension
Quadriparesis AVM
Small, reactive pupils Anticoagulants
Bilateral horizontal gaze paresis Tumor
Spontaneous downward eye movements (ocular bobbing) Amyloid angiopathy
Hemiparesis (contralateral) Pons: unilateral, basal Hypertension
Ataxia (contralateral) AVM
Hemisensory loss (contralateral) Pons: lateral tegmentum Hypertension
Conjugate gaze palsy (ipsilateral) AVM
Internuclear ophthalmoplegia (ipsilateral) Tumor
Ataxia
Gait ataxia Cerebellum (dentate nucleus) Hypertension
Veering to one side Anticoagulants
Vertigo AVM
Vomiting Tumor
Miosis (ipsilateral) Amyloid angiopathy
VIth nerve or conjugate gaze palsy (ipsilateral) Drugs
Headache Intraventricular Hypertension
Vomiting AVM
Depressed consciousness Tumor
Bilateral extensor plantar responses Amyloid angiopathy
Asymmetric motor, sensory, or visual signs sometimes
242 Vascular Diseases of the Nervous System

Imaging the symptomatic parenchymal lesion may compress and obscure the underlying lesion.
CT brain scan Amyloid angiopathy is a common cause of lobar
Fresh blood appears denser than normal brain (whiter). This hemorrhage in the elderly. The features include:
occurs immediately, and persists for 5–10 days (278–281); • A large area of rather patchy lobar hemorrhage extending
the whiteness gradually disappears to leave a low density out to the cortical surface.
(black) area (often slit shaped) which is the cerebromalacic • Evidence of previous hemorrhage elsewhere in the brain (on
cyst (282, 283). This process varies depending on the size MRI definitely identifiable as prior hemorrhage, whereas on
of the original hemorrhage – small hemorrhages disappear CT only identifiable as a prior ‘stroke’ unless recent).
more rapidly. Once the whiteness has disappeared it is • Recurrent hemorrhagic strokes in the same or other parts
impossible to tell (on CT) if the original lesion was an of the brain, usually, but not necessarily, in an elderly
infarct or a hemorrhage. person with some degree of cerebral atrophy.

MRI brain scan These features have been seen in pathologically-proven


On MRI the appearance of blood also changes with time (284) cases of amyloid angiopathy, but the condition is probably
and is rather complex and beyond the scope of this book. under recognized in life and many hemorrhages which are
Suffice it to say that on T2W image an area of high signal due to amyloid are diagnosed simply as PICH.
(bright) surrounded by a dark ring is most likely to be blood.
On T1W imaging, concentric bright and dark rings around a Cerebral angiography
lesion should suggest hemorrhage. The blood breakdown • Role: to detect underlying arteriovenous malformations,
products are visible on MRI for years after the hemorrhage, saccular aneurysms and intracranial venous thrombosis
allowing late distinction of the nature of the lesion (285). that are amenable to specific treatment.
However, timely use of CT soon after the stroke is surely better • Indications:
patient management than ‘salvage’ by MRI. The one caveat to – Younger than 50 years of age (provided fit for intervention).
the use of MRI very early after stroke is that occasionally it may – White matter, subcortical and superficial hematomas; i.e.
be very difficult to distinguish hemorrhage from infarct, though the pattern of hemorrhage is compatible with a saccular
in practice this is probably not often a problem. aneurysm (in that situation the angiogram should be
performed as quickly as possible) or vascular
Identifying the underlying cause malformation, or MR scanning shows ‘flow void’
CT and MRI brain scan phenomena consistent with arteriovenous malformations.
Some indication of the likely cause may come from the site and – The patient is not hypertensive.
shape of the hemorrhage. Primary hemorrhages most • Timing: angiography rarely needs to be urgent, re-
commonly arise in the basal ganglia and may track into the bleeding is uncommon and often after months or years,
lateral ventricles. They are not usually closely related to any if at all. The only important exception is when a saccular
large vessel. They can also be lobar or located in the posterior aneurysm is suspected and, even then, MR angiography
fossa. Close inspection should be made for calcification may be sensitive enough. Repeating angiography after a
(suggests AVM or tumor), multiple masses elsewhere in the negative first study may still uncover small arteriovenous
brain (suggests hemorrhage into a metastasis), serpiginous malformations in 10–20% presumably because at the
vessels visible after i.v. contrast (suggests AVM), close proximity initial study the lesion was being compressed by mass
to a major artery such as the MCA (suggests an aneurysm). effect from the hematoma.
The lack of visibility of any of these does not exclude their • Risks: arterial dissection and contrast hypersensitivity are
presence, particularly in the early stages when the mass of blood among the greatest dangers.

278 279 278 Plain (non-contrast) cranial


CT scan on the day of stroke
onset showing a homogeneous
area of high attenuation,
representing an acute
hematoma, in the right posterior
putamen.The surrounding rim
of low density is mild brain
edema.

279 T2W MRI showing a


right basal ganglia hemorrhage
10 days after stroke arrest.
Note the increased (bright)
signal centrally (methemoglobin)
and the dark low signal ring
encircling this due to
hemosiderin (compare
with 285).
Primary Intracerebral Hemorrhage (PICH) 243

280, 281 CT brain scans of a large 280 281


recent right temporal hemorrhage in
a patient on warfarin. Note the
appearance and extension into the
lateral ventricles.

282 Plain cranial CT scan in the


same patient as in 278 14 days after
stroke onset showing a residual low
density area in the right posterior
putamen, representing a resolved
hematoma.This sequence of CT
scans shows how difficult it can be
to distinguish radiologically cerebral 282 283
infarction from hemorrhage if the
CT scan is not done early.

283 CT brain scan (with contrast)


of a 2 week old frontal hematoma
(arrow).This patient had presented
with several days of headache and
confusion. Note that the ventricles
are also dilated indicating
hydrocephalus.The cause was a
subarachnoid hemorrhage due to
rupture of an anterior
communicating artery aneurysm
with a focal frontal hematoma and
secondary hydrocephalus. Note that
there is no longer any ‘whiteness’ to
identify the hematoma as blood, and
it just looks like a small mass lesion.
The contrast was given because the
initial diagnosis was of a tumor.

284 MRI (T2W) of a recent 284 285


hemorrhage in the left thalamus.The
dark central area is due to the
deoxyhemoglobin in intact red
blood cells and indicates that the
hemorrhage is only a few days old.

285 MRI (T2W) of a patient at


1 year after a severe head injury.The
black areas in the right frontal lobe
(arrow) are hemosiderin indicating
previous brain hemorrhage, in this
instance traumatic.The black areas
will persist indefinitely and are quite
specific for hemorrhage, thus MRI
can be used to identify hemorrhage
a long time after the event when CT
cannot.
244 Vascular Diseases of the Nervous System

N.B. Vascular malformations and aneurysms may still be • Intraventricular recombinant tissue plasminogen activator
the cause of hemorrhages in the basal ganglia or in the for lysis of intraventricular hemorrhage (experimental).
posterior fossa in patients over 65, and patients with pre- • Prevention of recurrence:
existing hypertension. – Diagnosis and control of hypertension.
– Surgical resection or embolization of arteriovenous
Blood tests malformation.
• Full blood count. – Surgical clipping or coiling of intracranial aneurysm.
• ESR.
• Coagulation studies: platelet aggregation, APTT, PT, CLINICAL COURSE
thrombin time (TT), INR. Intracerebral bleeding may continue for 24–48 hours after
• Autoantibody screen. onset, and appears more likely in patients who have a
• Blood DNA analysis by PCR: amplification of exons 16 bleeding diathesis (e.g. low platelet counts and low levels of
and 17 of the APP and exon 2 of cystatin C. fibrinogens, liver disease) and irregularly shaped hematomas.
Early deterioration in the first week may also be due to
DIAGNOSIS recurrent hemorrhage, hydrocephalus (if the hematoma
• Suspect when focal neurologic signs develop over a few obstructs CSF outflow or blood has entered the CSF),
minutes, without preceding warning attacks, in a person epileptic seizures, hypoglycemia, hypoxia (e.g. pneumonia,
with risk factors for bleeding such as hypertension, PE), and electrolyte imbalance (e.g. hyponatremia).
bleeding diathesis, and drug use.
• Confirmed by CT or MRI brain scan or at autopsy. PROGNOSIS
• Etiologic diagnosis (i.e. cause of the hemorrhage) Death and disability
determined by the site of hemorrhage, age of the patient, About 25% of patients die during the first day, and 50%
and other associated clinical and laboratory features. within the first month, usually as a consequence of
supratentorial hemorrhage large enough to cause
TREATMENT transtentorial herniation, or hemorrhage in the posterior
• General support (see Stroke, p.196). fossa causing direct brainstem compression and herniation
• Compression stockings, if required, for prevention of upwards and downwards. Amongst survivors, the outlook
DVT and PE; the safety of heparin and aspirin is for improvement in neurologic deficits is better for PICH,
unknown in patients with PICH. which separates and disconnects normal brain tissue, than
• Cease any antihemostatic drugs. for brain infarction of similar size, which results in tissue
• If anticoagulant-related hemorrhage, consider neutraliz- destruction. About half of survivors regain independence.
ation with intravenous vitamin K 10–20 mg (no more Survival and functional outcome depends largely on five
than 5 mg/minute) followed by infusion of a concen- factors:
trate of coagulation factors II, VII, IX, X or fresh frozen • Location of the hematoma: worse outcome with
plasma; infusion of the factors alone restores the coagula- posterior fossa, thalamic and putaminal hematoma.
tion system more rapidly than whole plasma and is safer • Size of the hematoma: >2 cm (>0.8 in) diameter on CT
from the point of view of transmission of virus particles. associated with raised intracranial pressure; >4 cm
• If fibrinolytic-related hemorrhage, control any (>1.6 in) diameter on CT associated with death unless
hypertension and infuse cryoprecipitate; the use of decompressed.
antifibrinolytic drugs is controversial. • Level of consciousness on admission (e.g. Glasgow coma
• Other bleeding diatheses should be treated specifically scale): stupor or coma is a grim prognostic sign except in
when possible. thalamic hemorrhage.
• Blood pressure should be lowered gently when it is very • Later progression of neurologic signs and development
high (e.g. >26.7 kPa (200 mmHg) systolic, >16 kPa of raised intracranial pressure.
(120 mmHg) diastolic). • Age of the patient: worse prognosis in the elderly.
• Reduce intracranial pressure if elevated: osmotic agents
such as mannitol (20–25% solution, 0.75–1 g/kg initially Recurrent intracerebral hemorrhage
then 0.25–0.5 g/kg every 3–5 hours depending on the • About 7% of 30-day survivors suffer a recurrent stroke
clinical findings, osmolality and central venous pressure). in the first year, of which at least 25% are hemorrhagic.
Hyperventilation only has a role in temporarily • About 70% of recurrences are fatal.
controlling intracranial pressure for patients in whom • Lobar hemorrhage at the junction of the gray and white
surgical evacuation is planned. Corticosteroids are more matter, often due to amyloid angiopathy, and poorly
hazardous than effective. controlled hypertension are risk factors for recurrence.
• Ventricular drain if symptomatic hydrocephalus.
• Surgical decompression and evacuation of a superficial
lobar or cerebellar hematoma which is causing
progressive neurologic deterioration and/or impairment
of consciousness may have a role in a patient who might
otherwise recover, but more evidence is needed from
large randomized trials. Surgical intervention in the
presence of cerebral amyloid angiopathy may be
hazardous because of brittleness and lack of contractility
of the sclerotic vessels, but this remains to be confirmed.
Arteriovenous Malformation (AVM) 245

ARTERIOVENOUS MALFORMATION • Atrophic changes develop in the adjacent nervous tissue,


(AVM) because of direct pressure and ischemia, due to vascular
steal.
• Associated vascular anomalies:
DEFINITION – Saccular cerebral aneurysms coexist in about 15%
Congenital anomalies that consist of a complex tangle of (6–58%) of patients. They may occur in vessels remote
abnormal arteries and veins which lack an intervening from the lesion, proximally or distally along the course
capillary bed but are linked by one or more fistulas. of feeding arteries, or within the nidus of the
malformation itself. At least two-thirds of associated
EPIDEMIOLOGY aneurysms occur on the proximal feeding arteries of the
The most common form of vascular malformation in the AVM or within the substance of the nidus. The
brain and spinal cord: pathogenesis of related aneurysms is believed to be either
• Incidence: 1 per 100 000 per year. a consequence of high flow through the AVM or due to
• Point prevalence: 18 per 100 000. a shared developmental abnormality that weakens the
• Age: symptom onset at any age, but typically before the cerebral vasculature.
age of 40 years. – Anomalous patterns of venous drainage: kinking, ectasia,
• Gender: M=F. and venous aneurysms are frequent and may compress
adjacent brain tissue, lead to venous thrombosis, or even
PATHOLOGY rupture.
An abnormal fistulous connection, without an intervening
capillary bed, between one or more hypertrophied feeding Sequelae
arteries and dilated draining veins (286). The small arteries • Intracerebral hemorrhage (about 2% per year).
have a deficient muscularis: • Subarachnoid hemorrhage.
• Location: in the dura, surface of the brain or spinal cord, • Obstructive hydrocephalus (287).
or in brain or spinal cord. • Venous hypertension.
• Size: half are small (<2.5 cm [<1 in] diameter); one-third • Mass effect.
are medium sized (2.5–5 cm [1–2 in]) and one-fifth are • Carotico-cavernous fistula.
large (>5 cm [>2 in]). Most seem to grow during life but • Epileptic seizures (about 1% per year).
some do not and others shrink or disappear, probably as
a result of hemorrhage. ETIOLOGY
• Blood supply: principally from parenchymal arteries, Developmental derangements occur at the embryonic stage
branches of the internal carotid and vertebrobasilar of vessel formation, at the fetal stage, or after birth.
systems, but about one-quarter receive contributions
from extracranial carotid or vertebral arteries. Some
arteries that supply an AVM continue beyond the fistula
to supply adjacent brain tissue (so-called en passage
feeders). The fistulas allow high flow, rapid arterio-
venous shunting thereby inducing arterial hypotension
in vessels feeding the malformation and neighboring
areas of brain.

286 287

286 Histologic section of an arteriovenous malformation showing a


network of tangled thin walled blood vessels interposed between
arteries and veins and which do not have the structure of normal 287 Brain section, coronal plane, showing a large arteriovenous
arteries or veins. malformation in the left medial frontal lobe and dilated lateral
ventricles. Hydrocephalus may result from involvement of the vein of
Galen, ventricular compression, or the meningeal fibrosing effect of
subarachnoid hemorrhage.
246 Vascular Diseases of the Nervous System

CLINICAL FEATURES – Meningism.


As few as 12% of AVMs are symptomatic during life. – Root pain.
Symptoms include a combination of: – Signs of acute cord compression if localized hematoma.
• Sudden focal neurologic deficit due to: • Spinal bruit.
– Intracerebral hemorrhage (see p.238): the most common
form of presentation (about half [30–82%] of cases). Diagnosis
Accounts for about 2% of all strokes. Risk factors for • Myelography (previously).
intracerebral hemorrhage: small AVM; exclusively deep • MRI (now the initial investigation of choice).
venous drainage; high intranidal pressure (high pressures • Spinal angiography.
in feeding arteries or restriction of venous outflow).
– Subarachnoid hemorrhage (see p.249). Treatment
• Epileptic seizures: partial or secondary generalized (see If AVM is causing progressive neurologic dysfunction or has
p.69): a common presentation (about one-third bled, consider:
[16–53%] of cases). • Embolization.
• Transient focal neurologic deficits, resembling transient • Surgical resection.
ischemic attacks, due to vascular steal or
microhemorrhage; more common with brainstem AVM Sturge–Weber syndrome (see p.151)
(about 10% of cases). Rarely (4–8% of cases) are the focal
neurologic symptoms progressive. Hereditary hemorrhagic telangiectasia
• Headache: a presenting symptom in about one-fifth (Osler–Rendu–Weber syndrome) (see p.153)
(7–48%) of patients. The headache is often non-specific,
and rarely ‘migrainous’ in occipital lobe AVMs. DIFFERENTIAL DIAGNOSIS
• Self-audible bruit, particularly with dural AVM. Other vascular malformations
• Progressive neurologic deficit caused by the mass effect Venous malformation
of the AVM (rare). • Consists of a collection of venous channels of various sizes.
• Hydrocephalus (obstructive, due to mass effect of the • Presents usually with small, low pressure, intracerebral or
AVM; or communicating, due to subarachnoid intraventricular hemorrhage that carries little morbidity.
hemorrhage).
• Intracranial hypertension, with headache or visual Cavernous malformation
symptoms, due to drainage of the malformation into the • Sharply circumscribed lesions, consisting of thin-walled
superior sagittal sinus, causing venous hypertension. sinusoidal vessels, that are of varying size and are often
• Orbital tumor (mass lesion in orbit). multiple (288).
• Carotico-cavernous fistula (ruptured dural AVM): • May be familial.
sudden onset of unilateral pulsating exophthalmos, often • Present usually with epileptic seizures (partial or
with monocular visual loss, a self-audible orbital bruit, secondary generalized) and rarely with small, low
orbital pain, chemosis, and involvement of cranial nerves pressure, intracerebral or intraventricular hemorrhage.
III, IV, VI, and the first (and sometimes second) division
of cranial nerve V. Telangiectases
Collections of dilated capillaries which usually have no
SPECIAL FORMS clinical significance unless as part of hereditary hemorrhagic
Spinal arteriovenous malformation telangiectasia (289).
Epidemiology
• May present at any age. Arteriovenous fistulas
• M>F (2:1). • Trauma.
• Occlusion of venous sinus or sinuses with formation of
Pathology neovascular collaterals.
• Extramedullary more common than intramedullary • Occlusion of branch arteries with formation of arterial
location. collaterals.
• Complications of AVM: spinal cord compression; venous
infarction; vascular ‘steal’ from the cord to the AVM; low INVESTIGATIONS
pressure intramedullary hemorrhage; subarachnoid CT brain scan
hemorrhage. Non-contrast scan may show calcification or hemorrhage or
occasionally enlarged vessels; contrast-enhanced scan may
Clinical features reveal enhancement of the AVM and serpiginous vessels.
• Spinal cord syndrome:
– Gradual onset and progressive course is common but MRI brain scan
onset may be acute. MRI is more sensitive; it can demonstrate flow voids of
– Back pain is common. multiple serpiginous arteries and large draining veins (290).
– Myelopathy, signs may indicate a rather long cord lesion The size of the AVM (i.e. small) and site of venous drainage
over several levels. (i.e. deep), as detected by MRI, increase the risk of a first
– Neurologic deficit may be exacerbated by physical (incident) hemorrhage.
exercise.
• Spinal subarachnoid hemorrhage: Intra-arterial angiography
– Sudden back or neck pain. The gold standard for demonstrating AVMs and their
Arteriovenous Malformation (AVM) 247

supply arteries and draining veins (291, 292). Typically 288


early venous drainage is seen due to the rapid flow through
the abnormal arteries. There are multiple, closely meshed
tortuous arteries which may arise from several major arteries.
AVMs may be very small or very large.
Dural AVMs are supplied by the external carotid artery
or dural branches of the vertebral artery and may
anastomose with the intracranial supply. They typically lie
on the surface of the brain.
Angiography should be delayed (if possible) after a
hemorrhage as there is more chance of seeing the full extent
of the AVM once the hematoma has resolved and is no
longer compressing and obliterating the AVM (though
obviously if emergency surgery is necessary an angiogram
should be done before that).
A high intranidal pressure is a risk factor for incident
hemorrhage. The pressure of the feeding arteries can be
measured directly only during microcatheter (superselective)
angiography. It can crudely be estimated during routine
angiography by assessing the size of the feeding arteries or
by determining how long it takes for contrast medium to
pass through the AVM.

288 T2W MRI of a right 289 290


temporal cavernous
hemangioma (arrow). Note
the areas of bright and dark
signal which are characteristic
of old hemorrhage. Flow
voids are not usually seen
because the vessels are too
small.

289 CT scan with contrast


of a right parietal capillary
hemangioma. Note the
slight calcification (arrow)
and peripheral site.The
appearance was similar
pre-contrast.

290 MRI scan, PDW


image, in an 18 year old
female with a first ever
partial motor seizure
involving the left face and
arm. Note the black flow 291 292
voids (arrows) in the small
arteriovenous malformation
in the right frontal lobe
cortex.

291, 292 Intra-arterial


digital subtraction
angiogram of an AVM. Early
film (291), late film (292).
Note the enlarged tortuous
mesh of arteries (arrow)
and early draining vein
(arrowhead).
248 Vascular Diseases of the Nervous System

MRA and spiral CT may show the AVM but usually the TREATMENT
lesion is visible on MRI (293, 294) or contrast CT and neither Treatment decisions are based on the risk of bleeding for
angiographic technique supplies enough information to replace each individual AVM and the risks of the proposed
conventional angiography, so are probably not of much help. treatment. Advances in anesthetic and microsurgical
N.B. All forms of imaging may fail to show small, techniques have resulted in substantial reductions in
compressed or obliterated vascular malformations. operative morbidity. Operative mortality is now less than 1%
in carefully selected patients.
DIAGNOSIS
Clinical clues to the diagnosis Conservative
• Known hereditary hemorrhagic telangiectasia or • AVMs discovered incidentally in the elderly.
Sturge–Weber syndrome. • Large or critically located AVMs:
• Subarachnoid hemorrhage with a past history of seizures. – Control hypertension.
• Carotid, orbital or skull bruit in a patient presenting with – Avoid antithrombotic agents, including aspirin.
headache, seizures or intracranial hemorrhage. – Anti-epileptic drugs for symptomatic seizures.
– Elective cesarean section at 38 weeks gestation for
Diagnosis is established by MRI or angiography pregnant women with AVM.

PROGNOSIS Surgical excision


• The course is difficult to predict: the AVM may remain Superficial (and thus surgically accessible), small AVMs that
static, grow, or even regress. link a single cortical arterial branch through a shunt to a
• The long term crude annual case fatality is 1–1.5%. cortical vein are the best surgical candidates.
• The annual risk of first (incident) bleeding is about 2–3%
and is similar in patients with or without a previous Spetzler and Martin (1986) classification for evaluating the
hemorrhage. risk of surgery in patients with AVMs:
• The case fatality for a first bleed is about 10–15%, and Graded feature Points assigned
the overall morbidity is about 50%. Size of AVM:
• The annual risk of future re-bleeding is probably higher, Small (maximal diameter <3 cm [<1.2 in]) 1
particularly in the first year after initial hemorrhage, and Medium (maximal diameter 3–6 cm [>1.2–2.4 in]) 2
one study has suggested that the risk is as high as about (maximal diameter >6 cm [>2.4 in]) 3
18% per year (among patients who have had a hemorrhage
Location:
at initial presentation). The risk of re-hemorrhage appears
Non-eloquent area of brain 0
to be greater in patients over 60 years, if there is an
Eloquent area of brain* 1
associated saccular aneurysm (the risk is about 7% per year),
if there is only a single draining vein, or if venous drainage Pattern of venous drainage:
is impaired or confined to the deep venous system. Superficial only 0
• For untreated AVMs, the annual risk of developing de Any deep 1
novo seizures is 1%.
*Sensorimotor, language, or visual cortex; hypothalamus or thalamus;
internal capsule; brain stem; cerebellar peduncles; or cerebellar nuclei.

293 294 293 T2W MRI of an AVM


in the left occipital lobe.
Note the dark areas
(arrows) which are ‘flow
voids’ marking the
abnormal arteries.

294 PDW MRI of a left


temporal AVM. Again note
the serpiginous dark
structures which are flow
voids in abnormal arteries
(arrows).
Subarachnoid Hemorrhage (SAH) 249

The assigned grade of the AVM (1–5) corresponds SUBARACHNOID HEMORRHAGE (SAH)
numerically to the cumulative score. A score of 4 or 5 is
associated with the highest risk of persistent neurologic
deficits after surgery. DEFINITION
The spontaneous extravasation of blood into the
Stereotactic radiosurgery (radiotherapy) subarachnoid space when a blood vessel near the surface of
Used for small (<3 cm [<1.2 in] diameter), deep AVMs the brain leaks. It is a condition, not a disease, that can be
<2.5–3 cm (1–1.2 in] in diameter. Radiotherapy is effective produced by many causes.
in obliterating about 80–90% of these AVMs within a latency
interval of 2–3 years. EPIDEMIOLOGY
A stereotactic frame is screwed into the skull vault. • Incidence: 6–8 per 100 000 person years outside
Cranial CT scan and cerebral angiogram are undertaken Finland; almost three times higher in Finland.
with the frame in place. Using the frame and coordinates • Age: typically over 40 years of age.
the location of the nidus can be plotted. It may involve the • Gender: F>M = 1.6 (1.1–2.3):1.
use of a gamma knife, proton beam, or linear accelerator. A
single session therapeutic radiation dose is applied to the PATHOLOGY
nidus and a little of the penumbra, via multiple focused Blood in the subarachnoid space, which may be diffuse or
beams to cause vascular injury, subsequent thrombosis, and localized and may have extended into the brain parenchyma.
delayed obliteration of the AVM. This may cause some Most commonly, subarachnoid blood is maximal adjacent
necrosis of underlying normal brain tissue; the amount to a ruptured saccular aneurysm at the base of the brain
depends on how large the penumbra is. The fact that (295–297). In 10% of cases, the center of the hemorrhage
radiotherapy takes months to years to have an effect is unlike is around the midbrain (perimesencephalic), usually ventral
other treatments. As a result, the major disadvantage is that to it, but there is no other pathology; the angiogram is
patients remain at risk for hemorrhage during the latency
interval until the AVM obliterates.
295
Endovascular embolization with rapidly
polymerizing liquid glues
Preferable for deep and large (>3.5 cm [1.4 in]) lesions,
often in combination with open surgery.

295 Section of a saccular aneurysm at the base of the brain, taken at


autopsy, showing disruption of the internl elastic lamina and
hemorrhage.

296 Section of the brain in the axial plane showing subarachnoid


hemorrhage in the anterior interhemispheric fissure and intraventricular
extension of the
hemorrhage due to a 296 297
ruptured anterior
communicating artery
aneurysm. (Courtesy of
Professor BA Kakulas, Royal
Perth Hospital,Western
Australia.)

297 Photograph of the


base of the brain at
autopsy, with the right
temporal lobe resected,
showing a ruptured
aneurysm of the right
middle cerebral artery
(arrow) and blood
in the Sylvian fissure.
(Courtesy of Professor
BA Kakulas, Royal Perth
Hospital,Western
Australia.)
250 Vascular Diseases of the Nervous System

normal, showing no evidence of an aneurysm and the • Dural arteriovenous fistula.


patients do not come to autopsy: they recover and go on to • Spinal arteriovenous malformation.
live a normal life. Rarely, other vascular pathologies, such as • Saccular aneurysm of a spinal artery.
arterial dissection, are identified. • Head trauma: may cause SAH and may also be a
consequence of SAH.
Saccular aneurysms, or berry aneurysms: • Mycotic aneurysms.
• Small, thin-walled blisters protruding from the arteries • Metastasis of cardiac myxoma.
of the circle of Willis or its major branches (298). • Cocaine abuse.
• Located at bifurcations and branchings: • Sickle cell disease.
– Anterior communicating artery complex: 41%. • Coagulation disorders.
– Internal carotid artery: 31%; origin of the posterior • Pituitary apoplexy.
communicating artery from the stem of the ICA; • Spinal meningioma.
terminal bifurcation of the ICA, into the middle and • Rupture of circumferential artery of the brain stem.
anterior cerebral arteries.
– Middle cerebral artery: first major bifurcation: 18%. CLINICAL FEATURES
– Posterior circulation: tip of basilar artery: 10%. Symptoms
• Vary in size from 0.2–3 cm (0.1–1.2 in) in diameter; • Sudden loss of consciousness (one-half of patients).
mean 7.5 mm (0.3 in). • Headache (two-thirds of patients): may be the only symptom.
• Vary in form: round with a narrow stalk, broad based – Onset: sudden, in a split second, like a ‘blow’ or
without a stalk; narrow cylinders. ‘explosion’ on/in the head: gradual over minutes rather
• Aneurysms are multiple in about 25% of cases. than seconds in about 10–15%; maximal in seconds.
• Usually rupture through the dome of the aneurysm. – Location: diffuse and poorly localized, but may be
• The risk factors for the formation and the rupture of occipital or retro-orbital.
aneurysms probably differ. The formation of aneurysms – Severity: unusually severe, ‘the worst headache in my life’.
if probably due to a developmental or acquired (possibly – Precipitants: uncommon; weight lifting, sexual intercourse.
due to hypertension, smoking and atheroma) defect in – Duration: hours (possibly minutes) to weeks, may be
the media and elastica of the arterial wall; perhaps focal biphasic in SAH due to vertebral artery dissection.
destruction of internal elastic membrane by hemo- – It is not clear how frequently patients with aneurysmal
dynamic forces at the apices of the bifurcations. SAH have had preceding and unrecognized ‘warning
• The rupture of an aneurysm may be due to acute rises in leaks’.
blood pressure, such as with heavy physical exertion and • Nausea.
sexual intercourse, but this remains to be established. • Vomiting.
• Neck stiffness: takes about 3–12 hours after onset to
ETIOLOGY AND PATHOPHYSIOLOGY develop, in response to blood in the subarachnoid space
Saccular aneurysms (85% of cases) which acts as a meningeal irritant.
Conditions associated with saccular aneurysms • Pain and stiffness in the back and legs, hours to days after
• Disorders of connective tissue: onset: blood irritating lumbosacral nerve roots.
– Marfan’s syndrome. • Photophobia.
– Ehlers–Danlos syndrome type IV. • Epileptic seizures: uncommon.
– Pseudoxanthoma elasticum. • Family history of SAH: 6–9% of patients. In first-degree
– Alpha1-antitrypsin deficiency. relatives of patients with SAH, the risk of SAH is three
– Neurofibromatosis. to seven times higher than in second-degree relatives or
• Disorders of angiogenesis: hereditary hemorrhagic in the general population.
telangiectasia.
• Associated hypertension: Signs
– Coarctation of the aorta. • None (very often).
– Polycystic kidney disease: hypertension and develop- • Meningism (but absence of neck stiffness does not
mental factors contribute to the development of intra- exclude SAH).
cranial aneurysms. • Focal neurologic signs:
• Hemodynamic stress: – IIIrd nerve palsy: aneurysm of internal carotid artery, at
– Anomalies of the circle of Willis. the origin of the posterior communicating artery; rarely
– Arteriovenous malformations. aneurysm of the basilar artery or superior cerebellar
– Moyamoya syndrome. artery.
– VIth nerve palsy: non-specific rise of CSF pressure.
Non-aneurysmal perimesencephalic hemorrhage – IX–XIIth nerve palsy: compression by subadventitial
(10% of cases) vertebral artery dissection.
– Hemiparesis: large subarachnoid clot in Sylvian fissure
Arterial dissection (MCA aneurysm).
– Paraparesis: aneurysm of anterior communicating artery.
Rare conditions – Cerebellar ataxia, Wallenberg’s syndrome, or both:
• Cerebral arteriovenous malformation: very few rupture vertebral artery dissection.
only into the subarachnoid space; most form an – N.B. Intraparenchymal hematomas may give rise to other
intracerebral hematoma with or without extension into deficits, depending on their site.
the subarachnoid space. • Subhyaloid hemorrhages in optic fundi (299) (20%).
Subarachnoid Hemorrhage (SAH) 251

• Fever ( if due to blood in subarachnoid space, the heart rate • Carotid or vertebral artery dissection.
is normal; if due to infection, the heart rate is increased). • Intracranial venous thrombosis.
• Raised blood pressure. • Occipital neuralgia.
• Kernig’s sign: passive extension of the knee with the hip • Acute obstructive hydrocephalus.
flexed elicits pain in the back and leg and resistance to
hamstring stretch. Primary intraventricular hemorrhage: causes
• Brudzinki’s sign: forward flexion of the neck elicits Uncommon aneurysms
flexion at the hip and knee. • Posterior inferior cerebellar artery.
• Altered consciousness. • Anterior inferior cerebellar artery.

DIFFERENTIAL DIAGNOSIS Arteriovenous malformations


With neck rigidity • Ependymal lining.
• Acute painful neck conditions: bony and ligamentous • Choroid plexus.
injury, extrapyramidal rigidity, cervical lymphadenitis. • Dural fistula of the superior sagittal sinus.
• Meningitis/encephalitis.
• Stroke: ischemic or hemorrhagic; cerebellar or Occlusive arterial disease
intraventricular (see below). • Moyamoya syndrome: idiopathic, atherosclerotic, or with
• Recent head injury. associated aneurysm.
• Lacunar infarction.
Without neck rigidity
If acute severe headache (1 minute–1 hour) is the only Tumors
symptom, the chance of SAH is about 12% and of a Pituitary tumor; ependymoma; meningioma.
ruptured aneurysm is about 6%. Other diagnoses include:
• Migraine. Infectious diseases
• Thunderclap headache. Brain abscess; parasitic granuloma.
• Pressor responses.
• Benign orgasmic cephalgia. Drugs
• Benign exertional headache. Cocaine; amphetamine.
• Reaction while on monoamine oxidase inhibitors.
• Pheochromocytoma. Wernicke’s encephalopathy
• Expanding intracranial aneurysm.

298 299

299 Ocular fundus of a patient with subhyaloid hemorrhage,


appearing as sharply demarcated linear streaks of brick red-colored
blood or flame-shaped hemorrhage in the preretinal layer, adjacent
to the optic discs and spreading out from the optic disc. (Courtesy of
Mr M Wade, Department of Medical Illustrations, Royal Perth Hospital,
Western Australia.)

298 Dissection of the arteries on the surface of the base of the brain
showing the circle of Willis and bilateral aneurysms of the middle
cerebral arteries (arrows). (Courtesy of Professor BA Kakulas, Royal
Perth Hospital,Western Australia.)
252 Vascular Diseases of the Nervous System

INVESTIGATIONS Lumbar puncture


Subarachnoid hemorrhage or not? Performed if a CT brain scan is negative for subarachnoid
CT brain scan blood (about 3–5% of cases).
The primary imaging modality to diagnose SAH. It may show:
• High density (whiteness) due to blood in the CSF spaces If the onset of sudden headache was less than 12 hours ago
in 95–97% of cases of SAH (300). • Wait for at least 12 hours to have elapsed since the onset
• The subarachnoid blood can be very subtle. Look in the of symptoms before doing a lumbar puncture (unless
occipital poles of the lateral ventricles for sediment and meningitis is suspected).
for ‘fissures which have disappeared’ (because they
contain just enough blood to raise their density from the If the onset of sudden headache was more than 12 hours ago
black of CSF to the gray of normal brain, but not enough • Do a lumbar puncture immediately (after brain CT).
to make the fissure look white with blood) (301). • If the CSF is unequivocally xanthochromic (with or
• The distribution of blood may help to localize the site of without red cells), no further CSF tests are necessary: the
hemorrhage (e.g. if in the anterior interhemispheric fissure, diagnosis of SAH is confirmed.
rupture of an anterior communicating artery aneurysm is • If the CSF is blood-stained:
likely [296], and if in the sylvian fissure, rupture of a – Spin down the CSF immediately (if it contains red blood
middle cerebral artery aneurysm is likely) (302). cells from a traumatic tap and is allowed to stand,
Spontaneous subdural and subarachnoid blood suggests oxyhemoglobin will be formed in vitro, bilirubin will
rupture of a posterior communicating artery aneurysm. not, but its absence cannot exclude SAH).
• The actual aneurysm may occasionally be seen if large. – Perform spectrophotometric analysis of the supernatant
• Complications such as hydrocephalus (with dilatation of for xanthochromia, (unless the yellow color is evident to
the ventricles) (300) or cerebral ischemia (with low the naked eye). Distinction between a traumatic tap and
density areas in an arterial territory) (303) may be seen. SAH is not possible with the naked eye. If spectro-
• Perimesencephalic hemorrhage typically results in blood photometry has to be deferred for practical reasons, store
around the brain stem, not usually a heavy load, and the CSF wrapped in tin foil, because daylight may induce
hydrocephalus occurs but is unusual. breakdown of bilirubin.
• If the CSF is clear, colorless and acellular, still do
There are several important caveats: spectrophotometry: the presence of pigment may not be
• A normal CT scan does not exclude SAH. A lumbar visible with the naked eye.
puncture must be performed if the CT is normal (about
3–5% of cases of SAH) but SAH is still suspected. If the onset of sudden headache was more than 2 weeks ago
• Blood can disappear very quickly from the CT scan: if Consider doing a lumbar puncture if <4 weeks ago because
the headache started more than 3 days ago, the chances xanthochromia may still be detectable up until 4 weeks, but nor-
of seeing blood decline rapidly and lumbar puncture mal spectrophotometric testing cannot exclude SAH.
becomes more important (after CT has excluded
hydrocephalus) to look for xanthochromia. Imaging the underlying cause of SAH
Intra-arterial angiography
MRI brain scan Indicated if CT brain scan shows subarachnoid hemorrhage
MRI is not useful to diagnose SAH: the blood is difficult to (95–97% of cases) or lumbar puncture performed more than
see and it is less easy to use the technique with acutely ill 12 hours (and up to 2–4 weeks) after headache onset reveals
confused patients. xanthochromia (yellow color) of the CSF supernatant fol-
lowing centrifugation and spectrophotometry. Performed with

300 301 300 CT brain scan showing


obvious recent SAH with
blood (white) in the basal
cisterns (arrows). Note there
is also secondary
hydrocephalus (dilatation of
temporal horns of lateral
ventricles).

301 Subtle SAH: blood is


visible in the Sylvian fissures
but is less obvious than in
300.The sulci are ‘missing’, i.e.
they would normally be filled
with black material.
Subarachnoid Hemorrhage (SAH) 253

multiple views of each major artery to identify the sympto- etiologies such as mycotic or traumatic.
matic aneurysm (304–306) and any incidental ones (306): • A negative angiogram should be repeated after a few
• The gold standard and most surgeons require this prior weeks as some aneurysms do not show up on the initial
to operation. one, except where the CT scan shows typical
• Most aneurysms arise from the arteries near or on the perimesencephalic hemorrhage. Indeed, CT angiography
circle of Willis, or at the major bifurcation points. only is the best diagnostic strategy for patients with
• Aneurysms arising elsewhere should suggest unusual perimesencephalic hemorrhage.

302 Cranial CT 302


scans showing
hemorrhage into the
left Sylvian fissure;
left posterior
inferior frontal lobe;
left anterior
temporal lobe;
subarachnoid space
(interpeduncular
cistern, ambient
cisterns, inter-
hemispheric fissure);
third and lateral
ventricles; and
cerebral aqueduct,
together with
hydrocephalus in a patient 303 304
with a ruptured left middle
cerebral artery aneurysm.

303 CT brain scan of a


patient presenting 2 weeks
after symptom onset.The
subarachnoid blood is no
longer visible, but there is
some low density in the left
parieto-occipital cortex
(arrow) which is ischemic
brain secondary to ‘vasospasm’
complicating
the SAH.

304 Intra-arterial angiogram of


the left internal carotid artery
showing an aneurysm at the
trifurcation of the middle 306
cerebral artery (arrow).
305
305 Catheter cerebral
angiogram, coronal plane,
showing the left middle
cerebral artery aneurysm
(arrow).

306 Intra-arterial angiogram


of the internal carotid artery
showing three aneurysms: a
large internal carotid tip
(arrow); a small pericallosal
(arrowhead) and a small
middle cerebral (short arrow).
Multiple aneurysms are not
uncommon. Usually the largest
one is the symptomatic one.
254 Vascular Diseases of the Nervous System

Others General care


• MRI, MRA and dynamic spiral CT angiography are still Nursing
undergoing evaluation for use in the diagnosis of Continuous observation (Glasgow coma scale, pupils, any
aneurysms (307, 308), though may be useful when focal deficits).
there are localizing signs such as a IIIrd nerve palsy and
when there is a perimesencephalic pattern of Nutrition
hemorrhage. Their use in screening for asymptomatic • Oral route:
aneurysms is controversial and still under evaluation. – Only if intact cough and swallowing reflexes.
• Angiography may show other features such as narrowed – Keep stools soft by adequate fluid intake and restricting
arteries due to vasospasm in the acute phase after SAH. milk content; if necessary add laxatives.
• Transcranial doppler ultrasound (TCD) is widely used in • Nasogastric tube:
some countries to identify patients with vasospasm (as – Deflate endotracheal cuff (if present) on insertion.
evidenced by raised arterial blood velocities) after SAH. – Confirm proper placement by x-ray.
– Begin with small test feeds of 5% dextrose.
Complications of SAH – Feed in sitting position and check gastric residue hourly
Electrocardiography (EKG) changes (to prevent aspiration).
• ST segment and T wave changes, U-wave abnormalities, – Crush tablets and flush down (if needed, phenytoin levels
QT prolongation and sinus arrhythmias are common won’t be adequate in conventional doses).
after SAH. • Total parenteral nutrition: use only as a last resort.
• Life-threatening arrhythmias such as ventricular
fibrillation or ‘torsade de pointe’ are rare. Blood pressure
• Mechanism is unexplained but thought to be sustained Do not treat hypertension unless there is clinical or
sympathetic stimulation, perhaps caused by dysfunction laboratory evidence of progressive hypertensive end-organ
of the insular cortex. damage. Existing antihypertensive drugs can be continued.

Urea and electrolytes Fluids and electrolytes


Hyponatremia is a sign of hypovolemia, due to excessive • Give at least 3 l per day (normal saline) to compensate
natriuresis (salt wasting), rather than inappropriate secretion for ‘cerebral salt wasting’ and to prevent hypovolemia,
of antidiuretic hormone. because this predisposes to cerebral ischemia.
• Compensate for negative fluid balance and for fever.
DIAGNOSIS • Empty bladder by catheter every 6–8 hours if post-void
Clinical plus CT or CSF evidence of subarachnoid blood. residual >100 ml (see p.197)

Grading system for patients with SAH, according to the Pain: headache
World Federation of Neurological Surgeons (WFNS): • Start with paracetamol (acetaminophen) 1 g every 3–4
hours and/or dextropropoxyphene (propoxyphene);
WFNS grade Glasgow Focal deficit avoid aspirin.
coma scale • Midazolam if the pain is accompanied by anxiety (5 mg
I 15 Absent i.m. or infusion pump).
II 14–13 Absent • For severe pain, add codeine 20 mg orally, piritramide
III 14–13 Present 2 mg subcutaneously, then, as a last resort, morphine
IV 12–7 Present or absent 2 mg intravenously, with 1–2 mg increments.
V 6–3 Present or absent
Prevention of deep venous thrombosis and
It is preferable to record the patient’s condition in pulmonary embolism
descriptive terms (e.g. level of consciousness and any focal Apply compression stockings or intermittent compression
neurologic deficits) rather than in codified terms. In by pneumatic devices.
addition, the cause of any neurologic deficit should be
identified (e.g. acute hydrocephalus, intracerebral hema- Prevention of re-bleeding
toma, cardiorespiratory difficulties). • Re-bleeding of a ruptured aneurysm cannot be predicted
reliably.
TREATMENT • Surgical clipping of the aneurysm as early as possible
Indications for immediate operation (within 3 days) is usual practice but it is not supported
• Large intracerebral hematoma associated with by evidence from randomized trials.
deterioration in conscious level. • Antifibrinolytic drugs (e.g. tranexamic acid) reduces re-
• Symptomatic hydrocephalus: gradual obtundation within bleeding by two-thirds but the increased risk of cerebral
24 hours of SAH, downward deviation of the eyes, and infarction negates any beneficial effect.
slow pupillary responses. • Endovascular application of coils into the aneurysm
• Prevent re-bleeding (see below). causing occlusion of the remaining lumen of the
aneurysm by reactive thrombosis: currently being
evaluated in clinical trials.
Subarachnoid Hemorrhage (SAH) 255

Prevention of delayed cerebral ischemia Management


• Avoidance of antihypertensive drugs. • If respiratory arrest: resuscitate and ventilate; within
• Adequate intake of fluid and sodium. hours either spontaneous respiration will return or all
• Calcium antagonists routinely: oral nimodipine reduces other brainstem functions will be lost.
the risk of a poor outcome by about one-third. • Repeat CT scan.
• Fludrocortisone to limit sodium loss. • Consider emergency clipping or coiling of aneurysm after
• Free radical scavengers: tirilazad mesylate: no consistent recovery, as the majority will re-bleed again, with high
improvement of outcome in four trials of tirilazad case fatality; any intracerebral hematoma can be removed
involving more than 3500 patients. at the same time.
• Aspirin and other antiplatelet agents, after clipping the
aneurysm: unknown benefit; currently being trialed. Acute hydrocephalus
• Intrathecal thrombolytics: unknown benefit. • Anticipate if intraventricular hemorrhage or extensive
hemorrhage in perimesencephalic cisterns is present.
CLINICAL COURSE AND COMPLICATIONS • Conscious level gradually deteriorates, particularly on the
Sudden deterioration first day after the bleed.
• Re-bleeding (two-thirds). • Pupils become small and unreactive, and eyes deviate
• Epileptic seizure. downwards (upgaze paresis) due to pressure on rostral
• Delayed cerebral ischemia, with atypical, sudden onset. midbrain.
• Ventricular fibrillation.
• Undetermined.
307
Subacute deterioration
• Edema surrounding intracranial hematoma.
• Cerebral ischemia.
• Hydrocephalus.
• Unsuspected re-bleeding.
• Systemic complications (hyponatremia, arrhythmia,
hypotension, infection, neurogenic pulmonary edema).

Re-bleeding
• Risk of re-bleeding after rupture of an intracranial
aneurysm, without medical or surgical intervention:
about 50% in the first month; about 20% on the first day,
and about 40% in the first month for survivors of the first
day.
• Sudden apnea, in a patient with SAH, usually signifies re-
bleeding.

307 MRI brain scan, proton density image, axial plane, showing
hypodense flow voids in the middle cerebral arteries and a left middle
cerebral artery aneurysm (arrow).

308 MR angiography, coronal plane, showing the left middle cerebral


artery aneurysm (arrow).

308
256 Vascular Diseases of the Nervous System

Management • Treatment with antihypertensive drugs.


• Repeat the CT scan and compare the bicaudate index • Treatment with antifibrinolytic drugs.
with that on any previous scan.
• Spontaneous improvement occurs within 24 hours in half Management
the patients (except those with massive intraventricular • Establish diagnosis: clinical and serial brain CT or MRI
hemorrhage); take action if patient further deteriorates scans (rule out other causes of deterioration).
or fails to improve within 24 hours. • Stop nimodipine.
• Lumbar punctures are reasonably safe if there is no brain • Immediately administer a plasma volume expander: 500 ml
shift, and effective in about half the patients who have of a colloid solution such a hetastarch or hemaccel.
obstruction in the subarachnoid space and not in the • Insert a subclavian vein catheter or pulmonary arterial
ventricular system. balloon catheter; maintain central venous pressure
• External drainage of the ventricles is very effective in between 1.1–1.3 kPa (8–12 mmHg), or pulmonary
restoring the level of consciousness but carries a high risk wedge pressure between 1.9–2.2 kPa (14–18 mmHg).
of re-bleeding (consider emergency clipping or coiling • Maintain fluid intake with at least 3 l of normal saline
of the aneurysm at the same time), and of infection (this (0.9%) per 24 hours.
may to some degree be prevented by prophylactic • Correct hyponatremia.
antibiotics or subcutaneous tunnelling). • Keep hematocrit around 40%.
• Keep arterial pressure 2.7–5.3 kPa (20–40 mmHg)
Hyponatremia above baseline values; may need inotropic support in
• Almost invariably caused by sodium depletion, not by intensive care unit.
sodium dilution (SIADH).
• Associated hypovolemia increases risk of delayed cerebral Cardiac arrhythmias
ischemia. Rarely need to be treated.

Management PROGNOSIS
• Give isotonic saline (with or without albumin to expand • Half of patients die; sudden death occurs in about 15%
plasma volume) or a mixture of glucose and saline; no of patients: usually ruptured posterior circulation
free water. aneurysms and intraventricular hemorrhage.
• If necessary add fludrocortisone acetate, 400 µg/day in • Half of survivors remain severely disabled.
two doses, orally or intravenously. • Delayed ischemia is a major cause of death and disability.
• Keep central venous pressure between 1.1–1.3 kPa • Outcome in patients with familial SAH is worse than in
(8–10 mmHg), or pulmonary capillary wedge pressure patients with sporadic SAH.
between 1.1–1.6 kPa (8–12 mmHg). • Non-aneurysmal perimesencephalic SAH: excellent
prognosis; re-bleeding and ischemia do not occur.
Delayed cerebral ischemia
Up to one-third of patients with a ruptured aneurysm Adverse prognostic factors
develop cerebral ischemia, mainly between day 5 and day Decreased level of consciousness on admission ++++
14 after the initial bleed. Cerebral ischemia or infarction is Large amount of blood in subarachnoid space on +++
not confined to the territory of a single cerebral artery or CT scan
one of its branches. The pathogenesis is complex. Increasing age ++
Vasospasm is often implicated because its peak frequency Loss of consciousness at onset +
from days 5–14 coincides with that of delayed ischemia and High blood pressure on admission +
because it is often generalized, like the clinical and Pre-existing medical condition +
radiologic findings, but it is not the only factor in the Aneurysm in posterior rather than anterior circulation +
pathogenesis of cerebral ischemia. Use of anticoagulants +
Angiographic evidence of vasospasm occurs in up to 70% Intravenous drug abuse +
of patients with aneurysmal SAH, but neurologic Associated intracranial hematoma +
deterioration secondary to vasospasm occurs in about one- Level of creatinine kinase BB in CSF +
third of all patients, and about half of these patients with
symptomatic vasospasm die or suffer neurologic disability
as a direct result of the ischemia.

Determinants of delayed cerebral ischemia


• SAH caused by a ruptured aneurysm.
• Arterial narrowing.
• Depressed level of consciousness for more than 1 hour
after the onset.
• Amount of subarachnoid blood on early CT scanning,
not the distribution.
• Acute hydrocephalus.
• Hyponatremia and hypovolemia.
• Detection of emboli on transcranial Doppler monitoring,
after surgical clipping.
Cerebral Venous Thrombosis 257

CEREBRAL VENOUS THROMBOSIS Cerebral veins


Three groups of veins drain the blood supply from the
brain: superficial cerebral veins (or cortical veins), deep
DEFINITION cerebral veins and posterior fossa veins.
Occlusion of cortical cerebral veins or dural venous sinuses,
or both, by thrombus (phlebothrombosis). PATHOLOGY
• Isolated cortical venous thrombosis is rare, most patients
EPIDEMIOLOGY have dural sinus thrombosis with or without cortical vein
• Incidence: unknown (previous studies have relied on involvement (310, 311).
autopsy data, and more recently cerebral angiography) • The thrombus itself is like other venous thrombi
but uncommon. elsewhere in the body. When it is fresh, it is a red
• Low prevalence in autopsy series; 0.1–0.9% of autopsies. thrombus rich in red blood cells and fibrin but poor in
• Age: any age, but young and middle-aged women at platelets; when it is old, it is replaced by fibrous tissue
greatest risk: mean age 40 years. sometimes showing recanalization.
• Gender: higher incidence in females (1.3:1). • Hemorrhagic infarction of the brain is common.

RELEVANT VENOUS ANATOMY


Blood from the brain is drained by cerebral veins which
empty into dural sinuses that drain mostly into the internal
jugular veins. There is extensive collateral circulation in the
cerebral venous system.

Dural sinuses 309


The most commonly affected by thrombosis are the
superior sagittal sinus, lateral sinus, cavernous sinus and
straight sinus (309).

309 MR venogram, left postero-lateral view, showing the normal


appearance of the venous sinuses in the head.

310, 311 Autopsy specimen of brain, vertex of the brain (310), and
coronal section through the frontal lobes (311), showing bilateral
parasagittal hemorrhagic infarction due to superior sagittal sinus
thrombosis.

310

311
258 Vascular Diseases of the Nervous System

ETIOLOGY Infective
Predisposing factors can be identified in up to 80% of • Bacterial: septicemia, endocarditis, typhoid, tuberculosis.
patients. • Viral: measles, hepatitis, encephalitis, herpes, HIV, CMV.
• Parasitic: malaria, trichinosis.
Local conditions directly affecting the veins and • Fungal: aspergillosis.
sinuses
Non-infective Uncertain etiology (20–25%)
• Head trauma (open or closed, with or without fracture).
• Intracranial surgery. PATHOGENESIS
• Brain infarction or hemorrhage. Thrombus formation due to venous stasis, increased clotting
• Tumor invasion of dural sinuses (meningioma, meta- tendency, changes in the vessel wall, and less frequently,
stases, glomus tumor). embolization.
• Catheterization of, and infusions into, the internal
jugular veins (e.g. for parenteral nutrition). CLINICAL SYNDROMES
Occur in isolation or combination.
Infective
• Direct septic trauma. Sagittal and/or transverse sinus thrombosis
• Local regional infection (scalp, sinuses, ears, mastoids, Syndrome of progressive idiopathic intracranial hypertension
nasopharynx). • Headache.
• Intracranial infection: • Obscurations of vision.
– Bacterial meningitis. • Papilloedema.
– Meningovascular syphilis.
– Subdural empyema. Cortical venous thrombosis
– Bacterial or fungal brain abscess. Syndrome of subacute or chronic focal or generalized
encephalopathy
Systemic conditions Evolution over a few days, weeks or months of headache,
Non-infective confusion, drowsiness, epileptic seizures (partial and
• Surgery: any surgical procedure, with or without deep secondary generalized), raised intracranial pressure, and
venous thrombosis. coma with or without focal neurologic signs.
• Hormonal:
– Pregnancy or more commonly the puerperium. Stroke-like syndrome (frequently with seizures)
– Oral contraceptives (estrogens or progestogens A cortical stroke syndrome (i.e. non-lacunar) occurring in
[progestins]) (OCP): age-adjusted odds ratio of 13 for patients suffering from any one of the etiologic conditions
OCP use and risk of CVST; age-adjusted odds ratio of should suggest venous thrombosis, particularly if the
30 for OCP use combined with prothrombin G20210A evolution of the focal neurologic deficit is rather slow, if
gene mutation and risk of CVST. seizures occur, and if there is hemorrhagic infarction or
• Medical: frank hemorrhage in the territory of drainage of a cortical
– Severe dehydration of any cause (including angiography vein in the cortex/subcortex.
or myelography).
– Hyperviscosity syndrome: multiple myeloma; Thunderclap headache
Waldenstrom’s macroglobulinemia. Abrupt onset of very severe pain in the head, perhaps caused
– Hypercoagulable state: by intraparenchymal hemorrhage, venous hemorrhagic
Red blood cell disorders: polycythemia; post-hemorr- infarction, or venous subarachnoid hemorrhage.
hagic anemia; sickle cell disease/trait; paroxysmal
nocturnal hemoglobinuria. Cavernous sinus thrombosis
Platelet disorders: essential thrombocythemia. Syndrome of chemosis, proptosis and painful ophthalmoplegia
– Coagulation disorders (see p.222): deficiency of anti- Usually due to suppuration spreading from face, orbit or
thrombin III, protein C, or protein S; circulating lupus paranasal sinuses (staphylococcal, streptococcal, mucormy-
anticoagulant; activated protein C resistance (factor V cosis), causing:
Leiden gene mutation); prothrombin G20210A gene • Pain around the eye and face.
mutation (see p.223); disseminated intravascular coagu- • Conjunctival and eyelid edema, episcleral congestion.
lation; heparin- or heparinoid-induced thrombo- • Proptosis.
cytopenia; antifibrinolytic treatment. • Blindness and papilledema.
– Extracranial malignancy (non-metastatic effect): any • Cranial nerve III, IV, VI, V1 and perhaps V2 palsies.
visceral carcinoma, carcinoid, lymphoma, leukemia, L-
asparaginase therapy. Thrombosis may spread to the contralateral cavernous
– Connective tissue disease: SLE; Wegener’s granuloma- sinus and other dural sinuses. Suppuration may spread to
tosis; giant cell arteritis. cause subdural empyema, bacterial meningitis, or infective
– Cardiac: congenital heart disease; congestive heart failure; arteritis and thrombosis of the terminal carotid and middle
pacemaker. cerebral artery origin.
– Gastrointestinal: inflammatory bowel disease; cirrhosis.
– Various others: Behçet’s disease (312–314); sarcoidosis;
nephrotic syndrome; androgen therapy; diabetes mellitus;
parenteral injections; hyperhomocystinemia.
Cerebral Venous Thrombosis 259

Deep cerebral vein thrombosis 312


Variable clinical features. May have an ‘encephalitic’
presentation characterized by the gradual onset of headache,
confusion, obtundation and vomiting with or without the
development of focal neurologic signs such as amnesia or
long tract signs due to bilateral thalamic infarction or
unilateral temporoparietal infarction (that may become
hemorrhagic) depending on the collateral venous circulation
and whether the thrombus propagated from the lateral sinus
into the vein of Labbé.

DIFFERENTIAL DIAGNOSIS
• Subdural empyema.
• Bacterial meningitis.
• SAH.
• Encephalitis.
• Brain abscess.
• Stroke: arterial occlusion or dissection, SAH. 313
• Benign thunderclap headache, ‘crash’ migraine.

INVESTIGATIONS
Cranial CT and MRI imaging
The appearance of the brain in cerebral venous thrombosis
depends on whether the venous sinuses or cortical veins (or
both) are involved. Thrombosis of the cortical veins usually
results in venous infarction (315), whereas isolated venous

312–314 Tongue (312) and and genital (scrotal) (313, 314) ulcers
in a young man with Behçet’s disease who presented with cerebral
venous thrombosis.

315 CT scan without contrast of a 6 hour old venous infarct


(compare with the arterial infarct of similar age in 227). Note the well
defined edges, marked swelling, central hemorrhage.There was no sinus
involvement.
315
314
260 Vascular Diseases of the Nervous System

sinus thrombosis may not cause infarction but results in Other investigations
generalized brain swelling and raised ICP (316, 317). The All other investigations are directed towards demonstrating
imaging of venous thrombosis is therefore directed at: the underlying cause (see Etiology, above).
• Recognizing correctly the venous origin of any
parenchymal lesion. Coagulation studies (see p.220)
• Diagnosing the extent of any sinus involvement. Indicated in all patients, and particularly if family or past
• Identifying any predisposing factors (such as tumors history of thrombotic episodes, or unexplained infarction:
pressing on the sinus or local infection). • Protein C and S.
• Antithrombin III.
Venous sinus thrombosis • Prothrombin G20210A gene mutation.
This may be imaged by CT and MRI and appear as: • Factor V Leiden mutation.
• A high density (or altered signal and loss of flow void on • Antiphospholipid antibodies.
MRI) in the sinus (due to the thrombus) (316–319). • Fibrinogen.
• A triangular filling defect in the sinus after i.v. contrast • Plasminogen.
(‘empty delta’ sign (251, 252). High splitting of the
superior sagittal sinus can mimic a delta sign, as can an Chest x-ray
adjacent epidural abscess. Chest x-ray or other imaging, inflammatory markers,
• Generalized brain swelling (cerebral edema); autoantibodies, and tissue biopsy if suspected malignancy or
• Local infection such as mastoiditis or sinusitis as a connective tissue disease.
precipitating cause.
DIAGNOSIS
Venous infarction Angiography remains the gold standard for diagnosis but
• May be single or multiple. MRI with MR venography allows accurate diagnosis in most
• Typically more clearly demarcated and of lower density cases.
than arterial infarction of the same age (315).
• More frequently hemorrhagic than arterial infarcts. TREATMENT
• The hemorrhage typically starts in the center and extends • Any underlying cause (e.g. infection) should be identified
towards the periphery, (whereas in arterial infarcts the and treated.
hemorrhage is typically around the edge). • The risks and benefits of anticoagulant therapy with
• Infarction occupies territories which are not typically intravenous heparin followed by oral anticoagulation for
arterial, or involve adjacent areas of brain (e.g. occipital 3–6 months remain uncertain, based on a systematic
lobe and cerebellum) which is difficult to explain from review of the only two randomized controlled trials in a
the arterial anatomy. total of 70 patients. Anticoagulants may be useful in
• More swollen than an arterial infarct of the same age. patients with dural sinus and even cortical venous
• The brain beyond the visible boundaries of the infarct thrombosis and may not necessarily be harmful, even in
also may appear swollen. the presence of hemorrhagic infarction.
• The cord sign refers to a thrombosed vein, present on • If patients deteriorate, local thrombolysis with intrasinus
pre-contrast views. urokinase may also have a role but this remains to be
evaluated in controlled trials.
Normal scans are more likely when the thrombotic
process has been confined to the deep venous system, PROGNOSIS
including the vein of Galen and the straight sinus; and when Mortality rate <10%. If patients survive, the prognosis for
the patient has presented with signs of intracranial recovery of function is generally favorable and much better
hypertension alone. than in arterial occlusion.
MRI of the brain is now the diagnostic procedure of
choice because it is non-invasive, sensitive to blood flow, and Prognostic factors
readily visualizes the thrombus itself. • Topography of thrombosis: deep cerebral vein and
cerebellar vein thrombosis carry a much higher risk than
Four vessel conventional or digitalized intra-arterial cortical vein thrombosis.
cerebral angiography • Underlying cause: septic cerebral venous sinus
This technique, which visualizes the entire venous phase on thrombosis still has a mortality rate of 30% in cavernous
at least two projections (frontal and lateral), has been the sinus thrombosis and up to 78% in SSS thrombosis.
gold standard for diagnosis, revealing the partial or
complete lack of filling of veins or venous sinuses.

CSF
• Pressure is usually increased.
• May be normal or contain a modest excess of red blood
cells, lymphocytes, neutrophils and protein.
• It is usually not necessary to perform a lumbar puncture
and CSF examination in patients with suspected cerebral
venous sinus thrombosis.
Vascular Dementia 261

VASCULAR DEMENTIA EPIDEMIOLOGY


• Incidence: 2.5 per 1000 undemented individuals per year.
• Prevalence: about 22 per 1000 population (up to 40% of
DEFINITION cases of dementia: the second most common cause of
An acquired syndrome of cognitive impairment (dementia) dementia after Alzheimer’s disease); about 62 per 1000
that is characterized by the abrupt onset and stepwise people over the age of 59 years. Estimates of the
progression of deficits of memory and at least two other prevalence of vascular dementia may be unreliable because
cognitive functions (e.g. abstract thinking), sufficient to of different diagnostic and pathologic criteria used in
interfere with the person’s usual social activities, and caused different studies.
by vascular diseases of the brain. • Age: elderly.
• Gender: M=F.

316 317

317 Same patient as 316,T1W midline sagittal MRI shows the


thrombus in the sagittal sinus (arrows).There should normally be a flow
void in the sagittal sinus.

316 CT scan without contrast shows thrombosed sagittal and straight


sinuses (arrows) without any actual infarction complicating
meningococcal meningitis.

318, 319 MRI brain scans 318 319


showing an hypointense
(dark) (318), and
hyperintense (white) (319)
defect in the venous sinus at
the torcula (arrows) due to
loss of the flow void as a
result of venous sinus
thrombosis.
262 Vascular Diseases of the Nervous System

PATHOLOGY AND PATHOGENESIS of the periventricular white matter, rather than any atheroma
Parenchymal pathology of the large arteries which is variable in extent and severity.
The site of brain tissue loss is a more important determinant White matter lesions are found in about 80% of patients with
of cognitive function than the volume of tissue lost. vascular dementia, about 15% of patients with early-onset
Alzheimer’s disease and about 75% of patients with late-onset
Single strategically placed infarcts or hemorrhages Alzheimer’s disease. They are associated with hypertension
A single infarct or hemorrhage, if located in a strategically and, in some studies, with heart disease and diabetes. They
important part of the brain, such as the circuits involving the are thought to cause dementia by disconnecting the
dorsolateral frontal convexity-caudate nucleus-globus pathways between the cortical and subcortical areas.
pallidus-thalamus, can produce a vascular dementia.
Examples include infarction or hemorrhage of the: Diffuse laminar necrosis (global cerebral ischemia)
• Dominant angular gyrus, situated in the inferior parietal
lobule, usually by embolic occlusion of the angular branch Vascular pathology
of the inferior division of the MCA, and characterized by Hypoxic-ischemic lesions
the acute onset of fluent dysphasia, dysgraphia, memory • Large artery atherosclerosis.
loss and visuospatial disorientation. • Small vessel hyaline wall thickening (arteriolosclerosis),
• Thalamus, particularly the dominant dorsomedial microatheroma and lipohyalinosis.
thalamus, by occlusion of the paramedian thalamic • Embolism from the heart.
(thalamoperforate) branches of the posterior cerebral • Non-atheromatous angiopathies.
artery, causing memory loss, slowness, apathy, ocular – Granular degeneration of the media of small arteries
palsies, and drowsiness. (CADASIL).
• Caudate nucleus and globus pallidus, usually by – Cerebral vasculitis
thrombotic or embolic occlusion of the penetrating lateral – Neoplastic angioendotheliomatosis (malignant lymphoma
lenticulostriate branches of the middle cerebral artery. of blood vessels).
• Basal forebrain and dorsolateral pre-frontal cortex. – Mural dissections.
• Hippocampus, usually by embolic occlusion of the – Dural arteriovenous malformation.
cortical branches of the posterior cerebral artery, or • Hematologic disease (thrombophilia).
diffuse cerebral ischemia.
Hemorrhagic lesions
Multiple infarcts or hemorrhages • Subdural hematoma.
Cortical/subcortical infarcts or hemorrhages may cause a • SAH: anterior communicating artery aneurysm.
‘cortical’ type dementia with signs of amnesia, aphasia, • Intracerebral hemorrhage (see p.238):
apraxia, and agnosia. The infarcts are commonly the result – Amyloid angiopathy.
of thromboembolism from the heart or a large artery (aortic – Hypertensive small vessel disease.
arch, carotid and vertebrobasilar) to the anterior, middle, or
posterior cerebral arteries or their branches, but can also be RISK FACTORS FOR VASCULAR DEMENTIA
caused by large vessel disease causing hypoperfusion and Age and stroke are the most important risk factors for
infarcts in the borderzones between major arterial territories, developing dementia:
and small vessel disease such as microatheroma/lipo- • Increasing age;
hyalinosis and vasculitis. Multiple cortical/subcortical • Past history of stroke (symptomatic stroke increases the
hemorrhages are most commonly due to amyloid angiopathy risk of dementia more than ninefold) or myocardial
but can also be seen with vasculitis, bleeding diatheses, infarction (one-third of patients).
metastases, hemorrhagic infarction and trauma.
Small deep (‘lacunar’) infarcts may cause a ‘subcortical’ Other putatitive risk factors include:
dementia characterized by signs of psychomotor slowing, • Hypertension (60% of patients).
poor concentration, indecision and mental apathy. Other • Smoking (35%).
features, besides cognitive impairment, include hemiparesis, • Diabetes (20% of patients).
small stepping gait (marche á petits pas), dysarthria, and • Hyperlipidemia (20%).
dysphagia. Typical patients are elderly, ex- or current • Alcohol abuse.
smokers, with hypertension and diabetes. Rarely, they are • Family history (CADASIL).
part of the syndrome of cerebral autosomal dominant • Lower education level.
arteriopathy with subcortical infarcts and leuko- • Residing in a rural area.
encephalopathy (CADASIL), which is a hereditary disease • Living in an institution.
with linkage to chromosome 19, and features of a subcortical • Brain atrophy.
dementia, recurrent stroke-like events and visible white • Cortical infarcts.
matter lesions on brain imaging. • Brain white matter lesions.
• Left hemisphere stroke.
Diffuse white matter infarction • Early urinary incontinence.
(Subcortical arteriosclerotic leukocencephalopathy, • Falls.
Binswanger’s disease). Diffuse or multifocal, often • Abnormal EKG (80% of patients compared with 30% of
periventricular, areas of demyelination, axonal loss, and people with Alzheimer’s disease).
reactive gliosis in the white matter probably due to anoxia as
a result of arteriosclerotic changes (hyalinization, fibrosis and
thickening) in the long penetrating end arteries and arterioles
Vascular Dementia 263

CLINICAL FEATURES Lewy body type dementia (see p.430)


Slowly progressive intellectual impairment, with recurrent • Marked fluctuations of symptoms.
stroke-like events. • Prominent extrapyramidal signs, particularly rigidity and
bradykinesia.
History • Early and florid visual hallucinations and delusions.
• Presenting symptoms: sudden onset and stepwise course • Poor tolerance of neuroleptic drugs.
of cognitive decline with history of transient ischemic
attacks, strokes, or both. Epileptic seizures occur in 10% Parkinson’s disease dementia (see p.420)
of patients. Incontinence of urine and feces is not • Possibly affects 10–20% of patients with Parkinson’s
uncommon. disease.
• Past history of vascular risk factors: hypertension, • Dementia is subcortical (apathy, poor concentration,
diabetes, heart disease, smoking. indecision, slowness of central processing).
• Family history: CADASIL. • Dementia is usually preceded by parkinsonian features.
• Gait changes of vascular dementia and Parkinson’s
Neurologic examination disease closely resemble each other.
• Focal neurologic deficits such as pyramidal tract signs • Dopaminergic medication usually improves the
(hemiparesis, extensor plantar response, pseudobulbar parkinsonian features.
palsy), extrapyramidal signs, hemisensory loss,
hemianopia, and dysarthria. Progressive supranuclear palsy (see p.432)
• Gait abnormality: start and turn hesitation, shuffling, • Subcortical dementia.
reduced arm swing. • Preceding parkinsonian features.
• Grasp reflexes • Paralysis of vertical gaze.
• Hypertension and hypertensive retinopathy. • Truncal ataxia.
• A source of thromboembolism such as AF, valvular heart • Parkinsonian features do not respond to dopaminergic
disease, heart failure, carotid artery disease. medication.

Neuropsychologic examination Multiple systems atrophy (see p.435)


• Essential to ascertain the presence or absence of a • Dementia is slowly progressive.
cognitive deficit, the nature of the deficit (i.e. in what • Accompanying signs of dysfunction of any or all of the:
domains) and the degree of the deficit. – Pyramidal tract.
• Concentration and executive function: poor learning – Extrapyramidal system.
strategies, impaired word list generation, emotional – Autonomic system.
blunting and lability, poor insight and judgement. – Cerebellum.
• Memory: impaired learning of verbal and visual
information, reduced recall following a delay. Frontal lobe tumors
• Verbal output: dysarthria, reduced grammatical Early symptoms may be behavioral disturbance (disinhibited
complexity of spontaneous speech. and irrational) or mental apathy and indecision. Later signs
• Depression: present in 25% of patients; depressive may be those of raised intracranial pressure (see p.477).
symptoms in 60%.
• Anxiety: common. Uncommon but treatable causes of dementia
• Delusions: present in up to one-half of patients. • Hypothyroidism.
• Personality alterations: apathetic, listless, lifeless, quiet • Vitamin B12 deficiency.
and labile. • Neurosyphilis.
• Normal pressure hydrocephalus.
DIFFERENTIAL DIAGNOSIS • Frontal lobe tumors.
Vascular dementia and Alzheimer’s disease (mixed or • Cerebral ischemia due to cerebral vasculitis,
‘double dementia’) hyperviscosity syndrome and severe bilateral carotid
Clinical course may be slowly progressive and it may be stenosis.
difficult to interpret the relevance of brain lesions, consistent • HIV dementia (see p.301).
with infarction, seen on neuroimaging (e.g. white matter
lesions). INVESTIGATIONS
CT or MR imaging of the brain
Alzheimer’s disease (see p.407) Excludes other causes of dementia (e.g. frontal tumor,
• More likely to have retrieval errors and irrelevant hydrocephalus) and almost always identifies one or more
intrusions on memory tests and to perform more poorly vascular lesions. CT brain scan reveals more or less
on a story recall test. symmetric periventricular and subcortical hypodensity in the
• Less likely to have a history of previous strokes, cerebral white matter, with or without ventricular dilatation
hypertension, and alcohol abuse, depression, or an and focal hypodensities and thought to be due to ‘small
abnormal EKG (30% of cases). vessel’ ischemia and infarction.
264 Vascular Diseases of the Nervous System

MR T2W or proton density imaging demonstrates the Hachinski ischemia scale


same features even better as high signal areas (320, 321). Item Score value
These radiologic appearances (leukoaraiosis [leuko=white, Abrupt onset* 2
araiosis=rarefaction]) are rather non-specific and can be Stepwise course* 1
found in apparently normal elderly people. However, they Fluctuating course 2
are more commonly associated with gradual dementia, Preservation of personality 1
unsteadiness of gait, and recurrent ischemic, particularly Nocturnal confusion 1
lacunar, or occasionally hemorrhagic strokes, and are more Depression 1
frequent in patients with hypertension, other vascular risk Somatic complaints* 1
factors, atherosclerosis and cerebral atrophy. Emotional incontinence* 1
Additional features may include multiple small ‘holes’ in History of hypertension* 1
the basal ganglia and pons, and cortical infarcts may also be Evidence of associated atherosclerosis 1
present. History of stroke* 2
Focal neurologic symptoms* 2
Blood tests Focal neurologic signs* 2
• Full blood count and ESR.
• Plasma viscosity. *Discriminating items after validation at post mortem examination.
• Serum urea and electrolytes.
• Plasma glucose. A score of 7 or higher is said to be diagnostic of vascular dementia but
• Serum cholesterol and triglycerides. this scale has poor interrater reliability.
• Liver function tests.
• Serum protein electrophoresis. NINDS–AIREN CRITERIA FOR VASCULAR
• Antinuclear antibodies. DEMENTIA
• Thyroid function tests. Definite vascular dementia
• Syphilis serology. • Clinical criteria for probable vascular dementia.
• Vitamin B12. • Autopsy demonstration of appropriate ischemic brain
• Coagulation studies (younger patients) (see p.220): injury and no other cause of the dementia.
– Antiphospholipid antibodies.
– Proteins C and S. Probable vascular dementia
– Antithrombin III. Dementia
– Factor V Leiden mutation. • Decline from a previous higher level of cognitive
functioning.
Other • Impairment of two or more cognitive domains.
• EKG. • Deficits severe enough to interfere with activities of daily
• Echocardiography: if prosthetic heart valves, rheumatic living and are not due to physical effects of stroke alone.
valvular heart disease, or suspected left atrial myxoma. • Patients must not be delirious; must not have psychosis,
• Doppler ultrasonography of the carotid arteries. aphasia or sensory-motor impairment that precludes
• Single photon emission computed tomography (SPECT): neuropsychologic testing; and the patient must not have
asymmetric patchy areas of reduced cerebral blood flow. any other disorder capable of producing a dementia
• Positron emission tomography (PET): fluorodeoxyglucose syndrome.
PET reveals multifocal regions of reduced cerebral
metabolism (due to local infarction or a distant effect of Cerebrovascular disease
diaschisis) in contrast to symmetric biparietal • Focal neurologic signs consistent with stroke.
hypometabolism typically observed in Alzheimer’s disease. • Neuroimaging evidence of extensive vascular lesions.

DIAGNOSIS Relationship between the dementia and the


The purpose of the clinical and laboratory assessment is to cerebrovascular disease
establish the diagnosis of vascular dementia, the cause of the As evidenced by one or more of the following:
cerebrovascular disease, and other factors that may be • Onset of dementia within 3 months of a recognized stroke.
contributing to the cognitive compromise. • Abrupt deterioration, or fluctuating or stepwise
The diagnosis of vascular dementia is based on a decline progression of the cognitive deficit.
in cognitive function (dementia) that correlates with a clear
history of strokes. However, the association between Possible vascular dementia
declining cognitive function (dementia) and cerebrovascular • Dementia with focal neurologic signs but without
disease may not be causal; it may be merely contributory or neuroimaging confirmation of definite cerebrovascular
even coincidental. Various diagnostic criteria exist to aid the disease.
diagnosis but many have not been validated. • Dementia with focal neurologic signs but without a clear
temporal relationship between dementia and stroke.
• Dementia with focal neurologic signs but with a subtle
onset and variable course of cognitive deficits.
Hypertensive Encephalopathy 265

TREATMENT HYPERTENSIVE ENCEPHALOPATHY


• Treat any underlying causes and risk factors that may be
remediable:
– Cerebral vasculitis: corticosteroids and cyclophosphamide. DEFINITION
– Hyperviscosity syndrome. A relatively rapidly evolving clinicoradiologic syndrome
– Neoplastic angioendotheliomatosis: chemotherapy. caused by capillary leakage related to abrupt hypertension,
• Control vascular risk factors such as hypertension, and characterized by confusion, cognitive changes, seizures
smoking, hypercholesterolemia and diabetes. and sometimes cortical blindness; and MRI evidence of
• Long term antiplatelet therapy such as low dose aspirin. diffuse or posterior (parieto-occipital) white matter changes.
• Physiotherapy: gait, spasticity.
• Speech therapy: dysarthria, dysphasia, dysphagia. EPIDEMIOLOGY
• Psychologic therapy: visual-spatial neglect. • Incidence: rare.
• Age: any age, mean age: 40 years.
No benefit is obtained with hemorrheologic agents • Gender: M=F.
(pentoxifylline), nootropic compounds (oxiracetam),
calcium channel blockers (nimodipine) or neuroprotective PATHOLOGY
agents. Macroscopic
The brain appears normal or shows evidence of cerebral
CLINICAL COURSE AND PROGNOSIS swelling or hemorrhages in various sizes. Brain hemorrhage
• Characteristically, a progressive stepwise course of is not a sequela of the encephalopathy but is due to the
cognitive decline but in up to half of patients it can primary disease causing the encephalopathy or one of the
follow a slow progressive course. complications of the disease; so it is a complication of
• 50% survival: 6.7 years (cf. 8.1 years for Alzheimer’s uncontrolled hypertension, eclampsia and potential bleeding
disease). disorders.
• Cause of death: heart disease or recurrent stroke.
Microscopic
Widespread minute infarcts, with a predilection for the basis
pontis, due to fibrinoid necrosis of the walls of arterioles and
capillaries, and occlusion of their lumens by fibrin thrombi.
Similar vascular changes are found in other organs,
particularly the retinae and kidneys.

320 T2W MRI in a 78 year 320 321


old female with vascular
dementia due to
Binswanger’s disease. Note
the cerebral atrophy and the
diffuse hyperintense areas
adjacent to the frontal and
posterior horns of the lateral
ventricles, representing
subcortical ischemic
leukoencephalopathy due to
‘small vessel disease’.

321 T2W MRI at the level


of the lateral ventricles in a
75 year old female with
cognitive impairment due to
multi-infarct dementia. Note
the atrophy and multiple
hyperintense areas in the
periventricular white matter
in keeping with ‘small vessel
disease’.
266 Vascular Diseases of the Nervous System

ETIOLOGY CLINICAL HISTORY


• Abrupt increase in blood pressure usually in patients with • Subacute onset:
known hypertension (most common). Encephalopathy – Headache: severe.
is rare in chronic hypertension unless the diastolic blood – Nausea, vomiting.
pressure exceeds 20 kPa (150 mmHg) because the upper – Confusion.
limit of anticoagulation is ‘set’ higher. – Declining conscious state.
• Renovascular hypertension. – Blurred vision or blindness.
• Parenchymal renal disease. – Seizures: partial or generalized.
• Acute glomerulonephritis. – Weakness: focal or generalized.
• Scleroderma and other connective tissue diseases. • Past history of hypertension: duration, severity and level
• Vasculitis. of control.
• Drugs: sympathomimetic agents (amphetamines, • Extent of pre-existing end-organ damage (heart, brain,
cocaine, phencyclidine hydrochloride, lysergic acid retina, kidneys).
diethylamide, diet pills), tricyclic antidepressants. • Coexisting acute or chronic illnesses.
• Ingestion of tyramine in conjunction with a monamine • Family history of hypertension.
oxidase inhibitor. • Medications: prescription, over-the-counter, illicit;
• Withdrawal of antihypertensive drugs (centrally acting antihypertensive drug compliance.
agents and beta-antagonists).
• Pre-eclampsia, eclampsia. EXAMINATION
• Pheochromocytoma. • Disorientation.
• Renin-secreting tumor. • Obtundation.
• Head injury. • Focal neurologic signs (e.g. cortical blindness due to
• Autonomic hyperactivity in Guillain–Barré or spinal cord bilateral occipital lobe dysfunction).
syndromes. • Seizures: generalized or focal.
• Bilateral carotid endarterectomy (baroreceptor reflex • Hypertensive retinopathy (including papilledema) (322,
failure). 323).
• Pulses (if unequal, suspect aortic dissection).
PATHOPHYSIOLOGY • Blood pressure supine and erect: can be apparently
Increases in circulating levels of vasoconstrictor substances, normal, particularly in young or pregnant patients in
such as norepinephrine, angiotensin II, or antinatriuretic whom the base-line pressures are normally low. A single
hormone lead to an abrupt increase in systemic vascular blood pressure reading may be deceptive. Blood pressure
resistance and blood pressure. Severely elevated blood should be measured frequently and compared with base-
pressure precipitates endothelial damage, platelet and fibrin line values.
deposition, arteriolar fibrinoid necrosis and loss of • Cardiac and chest auscultation (suspect congestive heart
autoregulatory function with resultant end-organ (brain, failure).
retina, kidney, heart) ischemia. Ischemia, in turn, triggers
the further release of vasoactive substances, thus initiating DIFFERENTIAL DIAGNOSIS
a vicious cycle of further vasoconstriction and myointimal • Stroke: there may be doubt about the onset of symptoms
proliferation. (i.e. if the patient is confused or obtunded), the blood
Failure of autoregulation of cerebral blood flow leads to pressure may be only moderately elevated, and focal or
a breakdown of the blood–brain barrier with transudation lateralizing neurologic signs may accompany the
of fluid, causing widespread cerebral edema, and petechial symptoms of more diffuse brain disturbance (headache,
hemorrhages, sometimes culminating in frank, fatal confusion, visual disturbances, and seizures). Even a
intracranial hemorrhage. severely elevated blood pressure is not specific because it
Encephalopathy may occur with a diastolic blood can be a consequence of stroke and a cause of stroke.
pressure of 13.3 kPa (100 mmHg) or less if autoregulation • SAH.
is normal and easily exceeded by a rapid rise in blood • Epilepsy.
pressure from a normally low level. • Encephalitis.
• Vasculitis of the CNS.
RISK FACTORS • Brain tumor.
• High blood pressure (e.g. poor antihypertensive drug • Reversible posterior multifocal leukoencephalopathy:
compliance or blood pressure control). – Puerperal eclampsia.
• Pre-existing dysfunction of the vascular endothelium, as – Renal failure.
may occur in pre-eclampsia and eclampsia or with – Immunosuppressive agent treatment: cyclosporin: has
immunosuppressive therapy, may predispose patients to direct toxic effects on vascular endothelial cells;
failure of autoregulation and encephalopathy after interferon-alfa; tacrolimus; levamisole; fluorouracil.
relatively slight increases in blood pressure. • Multiple sclerosis.
• Progressive multifocal leukoencephalopathy.
Hypertensive Encephalopathy 267

INVESTIGATIONS Hospital admission, preferably to an intensive care unit,


Imaging should be arranged. An arterial line should be placed as
CT or MRI brain scan soon as possible to confirm the cuff readings and to guide
Scans may show: therapy (along with the clinical response).
• Areas of infarction/ischemia in the distribution of an Sodium nitroprusside is the drug of choice. It is
arterial territory. administered by continuous infusion (0.5–10 μg/kg/min)
• Typical areas of involvement are the posterior parieto- and therefore the blood pressure demands constant
occipital lobes bilaterally. surveillance, preferably intra-arterially. Adverse effects
• The abnormalities may resolve as the patient recovers. include hypotension, nausea, vomiting, and apprehension.
• MRI is more sensitive than CT to these changes, but Effective alternatives include labetalol (20–80 mg i.v.
confused patients tolerate MRI less well than CT. bolus every 5–10 min, up to 300 mg; onset in 5–10 min,
duration of action 3–6 hours), diazoxide (50–100 mg i.v.
Causes and consequences of hypertension bolus every 5–10 min up to 600 mg; onset in 1–5 min,
• FBP. duration of action 6–12 hours), and nifedipine sublingually,
• ESR: ?arteritis. but the latter can cause precipitous falls in blood pressure.
• Serum urea and electrolytes: ?hypernatremia, Clonidine and methyldopa should be avoided because they
?hypokalemia, ?renal impairment. depress the CNS, making it difficult to distinguish further
• Urinalysis: blood, protein, casts, glomerular red blood CNS deterioration.
cells, 24 hour urinary catecholamines. Diuretics and fluid restriction should not be prescribed
• EKG: left ventricular hypertrophy. routinely because most patients are volume depleted,
• Chest x-ray: cardiomegaly, coarctation of the aorta. presumably due to a pressure-related diuresis.
• Renal ultrasound. If neurologic function deteriorates after blood pressure
• Antinuclear antibodies. reduction, antihypertensive therapy should be suspended
and the blood pressure allowed to increase. Subsequent
DIAGNOSIS blood pressure reductions should be effected more slowly.
A diagnosis of exclusion, requiring that the differential Magnesium sulfate intravenously is effective for eclampsia.
diagnoses (above) be ruled out. A loading dose of phenytoin 15–18 mg/kg (no faster than
50 mg/min) may be required if seizures occur.
TREATMENT
The goal of therapy is to reduce systemic vascular resistance PROGNOSIS
and thus reduce the mean arterial pressure gradually by no Usually reversible within hours of controlling the blood
more than 20–25% or to a diastolic blood pressure of pressure, but the course can be prolonged for days to weeks.
13.3 kPa (100 mmHg), whichever value is higher, during If the hypertension cannot be controlled, the outcome is
the first hour. The reason for the cautious reduction is usually fatal.
because the lower limit of cerebral bloodflow autoregulation
is reached when blood pressure is reduced by about one-
quarter and rapid reductions of blood pressure by more
than a half can precipitate cerebral ischemia or infarction.
The patient should be placed on a cardiac monitor and
intravenous access established.

322 323

322 Optic fundus showing papilledema, narrow arterioles, and a 323 Optic fundus showing features of grade IV hypertensive
macular star in a patient with hypertensive encephalopathy. retinopathy (malignant hypertension): superficial flame-shaped retinal
hemorrhages near the optic disc, soft exudates, retinal edema and
papilledema. (Courtesy of Mr M Wade, Department of Medical
Illustrations, Royal Perth Hospital,Western Australia.)
268 Vascular Diseases of the Nervous System

PITUITARY APOPLEXY 324

DEFINITION
Acute massive infarction or hemorrhage of the pituitary
gland.

EPIDEMIOLOGY
• Incidence: rare but probably underestimated because of
incomplete non-fatal pituitary infarction. Infarction of
more than 25% of the pituitary gland is reported to be
present in 1–3% of patients in large unselected autopsy
series.
• Age: evenly distributed between 20 and 70 years of age;
range 6–83 years. 324 Facial appearance of a patient with Cushing’s disease due to an
• Gender: M>F (2:1). ACTH-producing pituitary adenoma (which bled) featuring obesity,
hirsutism (a fine ‘downy’ coat), plethora and ecchymoses (due to
ANATOMY weakening and rupture of collagenous fibers in the dermis, exposing
Arterial blood supply to the pituitary: internal carotid artery heavily vascularized subcutaneous tissues).
via the superior and inferior hypophyseal arteries bilaterally.

PATHOLOGY VI; sympathetic chain; internal carotid and branches;


• Pituitary infarction (sometimes hemorrhagic) or hemorr- hypothalamus), leakage of blood or necrotic tissue into the
hage, most often in an enlarging pituitary adenoma. subarachnoid space and compromise of neurologic function.
• The pituitary may or may not have a pre-existing tumor, The initial event in post partum pituitary necrosis is
and if present, the tumor may be primary or metastatic. probably occlusive arterial spasm of the arteries to the anterior
• Primary pituitary tumors are most commonly a pituitary lobe and stalk. After a period of total ischemia, attempts at re-
adenoma. establishment of circulation subsequently lead to vascular
• Chromophobe adenomas predominate over eosinophilic congestion and thrombosis of the anterior lobe. The inferior
adenomas, merely reflecting the relative prevalence of hypophyseal arterial flow is relatively spared accounting for the
these tumors. Rarely, basophilic adenomas, cranio- preservation of the infundibular process in most circumstances.
pharyngiomas, and primary pituitary carcinoma occur.
• Metastatic pituitary tumors are most commonly breast CLINICAL FEATURES
and lung cancers, in patients with widespread disease. There are three main clinical syndromes.
• Most pituitary tumors are endocrinologically silent; a
minority are hormone-secreting: acromegaly, Cushing’s Acute apoplexy
disease. • Sudden severe retro-orbital headache.
• Cranial nerve dysfunction:
ETIOLOGY – II: impaired visual acuity; visual field defects (chiasmal
Predisposing factors compression).
• Pituitary tumor of any type. – III, IV, VI: ophthalmoplegia, diplopia.
• Pregnancy. – V1: facial pain.
• Diabetes. • Proptosis.
• Following obstetric hemorrhage: infarction of non- • Eyelid edema.
tumorous pituitary. • Horner’s syndrome.
• Raised intracranial pressure. • Nausea and vomiting.
• Bleeding disorders. • Altered consciousness, progressing to coma.
• Anticoagulants. • Focal hemispheric dysfunction: hemiparesis, seizures
• Upper respiratory infections. (carotid sinus compression, vasospasm due to SAH).
• Radiation therapy of pituitary tumors. • Fever.
• Trauma.
• Carotid angiography. Subacute meningeal irritation (blood/necrotic tumor
• Atherosclerosis. in subarachnoid space)
• Sickle cell trait. • Fever.
• Acromegaly. • Neck stiffness.
• Cushing’s disease (324). • Encephalopathy.
• Adrenalectomy.
• Mechanical ventilation. Subacute or chronic pituitary insufficiency months or
years after the event
PATHOPHYSIOLOGY • Diabetes insipidus (uncommon).
Infarction of the pituitary with secondary hemorrhage and • Syndrome of inappropriate antidiuretic hormone secretion.
edema destroys the pituitary and causes rapid expansion of the • Anterior hypopituitarism:
pituitary lesion, acute compression of adjacent structures (optic – Absence of lactation.
chiasm and tracts; cavernous sinus; cranial nerves III, IV, V, – Persistent amenorrhea.
Further Reading 269

– Lethargy. CSF
– Acute or delayed adrenal failure. • Cells: may contain red and/or white blood cells, and
necrotic tumor cells.
DIFFERENTIAL DIAGNOSIS • Protein: mildly elevated.
• Meningitis: viral, bacterial. • Gram stain, microscopy for organisms, and culture: negative.
• Meningoencephalitis: viral, bacterial.
• Brain abscess. Cerebral angiography
• Cavernous sinus thrombosis. Used if an intracavernous carotid aneurysm needs to be
• Carotid aneurysm. excluded.
• SAH: the association of SAH with ocular motor palsies in
pituitary apoplexy can be distinguished from SAH due to TREATMENT
rupture of an aneurysm by the presence of mixed, and Vigorous support
usually, bilateral ophthalmoplegias, multiple cranial nerve • Careful evaluation and monitoring of hormonal status.
palsies, and the presence of an afferent pupillary defect or • Immediate corticosteroid therapy in ‘stress dosages’
chiasmal pattern of field loss with pituitary apoplexy, (hydrocortisone 100 mg i.v. every 6–8 hours) due to the
although the latter goes unnoticed in obtunded patients. high incidence of acute adrenal insufficiency, and other
• Intracerebral hemorrhage. hormone replacement as appropriate.
• Transtentorial uncal herniation. • Fluid and electrolyte balance (cf. adrenal insufficiency,
• Mesencephalic infarction. diabetes insipidus).
• Parasellar tumor (meningioma or nasopharyngeal tumor). • Conservative management if consciousness, vision and
• Metastatic pituitary tumors. endocrine function are maintained.

INVESTIGATIONS Surgery
Cranial CT or MRI brain scan Early transphenoidal neurosurgical decompression of tissues
Plain scans often show a midline, hyperdense mass in the compromised by the rapidly expanding intrasellar mass is
pituitary fossa and an expanded sella due to hemorrhagic indicated if symptoms progress such as deteriorating visual
infarction of the pituitary gland. The mass may compress acuity, declining consciousness, or evidence of hypothalamic
the cavernous sinus and optic chiasm. Subarachnoid blood damage.
may be evident. The CT scan may be normal initially
however, in patients with small pituitary tumors in whom PROGNOSIS
radiographs of the pituitary fossa are also normal. • Highly variable and unpredictable clinical course.
• Potentially fatal.
Hypothalamic and pituitary function tests • Ophthalmoplegia often resolves spontaneously but
• Luteinizing hormone. recovery of vision is optimized by early decompression.
• Follicle stimulating hormone. • Multiple pituitary hormone deficiencies occur in most
• Thyrotrophin (thyroid stimulating hormone). patients after pituitary apoplexy and if unrecognized may
• Growth hormone. contribute to morbidity and mortality.
• Prolactin: low prolactin is very characteristic.
• Adrenocorticotropin.
• Cortisol.
• Thyroid-releasing and gonadotrophin-releasing hormone
stimulation tests.

FURTHER READING Hankey GJ (2001) Management of the first-time TIA.


Emergency Medicine, 13: 72–84.
Antiplatelet therapy
Algra A, van Gijn J (1996) Aspirin at any dose above
Hankey GJ, Warlow CP (1994) Transient ischaemic 30 mg offers only modest protection after cerebral
attacks of the brain and eye. WB Saunders/Ballière ischaemia. J. Neurol. Neurosurg. Psychiatry, 60:
NEUROVASCULAR SYNDROMES Tindall, London. 197–199.
Bamford J, Sandercock P, Dennis M, Burn J, Warlow Wolf PA, Clagett P, Easton JD, et al. (1999) Algra A, van Gijn J, Koudstaal PJ (1999) Secondary
C (1991) Classification and natural history of Preventing ischemic stroke in patients with prior prevention after cerebral ischaemia of presumed
clinically identifiable subtypes of cerebral infarction. stroke and transient ischemic attack. A statement arterial origin: is aspirin still the touchstone? J.
Lancet, 337: 1521–1526. for healthcare professionals from the Stroke Council Neurol. Neurosurg. Psychiatry, 66: 557–559.
Tatu L, Moulin T, Bogousslavsky J, Duvernoy H of the American Heart Association. Stroke, 30: Antiplatelet Trialists' Collaboration (1994) Collaborative
(1996) Arterial territories of the human brain: 1991–1994. overview of randomised trials of antiplatelet therapy-I:
Brainstem and cerebellum. Neurology, 47: Prevention of death, myocardial infarction, and stroke by
1125–1135. Epidemiology prolonged antiplatelet therapy in various categories of
Tatu L, Moulin T, Bogousslavsky J, Duvernoy H Brown RD, Petty GW, O’Fallon WM, et al. (1998) patients. BMJ, 308: 81–106.
(1998) Arterial territories of the human brain. Incidence of transient ischemic attack in Rochester, Derry S, Loke YK (2000) Risk of gastrointestinal
Cerebral hemispheres. Neurology, 50: 1699–1708. Minnesota, 1985–1989. Stroke, 29: 2109–2113. haemorrhage with long term use of aspirin: meta-
analysis. BMJ, 321: 1183–1187.
TRANSIENT ISCHEMIC ATTACKS (TIA) OF THE Diagnosis De Schryver ELLM, on behalf of the European/ Australian
BRAIN AND EYE Gunatilake S (1998) Rapid resolution of symptoms Stroke Prevention in Reversible Ischaemia Trial
General and signs of intracerebral haemorrhage: case (ESPRIT) Group (2000) Design of ESPRIT: An
Albers GW, Hart RG, Lutsep HL, et al. (1999) reports. BMJ, 316: 1495–1496. international randomised trial for secondary prevention
Supplement to the guidelines for the management Prognosis after non-disabling cerebral ischaemia of arterial origin.
of transient ischemic attacks. A statement from the Johnston SC, Gress DR, Browner WS, Sidney S Cerebrovasc. Dis., 10: 147–150.
Ad Hoc Committee on Guidelines for the manage- (2000) Short-term prognosis after emergency Hankey GJ, Sudlow CLM, Dunbabin DW (2000)
ment of transient ischemic attacks, Stroke Council, department diagnosis of TIA. JAMA, 284: Thienopyridines or aspirin to prevent stroke and
American Heart Association. Stroke, 30: 2901–2906. other serious vascular events in patients at high risk
2502–2511. of vascular disease. Stroke, 31: 1779–1784.
270 Vascular Diseases of the Nervous System

Mohr JP, Thompson JLP, Lazar RM, et al. (2001) A Epidemiology Smith Hammond CA, Goldstein LB, Zajac DJ, et al.
comparison of warfarin and aspirin for the preven- Chang CL, Donaghy M, Poulter N, et al. (1999) (2001). Assessment of aspiration risk in stroke patients
tion of recurrent ischemic stroke. N. Engl. J. Med., Migraine and stroke in young women: case-control with quantification of voluntary cough. Neurology,
345: 1444–1451. study. BMJ, 318: 13–18. 56: 502–506.
The Stroke Prevention in Reversible Ischemia Trial Gillum LA, Mamidipudi SK, Johnston SC (2000) Vestergaard K, Andersen G, Gottrup H, et al. (2001).
(SPIRIT) Study Group (1997) A randomised trial Ischemic stroke risk with oral contraceptives. A meta- Lamotrigine for central poststroke pain. A randomised
of anticoagulants versus aspirin after cerebral is- analysis. JAMA, 284: 72–78. controlled trial. Neurology, 56: 184–190.
chemia of presumed arterial origin. Ann. Neurol., Sudlow CLM, Warlow CP, for the International Stroke Wijdicks EFM (2000) Management of massive
42: 857–65. Incidence Collaboration (1997) Comparable studies hemispheric cerebral infarct. Is there a ray of hope?
Wilterdink JL, Easton JD (1999) Dipyridamole plus of the incidence of stroke and pathological types. Mayo Clin. Proc., 75: 945–952.
aspirin in cerebrovascular disease. Arch. Neurol., 56: Results from an international collaboration. Stroke,
1087–1092. 28: 491–499. Rehabilitation
Bakheit AMO, Thilmann AF, Ward AB, et al. (2000) A
Carotid revascularization Pathophysiology randomised, double-blind, placebo-controlled, dose-
Alamowitch S, Eliasziw M, Algra A, et al. (2001) Risk, Hademenos GJ, Alberts MJ, Awad I, et al. (2001) Ad- ranging study to compare the efficacy and safety of
causes, and prevention of ischaemic stroke in elderly vances in the genetics of cerebrovascular disease three doses of botulinum toxin type A (Dysport) with
patients with symptomatic internal-carotid-artery and stroke. Neurology, 56: 997–1008. placebo in upper limb spasticity after stroke. Stroke,
stenosis. Lancet, 357: 1154–1160. Kristián T, Siesjö BK (1998) Calcium in ischemic cell 31: 2402–2406.
Barnett HJM, Taylor DW, Eliasziw M, et al. for the North death. Stroke, 29: 705–18. Kwakkel G, Wagenaar C, Koelman TW, et al. (1997)
American Symptomatic Carotid Endarterectomy Trial Effects of intensity of rehabilitation after stroke. A
collaborators (1998) Benefit of carotid endarterectomy Imaging research synthesis. Stroke, 28: 1550–1556.
in patients with symptomatic moderate or severe Culebras A, Kase CS, Masdeu JC, et al. (1997) Practice Langhorne P, Legg L, on behalf of the Outpatient
carotid stenosis. N. Engl. J. Med., 339: 1415–1425. guidelines for the use of imaging in transient ischemic Therapy Trialists (1999) Therapy for stroke patients
Benavente O, Moher D, Pham B (1998) Carotid attacks and acute stroke. A report of the Stroke living at home. Lancet, 354: 1730–1731.
endarterectomy for asymptomatic carotid stenosis: a Council, American Heart Association. Stroke, 28:
meta-analysis. BMJ, 317: 1477–1480. 1480–1497. Community care
Biller J, Feinberg WM, Castaldo JE, et al. (1998) Keir SL, Wardlaw JM (2000) Systematic review of Han B, Haley WE (1999) Family caregiving for patients
Guidelines for carotid endarterectomy. A statement for diffusion and perfusion imaging in acute ischemic with stroke. Review and analysis. Stroke, 30:
healthcare professionals from a special writing group stroke. Stroke, 31: 2723–2731. 1478–1485.
of the Stroke Council, American Heart Association. Markus HS (1999) Transcranial Doppler ultrasound. J. Mant J, Carter J, Wade DT, Winner S (2000) Family
Stroke, 29: 554–562. Neurol. Neurosurg. Psychiatry, 67: 135–7. support for stroke: a randomised controlled trial.
CAVATAS Investigators (2001) Endovascular versus Wardlaw JM (2001) Radiology of stroke. J. Neurol. Lancet, 356: 808–813.
surgical treatment in patients with carotid stenosis Neurosurg. Psychiatry, 70 (Suppl 1): i7–i11.
in the carotid and vertebral transluminal angio- Prognosis and prognostic factors
plasty study (CAVATAS): a randomized trial. Acute treatment Bhalla A, Sankaralingam S, Dundas R, et al. (2000) Influence
Lancet, 357: 1729–1737. Barber PA, Demchuk AM, Zhang J, Buchan AM (2000) of raised plasma osmolality on clinical outcome after acute
Cina CS, Clase CM, Haynes RB (2001) Carotid Validity and reliability of a quantitative computed stroke. Stroke, 31: 2043–2048.
endarterectomy for symptomatic carotid tomography score in predicting outcome of hyperacute Hajat C, Hajat S, Sharma P (2000) Effects of poststroke
stenosis(Cochrane Review). In: The Cochrane Library, stroke before thrombolytic therapy. Lancet, 355: pyrexia on stroke outcome. A meta-analysis of studies
Issue 4, Oxford, Update Software. 1670–1674. in patients. Stroke, 31: 410–414.
European Carotid Surgery Trialists’ Collaborative Group Brott T, Bogousslavsky J (2000) Treatment of acute Hankey GJ, Jamrozik K, Broadhurst RJ, Forbes S, Burvill
(1998) Randomised trial of endarterectomy for recently ischemic stroke. N. Engl. J. Med., 343: 710–722. PW, Anderson CS, Stewart-Wynne EG (1998) Long-
symptomatic carotid stenosis: final results of the MRC Chen Z, Sandercock P, Pan H, et al. (2000) Indications term risk of first recurrent stroke in the Perth
European Carotid Surgery Trial (ECST). Lancet, 351: for early aspirin use in acute ischaemic stroke. A Community Stroke Study. Stroke, 29: 2491–2500.
1379–1387. combined analysis of 40 000 randomized patients from Hankey GJ, Jamrozik K, Broadhurst RJ, Forbes S, Burvill
Goldstein LB, Samsa GP, Matchar DB, Oddone EZ the Chinese Acute Stroke Trial and the International PW, Anderson CS, Stewart-Wynne EG (2000) Five-
(1998) Multicenter review of preoperative risk factors Stroke Trial. Stroke, 31: 1240–1249. year survival after first-ever stroke and related
for endarterectomy for asymptomatic carotid artery Davenport R, Dennis M (2000) Neurological emergencies: prognostic factors in the Perth Community Stroke
stenosis. Stroke, 29: 750–753. acute stroke. J. Neurol. Neurosurg. Psychiatry, 68: Study. Stroke, 31: 2080–2086.
Pritz MB (1997) Timing of carotid endarterectomy 277–288. Langhorne P, Tong BLP, Stott DJ (2000) Association
after stroke. Stroke, 28: 2563–2567. European Stroke Initiative (2000) European Stroke between physiological homeostasis and early recovery
Rothwell PM (2001) Carotid endarterectomy and Inititiative recommendations for stroke management. after stroke. Stroke, 31: 2526–2527.
prevention of stroke in the very elderly. Lancet, 357: Cerebrovasc. Dis., 10: 335–3351.
Gorelick P (2000) Neuroprotection in acute ischaemic Secondary prevention
1142–1143. Blood Pressure Lowering Trialists’ Collaboration (2000)
Rothwell PM, Slattery JM, Warlow CP (1996) Systematic stroke: a tale of for whom the bell tolls. Lancet,
355: 1925–1926. Effects of ACE inhibitors, calcium antagonists, and other-
comparison of the risks of stroke and death due to carotid blood pressure-lowering drugs: results of prospectively
endarterectomy for symptomatic and asymptomatic Gubitz G, Counsell C, Sandercock P, Signorini D
(2001) Anticoagulants for acute ischaemic stroke designed overviews of randomised trials. Lancet, 355:
carotid stenosis. Stroke, 27: 266–269. 1955–1964.
Rothwell PM, Warlow CP (1995) Is self-audit reliable? (Cochrane Review). In: The Cochrane Library, Issue
4. Oxford: Update Software. Bucher HC, Griffith LE, Guyatt GH (1998) Effect of HMG
Lancet, 346: 1623. CoA reductase inhibitors on stroke: a meta-analysis of
Rothwell PM, Warlow CP (1999) Prediction of benefit from Stroke Unit Trialists’ Collaboration (2001) Organised
inpatient (stroke unit) care for stroke (Cochrane randomised, controlled trials. Ann. Intern. Med., 128:
carotid endarterectomy in individual patients: a risk- 89–95.
modelling study. Lancet, 353: 2105–2110. Review). In: The Cochrane Library, Issue 4. Oxford:
Update Software. Eastern Stroke and Coronary Heart Disease Collaborative
Rothwell PM, Warlow CP, on behalf of the European Research Group (1998) Blood pressure,
Carotid Surgery Trialists’ Collaborative Group Wardlaw JM, del Zoppo G, Yamaguchi T (2001)
Thrombolysis for acute ischaemic stroke (Cochrane cholesterol, and stroke in eastern Asia. Lancet, 352:
(1999) Interpretation of operative risks of 1801–1807.
individual surgeons. Lancet, 353: 1325. Review). In: The Cochrane Library, Issue 4. Oxford:
Update Software. Gubitz G, Sandercock P (2000) Prevention of
Whitty CJM, Sudlow CLM, Warlow CP (1998) ischaemic stroke. BMJ, 321: 1455–1459.
Investigating individual subjects and screening Wardlaw JM (2001) Overview of Cochrane throm-
bolysis meta-analysis. Neurology, 57 (Suppl 2): Hankey GJ (1999) Smoking and risk of stroke. J.
populations for asymptomatic carotid stenosis can Cardiovasc. Risk, 6(4): 207–211.
be harmful. J. Neurol. Neurosurg. Psychiatry, 64: S69–S76.
Hankey GJ, Eikelboom JW (1999) Homocysteine and
619–623. Complications vascular disease. Lancet, 354: 407–413.
Writing Group (1998) Carotid stenting and Brittain KR, Peet SM, Castleden CM (1998) Stroke The Heart Outcomes Prevention Evaluation Study
angioplasty. Stroke, 29: 336–348. and incontinence. Stroke, 29: 524–528. Investigators (2000) Effects of an angiotensin-
STROKE Carson AJ, MacHale S, Allen K, et al. (2000) Depression converting enzyme inhibitor, ramipril, on death
General after stroke and lesion location: a systematic review. from cardiovascular causes, myocardial infarction,
Hankey GJ, Warlow CP (1999) Treatment and Lancet, 356: 122–126. and stroke in high-risk patients. N. Engl. J. Med.,
secondary prevention of stroke: evidence, costs, and Davenport RJ, Dennis MS, Wellwood I, Warlow CP 342: 145–153.
effects on individuals and populations. Lancet, 354: (1996) Complications following acute stroke. Stroke, The INDANA (Individual Data Analysis of
1457–1463. 27: 415–420. Antihypertensive intervention trials) Project
Hankey GJ (2002) Stroke: Your Questions Answered. Kim JS (2001) Delayed onset mixed involuntary Collaborators (1997) Effect of antihypertensive
Churchill Livingstone, Edinburgh, UK. movements after thalamic stroke. Clinical, radiological treatment in patients having already suffered from
Keir SL, Lindley RI (1999) Stroke medicine for the and pathophysiological findings. Brain, 124: stroke. Gathering the evidence. Stroke, 28:
geriatrician. Reviews in Clinical Gerontology, 9: 299–309. 2557–2562.
23–38. Koh M, Phan TG, Wijdicks EFM (2000) Management Lancaster T, Stead L, Silagy C, et al. (2000) Effectiveness
Warlow CP, Dennis MS, van Gijn J, Hankey GJ, of cerebellar infarction with mass effect. The of interventions to help people stop smoking: findings
Sandercock PAG, Bamford J, Wardlaw J (2000) Neurologist, 6: 172–176. from the Cochrane Library. BMJ, 321: 355–358.
Stroke: A practical guide to management 2nd edn. Korpelainen JT, Nieminen P, Myllylä VV (1999) Sexual PROGRESS Collaborative Group (2001) Randomised
Blackwell Scientific Publications, Oxford, UK. functioning among stroke patients and their spouses. trial of a perindopril-based blood pressure-lower-
Stroke, 30: 715–719. ing regimen among 6105 individuals with previous
O’Connell JE, Gray CS (1994) Treating hypertension stroke or transient ischaemic attack. Lancet, 358:
after stroke. BMJ, 308: 1523–1524. 1033–1041.
Further Reading 271

Prospective Studies Collaboration (1995) Cholesterol, Ross R (1999) Atherosclerosis – an inflammatory disease. Hart RG, Halperin JL (1999) Atrial fibrillation and
diastolic blood pressure and stroke: 13000 strokes in N. Engl. J. Med., 340: 115–126. thromboembolism: a decade of progress in stroke
450 000 people in 45 prospective cohorts. Lancet, Rothwell PM, Villagra R, Gibson R, Donders RCJM, prevention. Ann. Intern. Med., 131: 688–695.
346: 1647–1653. Warlow CP (2000) Evidence of a chronic systemic Lip GYH (1999) Thromboprophylaxis for atrial fibrillation.
Rodgers A, Neal B, MacMahon S (1997) The effects of cause of instability of atherosclerotic plaques. Lancet, Lancet, 353: 4–6.
blood pressure lowering in cerebrovascular disease. 355: 19–24. Thomson R, Parkin D, Eccles M, et al. (2000) Decision
Neurology Reviews International, 2(1): 12–15. Wald NJ, Law MR, Morris JK, Zhou X, Wong Y, Ward analysis and guidelines for anticoagulant therapy to
Sacco RL, Wolf PA, Gorelick PB (1999) Risk factors and ME (2000) Chlamydia pneumoniae infection and prevent stroke in patients with atrial fibrillation. Lancet,
their management for stroke prevention: Outlook mortality from ischaemic heart disease: large 355: 956–962.
for 1999 and beyond. Neurology, 53 (Suppl 4): prospective study. BMJ, 321: 204–207.
S15–S24. INFECTIVE ENDOCARDITIS
Sandercock P (2001) Statins for stroke prevention? Thrombosis Dajani AS, Bisno AL, Chung KJ, et al. (1990) Prevention
Lancet, 357: 1548–1549. Broze GJ Jnr (1992) The role of tissue factor pathway of bacterial endocarditis: recommendations of the
Viscoli CM, Brass LM, Kernan WN, et al. (2001) A inhibitor in a revised coagulation cascade. Semin. American Heart Association. JAMA, 264: 2919–2922.
clinical trial of estrogen-replacement therapy after Haematol., 29(3): 159–69. Heiro M, Nikoskelainen J, Engblom E, et al. (2000)
ischemic stroke. N. Engl. J. Med., 345: Coller BS (1997) Platelet GPIIb/IIIa antagonists: the first Neurologic manifestations of infective endocarditis. A 17-
1243–1249. anti-integrin receptor therapeutics. J. Clin. Invest., 99: year experience in a teaching hospital in Finland. Arch.
White HD, Simes RJ, Anderson NE, et al. (2000) 1467–1471. Intern. Med., 160: 2781–2787.
Pravastatin therapy and the risk of stroke. N. Engl. George JN (2000) Platelets. Lancet, 355: 1531–1539. Heiro M, Nikoskelainen J, Hartiala JJ, et al. (1998) Diagnosis
J. Med., 343: 317–326. Furie B, Furie BC (1992) Molecular and cellular biology of infective endocarditis. Sensitivity of the Duke vs Reyn
See TIA (above) for further reading about antiplatelet of blood coagulation. N. Engl. J. Med., 326: 800–806. Criteria. Arch. Intern. Med., 158: 18–24.
therapy and carotid revascularization, and Hirsh J, Weitz JI (1999) New antithrombotic agents. Ling R, James B (1998) White-centred retinal haemorrhages
Cardioembolic stroke (below) for further reading about Lancet, 353: 1431–1436. (Roth spots). Postgrad. Med. J., 74: 581–582.
anticoagulation. Rosenberg RD, Rosenberg JS (1984) Natural Mylonakis E, Calderwood SB (2001) Infective endo-
anticoagulant mechanisms. J. Clin. Invest., 74(1): 1–6. carditis in adults. N. Engl. J. Med., 345:
Miscellaneous Topol EJ, Byzova TV, Plow EF (1999) Platelet GPIIb- 1318–1330.
Yonehawa Y, Taub E (1999) Moyamoya disease: status IIIa blockers. Lancet, 353: 223–227.
1998. The Neurologist, 5: 13–23. van Kooten F, Ciabattoni G, Patrono C, Dippel DW, ANTIPHOSPHOLIPID SYNDROME AND OTHER
Koudstaal PJ (1997) Platelet activation and lipid PROTHROMBOTIC STATES
ATHEROSCLEROTIC ISCHEMIC STROKE peroxidation in patients with acute ischemic stroke. Bushnell CD, Goldstein LB (2000) Diagnostic testing
The French Study of Aortic Plaques in Stroke Group Stroke, 28: 1557–1563. for coagulopathies in patients with ischemic stroke.
(1996) Atherosclerotic disease of the aortic arch as Stroke, 31: 3067–3078.
a risk factor for recurrent ischemic stroke. N. Engl. Clinical Greaves M (1999) Antiphospholipid antibodies and
J. Med., 334: 1216–1221. Min WK, Park KK, Kim YS, et al. (2000) thrombosis. Lancet, 353: 1348–1353.
Hassan A, Markus HS (2000) Genetics and ischaemic Atherothrombotic middle cerebral artery territory Haviv YS (2000) Association of anticardiolipin
stroke. Brain, 123: 1784–1812. infarction. Topographic diversity with common antibodies with vascular injury: possible
occurrence of concomitant small cortical and mechanisms. Postgrad. Med. J., 76: 625–628.
Atherosclerotic plaque subcortical infarcts. Stroke, 31: 2055–2061. Markus HS, Hambley H (1998) Neurology and the
Davies MJ (1996) Stability and instability: two faces of blood: haematological abnormalities in ischaemic
coronary atherosclerosis. Circulation, 94: 2013–2020. CARDIOEMBOLIC STROKE stroke. J. Neurol. Neurosurg. Psychiatry, 64:
Davies MJ (1997) The composition of coronary artery General 150–159.
plaques. N. Engl. J. Med., 336: 1312–1314. Oppenheimer SM, Lima J (1998) Neurology and the Stewart AJ, Penman ID, Cook MK, Ludlam CA
Fuster V, Fallon JT, Badimon JJ, Nemerson Y (1997) The heart. J. Neurol. Neurosurg. Psychiatry, 64: 289–97. (1999) Warfarin-induced skin necrosis. Postgrad.
unstable atherosclerotic plaque: clinical significance and Med. J., 75: 233–235.
therapeutic intervention. Thromb. Haemost., 78(1): Atrial fibrillation
247–255. Yamanouchi H, Nagura H, Mizutani T, et al. (1997) DISSECTION OF THE CAROTID AND
Kullo IJ, Edwards WD, Schwartz RS (1998) Vulnerable Embolic infarction in nonrheumatic atrial fibrillation: VERTEBRAL ARTERIES
plaque: pathobiology and clinical implications. Ann. A clinicopathologic study in the elderly. Neurology, Crum B, Mokri B, Fulgham J (2000) Spinal
Intern. Med., 129: 1050–1060. 48: 1593–1597. manifestations of vertebral artery dissection.
Rauch V, Osende JI, Fuster V, et al. (2001) Throm- Neurology, 55: 304–306.
bus formation on atherosclerotic plaques: patho- Other embolic sources Guillon B, Levy C, Bousser MG (1998) Internal carotid
genesis and clinical consequences. Ann. Intern. De Castro S, Cartoni D, Fiorelli M, et al. (2000) artery dissection: an update. J. Neurol. Sci., 153:
Med., 134: 224–238. Morphological and functional characteristics of 146–158.
patent foramen ovale and their embolic implications. Guillon B, Tzourio C, Biousse V, et al. (2000) Arterial
Inflammation Stroke, 31: 2407–2413. wall properties in carotid artery dissection. An
Bittner V (1998) Atherosclerosis and the immune system. Freed LA, Levy D, Levine RA, et al. (1999) Prevalence ultrasound study. Neurology, 55: 663–666.
Arch. Intern. Med., 158: 1395–1396. and clinical outcome of mitral-valve prolapse. N. Hill MD, Hwa G, Perry JR (2000) Extracranial cervical
Cook PJ, Honeybourne D, Lip GYH, Beevers G, Wise R, Engl. J. Med., 341: 1–7. artery dissection. Stroke, 31: 799.
Davies P (1998) Chlamydia pneumoniae antibody Gilon D, Buonanno FS, Joffe MM, et al. (1999) Lack Leys D, Moulin Th, Stojkovic, Begey S, Chavot D,
titers are significantly associated with acute stroke and of evidence of an association between mitral-valve DONALD investigators (1995) Follow-up of patients
transientcerebral ischemia. The West Birmingham prolapse and stroke in young patients. N. Engl. J. with history of cervical artery dissection. Cerebrovasc.
Stroke Project. Stroke, 29: 404–410. Med., 341: 8–13. Dis., 5: 43–49.
Danesh J, Collins R, Peto R (1997) Chronic infections Libman R, Wein T (1999) ‘Newer’ cardiac sources of Schievink WI, Mokri B, O’Fallon WM (1994) Recurrent
and coronary heart disease: is there a link? Lancet, 350: embolic stroke. The Neurologist, 5: 231–246. spontaneous cervical-artery dissection. N. Engl. J.
430–436. Mas J-L, Arquizan C, Lamy C, et al. (2001) Recur- Med., 330: 393–397.
Danesh J, Whincup P, Walker M, et al. (2000) Low grade rent cerebrovascular events associated with patent Schievink WI, Wijdicks EF, Michels VV, et al. (1998)
inflammation and coronary heart disease: prospective foramen ovale, atrial septal aneurysm, or both. N. Heritable connective tissue disorders in cervical artery
study and updated meta-analysis. BMJ, 321: 199–204. Engl. J. Med., 345: 1740–1746. dissections. A prospective study. Neurology, 50:
Danesh J, Whincup P, Walker M, et al. (2000) Chlamydia 1166–1169.
pneumoniae IgG titres and coronary heart disease: Imaging
Blackshear JL, Brott TG (1999) Transesophageal Shievink WI (2001) Spontaneous dissection of the
prospective study and meta-analysis. BMJ, 321: carotid and vertebral arteries. N. Engl. J. Med.,
208–213. echocardiography in source-of-embolism evaluation:
the search for a better therapeutic rationale. Mayo Clin. 344: 898–906.
DeGraba TJ (1997) Expression of inflammatory mediators Silbert PL, Mokri B, Schievink WI (1995) Headache and
and adhesions molecules in human atherosclerotic Proc., 74: 941–945.
Channon KM, Banning AP (1999) Echocardiography in neck pain in spontaneous internal carotid and
plaque. Neurology, 49 (Suppl 4): S15–S19. vertebral artery dissections. Neurology, 45:
Grau AJ, Buggle F, Becher H, et al. (1998) Recent stroke and thromboembolism: transoesophageal
imaging for all? Q. J. Med., 92: 619–621. 1517–1522.
bacterial and viral infection is a risk factor for
cerebreovascular ischemia: Clinical can biochemical Acute treatment CENTRAL NERVOUS SYSTEM VASCULITIS
studies. Neurology, 50: 196–203. Berge E, Abdelnoor M, Nakstad PH, et al. (2000) Demiroglu H, Özcebe OI, Barista I, et al. (2000)
Gurfinkel E, Bozovich G, Daroco A, Beck E, Mautner Low molecular-weight heparin versus aspirin in Interferon alfa-2b, colchicine, and benzathine penicillin
B, for the ROXIS Study Group (1997) Randomised patients with acute ischaemic stroke and atrial versus colchicine and benzathine penicillin in Behçet's
trial of roxithromycin in non-Q-wave coronary fibrillation: a double-blind randomised study. disease: a randomised trial. Lancet, 355: 605–609.
syndromes: ROXIS Pilot Study. Lancet, 350: 404–407. Lancet, 355: 1205–1210. Hankey GJ (1991) Isolated angiitis/angiopathy of the
Koenig W (2000) Heart disease and the inflammatory central nervous system. Cerebrovasc. Dis., 1: 2–15.
response. BMJ, 321: 187–188. Secondary prevention Kontogiannis V, Powell RJ (2000) Behçet’s disease.
Markus HS, Mendall MA (1998) Helicobacter pylori Cromheecke ME, Levi M, Colly LP (2000) Oral Postgrad. Med. J., 76: 629–637.
infection: a risk factor for ischaemic cerebrovascular anticoagulation self-management and management by a Lanthier S, Lortie A, Michaud J, et al. (2001) Isolat-
disease and carotid atheroma. J. Neurol. Neurosurg. specialist anticoagulation clinic: a randomised cross-over ed angiitis of the CNS in children Neurology, 56:
Psychiatry, 64: 104–107. comparison. Lancet, 356: 97–102. 837–842.
Ridker PM, Cushman M, Stampfer MJ, Tracy RP, EAFT (European Atrial Fibrillation Trial) Study Group Moore PM (1998) Central nervous system vasculitis. Curr.
Hennekens CH (1997) Inflammation, aspirin, and the (1993) Secondary prevention in non-Rheumatic atrial Opin. Neurol., 11: 241–246.
risk of cardiovascular disease in apparently healthy men. fibrillation after transient ischaemic attack or minor stroke.
N. Engl. J. Med., 336: 973–979. Lancet, 342: 1255–1262.
272 Vascular Diseases of the Nervous System

Moore PM, Richardson B (1998) Neurology of the Koivisto T, Vanninen R, Hurskainen H, et al. (2000) Diagnosis
vasculitides and connective tissue diseases. J. Neurol. Outcomes of early endovascular versus surgical treat- Chui HC, Mack W, Jackson JE, et al. (2000) Clinical
Neurosurg. Psychiatry, 65: 10–22. ment of ruptured cerebral aneurysms. A prospective criteria for the diagnosis of vascular dementia. A
Numano F, Okawara M, Inomata H, Kobayashi Y (2000) randomised study. Stroke, 31: 2369–2377. multicenter study of comparability and interrater
Takayasu’s arteritis. Lancet, 356: 1023–1025. Roos YBWEM, Rinkel GJE, Vermeulen M, Algra A, van reliability. Arch. Neurol., 57: 191–196.
Sakane T, Takeno M, Suzuki N, Inaba G (1999) Behçet’s Gijn J (2001) Antifibrinolytic therapy for aneurysmal Pohjasvaara T, Mäntylä R, Ylikoski R, et al. (2000)
disease. N. Engl. J. Med., 341: 1284–1291. subarachnoid haemorrhage (Cochrane Review). In: The Comparison of different clinical criteria (DSM-III,
Cochrane Library, Issue 4. Oxford: Update Software. ADDTC, ICD-10, NINDS-AIREN, DSM-IV) for
GIANT CELL ARTERITIS the diagnosis of vascular dementia. Stroke, 31:
Hayreh SS (2000) Steroid therapy for visual loss in Prognosis 2952–2957.
patients with giant-cell arteritis. Lancet, 355: Hop JW, Rinkel GJE, Algra A, van Gijn J (1997) Case- Wetterling T, Kanitz R-D, Borgis K-J (1996)
1572–1574. fatality rates and functional outcome after Comparison of different diagnostic criteria for
Jover JA, Hernández-García C, Morado IC, et al. subarachnoid hemorrhage – a systematic review. vascular dementia (ADDTC, DSM-IV, ICD-10,
(2001). Combined treatment of giant-cell arteritis Stroke, 28: 660–664. NINDS-AIREN). Stroke, 27: 30–36.
with methotrexate and prednisone. A randomised,
double-blind, placebo-controlled trial. Ann. Intern. UNRUPTURED ANEURYSMS HYPERTENSIVE ENCEPHALOPATHY
Med., 134: 106–114. Broderick JP (2000) Coiling, clipping, or medical Vaughan CJ, Delanty N (2000) Hypertensive
Swannell AJ (1997) Polymyalgia rheumatica and management of unruptured intracranial aneurysms: time emergencies. Lancet, 356: 411–417.
temporal arteritis: diagnosis and management. BMJ, to randomise? Ann. Neurol., 48: 5–6.
314: 1329–1332. International Study of Unruptured Intracranial Aneurysms PITUITARY APOPLEXY
Zeidler M, Hughes T, Zeman A (2000) Confused by Investigators (1998) Unruptured intracranial aneurysms Anderson JR, Antoun N, Burnet N, Chatterjee K,
arteritis. Lancet, 355: 374. – risk of rupture and risks of surgical intervention. N. Engl. Edwards O, Pickard JD, Sarkies N (1999)
J. Med., 339: 1725–1733. Neurology of the pituitary gland. J. Neurol.
PRIMARY INTRACEREBRAL HEMORRHAGE Wardlaw JM, White PM (2000) The detection and Neurosurg. Psychiatry, 66: 703–721.
Broderick JP, Adams HP Jr, Barsan W, et al. (1999) management of unruptured intracranial aneurysms. Biousse V, Newman NJ, Oyesiku NM (2001) Precip-
Guidelines for the management of spontaneous Brain, 123: 205–221. itating factors in pituitary apoplexy. J. Neurol. Neu-
intracerebral haemorrhage. A statement for healthcare rosurg. Psychiatry, 71: 542–545.
professionals from a special writing group of the Stroke CEREBRAL VENOUS THROMBOSIS
Council, American Heart Association. Stroke, 30: Allroggen H, Abbott RJ (2000) Cerebral venous sinus
905–915. thrombosis. Postgrad. Med. J. 76: 12–15.
Fernandes HM, Gregson B, Siddique S, Mendelow AD Benamer HTS, Bone I (2000) Cerebral venous throm-
(2000) Surgery in intracerebral hemorrhage. The bosis. J. Neurol. Neurosurg. Psychiatry, 69:
uncertainty continues. Stroke, 31: 2511–2516. 427–430.
Knudsen KA, Rosand J, Karluk D, Greenberg SM Biousse V, Ameri A, Bousser M-G (1999) Isolated
(2001) Clinical diagnosis of cerebral amyloid intracranial hypertension as the only sign of cerebral
angiopathy: Validation of the Boston criteria. venous thrombosis. Neurology, 53: 1537–1542.
Neurology 56: 537–539. de Bruijn SFTM, Stam J, for the CVST Study Group
Querishi AI, Tuhrim S, Broderick JP, et al. (2001) (1999) Randomized, placebo-controlled trial of
Spontaneous intracerebral hemorrhage. N. Engl. J. anticoagulant treatment with low-molecular-weight
Med., 344: 1450–1460. heparin for cerebral sinus thrombosis. Stroke, 30:
Sacco RL (2000) Lobar intracerebral haemorrhage. N. 484–488.
Engl. J. Med., 342: 276–279. de Bruijn SFTM, Budde M, Teunisse S, et al. (2000)
Long-term outcome of cognition and functional
ARTERIOVENOUS MALFORMATION health after cerebral venous sinus thrombosis.
Al-Shahi R, Warlow C (2001) A systematic review of Neurology, 54: 1687–1689.
the frequency and prognosis of arteriovenous mal- de Bruijn SFTM, de Haan RJ, Stam J, for the Cere-
formations of the brain in adults. Brain, 124: bral Venous Sinus Thrombosis Study Group
1900–1926. (2001) Clinical features and prognostic factors of
The Arteriovenous Malformation Study Group (1999) cerebral venous sinus thrombosis in a prospective
Arteriovenous malformations of the brain in adults. series of 59 patients. J. Neurol. Neurosurg. Psychi-
N. Engl. J. Med., 340: 1812–1818. atry, 70: 105–108.
SUBARACHNOID HEMORRHAGE VASCULAR DEMENTIA
Epidemiology General
The ACROSS Group (2000) Epidemiology of Amar K, Wilcock G (1996) Vascular dementia. BMJ,
aneurysmal subarachnoid haemorrhage in Australia 312: 227–231.
and New Zealand. Incidence and case fatality from
the Australasian Cooperative Research on Subarach- Epidemiology
noid Haemorrhage Study (ACROSS). Stroke, 31: Bousser MG, Tournier-Lasserve E. (2001) Cerebral
1843–1850. autosomal dominant arteriopathy with subcortical
Ingall T, Asplund K, Mähönen M, et al. (2000) A infarcts and leukoencephalopathy: from stroke to
multinational comparison of subarachnoid vessel wall physiology. J. Neurol. Neurosurg. Psy-
haemorrhage epidemiology in the WHO Monica chiatry, 70: 285–287.
Stroke Study. Stroke, 31: 1054–1061. Hebert R, Lindsay J, Verreault R, et al. (2000)
Vascular dementia. Incidence and risk factors in the
Etiology Canadian Study of Health and Aging. Stroke, 31:
Schievink WI, Wijdicks EFM (2000) Origin of pretruncal 1487–1493.
nonaneurysmal subarachnold haemorrhage: Ruptured
vein perforating artery, or intramural haematoma? Mayo Pathogenesis
Clin. Proc., 75: 1169–1173. de Groot JC, de Leeuw F-E, Oudkerk M, et al. (2000)
Cerebral white matter lesions and cognitive function:
Diagnosis The Rotterdam scan study. Ann. Neurol., 47:
Edlow JA, Caplan LR (2000) Avoiding pitfalls in the 145–1451.
diagnosis of subarachnoid haemorrhage. N. Engl. J. Garde E, Mortensen EL, Krabbe K, et al. (2000)
Med., 342: 29–36. Relation between age-related decline in intelligence
Ruigrok YM, Rinkel GJE, Buskens E, et al. (2000) and cerebral white-matter hyperintensities in healthy
Perimesencephalic haemorrhage and CT angiography. octogenarians: a longitudinal study. Lancet, 356:
A decision analysis. Stroke, 31: 2976–2983. 628–634.
van Gijn J (1997) Slip-ups in the diagnosis of subarachnoid Merino JG, Hachinski V (2000) Leukoaraiosis. Reifying
haemorrhage. Lancet, 349: 1492–1493. rarefaction. Arch. Neurol., 57: 925–926.
van Gijn J, Rinkel GJE (2001) Subarachnold haemorrhage: Reed BR, Eberling JL, Mungas D, et al. (2000)
diagnosis, causes and management. Brain, 124: Memory failure has different mechanisms in
249–278. subcortical stroke and Alzheimer’s disease. Ann.
Neurol., 48: 275–284.
Treatment van Gijn J (1998) Leukoaraiosis and vascular dementia.
Brilstra EH, Rinkel GJE, Algra A, van Gijn J (2000). Neurology, 51 (Suppl 3): S3–S8.
Rebleeding, secondary ischaemia, and timing of van Gijn J (2000) White matters: small vessels and slow
operation in patients with subarachnold haemorrhage. thinking in old age. Lancet, 356: 612–613.
Neurology, 55: 1656–1660.
Feigin VL, Rinkel GJE, Algra A, Vermeulen M, van Gijn Clinical features
J (2001) Calcium antagonists for aneurysmal Dichgans M, Mayer M, Uttner I, et al. (1998) The
subarachnoid haemorrhage (Cochrane Review). In: The phenotypic spectrum of CADASIL: clinical findings
Cochrane Library, Issue 4. Oxford: Update Software. in 102 cases. Ann. Neurol., 44: 731–739.
Chapter Eleven 273

Infections of the
Nervous System
BACTERIAL INFECTIONS

ACUTE PYOGENIC (BACTERIAL) • Incidence rates are influenced by country, ethnic group,
MENINGITIS social class and deprivation, and immunization
programmes.
• Lifetime prevalence: 1(95% CI: 0.8–2) per 1000.
DEFINITION • Age: any age (see Table 33); most common in the first
Inflammation of the leptomeninges (pia and arachnoid month of life.
membranes) caused by bacterial infection.

EPIDEMIOLOGY
• Annual incidence: 1.9 per 100 000 for Neisseria menin-
gitidis; 1.6 per 100 000 for Haemophilus influenzae; 1.0
per 100 000 for Streptococcus pneumoniae.

Table 33 Empirical therapy for suspected bacterial meningitis*

Age Likely bacterial pathogens Empirical therapy**


0–4 weeks Escherichia coli (50%) Amoxycillin + cefotaxime, or amoxycillin + aminoglycoside
Group B Streptococci(30%)
Listeria monocytogenes (10%)
Klebsiella pneumoniae
4–12 weeks Escherichia coli Amoxycillin + a third generation cephalosporin
Group B Streptococci (S. agalactiae) (cefotaxime or ceftriaxone)
Listeria monocytogenes
Haemophilus influenzae
Streptococcus pneumoniae
Neisseria meningitidis
3 months–18 years Haemophilus influenzae Third generation cephalosporin (cefotaxime or ceftriaxone)
Neisseria meningitidis ± amoxycillin***, or amoxycillin + chloramphenicol
Streptococcus pneumoniae
18–50 years Streptococcus pneumoniae (40%) Third generation cephalosporin (cefotaxime or ceftriaxone)
Neisseria meningitidis (30%) ± amoxycillin**
>50 years Streptococcus pneumoniae Amoxycillin + a third generation cephalosporin
Neisseria meningitidis (cefotaxime or ceftriaxone)
Aerobic Gram-negative bacilli
Listeria monocytogenes
*Applies only to the immunocompetent patient
**Add vancomycin to empirical regime when pneumococcal meningitis highly resistant to penicillin or cephalosporin is suspected
***Add if L. monocytogenes meningitis suspected (i.e. deficiencies in cell-mediated immunity)
274 Infections of the Nervous System: Bacterial

PATHOLOGY meningitidis predominated in older children and young


The fundamental process is inflammation of the lepto- adults (5–29 years old) and S. pneumoniae in older adults.
meninges (325, 326). Complications include vasculitis, During the 1990s widespread use of conjugate H.
cerebral infarction, hydrocephalus (327), brain abscesses, influenzae type b (Hib) vaccines, consisting of protein
and cerebral edema. combined with the Hib polysaccharide capsule, has virtually
eliminated Hib disease from several developed countries
ETIOLOGY (and is now being introduced in developing countries). This
The most common organisms causing meningitis are: reduction means that S. pneumoniae and N. meningitidis
• Haemophilus influenzae: 45% of cases. have become the predominant causes of meningitis in
• Streptococcus pneumoniae (pneumococcus): 18%. children. There are at least 13 known groups of
• Neisseria meningitidis (meningococcus): 14%. meningococcal disease, but the most important are group
B (which is the most common, accounting for about 60%
However, there are important differences in the patterns of cases) and group C (which accounts for about 40% of
of organisms encountered in different age groups (see Table cases and causes most deaths). Meningitis due to
33). Listeria monocytogenes, although uncommon, occurs meningococcus group C is most common in babies, and
especially during pregnancy and the neonatal period. next most common in adolescents (15–17 years old). A
Among neonates (<1 month of age), group B streptococci second epidemiologic trend is the worldwide increase in
(S. agalactiae) and coliforms (particularly Escherichia coli, infection with strains of S. pneumoniae that are resistant to
but also Proteus and Pseudomonas) predominate. penicillin and other β-lactam antibiotics, mediated by
In the 1980s, H. influenzae was the most common cause alterations in the penicillin-binding proteins involved in the
of meningitis in children 1 month–4 years of age, N. synthesis of bacterial cell walls.

325 326

327

325 Base of the brain of a patient who died from acute pyogenic
meningitis showing purulent meningeal exudate. (Courtesy of
Professor BA Kakulas, Royal Perth Hospital,Western Australia.)

326 Histologic section of the meninges and cerebral cortex of a


patient who died of acute pyogenic meningitis showing neutrophilic
infiltration of the subarachnoid space, pia, arachnoid and
Virchow–Robin spaces of the outer part of the brain. Bacteria are
numerous, lying free in the subarachnoid space and inside neutrophilic
leukocytes (phagocytosis).

327 CT brain scan showing early hydrocephalus in a patient with


pneumococcal meningitis.Their symptoms had started 18 hours
previously.
Acute Pyogenic (Bacterial) Meningitis 275

Risk factors base of the brain or in the arachnoid granulations causing


CSF leak due to: hydrocephalus. Infection may become loculated to form
• Recent craniotomy: risk of staphylococcal meningitis. abscesses. Cerebral edema may develop also and raise
• Recent open skull fracture: risk of pneumococcal meningitis, intracranial pressure.
and Gram-negative bacillary (coliform) pathogens, such as
Klebsiella spp., E. coli and Pseudomonas spp. CLINICAL FEATURES
Meningeal inflammation
PATHOGENESIS Fever, headache, meningismus (neck stiffness), and signs of
Organisms may reach the meninges by direct spread from cerebral dysfunction (confusion, delirium, vomiting or
adjacent structures in the skull and spine (middle ear, declining consciousness) are found in about 85% of patients
paranasal sinuses, skull fractures) and via the blood stream. at presentation. Meningismus is accompanied by Kernig’s
Bacterial cell wall materials and bacterial products such and/or Brudzinski signs in about half of adults.
as endotoxin stimulate the local release of proinflammatory
cytokines such as tumor necrosis factor alpha, interleukin 1 Complications
and interleukin 6 from host cells and thereby initiate the • Cranial nerve palsies, particularly nerves III, IV, VI and
inflammatory response. Hyperemia of the meninges is VII (30% of cases).
followed by migration of neutrophils into the walls of blood • Deafness due to cochlear damage either by a direct effect of
vessels and into the subarachnoid space where the bacterial toxins or by an indirect cytokine-mediated effect.
inflammatory infiltrate extends along cranial and spinal • Epileptic seizures (30%) or focal neurologic signs
nerves. Foci of necrosis develop in vessel walls, sometimes (10–20% of cases), due to inflammation and thrombosis
with thrombosis sufficient to cause cerebral infarction and of cortical arteries and veins and venous sinuses (causing
seizures. Fibrino-purulent exudate accumulates in the cerebral infarction), or subdural effusion.
subarachnoid space and may block the flow of CSF at the • Signs of increased intracranial pressure (coma, cranial nerve
III palsy, hypertension, bradycardia) due to hydrocephalus
or cerebral edema, or subdural effusion. Papilledema is
328, 329 328 unusual at presentation (<1% of cases), and should suggest
Purpuric skin rash. an alternative diagnosis at that time, but is more common
Rash in an among acutely ill patients who are deteriorating.
unconscious child
(328) and adult ETIOLOGIC CLUES
(329) with • Sources of infection: evidence of suppuration may be
meningococcal present in the ears (otitis media), paranasal sinuses, skin,
meningitis and lungs, and heart (infective endocarditis).
septicemia. • Skin rash: primarily on the limbs, that is typically
(Courtesy of Dr erythematous and macular early in the infection, but may
AM Chancellor, quickly evolve into a petechial phase with further
Tauranga, New coalescence into a purpuric form, is present in about 50%
Zealand.) of patients with meningococcemia, with or without
meningitis (328, 329).
• Features of a rhombencephalitis, such as ataxia, cranial
nerve palsies, and nystagmus early in the clinical course
may indicate Listeria monocytogenes meningitis.

Atypical presentations
Neonates and infants
Change in affect or state of alertness, irritability, lethargy,
listlessness, feeding difficulties, weak suck, high-pitched
crying, fretfulness, vomiting, diarrhea, respiratory distress,
temperature instability (fever or hypothermia), or jaundice.
329 Neck stiffness may be absent. A bulging fontanelle is found
in one-third of cases and usually occurs late in the course of
the illness.

Elderly patients (particularly with diabetes or


cardiopulmonary disease)
May present insidiously with lethargy or obtundation, no
fever, and variable signs of meningeal inflammation.

Neutropenic patients
Symptoms and signs may be subtle because of the impaired
ability to mount an inflammatory response in the
subarachnoid space.
276 Infections of the Nervous System: Bacterial

DIFFERENTIAL DIAGNOSIS Other


Meningitis due to non-bacterial causes • Subarachnoid hemorrhage (see p.249).
Aseptic meningitis (see p.291) • Acute prolapsed cervical disc (see pp.545, 550).
The physical signs are not so marked and the illness is not • Brain abscess and subdural empyema (see p.284).
as severe and prolonged as bacterial meningitis. A CSF • Migraine (see p.95).
finding of >2000 white cells/mm3, >1180 neutro- • Acute tonsillitis, parotitis, cervical lymphadenitis and
phils/mm3, protein >22 g/l (>220 mg/dl), glucose pneumonia: may cause neck pain in children but
<1.9 mmol/l (<34 mg/dl), or a glucose ratio below 0.23 meningism is uncommon.
are individual CSF predictors of bacterial, rather than viral
meningitis, with 99% certainty or better. INVESTIGATIONS
Brain imaging
Tuberculous meningitis The imaging modality of choice is either CT or MRI; typically
CSF white cell count predominantly lymphocytic and seldom neither will show any abnormality in the early stages of uncom-
>1000/mm3; CSF protein can be very high (>10 g/l plicated meningitis. The main reason for imaging in suspected
[1000 mg/dl]). meningitis is to exclude other causes of headache, focal
neurologic signs or papilledema (such as a mass lesion), and to
Fungal meningitis (Cryptococcus neoformans, Candida, ensure that it is safe to do a lumbar puncture (LP).
aspergillus, histoplasma) In cases presenting later, or with proven bacterial
Insidious onset, often immunosuppressed (HIV infection, meningitis, the following features may be seen:
lymphoma, leukemia, other malignancies), with capsular • Hydrocephalus (dilated ventricles) (327), enlarged CSF
antigen present in serum and CSF. spaces (basal cisterns and interhemispheric fissure).
• Sediment in the posterior horns of the lateral ventricles
Protozoal meningitis (pus) (330).
Toxoplasmosis causes meningo-encephalitis and brain • Absence of the basal cisterns (due to inflamed meninges
abscesses in people who are usually immunosuppressed with and pus).
HIV infection, malignancy or immunosuppressive therapy. • Areas of altered density in the brain parenchyma
representing infarction (secondary to vasculitis) or
cerebritis (these areas may enhance).

Table 34 Cerebrospinal fluid profiles in various forms of meningo-encephalitis


Infection Cells/mm3 Protein Glucose Bacteriology
(g/l [mg/dl]) (mmol/l [mg/dl])
Normal 0–4 mononuclears 0.15–0.45 (15–45) 2.9–4.6 (52–83) Negative
Bacterial meningitis: acute 103 polymorphs Increased Markedly decreased Bacteria +ve
Bacterial meningitis: 102–103 mononuclears or Normal or increased Normal or decreased Bacteria +ve or -ve
partially treated polymorphs
Aseptic meningitis* 10–102 mononuclears Normal or increased Normal** Viruses
Meningoencephalitis 10–102 mononuclears Normal or increased Normal** Viruses or -ve
Tuberculous meningitis 10–102 mononuclears Increased Decreased Acid-fast bacilli +ve
Fungal meningitis 10–102 mononuclears Increased Decreased Organism +ve
Carcinomatous meningitis 10–102 mononuclears Increased Normal or decreased Malignant cells
Syphilitic meningitis 10–102 mononuclears Increased Normal** Serologic tests +ve
Raised gamma globulins
Subacute sclerosing Normal Increased Normal Measles antibody titer
panencephalitis Raised gamma globulins very high
Creutzfeldt–Jakob disease Normal Normal Normal Normal
Progressive multifocal Normal Normal Normal Normal
leukoencephalopathy
* Polymorphonuclear leukocytes may predominate early
** CSF glucose may be low in mumps and herpes simplex virus infections and acute syphilitic meningitis
Acute Pyogenic (Bacterial) Meningitis 277

• After i.v. contrast, periventricular enhancement • Serology for borrelia, brucellosis, legionella, leptospirosis,
(indicating ventriculitis) or enhancement in the basal and toxoplasmosis.
cisterns (from inflamed meninges).
• MRI is more sensitive to the enhancement following Other
contrast than CT. • Culture (bacterial and viral) from septic sites, biopsy
• Evidence of local infection such as sinusitis or mastoiditis material, throat swab and feces.
should be sought (opacified sinuses). • Chest x-ray may show signs of infection by pneumococci,
• Late complications include generalized atrophy, mycoplasma, legionella and tuberculosis (TB).
infarction, subdural empyema, mycotic aneurysms and • Skin scrapings may identify meningococci that are
loculated CSF collections. present in the skin rash. The lesion is scraped with the
• In cases of recurrent meningitis, a connection between point of a sterile needle until blood just starts to appear.
the nasal or middle ear spaces and the CSF should be The blood is then blotted on to microscope slides,
suspected and a contrast CT cisternogram performed to allowed to dry, and examined for Gram-negative
identify a leak when the patient is well. diplococci. This can provide results rapidly and, unlike
examination of blood and CSF, is not greatly affected by
CSF (and simultaneous blood glucose) (see Table 34) prior antibiotic treatment.
If no focal neurologic signs are present, LP should be
performed immediately and the CSF examined for cell DIAGNOSIS
count, protein, glucose, polymerase chain reaction (PCR), The diagnosis is based on the finding of an increased
and cultured: number of white cells in the CSF (CSF pleocytosis) and
• White cell count: usually 1000–5000/mm3 with a demonstration of bacteria in the CSF whether by Gram
neutrophil predominance; lymphocyte predominance in stain, CIE, culture or PCR, in a patient with consistent
10%, particularly L. monocytogenes meningitis and newborns clinical features.
with Gram-negative bacillary meningitis. A very low CSF
white cell count (0–20/mm3) in conjunction with high MANAGEMENT
CSF bacterial concentrations indicates a poor prognosis. Antibacterial chemotherapy
• Gram stain: positive in 60–90% of untreated cases and Principles
40–60% of partially treated patients. Specificity nearly 100%. • The need to commence antibiotic treatment as soon as
• Protein: raised (1–5 g/l [100–500 mg/dl]) in virtually the diagnosis is suspected, even before admission to
all patients. hospital or performing a lumbar puncture.
• Glucose: low (below 2.22 mmol/l [40 mg/dl]) in 60% • The need for bactericidal activity in CSF.
of patients. The CSF: serum glucose ratio is below 0.31 • Factors influencing bactericidal activity in CSF:
in 70% of cases. – Permeability of the blood–brain barrier.
• Culture: positive in 70–85% of untreated and <50% of Continued overleaf
partially treated meningitis.

Latex agglutination test for bacterial antigen (H. influenzae 330


type b, S. pneumoniae, N. meningitidis, Escherichia coli K1,
and S. agalactiae)
• Indicated if clinical features and CSF profile are
consistent with bacterial meningitis but CSF gram stain
is negative.
• Sensitivity: 50–100%, specificity: high.

Polymerase chain reaction


The most sensitive method but frequently false positive
results are obtained. Further refinements of this technique
are required to confirm its usefulness in patients with
bacterial meningitis in whom the CSF Gram stain, bacterial
antigen tests and culture are negative.

Reagent strips
If no facilities for laboratory examination of CSF are available,
the CSF can be tested with reagent strips that measure protein
and glucose concentrations and leukocyte counts in the urine.

Blood
• Full blood count and ESR: a neutrophil leukocytosis and
raised ESR are typical but not invariable nor specific:
severe viral infections can cause similar changes.
• Blood cultures are essential because bacteria are often 330 CT brain scan from an elderly lady who had been unwell for
easier to isolate from the blood than the CSF. several days with headache and confusion.There is debris in the
• Acute and convalescent viral serology in blood and CSF, occipital horns of the lateral ventricle (arrows) which is pus due to
including HIV serum antibody in at-risk individuals. pyogenic meningitis.
278 Infections of the Nervous System: Bacterial

Principles (continued) • Hyperventilation, to maintain arterial pCO2 between 3.6


– Characteristics of the antibiotic (molecular size, protein and 4 kPa (27 and 30 mmHg), but this may lead to a
binding, lipid solubility, degree of ionization). reduction in cerebral blood flow below ischemic thresholds.
– CSF pH, protein concentration, temperature. • Barbiturate therapy (pentobarbitone [pentobarbital]), in high-
• Potential hazards of bactericidal activity in CSF: release dose titrated to development of a burst-suppression pattern on
of biologically active cell-wall products, increasing the the EEG, may help to protect the brain from ischemic insult
production of cytokines in CSF, exacerbating inflam- (by causing vasoconstriction in normal tissues, thus shunting
mation and damaging the blood–brain barrier. blood to ischemic tissue) and decrease metabolic demands of
See management algorithm (331). the brain. This is of unproven benefit in bacterial meningitis.

Types of therapy Treatment of complications


• Empirical (see Table 33). Specific treatment is indicated for specific complications
• Specific (see Table 35): an important epidemiologic trend such as seizures and hydrocephalus.
is the worldwide increase in infection with strains of
S. pneumoniae that are resistant to penicillin and other Prophylactic treatment
β-lactam antibiotics, mediated by alterations in the Prophylaxis prevents secondary cases by eradicating
penicillin-binding proteins involved in the synthesis of nasopharyngeal carriage in the short term
bacterial cell walls.
Meningococcal infections
Supportive measures • Rifampicin (rifampin) 10 mg/kg/day for 4 days is recom-
• General nursing care. mended for close contacts (usually household) and the patient.
• Attention to the airway. • The meningococcal C vaccine is a conjugate vaccine
• Maintenance of adequate hydration: fluids should be administered to infants, children and young adults. It
replaced and maintained, and not restricted. overcomes the problems of the existing polysaccharide
• Antipyretic measures. vaccine which offers limited protection against groups B
and C, and does not protect infants <2 years old (but is
Adjunctive therapy 75–90% effective in older children and adults).
Dexamethasone
Insufficient evidence exists to support the routine use of Haemophilus infections
dexamethasone as an adjunct treatment in adults with Rifampicin (rifampin) 20 mg/kg/day for 4 days is recom-
bacterial meningitis. Selective use may be considered in mended for families and other close contacts with children
patients at high risk with severely impaired mental status, <4 years of age (i.e. at high risk) and for the patient.
cerebral edema or substantially raised intracranial pressure.
PROGNOSIS
Lowering intracranial pressure • Overall case fatality rate is 25%.
• Elevate the head of the bed to 30° to maximize venous • Case fatality rate varies according to the patient’s age and
drainage with minimal compromise of cerebral perfusion. the type of bacteria: 31% for S. pneumoniae meningitis in
• Hyperosmolar agents, such as mannitol and oral glycerol adults over 60 years of age compared with 3% for
(glycerin), increase the osmolality of the intravascular children younger than 5 years of age. Overall case fatality
space with respect to the brain and facilitate diffusion of is higher for S. pneumoniae (19%) than for either N.
water from the brain to the intravascular space. meningitidis (13%) or H. influenzae (3%).

331
331 Algorithm for the initial Bacterial meningitis suspected
N.B.There is an increasing trend to
management of patients with administer antibiotics immediately after
acute bacterial meningitis. suspecting bacterial meningitis (i.e. before
Consider empirical antibiotic therapy
performing a lumbar puncture, and even
before referral to hospital.)

Absent Focal neurologic signs or papilledema Present

Obtain blood cultures and do lumbar Obtain blood cultures


puncture immediately

Empirical antimicrobial therapy (Table 33)


CSF consistent with bacterial meningitis

Cranial CT scan
Gram stain or bacterial antigen test positive

Yes
No
No mass lesion Mass lesion present
Empirical antimicrobial therapy (Table 33) Specific antimicrobial therapy (Table 35)
Subacute and Chronic Meningitis and/or Encephalitis 279

SUBACUTE AND CHRONIC Bacterial meningitis (see p.273): inadequately treated


MENINGITIS AND/OR ENCEPHALITIS
Chronic bacterial infection following:
• Head injury.
DEFINITION • Intracranial surgery.
A syndrome characterized by various combinations of fever, • Use of intracranial shunts.
headache, lethargy, stiff neck, confusion, nausea, and
vomiting with accompanying CSF pleocytosis, of greater Parameningeal foci
than 4 weeks duration. • Otitis.
• Mastoiditis.
EPIDEMIOLOGY • Sinusitis.
• Incidence: uncommon. • Brain abscess.
• Age: any age. • Subdural empyema.

ETIOLOGY AND PATHOPHYSIOLOGY Non-infectious


Infection Connective tissue/granulomatous diseases
De novo infection • Systemic lupus erythematosus.
• Bacterial: • Behçet’s syndrome (oral and genital ulcers, iritis,
– Actinomycosis. meningitis).
– Brucellosis. • Sjögren’s syndrome.
– Listeria. • Sarcoidosis (see p.350).
– Nocardia. • Systemic vasculitis.
– Whipple’s disease (see p.289). • Isolated granulomatous angiitis of the nervous system
• Mycobacterial: TB. (see p.227).
• Spirochete: • Vogt–Koyanagi–Harada disease (uveitis, alopecia, vitiligo,
– Borrelia burgdorferi (see p.316). dysacousis, tinnitus).
– Leptospirosis.
– Syphilis (see p.312) (332). Malignant meningitis
• Viral: • Carcinoma: seeding of tumor cells in the CSF with, or
– HIV (see p.301). more often without, a solid intracranial or spinal primary
– Mumps. or secondary tumor. The common primary tumors are
– Cytomegalovirus. carcinoma of the breast, bronchus and nasopharynx;
– Lymphocytic choriomeningitis (LCM). leukemia, lymphoma and melanoma; primary intracranial
• Fungal: tumors rarely seed through the CSF.
– Cryptococcus. • Lymphoma (including intravascular lymphoma [neo-
– Coccidioides. plastic angioendotheliosis]).
– Histoplasma. • Leukemia.
– Candida. • Glioma.
– Aspergillus. • Melanoma.
– Blastomyces.
• Protozoal: toxoplasmosis. Chemical meningitis (see p.291)
• Helminthic (cestode worms): cysticercosis. Craniopharyngioma (see p.377), epidermoid cyst, drugs, dye.

Mollaret’s meningitis (see p.291)


Steroid-responsive chronic meningitis
Table 35 Antimicrobial therapy for bacterial meningitis Chronic benign lymphocytic meningitis
based on pathogen identification by positive Gram stain
and/or bacterial antigen test
Bacterial pathogen Therapy
Haemophilus Third generation cephalosporin 332
influenzae type B (cefotaxime or ceftriaxone): for 7 days
Neisseria meningitidis Penicillin or amoxycillin (amoxicillin) or
third generation cephalosporin: for 7 days
Streptococcus Vancomycin + third generation cephalosporin•:
pneumoniae for 10–14 days
Listeria Amoxycillin (amoxicillin) or penicillin G**:
monocytogenes for 14–21 days
Group B streptococci Amoxycillin (amoxicillin) or penicillin G**:
(S. agalactiae) for 14–21 days
Escherichia coli Third generation cephalosporin
(cefotaxime or ceftriaxone): for 21 days
*Consider addition of rifampicin (rifampin)
**Consider addition of aminoglycoside
332 Skin rash on the sole of the foot of a patient with syphilitic
meningitis.
280 Infections of the Nervous System: Bacterial

CLINICAL FEATURES clinically and radiologically involved leptomeninges and


Indolent onset and course. brain.

Meningitis DIFFERENTIAL DIAGNOSIS


• Headache. Chronic recurrent meningitis (vs. chronic persistent
• Fever. meningitis)
• Malaise. • Mollaret’s meningitis.
• Confusion. • Anatomic defects (e.g. skull fractures, post-operative).
• Weight loss. • Congenital defects (e.g. meningomyelocele, dermal sinus).
• Meningism. • Parameningeal focus (e.g. otitis, mastoiditis, sinusitis,
brain abscess, subdural empyema).
Associated syndromes • Immunosuppression.
• Seizures. • Tumors (e.g. craniopharyngioma, epidermoid, ependymoma).
• Hydrocephalus (obstructive or communicating). • SLE.
• Confusion, dementia.
• Papilledema. Chronic persistent meningitis (see Etiology, above)
• Syndrome of inappropriate antidiuretic hormone (SIADH). CSF cell count: <50 cells/mm3
• Migraine with CSF pleocytosis. • Behçet’s syndrome.
• Cranial neuropathies. • Benign lymphocytic meningitis.
• Radiculopathies. • Carcinoma.
• Spinal root pain, weakness, sensory loss. • Sarcoidosis.
• Myelopathy. • Vasculitis.

332)
Other manifestations of the underlying cause (3 CSF cell type
• Neutrophils:
INVESTIGATIONS – Bacterial meningitis.
Blood – Chemical (e.g. craniopharyngioma).
• Full blood count. – Fungal meningitis.
• ESR. – SLE.
• Serum biochemistry (e.g. urea and electrolyies, liver • Eosinophils:
function tests). – Chemical (e.g. intrathecal drugs).
• Antinuclear antibodies, anti-DNA antibody. – Coccidioides.
• Rheumatoid factor. – Lymphoma.
• Serum protein electrophoresis. – Parasites.
• Serum immunoglobulins. • Lymphocytes: all other causes.
• Serum complements.
• Serum angiotensin-converting enzyme. CSF glucose: low
• Serology should be repeated for borrelia, brucellosis, • Bacterial meningitis.
fungal antibodies, HIV, and leptospirosis. • Carcinoma.
• Fungi.
CSF • Syphilis.
Testing should be repeated on several occasions: • Sarcoid.
• Microscopy, cell count, Gram stain, culture, protein, • Subarachnoid hemorrhage.
glucose, VDRL, TPHA, oligoclonal bands, IgG/albumin • Viral (mumps, herpes, LCM).
ratio, cytology for inflammatory and malignant cells, • Mollaret’s meningitis.
cryptococcal antigen and other antigens and antibodies • Benign lymphocytic meningitis.
as appropriate from the list above (see Etiology, above). • Epidermoid cyst.
• Malignant cells are more likely to be seen if the CSF is
fresh and collected on several occasions. TREATMENT
• Lymphocytes usually predominate but occasionally • Specific treatment of underlying cause.
neutrophils do. • Empirical:
• CSF lymphocytes should be typed if there is any – Anti-TB (see p.282) and antifungal (see p.320) therapy:
suspicion of lymphoma. if no definite cause identified, and if the patient is unwell
and deteriorating.
Other – Corticosteroids: indicated if there is no favorable response
• Urine, sputum, and gastric washings for TB, fungi. clinically to empirical anti-TB and fungal therapy, and if
• Chest x-ray: if suspected TB, sarcoidosis or neoplasm. fungi have not been identified, because some patients have
• Ultrasound abdomen and pelvis: suspected hepatic a steroid-responsive chronic meningitis.
metastases or pelvic infection. – Relapse may occur when treatment is withdrawn.
• Cranial CT or MRI scan, or spinal MRI scan to exclude a • Malignant meningitis and the primary tumor are seldom
focal parameningeal collection of pus such as an treatable, and death occurs in weeks or months.
intracranial or spinal subdural empyema, or hydrocephalus.
• Biopsy with microscopy, cytology and culture of clinically PROGNOSIS
involved extraneural sites (e.g. lymph nodes, liver) or Depends on the underlying cause and response to treatment.
Tuberculous Meningo-encephalitis 281

TUBERCULOUS • The basal cisterns become blocked, resulting in a


MENINGO-ENCEPHALITIS meningeal obstructive type of hydrocephalus.
• The CSF in the aqueduct, or fourth ventricle more rarely,
becomes blocked by marked ependymitis, leading to
DEFINITION obstructive hydrocephalus.
Infection of the meninges and underlying brain by the acid-
fast organism Mycobacterium tuberculosis and exceptionally ETIOLOGY
by Mycobacterium bovis. Mycobacterium tuberculosis and exceptionally by
Mycobacterium bovis.
EPIDEMIOLOGY
• Incidence: increasing, concurrent with the HIV Risk factors
epidemic. Higher in developing countries, particularly • HIV infection: 500 times higher incidence of TB meningitis
the Indian subcontinent and sub-Saharan Africa, with than in the general population; HIV increases the lifetime
higher prevalence of HIV infection. risk of developing TB to one in three.
• Age: any age. • Alcohol abuse.
• Gender: either sex. • Diabetes mellitus.
• Malignancy.
PATHOLOGY • Recent corticosteroid use.
Macroscopic • Populations with a high prevalence of pulmonary
• Discrete, small, white tubercles scattered over the convexi- tuberculosis (i.e. poorer backgrounds, India, Africa).
ties and base of the brain. The ependyma and choroid
plexus are also studded with minute glistening tubercles. PATHOGENESIS
• Thick, gelatinous exudate over the basal meninges, Meningitis may occur as part of generalized TB with a single
obliterating the pontine and interpeduncular cisterns and focus (tuberculoma) in the brain or as a terminal event in
extending to the meninges in the floor of the third ventricle miliary TB. Two stages:
and subthalamic area, the optic chiasm, the under-surfaces • Bacterial seeding of the meninges and subpial regions of
of the temporal lobes, and around the medulla and spinal the brain, forming tubercles.
cord. The convexities are relatively spared. • Rupture of one or more of the foci (tuberculomas) with
discharge of bacteria into the subarachnoid space.
Microscopic
• Meningeal tubercles: a central zone of caseation CLINICAL FEATURES
surrounded by epithelioid cells and some giant cells, Subacute or chronic onset over weeks.
lymphocytes, plasma cells, and connective tissue.
• Exudate: composed of fibrin, lymphocytes, plasma cells, and Syndromes
other mononuclear cells, some polymorphonuclear leuko- • Meningo-encephalitis:
cytes and areas of caseation necrosis. The exudate is not – Headache, lethargy, confusion, drowsiness, fever, stiff
confined to the subarachnoid space (as is the case with the neck, Kernig and Brudzinski signs.
pyogenic meningitides) but frequently spreads along the pial – Cranial neuropathies: usually oculomotor palsies, less
vessels and invades the underlying brain, leading to a pure often facial palsies and deafness:
meningoencephalitis. The inflammatory exudate involves the • Focal neurologic deficit: due to brain infarction caused
cranial nerves as they traverse the subarachnoid space (333). by infectious arteritis.
• Arteries become inflamed and occluded, leading to focal • Raised intracranial pressure: due to hydrocephalus.
brain infarction.
Associated conditions
Systemic tuberculosis (two-thirds of patients): pulmonary,
333 bone or renal.

OTHER FORMS
Tuberculous serous meningitis
• Due to a meningeal reaction to an adjacent tuberculous
focus.
• Asymptomatic, or headache, lethargy and confusion with
mild meningeal signs.
• CSF: modest pleocytosis in some, normal or elevated
protein, normal glucose.
• Self-limiting usually, but may progress to fatal generalized
TB meningitis.

Tuberculoma
• Tumor-like masses of tuberculous granulation tissue in
333 Microscopic examination of the mononuclear inflammatory cell the brain parenchyma.
exudate involving the trigeminal nerve root as it traverses the • Asymptomatic or symptoms of a space-occupying lesion.
subarachnoid space of a patient with tuberculous meningitis. An artery • CSF: mild pleocytosis and raised protein; normal glucose.
in the lower part of the field has become inflamed and occluded.
282 Infections of the Nervous System: Bacterial

Myeloradiculitis acquiring the infection, or where the infection is over-


• Spinal cord or nerve root compression by an epidural whelming or the patient is immunosuppressed.
mass of granulation tissue or angulation of the vertebral
column (see Spinal epidural abscess, p.555). Other
• May accompany vertebral body disease (Pott’s paraplegia). • Sputum smears and cultures for acid-fast bacilli.
• Chest x-ray.
DIFFERENTIAL DIAGNOSIS • Gastric washings.
• Meningitis: fungal (e.g. cryptococcal, see p.318; see • Urine examination.
Chronic meningitis, p.279); pyogenic.
• Meningeal carcinomatosis (malignant meningitis). DIAGNOSIS
• Brain tumor (e.g. brainstem glioma). Clinical and microbiologic: demonstration of acid-fast
bacilli, either on microscopy of smears of CSF sediment
INVESTIGATIONS stained by the Ziehl–Neelsen method or by culture, in a
Blood patient with clinical evidence of meningo-encephalitis.
Hyponatremia is common due to the syndrome of inap-
propriate ADH secretion (SIADH) or TB of the adrenals. TREATMENT
Initial anti-TB chemotherapy treatment
Brain imaging 1 Rifampicin (rifampin) 600 mg daily as a single oral dose
Similar features will be seen on CT or MRI, though MRI is (15–20 mg/kg/day in children); plus
more sensitive: 2 Pyrazinamide 30–50 mg/kg as a single oral dose
• Tuberculomas may be single or multiple, usually small (10 mg/kg/day in children); plus
(2–3 mm diameter) and can coalesce to form a larger lesion 3 Isoniazid 400 mg/day (5 mg/kg in adults; 10 mg/kg in
(334, 335). They may be low or high density on CT, may children) as a single oral dose; plus
adhere to the dura, cause hyperostosis (mimicking a 4 Pyridoxine 20–50 mg daily, oral should always be given
meningioma), and may calcify (<20%). They may produce with isoniazid to prevent peripheral neuropathy.
symptoms from intracranial mass effect and edema.
• Tuberculous meningitis causes basal pachymeningitis and Alternative initial treatments
hydrocephalus, usually of the meningeal obstructive • Streptomycin 1 g i.m. daily (20–40 mg/kg/day in
variety (336, 337). The basal cisterns are difficult to see children) can be used instead of rifampicin (rifampin) if
as they are filled with exudate. They enhance intensely rifampicin causes intolerable adverse effects.
(particularly on MRI) with contrast. On MRI the basal • Ethambutol may be substituted for pyrazinamide. The
cisterns may appear brighter than normal on T1W image dose is 25 mg/kg/day as a single oral dose for the first
because of the protein content of the exudate. The month, then 15 mg/kg/day. Ethambutol should be
cisterns may calcify (best seen on CT). avoided in children, and patients with renal failure. It is
• Tuberculoma and tuberculous meningoencephalo- given in divided doses, after meals, because of its
vasculitis can simulate an infiltrating brainstem glioma tendency to produce gastric irritation. Visual acuity and
radiologically. red-green color discrimination are regularly checked
• With treatment the tuberculomas may shrink and disappear because of the risk of optic neuropathy.
(or calcify) but the hydrocephalus does not change.
Additional treatments
CSF (lumbar puncture) Dexamethasone (adults 12 mg/day; children <25 kg,
• Pressure: increased. 8 mg/day) given for 3 weeks (in conjunction with anti-TB
• Cells: 50–500 white cells/mm3; initially polymorpho- therapy) then tapered over a further 3 weeks may reduce the
nuclear leukocytes and lymphocytes in equal numbers incidence of sequelae in patients who are culture-positive,
but after several days, lymphocytes predominate. particularly those who have a decreased conscious level at
• Organisms: Ziehl–Neelsen stain of smears of CSF sedi- presentation. Prednisolone, 60–80 mg/day tapering after
ment reveals acid-fast tubercle bacilli in about 10–20% of 2 weeks to finish at 4–6 weeks, is an alternative.
patients; culture requires 3–4 weeks to become manifest
(so treatment must be instituted on suspicion). Treatment after 2 months
• Protein: 1.0–2.0 g/l (100–200 mg/dl); higher if CSF If the mycobacterium is shown to be sensitive to all three
flow is blocked around the spinal cord. antibiotics (rifampicin [rifampin], pyrazinamide and
• Glucose: <2.22 mmol/l (<40 mg/dl), but the glucose isoniazid) in vitro, pyrazinamide can be discontinued, but
falls slowly over several days and rarely falls to the rifampicin and isoniazid (and pyridoxine) should be
extremely low levels seen in pyogenic meningitis. continued together for about another 7 months; the dose
of isoniazid can be reduced to 300 mg once daily. The
Mantoux test duration of treatment depends on many factors, including
10 tuberculin units are given intradermally on the flexor the degree of supervision and compliance.
surface of the forearm. An area of induration measuring N.B. Intracranial tuberculomas require a similar course
5 mm or more read at 48–72 hours following the injection of chemotherapy as outlined above. The tuberculomas may
is a positive Mantoux. This indicates previous or recent transiently increase in size despite clinical improvement soon
exposure to M. tuberculosis and does not prove active TB; after the start of antimicrobial treatment, or disappear and
i.e. it may also indicate previous TB, healed infection, or calcify. If they persist, particularly exerting ‘mass effect’,
BCG vaccination. The test may also be negative in active surgical excision may be required.
TB, if the test is done within 6 weeks of the patient
Tuberculous Meningo-encephalitis 283

Adverse effects – Serum levels of streptomycin should be monitored and


• Rifampicin (rifampin): if renal impairment occurs, the dose should be reduced.
– Fever, skin rash, nausea, vomiting, diarrhea, thrombo- • Ethambutol: blurred vision, optic atrophy, peripheral
cytopenia, hepatitis. neuropathy, hepatitis.
– Saliva and urine turn orange-red and soft contact lenses
may discolor. CLINICAL COURSE AND PROGNOSIS:
– An early transient rise in liver enzymes is common. • Untreated, the course is progressive and invariably fatal
• Pyrazinamide: fever, urticaria, flushing, nausea, vomiting, within weeks to months.
arthralgia, hepatitis, hyperuricemia. • Overall mortality: about 10%; higher in infants, the
• Isoniazid: elderly and HIV-infected patients.
– Skin rash, nausea, vomiting, arthralgia, peripheral • Survival is enhanced by early diagnosis.
neuropathy, confusional and psychotic states, seizures, • Neurologic impairments persist in 20–30% of survivors,
hepatitis, pellagra. most commonly cognitive dysfunction, epileptic seizures,
– If symptoms of hepatitis or abnormal liver function tests visual and oculomotor disorders, deafness and hemiparesis.
occur, stop the isoniazid. • Focal weakness, Glasgow coma scale, and somatosensory
• Streptomycin: evoked potentials are the best predictors of outcome at
– Skin rash, deafness, tinnitus, vertigo, ataxia, nephro- 6 months (in one recent study).
toxicity, and exacerbation of myasthenia gravis.

334, 335 CT brain scans with 334 335


contrast showing calcified
nodules (arrows) in the
parenchyma (tuberculomas) and
a degree of hydrocephalus.The
white linear structure in the
anterior horn of the right lateral
ventricle is a shunt (335).

336, 337 CT brain scans 336 337


showing hydrocephalus due to
tuberculous meningitis in a
person from the Indian
subcontinent. Note the dilated
ventricles right down to the
fourth where there is good
communication with the basal
cisterns, i.e. this is a
communicating hydrocephalus.
284 Infections of the Nervous System: Bacterial

INTRACRANIAL ABSCESS • Streptococcus pneumoniae (pneumococcus): carried in the


(BRAIN ABSCESS AND nasopharynx from where it can spread by droplets to
SUBDURAL EMPYEMA) cause sinusitis, otitis media and pneumonia. Severe
infection tends to occur in patients who have had a
splenectomy or have hypogammaglobulinemia, comple-
DEFINITION ment deficiencies or sickle cell disease.
• Brain abscess: a localized collection of pus within the • Enterobacter/coliforms: increasing incidence (10–30% of
brain, and usually supratentorial. cases).
• Subdural empyema: a thin layer of pus between the dura • Staphylococcus aureus (head injury, neurosurgical
and arachnoid membranes over the surface of one side procedure, ventricular shunt, CSF fistula): decreasing as
of the brain, frequently spreading into the inter- a cause (10–30% of cases).
hemispheric fissure (parafalcine) and over to the surface • Fusobacterium spp.: (<10% of cases).
of the other side of the brain. • Haemophilus spp.: (<1%).
• Fungi (Aspergillus and Candida spp.): (especially if
EPIDEMIOLOGY immunocompromised).
• Incidence: uncommon. • Mycobacterium tuberculosis.
• Point prevalence 2 per 100 000. • Protozoa: Toxoplasma gondii, helminths (e.g. Strongyloides
• Age: any age. stercoralis) (especially if immunocompromised).
• Gender: M=F. • Ameba.

PATHOLOGY Anaerobic organisms


Bacterial infection of a part of the brain causes inflammation • Gram-negative rods: Bacteroides, Prevotella, Porphyomonas
and edema around the periphery of the infected brain spp.: increasing incidence: comprise 20–50% of cases.
parenchyma (so-called ‘cerebritis’), which is followed by • Peptostreptococcus spp., particularly P. micros: (10–40% of
necrosis in the center of the lesion, with the formation of cases).
pus in the abscess cavity.
Several bacterial strains are frequently present in a single
PATHOPHYSIOLOGY abscess but sometimes nothing is seen on Gram stain or
Local spread of infection/complications grown in aerobic or anaerobic culture.
• Trauma: penetrating head wounds with CSF leak.
• Suppurative middle ear, mastoid, paranasal sinus or CLINICAL FEATURES
dental infections (the most common local events leading Brain abscess
to brain abscess formation). The usual presenting features are the subacute onset and
• Bacterial meningitis. progressive evolution of:
• Orbital cellulitis. • Fever.
• Headache.
Local spread of infection usually causes a subdural • Lethargy and malaise.
empyema or a single/solitary intracerebral abscess. • Seizures.
• Focal neurologic signs.
Hematogenous spread • Symptoms and signs of raised intracranial pressure, such
• Septicemia. as papilledema, may follow as the abscess acts as an
• Right to left shunt: expanding space-occupying lesion.
– Intracardiac: cyanotic congenital heart disease. • Meningism and systemic evidence of bacterial infection
– Intrapulmonary: pulmonary arteriovenous malformation. may be present.
• Infective endocarditis. • Features of the underlying source (e.g. dental or ear
• Suppurative chest infection (abscess, bronchiectasis, infection) may be present.
empyema).
Subdural empyema
Hematogenous spread of infection usually causes Supratentorial
multiple intracerebral abscesses. • Severe headache, often unilateral.
• Dysfunction of, and focal seizures referable to, the
Other predisposing factors underlying hemisphere.
Immune system compromise (e.g. HIV infection). • Cranial nerve III and VI palsies due to compression.

ETIOLOGY Infratentorial (rare)


Brain abscesses are often polymicrobial, particularly if they • Severe headache.
arise from a dental source. • Meningism.
• Cerebellar and brainstem signs.
Aerobic organisms • Features of raised intracranial pressure.
• Streptococcus milleri (causes suppurative dental and sinus
infection in the middle-aged and elderly): up to 80% of
cases of brain abscess are caused by a streptococcus.
Intracranial Abscess (Brain Abscess and Subdural Empyema) 285

DIFFERENTIAL DIAGNOSIS mastoid. It has some mass effect and surrounding white
• Bacterial meningitis. matter edema, and shows enhancement in a thin walled
• Tuberculous meningitis. ring after i.v. contrast (338, 339).
• Encephalitis. • MRI, T1W, reveals a central region of marked low
• Intracranial tumor: both abscess and tumor frequently intensity surrounded by a discrete ring that is isointense
appear as ring-enhancing lesions on neuroimaging to mildly hyperintense. This area is surrounded by a
studies. A raised C-reactive protein and increased uptake region of mild hypointensity. These regions correlate
of tracer on 99mTc-HMPAO leukocyte scintigraphy with the necrotic center, the abscess capsule, and
raises the likelihood of abscess; steroid therapy should be surrounding edema respectively. N.B. Some tumors can
discontinued for 48 hours prior to leukocyte scintigraphy. look very much like abscesses and vice versa, and
• Cerebral infarction. abscesses do not always cause pyrexia. The only sure way
of differentiating an abscess from a tumor is by biopsy
INVESTIGATIONS which may be necessary prior to starting steroids.
Full blood count and other blood tests (Steroids may be acceptable for a brain tumor but not so
In general, laboratory tests are of little value in the diagnosis good for abscesses!)
of brain abscess. Only 30% of patients have peripheral
leukocyte counts greater than 11 000 cells/ml. However, Subdural empyema
blood tests can be helpful in identifying the underlying On CT or MRI:
cause or predisposing condition (e.g. HIV serology, • A collection in the subdural space which may be very
toxoplasma serology, serum immunoglobulins, serum difficult to see on the unenhanced scan as it is often
complement). isodense with brain or so thin that it is not visible.
• Often adjacent to an infected nasal sinus or mastoid.
Cranial CT or MRI scan without and with contrast • Swelling (edema) of the adjacent underlying cerebral
Chronic sinusitis, mastoiditis, osteomyelitis, fracture. hemisphere is usually present, sometimes with effacement
of cortical sulci, compression of horns of lateral
Abscess ventricles, and midline shift. The appearance may be
• CT or MR can be used, though MR is probably more extremely subtle, for example only slight widening of the
sensitive particularly in the very early ‘cerebritis’ stage. interhemispheric fissure as the only clue (if in doubt give
• CT shows a low density area (sometimes multiple) contrast and preferably do an MRI).
usually at the gray/white matter junction, and which (Continued overleaf)
may lie adjacent to an infected (opacified) nasal sinus or

338 339

338 CT brain scan with contrast showing a rounded area in the left
temporal region with ring enhancement (arrows) and some
surrounding edema typical of an abscess. Note that the only certain 339 CT brain scan of a more established abscess containing an
way of establishing the diagnosis is by biopsy, as many tumors can have air–fluid interface (arrows) due to the presence of gas-forming
a similar appearance. organisms. Note the considerable mass effect with midline shift.
286 Infections of the Nervous System: Bacterial

On CT or MRI (continued): Culture (aerobic and anaerobic) of pus


• After intravenous contrast injection, a thin rim of intense Pus is cultured from the abscess, sputum, sinuses and blood.
contrast enhancement over the cerebral hemisphere may Prolonged incubation of specimens under a wide range of
become evident (340), and enhancement in the brain if cultural conditions is necessary to ensure isolation and
the infection has spread to cause cerebritis. identification of all organisms.
• Magnetic resonance imaging without and with
gadolinium enhancement is superior to CT due to the Lumbar puncture
absence of bone artefact; increased contrast between LP is contraindicated when a brain abscess is suspected. If
bone, CSF and brain parenchyma; and the ability to done inadvertently, the CSF may be normal or show no
produce images in multiple planes. Furthermore, MRI organisms, a mild CSF pleocytosis, mild elevation of CSF
can better characterize subdural collections, allowing protein, and a normal glucose. Cultures of CSF are usually
differentiation of sterile, bloody and infected collections. negative (i.e. similar to acute viral encephalitis, and viral or
• Complications include meningitis (see above), cerebral tuberculous meningitis).
infarction, cerebral abscess, venous thrombosis.
DIAGNOSIS
Radionuclide scan The diagnosis of brain abscess is suggested but not confirmed
Increased uptake by the collection of pus on 99mTc- by the presence of a suggestive clinical picture, and imaging
HMPAO leukocyte scintigraphy. evidence of an asymmetric capsule, multiple lesions, the
location of the lesion at the corticomedullary junction, and
Carotid angiography associated leptomeningeal enhancement. Ultimately, aspiration
Rarely required, but may be useful if a subdural empyema and biopsy are often necessary to confirm the diagnosis of
is suspected but not imaged on CT or MRI, to show the brain abscess. Isolation and identification of all organisms is
avascular gap between the skull and underlying cerebral vital to allow rational decision-making about optimal therapy.
hemisphere, or in the interhemispheric fissure.
TREATMENT
Exploratory burr holes Small abscesses (<2.5 cm [<1 in])
These may be necessary when a subdural empyema is Antibiotics
strongly suspected but not imaged by serial application of Antibiotics that enter the brain at high concentrations include
the above techniques. chloramphenicol, selected third-generation cephalosporins,
metronidazole, methicillin, nafcillin, penicillin, trimethoprim
Chest x-ray plus sulfamethoxazole, and vancomycin.
• Heart abnormality (e.g. cyanotic congenital heart disease). Despite bacteriocidal concentrations of these antibiotics,
• Pulmonary arteriovenous malformation. bacteria may still be cultured from abscess aspirates,
• Lung infection (e.g. abscess, bronchiectasis, empyema). probably because of the acidic environment favoring
bacterial growth and inhibiting antibiotic action.
Blood cultures
Initial empirical antibiotic therapy
• Penicillin G 200 mg/kg/day intravenously (i.v.) in 4
340 hourly boluses (about 33 mg/kg every 4 hours); plus
• Chloramphenicol 3 g daily i.v. in 8 hourly doses (i.e. 1 g
every 8 hours) (75 mg/kg/day in children); plus
• Metronidazole 2 g daily i.v. in 6 hourly boluses (i.e.
500 mg infused over 1 hour every 6 hours) (7.5 mg/kg
6 hourly in children).

If staphylococcal infection is suspected (i.e. post-


neurosurgery, ventricular shunts, and so on), replace
penicillin G in the above regime with:
• Flucloxacillin 6 g daily i.v. in 4 hourly doses (i.e. 1 g
every 4 hours) (children 50 mg/kg/day); or
• Fusidic acid, 1.5 g daily i.v. (i.e. 500 mg infused over 6
hours, every 8 hours), particularly if osteomyelitis is present.

Alternatives
• Cefotaxime + metronidazole (+ penicillin G).
• Ampicillin + gentamicin + metronidazole.

Specific antibiotic therapy


Specific antibiotic therapy (choice of antibiotics and dose)
will depend on bacterial culture, sensitivities and serum
levels. Intravenous antibiotic therapy should be continued
340 CT brain scan with contrast shows a frontal subdural empyema until the patient is at least neurologically stable, and
(arrowheads) secondary to frontal sinusitis. Note the enhancing preferably fully recovered. It should then be possible to
membrane on its inner edge. change to oral therapy for several weeks.
Tetanus 287

Large abscesses (>2.5 cm [>1 in]) TETANUS


Rarely cured by antibiotics alone; indeed long-term
combined use of several antibiotics may allow for the
uncontrolled growth of resistant organisms or may lead to DEFINITION
greater toxicity to the patient. Furthermore, because the A serious preventable disease caused by infection of the skin
clinical and radiologic diagnosis of brain abscess is not and deep and necrotic wounds by Clostridium tetani, a
always certain, be aware that antibiotic therapy may be given Gram-positive bacillus, which secretes a neurotoxin.
for a brain tumor that is mistaken as an abscess.
EPIDEMIOLOGY
Neurosurgery • Incidence: 1 million cases annually world-wide. Rare in
With the advent of image-guided stereotactic techniques, developed countries because of immunization; only 36
aspiration of a brain abscess can be performed quickly and cases were reported (of an estimated 170 cases) in the
safely under local anesthesia. United States in 1994.
Aspiration provides diagnostic material, immediate • Age: more than half of cases occur in people 60 years of
decompression, and removes acidic and hypoxic necrotic age or older.
material, providing a more favorable environment for
antibiotic therapy. Aspiration is indicated in the stages of ETIOLOGY AND PATHOPHYSIOLOGY
cerebritis, multiple lesions, deep seated lesions, and lesions The source of infection is usually a wound (about 65%), often
in eloquent areas of the brain. a minor one such as a thorn, splinter of wood, or a piece of
Open surgical excision is limited to large superficial lesions metal. Chronic skin ulcers account for about 5% of cases, and
at risk of causing herniation and which are well encapsulated there is no obvious source in the remainder.
and in non-eloquent lesions, fungal abscesses, multiloculated Clostridium tetani are Gram-positive, spore-forming bacilli
abscesses, and post-traumatic abscesses which often contain that exist primarily in soil. Their prevalence in soil samples
contaminated foreign bodies. ranges from 2–23%.
Pre-operative antibiotics are not given because they Under anaerobic conditions, the spores germinate and
increase the risk of sterile cultures. Antibiotic therapy as produce two toxins: tetanolysis (a hemolysin with recognized
directed by culture and sensitivity is given for 6 weeks. pathologic activity) and tetanospasmin, which is responsible
Neuroimaging studies are obtained at weekly intervals during for tetanus. Tetanospasmin is synthesized as a single 151-kd
therapy and monthly thereafter until the abscess has resolved. chain and cleaved to two chains joined by a single disulfide
bond. The heavy chain (100 kd) is responsible for specific
Subdural empyema binding to neuronal cells and transport proteins. The light
At some stage it is usually necessary to drain surgically the chain (50 kd) is a zinc endopeptidase that cleaves an integral
purulent loculation, but the decision of when and whether membrane protein of small synaptic vesicles, synaptobrevin,
to undertake burr hole drainage or a formal craniotomy at a single site and blocks the release of neurotransmitters.
should be made in conjunction with the neurosurgeon. The Once the toxin is synthesized it moves from the
method of drainage probably matters less than its timing contaminated site to the spinal cord or brainstem. The toxin
and adequacy. reaches the CNS by intra-axonal transport, moving at a rate
The local instillation of antibiotics (e.g. amphotericin B) of 75–250 mm (3–10 in) per day. So, the process takes 2–14
as an adjunct to the treatment of otherwise intractable brain days. With facial injuries the interval is short. Once the toxin
abscess and subdural empyema appears to be promising. reaches the CNS, local or cephalic tetanus may occur initially,
followed by generalized tetanus. The toxin produces
Orbital, ear, mastoid and sinus infection presynaptic blockade of the synapses of inhibitory Renshaw
These must be treated accordingly. cells and 1a fibers of alpha motor neurons that handle the
transmission of γ-aminobutyric acid and glycine, but not of
Antiepileptic therapy the synapses of Renshaw cells that handle acetylcholine
Institute if recurrent seizures. transmission. Instability of the autonomic nervous system also
occurs. The toxin binding appears to be irreversible; recovery
PROGNOSIS depends on the sprouting of new axonal terminals.
Brain abscess
• Case fatality rate: CLINICAL FEATURES
– Pre-CT era: up to 30%, together with a high morbidity As described by Gowers (1888):
rate among survivors.
– Post-CT era: <6%, due to: improvements in ‘Tetanus is ... characterized by persistent tonic spasm with
microbiologic isolation techniques, more effective violent brief exacerbations. The spasm almost always
antibiotics, availability of CT and MRI that have resulted commences in the muscles of the neck and jaw, causing
in earlier diagnosis and more effective treatment at stages closure of the jaws (trismus, lockjaw) and involves muscles
when patients are relatively well neurologically. of the trunk more than those of the limbs.’
• Predictors of a poor outcome in terms of morbidity and
mortality: Initially, within days or weeks of the injury, the patient
– Severely impaired mental status and neurologic complains of stiffness, pain and rigidity in the voluntary
impairment on admission. muscles of the jaw, face and abdomen. The disease may be
– Brain abscess in infants, particularly those that are large mild and progress no further or the muscle stiffness, pain and
(>5 cm [>2 in]) or multiple. rigidity may spread to involve the neck, pharynx, back and
• Epilepsy is a complication in more than 50% of survivors. sometimes limbs. Involuntary spasms of the affected muscles
288 Infections of the Nervous System: Bacterial

may occur and be painful, frightening and cause difficulty – Paralysis and ventilation are required if spasms become
opening the jaw (trismus), a grimacing facial appearance ‘risus severe.
sardonicus’ due to contraction of the facial muscles (341), • Maintain nutrition and fluid balance with oral feeds if
and difficulty swallowing, sometimes prompting the patient mild, or i.v. line or nasogastric tube if spasms are
to complain of a ‘sore throat’. The patient is usually afebrile moderate but not severe. If severe, the patient must be
unless there is concurrent local infection. intubated and ventilated.
With severe tetanus, all muscles contract, with the stronger • Human antitetanus immunoglobulin (500 units) is
overpowering the weaker. There is opisthotonos, flexion of the recommended, although its efficacy is controversial.
arms, extension of the legs, periods of apnea due to spasm of • Antitetanus toxin.
the intercostal muscles and diaphragm, and rigidity of the • Antimicrobial chemotherapy: metronidazole (0.5 g every
abdominal wall. Spasms are precipitated by startle, cough, 6 hours or 1.0 g every 12 hours intravenously) is as good
touch, and a full bladder. Laryngeal spasm can be precipitated as, or better than, penicillin G which is no longer the
by swallow or the passage of a nasogastric tube. Late in the drug of choice. Also, penicillin is an antagonist of
disease autonomic dysfunction develops with tachycardia and γ–aminobutyric acid, just as is tetanus toxin.
hypertension alternating with bradycardia and hypotension, • Prevent gastric stress ulcers: H2 receptor antagonists.
cardiac arrhythmias, sweating, fever, salivation and gastric stasis. • Prevent deep vein thrombosis: low dose heparin
Ophthalmoplegia and facial weakness are rare. 5000 units subcutaneously twice daily.
• Control autonomic dysfunction: beta-blockade or
DIFFERENTIAL DIAGNOSIS combined alpha and beta-blockade as necessary for labile
• Dystonic reactions to neuroleptic drugs (which typically in- blood pressure and heart rate.
volve lateral turning of the head, often with protrusion of the
tongue [symptoms that are rarely, if ever, seen in tetanus]). PROGNOSIS
• Strychnine poisoning. • The disease progresses over about a week, stabilizes for
• Local infection in the pterygomandibular space (e.g. dental another week, and then recovers over several weeks.
or in the masseter muscle) with trismus. • The severity is very variable.
• Dislocation of the mandible leading to ‘lockjaw’. • Case fatality ranges from 10 to >50% worldwide. At least
• Facial or jaw trauma. half of deaths from tetanus worldwide occur in neonates.
• Rabies. These deaths are preventable through antepartum
• Hysteria. maternal immunization.
• In the USA, 75% of deaths occur in people who are 60 years
INVESTIGATIONS of age or older (who make up 59% of all cases of tetanus).
• Laboratory tests are of virtually no value except to
exclude strychnine poisoning. PREVENTION
• Blood counts and blood biochemistry findings are Tetanus is a serious preventable disease that remains a threat
unremarkable. even in developed countries, particularly in older people. A
• Imaging studies of the head and spine reveal no case of tetanus reflects the failure of our health care delivery
abnormalities. system to provide immunization. Routine boosters every
• A LP is not necessary; the CSF is normal except for a 10 years should be emphasized in older people.
raised opening pressure, particularly during spasms. About 70% of a random sample of Americans aged 6 or
more years had protective levels of tetanus antibodies. The
DIAGNOSIS prevalence of protective antibodies is less than 50% in people
Clinical. aged 60–69 years, and about 30% in people aged 70 years
and older.
TREATMENT
• Nurse in quiet surroundings.
• Excise and debride any wound. 341
• Control muscle spasms: benzodiazepines are the main-
stay of treatment; intravenous diazepam 10–40 mg every
1–8 hours may be required to prevent spasms that last
more than 5–10 seconds. At high doses, lactic acidosis
can occur, possibly as a result of the solvent vehicle,
propylene glycol. There are γ-aminobutyric acid agonists
that indirectly antagonize the toxin, but they do not
restore glycinergic function.
• Maintain ventilation and oxygenation and prevent
pulmonary aspiration of gastric contents:
– Control the muscle spasms.
– Tracheostomy is indicated if rigidity and spasms cannot
be controlled and interfere with swallowing or breathing.

341 Facial photograph of a patient with facial muscle stiffness and


grimacing (risus sardonicus) and difficulty opening the jaw (trismus) due
to tetanus.
Whipple’s Disease 289

WHIPPLE’S DISEASE ETIOLOGY AND PATHOPHYSIOLOGY


Infection by T. whippelii, a Gram-positive bacillus.

DEFINITION CLINICAL FEATURES


A rare, chronic, relapsing, multisystem granulomatous The most common clinical presentation is the insidious
disorder that is caused by infection by Tropheryma whippelii, onset of a malabsorption syndrome (e.g. diarrhea),
a Gram-positive bacillus; it primarily involves the intestine sometimes preceded by migratory polyarthralgia and fever.
causing chronic diarrhea, together with fever and migratory Cardiac (pericarditis, myocarditis, marantic endocarditis)
polyarthralgias. About 5% of patients present with and CNS involvement is also common, and a few present
neurologic manifestations and 6–43% eventually develop with primarily neurologic manifestations.
symptomatic CNS involvement.
Neurologic triad
HISTORY • Dementia, apathy and personality change: progressive.
Described in 1907 by George Whipple. The patient was a • Myoclonus (facial) and tonic-clonic seizures; oculomastica-
missionary who had weight loss, diarrhea, and abdominal pain tory myorhythmia (facial myoclonus) is pathognomonic:
associated with polyarthralgia and lymphadenopathy. At the eyes converge synchronously (convergent nystagmus)
autopsy, numerous argyrophilic rod-shaped organisms were with involuntary jaw, palatal and tongue movements.
present in mesenteric lymph nodes. In 1949 Black-Schaffer Rhythmic spinal myoclonus has also been described.
showed that the bacilli gave a strongly positive result when • Supranuclear ophthalmoplegia (voluntary vertical and, less
stained with periodic acid Schiff reagent. In 1991 and 1992 so, horizontal gaze palsy; not caused by cranial nerve
specific DNA sequences were amplified from affected tissue. involvement but by lesions in the brainstem).
The nucleotide sequence had phylogenetic similarities to the
actinomyces group, and the organism was named T. whippelii. Other features
• Ataxic gait.
EPIDEMIOLOGY • Hypothalamic dysfunction (sleep [hypersomnolence],
• Incidence: rare; fewer than 800 cases have been reported drinking [polydipsia], and eating [hyperphagia] disorders).
(<10 cases per year). • Coma.
• Age and gender: usually presents in middle-aged men. • Chronic meningitis.
• Blurred vision or visual loss due to vitritis, uveitis, retinitis,
PATHOLOGY retinal hemorrhage, choroiditis, papilledema, optic atrophy
• Symptomatic neurologic involvement occurs in at least or keratitis.
10% (6–43%) of patients at some stage in their illness. • Progressive peripheral neuropathy and myopathy are rare.
Autopsy more frequently reveals brain involvement, even
in the absence of neurologic symptoms. DIFFERENTIAL DIAGNOSIS
• Sites of predilection are the basal ganglia, insular cortex, Depends on the clinical features:
midbrain, pons and cerebellum. • Progressive supranuclear palsy.
• Microscopic examination of the brain reveals widespread • Pituitary tumor.
inflammation (encephalitis) throughout the cerebral • Alzheimer’s disease.
hemispheres and brainstem (particularly the striatum, • Vascular dementia.
other basal nuclei and pons) characterized by • Cerebral tumor: primary or metastatic.
mononuclear cell perivascular cuffing and infiltration of • Sarcoidosis.
white and gray matter, marked astrocytosis, proliferation • Syphilis.
of microglia, and focal necrosis of many nerve cells.
• With hematoxylin and eosin (H&E) stain some nerve INVESTIGATIONS
cells appear swollen and the cytoplasm has a very fine, Full blood count, ESR and other blood tests
granular, bluish-purple staining appearance. Minimal neutrophil pleocytosis; ESR and serum ACE may
• Fat stains demonstrate nerve cells containing numerous, be raised.
rather coarse, varying-sized pale granules and globules of fat.
• PAS stain shows that the cytoplasm of neurons, astrocytes Molecular DNA analysis
and perivascular inflammatory cells contain abundant Amplification of sequences of bacterial 16S ribosomal RNA
foamy macrophages filled with masses of coalescent PAS- (rRNA) specific for Whipple bacillus (T. whippelii) from
staining granules. peripheral blood mononuclear cells using PCR.
• Electron microscopy shows numerous bacilli in the
distended macrophages and in the intercellular spaces. Cranial CT scan
The bacilli measure 1.5–3.0 μm in length and 200 nm in May be normal, show cortical atrophy, areas of low attenuation
diameter. The bacilli are lined by a thin surface membrane with or without contrast enhancement, areas of mixed density
and a thick (20 nm) cell wall. Whipple’s bacteria are most with multifocal enhancement and regions of vasogenic edema.
frequently found in microglial and ependymal cells in the
brain but they are also found within astrocytes, pericytes, MRI brain
and choroid plexus cells. The internal layers of the retina May be normal in the early stages or show diffuse abnor-
also show many large, pale, granule-containing cells. malities on T2W images. Areas of increased T2W signal inten-
sity on MRI scans are a sensitive indicator of disease activity
and the lesions may enhance intensely with contrast medium.
290 Infections of the Nervous System: Bacterial

The lesions may be seen in the hypothalamus, midbrain and • Chronic migratory arthralgias or polyarthralgias.
basal ganglia and may have slight mass effect (342, 343). • Unexplained lymphadenopathy, night sweats, or malaise.
• Also must have any one of four neurologic signs, not due
CSF to another etiology:
CSF is normal or more commonly contains excess white cells – Supranuclear vertical gaze palsy.
and protein, oligoclonal banding and increased IgG. It may – Rhythmic myoclonus.
reveal sickle-particle-containing cells. Staining of the CSF with – Dementia with psychiatric symptoms.
periodic acid Schiff reagent may give a positive result. PCR can – Hypothalamic manifestations.
be used to amplify sequences of bacterial 16S rRNA specific • A favorable response to trimethoprim-sulfamethoxazole
for Whipple bacillus (T. whippelii) in the CSF. or chloramphenicol therapy helps to confirm the diagnosis.

Other TREATMENT
• EEG: non-specific. Antimicrobial treatment
• Biopsy of jejunal mucosa, lymph nodes, vitreous or brain. • Oral trimethoprim (160 mg) and sulfamethoxazole
(800 mg) twice daily for 1 year plus folate (folic acid)
DIAGNOSIS supplementation.
Definite CNS Whipple’s disease • If allergic: intravenous penicillin G (2 million units,
Must have any one of the following three criteria: 4 hourly) and oral doxycycline (100 mg bd).
• Oculomasticatory myorhythmia (OMM: pendular
vergence oscillations of the eyes that are synchronous The choice of antimicrobial treatment is based mainly on
with masticatory myorhythmia) and oculo-facial-skeletal taxonomy, which has been known for only the past few years.
myorhythmia (OFSM: similar to OMM, also involves The organism has not been cultured, and therefore in vitro
myorhythmia of non-facial skeletal muscle). susceptibilities are not available. There are no data on compari-
• Positive tissue biopsy: ultrastructural findings of son of antibiotics. However, empirical clinical experience sug-
distinctive periodic acid Schiff-positive bacillary rods and gests that patients initially treated with drugs that do not pene-
sickle-shaped inclusion bodies in macrophages in the trate the blood–brain barrier are at risk of neurologic relapse.
CSF, vitreous or in a jejunal or brain biopsy.
• Positive PCR amplification of sequences of bacterial 16S Myoclonus (see p.124)
rRNA gene corresponding to the Whipple’s disease • Valproate 500 mg bd.
bacillus (T. whippelii) in infected tissues. • Clonazepam 0.5 mg nocte, increasing up to 8 mg per
• If histologic or PCR analysis is not performed on CNS day (or tolerance).
tissue, then the patient must also demonstrate neurologic
signs. If histologic or PCR analysis is performed on CNS PROGNOSIS
tissue, then the patient need not demonstrate neurologic • The clinical course is usually one of progression to death
signs (i.e. asymptomatic CNS infection). within 6–12 months but can be fulminant, leading to
death within weeks, in spite of treatment with appro-
Possible CNS Whipple’s disease priate antibiotics.
Must have any one of four systemic symptoms, not due to • Involvement of the neuraxis carries a poor prognosis,
another known etiology: even when the intestinal disease has been eradicated.
• Fever of unknown etiology. • Relapses are most common with CNS Whipple’s disease
• Gastrointestinal symptoms (steatorrhea, chronic diarrhea, and are often resistant to antibiotics.
abdominal distension, or pain).

342 343 342, 343 T2W axial


(342) and T1W axial (343)
post contrast MRI in a
patient with biopsy-proven
Whipple’s disease. Note
the areas of increased signal
with mass effect and some
enhancement in the basal
ganglia.The lesion was
initially mistaken for an
infarct but, because it
involves the carotid and
basilar territory, this would
be distinctly unusual.
Acute Aseptic Meningitis 291

VIRAL INFECTIONS

ACUTE ASEPTIC MENINGITIS CLINICAL FEATURES


Usually a rapid onset over hours of fever, headache, malaise,
neck stiffness, photophobia, lethargy, myalgia and irritability.
DEFINITION The patient usually remains coherent and can be roused
Acute meningeal irritation, benign and self-limiting, with easily. The occurrence of depressed consciousness, focal
sterile pleocytic CSF and complete recovery. neurologic signs, or epileptic seizures usually implies
encephalitis. The physical signs are not so marked and the
EPIDEMIOLOGY illness is not as severe and prolonged as bacterial meningitis.
• Incidence: 2–27 cases per 100 000 per year are reported. The pathogen is seldom identified clinically:
The incidence is probably higher by several multiples • Myalgia and myocarditis point to coxsackie.
because of under-reporting. • Skin rash to enteroviruses, or herpes zoster virus (344).
• Lifetime prevalence: 0.9 (95% CI: 0.6–1.0) per 1000. • Parotitis and orchitis to mumps.
• Infections occur throughout the year, with a preponderance • Arthralgia and lymphadenopathy to HIV.
in summer and autumn in temperate climates.
• Age: mostly in children and young adults. SPECIAL FORMS
Mollaret’s meningitis
ETIOLOGY AND PATHOPHYSIOLOGY A benign recurrent ‘non-infectious’ meningitis of hitherto
Virus infection: at least 70% of cases unknown cause characterized by fever, malaise and CSF pleo-
• Enteroviruses (over half cases of viral meningitis): cytosis (lymphocytes, neutrophils and endothelial cells), which
– Coxsackie-B. may respond to corticosteroids, colchicine or phenylbutazone.
– Echovirus. New diagnostic techniques, such as the PCR for detecting viral
• Herpes simplex virus (HSV) type 1 and type 2. DNA in CSF have shown HSV, predominantly type 2, to be
• Varicella-zoster. a major cause of benign recurrent lymphocytic meningitis.
• Mumps.
• Lymphocytic choriomeningitis. DIFFERENTIAL DIAGNOSIS
• HIV. • Meningitis:
– Early bacterial meningitis (see p.273).
Other causes – Partially treated bacterial meningitis.
• Chemical and physical agents: – Meningitis caused by fastidious bacteria.
– Myelography with oil-based contrast media. – Fungal and parasitic meningitis (fungi and parasites do
– Intrathecal drug administration. not grow readily in routine culture).
– Craniopharyngiomas. – Parameningeal inflammation, infection or neoplasia.
– Cholesterol crystals discharged from cholesteatoma. • Subarachnoid hemorrhage (see p.249).
– Pituitary apoplexy.
• Parasites. INVESTIGATIONS
• Granulomatous inflammation: sarcoidosis. Blood
• Connective tissue disease. Serologic studies of acute and convalescent serum samples (e.g.
• Malignant infiltration. coxsackie, echovirus, HSV, varicella-zoster, mumps, HIV, measles,
• Drugs: adenovirus, Epstein–Barr virus [EBV], cytomegalovirus [CMV]).
– Cytotoxic drugs.
– Non-steroidal anti-inflammatory drugs such as ibuprofen. CT or MRI brain scan
– Immunoglobulins. Typically neither scan will show any abnormality. The main
– Antibiotics such as penicillin, isoniazid, ciprofloxacin, reason for imaging is to exclude other causes of headache,
trimethoprim-sulfamethoxazole. such as a space-occupying lesion or raised intracranial
pressure, and ensure that it is safe to do an LP.
PATHOPHYSIOLOGY
Viral infections of the CNS are complications of systemic viral
infections. The virus gains access to the brain by the bloodstream 344
or, less commonly, by travelling along peripheral nerves. In order
for viruses to enter the CNS from the blood they must cross the
endothelial cell junctions of the blood–brain barrier (the ability
to do this is dependent on surface adhesion molecules on the
cells, and surface charges and cellular receptor of the virus).
Certain viruses preferentially infect the meninges, choroid
plexus, and ependyma rather than cerebral parenchyma, causing
meningitis; others infect neurons and glia to cause encephalitis.
There is, however, considerable overlap, and some viruses may
cause meningo-encephalitis. Drug-induced meningitis is either
a direct chemical irritation or a hypersensitivity reaction.

344 Herpes zoster skin rash in a patient with aseptic meningitis.


292 Infections of the Nervous System: Viral

CSF VIRAL ENCEPHALITIS


• Pressure: normal or raised slightly.
• Appearance: clear to the naked eye.
• Microscopy: up to 500–1000 white cells/mm3, mainly DEFINITION
lymphocytes; polymorphs may predominate in some cases Inflammation of the brain caused by a virus, often accompanied
early. In such cases it is prudent to re-examine the CSF by inflammation of the meninges (meningo-encephalitis).
12–24 hours later to identify a lymphocytosis and exclude
a bacterial cause. EPIDEMIOLOGY
• Protein: raised slightly. The most common cause of encephalitis world-wide but in
• Glucose: normal or reduced a little; CSF glucose can be certain locations and seasons other organisms such as
low in antibiotic-induced meningitis. malaria and other protozoans, rickettsiae and fungi may be
• Viral antigen and IgM antibody. more common causes of encephalitis.
• Culture: negative. • Incidence: 1 in 20 000 per year.
• PCR: may detect viral DNA. • Lifetime prevalence: 0.4 (95% CI: 0.2–0.7)/1000.
• Age: most common in the first decade of life, with a peak
Many laboratory tests have been applied to CSF (i.e. incidence of 1 in 500–1000 infants in the first 6 months
creatine kinase, lactate, lysozyme, and C-reactive protein) to of life, but any age may be affected.
try and differentiate a viral from a bacterial cause but none is • Gender: M=F.
sufficiently discriminating to be useful; nor is the blood count,
blood biochemistry and EEG. PATHOLOGY
Acute viral encephalitis
DIAGNOSIS • Cortical vessels in the gray matter and at the junction of
• Appropriate clinical features and a raised white cell count gray and white matter: striking endothelial and capillary
in sterile CSF. inflammation. Perivascular lymphocyte infiltration results
• Provided other similar treatable diseases have been excluded from either the passive transfer of virus across the
(see Differential diagnosis), it is not necessary in most cases endothelium at pinocytic junctions of the choroid plexus
to establish an exact etiologic diagnosis for treatment pur- or active replication of virus in capillary endothelial cells.
poses because the disease is usually benign and self-limiting. • Gray matter: lymphocytic infiltration and neuronophagia
may occur; astrocytosis and gliosis if progressive disease.
TREATMENT
Symptomatic treatment with analgesics and antiemetics is all Specific cell types infected by specific virus and their pathologies
that is required. Antibiotics should not be given. • Poliovirus (motor neurons), rabies (limbic system), and
togavirus infect neurons and cause necrosis.
CLINICAL COURSE AND PROGNOSIS • Lymphocytic choriomeningitis virus infects choroid plexus,
Determined by the underlying cause. Most cases do not meninges, and ependyma and causes fatal choriomeningitis.
progress; resolution begins within a few days and is complete • Mumps virus can infect the ependymal cells of newborns.
within 2 weeks in most patients. A few will have persistent • Papovavirus infects oligodendrocytes and causes
malaise and myalgia for some weeks. demyelination.
• Parvovirus infects endothelial cells and produces a
hemorrhagic encephalopathy.
345 • Varicella-zoster virus infects multiple cell types, including
blood vessels causing a vasculopathy leading to
ischemic/hemorrhagic infarction and oligodendrocytes
causing demyelination.
• HSV infects neurons of the temporal and inferior frontal
lobes (345).

Unique histopathologic features


• Cowdry type A inclusion bodies: herpes virus.
• Negri bodies: rabies.

Postinfectious encephalomyelitis
Perivascular inflammation and demyelination.

ETIOLOGY
Most cases are sporadic accompaniments of common
infections such as mumps, measles, herpes simplex and
varicella-zoster, and less commonly EBV, but in as many as
one-third of all cases no specific etiology can be established.
345 T2W MRI brain scan of the temporal regions in a patient with
herpes simplex encephalitis. Note the increased signal in the medial Acute viral encephalitis
temporal gray and white matter, more on the right (arrows), associated Endemic
with some swelling.These features are typical of HSV encephalitis though • HSV: 10% of cases (see p.295).
are not specific, and a normal MRI does not exclude encephalitis. • Rabies.
Viral Encephalitis 293

Epidemic or sporadic Preferential sites of involvement


• Japanese B encephalitis: probably the most common Certain viruses preferentially infect neurons and glia, and
epidemic infection outside North America. some exhibit tropism towards specific neuronal cell types:
• Arthropod-borne viruses (arboviruses – viruses transmit- • Frontal and temporal lobes: HSV.
ted by insects): • Cerebellum: varicella-zoster and EBV.
– St Louis encephalitis. • Limbic system: rabies.
– Eastern equine encephalitis.
– Venezuelan equine encephalitis. Direct virus-induced cellular damage, secondary
– La Crosse virus, the most important mosquito-borne immunologically mediated events, or both?
virus in the USA; found in all 13 states east of the Missis- Serum IgG and IgM antineuronal antibodies in patients
sippi river. Encephalitis caused by La Crosse virus is with encephalitis suggest a role for autoimmune mechan-
transmitted by mosquitos and most commonly affects isms; e.g. in EBV encephalitis either EBV-transformed B
children and young adults. There is no therapy but most cells secrete antineuronal antibodies or antibodies against
patients recover (death occurs in <1%). The primary EBV proteins cross-react with neuronal antigens.
reservoirs of the virus are small forest mammals.
• Enteroviruses: coxsackie virus, echovirus and poliovirus Postinfectious encephalomyelitis
(usually cause aseptic meningitis). An autoimmune phenomenon, initiated by the viral
• Paramyxoviruses: measles, mumps, respiratory syncytial pathogen and characterized by immunoregulatory failure
virus, parainfluenza. rather than immunosuppression. There is an apparent latent
• CMV. phase between the acute illness and the onset of neuro-
• EBV. logical symptoms due to demyelination.
• Rubella.
• Influenza. CLINICAL FEATURES
• Adenovirus. • Can be enormously variable.
• Yellow fever. • Onset is usually insidious but may occasionally be
cataclysmic.
Postinfectious encephalomyelitis • Prodrome of fever, malaise, myalgia, irritability, mild
• Influenza A. upper respiratory tract infection, rash or parotitis lasts
• Measles (1 in 1000 cases; in countries where vaccination 4–10 days.
is not routine). • Fever (not invariable).
• Varicella-zoster virus. • Headache.
• EBV. • Stiff neck (meningismus).
• Mumps. • Photophobia.
• Rubella. • Disorientation.
• Vaccination (vaccinia, rabies). • Behavioral disturbances.
• No recognizable preceding illness sometimes. • Focal neurologic signs such as dysphasia and hemiparesis
(frontal lobes), epileptic seizures (cerebral cortex), ataxia
PATHOGENESIS (cerebellum), and memory disturbance (temporal
The rare occurrence of encephalitis after viral infection lobes/limbic system) depending on which part of the
depends on the nature and virulence of the organism and brain is involved. It is vital to distinguish between
the host immune response. generalized and focal neurologic dysfunction, and to
determine whether focal neurologic signs could be
Access routes to the brain explained by the tropism of certain viruses to infect
Viruses generally gain access to the CNS by the certain CNS cell types (see Pathology and pathogenesis).
hematogenous or peripheral intraneuronal routes. • Depressed consciousness.
• Raised intracranial pressure.
Haematogenous
Viruses may enter the body through the skin following an Other possible features
insect bite (as with arbovirus infection) or via the respiratory • Season of the year and climate:
or gastrointestinal route where they replicate locally. – Summer and damp climates: mosquito propagation (cf.
Transient viremia may ensue if the inoculum spills into the arthropod-borne virus infection).
blood, where it is transported to the reticuloendothelial – Late summer/early autumn in temperate climate:
system (particularly the liver, spleen, lymph nodes and enteroviral infections.
sometimes muscle) whence, following further replication, • Travel to or from an area that harbors known vectors (i.e.
secondary viremia may occur and the organism spreads to arboviruses).
other sites including the CNS. • Insect bite: tick or mosquito (cf. arbovirus).
• Animal bite: potentially rabid (cf. rabies).
Neuronal • Skin rash: varicella or measles.
Organisms, such as herpes simplex virus and rabies may • Parotitis: mumps.
ascend neurons centripetally and directly invade brain • Hepatosplenomegaly: EBV, CMV.
parenchyma. Clinical manifestations are caused by cell • Immunocompromised (neonates, organ transplant
dysfunction from viral invasion and associated inflammatory recipients, HIV infection): CMV.
change.
294 Infections of the Nervous System: Viral

The disease may be more severe in the very young, very CSF
old and immunocompromised. Abnormal in 95% of patients:
• Pressure: increased.
SPECIFIC FORMS • Microscopy: cell count: raised; 10– several hundred white
• Herpes simplex encephalitis (see p.295). cells/mm3.
• Varicella-zoster encephalitis (see p.299). • Cell type: lymphocytes; polymorphs may predominate
early, red cells may be present if there is a necrotizing
DIFFERENTIAL DIAGNOSIS component, as in herpes simplex encephalitis.
Other intracranial infections • Organisms: not seen.
• Viral meningitis: usually only fever, headache, neck • Protein: raised.
rigidity and photophobia without signs of brain • Glucose: normal.
dysfunction such as mental change and focal neurologic • Viral antigen or IgM antibody.
signs. • IgG index and oligoclonal bands.
• Bacterial meningitis. • PCR: a positive result increases the likelihood of a
• Mycoplasma pneumoniae encephalitis. definite diagnosis of viral meningo-encephalitis by 88
• Malaria and other protozoal and fungal infections. times (95% CI: 21–378) compared with a negative PCR
• Cerebral abscess. result.
• Culture: of little value for virus isolation.
Non-infectious conditions • Interferon alpha in the CSF: only moderately sensitive
• Cerebral tumor or metastases. but highly specific for virus infection of the CNS; not
• Connective tissue disease. widely available.
• Drug abuse.
• Reye’s syndrome Brain biopsy
• Hypoxic-ischemic encephalopathy • No role in confirming the diagnosis of viral encephalitis
• Hemorrhagic shock and encephalopathy (neither sensitive nor safe).
• Metabolic/toxic encephalopathy • Biopsy has a role in clarifying the differential diagnosis
• Mitochondrial encephalomyopathy, lactic acidosis, and of a mass lesion that could be an abscess, granuloma,
stroke-like episodes (MELAS). tumor or inflammatory mass, particularly if no response
• Deep cerebral venous thrombosis. to acyclovir.

INVESTIGATIONS DIAGNOSIS
Blood Blood viral serology demonstrating seroconversion or
• Full blood count. seroboosting in paired acute and convalescent serum is still
• Urea and electrolytes: hyponatremia secondary to the mainstay of diagnosis, but PCR of infected tissue
SIADH. (blood, CSF) is soon likely to provide rapid and accurate
• Blood film: malarial parasites. diagnosis.
• Blood cultures.
• Viral culture of samples of respiratory secretions, blood, TREATMENT
CSF, urine and stool, taken within 4 days of onset of the Antiviral chemotherapy
illness if possible; throat swab in viral transport medium. Acyclovir must be commenced immediately if there is any
• Blood viral serology (paired acute and convalescent possibility of HSE (see p.295).
samples to demonstrate seroconversion or seroboosting)
and PCR (detection of viral antigen by amplification of Symptomatic
nucleic acid sequences): herpes simplex, measles, mumps, Supportive care: adequate oxygenation, hydration and
rubella, influenza, parainfluenza, adenovirus, varicella- nutrition; maintain fluid and electrolyte balance, control the
zoster, CMV, EBV. temperature and seizures, and treat dysfunction of other
organs.
Cranial CT scan
Normal, or diffuse or focal areas of low attenuation, with or Steroids
without space effect. The findings are non-specific and can In severe cases of acute post infectious encephalomyelitis,
look just like an infarct. Changes may not become apparent corticosteroids are given empirically (prednisolone 20 mg
for 5 or 6 days, if ever. CT is useful to exclude other possible four times daily).
causes of headache and drowsiness but will not give positive
confirmatory evidence of encephalitis. Complications
• Cerebral edema and raised intracranial pressure:
MRI brain dexamethasone, mannitol, glycerol (glycerin), and
More sensitive than CT, particularly early in the illness, and intubation and hyperventilation may be required.
may help identify other pathologies in the differential Dexamethasone is often used despite the theoretical
diagnosis, such as tumors and abscesses. disadvantage that interferon synthesis may be inhibited.
There is no consensus on the correct treatment.
EEG • Epileptic seizures: anti-epileptic drugs such as sodium
Abnormal, most often non-specific diffuse slow wave valproate or carbamazepine, if seizures occur.
activity, sometimes with epileptiform activity. • Secondary infections: treat as necessary.
Herpes Simplex Virus Encephalitis (HSE) 295

PROGNOSIS HERPES SIMPLEX VIRUS


• Mortality: about 3%. ENCEPHALITIS (HSE)
• Serious morbidity: about 7%.
• At 10 year follow-up: almost half the survivors have
motor dysfunction and educational dysfunction; well DEFINITION
over a third have neurologic dysfunction, and almost a An acute asymmetric necrotizing infection of the brain
fifth have behavioral, self-care and sensory dysfunction. parenchyma, primarily the frontal and temporal lobes and
the limbic system, by HSV, usually type 1.
Adverse prognostic factors
• Young age: children younger than 3 years of age: 34% EPIDEMIOLOGY
case fatality. The most common cause of sporadic, non-epidemic, acute
• Low conscious level. focal encephalitis in immunocompetent adults in developed
• Abnormal oculocephalic responses. countries; comprising about 10% of cases of encephalitis in
• High ratio of albumin in CSF to that in serum. the United States.
• Laboratory evidence of infection of the CSF. • Incidence: 1 per 500 000 people per year (an
• Organism: underestimate; milder cases go unrecognized). The
– Herpes simplex virus encephalitis: 12-fold excess incidence is higher in immunocompromised individuals
mortality. (e.g. those with HIV infection) but most patients are
– M. pneumoniae encephalitis: sevenfold excess mortality. immunocompetent.
– Measles encephalitis: 15% mortality, most survivors • It occurs throughout the year.
disabled. • Age: any age; one-third <20 years of age, and one-half
– Varicella-zoster encephalitis: 10% mortality with diffuse >50 years.
form. • Gender: M=F.
– EBV and mumps encephalitis: excellent prognosis.
PATHOLOGY
PREVENTION Acute focal hemorrhagic necrotizing inflammation
Immunization of the whole child population is necessary for predominantly in the temporal and inferior frontal lobes
effective prevention. (orbital surface) but adjacent frontal, parietal, and occipital
lobes and cingulate gyri may also be involved.
Pathologic findings are due to the direct cytopathic
effects of viral infection and include neuronal necrosis,
reactive astrocytosis, perivascular lymphocyte cuffing and
characteristic Cowdry type A intranuclear inclusion bodies.

ETIOLOGY AND PATHOGENESIS


HSV type 1 (HSV-1, >90% of cases) and type 2 (HSV-2, up
to 10% of cases):
• Both viruses are widespread in the adult population, with
a rate of seropositivity of 60–100% for HSV-1 and
10–80% for HSV-2. Levels of seropositivity are related
to socio-economic status and geographic area.
• Primary HSV infection occurs relatively early in life and
is usually asymptomatic.
• Pathogenesis of HSE is only partly understood, and
includes primary HSV infection, re-infection and
reactivation of latent virus infection.

Neonatal HSE
• HSV-1 or HSV-2.
• HSE caused by HSV-2 in neonates with multiple organ
disseminated disease is probably blood-borne and is
associated with a diffuse encephalitis, resulting in
generalized encephalomalacia.
• Isolated HSE in neonates (without systemic disease) is
transmitted by the peripheral intraneuronal route; tends to
infect one temporal lobe and then the other as the disease
progresses, as is also the case in older children and adults.
296 Infections of the Nervous System: Viral

Childhood (>2 years) and adult HSE • Brain tumor.


• HSV-1 in the vast majority (>90%) of cases; HSV-2 in • Granuloma.
<10% (HSV-2 tends to cause viral meningitis in adults, • Other forms of viral encephalitis: e.g. arbovirus
particularly in association with primary genital infection). (appropriate travel history), mumps, enterovirus.
• HSV-1 enters the nervous system by way of the olfactory • Deep cerebral venous thrombosis.
bulb, or reactivation of a latent infection in the trigeminal • MELAS.
and other cranial nerve ganglia, with centripetal spread
of virus to the frontal or temporal lobes of the brain INVESTIGATIONS
through its branches that innervate the meninges. The Cranial CT scan
virus causes encephalitis by direct invasion of brain CT is useful to exclude other possible causes of headache
parenchyma but the mechanism of reactivation in HSE and drowsiness. It may be normal initially but commonly
is poorly understood. (although not always) it eventually shows low density lesions
due to edema and necrosis, sometimes with hemorrhagic
CLINICAL FEATURES change, in the inferior frontal and medial temporal lobe, and
• Onset is commonly quite abrupt but may be insidious. which may demonstrate enhancement after injection of
• Concurrent cold sores or genital herpes are uncommon. intravenous contrast. The changes may not become
• Prodrome of fever, headache, lethargy and nausea over a apparent however for 5 or 6 days and can look just like an
few days. infarct (346, 347). Herpes encephalitis is therefore a
• Behavioral change. diagnosis which needs a low index of suspicion as there is
• Disorientation. an effective treatment.
• Altered conscious level.
• Focal neurologic signs such as anosmia or altered MRI brain scan
olfactory perception, dysphasia, hemiparesis, memory Hypointense lesions on T1W images and hyperintense
loss, olfactory or gustatory hallucinations, bizarre lesions on proton density and T2W images (T2WI) at an
behavior and complex partial seizures. earlier stage than by CT. The T2WI typically shows
increased signal in the temporal and inferior frontal lobes
Other, unusual presentations of HSV infections of the (more ‘whiteness’ on T2WI) accompanied by swelling of
nervous system include: the affected cortex, and possibly evidence of hemorrhage
• Mild/subacute encephalitis. (black bits on the T2WI). The appearance can be quite
• Psychiatric syndromes. asymmetric (348, 349).
• Brainstem encephalitis. MRI brain is more sensitive than CT to the abnormalities
• Benign recurrent meningitis. of encephalitis particularly early in the illness and is the
• Myelitis. imaging investigation of choice. MRI also may show more
extensive involvement than CT (350, 351) and help
DIFFERENTIAL DIAGNOSIS identify other pathologies such as cerebral tumors or
• Meningitis: bacterial, fungal and mycobacterial. abscesses.
• Brain abscess.
• Subdural empyema. EEG
• Stroke: frontotemporal infarction, hemorrhagic infarction EEG may be normal early in disease but usually is abnormal
or hemorrhage. and shows non-specific slow wave activity in the first few
• Subdural hematoma. days. Later, more characteristic periodic lateralized

346 347 346, 347 Cranial CT scan in


the axial plane of a patient
with herpes simplex
encephalitis involving the
right inferior frontal and
antero-medial temporal lobe.
The lesion extends to involve
the external capsular region
and the associated mass
effect is seen to compress
the frontal horn of the right
lateral ventricle.These scans
(with 348, 349) highlight the
difficulty of distinguishing focal
encephalitis from infarction
sometimes.
Herpes Simplex Virus Encephalitis (HSE) 297

epileptiform discharges (PLEDs) which consist of 50–100 mV, occurring every 1 to several seconds (352).
paroxysmal, high voltage, sharp waves, sharp and slow The PLEDs are maximal over the affected temporal region
waves, spikes, or multiple spikes with amplitude of and, if bilateral, suggest HSE.

348, 349 MRI brain scan, 348 349


T2W images in the axial
plane of a patient with
herpes simplex encephalitis
involving the right inferior
frontal and antero-medial
temporal lobe.The lesion
extends to involve the
external capsular region and
the associated mass effect is
seen to compress the frontal
horn of the right lateral
ventricle.

350, 351 MRI brain scan, 350


T2W images in the axial 351
(350) and coronal (351)
planes showing extensive
high density due to cystic
gliotic change in the anterior
three-quarters of the right
temporal lobe, the medial left
temporal lobe, the inferior
and lateral right frontal lobe,
the right insular cortex, and
the right cingular gyrus of a
patient with severe amnesia
due to bilateral herpes
simplex encephalitis several
years previously.The right
cerebral peduncle has
atrophied.

352 Electroencephalogram of a patient with herpes simplex 352


encephalitis showing complex discharges composed of spiky
elements mixed with slower waveforms diffusely over the left
hemisphere and recurring at not quite regular intervals.
Periodic lateralized epileptiform discharges (PLEDs) usually
indicate an acute or rapidly progressive focal brain lesion such
as infarction, hemorrhage, glioblastoma, abscess or encephalitis.
Rarely, a severe metabolic disturbance may be present.
Occasionally, PLEDs may be bilateral, such as in patients with
severe brain injury due to hypoxia, encephalitis, and status
epilepticus. PLEDs are usually short lived and disappear within
a few weeks, irrespective of etiology and intervention, but a
chronic seizure disorder may remain.
298 Infections of the Nervous System: Viral

CSF TREATMENT
• May be normal in about 5% of cases. Prompt antiviral chemotherapy
• Pressure: increased. Acyclovir must be commenced immediately in any patient
• Microscopy: cell count raised; 5–1000 (typically 5–100) with suspected HSE. The dose is 10 mg/kg given by
white cells per mm3. intravenous infusion over 1 hour, every 8 hours for
• Cell type: lymphocytes; polymorphs may predominate 10–14 days. Acyclovir is extremely well tolerated with few
early, red cells may be present. adverse effects in patients with normal renal function, and
• Organisms: not seen. so it is often appropriate to treat patients with suspected
• Protein: normal or raised up to 2 g/l. HSE presumptively pending the results of further
• Glucose: normal. investigations. New anti-HSV agents with enhanced
• IgG index and oligoclonal bands: raised IgG; oligoclonal bioavailability, such as famciclovir are also available.
bands may be present.
• HSV antibody titers: an increased CSF:serum quotient Symptomatic
for the HSV antibody titer, adjusted for CSF-blood Adequate oxygenation with assisted ventilation if necessary,
barrier integrity, indicates a specific intrathecal antibody optimal fluid and electrolyte balance and hydration,
response. nutrition and analgesia.
• HSV antigen: virus isolated in less than 5% of cases.
• PCR for detection of HSV DNA in CSF: sensitive but Complications
not 100% reliable. If positive, it remains positive for at • Cerebral edema and raised intracranial pressure:
least 5 days after initiation of acyclovir therapy; the dexamethasone (4 mg 6 hourly i.v. or orally), mannitol
method of choice for diagnosis of HSE. (1 g/kg i.v. as a 20% solution over 1 hour), glycerol
• Viral culture: low sensitivity in detecting HSV in adults (glycerin) and intubation and hyperventilation may be
but HSV can be retrieved from CSF in about half of required but remain controversial. Dexamethasone is
babies with encephalitis or disseminated disease. often used despite the theoretical disadvantage that
interferon synthesis may be inhibited. There is no
Blood viral serology consensus on the correct treatment.
Acute and convalescent serum samples to demonstrate • Epileptic seizures: anti-epileptic drugs such as sodium
seroconversion or seroboosting. valproate or carbamazepine, if seizures occur.
• Secondary infections: treat as necessary.
Brain biopsy
The definitive diagnostic technique (characteristic histologic CLINICAL COURSE AND PROGNOSIS
changes, immunofluorescence for HSV antigens, culture of Untreated, HSE is rapidly progressive, typically leading to
HSV, and detection of viral DNA by in situ hybridization), brain edema and death after 7–14 days; the 1 month
but has little role because it is neither sensitive or specific mortality rate exceeds 70%, and only about 2.5% regain
(or safe), and in many cases it is appropriate to treat on the normal function.
basis of clinical, EEG and radiologic findings and observe Early aggressive antiviral therapy with acyclovir reduces
the clinical response to acyclovir. It does have a role in mortality to 28% at 18 months, and is superior to that of
clarifying the differential diagnosis of a mass lesion that vidarabine which reduces mortality to 28% at 1 month and
could be an abscess, granuloma, tumor or inflammatory 44% at 6 months. At 2 years after treatment with acyclovir
mass, particularly if no response to acyclovir. about 38% are normal or have mild impairment, 9% have
moderate sequelae, and 53% have died or are severely
DIAGNOSIS impaired. Among vidarabine recipients, only 15–20% are
The diagnosis is suggested by neurologic, electrophysiologic judged to be normal on long term follow-up.
and neuroimaging signs of fronto-temporal lobe necrosis Outcome is influenced by:
with serologic evidence of seroconversion or seroboosting • Age.
in paired acute and convalescent serum. However, a specific • Level of consciousness (if Glasgow coma score is <6,
diagnosis can only be achieved by virologic studies of the outcome is uniformly poor).
CSF, PCR of the CSF, or by examination of brain tissue • Duration of untreated disease (if treatment is com-
obtained by biopsy or at post mortem. Nested PCR menced within 4 days of onset of symptoms, the chance
amplification is a rapid, sensitive, inexpensive, and relatively of survival increases from 72% to 92% at 1 month).
non-invasive method for establishing the initial diagnosis of • Infants with HSV-1 encephalitis have a significantly
HSE and for monitoring the response to therapy. Although better neurologic outcome than those with HSV-2
nested PCR is more susceptible to contamination artefact infection of the CNS.
(false positive) than traditional methods such as virus
isolation or serology, diagnostic PCR has been successfully Relapse
incorporated into many laboratories and is likely to become Relapse of HSE after acyclovir or vidarabine therapy can
the gold standard. HSV sequences can be detected for up occur in up to 5% of cases. In the few cases in which HSV
to 5 days after commencing acyclovir and can differentiate isolates were tested for resistance to acyclovir, they remained
between HSE due to HSV-1 and HSV-2. sensitive. Re-institution of treatment with a higher dose of
acyclovir (15 mg/kg every 8 hours) for a longer course
(21 days) or in combination with vidarabine has been tried
in a few patients but the results are inconclusive.
Varicella-Zoster Virus Encephalomyelitis 299

VARICELLA-ZOSTER VIRUS • Arteritis: involves small vessels with fibrinoid necrosis and
ENCEPHALOMYELITIS granulomatous inflammatory infiltrates causing throm-
bosis and multifocal deep white matter lesions
(demyelination or infarction). In purely ischemic lesions,
DEFINITION characteristic Cowdry A inclusions may be scant or
Inflammation of the brain or spinal cord caused by infection absent. Rarely, mycotic aneurysms may form and
with the varicella-zoster virus (VZV), an α-herpes virus. rupture, causing hemorrhage.

EPIDEMIOLOGY Brain infarction does not usually occur until weeks to


• Incidence: rare, about 1 case per million population per months after trigeminal nerve distribution herpes zoster rash
year. (shingles).
• Prevalence: about 0.1% of VZV infections involve the
nervous system and 90% of these are in the form of ETIOLOGY AND PATHOPHYSIOLOGY
encephalitis. VZV encephalitis has become more Chickenpox
prevalent in the era of acquired immunodeficiency Primary VZV infection causes the typical rash of
syndrome and other immunosuppressive diseases. chickenpox. During chickenpox, VZV establishes latency in
• Age: in immunocompetent individuals, herpes zoster is multiple dorsal root ganglia.
predominantly a disease of the elderly.
• Gender: M=F. Shingles
After a variable, but usually long period, the VZV reactivates
PATHOLOGY to cause shingles (herpes zoster), which is a disease of the
A combination of ischemic and demyelinative features. peripheral nervous system causing severe radicular pain,
herpetic vesicles on the skin usually in the distribution of one
Multifocal demyelinating leukoencephalomyelitis to three dermatomes (353–355), and pain after the vesicles
Infection of oligodendrocytes causes demyelinative lesions have healed (post herpetic neuralgia). Focal muscle weakness
in the deep white matter which are smaller and less
coalescent than those seen in PML. Target-like lesions with
central necrosis and surrounding myelin pallor, and Cowdry 353
type A intranuclear inclusions in surrounding glia and
neurons are the pathologic hallmarks of varicella-zoster
multifocal leukoencephalitis. Cowdry type A inclusions are
virtually specific for herpes viruses but their absence does
not rule out subtle infections.

Angiopathy causing cerebral infarction and


hemorrhage
• Non-inflammatory angiopathy: involves mainly large
cerebral arteries with non-inflammatory hyperplasia of
intima and media, sometimes resulting in thrombosis and
multifocal large ischemic and hemorrhagic infarcts in the
cerebral cortex and subcortical white matter.

354

355

353–355 Herpes zoster infection (shingles) in the distribution of the


ophthalmic division of the trigeminal nerve.
300 Infections of the Nervous System: Viral

(zoster paresis) occurs in about 5% of cases, of whom about Cranial CT scan


60% recover completely, 25% incompletely, and 15% do not CT shows focal or multifocal areas of low attenuation and
improve. Encephalitis may rarely follow shingles. swelling, and sometimes petechial or frank hemorrhage.
Post herpetic pain probably occurs because reactivation Intracranial calcification may occur as a sequelae to zoster
of VZV in the dorsal root ganglia appears to cause structural encephalitis.
damage which results in chronic stimulation of the CNS.
This re-sets the homeostatic mechanisms controlling pain, MRI brain
so that the patient perceives chronic pain, severe pain from MR may reveal a spectrum of lesions from multifocal ischemic
minor stimuli, and allodynia (abnormal sensations). and hemorrhagic infarction of varying size, depending on the
caliber of the vessel involved, to a leukoencephalopathy
Encephalomyelitis characterized by multifocal small ovoid lesions involving white
Caused by reactivation of the VZV, latent in sensory ganglia matter more than gray matter, and often more concentrated
after primary infection. Several mechanisms have been at gray-white matter junctions, due to both demyelination
proposed, including hematogenous, transaxonal and necrosis caused by a small vessel vasculopathy. There are
(transneural) and/or vascular spread to the CNS, and an no specific distinguishing features however.
immune-mediated effect. VZV may enter the CNS via
transaxonal spread from ocular or dermatomal zoster and CSF
directly invade and infect small blood vessels, ultimately Findings are similar to viral encephalitis (see p.292).
leading to ischemic infarction and necrosis of brain or spinal Enzyme immunoassay for antibody to VZV and PCR for
cord, or directly infect oligodendrocytes, leading to detection of VZV DNA.
demyelination. The finding of an active ‘centrifuge wave’ of
infection at the periphery of lesions and their random EEG
distribution in the brain however, suggests hematogenous Abnormal, most often non-specific focal or diffuse slow
spread during viremia. wave activity, occasionally with epileptiform activity.

Risk factors Cerebral angiography


Immunocompromised: elderly, acquired immunodeficiency May reveal segmental narrowing of large and medium-sized
syndrome (AIDS), immunosuppressive therapy. arteries in cases of angiopathy.

CLINICAL FEATURES Brain biopsy


In about half of cases, usually young people with chicken Necrosis and gliosis consistent with infarction;
pox, the encephalitis predominantly affects the cerebellum, immunohistochemical localization of viral antigens and in
causing cerebellar ataxia, dysarthria, headache and situ hybridization or PCR detection of VZV DNA.
drowsiness. The skin rash often precedes the neurologic
symptoms by about a week but occasionally the rash of DIAGNOSIS
chicken pox may follow. • The finding of brain infarction (ischemic or hemorr-
In immunocompromised and older individuals zoster hagic) and/or demyelination lesions in an immuno-
encephalomyelitis and granulomatous arteritis (the latter after compromised patient with or without a history of zoster
zoster ophthalmicus) usually develops concurrent with rash rash, and with a fever and encephalopathy, should
or weeks to months after acute herpes zoster rash (353–355) suggest the diagnosis of VZV encephalitis.
and is characterized by fever, seizures, mental status changes • Serologic evidence of seroconversion or seroboosting in
and multifocal or isolated focal neurologic deficits. paired acute and convalescent serum or the detection of
antiviral antibody and VZV DNA in CSF can confirm the
DIFFERENTIAL DIAGNOSIS diagnosis.
• Other forms of viral encephalitis (see p.292): CMV, • Brain biopsy or autopsy evidence of immunohistochemical
HIV-1 related encephalitis, HSE (see p.295). localization of viral antigens and in situ hybridization or
• Hemorrhagic multifocal leukoencephalopathy secondary PCR detection of VZV DNA are required for the
to angiopathy caused by toxoplasmosis, aspergillus, unequivocal demonstration of virus-specific antigens or
CMV, HSV, ameba, HIV-1 (see p.301). nucleic acids.
• Isolated angiitis of the CNS (see p.227).
• Stroke: ischemic (see p.202) or hemorrhagic (see p.238). TREATMENT
• Progressive multifocal leukoencephalopathy: typically Encephalomyelitis
causes hemiparesis, aphasia, visual-field defects and • General management is as for viral encephalitis (see
confusional states; MRI reveals larger and more p.292).
coalescent zones of demyelination than VZV • Specific antiviral therapy with intravenous acyclovir
encephalitis; and CSF PCR detects JC virus DNA. 10 mg/kg every 8 hours for 10–14 days, or alternatively
famciclovir.
INVESTIGATIONS
Blood viral serology PROGNOSIS
Acute and convalescent serum samples to demonstrate Patients with the cerebellar form usually recover completely
seroconversion or seroboosting. but at least 10% of those with the general form die.
HIV-Associated Cognitive/Motor Complex (HIV-CMC) 301

HUMAN IMMUNODEFICIENCY into the brain and viral entry into the brain. Certain strains
VIRUS (HIV)-ASSOCIATED of HIV-1, characterized by specific sequences in the
COGNITIVE/MOTOR COMPLEX envelope region, may have a predilection for infecting the
(HIV-CMC) brain and causing dementia.

CLINICAL FEATURES
DEFINITION Subtle early in the course, and may only be identified by
A distinct neurologic syndrome of subcortical dementia formal neuropsychologic evaluation.
characterized by slowness and imprecision of cognition and Three main categories:
motor control, and called the AIDS dementia complex or • Cognitive: initially a predominantly subcortical dementia
HIV-1-associated cognitive/motor complex. It is the most characterized by forgetfulness and slowed mental and
important ‘primary’ neurologic complication of HIV infection. motor abilities.
• Motor: loss of balance and occurrence of spastic leg
EPIDEMIOLOGY weakness.
HIV dementia usually occurs late in the course of HIV • Behavioral: apathy and social withdrawal (often mistaken
disease, when CD4 lymphocyte counts are less than as depression); sometimes organic psychosis, such as
200 cells/mm3, but in 3–10% of patients it is the first acute mania.
manifestation of AIDS.
• Incidence: The disease stages have been classified from normal
– 2 (95% CI: 0.8–5) per 100 000 per year. (stage 0) to profoundly impaired (stage 4).
– 7% annual incidence during the first 2 years after AIDS
diagnosis. Examination findings
– 7.3 cases per 100 person years for people with CD4 • Cognitive: psychomotor slowing, and abnormalities of
counts of ≤100. reaction time, memory, executive function and complex
– 3.0 cases per 100 person years if CD4 count 101–200. attention.
– 1.3 to 1.7 cases per 100 person years if CD4 count • Eye movements: abnormal: saccadic pursuit and
201–500. hypometric saccades.
– 0.5 cases per 100 person years if CD4 count >500. • Limbs: bradykinesia; impairment of rapid alternating and
• Prevalence: 0.4% during the asymptomatic phase, repetitive movements and tandem (heel-to-toe) gait.
7.5–27% during the late stages of HIV disease; at least • Reflexes: brisk and symmetric; primitive reflexes may
15% of AIDS patients develop moderate to severe manifest in later stage disease.
dementia, and up to another 20–25% have less severe
cognitive dysfunction. Concurrent illness
Subclinical HIV-associated dementia may be ‘unmasked’ by
PATHOLOGY intercurrent illnesses, such as depression, metabolic derange-
Key features ments, systemic and cerebral opportunistic infections (e.g.
• Multiple foci of microglia, macrophages, and toxoplasmosis, PML, cryptococcal meningitis), and lym-
multinucleate giant cells or the presence of HIV-infected phoma, which are important to look for (and treat).
cells in the CNS (356).
• Other abnormalities: inflammatory cell infiltrate, multi-
nucleate giant cells (HIV encephalitis), reactive gliosis and
diffuse white matter pallor (HIV leukoencephalopathy).

The pathologic changes are often less prominent than the


clinical symptoms. 356
PATHOGENESIS
Complex and obscure but determined by :
• The effectiveness of the immune defences and their
capacity to suppress viral replication within the brain.
• The genetic variance of the virus and its macrophage
tropism (i.e. capacity to replicate well in macrophages
and related cells but not well in lymphocyte-derived
chronic cell lines).
• The genetic predisposition of the individual.

Probably due to indirect effects of HIV infection on the


CNS. Infected and activated macrophages and microglia
produce increased intracerebral cytokines and neurotoxins
leading to secondary neuronal damage, principally in the 356 Histologic section of the brain of a patient who died with the
subcortical and basal ganglia regions, but also in neocortical HIV-associated cognitive/motor complex showing an inflammatory
areas. White matter pallor may be due to demyelination or cell infiltrate with multiple foci of macrophages and multinucleate
changes in the blood–brain barrier that are mediated by giant cells. (H&E stain, magnification ×16.) (Courtesy of Professor
cytokines and contribute to extravasation of serum proteins BA Kakulas, Royal Perth Hospital,Western Australia.)
302 Infections of the Nervous System: Viral

DIFFERENTIAL DIAGNOSIS DIAGNOSIS


Other causes of subcortical dementia: A diagnosis of exclusion(359); there are no specific
• Vascular dementia. laboratory or imaging results.
• Hydrocephalus. Blood and CSF studies are helpful to screen for systemic
• Subcortical tumor. infections (VDRL, cryptococcal antigen). The diagnosis is
• Huntington’s disease. likely if the patient has a subcortical dementia, positive HIV
• Parkinson’s disease. serology and typical neuroimaging and CSF findings.
• Syphilis (see p.312).
• Hypothyroidism. MANAGEMENT
• Vitamin B12 deficiency. Highly active antiretroviral therapy (HAART)
• Non-convulsive status epilepticus. • Simultaneous treatment with at least three combination
• Progressive multifocal leukoencephalopathy. antiretroviral medications, including various com-
• Subacute/chronic meningitis (see p.279). binations of nucleoside reverse transcriptase (RT)
• Cryptococcal meningitis (see p.318). inhibitors, non-nucleoside RT inhibitors, and at least one
protease inhibitor (PI).
INVESTIGATIONS • About 30% of patients with HIV-CMC show
Neuropsychologic assessment improvement in neurocognitive function after 3 months
Characteristically reveals impairment in cognitive domains of HAART, and improvement on psychomotor speed
of memory, complex attention and executive function. over the course of 1 year.
• May reduce plasma and CSF HIV RNA viral loads to
Blood undetectable levels, but viral suppression is often
• Full blood count and ESR. incomplete and drug resistance may develop
• Thyroid function tests. concomitantly. Another problem is that the newer
• Vitamin B12 level. antiretroviral medications, such as the majority of the PIs,
• VDRL/RPR, TPHA. do not cross the blood–brain barrier readily.
• HIV serology. • Adverse effects of antiretroviral therapy include mito-
• Cryptococcal antigen. chondrial toxicity, hypersensitivity, lipodystrophy and
• CD4 cell count: usually around or less than 100/µl at more drug-specific adverse effects (Carr and Cooper,
time of diagnosis. 2000).
• Plasma HIV RNA viral load.
Clinical and laboratory follow-up
Cranial CT and MRI scan Repeat clinical, neurologic and neuropsychologic assessment
Scans are rarely normal. Both often show various degrees of every 4–6 weeks to ascertain any treatment benefit. Involve
cortical atrophy, ventricular enlargement, widened sulci, and physiotherapist, occupational therapist and social worker
diffuse or multifocal white matter abnormalities (357, 358). early.
The amount of atrophy on brain MRI tends to correlate
with the severity of HIV dementia. MRI may also reveal Counselling
areas of high signal in the white matter on T2 and PDWI. Frequently required for patient and carers early in the illness.
The white matter changes may be focal or diffuse,
symmetric or not, and not associated with mass effect or Medical power-of-attorney
enhancement. MRI is more sensitive in detecting Recommend to patient early in the illness.
confounding illnesses such as PML.
Respite and palliative care
CSF Regular respite care can be helpful for the patient and carers
• Cells: mild pleocytosis in 25%. during the illness. Palliative care facilities are usually required
• Protein: elevated in 55% (common CSF findings in non- to manage late complications of dementia.
demented patients also).
• Glucose: normal. CLINICAL COURSE
• Cryptococcal antigen: negative. Progressive, may be insidious or rapid.
• VDRL, TPHA: negative.
• Total immunoglobulin fraction: increased. PROGNOSIS
• Immunoglobulin: anti-HIV IgG. Until recently there was about a 67% cumulative mortality
• Oligoclonal bands: may be present. over 6 months for moderate/severe (stages 2–4: motor
• β2 microglobulin, neopterin and quinolinate levels: abnormalities in addition to psychomotor retardation)
markers of immune activation. β2 microglobulin is a low patients. This is three times the rate for Pneumocystic carinii
molecular weight protein expressed on the surface of pneumonia. However, HAART has substantially improved
nucleated cells (particularly lymphocytes and macro- survival. In addition to clinical status, both viral load (which
phages) and is elevated in the CSF of patients with HIV reflects the amount of viral replication occuring in an
dementia, independent of serum level. CSF β2 micro- infected person) and CD4 cell count (depletion reflects
globulin and neopterin levels correlate with severity of ongoing viral replication and immune compromise) are
dementia. useful prognostic markers.
• Culture.
HIV-Associated Cognitive/Motor Complex (HIV-CMC) 303

357 358

357 CT brain scan of a patient with HIV-associated 358 T2W MRI scan from a 25 year old HIV positive
cognitive/motor complex showing widened cortical sulci, patient. Note the atrophy, which in a young person
enlargement of the lateral ventricles, and diffuse low should raise the possibility of AIDS, and the subtle
attenuation in the periventricular deep white matter. white matter hyperintensities (arrows).

359
359 Algorithm for the management CNS abnormalities1 and HIV infection
of brain lesions in patients with HIV
infection. CT or MRI without and with contrast

Focal enhancing lesions


Focal non-enhancing
Normal or atrophy white matter lesion(s)

Single lesion Multiple lesions Large lesion with mass


Further studies: Consider stereotactic effect and impending
bloods, LP, etc biopsy herniation

Toxoplasma serology Toxoplasma serology

Cryptococcal Decompression
HIV dementia2 PML3 Negative Positive Open biopsy
meningitis,
neurosyphilis

Stereotactic biopsy
Anti-toxo therapy5

Others: Improvement in
Lymphoma4 toxoplasmosis, 2–3 weeks?
cryptococcosis,
tuberculosis
No Yes

1Headache, altered mental state, focal neurologic signs Toxoplasmosis:


2Highly Stereotactic biopsy
active antiretroviral therapy (HAART), clinical trial continue therapy
3Antiretroviral, Ara-C (intravenous, intrathecal), clinical trial
4Radiation therapy
5Pyrimethamine + sulfadiazine or pyrimethamine + clindamycin
Follow-up MRI or
CT scan
CNS = central nervous system PML = progressive multifocal leukocencephalopathy
CT = computed tomography LP = lumbar puncture
MRI = magnetic resonance imaging HIV = human immunodeficiency virus
304 Infections of the Nervous System: Viral

SUBACUTE SCLEROSING • Mutations of the measles virus, resulting in a lack of


PANENCEPHALITIS (SSPE) production of an M (matrix) protein in the measles virus,
which allows the virus to remain relatively protected in
an intracellular form and environment, away from the
DEFINITION extracellular host defence mechanisms, and spread by
A rare slowly progressive disease of the CNS in children and cell-to-cell contact.
young adults that is probably caused by chronic infection
by defective measles virus not expressing its M protein. CLINICAL FEATURES
• History of primary measles infection at a very early age;
EPIDEMIOLOGY often before 2 years.
• Incidence: extremely rare, about 1 case per million • Latent interval of 6–8 years after measles infection, or in
children per year; measles vaccination has greatly reduced rare cases live measles vaccination.
the incidence. Many ethnic groups are affected. • First stage:
• Age: affects children and adolescents; mean age of onset – Cognitive decline: less proficient at school, language
of 7–8 years; rarely beyond 18 years. difficulties.
– Behavioral changes: temper outbursts, loss of interest in
PATHOLOGY usual activities, and other changes in personality.
• Cerebral cortex and white matter of the cerebral • Second stage:
hemispheres and brainstem (360): – Intellectual deterioration: severe and progressive.
– Nerve cell destruction, neuronophagia, and perivenous – Myoclonic jerks; widespread.
cuffing by lymphocytes (361) reflect the viral nature of – Seizures: focal or generalized.
the infection. – Visual disturbance sometimes: due to progressive
– Degeneration of medullated fibers (myelin and axis chorioretinitis.
cylinders) of the white matter, accompanied by • Third stage: neuromotor retardation with spasticity,
perivascular cuffing with mononuclear cells and fibrous rigidity, autonomic dysfunction and unresponsiveness.
gliosis (sclerosing encephalitis). • Final stage: insensate, virtually ‘decorticated’.
– Eosinophilic inclusions in the cytoplasm and nuclei of
neurons and glial cells are the hallmark of the disease DIFFERENTIAL DIAGNOSIS
(362–364). • Lipid storage diseases.
– Virions, thought to be measles nucleocapsids, may be • Schilder disease.
seen with electron microscopy in inclusion-bearing cells. • HIV infection.
• Immunohistochemical studies reveal measles virus • Neurosyphilis.
antigen (365). • Creutzfeldt–Jakob disease (CJD).
• Paired helical filament-tau immunohistochemistry and
electron microscopy reveal early stages of neurofibrillary INVESTIGATIONS
tangle formation in hippocampal neurons. Serum
High titers of measles virus specific IgG antibody; negative
ETIOLOGY AND PATHOPHYSIOLOGY syphilis and HIV serology.
• Measles virus infection acquired pre-natally or in infancy
or early childhood: measles virus infection below 1 year
of age is a risk factor.
• Persistent measles virus infection of the brain.

360 361

360 Section of the brain at autopsy, coronal plane, showing severe 361 Histologic section of brain showing perivenous cuffing by
generalized atrophy of the brain due to subacute sclerosing lymphocytes and destruction of neurons.
panencephalitis.
Subacute Sclerosing Panencephalitis (SSPE) 305

EEG CSF
Characteristic periodic bursts (every 3–10 seconds) of • Cell count: normal.
symmetrically bisynchronous slow and sharp wave • Protein: normal or raised.
complexes (Rodermacker complexes), with an amplitude • Immunoglobulin levels: high.
around 500 μV, and often associated with myoclonic jerks, • Oligoclonal bands of IgG: present (measles virus-specific
appear early in the clinical disease process and may be antibody).
diagnostic (366). The periodic discharges are more • Measles virus antibody titer: extremely high titers of
complicated in outline and separated by longer intervals measles specific IgG.
than in CJD. Unlike other types of periodic phenomena,
they may occur on a fairly normal background in SSPE.

362 363

364 365

362–364 The histologic hallmark of subacute sclerosing 366


panencephalitis: eosinophilic inclusions in the cytoplasm and nuclei of
neurons and glial cells.

365 Brain immunohistocytochemistry showing measles virus antigen.

366 Electroencephalograph, sensitivity 30 μV, of a 6 year old girl with


subacute sclerosing panencephalitis showing periodic discharges taking
the form of double or triple sine waves separated by long intervals and
a rather flat, abnormal background.
306 Infections of the Nervous System: Viral

CT brain scan doses, is said to prolong survival by 1–2 years.


May be normal in the early stages, and at all stages in rapidly • Intraventricular α-interferon, 100 000 U/m2 with daily
progressive disease; may show generalized brain swelling and increments up to 106 U/m2 on the fifth day, to a total
ventricular compression (367). In slowly progressive cases, dose of 30 × 106 U/m2 over a 6 week period,
CT may show marked generalized cerebral atrophy with administered via an Ommaya reservoir located under the
widened cortical sulci and a slightly dilated ventricular scalp, into the frontal horn of the right lateral ventricle,
system, and low density areas in the basal ganglia and in combination with oral inosiplex (isoprinosine) may
periventricular white matter (367, 368). prolong survival.

MRI brain PROGNOSIS


• T2W images are frequently abnormal, unlike CT. • Evolves over some months; steadily progressive.
• Lesions of high signal intensity (bright) are seen on • Death usually occurs within 1–3 years.
T2WI and PDWI in the basal ganglia, periventricular • In about 10% of cases the course is fulminating leading
regions, cerebellum and pons. The periventricular and to death within 3 months, and in another 10% the course
subcortical white matter lesions are most common is quite prolonged, with one or more remissions.
(369). They tend to start in the cortex-subcortical white • Occasional patients recover and may return to school or
matter (370) and progress to involve the periventricular work, but usually with some neurologic deficit.
white matter and lead to diffuse cerebral atrophy.
• Other findings include glial and parenchymal contrast
enhancement, local mass effect of parenchymal lesions, 367 367 CT brain scan
and involvement of the splenic portion of the corpus in a patient with
callosum. biopsy-proven
• Cortical and subcortical lesions have some correlation subacute sclerosing
with the clinical findings but the extent and location of panencephalitis.
the periventricular white matter lesions and cerebral Note the
atrophy do not reflect the neurologic status of many generalized brain
patients. swelling (the
ventricles are small
DIAGNOSIS with effacement of
History of exposure to measles at a very young age, cortical sulci) and
appropriate clinical findings and course, periodic complexes low density area in
on EEG, elevated gamma globulin in CSF and elevated the right frontal
measles antibody titers in serum and CSF are sufficient to region.
make the diagnosis.

TREATMENT
• No specific treatment.
• Inosiplex (isoprinosine) (an antiviral or immuno-
modulator drug): 100 mg/kg/day orally, in 4 hourly

368 369 370

368, 369 CT (368) and MRI (369) brain scans,T2W image, axial plane showing bifrontal and 370 T2W MRI from the same patient as in
right occipital signal abnormality in a patient with subacute sclerosing panencephalitis. 367 a few weeks later.The brain appears
swollen, particularly the gray matter, with loss
of clarity of the gray-white matter junctions.
Progressive Multifocal Leukoencephalopathy (PML) 307

PROGRESSIVE MULTIFOCAL • The occurrence of PML in the absence of cellular


LEUKOENCEPHALOPATHY (PML) immunodeficiency is quite extraordinary. Indeed, only a
small minority of people with impaired cellular immunity
ultimately develop PML suggesting that something more
DEFINITION than the presence of JCV and cellular immunodeficiency
A demyelinating disease of the CNS that results from infection are required for PML to develop.
of oligodendrocytes with JC virus (JCV), a papovavirus.
CLINICAL FEATURES
EPIDEMIOLOGY Subacute onset over weeks.
• Prevalence: PML is uncommon; even among AIDS
patients it only develops in 2–5% of patients. Focal neurologic deficit
• Seropositivity for IgG antibodies against JCV is very • Mono- or hemiparesis (30–50%).
common, being present in about 10% of children aged 1–5 • Visual deficits such as homonymous hemianopia or
years, 50% of children aged 10 years, and 80–90% of middle- quadrantanopia, cortical blindness and visual agnosia
aged adults. Acute infection appears to be asymptomatic. (20–30%).
• Age and gender: before the AIDS epidemic, PML chiefly • Speech defects (20–30%).
affected elderly individuals and men and women equally. • Limb incoordination (10–30%).
Since then, PML has become a disease of the young and • Gait disturbances.
middle-aged populations affected by AIDS and the male • Rapid cognitive decline in conjunction with other focal
to female ratio is now about 5:1. However, it is likely that neurologic deficits (30%).
the changing pattern of HIV infection with increasing • Headache (10%).
numbers of women affected by AIDS as opposed to • Preserved consciousness and without fever.
homosexual men will return this ratio towards parity.
The clinical features in patients with AIDS are not
PATHOLOGY significantly different from those who have other immuno-
Macroscopic suppressive disorders.
• The cardinal feature is demyelination.
• Multiple areas of pronounced demyelination in the white 371
matter of the cerebral hemispheres (371), brainstem and
spinal cord, with frequent involvement of the gray–white
matter junction and cortical gray matter in the most
severe cases.
• A higher incidence of large confluent lesions and marked
perivascular inflammatory infiltrates have been described
in patients with AIDS.

Microscopic
The histopathologic hallmarks are a triad of:
• Multifocal demyelination.
• Hyperchromatic, enlarged oligodendroglial nuclei.
• Enlarged bizarre astrocytes with lobulated hyperchromatic 371 Microscopic low power section of cerebral cortex and
nuclei. subcortical white matter, myelin stain, showing multiple foci of
demyelination (i.e. lack of staining) in a patient with progressive
Large ‘ballooned’ oligodendroglial cells are seen, with multifocal leukoencephalopathy.
nuclear inclusions containing many virions (372). In situ
hybridization techniques for JCV antigen is more sensitive
than light microscopy for detecting virion in infected 372
oligodendrocytes. Electron microscopy reveals the JCV in
the nucleus of oligodendroglial cells.

ETIOLOGY AND PATHOPHYSIOLOGY


• Infection with JCV, a human papillomavirus, in patients
with cellular immunodeficiency, and perhaps some other
unknown factor.
• It is unclear whether PML results from reactivation of
latent JCV within the brain or from transport of virus to
the CNS from other infected organ systems; JCV DNA
has been detected in peripheral blood lymphocytes in the
vast majority of patients with AIDS-associated PML.
• AIDS is the underlying cause of immunodeficiency in about
75% of patients with PML (PML is the initial manifestation
of AIDS in about one-third of these cases); the remainder 372 Histologic section of cerebral white matter showing large
have other disorders such as Hodgkin’s disease, chronic ‘ballooned’ oligodendroglial cells with hyperchromatic, enlarged nuclei
lymphocytic leukemia and lymphosarcoma. and nuclear inclusions which contain many virions.
308 Infections of the Nervous System: Viral

DIFFERENTIAL DIAGNOSIS MRI brain


• HIV dementia: the demyelination of PML may be T2W spin-echo MRI scan is the preferred imaging
radiographically indistinguishable from that of HIV technique as it demonstrates more lesions and the lesions
dementia but PML is clinically associated with focal are seen more prominently.
neurologic signs, dementia is uncommon, and the clinical
course is usually rapid over weeks. Radiographically a MRI
greater proportion of PML lesions involve the subcortical • A focal region of low intensity on T1WI and high
white matter, the T1W images are hypointense (isointense intensity on T2WI anywhere in the brain, but most
in HIV dementia), faint and peripheral enhancement commonly involving the parieto-occipital region.
occurs rarely, and infratentorial lesions occur more • Usually involves white matter but may involve gray matter.
frequently (30% of cases). • May be single or multiple, patchy or confluent.
• Multiple infarcts: systemic lupus erythematosus or HIV- • May have mass effect.
associated granulomatous angiitis. • Usually without enhancement; about 5–10% of cases
• Multiple sclerosis. show contrast-enhancement which is typically faint and
• Cerebral toxoplasmosis. peripheral.
• Neurosyphilis.
• CMV ventriculoencephalitis and other viral opportunistic CSF
infections: typically CMV lesions are located in the CSF is usually normal in the absence of HIV infection but
periventricular white matter and centrum semiovale, and there may be a mild lymphocytic pleocytosis, slight elevation
subependymal enhancement is observed. of CSF protein and the presence of myelin basic protein.
• CNS lymphoma. JCV may be detected in CSF by PCR.
• Multifocal necrotizing leukoencephalopathy with a
predilection for the pons. Other
• Immunosuppression-induced leukoencephalopathy: • EEG: may show focal slowing but is non-diagnostic.
tacrolimus (FK506), cyclosporin, fluorouracil, levamisole. • Serum antibodies to JCV: not helpful in establishing the
• Metachromatic leukodystrophy. diagnosis because more than 80% of the population show
seropositivity to antibodies against JCV by adulthood.
INVESTIGATIONS • Plasma levels of HIV RNA (‘virus load’): critical to assess
Cranial CT and MRI scan the effectiveness of antiretroviral therapy.
Single or multiple confluent, non-contrast-enhancing areas of
hypodensity without mass effect, predominantly located in the DIAGNOSIS
parieto-occipital white matter, but also in the basal ganglia, Despite the characteristic radiologic findings, the only
external capsule and posterior fossa (brainstem and cerebellum), definitive method of diagnosis is brain biopsy, but the
and without mass effect (373–375). The lesions may have a causative virus, the JCV, may be detected in CSF by PCR.
‘scalloped’ appearance as a result of the subcortical arcuate
fibers lying directly beneath the cortex. In about 5–10% of cases
however, PML is confined to the posterior fossa.

373 374 375

373, 374 T2W axial (373) and T1W coronal (374) MRI with contrast showing a 375 T2W MRI of the brain showing
non-enhancing area on T1W image, high intensity on T2W image in the parietal region biopsy-proven progressive multifocal
typical of PML. (Courtesy of Professor J Best, Department of Medical Radiology, Royal leukoencephalopathy in an HIV-infected
Infirmary, Edinburgh, UK.) patient. Note the T2W high signal
abnormalities involving the parietal subcortical
white matter and sparing the cerebral cortex.
Poliomyelitis (Infantile Paralysis) 309

TREATMENT POLIOMYELITIS (INFANTILE


HAART, with its use of multiple antiretroviral drugs PARALYSIS)
including various combinations of nucleoside RT inhibitors,
non-nucleoside RT inhibitors, and PI antiretrovirals has
become the standard practice for HIV treatment. DEFINITION
The HAART regimen is guided by viral load Infection of the spinal cord and brain by the poliovirus.
measurements, with the goal of achieving non-detectable
HIV RNA viral loads in the patient’s plasma. EPIDEMIOLOGY
These practices have resulted in dramatic improvement Distribution and incidence: polio had a worldwide
in life expectancy of HIV patients. Intravenous and distribution prior to vaccination. Epidemics occurred during
intrathecal cytarabine (Ara-C) therapy does not appear to the summer months and were more frequent in temperate
offer any survival advantage over best antiretroviral therapy. climates. The incidence was greater in areas of poor sanitation.
Following the development of the Salk trivalent inactivated
CLINICAL COURSE AND PROGNOSIS vaccine (1955) and the Sabin trivalent oral live attenuated
HIV-associated PML: median survival of 2–4 months vaccine (1961) and introduction of mass vaccination
previously. However, cohorts treated with HAART have programmes in the late 1950s and early 1960s, the incidence
experienced a median survival of more than 46 weeks. of paralytic poliomyelitis has declined dramatically in countries
About 10% of patients have a more benign course, with with these programmes. In England and Wales there were 21
remission and prolonged survival, or even spontaneous cases of paralytic poliomyelitis between 1985–91. The
recovery on rare occasions. majority (13) were vaccine-associated, of whom nine occurred
in the recipient; three occurred in previously healthy
unimmunized adolescent or adult contacts of infants who had
received their first vaccination, two cases were infants with
unsuspected immune deficiency and five were imported and
the source of infection was unknown.
The remaining principal reservoirs of the wild poliovirus
are West and Central Africa and South Asia, which result in
nearly 100 000 new paralytic cases per year.
• Prevalence: the World Health Organization (WHO)
estimates there are 12 million people worldwide with
some degree of disability caused by poliomyelitis.
• Lifetime prevalence: 0.7 (0.4–1.0) per 1000.
• Age: any age.
• Gender: either sex.

PATHOLOGY
Site
• Neurons in the anterior, intermediate, intermediolateral
and posterior horns of the spinal cord are most
commonly affected.
• The precentral gyrus, globus pallidus, thalamus,
hypothalamus, brainstem nuclei (particularly the nucleus
ambiguus, V, VII, XII and vestibular nuclei) and
brainstem reticular formation, cerebellar vermis, and
dorsal root ganglia may also be affected.

Microscopic findings
• Early: neuronal chromatolysis, particularly affecting the
cytoplasmic Nissl substance, and a perivascular
inflammatory cell infiltrate.
• Later: the nucleus disintegrates, followed by necrosis and
lysis of the whole cell.

ETIOLOGY AND PATHOPHYSIOLOGY


Poliovirus infection
A neurotrophic enterovirus of high infectivity:
• Three subtypes; prior to vaccination, type I caused 85%
of cases of paralytic polio.
• Route of infection: oral–oral and fecal–oral.
• Viruses multiply in the pharynx and intestine during the
1–3 week incubation period before blood-borne dis-
semination occurs. The virus continues to be excreted in
saliva for 2–3 days, and in feces for a further 2–3 weeks.
(Continued overleaf)
310 Infections of the Nervous System: Viral

Poliovirus infection (continued) Chronic poliomyelitis


• In countries with mass vaccination programmes, about • Motor neuron disease.
half of cases are caused by the live vaccine virus in adult • Spinal muscular atrophy.
contacts of vaccinated infants, one-quarter occur in the
vaccine recipient, and the remaining quarter are thought INVESTIGATIONS
to be due to wild virus infection in non-immunized
people. Swabs
• Nasopharynx swab: virus may be isolated for 5 days after
Possible risk factors for development of onset of symptoms.
paralytic disease • Stool swab: virus may be isolated for 5 weeks after onset
Physical activity and intramuscular injections during the of symptoms.
prodrome.
Blood viral serology
CLINICAL FEATURES Following type 1 poliovirus there is an acute rise in serum IgM
Non-paralytic or pre-paralytic poliomyelitis titer with IgG antibody developing over about 3 months.
Asymptomatic or a self-limited ‘flu-like’ illness characterized
by fever, pharyngitis, myalgia, nausea, anorexia, vomiting, CSF
headache and neck stiffness due to meningitis, which Pleocytosis (neutrophils in the first few days then
subsides within 1 or 2 weeks: 95% of all infections. lymphocytes), increased protein, normal glucose, virus rarely
isolated by PCR. If the virus is isolated, it is serotyped using
Acute anterior horn cell poliomyelitis stain-specific antisera, to differentiate wild-type from
Spinal form (>60% of cases) vaccine-induced disease.
• Severe muscle pain and spasms.
• Fasciculations and asymmetric weakness of lower limbs EMG
more than upper limbs, maximal within 48 hours, • Early: reduced recruitment and interference pattern with
particularly in children. Further weakness may occur after voluntary muscle activation.
a short period of stability if fever is still present, but • Later: at rest: electrically silent, fibrillations develop in
generally paralysis stops progressing within 5–7 days. 2–4 weeks and persist indefinitely; fasciculations may also
• Muscle tone: flaccid. be seen during voluntary activation: motor unit action
• Reflexes initially brisk, then become absent. potentials are reduced in number but return during
• No objective sensory loss, but paresthesiae are common. recovery and then become abnormally large in amplitude
and of increased duration and polyphasic, due to
Bulbar form (20% of cases) reinnervation.
• May occur in children, adults tend to have spinal as well • Motor conduction velocities: remain normal.
as bulbar involvement.
• Most commonly the lower cranial nerve nuclei are DIAGNOSIS
affected. Asymmetric purely motor flaccid paralysis with an aseptic
• Dysphagia. meningitis; virus isolation from nasopharynx or stool.
• Dysphonia.
• Tongue wasting and fasciculation (376).
• Respiratory failure.
• Vasomotor disturbances (hypo- and hypertension,
circulatory collapse).

Encephalitic form (rare)


• Agitation, confusion, stupor and coma.
• Autonomic dysfunction is common. 376
• High mortality rate.

DIFFERENTIAL DIAGNOSIS
Acute poliomyelitis
• Other enterovirus infection:
– Coxsackie A and B.
– Echovirus.
– Enterovirus 70 and 71.
• Guillain–Barré syndrome: acellular CSF, prolonged nerve
conduction velocities.
• Acute intermittent porphyria.
• HIV.
• Diphtheria.
• Lyme disease.
• Triorthocresylphosphate poisoning.
• Botulism. 376 Tongue wasting and fasciculation due to necrosis of neurons in
the hypoglossal (XIIth cranial) nerve nucleus.
Poliomyelitis (Infantile Paralysis) 311

TREATMENT PROGNOSIS
Acute attack Clinical course
• Nurse in isolation; cases are most infectious 7–10 days • After the initial insult, paralysis remains stable for several
before and after the onset of symptoms. days or weeks before a slow recovery occurs over several
• Observe vital capacity, cardiovascular responses and other months or years, caused by sprouting and re-innervation
bulbar functions. of muscle by surviving motor neurons, before stabilizing.
• Relieve pain. • About 10% of patients with bulbar paralysis die, usually
• Prevent joint contractures and ankylosis: frequent passive from vasomotor disturbances or respiratory failure.
movements, physiotherapy.
• Negative pressure ventilation (iron lung) which allows Post-polio progressive muscular atrophy
concurrent physiotherapy and postural drainage. Slowly progressive, painless, weakness and wasting of
• Tracheostomy in severe bulbar weakness to protect the muscles that were often severely affected by the original
airway; under these circumstances intermittent positive disease, after at least 10–15 years, is a rare, late complication
pressure ventilation is preferable during the acute illness. of polio.

Prevention Post-polio syndrome


Vaccination Progressive muscle pain, weakness, wasting and fatigue
• Salk trivalent inactivated polio vaccine is administered by occur in about 60% of polio survivors but they are non-
injection and stimulates serum IgM, IgG, and IgA but specific symptoms, particularly in an elderly population, and
not secretory IgA, immunity being induced by antibody are not due to progressive neuromuscular failure. In two-
transuding into the oropharynx. It is highly effective and thirds of symptomatic patients, an underlying cause that is
remains indicated in pregnant, immunosuppressed and unrelated to earlier polio can be readily identified, most
unvaccinated individuals over the age of 50 years. commonly degenerative joint disease, but also diabetes,
• Sabin trivalent oral live attenuated polio vaccine is taken excessive weight gain, alcoholism, depression, acute lumbar
orally and causes an active attenuated infection of the disc herniation and radiculopathy and motor vehicle trauma.
oropharynx and intestinal endothelium leading to local In the other one-third, in whom a ready explanation is not
secretory IgA in addition to serum antibody production. apparent, the cause remains unknown but the symptoms are
Furthermore, the attenuated virus is excreted in the feces similar to patients with chronic fatigue syndrome (a disorder
producing herd immunity. often associated with depression) and tension myalgia/fibro-
• Individuals born before 1958 may not have been myalgia (a disorder that is common in middle-aged,
immunized and should be. deconditioned people that is not usually associated with
• Oral polio vaccine is recommended for infants from depression but in which muscle pain, usually at tendon
2 months of age. The primary course consists of three insertion, is worsened with, and may persist after, use or
separate doses with intervals of 1 month between doses. overuse). In neither is there any evidence of intrinsic nerve
• Unimmunized contacts of recently immunized infants or muscle disease.
are at particular risk of infection when changing nappies The chronic use of weak muscles or muscles acting in a
and must be vaccinated. compensatory fashion may well predispose people to this
type of problem, If this is the cause of the post-polio
In 1988, the WHO declared its commitment to the syndrome, it leads directly to therapeutic strategies. Patients
complete eradication of acute paralytic poliomyelitis by the with fibromyalgia can be effectively treated with
year 2000. This has been achieved in most Western conditioning, physical medicine/rehabilitation approaches,
countries using strategies relying on: and tricyclic antidepressants. There is no basis for the use of
• Mass vaccination campaigns using live attenuated oral drugs designed to improve neuromuscular function because
polio vaccine in all children under the age of 5 years. there is no evidence of progressive neuromuscular
• Enhanced surveillance. deterioration.
• Targeting immunization to areas and populations where
poliovirus transmission is likely to persist.

However, wild-type poliomyelitis continues to occur in


regions with high rates of endemic poliomyelitis and, rarely,
in outbreaks in areas believed to be free of disease.
312 Infections of the Nervous System: Other

OTHER INFECTIONS: SPIROCHETAL, FUNGAL,


PROTOZOAN, WORM AND PRION

NEUROSYPHILIS • General paralysis of the insane develops about


15–20 years after the primary infection. It is due to
widespread spirochetal invasion of the brain and chronic
DEFINITION meningovascular syphilis.
Syphilis is an infectious disease caused by Treponema
pallidum, which is a natural pathogen only in humans. Four CLINICAL FEATURES
stages can be distinguished in the natural history of syphilis: Clinically, neurosyphilis can be classified into well defined
primary, secondary or disseminated, latent and tertiary syndromes, representing different clinical expressions of the
syphilis. Neurosyphilis is one of the tertiary forms of syphilis. same disease.

EPIDEMIOLOGY Asymptomatic neurosyphilis


• Incidence: the incidence of syphilis is rising in the United • The manifestations of primary or secondary syphilis (e.g.
States, mainly due to an increase in the number of cases skin rash [379–382]) have resolved, and there are no
of cocaine/crack addiction. neurologic gray signs.
• Prevalence: in Florida, USA, the prevalence of syphilis in • The CSF is abnormal, such as positive serology, an
1987 was 66 per 100 000 persons, particularly among increased cell count and, occasionally, an increased
homosexual men. Neurosyphilis occurs in less than 3% protein content.
of all syphilis cases due to administration of antibiotics in
the first stages of the disease.
• Age: adults of any age. 377
• Gender: M>F.

PATHOLOGY
Meningovascular syphilis
• Diffuse thickening and lymphocytic infiltration of the
leptomeninges and perivascular spaces.
• Vasculitis (endarteritis obliterans) of terminal arterioles,
which show concentric proliferative thickening of the
endothelium as well as appearance of fibroblasts (377).
The most commonly involved artery is the MCA.

Tabes dorsalis
Degeneration with neuronal loss in the dorsal root entry
zones of the spinal cord and fibers in the dorsal columns
(378).
377 Cross section of a branch of the middle cerebral artery showing
ETIOLOGY AND PATHOPHYSIOLOGY Intimal hyperplasia due to meningovascular syphilis.
• Acquired infection, mainly by sexual intercourse with an
infected individual but also by vertical transmission at 378
delivery, by laboratory accidents or blood transfusions.
• If the CNS is to be invaded, this usually occurs within
2 years after primary infection.

Stages of neurosyphilis
• Acute syphilitic meningitis usually occurs within the first
2 years of the primary infection. May involve the brain
or the spinal cord.
• Meningovascular syphilis may occur any time from the
onset of the secondary syphilis rash to 10 years after the
primary infection but generally does not occur until
4–7 years after the primary infection. It is due to
granulomatous syphilitic infiltration and vasculitis around
the brainstem, spinal cord and spinal roots.
• Tabes dorsalis develops 20 years or so after the primary
infection. Degeneration of dorsal roots and fibers in 378 Transverse section of the spinal cord in a patient with tabes
dorsal columns interrupt the stretch reflex arc to cause dorsalis showing degeneration of the posterior columns (arrows)
hypotonia and loss of reflexes, and lightning (stabbing) secondary to Treponema pallidum infection and inflammation along the
pains mainly in the legs. dorsal roots. (Courtesy of Professor BA Kakulas, Royal Perth Hospital,
Western Australia.)
Neurosyphilis 313

Acute syphilitic meningitis Meningovascular syphilis


• Symptoms include headache, nausea and vomiting, neck Generally presents with a prodromic phase, weeks or
stiffness, seizures and changes in mental status; patients months before the onset of identifiable vascular syndromes.
are often afebrile. Specific clinical features include:
• Ocular or cranial nerve abnormalities, especially VII and • TIA or stroke-like syndrome (if focal inflammation of
VIII, may occur due to involvement at the base of the intracranial arteries).
brain. • Slowly progressive cognitive decline and personality changes
• Some patients can develop leptomeningeal granulomas, (if multifocal involvement of small intracranial arteries).
hydrocephalus or neuritis. • Epileptic seizures.
• Argyll Robertson pupils.
(Continued overleaf)

379, 380 Skin rash on the 379 380


axilla and trunk of a patient in
the initial stages of secondary
syphilis.The lesions are
bilaterally symmetric, pale
red/pink, non-pruritic,
discrete, round macules,
about 5–10 mm (0.2–0.4 in)
in diameter, distributed on
the trunk and proximal
extremities. After 1–2
months, red, papular lesions
3–10 mm (0.1–0.4 in) in
diameter also appear on the
palms, soles, face and scalp.

381, 382 Skin rash on the 381 382


legs and foot soles of a
patient in the later stages of
secondary syphilis.The lesions
are red/copper-colored,
papular and papulosquamous,
about 3–10 mm (0.1–0.4 in)
in diameter and were also
present on the palms, face
and scalp.
314 Infections of the Nervous System: Spirochete

Meningovascular syphilis (continued) to accommodation but not to light and dilate poorly with
• Optic neuritis, optic atrophy and chorioretinitis. a mydriatic. Iris atrophy may be present. They were
• Deafness: acute and subacute, unilateral at first. described by Douglas Argyll Robertson, an Edinburgh
• Other cranial neuropathies: acute and subacute. physician, and are probably due to a lesion in the pretectal
• Meningitis: acute, subacute and chronic. periaqueductal gray matter or ciliary ganglia. They are
• Spinal cord and/or nerve root syndromes (spinal cord almost pathognomonic of meningovascular syphilis, tabes
compression, infarction and transverse myelitis): acute dorsalis and general paralysis of the insane.
and subacute. Other causes of AR pupils include:
• Diabetic autonomic neuropathy.
Usually there is no fever. • Amyloid neuropathy.
• Hereditary motor and sensory (hypertrophic)
Tabes dorsalis polyneuropathy type III.
Symptoms • Late Holmes–Adie pupil.
• Lightning (stabbing) pains mainly in the legs and back. • Upper dorsal mid-brain lesions, usually a pineal region
• Abdominal pains and vomiting. tumor, causing Parinaud’s syndrome (failure of upward
• Loss of deep pain sensation in the legs. gaze, bilateral pupil dilatation with a poor response to
• Patch loss of superficial pain sensation in the face and light and often to accommodation as well, sometimes
limbs. accompanied by convergence-retraction nystagmus on
attempted upgaze) give rise to pseudo-AR pupils, which
Signs do not react to light but are neither small nor irregular.
• Ptosis. • Iris trauma.
• Argyll Robertson pupils in about 90% of cases.
• Optic atrophy. Positive non-specific or specific syphilis serology
• Sensorineural deafness. (VDRL, RPR, TPHA and FTA)
• Hypotonia and loss of deep tendon reflexes in the legs, • Yaws.
and sometimes arms. • Mycoplasma pneumoniae.
• Flexor plantar responses, but sometimes extensor plantar • Malaria.
responses. • Acute bacterial and viral infections.
• There is no primary motor involvement. • Smallpox vaccination.
• Patchy loss of sensation to pain over the bridge of the • Addiction: e.g. narcotics.
nose, sternum, and tibial prominences. • Autoimmune disease: e.g. systemic lupus erythematosus,
• Impaired position and vibration sense. anticardiolipin antibody syndrome.
• Painless destruction of weight-bearing joints (Charcot • Ageing.
joints) in the lower limbs in about 10% of patients due
to loss of deep pain sensation. INVESTIGATIONS
• Trophic foot ulcers. Blood serology
• High stepping unsteady gait due to proprioceptive loss Non-treponemal antibody
in the lower limbs Rapid plasma reagin (RPR) and Venereal Disease Research
• Painless urinary retention/incontinence is common. Laboratory slide flocculation test (VDRL): subject to false
• Impotence. negative reactions (e.g. VDRL in early infection, RPR in late
• Postural hypotension. disease and both in HIV infection; sensitivity varies from
30–70%) and false-positive reactions in certain clinical
General paralysis of the insane situations (e.g. pregnancy, autoimmune disease, i.v. drug use
Clinical features reflect widespread and diffuse parenchymal and HIV infection).
damage and include:
• Personality and affect changes. Specific treponemal antibody
• Progressive dementia: impaired recent memory, • Treponema pallidum hemagglutination test (TPHA).
disorientation, dyscalculia, impaired judgement. • Fluorescent treponemal antibody test (FTA).
• Sensorium abnormalities including illusions, delusions • Treponema pallidum immobilization test (TPI).
and hallucinations. • Enzyme immunoassay for treponemal antibodies.
• Dysarthria.
• Tremor. Syphilis cannot be differentiated serologically from yaws,
• Epileptic seizures. a spirochetal disease which is endemic in the Caribbean.
• Pyramidal tract signs.
• Argyll Robertson pupils. CT/MRI brain scan
• Multifocal areas of low density are seen on CT and
DIFFERENTIAL DIAGNOSIS increased signal on T2W MRI in the brain parenchyma,
Charcot joints consistent with infarction, due to meningovascular
May be seen in other neurologic conditions in which central syphilis.
or peripheral pain pathways are damaged: syringomyelia, • Widespread thickening of the meninges or extra-axial
diabetes, hereditary sensory neuropathy. enhancement suggestive of meningitis.
• Various enhancing parenchymal nodules (cerebral
Argyll Robertson (AR) pupils gumma).
AR pupils are small, irregular, unequal pupils that constrict
Neurosyphilis 315

CSF Herxheimer reaction prophylaxis


In active neurosyphilis, the CSF is characterized by: Oral prednisolone, 40 mg daily, for 1 day before and 3 days
• Lymphocytosis: up to 1000 cells/mm3. after beginning treatment.
• Protein: increased moderately.
• Glucose: normal or reduced. Lightning pains
• Specific antibody serology (FTA, TPHA): positive. Carbamazepine may be helpful.
• Non-specific VDRL/RPR: negative in up to 50% of
cases. FOLLOW-UP
• Oligoclonal bands: commonly present. • The CSF must be re-examined at 3 months, 6 months,
• PCR: amplifies specific T. pallidum DNA in the CSF or 1 year and 2 years after treatment is commenced. If the
the blood, which is then detected by agarose gel cell count remains elevated, then treatment should be
electrophoresis or by hybridization with probes. It repeated until it returns to normal.
detects the actual presence of the organism rather than • The CSF protein level may not return to normal until
the immune response of the host to the organism. 1–2 years after successful treatment.
• The blood and CSF serology often remains positive for
If the CSF cell count and protein are normal, then the life, although the VDRL titers should fall.
patient’s disease is ‘burnt out’ or the diagnosis of
neurosyphilis is incorrect. PROGNOSIS
It is unnecessary to examine the CSF if: Antimicrobial therapy can cure meningovascular syphilis and
• The clinical picture is not compatible with neurosyphilis arrest tabes dorsalis and GPI, but lightning pains and fixed
and is clearly due to a non-syphilitic cause. neurologic deficits are likely to remain.
• The patient has had effective treatment for syphilis in the
past. SPECIAL FORMS
• The blood serologic titers are low. Concurrent HIV infection
• Neurosyphilis may present at any time during the course
Cerebral angiography of HIV infection. Considerable overlap is present
Concentric or asymmetric constriction and occlusion of the between the spectrum of neurologic diseases caused by
intracranial portion of the internal carotid, proximal middle HIV and Treponema pallidum. Both may result in
and anterior cerebral and basilar arteries, and narrowing dementia, cerebrovascular disease, chronic meningitis,
with aneurysmal dilatation of small vessels are seen in myelopathy and cranial and peripheral neuropathies; and
meningovascular syphilis. both may produce similar CSF abnormalities, particularly
persistent pleocytosis. In addition, atypical presentations
DIAGNOSIS may occur with concurrent HIV infection.
• Suggestive clinical picture. • Patients without clear documentation of adequate
• Positive treponemal serology: fluorescent treponemal therapy for past syphilis should be assessed for
antibody absorption (FTA-ABS) (serologic evidence of neurosyphilis by LP. A positive CSF VDRL test is
exposure to T. pallidum). diagnostic of neurosyphilis but this assay has a sensitivity
• One of the following CSF abnormalities: of only 30–70%.
– White blood cell pleocytosis of >5 leukocytes/mm3. • Concurrent HIV infection may alter the natural history of
– CSF protein >0.45 g/l (at least 450 mg/dl protein). syphilis (e.g. fulminant, necrotizing encephalitis may occur),
– Positive CSF VDRL/RPR. the yield of tests (serologic tests are less sensitive in
• Positive PCR: blood or CSF. diagnosing late syphilis in HIV infection) and response to
conventional therapy (e.g. neurosyphilis may relapse in HIV-
TREATMENT infected patients after therapy with benzathine penicillin).
Since neurologic deficits, once established, may only slightly Consequently, neurosyphilis should be treated aggressively
improve with treatment, the goal of therapy is to halt the in patients with HIV infection and closely followed-up.
progression of the disease. • Penicillin-based regimens are preferred; procaine
penicillin 1.5 g daily i.m. plus probenecid (to increase
Active disease (raised CSF lymphocytes or protein) serum and CSF levels of penicillin) 500 mg four times a
• Aqueous crystalline penicillin G (benzylpenicillin) day by mouth for 21 days, or penicillin G 2.4 g every
12–24 million units (600 mg), i.v. daily for 10–14 days 4 hours by i.v. injection plus probenecid 500 mg four
followed by either further penicillin G, i.v. or daily times a day by mouth for 10 days. Ceftriaxone (1–2 g for
procaine penicillin, i.m. or weekly benzathine penicillin 14 days) is an alternative regimen, although experience
2.4 million units, i.m. for a total of 4 weeks; or, if is limited.
hospital admission undesired: procaine penicillin, 600 mg • Serum and CSF should be followed for at least 2 years to
once daily, i.m., for 21 days. monitor response to therapy. The goal is normalization
• If minor penicillin allergy occurs, cefotaxime or of CSF values and VDRL titers but the presence of
ceftriaxone can be prescribed. concurrent HIV infection may cause persistent CSF
• If more severe penicillin sensitivity occurs: tetracycline abnormalities.
(500 mg oral 6-hourly for 30 days) or erythromycin • If a patient with HIV infection has symptoms consistent
(500 mg oral 6-hourly for 30 days) can be given. with neurosyphilis, a positive serum VDRL, and a
reactive CSF profile, even in the absence of a positive
CSF VDRL, then treatment with intravenous high-dose
penicillin is probably indicated.
316 Infections of the Nervous System: Spirochete

LYME DISEASE • Mononeuritis multiplex.


(NEUROBORRELIOSIS) • Neuralgic amyotrophy (see p.595).
• Myositis.

DEFINITION Stage 3: Chronic stage


A multisystem non-venereal treponemal disease caused by a Many months, and even a year or more, after the initial
spirochete of the genus Borrelia, called Borrelia burdorferi, infection, other neurologic complications may occur:
and named after the town of Lyme, Connecticut, where a • Encephalopathy can present as severe encephalomyelitis
cluster of cases was first recognized in 1975. complicated by seizures or can be more mild, with mild
memory and mood disturbances, word letter reversals,
EPIDEMIOLOGY and spatial disorientation. Major depression, anxiety,
A worldwide zoonosis and currently the most common panic attacks and catatonia have been reported.
vector-borne illness in the United States. • Peripheral neuropathy is primarily axonal in nature,
• Incidence: 3.9 per 100 000 in 1992 in the United States includes both motor and sensory fibers, and can present
(a 19-fold increase since 1982). Reforestation and an as a radiculopathy, plexopathy, distal neuropathy, or
increasing deer population are believed to contribute to mononeuritis multiplex.
the increasing number of cases. The disease usually • Arthritis may not occur until 6 months after the onset of
appears in the summer in endemic areas: in the USA Lyme disease.
where it is known as Lyme disease, and in Europe where • Post borrelia syndrome: some patients treated for Lyme
it is called Bannwarth’s syndrome. disease continue to experience symptoms of fatigue,
• Age and gender: any. fibromyalgia, insomnia, or psychiatric disorder. The
etiology is uncertain.
ETIOLOGY
Caused by B. burgdorferi, a spirochete carried primarily by DIFFERENTIAL DIAGNOSIS
the hard-shelled deer ticks: Ixodes dammini, I. pacificus, and • Viral encephalitis (see p. 292).
I.ricinus in the Eastern United States, Western United • Bell’s palsy.
States, and Europe respectively. The ticks infest deer and • Chronic meningitis (see p.279).
small mammals, and are transmitted to man by tick bite. • Guillain–Barré syndrome (no CSF pleocytosis).

PATHOPHYSIOLOGY
Following intradermal inoculation, B. burgdorferi
proliferates within the dermis and then invades the blood
stream to cause a spirochetemia. After a few weeks it directly
invades the meninges to cause a basilar meningitis. It may
also cause an encephalopathy by invading the brain
parenchyma or by stimulating an immune response with the
production of quinolinic acid, a CNS excitotoxin.

CLINICAL FEATURES
Stage 1: Early, localized disease stage 383
Within a few days of tick bite, an erythematous papule
appears at the site of the bite and this expands into an
annular lesion with a partially clearing center (erythema
chronicum migrans), sometimes with multiple secondary
skin lesions and often accompanied by headache, myalgia,
general malaise, and lymphadenopathy. Sometimes the
illness is more indolent; some patients do not recall an insect
bite, and there may be no preceding skin lesion nor very
much systemic upset initially.

Stage 2: Early disseminated disease stage


A few weeks to a few months after infection there is often
resolution of earlier symptoms and evolution of arthritis
and/or the subacute onset of a chronic basilar lymphocytic
meningitis with or without encephalopathy, cranial neuritis and
radiculoneuropathy. The neurologic complications include:
• Meningitis (headache, nausea, photophobia).
• Subacute or chronic facial palsy: usually bilateral and
complete but may be unilateral and incomplete (383).
• Other cranial nerve palsies (II, V).
• Encephalitis: mild, subacute or chronic.
• Transverse myelitis.
• Polyradiculopathy with severe root pain, sensory and 383 Right lower motor neuron facial nerve palsy in a patient with
motor loss. neuroborreliosis.
Lyme Disease (Neuroborreliosis) 317

INVESTIGATIONS • Other tests, such as the immunocytochemical antigen


Blood binding test and the cell-mediated immunity response
• Peripheral white cell count: normal or lymphopenic. test, are available but not recommended for laboratory
• ESR: may be raised. diagnosis.
• Serology: serum titers of IgG antibodies to B. • In patients with neurologic complications that occur very
burgdorferi: elevated. However, a raised serum IgG does early in the course of infection, before a strong immune
not necessarily indicate active infection and the serum response develops (e.g. facial nerve palsy), an ELISA is
IgM may no longer be raised at the time that neurologic obtained at the time of presentation and if positive, the
complications occur (see below). CSF is examined for confirmation of the diagnosis. If the
ELISA is negative, the ELISA is repeated in 1 month and
Imaging the CSF is repeated if seroconversion occurs. If the CSF
The following imaging findings may occur: findings support the diagnosis, intravenous antibiotics
• A normal CT and MRI brain scan. are commenced, and if the CSF is normal, oral
• Areas of increased signal on T2WI/PDWI varying in size doxycycline therapy is started (see below).
from small punctate to large mass lesions, with a
predilection for the frontal and parietal lobes. PCR
• Contrast-enhancing parenchymal lesions, meninges and PCR is now available as a diagnostic test. The test amplifies
cranial nerves. specific B. burgdorferi DNA in the blood or CSF, which is
• Lyme disease should be considered in the imaging then detected by agarose gel electrophoresis or by
differential diagnosis of multiple sclerosis, ADEM and hybridization with probes. It detects the actual presence of
vasculitis. an organism rather than the host’s immune response to the
organism. However, it has a high false negative rate of
CSF 30–50%, perhaps because the organism prefers tissue over
• Cell count: raised, lymphocytic (in acute Lyme disease body fluids.
but uncommonly in chronic Lyme disease).
• Protein: raised. TREATMENT
• Glucose: normal glucose. Treat early and promptly.
• IgG/albumin ratio: raised.
• Oligoclonal bands: present. Cranial neuropathy (usually VIIth) with normal CSF
• Cytology: a lymphoplasmacytic pleocytosis with frequent Doxycycline and amoxycillin (amoxicillin) (especially in
mitotic figures may occur, simulating leptomeningeal children) with or without probenecid, orally, for 2–4 weeks.
lymphoma.
• Serology: borrelia antigens are sometimes, albeit rarely, Other neurologic complications
detected in CSF when serum tests are negative and when • Penicillin G, i.v. 200 mg/kg/day in 6-hourly doses for
CSF is otherwise normal. So, the diagnosis should not 14 days.
be excluded solely on the basis of normal routine CSF or • Tetracycline, oral, 500 mg four times daily for 30 days.
negative CSF antibody analyses. Exceptionally, the CSF • Alternatives to penicillin include intravenous ceftriaxone,
is normal in neuroborreliosis. cefotaxime, or doxycycline; or oral chloramphenicol.

DIAGNOSIS
Prompt and precise diagnosis is difficult because basic
microbiologic tests such as staining and culture of this
fastidious organism remain low yield procedures.

Serologic testing
• Detection of specific antibodies to B. burgdorferi in
blood and CSF by means of ELISA and the indirect
fluorescent antibody test, is commonly used to support
or refute the diagnosis but the tests are not well
standardized, false positive results are not uncommon
(particularly in patients with other infectious and
inflammatory diseases), a true positive result is evidence
of exposure and not necessarily active infection, a (false)
negative result may be present initially in cases with
eventually proven neuroborreliosis, and the test may
remain positive for years, despite clinical resolution after
therapy.
• A positive ELISA or indirect immunofluorescent
antibody result should be confirmed with the Western
blot assay but this test is laborious, expensive, and its
criteria for positivity have not been adequately
standardized. Quantitative criteria using a gel
densitometric approach have been recently devised to
help standardize this procedure.
318 Infections of the Nervous System: Fungal

CRYPTOCOCCAL MENINGITIS • Seizures and focal neurologic signs suggest the presence
of complications such as a granulomatous mass lesion in
one part of the brain (a cryptococcoma), granulomatous
DEFINITION meningovasculitis, or venous sinus thrombophlebitis.
Infection of the leptomeninges caused by the yeast • Confusion and impaired consciousness tend to develop
Cryptococcus neoformans. late and in severe cases.
• Extraneural manifestations, which may occur with or
EPIDEMIOLOGY without meningitis, include pulmonary infiltrates, skin
Cryptococcosis (formerly called torulosis) is one of the most lesions (abscesses, ulcers, or molluscum contagiosum-like
common fungal infections of the CNS and cause of lesions), prostatic abscess, hepatitis and lytic bone lesions.
meningitis, particularly in patients with HIV infection,
affecting about 10% of AIDS patients, usually late in the DIFFERENTIAL DIAGNOSIS
course. It is most common in Thailand and Central Africa. Meningitis
• Aseptic (normal CSF glucose levels; early HIV).
PATHOLOGY • Syphilis (see p.312).
• Granulomatous meningitis. • Listeria.
• Small granulomas and cysts within the cerebral cortex. • Tuberculosis (distinguished by fever, pulmonary lesions,
The cortical cysts contain large numbers of organisms in hyponatremia due to SIADH, and organisms in the CSF;
a gelatinous material. The solid granulomatous nodules late HIV; see p.281).
are composed of fibroblasts, giant cells, organisms, and • CNS vasculitis (normal CSF glucose levels, see p.227).
regions of necrosis. • Malignant meningitis (carcinomatous, lymphomatous;
• Large granulomas and cystic nodules sometimes form see Metastases to the CNS, p.396).
deep in the brain. • Other causes of subacute or chronic meningitis (see
p.279).
ETIOLOGY AND PATHOPHYSIOLOGY
• Cryptococcus neoformans is the etiologic agent. It is a Other uncommon CNS infections in AIDS
common soil fungus found in the roosting sites of birds, • Candida albicans infection.
particularly pigeons. The portal of entry is usually the • Coccidioidomycosis.
respiratory tract, and less often skin and mucous • Histoplasmosis.
membranes. • Mycobacterium tuberculosis infection.
• It is usually acquired in the community. • Herpes zoster infection.
• It may cause minimal inflammation in people with AIDS • Herpes simplex infection.
and impaired immune responses. • JC papovavirus infection (progressive multifocal
leukoencephalopathy).
Risk factors • Aspergillosis.
A debilitating disease that alters T lymphocyte-mediated • Amebiasis.
immune responses: • Trypanosoma cruzi infection.
• AIDS due to HIV infection. • Nocardia.
• Transplantation recipients.
• Lymphoma. CNS mass lesion
• Hodgkin’s disease. • Brain tumor.
• Leukemia. • Brain abscess.
• Carcinoma. • Brain granuloma.
• Tuberculosis.
INVESTIGATIONS
CLINICAL FEATURES CT or, preferably, MRI of the brain
• Cryptococcosis is a respiratory infection in patients who Findings include:
usually present with disseminated disease, most • Normal scan.
commonly as meningitis. • Dilated Virchow–Robin spaces which may enhance (in a
• Presenting features are often subacute in onset, subtle young immunosuppressed patient this should raise the
and non-specific; indeed, in the setting of HIV infection, possibility of cryptococcus infection).
cryptococcal meningitis can be truly cryptic with little or • Hydrocephalus (rare) (384–387).
no neurologic dysfunction. The classic symptoms signs • Multiple miliary enhancing nodules in the parenchyma
of meningitis such as neck stiffness and photophobia, are and leptomeninges involving the choroid plexus of the
often absent. trigone, cranial nerves and spinal nerve roots. Contrast
• Fever, headache, malaise, nausea and vomiting, and is required to demonstrate the miliary nodules.
mental changes are common but can be mild. • Diffuse meningeal enhancement rarely (differentiating it
• Some patients do not have fever, headache and stiff neck, from tuberculous and bacterial meningitis) (388–390).
but present with symptoms of gradually increasing • Multiple small areas of increased signal in the basal
intracranial pressure due to hydrocephalus, or with a ganglia that do not enhance are characteristic, but not
confusional state, dementia, cerebellar ataxia or spastic pathognomonic, of cryptococcus, and occur in
paraparesis. coccidioidomycosis and candidiasis.
• Cranial nerve palsies, psychiatric abnormalities, speech
disturbances and seizures are not common.
Cryptococcal Meningitis 319

384 385 386

387 388

389

384–387 Plain CT brain scan showing dilatation of all ventricles and


low density, due to edema, in the subependymal and periventricular
regions of a patient with communicating hydrocephalus due to
cryptococcal meningitis.

390

388–390 Cranial CT scan with contrast showing diffuse enhancement


of the basal meninges, particularly those surrounding the upper cervical
spinal cord at the level of the foramen magnum (390, arrows), and also
within the fourth ventricle and its lateral recesses (389, arrows).
320 Infections of the Nervous System: Fungal

CSF Mild case


• Gram stain: spherical cells, 5–15 μm in diameter, which • Fluconazole 800 mg loading dose by mouth, then
retain the Gram stain and are surrounded by a thick 400 mg daily by mouth.
refractile capsule. • Itraconazole may be used instead of fluconazole but
• India ink stain for cryptococcal antigen. The carbon experience with itraconazole is limited.
particles fail to penetrate the capsule, producing a white
halo around the double refractile wall of the yeast. Most patients respond clinically within 1–2 weeks, with
• Latex agglutination test or ELISA for the cryptococcal an early mortality of 5–10%. Continue therapy for
polysaccharide antigen. 4–6 weeks or until the CSF is sterile.
• Cryptococcal antigen titer (more than 1 : 8 is diagnostic).
• Culture: positive in 4–12 days. Isolation of yeast confirms Management of raised intracranial pressure
the diagnosis and allows for sensitivity tests in vitro. • Mechanical drainage (repeated LPs, intraventricular
• Opening pressure should be measured because elevated shunt).
intracranial pressure may have important prognostic • Acetazolamide treatment.
implications, particularly for visual impairment. • Corticosteroids are being studied in a controlled trial.
• White cell count: may be normal or only mildly elevated.
• Protein: normal or elevated. Maintenance therapy to prevent relapses
• Glucose: normal or decreased. Triazole antifungal agents (fluconazole 200–400 mg daily
by mouth, and itraconazole) have a more favorable toxicity
N.B. It is essential to test the CSF for cryptococcal profile and may be preferable for long term suppressive
organisms and antigen in patients with suspected cryptococcal treatment necessary to avoid the high incidence of relapse
meningitis because the CSF cell count, protein and glucose (more than 50%) in patients with AIDS. With maintenance
can all be normal, particularly in AIDS patients. Large volumes therapy, relapses are uncommon and usually related to non-
of CSF (20–40 ml) may be needed to find the organism. compliance with treatment, but may also occur because of
development of drug resistance and drug interactions which
Other lower fluconazole levels. Monitoring serum cryptococcal
• Serum cryptococcal antigen: positive. AIDS patients have antigen titers is not useful in predicting relapse.
higher antigen titers in blood than in CSF.
• Blood and urine cultures are often positive. CLINICAL COURSE/PROGNOSIS
A steadily progressive course occurs over several weeks or
DIAGNOSIS months. In a few patients, the course may be remarkably
Serum cryptococcal antigen is almost always positive in indolent with periods of clinical improvement and
cryptococcosis, but meningitis should be confirmed by CSF normalization of the CSF. In others, it may be fatal within
examination. Positive India ink staining or CSF cryptococcal a few weeks if untreated.
antigen titer will provide rapid diagnosis, which is confirmed Unfavorable prognostic factors are:
subsequently by CSF culture. • Altered mental status.
• High CSF cryptococcal antigen titer.
TREATMENT • Low CSF leukocyte count.
Treatment is determined by the severity of the illness. • Positive extrameningeal culture for cryptococcus, hypo-
natremia, and raised intracranial pressure (for vision).
Primary therapy (treatment of acute infection)
Severe case
Altered consciousness, CSF white blood cell count
>20/mm3, CSF antigen titer >1 : 1024 and a positive blood
culture:
• Amphotericin B 0.5–0.7 mg/kg body weight per day by
intravenous injection. Amphotericin is limited by
frequent adverse effects, such as fever, renal dysfunction
with renal tubular acidosis and, rarely, leukoencepha-
lopathy. A liposomal form of amphotericin B may have
less adverse effects.
• +5-flucytosine (100–150 mg/kg/day by mouth or
intravenously) for 2–3 weeks (the value of adding
flucytosine remains undetermined). Flucytosine adverse
effects include myelosuppression and gastrointestinal
toxicity.
• Followed by fluconazole (an oral triazole antifungal
agent) 400 mg/day by mouth, for an additional
8–10 weeks; fluconazole is as effective as amphotericin B
but fluconazole causes delayed clearance of cryptococcus
from the CSF.
Mucormycosis of the Nervous System 321

MUCORMYCOSIS OF THE RISK FACTORS/PREDISPOSING CONDITIONS


NERVOUS SYSTEM • Diabetes mellitus:
– Present in about three-quarters of cases.
– A risk factor for cutaneous and localized paranasal
DEFINITION infection.
An opportunistic, malignant infection of the brain vessels – The fungus thrives in an environment rich in glucose and
caused by zygomycete fungi, including those of the genera acid pH.
Absidia, Rhizomucor and Rhizopus. – Poor glycemic control and ketoacidosis impair phagocytic
activity of neutrophils.
EPIDEMIOLOGY – Diabetic ketoacidosis is associated with increased serum
• Incidence: rare. free-iron levels, which facilitates growth of rhizopus, an
• Age: often adolescents and young adults. iron-requiring organism.
• Gender: M=F. • Burns (for skin invasion).
• Neutropenia (for widespread mucor infection).
PATHOLOGY • Leukemia.
Several forms • Lymphoma.
• Orbital. • Cytotoxic drug treatment.
• Rhino-orbital. • Corticosteroid treatment.
• Rhino-orbito-cerebral. • Post organ transplant.
• Cutaneous. • Renal failure.
• Pulmonary. • AIDS.
• Gastrointestinal. • Amebiasis.
• Disseminated. • Malnutrition.
• Kwashiorkor.
Rhino-orbito-cerebral form • Intravenous drug abuse.
• Mucosal thickening of the paranasal sinuses on one or
both sides. CLINICAL FEATURES
• Soft tissue retro-orbital mass. Retro-orbital mucormycosis
• Numerous broad, irregular non-septate fungal hyphae History
with non-parallel margins, within the arterial wall and • Headache (75%).
often extending into the surrounding brain parenchyma. • Eye pain.
• Secondary intraluminal arterial thrombosis. • Facial swelling.
• Secondary hemorrhagic infarction of the brain. • Dark, blood-stained nasal discharge (45%).
• Intracranial granuloma from hematogenous spread are
unusual. Examination
• Cavernous sinus thrombosis following invasion of orbital • Sick (i.e. looks unwell).
apex. • Altered sensorium (30%)
• Brain abscess following direct intracranial extension. • Fever (55%)
• Orbito-fascio- cellulitis (35%): swelling, mucopurulent
ETIOLOGY AND PATHOPHYSIOLOGY discharge from the eye.
The fungi are acquired from the environment and infect the • Proptosis.
lungs, paranasal sinuses and, less commonly, the skin. • Edema of the eyelids.
The brain infection begins in the nasal turbinates and • Reduced visual acuity and direct light reflex.
paranasal sinuses and spreads along infected vessels through • Relative afferent pupillary defect.
the orbital apex to the retro-orbital tissues and through the • Retinal edema.
cribriform plate to the brain, where it may cause a cerebral • Rapid reduction in visual acuity, resulting in blindness in
vasculitis with secondary arterial and venous thrombosis and one eye.
hemorrhagic infarction. • Ophthalmoplegia (35%).
• Hemiparesis (30%).

Cerebral mucormycosis
Carotid territory hemorrhagic infarction of the brain (e.g.
hemiparesis, dysphasia).

DIFFERENTIAL DIAGNOSIS
• Cavernous sinus syndrome such as cavernous sinus
thrombophlebitis (see p.258).
• Retro-orbital tumor.
322 Infections of the Nervous System: Fungal

INVESTIGATIONS DIAGNOSIS
CT or MRI brain scan (391–393) The diagnosis of fungal infection due to mucormycosis is
• Typically starts as a rim of soft tissue thickness along the strongly suggested by painful proptosis and redness of the
walls of the paranasal sinuses. eye with blood-stained nasal discharge in the setting of
• Fluid levels, obliteration of the nasopharyngeal tissue diabetes mellitus and poor glycemic control. Although
planes, bone destruction. demonstration of non-septate hyphae on histopathologic
• Orbital extension causes proptosis, superior orbital vein examination further supports the clinical diagnosis, the
thrombosis. definitive diagnosis depends on growth of zygomycetes in
• Extension through the orbital apex causes cavernous fungal culture of the purulent discharge, lesion scrapings or
sinus thrombosis. biopsy material.
• Further intracranial extension causes brain parenchymal
low density on CT with mass effect and enhancement in
the anterior and temporal fossae.
• Intracranial abscesses are also seen. 391
• CT is best for demonstrating soft tissue invasion, necrosis
and early bone erosion.
• MRI, with or without gadolinium, is best for evaluating
intracranial extension including carotid artery thrombosis
and cavernous sinus thrombosis.

Carotid angiography
May show features of vasculitis, pseudoaneurysm formation,
arterial occlusion.

Blood
• Full blood count: leukocytosis (white cell count may be
used to monitor response to treatment).
• Glucose: hyperglycemia.
• Arterial blood gases: acidosis.
• Serology has no place in the diagnosis of mucormycosis.

Other
• Swab of purulent discharge from the eye and nose:
culture fungus on Sabouraud medium.
• External ethmoidectomy: scrapings or biopsy of the nasal 391 CT scan showing extensive muco-periosteal disease involving the
lesion to look for broad, irregular non-septate hyphae on maxillary antra (arrows) in a patient with non insulin-dependent
microscopy. diabetes mellitus, chronic renal failure, and cavernous sinus thrombosis
due to mucormycosis.The sphenoid and frontal sinuses were also
involved.

392 393 392, 393 CT brain scans


with contrast same patient
as 391). Coronal (392) and
axial (393) views of an
enhancing mass extending
from the paranasal sinuses
into frontal fossa (arrows)
due to mucormycosis.
Toxoplasmic Encephalitis 323

TREATMENT TOXOPLASMIC ENCEPHALITIS


• Control diabetes (e.g. correct hyperglycemia and
acidosis) with fluids and insulin.
• Amphotericin B: rapid infusion of 0.5–1 mg/kg/day DEFINITION
dissolved in 5% dextrose solution to a total cumulative Infection of the brain by Toxoplasma gondii, which is a
dose of 1.5–4 g over a prolonged period of 8 weeks protozoan intracellular parasite that reproduces sexually only
because of the fungistatic rather than fungicidal nature of in cats and differentiates into encysted forms.
the drug. Cessation of treatment is guided by clinical
symptoms and signs, including resolution of fever, wound EPIDEMIOLOGY
healing, decreasing white cell count, and negative cultures. Prevalence
Limitations are nephrotoxicity, bone marrow depression, Toxoplasma infection
cardiotoxicity and hypokalemic weakness. Liposomal Toxoplasma infection is widespread in the community, with
amphotericin can be used in much higher doses without marked geographic variation. About 50–80% of adults in
any significant risk of the above adverse effects. continental Europe are infected, compared with 30% in the
N.B. Azole group of antifungals, such as UK. Antibody prevalence is highest in warm areas with high
ketoconazole, are not recommended. rainfall where the infective sporocyst is more likely to
• Surgical debridement: in early cases without necrosis, survive. Contact with cats and eating raw or undercooked
simple nasal, transantral or ethmoidal biopsy and meat are also contributing factors.
curettage should be undertaken. Extensive necrosis Disease occurs sporadically, though outbreaks with a
requires more radical debridement. A blind eye should common source have been reported, such as following the
not be removed as routine because there are no data to consumption of unpasteurized milk.The highest incidence
suggest that orbital exenteration improves survival. of infection occurs in the late teenage years and early 20s.
• Hyperbaric oxygen: reported to inhibit the growth of
Zygomycetes in vitro. Toxoplasma encephalitis
Arises in the immunocompromised host (e.g. AIDS). In the
PROGNOSIS pre-prophylaxis era, toxoplasma encephalitis was the AIDS
• Orbital mucormycosis: amphotericin B has considerably index diagnosis in up to one-half of cases, and the most
reduced mortality from 90% in the untreated state to common cause of focal brain lesions in HIV/AIDS,
40%. occurring in 3–40% of patients, usually when HIV infection
• Rhino-orbito-cerebral mucormycosis: infection of the was advanced and CD4 count <200 cells/mm3.
brain and cerebral vessels is usually rapidly fatal but a Toxoplasma encephalitis is now rarely seen among
minority recover with appropriate emergent treatment. patients attending HIV clinics who are commenced on
prophylaxis when they become immunosuppressed; it is only
Adverse prognostic factors for survival seen in ‘lay’ presenters (i.e. unknown HIV infection) and
• Delayed diagnosis and treatment. patients who are poorly compliant with prophylactic therapy.
• Leukemia.
• Renal disease. PATHOLOGY
• Treatment with desferrioxamine. Toxoplasma infection in the immunocompetent host
• Bilateral sinus involvement. • Lymphadenopathy.
• Facial necrosis. • Chorioretinitis.
• Nasal deformity.
• Hemiparesis. Toxoplasma infection in the immunocompromised
host (e.g. AIDS)
Toxoplasmosis is at its most severe in immunocompromised
individuals.

Primary infection may cause


• Necrotizing encephalitis (widespread microscopic lesions).
• Brain abscesses, usually in the gray matter of the
diencephalon and cortex.
• Pneumonia.
• Myocarditis.
• Myositis.
• Reactivation of quiescent tissue cysts in the brain.
• Focal encephalitis.

ETIOLOGY AND PATHOPHYSIOLOGY


Humans ingest oocysts in cat litter or contaminated soil
(e.g. from gardening, washing vegetables, playing in sand
pits), or ingest tissue cysts in infected meat.
Bradyzoites are released from the ingested sporocyst and
penetrate intestinal cells, reach the lymphatic system, multiply
asexually to trophozoites and then migrate throughout the
body via lymphocytes. They disrupt and invade adjoining cells
324 Infections of the Nervous System: Protozoan

and, eventually, a tissue cyst forms, which is partly host in origin Other uncommon CNS infections in AIDS
and contains thousands of bradyzoites that are morphologically • Candida albicans infection.
identical to trophozoites but have a reduced metabolic rate. • Coccidioidomycosis.
These persist for the lifetime of the host in the brain and skeletal • Histoplasmosis.
and cardiac muscle, continually stimulating the immune system • Mycobacterium tuberculosis infection.
and conferring immunity. IgG antibody reaches a maximum • Herpes zoster infection.
concentration 2 months after infection and persists for life. • Herpes simplex infection.
Acute disease is caused by a type III hypersensitivity reaction, • Aspergillosis.
leading to intravascular thrombosis and tissue infarction. • Amebiasis.
Cyst reactivation is initiated by a reduction in cell mediated • Trypanosoma cruzi infection.
immunity (CD4 count <100 cells/mm3) and a type IV • Nocardia.
reaction, accompanied by round cell infiltration, granuloma
formation, and tissue necrosis. In chronic infection, cell- INVESTIGATIONS
mediated immunity forms the basis of the immune response. Serum antitoxoplasma antibodies
• Specific antitoxoplasma IgG is usually detectable and
CLINICAL FEATURES supports the diagnosis but a low titer or an absence of
Various types of presentations: antibody does not exclude the diagnosis; about 3% of
• Infection with Toxoplasma gondii is usually asympto- toxoplasmic encephalitis patients are seronegative.
matic, except for congenital infection and in the im- • Immunofluorescence assay may be less sensitive than
munocompromised host. ELISA in detecting antitoxoplasma IgG.
• Encephalitis usually evolves quite rapidly (time from
onset to presentation is only a few days). CT brain scan (394, 395)
• Subacute focal (hemisphere > brainstem or cerebellum) Single or multiple nodular low density areas (396), often at
encephalopathy: hemiparesis, aphasia, apraxia, hemi- the cortico-medullary junction or in the basal ganglia, with
sensory impairment, homonymous hemianopia, ataxia, little or no enhancement, or gyriform enhancement; or,
and cranial nerve palsies. isodense ring-enhancing nodules in the basal ganglia. These
• Diffuse encephalopathy: reduced alertness/confusion features are not diagnostic of toxoplamic encephalitis as
with subsequent evolution of focal neurologic signs over other opportunistic infections and lymphoma can produce
weeks to months. similar lesions.
• Constitutional (systemic) symptoms: headache, fever
(47%), lethargy (43%) and seizures (29%), and disorien- MRI brain scan
tation, altered mental state and coma with more diffuse • Often shows more lesions, that were not shown by CT.
cerebral dysfunction. • Multiple lesions of high intensity usually in the basal
• Acute onset of a focal neurologic deficit or seizures due ganglia (397, 398), with vasogenic edema and ring or
to intracerebral hemorrhage. nodular enhancement on T1WI (399).
• Single lesions seen on MRI are more likely to be due to
DIFFERENTIAL DIAGNOSIS lymphoma. Rarely, the CT and MRI may be normal in a
Focal brain lesions diffuse form of toxoplasmic encephalitis.
• Primary CNS lymphoma: slower onset (patients present
within 1–2 weeks of onset of symptoms) and greater CSF
involvement of the periventricular white matter. • Non-specific abnormalities.
• Progressive multifocal leukoencephalopathy: slower, • PCR to detect toxoplasma DNA in CSF is a promising
quite indolent, onset over several weeks, with lesions definitive diagnostic tool.
confined to the white matter.
• Tuberculous brain abscess. Brain biopsy
• Demyelination. To demonstrate tachyzoites histologically.
• Vasculitis related to varicella-zoster infection.
• Syphilis DIAGNOSIS
• CMV. • The diagnosis of CNS toxoplasmosis may be presumed
• Stroke. in patients with characteristic clinical and radiologic
findings and detectable antitoxoplasma antibody.
Encephalitis • The definitive diagnosis requires demonstration of
• CMV: non-specific clinical and neuroimaging features tachyzoites histologically or by detection of specific
but in some cases CT or MRI show diagnostic toxoplasma DNA by PCR.
subependymal prominence (signal change and contrast
enhancement) and sometimes small areas of gray matter CLINICAL COURSE
enhancement. A low serum sodium may also be a clue. Evolves rapidly if untreated.
• Herpes simplex.

Myelitis
• CMV.
• Herpes simplex.
• Varicella-zoster.
• Syphilis.
Toxoplasmic Encephalitis 325

394, 395 CT brain scan of 394 395


congenital toxoplasmosis (two
different patients). Note the tiny
calcified nodules near the lateral
ventricles (arrows) and the
prominent ventricles due to
atrophy.

396 CT brain scan with 396 397


contrast in a patient with HIV
infection and cerebral
toxoplasmosis.There is a solitary
lesion in the left occipital lobe
(arrows).

397, 398 T2W axial (397) and


T1W coronal (398) MRI with
contrast showing multiple lesions
in the basal ganglia and
peripherally due to toxoplasma.
(Courtesy of Professor J Best,
Department of Medical
Radiology, Royal Infirmary,
Edinburgh, UK.)

399 T1W MRI with contrast 398 399


(same patient as in 396)
confirms the presence of the
enhancing lesion, which resolved
with antitoxoplasma treatment.
(Courtesy of Professor J Best,
Department of Medical
Radiology, Royal Infirmary,
Edinburgh, UK.)
326 Infections of the Nervous System: Worm

MANAGEMENT CYSTICERCOSIS
• HIV-infected, toxoplasma-seropositive, and a single
lesion on CT: proceed to MRI and, if this also shows
only a single lesion, then biopsy should be considered DEFINITION
because of possibility of lymphoma. Otherwise consider The larval or intermediate stage of infection with the pork
an empirical trial of antitoxoplasma therapy, await tapeworm Taenia solium; a cysticercus is a fluid-filled
response, and if no improvement after 2–3 weeks, bladder that contains the invaginated head or scolex of the
consider stereotactic brain biopsy. larval form of Taenia solium.
• HIV-infected, brain lesions on CT, but no specific toxo-
plasma antibody or receiving prophylactic trimethoprim- EPIDEMIOLOGY
sulfamethoxazole: consider for brain biopsy because of • Prevalence: endemic (affects 2–4% of the general
the increased likelihood of other disease. population) in Mexico, Central and South America,
• HIV-infected, toxoplasma-seropositive, and multiple Poland, China, Africa, India and New Guinea where it is
ring-enhancing lesions on CT: start empiric antitoxo- the leading cause of adult-onset epilepsy. Becoming more
plasma therapy, await a response, and if no response, widespread in the United States, France, and Italy, and
consider brain biopsy (359). less commonly other countries in Europe and Australasia
because of the massive emigration from endemic areas.
PRIMARY THERAPY • Age: more common in adults than children.
• Pyrimethamine (200 mg loading dose orally followed by • Gender: M=F.
75 mg/day by mouth) + sulfadiazine (100 mg/kg per
day, in four divided doses up to 8 g/day by mouth or ETIOLOGY
intravenously) + folinic acid (leucovorin calcium) (7.5 mg Accidental ingestion of eggs produced by adult Taenia
daily by mouth). Or: solium, or pork tapeworms.
• Pyrimethamine and folinic acid (leucovorin calcium) (as
above) + clindamycin (600 mg four times a day by PATHOPHYSIOLOGY
mouth or intravenous injection). The adult Taenia solium, or pork tapeworm, which is 2–8 m
(6.5–26 ft) in length, resides (for up to 10 years or so) in
Pyrimethamine is the most important drug. Adverse the small intestine of the human, which is its only known
effects occur in >40% of patients, usually rash and nephro- host. It attaches to the intestine by its scolex or head. The
toxicity, which are particularly related to sulfadiazine. terminal, egg-bearing segments, or proglottids, occasionally
Hematologic toxicity of pyrimethamine (e.g. megaloblastic separate and are discharged in the feces. Infectious ova are
anemia) can be ameliorated with folinic acid (leucovorin also deposited in the perianal area and can contaminate
calcium) (5–10 mg/day). Continue treatment for at least 3 fingers and other parts of the body.
weeks, up to 6 weeks if the patient does not show a complete Food or water that is contaminated with Taenia solium
response. Up to 86% of responders show improvement by eggs (ova) from human feces may be ingested by the pig,
the seventh day of treatment. If clinical or radiologic which is the main intermediate host, or human beings
improvement does not occur within 1–2 weeks of starting (fecal–oral route), also serving as intermediate hosts.
empirical therapy for toxoplasmosis, an alternative diagnosis
should be pursued using stereotactic brain biopsy. Larval stage
Human ingestion of food and water contaminated by
Maintenance therapy cysticercus ova is the most common means by which
Relapse of CNS toxoplasmosis is common in patients with humans contract systemic cysticercosis. The ingested eggs
HIV infection so life-long maintenance therapy is indicated: hatch in the small intestine, develop into embryos or
• Sulfadiazine (500 mg four times a day by mouth) + oncospheres, and migrate through the mucosa of the small
pyrimethamine (25 mg daily by mouth) + folinic acid intestine and enter the portal circulation from where they
(leucovorin calcium) (7.5 mg daily by mouth). Or: are distributed to capillaries in the parenchyma of the brain,
• Clindamycin (600 mg four times a day by mouth) + pyri- subarachnoid space and ventricles, the eyes, and striated
methamine and folinic acid (leucovorin calcium) (as above). muscles. Once in capillaries these oncospheres develop into
cysticerci, or encysted larvae, within about 2 months. Viable
PRIMARY PREVENTION encysted larvae consist of cysts with the larva projecting into
In countries with a high background seroprevalence of the center of the cyst from the wall to which it is attached
toxoplasma, HIV-infected patients with a CD4 cell count by its scolex. No further development of the worm occurs
<200/µl should have prophylaxis for toxoplasmosis while it is encysted, and it can remain in this stage for many
(trimethoprim and sulfamethoxazole). Many primary years. The cyst maintains a diameter of 10–20 mm
prophylactic regimes for Pneumocystis carinii have antitoxo- (0.4–0.8 in). There is little or no inflammation and the
plasmal activity. Coinfected pregnant women whose CD4 cell patient is usually asymptomatic.
count is <200/µl warrant primary prophylaxis as maternal The same process occurs in pigs who ingest food and
reactivation may result in congenital toxoplasmosis, which has water contaminated by cysticercus ova. The cyst (in pig
a rapidly fatal course in the HIV-infected infant. HIV-infected muscle) must be ingested (by humans) for the larva to
patients who are toxoplasma-seronegative should be advised emerge and develop into an adult in the gut of the definitive
to wear gloves when changing cat litter or gardening and to host. Thus humans can be intermediate as well as definitive
avoid eating undercooked meat, particularly lamb or pork, hosts, whereas pigs are only intermediate hosts, since they
unless it has been thoroughly frozen (–20°C [–4°F]). cannot acquire the adult infection by eating cysts.
Cysticercosis 327

Active stage • Cysts may have a normal morphologic pattern or may be


After an incubation period of <1–30 years (average complicated by progressive inflammation with
4.8 years), the parasite (larva) degenerates and dies, and the granulomatous meningitis and proliferative endarteritis,
disease enters the active phase. The cyst loses osmotic regu- associated with obstruction of the foramina of Luschka
lation, and swells to a diameter of 40–50 mm (1.6–2 in). An and Magendie and involvement of the cranial nerves.
inflammatory reaction is generated against parasite antigens Intraventricular cysts may cause hydrocephalus when
and the cyst develops into a granulomatous abscess, with a located at the foramen of Monro.
fibrous capsule, a granulomatous inflammatory infiltrate with
epithelioid giant cells, and an interior consisting of pus and CLINICAL FEATURES
the bladder-wall remnants. This inflammatory response is Diverse and depend on the number of cysticerci, their
responsible for symptoms such as seizures, enhancement of location in the brain and the immune status of the patient.
the cysts after the administration of contrast, and a lympho-
cytic response in the spinal fluid. Cysticerci located in the Presenting features
basilar subarachnoid space may induce an intense inflam- May comprise any combination of:
matory reaction, leading to hydrocephalus, cranial nerve • Non-specific ‘flu-like’ illness in the initial stage of larval
palsies and stroke due to a basal vasculitis. invasion.
• Epileptic seizures: the most common manifestation of
Inactive stage the active stage; caused by parenchymatous cysts.
The abscess may then undergo progressive mineralization, • Raised intracranial pressure.
with collagen and calcium deposition (the inactive stage) • Meningoencephalitis.
and without surrounding edema or an inflammatory • Ventriculitis.
response. • Hydrocephalus.
Human ingestion of raw or undercooked ‘measled’ • Hemiparesis.
muscle of pig (pork) contaminated with mature cysticercus • Cranial nerve palsies.
usually causes asymptomatic bowel infestation with the • Acute cerebellar ataxia.
tapeworm, as the human, like the pig, becomes an • Behavioral disturbance.
intermediate host. This does not necessarily lead to, and is • Asymptomatic (the cysts being discovered radiologically).
not a prerequisite for, the development of systemic
cysticercosis. Ingested cysticerci invaginate into the mucosa Other features
of the small intestine and then develop to adulthood. The • History of travel to, or residence in, an area where
adult worm elongates up to a length of 2–8 m (6.5–24 ft), cysticercosis is endemic.
and may survive in situ for up to 25 years. The terminal part • Low grade fever.
of the worm containing the eggs is passed in the feces. • Palpable subcutaneous nodules in the soft tissues of the
limbs.
PATHOLOGY
• Multiple calcified lesions in the muscles of the thigh, leg, DIFFERENTIAL DIAGNOSIS
and shoulder and in the brain. • Hydatid disease with cysts produced by Echinococcus
• The brain is affected in 90% of cases of human cysti- granulosus.
cercosis: leptomeninges (especially the basilar cisterns) • Viral encephalitis: varicella, coxsackie, echovirus.
(55%), ventricular system (more often in the fourth • Brain tumor: primary or secondary.
ventricle) (15%), and in the parenchyma (30%). • Drug toxicity (e.g. phenytoin).
• Eosinophilic meningoencephalitis due to parasites:
Intraparenchymal cysticercosis – Angiostrongylus cantonensis.
• Cysticerci in the brain parenchyma are located in the gray – Gnathostoma spinigerum.
matter at the gray-white matter junction and are solitary – Paragonium westernensis.
in 25–75% of patients. They often appear as an enlarged
larva surrounded by an abscess wall with adjacent gliosis. INVESTIGATIONS
• The cysts may be alive, but the majority (60%) are dead Radiography
and calcified. • Plain skull x-ray shows calcification in the brain
parenchyma, typically 1–2 cm (0.4–0.8 in) in diameter,
Racemose cysticercosis surrounded by a calcified sphere of 7–12 cm (2.8–4.7 in)
• A proliferating form of larval tapeworm that consists of diameter.
multiple interconnected bladders of different sizes that • Soft tissue radiography of thighs, and limbs for evidence
lack scolices. of calcified larvae.
• Occurs in the subarachnoid space and cisterns at the base
of the brain, sylvian fissures, and wider parts of the fourth
ventricle.
• Cysts collect in grape-like clusters with tiny points of
attachment to the pia arachnoid, or the larvae migrate
within the ventricles.
• ‘Complex arachnoid cysts’: cysts that lie within major
sulci or fissures in which a wall composed principally of
inflamed meninges develops once the larva has aged,
decayed, and died.
328 Infections of the Nervous System: Worm

CT (400, 401) TREATMENT


CT shows single or multiple hypodense cysts that are Treatment depends on the presenting syndrome, nature of
localized or generalized and contain a small, asymmetrically the neurologic impairment, location and activity of cysticerci
located ‘off-center’, internal, hyperdense nodule (with or and host immune response.
without calcification) corresponding to the dead and Calcified neurocysticerci represent inactive cysts, and
hyalinized larval scolex (calcification occurs where cysts have neither require nor respond to anticysticercal drug treatment.
died). The cyst fluid is of CSF density. The cysts or nodules
enhance with intravenous contrast as the cysticerci Anticysticercal drugs
degenerate. This occurs early on, together with edema and • Praziquantel, an isoquinolone antihelminthic agent,
mass effect, but the enhancement wears off as the lesion ages 50 mg/kg of body weight, given daily for 15–30 days,
and the edema disappears. CT may reveal complications, depending on the number and size of the cysts.
which include hydrocephalus and basal meningitis. • Albendazole, an imidazole antihelminthic agent,
15 mg/kg body weight per day for 21 days, is the
MRI (402, 403) preferred drug as its plasma levels increase by 50% when
MRI shows similar features to CT, with the cysts of CSF used concurrently with dexamethasone, in contrast to
signal. MRI is more sensitive than CT for detecting edema praziquantel whose bioavailability is decreased by 50%.
(which indicates lesion activity; edema occurs as the parasite Albendazole produces an 80% reduction in total number
dies) and non-calcified cysticercosis lesions in the ventricles of cysts after a single course of therapy, compared to a
and subarachnoid space, such as in the basal cisterns. 65% reduction after praziquantel.

Blood Anti-inflammatory drugs


• Full blood count and film: leukocytosis due to peripheral A host inflammatory reaction to the death of cysticerci as
eosinophilia a result of anticysticercal drug treatment may lead to
• ESR: elevated. cerebral edema and increased intracranial pressure.
• Serum immunochemistry: ELISA for IgM antibodies Concurrent treatment with:
against the cysticercus antigen, and the enzyme-linked • Prednisolone (1–2 mg/kg/day oral for 7 days), or
immunoelectrotransfer blot (EITB), which has a • Dexamethasone.
sensitivity of greater than 93% and a specificity of 100%.
Surgery
CSF • A CSF shunting procedure may be required for
• CSF is negative in 49–63% of cases of the hydrocephalus due to cisternal cysticercosis and chronic
intraparenchymal form; pleocytosis, eosinophilia, elevated arachnoiditis.
protein, and occasionally hypoglycorrhachia occur in the • Excision/decompression of large subarachnoid or
encephalitic, racemose and basilar meningitic forms. intraventricular cysts that obstruct CSF flow.
• Immunochemistry: ELISA for IgM antibodies against the
cysticercus antigen shows a sensitivity of 87% and Anti-epileptic drug therapy
specificity of 95% in active disease. False positives can occur If required (i.e. symptomatic epileptic seizures).
in neurosyphilis and meningeal tuberculosis. The EITB
has a sensitivity of about 80% and a specificity of 100%. Prevention
• Disposal of human feces in a sanitary manner.
Other • Cook or freeze all meat or fish thoroughly before it is eaten.
Anal scrapings and fecal examination for ova and parasites: • Mass treatment with albendazole or praziquantel could
frequently negative. eliminate cestodes from the world, but this would be a
formidable task, given the widespread distribution of the
DIAGNOSIS pork tapeworm.
• A definitive diagnosis requires identification of the
proglottids or eggs of the larval form of T. solium in PROGNOSIS
stool. The gravid segments of the eggs must be examined Initially treatment may seem to exacerbate neurologic
for their characteristic morphologic features. symptoms, with an increase in cells and protein in the CSF,
• As this can be difficult to identify positively, a positive but then the patients improve and many become
diagnosis can be made by detecting antibodies to T. asymptomatic, with a striking decrease in the size and
solium by Western blot analysis. Biopsy can also be used number of cysts on CT scanning.
to make a positive identification. Cisternal and racemose cysticercosis are less responsive
to anticysticercal drugs than parenchymal cysticercosis.
The outcome of patients with epilepsy due to
neurocysticercosis is better after anticysticercal drug therapy
than when the primary disease is left to follow its natural course.
This is true for both those in whom the cysticerci remain
unchanged for long periods and for those in whom there is an
intense inflammatory reaction that eventually destroys the
cysticerci. Also, there are fewer seizures after medical treatment
than after surgical removal of the cystic lesion.
Cysticercosis 329

400 401

400, 401 Plain cranial CT scan showing multiple small hyperdense areas of calcification (arrows – dead cysts)
in a patient with inactive cysticercosis.

402 403

402, 403 MRI brain scan showing subtle inactive cysticercosis lesions (arrows).
330 Infections of the Nervous System: Prion

CREUTZFELDT–JAKOB • Inadequately sterilized neurosurgic instruments and


DISEASE (CJD) depth electrodes: 6 cases.
• Human gonadotrophin: 4 cases.
• Corneal transplant: 2 cases.
DEFINITION
A neurodegenerative condition that is the most common Sporadic, randomly distributed illness of unknown
clinicopathologic subtype of the transmissible spongiform cause (85% of cases)
encephalopathies or prion diseases.
The transmissible spongiform encephalopathies are all
characterized by the deposition in brain of an abnormal,
protease resistant, isoform of a membrane-bound
glycoprotein, the prion protein. Since the spongiform 404
change can be variable, the term prion disease is more
accurate.

EPIDEMIOLOGY
• Incidence: 1 per million per year.
• Age: median age of onset: 61 years; rare (but increasingly
reported [nvCJD]) younger than 30, and older than
80 years.
• Gender: M=F.

PATHOLOGY
Macroscopic
Normal or slightly atrophic brain.

Microscopic (404) 404 Microscopic section of the gray matter of cerebellum of a patient
A triad of: with CJD showing neuronal loss, reactive astrocytic proliferation and
• Neuronal loss. gliosis, and spongiform degeneration (vacuolation of the neuropil).
• Reactive astrocytic proliferation and gliosis.
• Spongiform degeneration (status spongiosus and
spongiform change: vacuolation of the neuropil [405, 405
406]): particularly in deeper laminae of the gray matter
of the frontal and temporal lobes, but also the gray
matter of the striatum, thalamus, tegmentum of the
upper brain stem, and cerebellar cortex.

Kuru plaques are also present: eosinophilic, round,


compact extracellular depositions of prion protein (PrP);
pathognomonic of a prion disease but found in only a few
sporadic cases.

Ultrastructural
Intraneuronal, complex, clear vacuoles whose membranous
septa look curled in profile.

ETIOLOGY
Inherited mutation in the PrP gene 406
Familial CJD (10–15% of cases)
Autosomal dominant pattern of inheritance; point
mutations or insertions in the prion protein gene located on
the short arm of chromosome 20. The codon 200Lys and
178Asp mutations (in association with codon 129Val/Met
polymorphism) are associated with neuropathologic changes
essentially indistinguishable from sporadic CJD.

Acquired
Iatrogenic transmission of CJD (<5% of cases)
• Human cadaveric pituitary-derived growth hormone
(hGH): >100 cases worldwide.
• The estimated risk of developing cadaveric hGH-induced
CJD for patients treated with cadaveric-derived hGH is
1 in 200–300. 405, 406 Neuronal vacuolation and spongiform change in the brain of
• Foreign dura mater grafts: 69 cases. a patient with CJD.
Creutzfeldt–Jakob Disease (CJD) 331

PATHOGENESIS • Most patients with iatrogenic CJD (treatment with


• Transmissible to experimental animals following cadaveric pituitary-derived hGH) are also homozygous,
inoculation or dietary exposure. mostly for the valine allele at this codon.
• Long incubation period, hence the initial concept of a slow • Heterozygosity for the PrP gene appears to be
virus infection, but no infective agent has been found. protective.

Prion hypothesis Risk factors


• The transmissible agent is a proteinaceous infectious • Iatrogenic routes of exposure.
particle, or prion. • Growth hormone deficiency.
• The prion appears to consist principally or entirely of a • Family history.
modified abnormal, partially protease-resistant isoform • Homozygosity for the valine or Met allele at codon 129
of a normal host-encoded cellular glycoprotein, the prion of the PrP gene.
protein (PrP). • Mutations in the PrP gene.
• Normal prion protein, described as PrPC, is a constituent • Occupational exposure to bovine prions.
of the surface of the neuronal cell.
• In the presence of mutations, now discovered in CJD CLINICAL FEATURES
and other transmissible spongiform encephalopathies • Variable (particularly with familial cases, and even within
(kuru, Gerstmann Straussler Scheinker syndrome [GSS], single pedigrees).
fatal familial insomnia [FFI], and atypical prion disease), • Insidious onset.
PrPC may change into a disease-related isoform, PrPSc, • Early behavioral abnormalities (initial symptom in about
which differs from the normal cellular isoform by a post- 10% of cases):
translational modification that appears to involve a – Personality change, withdrawal, apathy, depression, sleep
conformational change. disturbance.
• The post-translational change in protein structure from – Agitation, fear and paranoia may prompt a psychiatric
PrPC to the abnormal PrPSc is probably a critical referral.
pathologic event, causing either loss of function of PrPC • Rapidly progressive and profound dementia: forgetful,
or, less likely, toxicity due to gain of function of PrPSc. confused, visual distortion and hallucinations.
• The structural change of PrPC to PrPSc occurs as an • Myoclonus, usually stimulus-sensitive (>80% of patients).
inevitable event (predisposed by the change in primary • Motor abnormalities:
structure due to mutations in the PrP gene) in familial – Cerebellar ataxia.
disease, as an induced event by exogenous PrPSc in – Extrapyramidal signs: tremor, rigidity, bradykinesia,
transmitted cases (exogenous PrPSc interacts with dystonic posturing; choreoathetosis.
endogenous PrPC and induces the structural change), – Pyramidal signs: weakness, spasticity, hyper-reflexia, and
and as a rare spontaneous event in sporadic disease. Babinski signs.
Sporadic cases may also arise from a somatic mutation of • Generalized seizures may occur.
the PrP gene (in addition to spontaneous conversion of • Positive family history of CJD in 6–15% of cases.
the cellular isoform PrPC to PrPSc as a rare stochastic
event). Syndromes of iatrogenic CJD
• The prion hypothesis may explain the paradoxic • Psychiatric disturbance (e.g. anxiety and depression) and
observation that the disease can be genetically inherited, progressive ataxia and clumsiness before the appearance
transmitted, and sporadic. of dementia and myoclonus: iatrogenic CJD associated
• The clinical heterogeneity (see below) may reflect with peripheral inoculation of prions (i.e. cadaveric hGH
differences in host genotype (e.g. polymorphism at cases).
codon 129 of the PrP gene) and different isoforms of • Progressive dementia: iatrogenic CJD associated with
PrPSc (e.g. types 1 and 2). central inoculation of prions (i.e. foreign dura mater or
neurosurgic instruments).
Genetic susceptibility to acquired iatrogenic
and sporadic CJD Clinical variants (the spectrum)
• The general population has a common silent protein Heidenhain variant
polymorphism at position (codon) 129 of the human Early pathologic involvement of the occipital cortex leading
PrP gene, where either a methionine (Met) or valine may to cortical blindness.
be encoded (present). About 40% of whites are
homozygous for the more frequent Met alleles, 50% are Brownell–Oppenheimer variant
heterozygous for Met, and 10% are homozygous for the Early and prominent ataxia (cerebellar ataxia is also
valine allele. prominent in iatrogenic CJD due to pituitary-derived
• Homozygosity at PrP 129 (present in about 50% of the hormones).
general population) appears to confer susceptibility to
iatrogenic and sporadic disease. Thalamic variant
• Most patients with sporadic CJD are homozygous for Insomnia and dysautonomia.
either allele. The new variant cases of CJD (see below)
are Met 129 homozygous, suggesting that about 40% of Spastic paralysis with dementia (Worster–Drought)
the general population are susceptible.
332 Infections of the Nervous System: Prion

Amyotrophic form • GSS disease: autosomal dominant inheritance, prominent


Loss of anterior horn cells causing significant wasting, cerebellar ataxia, dementia.
weakness and areflexia due to lower motor neuron changes. • Psychiatric illness (anxiety, depression).

More chronic and indolent form INVESTIGATIONS (see Table 36)


Progresses over more than 2 years. CT or MRI brain
• Scans are normal (45%) or show cerebral atrophy (30%).
New variant CJD • T2W and PDWI MRI may reveal high-signal changes in
• Linked causally to bovine spongiform encephalopathy the basal ganglia (407) and thalamus (5%) (e.g. the
(BSE), nvCJD is believed to be caused by the oral ‘pulvinar sign’ in variant CJD) or in scattered areas such
transmission of the BSE agent through consumption of as the cortex (7%). The sensitivity and specificity of these
bovine tissues containing significant infectivity (e.g. beef findings are uncertain.
products containing bovine spinal cord). • Diffusion-weighted MRI, which rapidly detects tiny
• Young age of onset (below 42 years of age). changes in water mobility, such as shifts from the
• Early behavioral/psychiatric disturbance (anxiety, extracellular to the intracellular space when cellular
depression), cerebellar ataxia and dysesthesiae. homeostasis is disturbed, has been reported to not only
• Absence of typical EEG changes, although the EEG is show basal ganglia abnormalities but also gross
abnormal. abnormalities involving most of the cerebral hemispheres.
• High signal changes in the pulvinar of the thalamus on • MRI is probably the imaging modality of choice, though
T2W MRI imaging of the brain. both MRI and CT are insensitive. Their main role is to
• Clinical course is more prolonged and protracted, with exclude other differential diagnoses.
average duration of 13 months compared with a mean of
6 months in other types. EEG
• Specific neuropathologic profile with extensive forma- • Early: non-specific disorganization and generalized slow
tion of cerebellar prion plaques resembling those seen in wave activity.
kuru: they have a dense eosinophilic center and pale • Later (within 12 weeks of onset of symptoms):
periphery surrounded by a zone of spongiform change. – Slow background rhythm.
• Homozygous for Met at codon 129 of the PrP gene. – Periodic sharp wave complexes (408–411):
• Associated with a specific pattern of protease-resistant PrP bisynchronous, and most commonly anterior and central
on Western blot analysis. The biochemical signature of (but may be lateralized and localized: occipital
the prions is distinct from other types of CJD and preponderance in Heidenhain’s variant);
matches that of animals experimentally infected with BSE. duration: 100–600 ms;
amplitude: up to 300 mV;
DIFFERENTIAL DIAGNOSIS may be monophasic, biphasic, triphasic or multiphasic;
• Alzheimer’s disease with myoclonus, but the course is repetitive, occurring every 0.5–2.0 seconds;
usually more prolonged. may be associated with myoclonic jerks;
• Non-convulsive status epilepticus. may be activated by startle;
• Metabolic/toxic encephalopathy (e.g. drugs). present in >60% of cases, particularly sporadic CJD, rarely
• Bilateral subdural hematomata. in ‘peripheral’ iatrogenic CJD;
• CNS vasculitis. non-specific: also occur in several encephalopathies,
• Subacute sclerosing panencephalitis (SSPE). epilepsy and post ictal states, or during barbiturate
• Infiltrating corpus callosum glioma. overdose and deep anesthesia.
• Huntington’s disease. • 20–40% of patients do not exhibit a typical EEG (cf.
• Motor neuron disease, but normal cognition. nvCJD).

Table 36 Investigation findings in Creutzfeldt–Jakob disease


Disease ‘Typical EEG’ 14-3-3 +ve CSF High signal on MRI brain Other
Sporadic CJD + (65%) + (90%) + (>70%) (caudate and putamen)
Familial CJD ± ± ± PRNP analysis
Iatrogenic CJD
CNS route ± ? ?
Peripheral route – ± (50%) ±
New variant – (100%) ± (50%) + (>70%) (posterior thalamus) Tonsil biopsy
% of cases with a positive investigation, where known, are in parentheses
PRNP = prion protein
Creutzfeldt–Jakob Disease (CJD) 333

407 408

408 Electroencephalograph showing periodic triphasic wave


complexes that are rather simple in contour and recurring every
0.7–0.8 seconds, against a slow polymorphous background, in a patient
with rapidly progressive dementia and myoclonus due to CJD.
Occasionally, the periodic discharges begin unilaterally and may
resemble periodic lateralized epileptiform discharges.

407 T2W MRI from a patient with pathologically proven CJD. Note 409
increased signal (whiteness) of the basal ganglia (arrows). Normally the
basal ganglia become darker due to iron deposition with age.There is
also a minor degree of atrophy. Also note the increased signal in the
pulvinar (arrowheads).This was variant CJD.

409–411 Serial electroencephalographs, every 3 days, from a patient


with CJD showing the progressive evolution of periodic triphasic wave
complexes and the background rhythm.

410 411
334 Infections of the Nervous System: Prion

CSF DIAGNOSIS
• CSF has normal or slightly raised protein. Diagnostic criteria for classical CJD
• Immunoassay for the 14-3-3 brain protein. The 14-3-3 Sporadic CJD
protein is a non-specific marker of central nervous system Definite:
neuronal injury or death. A positive test is highly sensitive • Neuropathologically confirmed and/or
(90%) and specific for CJD when used in carefully • Immunocytochemically confirmed PrP positive (Western
selected patients with dementia. blot) and/or
• Neuron-specific enolase (NSE) concentrations may be • SAF.
increased early (>35 ng/ml; sensitivity 80%, specificity
92%) and when myoclonus and periodic sharp complexes Probable:
appear, and return to normal in the late stage. Raised • Progressive dementia.
NSE levels in CSF also reported in brain trauma, tumor, • Typical EEG.
and acute stroke including SAH. The enzyme is localized • At least two of the following clinical features:
in neurons and neuroendocrine cells and is synthesized – Myoclonus.
completely in the CNS. – Visual or cerebellar disturbance.
• Two-dimensional gel electrophoresis to detect two – Pyramidal/extrapyramidal dysfunction.
proteins, p130/131, with an apparent molecular weight – Akinetic mutism.
of 26 and 29 kDa and an isolectric point of 5.2 and 5.1:
highly specific; positive predictive value up to 100%, Possible:
negative predictive value about 69%. • Progressive dementia.
• Two of the clinical features listed above.
Molecular genetic analysis • No EEG done or atypical EEG.
Molecular genetic analysis of DNA in blood, CSF or brain. • Duration <2 years.
The prion protein gene (PRNP) blood test is a genetic test
for mutations that cause familial prion disease. The PRNP Accidentally transmitted CJD
is sequenced for the presence of pathogenic mutations, and • Progressive cerebellar syndrome in a pituitary hormone
the polymorphism risk factor found at codon 129 on the recipient.
PRNP gene. It is important to perform, even if no family • Sporadic CJD with a recognized exposure risk (e.g. dura
history is apparent, because mutations are commonly found mater transplant).
in apparently sporadic cases because of incomplete
penetrance, non-paternity, adoption, or the gene-carrying Familial CJD
parent having died before the age of onset of the prion • Definite or probable CJD plus definite or probable CJD
disease from another cause. in a first-degree relative.
Mutations occur at codons 102, 105, 117, 14, 178, 180, • Neuropsychiatric disorder plus disease-specific prion
198, 200, 210, 217, 232, and insertions in the PrP gene. protein (PRNP) mutation.

Tonsil biopsy Diagnostic criteria for nvCJD


Western blot analysis of tonsil material obtained by biopsy 1 (a) Progressive neuropsychiatric disorder.
of tonsil or lingual tonsillar remnants under local anesthetic (b) Duration of illness >6 months.
by means of a disposable tonsil-biopsy kit may allow (c) Routine investigations do not suggest an alternative
antemortem detection of prion protein expressed in the diagnosis.
lymphoreticular system. (d) No history of potential iatrogenic exposure.

Brain biopsy 2 (a) Early psychiatric symptoms.


Biopsy is mainly indicated to diagnose a suspected treatable (b) Persistent painful sensory symptoms.
cause of the clinical state, such as CNS vasculitis or SSPE. (c) Ataxia.
Rarely indicated to diagnose CJD because it: (d) Myoclonus or chorea or dystonia.
• Carries a small but definite risk of brain hemorrhage and (e) Dementia.
infection.
• May miss the diagnosis because of the patchy nature of 3 (a) EEG does not show the (typical) appearance of
prion immunostaining. classical CJD (or no EEG performed).
• Neurosurgic instruments have to be destroyed after biopsy (b) Posterior thalamic high signal on MRI brain scan
in all positive cases to prevent iatrogenic transmission. (407).

Neuropathology • Definite: 1(a) and neuropathologic confirmation of


The demonstration of proteinase-resistant PrP in brain is nvCJD.
the neuropathologic diagnostic marker. This can be • Probable: 1 and four-fifths of 2 and 3(a) and 3(b).
undertaken at post mortem by light microscopy of various • Possible: 1 and four-fifths of 2.
brain regions and antemortem, in some laboratories, by
immunohistochemistry with antibodies against the prion
protein and molecular genetic analyses of PrP performed on
DNA extracted from blood leukocytes (PrP Western blot
and/or preparation of scrapie-associated fibrils [SAF]) and
identifying a mutant PrP genotype or the presence of PrPSc.
Further Reading 335

TREATMENT PREVENTION
• At present, no effective curative therapy is available, • Avoid risk factors: hGH is now manufactured using
treatment is symptomatic. DNA recombinant technology, thereby eliminating the
• Patients should be nursed similarly to others with need for human sources.
infectious disease, using disposable pins and EMG and • Genetic counselling coupled with prenatal DNA
LP needles. screening is possible but the apparent incomplete
• Anti-epileptic drugs may be required for seizures, penetrance of some of the inherited prion diseases
nasogastric tube or percutaneous endoscopic gastrostomy increases the uncertainty of predicting the future for an
for feeding, intermittent or indwelling bladder catheters asymptomatic individual. Problems common to all
for urinary incontinence, and appropriate posturing and predictive testing programmes are also likely to arise.
regular turning to prevent bedsores.

CLINICAL COURSE AND PROGNOSIS


Rapid progressive decline to akinetic mutism and death over
2–12 months; about 10% have a protracted clinical course.

FURTHER READING Intracranial abscess Jeffery KJM, Read SJ, Peto TEA, Mayon-White
Arunkumar MJ, Rajsjekhar V, Chandy MJ, et al. RT, Bangham CRM (1997) Diagnosis of viral
(2000) Management and outcome of brain infections of the central nervous system: clin-
abscess in renal transplant recipients. Postgrad. ical interpretation of PCR results. Lancet, 349:
BACTERIAL INFECTIONS Med. J., 76: 207–211. 313–317.
Acute pyogenic (bacterial) meningitis Lipkin WI (1997) European consensus on viral
Begg N, Cartwright KAV, Cohen J, et al. (1999) Tetanus encephalitis. Lancet, 349: 299–300.
Consensus statement on diagnosis, investi- Farrar JJ, Yen LM, Cook T, et al. (2000) Tetanus. Pleasure SJ, Fischbein NJ (2000) Correlation of
gation, treatment and prevention of acute bac- J. Neurol. Neurosurg. Psychiatry, 69: 292–301. clinical and neuroimaging findings in a case of
terial meningitis in immunocompetent adults. Sanford JP (1995) Tetanus – forgotten but not rabies encephalitis. Arch. Neurol., 57:
J. Infect., 39: 1–15. gone. N. Engl. J. Med., 332: 812–813. 1765–1769.
Leen CLS (2000) Adjunctive therapy for bacteri- Solomon T, Dung NM, Kneen R, et al. (2000)
Whipple’s disease
al meningitis. Proc. R. Coll. Physicians Edin., Japanese encephalitis. J. Neurol. Neurosurg.
Dobbins WO (1995) The diagnosis of Whipple’s
30: 305–310. Psychiatry, 68: 405–415.
disease. N. Engl. J. Med., 332: 390–392.
Moller K, Skinhoj P (2000) Guidelines for man- Solomon T, Cardosa MJ (2000) Emerging ar-
Louis ED, Lynch T, Kaufmann P, Fahn S, Odel J
aging acute bacterial meningitis. BMJ, 320: boviral encephalitis. BMJ, 321: 1484–1485.
(1996) Diagnostic guidelines in central ner-
1290.
vous system Whipple’s disease. Ann. Neurol., Herpes simplex virus encephalitis
Quagliarello VJ, Scheld WM (1997) Treatment of
40: 561–568. Balfour HH (1999) Antiviral drugs. N. Engl. J.
bacterial meningitis. N. Engl. J. Med., 336:
Raoult D, Birg ML, La Scola B, et al. (2000) Cul- Med., 340: 1255–1268.
708–716.
tivation of the bacillus of Whipple’s disease. N. Cinque P, Cleator GM, Weber T, Monteyne P,
Rosenstein NE, Perking BA, Stephens DS, et al.
Engl. J. Med., 342: 620–625. Sindic CJ, van Loon AM, for the EU Con-
(2001) Meningococcal disease. N. Engl. J.
Ratnaike RN (2000) Whipple’s disease. Postgrad. certed Action on Virus Meningitis and En-
Med., 344: 1378–1388.
Med. J., 76: 760–766. cephalitis (1996) The role of laboratory inves-
Steigbigel NH (2001) Computed tomography of
Swartz MN (2000) Whipple’s disease – past, pre- tigation in the diagnosis and management of
the head before a lumbar puncture in suspect-
sent and future. N. Engl. J. Med., 342: suspected herpes simplex encephalitis: a con-
ed meningitis – is it helpful? N. Engl. J. Med.,
648–650. sensus report. J. Neurol. Neurosurg. Psychia-
345: 1768–1770
Verhagen WIM, Huygen PLM, Dalman JE try, 61: 339–345.
Tunkel AR, Scheld WM (1995) Acute bacterial
(1997) Central nervous system Whipple’s dis- DeBiasi RL, Tyler KL (1999) Polymerase chain re-
meningitis. Lancet, 346: 1675–1680.
ease. Ann. Neurol., 41: 560–561. action in the diagnosis and management of
Wood M (2000) Antimicrobial treatment of com-
munity-acquired meningitis in adults. Proc. R. central nervous system infections. Arch. Neu-
VIRAL INFECTIONS rol., 56: 1215–1219.
Coll. Physicians Edin., 30: 311–314.
Acute aseptic meningitis Lipkin WI (1997) European consensus on viral
Tuberculous meningo-encephalitis Attia J, Hatala R, Cook DJ, Wong JG (1999) encephalitis. Lancet, 349: 299–300.
Enarson DA (2000) Resistance to antituberculous Does this adult patient have acute meningitis?
medications. Hard lessons to learn. Arch. In- JAMA, 282: 175–181. Varicella-zoster virus encephalomyelitis
tern. Med., 160: 581–582. Moris G, Garcia-Monco JC (1999) The challenge Cohen JI, Brunell PA, Straus SE, Krause PR
Garg RK (1999) Tuberculosis of the central ner- of drug-induced aseptic meningitis. Arch. In- (1999) Recent advances in varicella-zoster
vous system. Postgrad. Med. J., 75: 133–140. tern. Med., 159: 1185–1194. virus infection. Ann. Intern. Med., 130:
Misra UK, Kalita J, Roy AK, et al. (2000) Role of 922–932.
Viral encephalitis Gilden DH, Kleinschmidt-DeMasters BK, La-
clinical, radiological, and neurophysiological
Balfour HH (1999) Antiviral drugs. N. Engl. J. Guardia JJ, Mahalingam R, Cohrs RJ (2000)
changes in predicting the outcome of tuber-
Med., 340: 1255–1268. Neurologic complications of the reactivation
culous meningitis: a multivariable analysis. J.
Davis LE (2000) Diagnosis and treatment of acute of varicella-zoster virus. N. Engl. J. Med., 342:
Neurol. Neurosurg. Psychiatry, 68: 300–303.
encephalitis. The Neurologist, 6: 145–159. 635–644.
Thwaites G, Chau TTH, Mai NTH, Drobniewski
DeBiasi RL, Tyler KL (1999) Polymerase chain re-
F, McAdam K, Farrar J (2000) Tuberculous
action in the diagnosis and management of
meningitis. J. Neurol. Neurosurg. Psychiatry,
central nervous system infections. Arch. Neu-
68: 289–299.
rol., 56: 1215–1219.
336 Infections of the Nervous System

Human immunodeficiency virus (HIV)- Progressive multifocal leukoencephalopathy PROTOZOAN INFECTIONS


associated cognitive/motor complex Berger JR, Concha M (1995) Progressive multi- Toxoplasmic encephalitis
Carr A, Cooper DA (2000) Adverse effects of an- focal leucoencephalopathy: the evolution of a Report of the Quality Standards Subcommittee of
tiretroviral therapy. Lancet, 356: 1423–1430. disease once considered rare. J. Neurovirol., 1: the American Academy of Neurology (1998)
Chang L, Ernst T, Leonido-Yee M, et al. (1999) 5–18. Evaluation and management of intracranial
Highly active antiretroviral therapy reverses Clifford DB, Yiannoutsos C, Glicksman M, et al. mass lesions in AIDS. Neurology, 50: 21–26.
brain metabolite abnormalities in mild HIV (1999) HAART improves prognosis in HIV-
dementia. Neurology, 53: 782–789. associated progressive multifocal leuko- WORM INFECTIONS
D’Souza MP, Cairns JS, Plaeger SF (2000) Cur- encephalopathy. Neurology, 52: 623–625. Cysticercosis
rent evidence and future directions for target- Dolin R (1999) Gaining ground in progressive Garg RK (1998) Neurocysticercosis. Postgrad.
ing HIV entry. Therapeutic and prophylactic multifocal leukoencephalopathy. Neurology, Med. J., 74: 321–326.
strategies. JAMA, 284: 215–222. 52: 440–441. Proanao JV, Madrazo I, Avelar F, et al. (2001)
Freedberg KA, Losina E, Weinstein MC, et al. Manji H, Miller RF (2000) Progressive multifocal Medical treatment for neurocysticercosis char-
(2001) The cost effectiveness of combination leucoencephalopathy: progress in the AIDS acterized by giant subarachnoid cysts. N. Engl.
antiretroviral therapy for HIV disease. N. Engl. era. J. Neurol. Neurosurg. Psychiatry., 69: J. Med., 345: 879–885.
J. Med., 344: 824–831. 569–571.
Geraci AP, Di Rocco A, Simpson DM (2001) PRION INFECTIONS
Neurological complications of AIDS. The Neu- Subacute sclerosing panencephalitis Creutzfeldt-Jacob disease
rologist, 7: 82–97. Anlar A, Saatci I, Kose G, Yalaz K (1996) MRI Brown P (2001) Bovine spongiform en-
Gulick RM (2000) Assessing the benefits of anti- findings in subacute sclerosing panencephali- cephalopathy and variant Creutzfeldt-Jakob
retroviral therapy. Ann. Intern. Med., 133: tis. Neurology, 47: 1278–1283. disease. BMJ, 322: 841–844.
471–473. Haywood AM (1997) Transmissible spongiform
Lederman MM, Valdez H (2000) Immune Poliomyelitis encephalopathies. N. Engl. J. Med. 337:
restoration with antiretroviral therapies. Im- Kidd D, Williams AJ, Howard RS (1996) Po- 1821–1828.
plications for clinical management. JAMA, liomyelitis. Postgrad. Med. J., 72: 641–647. Knight R (1999) The relationship between new
284: 223–228. Mulder DW, Rosenbaum RA, Layton DD Jr variant Creutzfeldt-Jakob disease and bovine
Montaner JSG, Hogg R, Raboud J, et al. (1998) (1972) Late progression of poliomyelitis or spongiform encephalopathy. Vox. Sang. 76:
Antiretroviral treatment in 1998. Lancet, 352: forme fruste amyotrophic lateral sclerosis? 203–208.
1919–1922. Mayo Clin. Proc., 47: 756–761. Petersen RB (1999) Antemortem diagnosis of
Piscitelli SC, Gallicano KD (2001) Interactions Windebank AJ, Litchy WJ, Daube JR, Iverson RA variant Creutzfeldt-Jakob disease. Lancet, 353:
among drugs for HIV and opportunistic infec- (1996) Lack of progression of neurologic 163–164.
tions. N. Engl. J. Med., 344: 984–996. deficit in survivors of paralytic polio: A 5-year Prusiner SB (2001) Shattuck Lecture – Neurode-
Price RW (1996) Neurological complications of prospective population-based study. Neurolo- generative diseases and prions. N. Engl. J. Med.,
HIV infection. Lancet, 348: 445–452. gy, 46: 80–84. 344: 1516–1526.
Sacktor NC, Skolasky RL, Lyles RH, et al. (1998) Venters GA (2001) New variant Creutzfeldt-Jakob
Highly active anti-retroviral therapy (HAART) SPIROCHETE INFECTIONS disease: the epidemic that never was. BMJ, 323:
improves cognitive impairment in HIV+ ho- Neurosyphilis 858–861.
mosexual men. J. Neurovirol., 4: 365. del Mar Saez de Ocariz M, Nader JA, Del Brutto Will RG (1999) Prion related disorders. J. R. Coll.
Sactor N, Lyles RH, Skolasky R, et al. (2001) OH, Santos Zambrano JA (1996) Cere- Physicians Lond. 33: 311–315.
HIV-associated neurological disease incidence brovascular complications of neurosyphilis: Will RG, Zeidler M, Stewart GE, et al. (2000) Di-
changes: multicenter AIDS cohort study, The return of an old problem. Cerebrovasc. agnosis of new variant Creutzfeldt-Jakob dis-
1990–1998. Neurology, 56: 257–260. Dis., 6: 195–201. ease. Ann. Neurol., 47: 575–582.
Stankoff B, Tourbah A, Suarez S, et al. (2001) Zeidler M, Sellar RJ, Collie DA, et al. (2000) The
Lyme disease
Clinical and spectroscopic improvement in pulvinar sign on magnetic resonance imaging
Garcia-Monco JC, Benach JL (1995) Lyme neu-
HIV-associated cognitive impairment. Neu- in variant Creutzfeldt-Jakob disease. Lancet,
roborreliosis. Ann. Neurol., 37: 691–702.
rology, 56: 112–115. 355: 1412–1418.
Prasad A, Sankar D (1999) Overdiagnosis and
Steinbrook R (2001) Providing antiretroviral ther- Zerr I, Schulz-Schaeffer WJ, Giese A, et al. (2000)
overtreatment of Lyme neuroborreliosis are
apy for HIV infection. N. Engl. J. Med., 344: Current clinical diagnosis in Creutzfeldt-Jakob
preventable. Postgrad. Med. J., 75: 650–656.
844–846. disease: identification of uncommon variants.
Steere AC (2001) Lyme disease. N. Engl. J. Med.,
Sterling TR, Vlahov D, Astemborski J, et al. Ann. Neurol., 48: 323–329.
345: 115–125.
(2001) Initial plasma HIV-1 RNA levels and
progression to AIDS in women and men. N. FUNGAL INFECTIONS
Engl. J. Med., 344: 720–725. Mucormycosis
Temesgen Z, Wright AJ (1997) Recent advances Jaitly R, Dhaduk N, Jensen ME, et al. (2000) Pri-
in the management of human immunodefi- mary cerebral mucormycosis: a case report and
ciency virus infection. Mayo Clin. Proc., 72: literature review. The Neurologist, 6: 232–237.
854–859.
Chapter Twelve 337

Inflammatory
Disorders of the
Nervous System
ACUTE DISSEMINATED EPIDEMIOLOGY
ENCEPHALOMYELITIS • Incidence: uncommon; most frequent after non-specific
(POST INFECTIOUS upper respiratory tract infections of undetermined
ENCEPHALOMYELITIS) (ADEM) etiology.
• Age: any age.
• Gender: either sex.
DEFINITION
An acute inflammatory demyelinating disease of the brain PATHOLOGY
and spinal cord characterized by widespread perivascular Macroscopic
inflammation and demyelination and caused by an The brain is congested and swollen (412).
autoimmune attack on the brain, most commonly as a result
of a viral infection which activates autoreactive T cells that Microscopic
recognize myelin-specific proteins. The disease is one of Perivascular inflammation (macrophages, plasma cells, and
immunoregulatory failure rather than immunosuppression. T lymphocytes) and demyelination of the white matter tracts
of the cerebral hemispheres, brainstem, spinal cord and optic
nerves.

ETIOLOGY
412 Viral infection
Exanthematous
• Measles: complicates 1 in 1000 cases of measles; in
countries where vaccination is not routine.
• Varicella-zoster: <1 : 10 000 (more commonly causes
acute ataxia).

Non-exanthematous
• Influenza A upper respiratory tract infection.
• Mumps.
• Epstein–Barr virus.
• Rubella: <1 : 20 000 (more commonly causes a toxic
encephalopathy).

Bacterial infection
• Mycoplasma pneumoniae.

Immunization
• Vaccination (vaccinia, rabies).
• Tetanus antitoxin.

No recognizable preceding illness sometimes

412 Ventral surface of a swollen, hemorrhagic brain affected by acute


hemorrhagic encephalitis.
338 Inflammatory Disorders of the Nervous System

PATHOPHYSIOLOGY Clinical
ADEM is thought to be an autoimmune disease, partly • Latent period after the initial illness ranges from 1–20
because of the inability to isolate an infectious agent from the days but, unlike ADEM, tends to last just a few days or
CNS, and because of experimental models of autoimmune may even be unnoticeable.
diffuse white matter encephalomyelitis that can be induced in • Fever (temperature up to 42°C) and malaise.
animals, or accidentally in humans, by injection of the myelin • Mild neck rigidity.
antigens, myelin basic protein, proteolipid protein, or myelin • Progressive focal neurologic signs and raised intracranial
oligodendrocyte glycoprotein. A very small inoculum of pressure.
activated T cell clones that recognize small fragments of
myelin basic protein and proteolipid protein can induce CNS Laboratory
inflammation and destruction of normal brain white matter • Peripheral blood neutrophil pleocytosis and high ESR.
by the immune system. The disease can range from acute • CT/MRI scan: abnormal; brain swelling, similar to
hemorrhagic leukoencephalitis with massive fibrin deposition ADEM, but with much more extensive lesions, and a
and a substantial neutrophilic hemorrhagic lesion to a chronic hemorrhagic component.
demyelinating disorder with a lymphocytic infiltrate, which is • CSF:
similar to multiple sclerosis. – High pressure (>300 mm water).
It is believed that a viral infection activates circulating – Cells: neutrophil pleocytosis (may be >1000); hundreds
autoreactive T cells that recognize myelin-specific proteins, of red cells.
and these T cells migrate into the CNS and recruit neutrophils – Protein: raised.
in a bystander fashion, triggering massive multifocal tissue – Glucose levels: normal.
destruction. The mechanism of T cell activation is unknown.
Prognosis
CLINICAL FEATURES Rapid clinical course (more rapid than ADEM), progressing
• Preceding upper respiratory tract or gastrointestinal tract to delirium and coma; patients are more likely to die.
infection is common.
• Latent interval between the acute viral illness and the Diagnosis
onset of neurologic symptoms. Diagnosed at autopsy in most cases.
• Acute/subacute onset.
• Fever, headache, malaise. DIFFERENTIAL DIAGNOSIS
• Skin rash. • Viral encephalitis (see p.292): direct infection of the CNS
• Progressive focal neurologic signs: (e.g. by arbovirus, herpes simplex virus, cytomegalovirus)
– Hemiparesis or paraparesis. rather than activation of autoreactive T cells by the
– Sensory defects, depending on location of lesions in brain primary infection. The presence of a persistent
and spinal cord. neutrophilic instead of lymphocytic CSF pleocytosis in
– Ataxia. ADEM is against a direct viral infection of the CNS.
– Optic neuritis. • Leptospirosis meningoencephalitis: a biphasic illness, with
– Other cranial nerve palsies. a prodrome of chills and conjunctival suffusion and the
• Raised intracranial pressure: presence of leptospiras in the blood and CSF. In the
– Headache. second phase, many develop neurologic complications
– Vomiting. with a neutrophilic pleocytosis in CSF.
– Papilledema. • Lyme disease (see p.316).
– Obtundation. • Brain abscess (see p.284).
• Brain tumor (see p.357).
SPECIAL FORMS • Multiple sclerosis (see p.340): may have the same under-
Acute hemorrhagic leukoencephalitis is lying pathophysiology and clinical presentation but has
• A more aggressive form of ADEM. a chronic relapsing and remitting, or progressive course.
• Meningitis: viral, bacterial, tuberculous, cryptococcal.
Epidemiology • Stroke (see p.192): massive carotid territory infarction
• Affects children and adults and both sexes equally. with temporal lobe swelling compressing the posterior
cerebral artery against the brainstem and causing
Pathology additional posterior infarction.
• Swollen hemorrhagic brain with herniation. • Transverse myelitis (see p.561): Mycoplasma pneumoniae.
• Widespread necrotizing hemorrhagic lesions in the white
matter and brainstem. INVESTIGATIONS
• Perivascular neutrophilic infiltrate with varying numbers CT brain scan
of lymphocytes. Scans show diffuse low attenuation throughout the gray and
• Blood vessels impregnated with fibrin (fibrinoid necrosis). white matter of one or both hemispheres with mass effect
• Widespread demyelination, usually in regular patches, (midline shift, subfalcine herniation, uncal herniation,
and around areas of hemorrhage. effacement of basilar cisterns, entrapment of lateral
• Gray matter is also frequently involved. ventricles) but may be normal.
Acute Disseminated Encephalomyelitis (Post Infectious Encephalomyelitis) (ADEM) 339

MRI brain scan PROGNOSIS


• MRI is more sensitive than CT. Some patients make remarkable recoveries, even after
• Areas of increased signal on T2WI in the white matter prolonged periods of profound coma, so very aggressive
are present which may be quite extensive and enhance supportive therapy should always be entertained.
with contrast (413).
• The lesions occur mostly in the cerebrum, but are also Major sequelae
found in the brainstem and cerebellum and spinal cord. • Measles: frequent.
When they predominate in the latter, they may mimic • Rubella: rare.
spinal cord or brainstem tumor. • Vaccinia: 10%.
• The lesions may not resolve completely, and result in • Varicella: very rare.
diffuse atrophy.
• No new lesions should appear on MRI after 6 months Case fatality rate
from the start of the disease. • Measles: 20%.
• Rubella: 20%.
CSF • Vaccinia: 10%.
CSF is normal in about one-third of patients. Two-thirds • Varicella: 5%.
show a mild mononuclear cell pleocytosis and protein
elevation. Viral and bacterial culture, PCR for viral DNA,
myelin basic protein (which may be found by
radioimmunoassay), and immunoglobulin G (which may be
elevated) are indicated.

Blood
• Full blood count and ESR.
• Blood biochemistry.
• Blood viral serology: acute and convalescent sera may
establish the specific infecting agent but usually are not
of help in differentiating acute encephalitis caused by
direct infection from encephalitis caused by immune-
mediated perivenular demyelination.
413
Other
• Chest x-ray.
• Throat and rectal swabs.
• Urinalysis.
• EEG: abnormal: non-specific diffuse slow wave activity.

DIAGNOSIS
• There are no consistent abnormalities in the blood or
urine.
• A definitive diagnosis requires pathologic examination.

TREATMENT
Prevention
Vaccination: the cessation of immunization with vaccinia
virus and the introduction of vaccines for measles, mumps
and rubella viruses has proved highly effective.

Acute treatment
• Supportive care: lowering temperature with antipyretic
agents, maintaining an adequate fluid intake, treating
epileptic seizures if they develop and reducing intracranial
pressure if raised.
• There is no conclusive evidence that hyperimmune
gamma globulin, corticosteroids or adrenocorticotropic
hormone (ACTH) are of benefit (or no benefit).

413 T2W MRI from a young patient who became confused and
neurologically unwell about 2 weeks after a dose of chickenpox. Note
the extensive white matter and basal ganglia increased signal which also
extended into the brainstem.
340 Inflammatory Disorders of the Nervous System

MULTIPLE SCLEROSIS (MS) Epidemiologic evidence points to environmental factors


at a young age:
• Prevalence of MS increases with increasing latitude.
DEFINITION • Risk of MS for an individual corresponds to the risk of
A chronic autoimmune inflammatory demyelinating disease their area of residence before the age of about 15 years.
of the CNS in which the lesions are disseminated in time
and space (different sites in the CNS are affected at different Genetic factors
points in time). Also likely to be relevant:
• A family history of MS is present in about 10% of people
EPIDEMIOLOGY with MS.
• Prevalence: higher in temperate climates further from the • The concordance rate for MS in monozygotic twins is
equator (e.g. Hobart, Tasmania, Australia, 76/100 000); 25%, 2.5% in dizygotic twins and 1.9% in non-twin
lower in tropical and subtropical climates close to the siblings.
equator (e.g. tropical Queensland, Australia, 12/100 000). • HLA associations with MS:
• Age: rare before puberty and beyond 60 years of age. – HLA-A3, HLA-D7, and HLA-DR2 common in
• Age of onset: <20 years: 20% of cases; 20–50 years: Caucasians with MS (HLA-DR2: present in >60% of
50–60% of cases; >50 years: 20–30% of cases. people with MS and 15–20% of controls).
• Gender: F>M (2 : 1). – HLA-DR4 common in Arabs with MS.
• Race: Northern European ancestry most common;
uncommon in Australian aboriginals, Maori, Chinese, PATHOPHYSIOLOGY
Japanese, black African. Exposure to an unidentified non-self antigen that ‘mimics’
constitutive peptides of myelin evokes an antigen-specific,
PATHOLOGY T cell mediated immune response. Lymphocytes, macro-
Multiple plaques of demyelination in the white matter of phages and humoral factors enter the CNS and the
the CNS. blood–brain barrier breaks down. B-lymphocytes produce
oligoclonal immunoglobulin G (IgG) in the CSF. Sensitized
Acute lesion T cells produce cytokines which may damage
• Demyelination of nerve fibers in the white matter of the oligodendrocytes and myelin. Nerve conduction is blocked
CNS. in demyelinated axons and can be restored by remyelination.
• Loss of oligodendrocytes. In contrast, axonal loss leads to a permanent loss of
• Perivenular infiltration of T and B lymphocytes, neurologic function, as the CNS axonal regenerative
macrophages (filled with myelin debris) and plasma cells. capacity is severely limited.
• Secondary axonal degeneration.

Chronic lesion
• Some axonal loss and remyelination.
• Glial cell proliferation resulting in discrete gray colored 414
areas of gliosis (or sclerosis) that are called plaques.

Sites of demyelination
Any part of the CNS, particularly:
• Periventricular white matter of the cerebral hemispheres
(414–416).
• Optic nerves (417).
• Cerebellum.
• Brainstem.
• Spinal cord (particularly subpial regions of the spinal
cord) (418, 419).

The peripheral nervous system is not affected.

ETIOLOGY
• Unknown, but likely to be a misdirected autoimmune
disease (because of the predilection of women, the
human leukocyte antigen (HLA) association, the
relapsing and remitting course, and the finding of
immunologically active cells in the brain, spinal cord and
CSF). This, and other evidence, suggests that MS is an
autoimmune disease resulting from an immune attack on
the myelin sheaths and axons in the CNS by autoreactive
T lymphocytes and autoantibodies. 414 Section of one cerebral hemisphere (parietal lobe) showing
• Environmental factors, such as a virus infection (e.g. a lack of staining due to periventricular demyelination in the white
herpes virus-6), may trigger immune-mediated matter. (Courtesy of Professor BA Kakulas, Royal Perth Hospital,
demyelination in genetically predisposed individuals. Western Australia.)
Multiple Sclerosis (MS) 341

CLINICAL FEATURES Nature


Onset The symptoms usually reflect dysfunction of the optic
• Subacute onset of neurologic symptoms over several nerves, brainstem or spinal cord.
hours to days. Infrequently the symptoms evolve quickly
over minutes or slowly over weeks or months. Reduced visual acuity
• The onset is monosymptomatic in about 50% of cases. Reduced visual acuity, which varies from a slight dulling of
The remainder have initial symptoms of multiple lesions color vision to complete monocular blindness, together with
within the CNS.

416

415, 416 Autopsy specimens of brain, coronal sections, showing


periventricular demyelination (arrows).

415

417 418

417 Optic neuritis.Transverse section of the optic nerve at autopsy, 419


showing demyelination of the optic nerve in a patient with optic
neuritis due to multiple sclerosis.

418 Transverse section of the thoracic spinal cord showing a large


plaque of demyelination in the dorsal columns (arrow) in a multiple
sclerosis patient who had a high stepping gait due to sensory ataxia in
the lower limbs. (Courtesy of Professor BA Kakulas, Royal Perth
Hospital,Western Australia.)

419 Transverse section of the spinal cord showing a large plaque of


demyelination (arrows) in almost one-half of the spinal cord in a
multiple sclerosis patient with a Brown–Sequard syndrome. (Courtesy
of Professor BA Kakulas, Royal Perth Hospital,Western Australia.)
342 Inflammatory Disorders of the Nervous System

pain around the eye that is exacerbated by eye movement or (caused at least in part by detrusor sphincter dyssynergia),
touching the eye, are symptoms of optic neuritis (see p.489). failure to store, or a combination of both. Frequency,
The signs include a central or paracentral scotoma in most urgency and precipitancy of micturition (85%), urge incon-
patients, particularly using a red target (most of the optic tinence (63%), hesitancy and interrupted stream (45%) and
nerve fibers transmit information from the macular), and a retention of urine may be early symptoms of spinal cord
swollen optic disc (papillitis) in the acute phase if there is demyelination and are very common in later stages. Bowel
demyelination of the anterior part of the optic nerve (420). dysfunction occurs in more than half of patients. Impotence
Optic disc pallor due to optic atrophy ensues later (421). is also common.

Weakness in one or more limbs or the face Mental changes


Weakness in one or more limbs or the face due to corticospinal After several years, many patients experience emotional
tract demyelination: may be described as heaviness, tiredness instability, anxiety, depression, euphoria and mildly impaired
or stiffness, dragging of one leg, or a tendency to fall. cognitive function (short-term memory, attention, and
speed of processing).
Altered sensation of the face, trunk or one or more limbs
• Trigeminal neuralgia (tic douloureux) (see p.509) (brain- Fatigue
stem). Many patients with long standing MS complain of fatigue.
• Lhermitte’s symptom: electric shock-like sensation down
the back on flexing the neck: (cervical spinal cord). Pain
• Numbness starting in the feet and spreading up to the Occurs in up to two-thirds of patients. In most it is chronic,
waist: (cervical or thoracic cord). but in about 10% it is acute and paroxysmal. Examples
• Reduced temperature sensation in a warm bath/shower include trigeminal neuralgia; brief (1 minute) dysesthetic
or when swimming: (spinothalamic tract). burning pain in one or more extremities that is provoked by
• Feeling of a tight band wrapped around the trunk or movement, tactile stimulation, or hyperventilation; and
limbs, or swelling of limbs: (posterior columns). painful tonic seizures. The latter can occur with or imme-
• Clumsy or functionally useless hand despite normal diately after the dysesthetic burning pain, or independently.
power and coordination: (posterior columns and dorsal They are brief, frequent and often intensely painful episodes
root entry zone). in which the limbs on one side may adopt a tetanic posture.
They may be precipitated by movement or sensory
Unsteady gait stimulation and usually remit completely after 4–6 weeks.
Weakness and spasticity, proprioceptive loss (sensory ataxia), The three main types of chronic pain are dysesthetic
spinocerebellar dysfunction, vestibular dysfunction, reduced extremity pain, chronic back pain and painful leg spasms.
or double vision, and other comorbidities such as pain and
arthritis. Epileptic seizures
Epileptic seizures may occur but are uncommon.
Double vision
Internuclear ophthalmoplegia due to lesion of the medial Precipitating and exacerbating factors
longitudinal fasciculus (MLF), which connects the nuclei of • Trauma (including surgery), infections, vaccinations,
cranial nerves III and VI (422). Elicited by asking the emotional stress, and fatigue may possibly precipitate
patient to look laterally to one side and noticing a slower attacks of MS.
rate of adduction, or a failure of adduction, of the adducting • Pregnancy and the early post partum period can mildly
eye (ipsilateral to the lesion) and horizontal jerk nystagmus increase the risk of exacerbation of symptoms of MS but
of the fully abducting eye. Subtle forms can be detected if the long term outcome is not affected.
the examiner watches the patient from the side, in the • Physical exercise (Uhthoff’s symptom), an increase in body
direction of attempted lateral gaze (i.e. not ‘front-on’) , and temperature (i.e. hot weather, hot baths), increased illu-
following the visual axes during rapid voluntary eye mination, eating, drinking, smoking and menstruation may
movements (saccades) from one side to the other and back; exacerbate neurologic symptoms. The pathophysiology of
and then watching from the other side. The lesion may be Uhthoff’s symptom is unknown, although a reversible
unilateral (which is more commonly due to cerebrovascular conduction block in demyelinated nerve fibers secondary to
disease than demyelination) or bilateral (which is more an increase in body temperature or to changes in blood
commonly due to demyelination). With rostral MLF lesions, electrolyte levels or pH is believed to play a role.
near the IIIrd nerve nucleus, vergence (ability to converge • Immunization can precipitate relapses of MS, so the
the eyes) may be impaired, whereas with caudal MLF potential benefits and risks of immunization need to be
lesions, near the VIth nerve nucleus, vergence is preserved. carefully considered.

Vertigo HISTORY
Sensation of rotation or spinning, causing nausea and ataxia • Establish the onset and nature of the neurologic
(intra-axial vestibular nerve, vestibular nucleus in the lateral symptoms, any associated or exacerbating factors, and
medulla, and pathways from the vestibular nucleus to the the clinical course.
vestibular cortex), rarely in isolation and often occurring • Enquire about previous neurologic symptoms such as
together with other brainstem symptoms. blurred vision, blindness, double vision, weakness, altered
feeling (numbness, tingling), and disturbances of bladder
Sphincter and sexual disturbances function.
Bladder dysfunction may be divided into a failure to empty
Multiple Sclerosis (MS) 343

PHYSICAL EXAMINATION Bacteria (brucellosis, chlamydia, tularemia), and myco-


• Assess the presenting neurologic impairments and bacteria (tuberculosis, atypical mycobacteria).
functional disabilities (e.g. spastic paraparesis). Fungi (histoplasmosis, coccidioidomycosis, cryptococcosus).
• Search for other ‘silent’ neurologic signs of previous Spirochetes (treponemal infections such as syphilis).
subclinical demyelination (e.g. optic atrophy, internuclear Parasites (toxoplasmosis, leishmaniasis).
ophthalmoplegia). – Occupational and environmental exposure to organic or
inorganic agents (e.g. methotrexate, talc, metals).
DIFFERENTIAL DIAGNOSIS – Neoplasia: lymphoma (e.g. intravascular lymphoma).
Multifocal neurologic syndrome – Autoimmune disorders (Wegener’s granulomatosis,
• Inherited ataxias (see pp.437, 441). Churg–Strauss syndrome).
• Vitamin B12 deficiency (see p.463). • Infection:
• Vasculitis: – Acute post infectious encephalomyelitis.
– Infection: Borrelia burgdorferi (Lyme disease), meningo- – Subacute leukoencephalitis cause by human herpes virus-6.
vascular syphilis. – Progressive multifocal leukoencephalopathy (JC
– Isolated granulomatous angiitis of the CNS. papovavirus).
– Polyarteritis nodosa. • Leukodystrophy.
– Systemic lupus erythematosus (facial rash, arthritis, • Brain, foramen magnum or spinal arteriovenous
pericarditis, pleuritis). malformation or tumor(s) (primary or metastatic).
– Behçet’s disease. • Cervical spondylitic myeloradiculopathy.
• Granulomatous inflammation: • Tethered spinal cord (see Spinal dysraphism, p.139).
– Sarcoidosis. • Multiple emboli to the CNS.
– Infection: • Multiple pathologies.

420 421

420 Optic neuritis. MRI orbits, axial plane, of the optic nerve, showing 421 Optic atrophy.
swelling of the optic nerve (arrow) in a patient with optic neuritis due
to multiple sclerosis.

422

422 Bilateral internuclear ophthalmoplegia.


344 Inflammatory Disorders of the Nervous System

Optic neuropathy endemic regions or with other risk factors (e.g. Afro-
• Leber’s hereditary optic neuropathy: a maternally Caribbeans). A progressive spastic paraplegia develops
inherited disease, usually leading to severe bilateral visual over a number of years. However, sphincter disturbance
loss, and associated with several mitochondrial DNA is the rule and considerable neuropathic lower limb pain
point mutations; the major ones at nucleotide positions is common.
11 778, 3460, and 14 484 (see p.491). • Tuberculosis.
• Other hereditary optic neuropathies. • Syphilis.
• Ischemic optic neuropathy. • Toxoplasmosis.
• Neurosyphilis. • Schistosomiasis.

Acute non-compressive spinal cord syndrome Intramedullary tumor of the spinal cord
Vascular • Astrocytoma.
• Anterior spinal artery infarction: paraparesis with loss of • Ependymoma.
pain and temperature sensation, develops over minutes • Lymphoma.
to hours, and usually persists if infarction occurs. • Lipoma.
• Intramedullary hemorrhage. • Hemangioma.
• Metastases.
Inflammation of the spinal cord
• MS: usually a partial cord syndrome (e.g. unilateral loss Metabolic
of pain and temperature, deafferentation of one limb, or • Vitamin B12 deficiency: presents over weeks or months as
an asymmetric incomplete paraparesis) develops over a subacute spinal cord syndrome with intense paresthesia
hours to days with partial or complete recovery over and a combination of pyramidal and dorsal column signs.
several weeks. • Adrenomyeloneuropathy: an X-linked inherited disorder
• Transverse myelitis: complete loss of sensory and motor that affects males and some heterozygous females with a
function below the level of the lesion, resulting in a progressive myelopathy. A peripheral neuropathy and
flaccid, areflexic paraplegia; develops over hours to days, adrenal insufficiency may be present.
commonly after an infection (e.g. upper respiratory
tract). Toxic/iatrogenic
• Acute necrotizing myelitis: tuberculosis, lymphoma, • Radiation myelopathy (see p.565): steadily progressive
carcinoma. spastic paraplegia, months to years after radiotherapy.
• Connective tissue disease: systemic lupus erythematosus. Pathologically there is necrosis of the irradiated cord
• Sarcoidosis. segments with obliterative changes in blood vessels in the
same region.
Infection of the spinal cord • Lathyrism: endemic in parts of India and presents as a
• Herpes zoster. subacute or chronic spastic paraparesis in people who
• Herpes simplex types I and II. regularly ingest chickling pea vetch over several months.
• HIV. It is thought to be caused by a toxin in the chickling pea.
• Tuberculosis.
• Syphilis. Degenerative
• Syringomyelia (see p.541).
Chronic non-compressive spinal cord syndrome • Motor neuron disease (see p.534): purely motor; usually
Inherited a combination of lower and upper motor neuron signs.
Hereditary spastic paraparesis: usually a family history of
autosomal dominant inheritance. INVESTIGATIONS
MRI of the brain
Vascular MRI is the imaging investigation of choice. T2W images
Dural arteriovenous malformation: the most common type show:
of spinal angioma. Usually affects the thoracolumbar • Areas of increased signal (brightness) in the white matter
segments and tends to present in middle-aged men as a which can be anywhere in the brain but are typically seen
chronic progressive myelopathy with symptoms that may in the immediate periventricular white matter and corpus
fluctuate or be aggravated by exercise. A combination of callosum (423–428).
upper and lower motor neuron signs may be present. • The areas of brightness can be of varying size, may show
some swelling, and may be multiple or only a few.
Inflammation of the spinal cord • A small proportion of patients with definite MS will have
• MS. a normal MR.
• Sarcoidosis. • It is important to differentiate other causes of bright
spots which are non-pathologic from MS plaques, for
Infection of the spinal cord example enlarged perivascular spaces which are usually
• Herpetic necrotizing myelitis. small. Normal young persons are allowed to have up to
• Cytomegalovirus. three white spots (if small).
• Varicella-zoster granulomatous myelitis.
• Human T lymphocyte virus-1 (HTLV-1) associated T1W images show:
myelopathy (‘tropical spastic paraparesis’) in adults from • Low signal areas (dark spots), but the T1W image is not
so sensitive and demonstrates fewer lesions than T2W.
Multiple Sclerosis (MS) 345

423, 424 MRI of brain in the 423 424


sagittal plane, proton density
image (423), and axial plane
T2W image (424), showing
multifocal areas of high signal
intensity adjacent to the
corpus callosum (423) and
lateral ventricles (424) due
to demyelination (arrows).

425 T2W MRI showing 425 426


multiple areas of increased
signal in the periventricular
white matter. The involvement
of the corpus callosum (lesions
right down at the top of the
lateral ventricles) is said to be
characteristic of MS.

426 T1W MRI following


contrast (same patient) shows
that some of the lesions
enhance (arrows) and some
do not, indicating that they are
of different ages and confirming
the most likely diagnosis to
be MS.

427, 428 MRI cervical spine,T2W image, 427 428


in the axial plane (427) and sagittal plane
(428), showing a focal area of high signal
intensity in the high left cervical spinal cord
posteriorly (arrows) due to demyelination,
in a patient who presented with symptoms
and signs of left dorsal column dysfunction
(ascending tight feeling in left leg like a
stocking around it).
346 Inflammatory Disorders of the Nervous System

• To differentiate MS from other causes of white matter • Viral serology and culture.
bright spots it is sometimes necessary to give intravenous • VDRL, TPHA.
contrast; MS will show patchy enhancement of some
abnormal areas reflecting its typical multiphasic course. Electrophysiologic studies
Other diseases such as ADEM (see p.337) are monophasic At least 90% of patients with clinically definite MS have a
so all the abnormal areas should either enhance or not. persistent abnormality detected by visual, auditory or
• Differential diagnosis: the appearance of MS on MR somatosensory evoked potentials, but the utility of these
while typical, is non-specific. A similar picture can occur studies in identifying other sites of previous subclinical
in other conditions (see below). demyelination has been superseded to a large extent by MRI
of the brain and spinal cord:
Differential diagnosis of brain MRI mimicking MS • Visual evoked potentials: delayed conduction in about
• Subcortical arteriosclerotic encephalopathy or 90% of clinically definite MS.
Binswanger’s disease. • Brainstem auditory evoked potentials: abnormal in half
• Multiple metastases. of clinically definite MS.
• Vasculitis. • Somatosensory evoked potentials: abnormal in 70% with
• Sarcoidosis. clinically definite MS.
• Leukodystrophies.
• Encephalitis: EEG
– Viral: HIV, progressive multifocal leukoencephalopathy, If considering a diagnosis of encephalitis.
subacute sclerosing panencephalitis (measles), acute
disseminated encephalomyelitis. DIAGNOSIS
– Bacterial: tuberculosis. • Clinical and MRI evidence of at least two CNS lesions
Spirochetal: syphilis, neuroborreliosis, or Lyme disease. that are consistent with demyelination occurring at
• Alzheimer’s disease. different sites in the CNS and at different times.
• Radiation therapy. • The diagnosis can be classified according to the degree
• Chemotherapy. of certainty as clinically ‘definite’, ‘probable’ or ‘possible’.
• Cyclosporine use.
• Hyperperfusion syndrome. N.B. MS should only be considered if all of the symptoms
• Chronic inflammatory demyelinating polyneuropathy. and signs cannot be explained by a single neurologic lesion
• Subacute combined degeneration of the spinal cord and the history, examination and special investigations fail to
(vitamin B12 deficiency). identify other conditions that can cause multiple CNS lesions.

CT scan of the brain DIAGNOSTIC CRITERIA FOR MS FOR RESEARCH


• May demonstrate plaques of demyelination, particularly PURPOSES (POSER ET AL., 1983)
after double-dose contrast, but it is non-specific and Clinically definite MS
rather insensitive, so is not recommended. 1 Two attacks and clinical evidence of two separate lesions, or
• Main role is to exclude other differential diagnoses such 2 Two attacks; clinical evidence of one lesion and
as tumors. paraclinical* evidence of another, separate lesion.

CSF The two attacks must involve different parts of the CNS,
• Cell count: increased (5–50 lymphocytes/mm3) in two- must be separated by a period of at least 1 month, and must
thirds of patients during an acute attack; normal each last a minimum of 24 hours.
(<3–4 cells/mm3) in two-thirds of patients in remission.
• Protein: mildly elevated, up to 1.0 g/l (100 mg/dl), in *Paraclinical evidence of a lesion: the demonstration by means
about one-third of patients. of various tests and procedures of the existence of a lesion of
• IgG/albumin ratio: raised (>25%), in two-thirds of the CNS which has not produced signs of neurologic
clinically definite MS. dysfunction but which may or may not have caused symptoms
• IgG index (compares IgG/albumin ratio in the CSF and in the past. Such tests and procedures include the hot bath
blood): abnormal in about 90% of patients with clinically test, evoked response studies, tissue imaging procedures
definite MS. (including MRI), and reliable, expert neurologic assessment.
• Oligoclonal IgG bands (429): present in about 90% of
patients with clinically definite MS; not specific; found in
other immune-mediated CNS diseases. 429

429 Isoelectric focusing in an agarose gel at stable pH (range 5.0–9.5)


demonstrating the presence of oligoclonal bands (arrows) in the CSF
(top) and not the serum (bottom).The oligoclonal bands are different
clones of IgG that have migrated electrophoretically in the stationary
pH gradient until a steady state is reached when all the components
are concentrated or focused as sharp bands at their respective
isoelectric points.The bands are visualized by immunofixation.To be
positive, two or more oligoclonal bands must be detected in the CSF
that are not present in the serum of the patient.
Multiple Sclerosis (MS) 347

Laboratory-supported definite MS • Interferon beta-1b (IFN-β-1b), 8 million international


1 Two attacks; either clinical or paraclinical evidence of one units (MIU) every second day, subcutaneously reduces the
lesion; and CSF oligoclonal bands or increased IgG number of relapses (i.e. the attack rate) by about one-
(serum levels of either must be normal). third, the severity of the relapses and the number of
2 One attack; clinical evidence of two separate lesions; and demyelinating lesions seen on MRI of the brain. Short
CSF oligoclonal bands or increased IgG. term adverse effects include systemic ‘flu-like’ symptoms,
3 One attack; clinical evidence of one lesion and injection site reactions, liver enzyme elevations, anemia,
paraclinical evidence of another, separate lesion; and CSF mild leukopenia, thrombocytopenia and possibly
oligoclonal bands or increased IgG. depressive symptoms. Active, severe depression, pregnancy
and breastfeeding are contraindications to its use.
Clinically probable MS Neutralizing antibodies to IFN-β are detectable in about
1 Two attacks and clinical evidence of one lesion (the two 38% of patients by the third year of treatment and seem to
attacks must involve separate parts of the CNS. Historic attenuate the treatment effect; further research is needed
information cannot be considered as a substitute for the to determine their clinical significance. IFN-β-1b is a
clinical evidence.); or recombinant interferon beta made in bacterial cells. The
2 One attack and clinical evidence of two separate lesions; or mechanism of action of IFN-β in MS remains unknown.
3 One attack; clinical evidence of one lesion and • Interferon beta-1a (IFN-β-1a), 6 MIU once a week,
paraclinical evidence of another, separate lesion. intramuscularly (Avonex; CSL Ltd) or 6–12 million units
three times a week subcutaneously (Rebif; Serono Australia
Laboratory supported probable MS Pty Ltd). Interferon β-1a is a recombinant interferon-β
Two attacks and CSF oligoclonal bands or increased IgG. made in mammalian cells which, like IFN-β-1b also
reduces the annual relapse rate by about one-third and
NEW DIAGNOSTIC CRITERIA FOR MS significantly delays the progression of disability, compared
(MCDONALD ET AL. [2001]) with placebo, in relapsing multiple sclerosis, and has similar
• Incorporate MRI into the overall diagnostic scheme, and adverse effects.
add guidelines for the diagnosis of primary progressive • The most important determinant of clinical efficacy is the
disease. total dose of interferon-β.
• Some problems with these criteria are discussed by Poser • Glatiramer acetate (previously called copolymer 1)
and Brinar (2001). (Copaxone; Aventis Pharma Pty Ltd), 20 mg per day by
daily subcutaneous injection, reduces relapse rate at 2 years
TREATMENT by about 29% and improves disability in relapsing-remitting
Acute treatment to accelerate recovery from the acute attack. MS. Adverse effects include a transient injection site
reaction, sometimes with focal lipoatrophy, and a transient
Mild relapse of relapsing-remitting MS self-limiting systemic reaction characterized by flushing or
No specific treatment required, as most resolve spontaneously. chest tightness with palpitations, anxiety or dyspnea. It is a
mixture of random polymers of alanine, glutamine, lysine
Moderate–severe relapse of relapsing-remitting MS and tyrosine. It approximates the antigenic structure of
• Methylprednisolone: 1000 mg in 100 ml 5% dextrose, myelin basic protein (MBP) sufficiently to be cross-reactive
infused intravenously over 30–60 minutes, daily for 3 days, with monoclonal antibodies and T cells generated to MBP,
or 500 mg daily for 5 days. This therapy accelerates the a putative target antigen in MS. Glatiramer acetate is an
rate of recovery (i.e. reduces the duration of relapse) but alternative to interferon-β therapy in relapsing-remitting
has no effect on long term outcome. Potential adverse MS, and should particularly be considered in patients who
effects include mood alterations, psychosis, acne, fluid do not respond to, or do not tolerate, interferon-β.
retention, hyperglycemia, and osteonecrosis. • Intravenous immunoglobulin 0.15–0.2 g/kg body-
• Prednisolone: 60 mg, oral, daily for 1 week, 30 mg daily weight administered monthly in relapsing-remitting MS
for the second week, and 15 mg daily for the third week is well tolerated and may improve clinical disability and
is a more convenient and less expensive alternative but reduce annual frequency of relapses by about 59%.
there is no definitive evidence that it is better than
placebo. The data from randomized trials indicate that, compared
• Adrenocorticotropic hormone injections, given in with placebo, the odds of freedom from relapse at the end
reducing doses over 4 weeks (40 U i.m., b.d. for week 1; of 2 years are:
40 U i.m., b.d. for week 2; 20 U i.m., daily for week 3), • 1.37 (95% CI: 0.82–2.34) for glatiramer acetate.
may be effective but are rarely used nowadays. • 1.68 (0.88–3.16) for interferon β-1a.
• 2.38 (1.25–4.25) for interferon β-1b.
Long term treatment to prevent relapses and • 2.07 (1.07–4.00) for intravenous immunoglobulin (i.v. Ig).
progression of disability • 2.04 (1.42–2.93) for azathioprine.
Relapsing-remitting MS with at least two relapses in the
previous 2 years A randomized trial is currently comparing early
Several new drug treatments to reduce the relapse rate are azathioprine with interferon-β treatment.
now available (see below) but they are all expensive, have to The cost of generic azathioprine 150 mg daily is about
be given by injection, their long term effects are not yet GBP 80 yearly, excluding the costs of blood counts and liver
known, and definite evidence of a clear effect on progression function tests, compared with about GBP 3300 per year for
of disability is lacking: i.v. Ig, excluding day case costs each month, and about GBP
10 000 per year with the other new drugs.
348 Inflammatory Disorders of the Nervous System

Recent accelerated deterioration in primary or subcutaneous reservoir and pump, connected by catheter
secondary progressive MS to the subarachnoid space between L3/4 with the
• Intravenous high dose methyl-prednisolone infusion catheter tip located around T12 or higher.
(500 mg over 5 days). • Surgery.
• The chemotherapeutic agent, mitoxantrone, may have a
future role in the management of patients with recent Bladder dysfunction
onset, severe and rapidly progressive disease. Detrusor hyper-reflexia:
• Clean intermittent self-catheterization (CISC): the most
Sustained deterioration in primary or secondary effective.
progressive MS • Anticholinergic agents (reduce urgency but may increase
• Interferon β-1b slows the progression of disability and residual volume):
reduces the accumulation of MRI brain lesions in patients – Oxybutynin hydrochloride 5 mg tablets, 25–5.0 mg
with secondary progressive MS. Further studies are every 6–8 hours.
needed to determine its place in this context. The effect – Propantheline bromide (Pro-Banthine) 15 mg, four
of interferon-β in primary progressive MS is unknown. times daily.
• Oral low-dose methotrexate (7.5–12.5 mg once a week – Amitriptyline 25–100 mg daily.
indefinitely) significantly reduces the rate of progression • Intravesical capsaicin, which has a toxic effect on the C-fiber
of disability in progressive MS. It is generally well afferents in the bladder wall that drive the abnormal spinal
tolerated, but requires regular monitoring of full blood detrusor reflex, reduces detrusor muscle hyperactivity and
count and liver function. Adverse effects include nausea, may help patients with severe detrusor hyper-reflexia whose
hair thinning, bone marrow suppression, hepatotoxicity, bladder has very limited storage capacity. The effect lasts
opportunistic infection and pneumonitis. It is important 1–5 months, and may respond to repeat infusions.
to name the day (e.g. Monday) of the week on which the • Indwelling suprapubic catheter if the combination of
methotrexate is to be taken in order to avoid the possi- medication and CISC is not effective or practical.
bility of the patient taking the dose daily instead of weekly. Nocturia. Desmopressin spray (DDAVP) reduces the
Further clinical trials are needed to determine whether volume of urine produced.
higher doses of oral methotrexate are more effective, and
to determine the relative benefits of methotrexate therapy Bowel dysfunction
and interferon-β therapy in progressive MS. • Diet.
• Lactulose.
Symptomatic relief • Suppositories.
Rehabilitation by a multidisciplinary team plays a crucial role • Loperamide for urgency.
in management of MS.
Sexual dysfunction
Spasticity • Erectile dysfunction:
• Attend to any factors which may exacerbate spasticity, – Psychotherapy and psychosexual counselling.
such as noxious stimuli due to urinary tract infection, – Sildenafil: a phosphodiesterase type-5 (PDE-5) inhibitor
infected pressure sores or ulcers, tight clothing or an which increases cyclic GMP levels in the penis and enhances
uncomfortable orthosis. the smooth muscle relaxant effects of the nitric oxide
• Educate patients to understand and manage their spasticity. (NO)/cyclic GMP pathway, increasing penile blood flow.
• Correct posture: avoid positions which favor the pattern There are three doses (25 mg, 50 mg, 100 mg). The usual
of spasticity. starting dose is 50 mg; the 25 mg dose is used in younger
• Physiotherapy: aims to inhibit spasticity by facilitating a men, men with renal or hepatic impairment and those men
normal pattern of movement, improving postural tone on CYP3A4 inhibitors (erythromycin, cimetidine, and
and re-learning selected movements. Muscle stretching retroviral drugs). The tablet should be taken at least 1 hour
is also important. before anticipated sexual activity and will remain effective
• Pharmacology: for up to 4 hours. A large meal and alchol intake may delay
– Oral agents: the absorption. It is effective in 70–80% of men with
Baclofen, 5 mg b.d., increasing slowly to 10–25 mg three erectile problems. Adverse effects are mild and include
times daily if tolerated and required: the most effective headache, facial flushing, indigestion and rhinitis. It is
oral agent. contraindicated in men who take nitrate medication
Diazepam 5–10 mg three times daily. (including glyceryl trinitrate and amyl nitrate) or any form
Dantrolene. of NO donors, because of potential hypotensive effects.
Vigabatrin. – Yohimbine, an L2 alpha agonist.
Tizanidine, an L2 alpha adrenergic antagonist. – Intracorporeal papaverine or prostaglandin injection.
Cannabis. – Surgically implanted prostheses.
– Intramuscular injections of botulinum toxin A injections • Lack of vaginal lubrication and loss of sensation:
for focal spasticity (i.e. hip adductors). Effective when lubricating gels.
combined with regular physiotherapy.
– Nerve blocks: preferably on predominantly motor nerves, Bulbar dysfunction
such as obturator nerves. Pre-test with a reversible local • Speech (dysarthria predominantly):
anesthetic such as bupivacaine before resorting to dilute – Speech pathologist assessment and guidance: advice on
phenol or alcohol. breathing and articulation patterns, and augmentative com-
– Intrathecal baclofen: test with an initial bolus injection munication aids.
for functional benefit and analgesia; abdominal wall • Swallowing dysfunction (oral and pharyngeal phases):
Multiple Sclerosis (MS) 349

– Speech pathologist (± videofluoroscopy) assessment and • Amantadine 100 mg in the morning and afternoon.
guidance: education, dietary modification, positioning • 4-aminopyridine (fampridine), and 3-4-diaminopyridine,
strategies (e.g. ‘chin tuck’ and ‘head turn’), thermal a potassium channel blocking agent, may have a role.
stimulation (i.e. using ice to stimulate the faucial arches,
which delays triggering of the swallow reflex). Pain
– Assisted feeding, via percutaneous gastrostomy, may be • Trigeminal neuralgia (see p.509):
required in severe cases where swallowing is no longer – Carbamazepine 200–400 mg three times daily.
safe and the patient and carer agree. – Baclofen 10–20 mg three times daily.
– Misoprostol, a prostaglandin E1 analogue.
Visual dysfunction • Dysesthetic burning pain in the extremities:
• Monocular blindness or scotoma, diplopia and oscillopsia: – Paroxysmal: avoid precipitating maneuvers such as
– Difficult to manage. movement, tactile stimulation, and hyperventilation, if
– Referral to low vision clinics can be helpful. possible; consider bromocriptine 2–5 mg b.d.
– Botulinum toxin injections of oculomotor muscles may – Chronic: tricyclic antidepressants, e.g. amitriptyline, may help.
reduce persistent oscillopsia. • Painful tonic seizures: muscle relaxants, e.g. baclofen, may help.
• Acquired pendular nystagmus may respond to converg- • Chronic back pain: physiotherapy incorporating heat
ing prisms and isoniazid (and possibly gabapentin). pads and transcutaneous electric nerve stimulation.
• Painful leg spasms: baclofen.
Cognitive dysfunction
Adequate assessment and clarification of the deficits allows CLINICAL COURSE
informed discussion with patient and carer, constructive Symptoms usually persist for several weeks and then gradually
planning to minimize or overcome these deficits and, in but incompletely resolve over 1 or 2 months. Vision fre-
some cases, cognitive rehabilitation. quently improves in 1 or 2 weeks and often returns to near
normal. Occasionally, symptoms are short lived and paroxys-
Depression mal (e.g. recurrent short episodes of ataxia). The course is
The treatment of depression is similar to that of a patient relapsing and remitting in about 80% of patients. Relapse may
who does not have MS but particular attention needs to be occur at any time. The average relapse rate is about 0.5 attacks
paid to adverse effects, as they may exaggerate existing per year but is very variable. The first 3 months following
problems such as sexual dysfunction: delivery of a child is associated with about a threefold increase
• Psychologic counselling. in relapse rate. Complete recovery usually follows the initial
• Tricyclic antidepressants. attack but later relapses are associated with increasing residual
• Serotonin reuptake inhibitors. disability. In some patients, the course becomes progressive.
A chronic progressive course occurs from onset in the other
Tremor and ataxia 20%, particularly if onset occurs after 40 years of age with
• Physiotherapy: improve the patient’s posture and seating spastic paraparesis due to spinal cord dysfunction. These
and supply adequate support. patients also tend to have a worse prognosis.
• Oral medication:
– Clonazepam 0.5–2.0 mg two or three times daily. PROGNOSIS
– Thioridazine (Mellaril) 10–50 mg two or three times daily. • Life expectancy from onset of symptoms is very variable.
– Ondansetron, a 5-hydroxytryptophan-3 (5-HT3) • Mean survival: 30 years; 10% die within 15 years and a
antagonist, 8 mg i.v. small percentage die within several months or years. In
– Carbamazepine. one study, the 25 year survival rate was 74%, compared
– Isoniazid and pyridoxine. with 86% for the general population.
– Propranolol. • Most deaths are due to advanced chronic disability and
– Buspirone. not acute attacks.
• Surgery. Stereotactic thalamotomy, lesioning the • After 25 years, one-third of MS patients are still working
ventrolateral nucleus of the thalamus, is beneficial in and two-thirds are still ambulating.
about half of cases but carries a risk of hemiparesis and
dysphasia. Electrode implantation and stimulation in the Adverse factors for long term survival free of disability
same area appears more promising. • Late age of onset.
• Botulinum toxin type A injections (40 mouse units) into • Male sex.
the flexor and extensor compartments of the forearm is • Cerebellar dysfunction (ataxia) at onset.
not helpful, particularly if there is pre-existing weakness. • Short interval between the first two relapses.
• Progressive clinical course.
Temperature lability • Spinal cord axonal loss and reduced N-acetyl aspartate as
• The rather cumbersome cooling suits may be helpful in measured by magnetic resonance spectroscopy.
selected cases.
• 4-aminopyridine (fampridine), a potassium blocking Favorable factors for long term survival free of disability
agent, improves symptoms and signs in MS, probably by • Early age of onset.
prolonging the repolarization phase of the action • Female sex.
potential and thus helping to restore nerve conduction • Sensory dysfunction (paresthesia) at onset.
in demyelinated nerve fibers. • Relapsing-remitting clinical course.
• Longer inter-attack interval.
Fatigue • Low initial relapse rate.
• Psychologic counselling. • Fewer lesions on baseline MRI.
350 Inflammatory Disorders of the Nervous System

NEUROSARCOIDOSIS Microscopic findings in the nervous system


• Non-caseating epithelioid granulomata (430):
– An aggregate of tightly clustered mononuclear
DEFINITION epithelioid histiocytes (phagocytes) with eosinophilic
A chronic, multisystem, granulomatous, inflammatory cytoplasm and oval nuclei surrounded by a rim of T
disease of unknown etiology in which any part of the helper-inducer lymphocytes and, to a far lesser extent, B
nervous system (and indeed any organ of the body) may be lymphocytes.
affected by exaggerated T helper lymphocyte immune – Giant cells of the Langhan’s or foreign-body variety may
responses to a variety of antigens (self or non-self). This be present in the granuloma. Tend to form in the
results in an accumulation of mononuclear inflammatory cells perivascular spaces and walls of small penetrating arteries
(mostly T helper lymphocytes and mononuclear phagocytes), and veins of the meninges and parenchyma.
non-caseating epithelioid granulomas, and derangements of • Lymphocytic infiltrate, often perivascular and associated
normal tissue architecture in affected tissues. with neuronal loss, reactive gliosis, loss of myelin, and
residual scars.
EPIDEMIOLOGY • Basal leptomeningitis.
• Prevalence: • More diffuse leptomeningeal and ependymal disease.
– Systemic sarcoidosis: 1–50 per 100 000 population. • Intra-axial granulomatous masses.
– Neurosarcoidosis: 5 per 100 000. • Granulomatous necrotizing angiitis that predominantly
• Incidence: systemic sarcoidosis: 10.9 per 100 000 for affects veins (causing a phlebitis or venulitis) and small
whites; 35.5 per 100 000 for blacks. arteries of the meninges and brain.
• Age: any age (3 months–old age), but commonly 20–55
years of age. PATHOGENESIS
• Gender: M=F. • Uncertain.
• An exaggerated helper-inducer T lymphocyte cellular
PATHOLOGY immune response to a variety of antigens or self-antigens.
Site • Since the activated helper-inducer T lymphocytes release
Sarcoidosis often affects multiple organ systems: mediators that attract and activate mononuclear
• Lungs (about 87%). phagocytes, it is likely that the process of granuloma
• Lymph nodes (28%). formation is secondary to the exaggerated helper-inducer
• Skin (18%). T cell process.
• Eyes: conjunctivae, iris (15%).
• CNS (5–16%) ETIOLOGY
– Intracranial: Hypotheses
Meningeal: diffuse granulomatous meningitis or • A class of antigens, non-self or self, trigger only the
meningo-encephalitis, or circumscribed granulomas, helper-inducer T cell arm of the immune response.
causing thickening of the arachnoid, anywhere over the • An inadequate suppressor arm of the immune response,
surface of the brain or spinal cord but particularly around such that helper-inducer T cell processes cannot be shut
the optic chiasm, hypothalamus, basal cisterns and down in a normal fashion.
subependymal region of the third ventricle. • Inherited (and/or acquired) differences in immune
Parameningeal. response genes, such that the response to a variety of
Parenchymal lesions: anywhere but particularly in the antigens is an exaggerated helper-inducer T cell process.
periventricular regions and in the Virchow–Robin spaces
where they form granulomatous masses up to several CLINICAL FEATURES
centimeters in diameter. Cerebral infarction may also Systemic manifestations
occur due to sarcoid angiitis. • Present in 98% of patients with neurosarcoidosis.
Hypothalamus. • Symptoms include exertional dyspnea and dry cough due
Pituitary. to interstitial lung disease.
Ependymal linings of ventricles and choroid plexus (may • Signs include skin rash (i.e. erythema nodosum), uveitis,
cause hydrocephalus). lymphadenopathy and arthritis.
Cranial nerves (any nerve, but mostly facial nerve,
involved by granulomatous infiltration or compression Neurologic manifestations
by mass lesions). • The presenting manifestation in about 2% (0.3–2.5%) of
– Spinal cord: granulomatous meningitis, vasculitis or cases of sarcoidosis, concurrent with systemic
circumscribed granulomas within the leptomeninges or involvement in about 5% of patients, and may precede
parenchyma of the spinal cord. systemic involvement by up to 18 months.
• Peripheral nervous system (6–18%): • Meningitis:
– Polyradiculopathy. – Aseptic meningitis.
– Symmetric polyneuropathy (pure sensory, pure motor, – Chronic granulomatous basal meningitis.
sensori-motor). • Epilepsy: secondary to meningitis; mass lesion; cerebral
– Mononeuritis multiplex. infarction.
• Muscle. • Raised intracranial pressure:
• Liver. – Intraparenchymal mass lesion.
• Kidneys. – Hydrocephalus.
• Testes.
Neurosarcoidosis 351

• Uni- or multi-focal neurologic deficits: INVESTIGATIONS


– Stroke syndrome. Blood
– Multifocal changes in CNS white matter. No peripheral blood findings are diagnostic of the disease:
– Vasculopathy: ophthalmoscopy may reveal periphlebitis, • Full blood count: anemia, increased number of
as yellowish-white focal or diffuse sheathing of retinal monocytes.
veins. Hard exudates, sometimes termed taches de bougie • Serum urea and electrolytes, glucose, liver function tests,
because of their resemblance to candle-wax drippings, uric acid: may be abnormal.
often accompany the periphlebitis and can leave white • Serum calcium: elevated.
choreoretinal scars. • Serum immunoglobulins: hypergammaglobulinemia.
• Single or multiple cranial neuropathies: • Serum angiotensin-converting enzyme (ACE): elevated
– Anterior optic neuropathy (431, 432). in about two-thirds of patients but it is neither sensitive
– Facial palsy: with enlarged parotid glands (uveoparotid (sensitivity varies from 56–86%) nor specific (high). The
fever). false positive rate in a normal population is about 2–4%.
• Neuroendocrine (hypothalamic and pituitary The level of serum ACE correlates with the severity of
dysfunction): diabetes insipidus (polyuria, polydipsia, the lung disease and the presence or absence of
disordered thirst). extrathoracic disease.
• Subacute or chronic myelopathy. • Serum lysozyme: elevated in about two-thirds of cases,
• Peripheral neuropathy: less specific than elevated ACE.
– Guillain–Barré syndrome. • Serum beta2-microglobulin: elevated in about two-
– Polyradiculopathy. thirds, less specific than elevated ACE.
– Symmetric polyneuropathy (pure sensory, pure motor,
sensori-motor).
– Mononeuritis multiplex.
• Myopathy.

430 Biopsy showing a sarcoid granuloma characterized by an 430


aggregate of tightly clustered mononuclear epithelioid histiocytes and
multinucleated giant cells.

431 432

431, 432 Ocular fundi of a patient with anterior optic neuropathy due to sarcoidosis (431, right eye; 432, left eye). He presented with subacute
onset of an island of blurred vision in the inferior visual field of the left eye and mild ocular pain bilaterally. Examination showed normal visual acuity
bilaterally, an enlarged blind spot and an arcuate scotoma infero-nasally on the left, and asymmetric optic disc swelling being greater in the left eye
(432). Note the nerve fiber layer hemorrhages in the left fundus.
352 Inflammatory Disorders of the Nervous System

Imaging • Bone marrow: non-caseating granuloma.


• Chest x-ray: may show bilateral hilar adenopathy (433) • Biopsy of muscle (even in the absence of myopathic
but occasionally a similar pattern is seen in lymphoma, symptoms), lymph node, conjunctiva, skin, lung, liver or
tuberculosis, brucellosis and bronchogenic carcinoma. brain may reveal the diagnosis.
• Gallium scan: more sensitive than chest x-ray and the • Nerve biopsy:
appearance of diffuse uptake in the lungs (or parotid, – An axonopathy sparing unmyelinated fibers.
salivary and lacrimal glands), even in the absence of – Marked loss of myelinated fibers.
clinical involvement, is relatively specific but not – Demyelination is not prominent nor is it in the usual
diagnostic. central predominant pattern seen in vasculitic
• CT brain scan without and with contrast: hydrocephalus, neuropathies.
leptomeningeal thickening and enhancement, intra-axial – Epineural and perineural granuloma with or without
mass lesions, extra-axial mass lesions. periangiitis and panangiitis.
• Brain biopsy: in most cases involving intracranial mass
MRI brain/spinal cord scan lesions which are difficult to access safely with stereotactic
Several patterns may be visible on cranial MRI (434) (CT brain biopsy, biopsy can be deferred until a trial of
is much less sensitive): steroids is given if sarcoidosis can be inferred clinically.
• Chronic basal leptomeningitis with thickened enhancing
meninges involving the hypothalamus, pituitary stalk, Other
optic nerve (435) and chiasm. • Skin: skin-test unreactivity (anergy).
• Communicating hydrocephalus. • Urine: 24 hour urinary calcium level: typically elevated.
• Involvement of the lenticulostriate arteries by spreading but not diagnostic, of sarcoidosis.
up the Virchow–Robin spaces causing thrombosis and
granulomatous angiitis. DIFFERENTIAL DIAGNOSIS
• Parenchymal nodules (granulomas) which may be iso- or Meningo-encephalitis
hyperdense, may calcify and appear as a mass lesion and • Granulomatous inflammation:
may or may not enhance. These may cause obstruction to – Infection by bacteria (brucellosis, chlamydia, tularemia),
the ventricles. They occur particularly around the skull base, mycobacteria (tuberculosis, atypical mycobacteria), fungi
pituitary, pons, hypothalamus and periventricular region. (histoplasmosis, coccidioidomycosis, cryptococcosus),
• Diffuse high signal areas in the white matter on T2W spirochetes (treponemal infections such as syphilis), and
images indistinguishable from MS may also occur. parasites (toxoplasmosis, leishmaniasis).
• Extra-axial sarcoid may mimic a meningioma by causing – Occupational and environmental exposure to organic or
a dural enhancing mass with hyperostosis. inorganic agents (e.g. methotrexate, talc, metals).
• Spinal sarcoid may mimic leptomeningeal metastases. – Neoplasia (lymphoma).
– Autoimmune disorders (Wegener’s granulomatosis,
Cerebral angiography Churg–Strauss syndrome).
Changes suggestive of cerebral angiitis may occur. – Histiocytosis X:
• Viral encephalitis (see p.292): HIV infection (see p.301).
CSF • Carcinomatous meningitis
• Mild pleocytosis (mostly lymphocytes). • Granulomatous angiitis of the CNS (see CNS vasculitis,
• Mild increased protein. p.227). The meningeal inflammatory reaction is
• Increased T4 : T8 lymphocyte ratio. consistently localized to blood vessel walls and typically
• Low CSF glucose in 20–39% of cases. produces extensive disruption of the vascular wall.
• Elevated CSF ACE, oligoclonal banding and increased • Meningiomatosis
IgG index in nearly one-third of patients may occur. • Limbic encephalitis associated with occult neoplasm (see
p.401).
EMG
• Compound muscle action potentials and sensory nerve Multifocal white matter disease
action potentials: normal or mild to moderately Multiple sclerosis.
decreased.
• Motor and sensory nerve conduction velocities: mild to Mass lesions
moderately decreased. • Primary brain tumor (see p.357): glioma (including optic
• EMG features of chronic partial denervation may be nerve glioma), meningioma, germ cell tumor, leukemia,
present. primary CNS lymphoma.
• Metastatic brain tumor (see p.396).
Kveim test • Granulomatous inflammation: syphilitic gumma, tuber-
A suspension of sarcoid tissue is injected intradermally and culoma, cryptococcoma, toxoplasmosis, histiocytosis X.
produces a sarcoid granuloma in about 75% of patients with • Spinal cord glioma, ependymoma, myelitis.
subacute active sarcoidosis and in about two-thirds of those
with chronic sarcoidosis of >2 years duration. False-positives Non-caseating granulomas
occur in <5% of cases. Non-specific: found in infections and malignancy.

Tissue Increased level of serum ACE


• Bronchoalveolar lavage: abnormally increased T4 : T8 • Youth.
lymphocyte ratio: relatively specific but not diagnostic. • Hyperthyroidism.
Neurosarcoidosis 353

• Diabetes. • Prednisone 60 mg (1 mg/kg) oral, daily for 4–6 weeks,


• HIV infection. followed by a slow taper over 3–6 months or longer.
• Leprosy. • If steroids fail, or cannot be tapered eventually, other
• Tuberculosis. immunosuppressive agents, such as cyclophosphamide,
• Coccidioidomycosis. azathioprine, and cyclosporine (4–6 mg/kg/day, which
• Histoplasmosis. inhibits interleukin-2 secretion by T cells) allow
• Amyloidosis. reduction of steroids to 30–50% of the original dose.
• Multiple myeloma. • Surgery may be indicated for hydrocephalus, expanding
• Hodgkin’s disease. mass lesions, or mass lesions causing increased
• Lymphoma (including intravascular lymphoma). intracranial pressure.
• Malignant histiocytosis. • Cranial irradiation of intracranial neurosarcoidosis that is
• Primary biliary cirrhosis. refractory to treatment with steroids may be successful.
• Whipple’s disease.
• Gaucher’s disease. PROGNOSIS
• Silicosis. • Most patients respond to treatment and can have
• Asbestosis. medication withdrawn over months but about one-third
will relapse, often in the same location as the initial
DIAGNOSIS disease.
A positive biopsy finding of non-caseating granulomas in • Neurosarcoidosis limited to peripheral nerves and cranial
the context of characteristic clinical features, blood test nerves may respond spontaneously.
(elevations of serum gamma globulin, ESR, or serum ACE), • Intracranial neurosarcoidosis, such as mass lesions or
chest x-ray evidence of enlarged hilar and mediastinal lymph hydrocephalus, have a more malignant course and a
nodes, gallium scan, defects in cell mediated immunity, and higher rate of relapse.
MRI and CSF findings, having excluded other causes of • Patients with relapsing disease and more ominous clinical
granulomatous inflammation. features (e.g. intracranial neurosarcoid such as mass lesions
or hydrocephalus) should probably remain on low-dose
TREATMENT steroids as a maintenance dose between exacerbations.
Inferred from experience with pulmonary sarcoidosis; no
controlled studies:

433 434

435
433 Chest x-ray showing bilateral hilar adenopathy due to sarcoidosis.

434 T1W MRI of the brain stem following contrast. Note the thin rim
of enhancement (arrows) around the brain stem. It is unusual to see
this in sarcoid, but its presence should certainly prompt the diagnosis,
though is not specific for sarcoid.

435 MRI of the orbits, coronal plane, after gadolinium injection


showing brightness, due to contrast enhancement, of the dural sheath
of the optic nerves (arrows) bilaterally in the patient in 431, 432, with
bilateral optic neuropathy due to sarcoidosis.
354 Inflammatory Disorders of the Nervous System

CAVERNOUS SINUS SYNDROME CLINICAL FEATURES


Cranial nerve palsy
• Oculomotor (IIIrd) nerve: almost all patients; pupil is
DEFINITION involved in three-quarters of cases and generally parallels
Involvement of two or more of the IIIrd, IVth, Vth (V1, the severity of the ophthalmoplegia.
V2), or VIth cranial nerves or oculosympathetic fibers on • Abducens (VIth) nerve: 95% of patients.
the same side. • Trochlear (IVth) nerve: 30% of patients; may be difficult
to confirm in the presence of a partial IIIrd nerve palsy.
EPIDEMIOLOGY • Trigeminal (Vth) nerve: 40% of patients.
• Incidence: rare. • Optic nerve or chiasmal dysfunction: 40% of patients.
• Age: any age; mean age: 40 years.
• Gender: M=F. Horner’s syndrome
• 5–10% of patients.
PATHOPHYSIOLOGY • Difficult to diagnose in the presence of IIIrd nerve palsy
The cavernous sinus (really a venous plexus) lies within a without pharmacologic pupillary testing.
dural envelope that funnels upper cranial nerves to the orbit.
Consequently, small lesions, adjacent to or within the HISTORY
cavernous sinus, can produce dramatic localizing signs. • Age of onset.
• Presence or absence of pain.
ETIOLOGY • Speed of progression.
Tumor: 30–70% of all cases • Past infections and tumors.
Malignant tumors: two-thirds
In marked contrast to the high incidence of benign tumors Tumor
causing chiasmal compression: Average age: 47 years:
• Nasopharyngeal cancer: 22–46% of tumors. • Nasopharyngeal cancer: preferentially involves the VIth
• Metastases: 16–33%. nerve and mandibular division of the Vth nerve.
• Lymphoma: 0–18%. • Metastases: often present with rapid complete ophthal-
• Pituitary adenoma: 9–22%. moplegia.
• Lymphoma: other signs may be present, such as
Benign tumors abdominal mass.
One-third. • Pituitary adenoma: lateral extension tends to involve the
• Meningioma: 4–9% of tumors. IIIrd nerve and ophthalmic division of the Vth nerve.
• Chordoma: 2–11%. Apoplectic onset usually, and involves the oculomotor
• Neuroma: 0–8%. nerves with a relative frequency ratio of about 4 (IIIrd
nerve), 2 (VIth nerve) and 1 (IVth nerve).
Aneurysm or fistula: 5–35% of all cases • Benign tumors: a single cranial nerve is involved for long
• Cavernous carotid artery aneurysm. periods.
• Carotid-cavernous fistulas.
Aneurysm
Trauma: 5–25% of all cases • Average age: 52 years.
• Typically progressive, painful involvement of multiple
Surgery: 10% of all cases cranial nerves.
• Multiple cranial nerves are affected in two-thirds, VIth
Inflammation (idiopathic cavernous sinusitis): nerve only in one-quarter, and IIIrd nerve only in 10%.
0–23% of all cases • Despite IIIrd nerve involvement, the pupil on the
The Tolosa–Hunt syndrome eponym, with its promise of involved side is often the same size or smaller than the
making a specific diagnosis on clinical grounds, has not normal pupil, perhaps because the parasympathetic fibers
proved reliable. One of the five patients who helped are less vulnerable within the cavernous sinus.
establish the Tolosa–Hunt syndrome later was shown to • Painful onset in almost all patients.
have a meningioma. A generic description, such as • Sudden onset in about 10% of cases.
idiopathic cavernous sinusitis, seems preferable. Overlap
with orbital inflammatory pseudotumor and idiopathic Trauma
multiple cranial neuropathy syndromes suggest a similar • Average age: 30 years.
underlying mechanism. • Usually severe head trauma with basal skull fractures.

Infection: 0–5% of all cases Inflammation (idiopathic cavernous sinusitis)


Spread of fungus or bacteria, usually from the sphenoid • Average age: 35 years.
sinus: • Painful, non-specific inflammation of the superior orbital
• Mucormycosis. fissure.
• Meningitis.
• Bacterial sphenoid sinusitis. Infection
• Septic cavernous sinusitis. • Underlying diabetes common.
• Rapid, complete ophthalmoplegia.
Diabetes: 1% of all cases • Life-threatening.
Cavernous Sinus Syndrome 355

DIFFERENTIAL DIAGNOSIS PROGNOSIS


Unilateral ophthalmoplegia Tumor
• Orbital inflammatory pseudotumor syndrome. • Malignant tumors: rapid deterioration.
• Idiopathic multiple cranial neuropathy syndrome. • Benign tumors: prolonged course.
• Wernicke’s encephalopathy.
• Myasthenia gravis. Aneurysm
• Typically progressive, prolonged course.
Rapid onset, severe ophthalmoplegia • One-quarter have some recovery of eye movements;
• Trauma. residual damage often consists of elevator paresis and
• Pituitary apoplexy. mild ptosis due to permanent damage to the superior
• Carotid aneurysm. division of the IIIrd nerve.
• Mucormycosis.
• Metastatic tumor. Inflammation (idiopathic cavernous sinusitis)
Remitting course, usually self-limiting.
Bilateral cavernous sinus syndrome: <10% of all cases
• Nasopharyngeal carcinoma. Infection
• Lymphoma. Life-threatening and usually fatal, despite intensive antibiotic
• Leukemia. treatment.
• Craniopharyngioma.
• Pituitary apoplexy. Close clinical follow-up is essential and the MRI should
• Mucormycosis. be repeated if a sustained remission is not forthcoming.

INVESTIGATIONS
• Contrast-enhanced MRI brain scan (436) is the key
investigation: it clearly demonstrates any tumor mass or
aneurysm large enough to cause a cavernous sinus
syndrome, and usually shows carotid-cavernous fistulas,
clues to bacterial or fungal nasal sinusitis and adjacent
basilar meningeal enhancement. Idiopathic cavernous
sinusitis is often not apparent on non-contrast imaging
but, after contrast injection, the affected cavernous sinus
usually appears mildly or moderately enlarged. Abnormal
signal tissue, which may have mass effect in the cavernous 436
sinus (similar to muscle on T1WI and to fat on T2WI),
may extend into the orbital apex. The differential diagnosis
includes sarcoid, meningioma, lymphoma, metastatic or
local spread of tumor, infections like actinomycosis.
• MR angiography or catheter contrast carotid
angiography.
• CSF, including cytology: if meningitis, lymphoma, or
leukemia is suspected.
• Tensilon test: if myasthenia gravis is suspected.

DIAGNOSIS
Idiopathic cavernous sinusitis is a diagnosis of exclusion,
made only after the passage of considerable time (at least 6
months) after the onset of acute painful ophthalmoplegia to
confirm complete or almost complete remission and
eliminate the possibility of a subacute infection or a subtle
tumor in the cavernous sinus. MRI enhancement of a mildly
enlarged cavernous sinus supports, but does not establish,
the diagnosis; it may also be caused by infection, lymphoma,
meningioma, dural arteriovenous malformation.

TREATMENT
• Specific medical (anti-microbial, anti-inflammatory) or
surgical treatment, depending on the cause.
• Idiopathic cavernous sinusitis may respond to
corticosteroid therapy but patients need to be carefully
observed for a facilitated fungal infection. 436 T1W coronal MRI with contrast of a 20 year old man with a
painful right ophthalmoplegia, visual loss, sensory disturbance in the V1
distribution.This section was taken just posterior to the orbital apex.
Note the enhancement (whiteness, arrow) around the right optic
nerve which extended from the orbital apex.
356 Inflammatory Disorders of the Nervous System

Drug treatment Rehabilitation


FURTHER READING Bryant J, Clegg A, Milne R (2001) Systematic re- Freeman JA, Langdon DW, Hobart JC, Thomp-
view of immunomodulatory drugs for the son AJ (1999) Inpatient rehabilitation in mul-
treatment of people with multiple sclerosis: is tiple sclerosis. Do the benefits carry over into
ACUTE DISSEMINATED there good quality evidence on effectiveness the community? Neurology, 52: 50–56.
ENCEPHALOMYELITIS and cost? J. Neurol. Neurosurg. Psychiatry, 70: Freeman JA, Thompson AJ (2001) Building an
Davis LE (2000) Diagnosis and treatment of acute 574–579. evidence base for multiple sclerosis manage-
encephalitis. The Neurologist, 6: 145–159. Campbell SK, Almeida GL, Penn RD, et al. ment: support for physiotherapy. J. Neurol.
Sahlas DJ, Miller SP, Guerin M, et al. (2000) (1995) The effects of intrathecally adminis- Neurosurg. Psychiatry, 70: 147–148.
Treatment of acute disseminated en- tered baclofen on function in patients with Kraft GH (1999) Rehabilitation still the only way
cephalomyelitis with intravenous im- spasticity. Physical Therapy, 75: 352–362. to improve function in multiple sclerosis.
munoglobulin. Neurology, 54: 1370–1372. Comi G, Filippi M, Barkhof F, et al. (2001) Effect Lancet, 354: 2016–2017.
of early interferon treatment on conversion to Solari A, Filippini G, Gasco P, et al. (1999) Phys-
MULTIPLE SCLEROSIS (MS) definite multiple sclerosis. A randomised study. ical rehabilitation has a positive effect on dis-
General Lancet, 357: 1576–1582. ability in multiple sclerosis patients. Neurolo-
Noseworthy JH, Lucchinetti C, Rodriguez M, Ebers GC (2001) Preventing multiple sclerosis? gy, 52: 57–62.
Weinshenker BG (2000) Multiple sclerosis. N. Lancet, 357: 1547. Wiles CM, Newcombe RG, Fuller KJ, et al.
Engl. J. Med., 343: 938–952. Edan G, Miller D, Clanet M, et al. (1997) Ther- (2001) Controlled randomized cross-over trial
apeutic effect of mitoxantrone combined with of the effects of physiotherapy on mobility in
Pathogenesis methylprednisolone in multiple sclerosis: a ran- chronic multiple sclerosis. J. Neurol. Neurosurg.
Confavreux C, Suissa S, Saddier P, et al. (2001) domised multicentre study of active disease Psychiatry, 70: 174–179.
Vaccinations and the risk of relapse in multi- using MRI and clinical criteria. J. Neurol. Neu-
ple sclerosis. N. Engl. J. Med., 344: 319–326. rosurg. Psychiatry, 62: 112–118. Prognosis
Lucchinetti C, Brück W, Parisi J, et al. (2000) Fischer JS, Priore RL, Jacobs LD, et al. (2000) Bjartmar C, Kidd G, Mörk S, et al. (2000) Neu-
Heterogeneity of multiple sclerosis lesions: im- Neuropsychological effects of interferon ß–1a rological disability correlates with spinal cord
plications for the pathogenesis of demyelina- in relapsing multiple sclerosis. Ann. Neurol., axonal loss and reduced N-acetyl aspartate in
tion. Ann. Neurol., 47: 707–717. 48: 885–892. chronic multiple sclerosis patients. Ann. Neu-
Weatherby SJM, Hawkins CP (2001) Multiple Hartung HP, Gonsette RE, the MIMS Study rol., 48: 893–901.
sclerosis: New pathophysiological and thera- Group (1998) Mitoxantrone in progressive Weinshenker BG (2000) Progressive forms of MS:
peutic concepts. Proc. R. Coll. Physicians multiple sclerosis: a placebo controlled ran- classification streamlined or consensus over-
Edinb., 31: 134–148. domised, observer-blind European Phase III turned? Lancet, 355: 162–163.
Clinical entities multicentre study. Clinical data. Multiple Scle- NEUROSARCOIDOSIS
Chan JW (2000) Optic neuritis in multiple scle- rosis (Abstract), 4: 380. James DG (2001) Sarcoidosis 2001. Postgrad.
rosis: an update. The Neurologist, 6: 205–213. INFß Multiple Sclerosis Study Group (1993) In- Med. J., 77: 177–180
Rudick RA (1998) A 29-year-old man with mul- terferon Beta–1b is effective in relapsing–re- Newman LS, Rose CS, Maier LA (1997) Sar-
tiple sclerosis. JAMA, 280: 1432–1439. mitting multiple sclerosis. I. clinical results of coidosis. N. Engl. J. Med., 336: 1224–1234.
a multicentre, randomised, double-blind, Nikhar NK, Shah JR, Tselis AC, Lewis RA (2000)
Diagnosis placebo-controlled trial. Neurology, 43: Primary neurosarcoidosis: a diagnostic and
McDonald WI, Compston A, Edan G, et al. 662–667 therapeutic challenge. The Neurologist, 6:
(2001) Recommended diagnostic criteria for Jacobs LD, Beck RW, Simon JH, et al. (2000) In- 126–133.
multiple sclerosis: guidelines from the Interna- tramuscular interferon beta-1a therapy initiat- Scott TF (1993) Neurosarcoidosis: Progress and
tional Panel on the diagnosis of multiple scle- ed during a first demyelinating event in mul- clinical aspects. Neurology, 43: 8–12.
rosis. Ann. Neurol., 50: 121–127. tiple sclerosis. N. Engl. J. Med., 343: Vinas FC, Rengachary S (2001) Diagnosis and
Poser CM, Brinar VV (2001) Problems with di- 898–904. management of neurosarcoidosis. J. Clinical
agnostic criteria for multiple sclerosis. Lancet, Polman CH, Uitdehaag BMJ (2000) Drug treat- Neuroscience, 8(6): 505–513.
358: 1746–1747. ment of multiple sclerosis. BMJ, 321: Zajicek JP, Scolding NJ, Foster O, et al. (1999)
Thompson AJ, Montalban X, Barkhof F, et al. 490–494. Central nervous system sarcoidosis – diagno-
(2000) Diagnostic criteria for primary pro- Prisms Study Group (1998) Randomised double- sis and management. Q. J. Med., 92: 103–117.
gressive multiple sclerosis: a position paper. blind placebo-controlled study of interferon
Ann. Neurol., 47: 831–835. Beta-1a in relapsing/remitting multiple scle- CAVERNOUS SINUS SYNDROME
Poser CM, Paty D, Scheinberg LS, et al. (1983) rosis. Lancet, 352: 1498–1504 Keane JR (1996) Cavernous sinus syndrome.
New diagnositic criteria for multiple sclerosis. Analysis of 151 cases. Arch. Neurol., 53:
Ann. Neurol., 13: 227–231. 967–971.
Chapter Thirteen 357

Tumors of the
Central Nervous
System
BRAIN TUMORS – Neuroblastoma.
• Pineal cells: pineal cell tumors (pineocytoma, pineo-
blastoma).
DEFINITION
Neoplastic lesions of the brain which may be benign or Cellular derivatives of the neural crest
malignant, and primary or secondary (metastatic). • Arachnoid cells:
– Meningioma (15%) (see p.374).
EPIDEMIOLOGY – Meningeal sarcoma.
• Incidence: • Schwann cells:
– Primary brain tumors (benign and malignant): – Schwannoma (7–8%) (see p.383).
12–15/100 000/year (adults); 2–5/100 000/year – Neurofibroma.
(children).
– Primary benign CNS tumor: 7 (95% CI: 3–13) per Other cells
100 000 per year. • Adenohypophyseal cells (see p.379): pituitary adenoma
– Primary malignant CNS tumor: 3 (95% CI: 0.7–7) per (10%), pituitary carcinoma.
100 000 per year. • Reticuloendothelial cells: primary cerebral lymphoma
– Secondary brain tumors (metastases): 4 (95% CI: 1–9) (see p.385).
per 100 000 per year. • Vascular cells: hemangioblastoma (see p.392).
• Age: • Glomus jugulare cells: glomus jugulare tumors.
– Adults: supratentorial tumors predominate (e.g. glioma, • Connective tissue cells: sarcomas.
meningioma, pituitary adenoma, metastases).
– Children: infratentorial tumors predominate (e.g. Embryonal remnants
medulloblastoma, ependymoma, astrocytoma, pinealoma). • Ectodermal derivatives: craniopharyngioma (2–3%) (see
After 10 years of age, the incidence of neuroepithelial p.377).
tumors increases steadily with increasing age to a peak • Notochord: chordoma.
incidence of 20/100 00/year at age 70 years. • Germ cells (see p.388): germinoma (0.5%).
• Gender: M=F overall. • Derived from the three germ layers: teratoma.
– Men: gliomas more common; women: meningiomas and
nerve sheath tumors are more common.

PATHOLOGY
A. Histologic classification of brain tumors. (A more recent
World Health Organization classification also exists
[Kleihues and Cavenee, 2000; De Angelis, 2001].)

Neuro-epithelial (50%): cellular derivatives of the


neural tube
• Glial cells (Gliomas, see p.364)
• Neurons:
– Embryonal tumors: medulloblastoma (20% of all
childhood tumors, <5% of adult brain tumors) and
primitive neuroectodermal tumors.
– Gangliocytoma/ganglioglioma.
– Dysembryoblastic neuroepithelial tumor (DNET).
358 Tumors of the Central Nervous System

Metastases to the brain (15%) (437) Cytotoxic edema


Carcinoma of the lung, breast and kidney, and malignant Cytotoxic edema is excessive intracellular fluid that is caused
melanoma (438). by an increase in permeability of the cell membrane due to
failure of the cell membrane pump that maintains ion and
B. An alternative classification of brain tumors. fluid homeostasis. Although cytotoxic edema
characteristically occurs in response to cerebral hypoxia and
Primary brain tumors ischemia, it also occurs in brain tumors.
Histologically malignant
• Glioma: ETIOLOGY
– Astroglial neoplasm: No specific etiologic factor (e.g. infectious agent, chemical,
Astrocytoma (439)*. electric field) has been identified.
Anaplastic astrocytoma.
Glioblastoma multiforme. Risk factors
– Oligodendroglioma*. • Ionizing radiation of the cranium, usually for childhood
– Ganglioglioma*. malignancy: the only unequivocal risk factor for glial and
– Ependymoma. meningeal neoplasms can increase the incidence of
• Lymphoma. meningioma by a factor of 10 and the incidence of glial
• Medulloblastoma. tumors by a factor of 3–7, with a latency of 10 years to
• Primitive neuroectodermal tumors. more than 20 years after exposure.
• Germ-cell tumors: • Family history of cancer: present in 16% of patients with
– Germinoma. primary brain tumor. Several neurocutaneous syndromes
– Teratoma. are associated with specific tumor types: neurofibromatosis
– Embryonal carcinoma. types 1 and 2 is associated with glioma, meningioma,
– Choriocarcinoma. acoustic neuroma (see p.383); tuberous sclerosis with
– Endodermal-sinus tumor (yolk-sac tumor). astrocytoma (see p.143); and von Hippel–Lindau disease
• Pineal cell tumors. with hemangioblastoma (see p.392); Turcot’s syndrome
• Chordomas. with glioblastomas, medulloblastomas, and colon
• Choroid-plexus carcinomas. carcinoma; and Li-Fraemeni syndrome with multiple
familial tumors including glioblastoma.
*It is not quite clear whether these should be considered
benign or malignant histopathologically. Heritable syndromes and CNS tumor associations
Neurofibromatosis
Histologically benign or malformative • Visual pathway gliomas.
• Meningioma. • Other glial tumors.
• Pituitary adenoma • Meningioma.
• Acoustic neuroma.
• Craniopharyngioma. Tuberous sclerosis
• Pilocytic astrocytoma. • Subependymomas.
• Epidermoid tumor. • Glial tumors.
• Colloid cyst.
• Choroid plexus papilloma. Turcot’s syndrome
• Hemangioblastoma. Medulloblastoma.

Metastatic brain tumors Naevoid basal cell carcinoma


• Single or multiple metastases. Medulloblastoma.
• Meningeal carcinomatosis.
Ataxia telangiectasia
C. Other pathology. Medulloblastoma.

Cerebral edema von Hippel–Lindau disease


Vasogenic edema Cerebellar hemangioblastomatosis.
Vasogenic edema is excessive extracellular fluid that is caused
by an increase in permeability of the capillary endothelium Head trauma has been followed by meningiomas and
(which allows plasma to move from the intravascular even gliomas at the site of previous trauma but this is likely
compartment into the extracellular space) and the absence to be coincidental rather than causal.
of any lymphatic drainage in the brain. Consequently, the
fluid accumulates and further increases the mass effect of PATHOGENESIS
the tumor. It is the most common type of cerebral edema Oncogenesis
associated with brain tumors and occurs predominantly in • Oncogenes may be expressed inappropriately in
the cerebral white matter. neoplastic cells, and potentiate or initiate cell mitosis. For
example, oncogenes such as c-sis, c-erbB, gli, N-ras, and
c-myc are expressed in astrocytomas, and encode growth
factors, or their receptors (e.g. the B subunit of platelet-
derived growth factor is encoded by the c-sis oncogene).
Brain Tumors 359

• Tumor-suppression genes may be lost by neoplastic cells. CLINICAL FEATURES


For example, deletions have been identified on Clinical features depend on the site of origin of the tumor
chromosome 3p in hemangioblastomas due to von in the CNS, the pathologic type of tumor, the rate and
Hippel–Lindau disease, chromosomes 10 and 17 in nature of its growth, and the degree of brain edema.
astrocytomas, chromosomes 17 and 22 in acoustic
neuromas, and chromosome 22 in meningiomas. Epileptic seizures
• Many malignant tumors produce factors that stimulate The presenting feature in 20–50% of patients with brain
and induce angiogenesis, such as endothelial-cell growth tumors and may be partial or generalized.
factor, thus increasing the blood supply to the tumor and
possibly enhancing its growth. Raised intracranial pressure syndrome
• Astrocytomas secrete immunosuppressive substances, This may be a result of the mass itself, associated cerebral
such as transforming growth factor β2 which render the edema, or obstruction of the CSF pathways causing
host immune response to the tumor as inadequate. hydrocephalus:
• Tumors may cause edema in the surrounding brain, per- • Headache is a presenting symptom in about 30% of cases
haps by secreting factors that increase vascular permeability. (see Idiopathic intracranial hypertension, p.477) but
tumor is a very rare cause of headache.
Neurologic dysfunction • Vomiting may occur with or without nausea and be
The brain and spinal cord lie in the cranial cavity of the skull projectile. It is usually associated with headache.
and the spinal canal and are protected by the strong bones • Altered higher mental function and level of consciousness may
of the skull and vertebral column. However, because the manifest as increasing lethargy and a general slowing down or
cranial cavity and spinal canal is a closed inexpandible take the form of an acute and profound deterioration.
system, lesions such as tumors which grow and occupy space • Other features include an unsteady gait, urinary incon-
within this closed system ultimately compress, displace and tinence, obscurations of vision (see p.477), papilledema
cause dysfunction of normal brain and spinal cord and other (in fewer than half of patients) and false localizing signs
structures. Headache may be caused by compression and such as bilateral abducens (VIth) nerve palsies.
displacement of pain sensitive structures such as blood
vessels and meninges or by raised intracranial pressure.

437 Coronal section through the brain showing multiple metastases 437
(arrows) in the medial thalamus and the superior and medial aspect of
the contralateral temporal lobe. (Courtesy of Professor BA Kakulas,
Royal Perth Hospital,Western Australia.)

438

439

438 Axial section through the brain showing multiple areas of 439 Axial section through the pons and cerebellum showing a cystic
hemorrhage into metastases of malignant melanoma. (Courtesy of astrocytoma of the cerebellum (arrow). (Courtesy of Professor BA
Professor BA Kakulas, Royal Perth Hospital,Western Australia.) Kakulas, Royal Perth Hospital,Western Australia.)
360 Tumors of the Central Nervous System

Focal neurologic symptoms and signs MRI brain scan


These reflect the anatomic site of the tumor (e.g. blindness • More sensitive than CT, particularly for demonstrating
with optic nerve glioma) or a consequence of it (i.e. low grade astrocytomas and multiple metastases, and
compression of the posterior cerebral artery causing occipital tumors in the skull base, pituitary fossa and suprasellar
lobe infarction): region, posterior fossa, craniocervical junction and spine
• Hemiparesis, hemianopia, or dysphasia of gradual onset • Gadolinium enhanced MRI may increase sensitivity
is typical of supratentorial tumors. further.
• Altered higher mental function and behavior manifesting • MRI is therefore particularly useful for the investigation
as apathy, lack of initiative, tiredness, weakness, dullness, of patients with adult-onset epileptic seizures and no
a change in personality and social behavior, forgetfulness, other clinical or CT abnormality.
and self-neglect, may reflect focal dysfunction of the • Magnetic resonance spectroscopy can quite accurately
frontal and temporal lobes (i.e. frontal lobe or corpus predict histologic diagnosis and may, in the future,
callosum tumor) or raised intracranial pressure due to obviate the need for stereotactic biopsy.
hydrocephalus, for example.
• Ataxia, dysarthria, and brainstem and cranial nerve signs Skull x-ray
suggest a posterior fossa tumor. Can be useful if CT or MRI is not available for
demonstrating:
Associated features • Calcification within a tumor (usually benign).
May include: • Displacement of a calcified pineal gland.
• A congenital syndrome associated with tumors: • Bone hyperostosis adjacent to meningioma.
neurofibromatosis, tuberous sclerosis, von • Enlargement and erosion of the pituitary fossa
Hippel–Lindau disease. (adenoma, craniopharyngioma, carotid aneurysm).
• Local effects of tumors: proptosis, epistaxis. • Osteolytic lesions in the skull (myeloma, primary bone
• Signs of systemic malignancy: cutaneous melanoma, tumor, secondary carcinoma).
clubbing, cachexia. • ‘Copper beating’ pattern of the skull vault or separation
of the skull sutures (children) due to raised intracranial
Specific tumor syndromes pressure.
See specific tumors (pp.364–405).
Other investigations guided by the clinical setting
DIFFERENTIAL DIAGNOSIS • Chest x-ray helps to eliminate an obvious primary or
• Chronic subdural hematoma (see p.177). secondary lung tumor.
• Brain abscess (see p.284)): fever. • Mammography: to exclude primary breast carcinoma.
• Demyelination: encephalitis, multiple sclerosis (see • Abdominal ultrasound: to exclude primary renal cell
p.340). carcinoma and liver metastases.
• Idiopathic intracranial hypertension: headache,
papilledema, no focal neurologic signs (see p.477). Tumor biopsy
• Stroke: ischemic or hemorrhagic: onset is usually sudden Indications
(as it can be also with hemorrhage into a brain tumor • To establish the tissue diagnosis if there is clinical and CT
[440]) (see p.192). or MRI brain scan evidence of a solitary or even multiple
• Metabolic or toxic encephalopathy: may cause seizures intracerebral lesions which could be neoplastic.
and altered consciousness. • To exclude treatable lesions such as brain abscess,
lymphoma, meningioma, and tuberculoma.
INVESTIGATIONS
CT brain scan with and without contrast
• Images most brain tumors and their consequences such
as mass effect, cerebral edema and hydrocephalus. 440
• The tumor density may be lower, the same or higher
than that of the normal surrounding brain. Surrounding
edema appears as low density.
• Following intravenous contrast injection, most
histologically malignant and metastatic tumors enhance,
as do many benign tumors.
• CT scan with contrast images almost all tumors above
the tentorium but not below the tentorium, when the
only CT sign may be obstructive hydrocephalus or
displacement of the fourth ventricle.
• If MRI is not available, it is sometimes necessary to
introduce contrast (e.g. metrizamide or air) into the
intrathecal space, by lumbar or cisternal puncture, to
outline posterior fossa structures, provided the
intracranial pressure is not raised.
440 Axial section through the pons and cerebellum showing
hemorrhage into a malignant glioma. (Courtesy of Professor
BA Kakulas, Royal Perth Hospital,Western Australia.)
Brain Tumors 361

N.B. A vascular malformation needs to be excluded distinguish between recurrence of tumor and post-radiation
before undertaking stereotactic biopsy. necrosis.

Not necessary if DIAGNOSIS


• Patient has features suggestive of a very poor prognosis The tumor type (e.g. glioma), but not the grade, can
for survival and functional improvement, even after commonly be diagnosed confidently from the combination
multimodality therapy (e.g. substantial disability due to of the clinical features, neuroanatomic location of the tumor
progressive major neurologic or cognitive deficits; and and neuroimaging characteristics.
elderly [but biologic age and the opinions of the patient However, in practice a tissue biopsy is required to
and family need to be considered in a decision for non- exclude other differential diagnoses that can simulate a brain
interventional management]). Care is required to be sure tumor (i.e. abscess, other inflammatory lesions, metastases,
that a treatable condition such as brain abscess or and other contrast-enhancing neuropathologies) and to
lymphoma has been excluded. establish the histologic diagnosis and grade of the tumor, so
• CT or MRI brain scan reveals multiple deposits that realistic discussions can take place with the patient and
characteristic of metastases and there is a known, family about the prognosis and management decisions. For
histologically-confirmed, primary tumor. example, a proportion of patients with apparently innocuous
• Characteristic clinical and neuroradiologic features of a focal lesions on MRI, and many patients diagnosed by CT
glioblastoma. to have low grade astrocytomas, will be shown to have a
non-contrast-enhancing malignant anaplastic astrocytoma
Can be delayed if with a worse prognosis.
Epileptic seizures only (which are mild or controlled by anti-
epileptic drug therapy) and a solitary intracerebral lesion. TREATMENT
Serial clinical assessment and CT scan is required to monitor A multidisciplinary neuro-oncology team approach
the clinical progress and rate of tumor growth. Biopsy is (neurosurgeons, radiation therapists, neuro-oncologists,
indicated if there is any deterioration. endocrinologists, rehabilitation teams, neuropsychologists,
social workers, nurses) is required.
Risks
MR or CT guided stereotactic biopsy has a morbidity of General
<5% and mortality of <1%; the 30 day morbidity associated Malignant brain tumor
with biopsy is usually transient or related to the rapid • Education of patient and family: involve GP, liaison
progression of the primary disease process. nurses, and palliative care team as appropriate.
• If the patient is considered to have a very poor prognosis,
CSF good primary medical practice (including judicious use
Seldom useful and lumbar puncture can be dangerous in the of glucocorticoid treatment) and the use of home
presence of an intracranial mass because of the risk of nursing and hospice care are generally appropriate. The
transtentorial herniation and/or brainstem coning. facility for additional neuro-oncologic follow up should,
however, be retained as an option as even without
Positron emission tomography (PET) treatment some of these patients survive longer than
Reveals areas of increased metabolism in tumors that are expected, and are at risk of major steroid-related
actively growing and this can be helpful when trying to complications and even misdiagnosis.

Table 37 Treatment and prognosis of brain tumors


Tumor Surgery Adjuvant therapy Prognosis
Glioblastoma Biopsy/PR RT ± CH for recurrence ≤12 months median survival
‘Low-grade’ glioma CR/PR ± RT – 5-year survival: 50%
Pilocytic astrocytoma CR – 5-year survival: 100%
PR – 5-year survival: >80%
Cerebral lymphoma Biopsy RT ± CH 12–18 months median survival
Metastases CR/PR RT 4 months median survival
Medulloblastoma/PNET CR RT ± CH for recurrence 5-year survival: >50%
Meningioma CR RT if aggressive Recurrence 1%/year even after ‘CR’
Acoustic neuroma CR – Excellent if CR
Craniopharyngioma CR/PR ± RT/brachytherapy Good if CR
± Cyst aspiration
Pituitary adenoma CR/PR ± RT Good long term survival
CR: complete resection; PR: partial resection; RT: radiotherapy; CH: chemotherapy; PNET: primitive neuroectodermal tumor
362 Tumors of the Central Nervous System

Medical • Solitary brain metastasis may be completely removed. This


Raised intracranial pressure, transtentorial herniation and provides a tissue diagnosis in the one-third of patients who
impending brainstem coning have no evidence of systemic cancer, achieves good
• Mannitol 0.5–1.0 g/kg intravenously as a 10–20% palliation of symptoms, and improves median survival in
solution over 10–20 minutes may ‘buy time’ to arrange those with stable extracranial disease from 7 months with
surgical decompression if considered appropriate. Beware radiotherapy alone to 12 months, and functionally
of precipitating acute heart failure, particularly in the independent survival from 4 months to 9 months. Older
elderly, due to fluid overload. Infusions should be age (>60 years) increases the hazard of dying by 2.7 times,
repeated no more than four or five times within 48 hours compared with younger patients, in both treatment groups.
because of electrolyte imbalance.
• Consider urgent CSF drainage via a burr hole. Partial removal/debulking
Infiltrative malignant tumors can be removed partially or
Cerebral vasogenic edema debulked (remove necrotic tissue and reduce the mass
• Dexamethasone 12 mg orally as a loading dose followed effect) quite safely and effectively with adjuncts such as
by 4 mg every 6–24 hours and gradually reducing over MRI, stereotactic craniotomy and operating microscopes.
a few weeks. A favorable response is often evident clini- The purpose is to establish a histologic diagnosis, relieve
cally within 24–48 hours, and sometimes within 4 hours. symptoms of raised intracranial pressure (and thereby reduce
• Analgesics and anti-emetics for headache, nausea and corticosteroid requirements, seizure frequency and intensity,
vomiting. Take care not to sedate the patient because this and focal neurologic deficits), and reduce tumor bulk before
complicates the evaluation of a genuine change in radiotherapy. However, it is controversial whether median
conscious level and other neurologic functions. survival times are improved.
Any benefits of cytoreductive surgery probably become
Epileptic seizures less with increasing age of the patient and increasing tumor
Anti-epileptic drug therapy with the usual medications such malignancy. The limited survival benefit bestowed by even
as valproate or carbamazepine, and advice about driving (see the most exacting cytoreductive operation for malignant
p.76). Anti-epileptic drug treatment is not required as glioma, and particularly glioblastoma, is related to the
‘routine prophylaxis’ if seizures have not occurred. invasive and infiltrative potential of malignant glioma.
Generally, peritumoral invasion extends for 2 cm (0.8 in)
Specific antitumor chemotherapy beyond either the macroscopic tumor margin or the borders
• Lymphoma: if widespread systemic disease. defined by enhanced CT. In most locations the likelihood
• Leukemia. of iatrogenic brain dysfunction prohibits excision of
• Choriocarcinoma. infiltrated peritumorous brain. Even when extensive
• Pituitary tumors (see p.379): e.g. bromocriptine, peritumorous brain resection has been undertaken tumor
carbergoline. recurrence at the margin of resection is almost inevitable.
• Primary malignant brain tumors: see Gliomas, p.364.
Ventriculoperitoneal shunt
Surgery Can relieve obstructive hydrocephalus.
Surgery has two roles: (1) to establish the histologic
diagnosis of the tumor; and (2) to treat the patient by Radiotherapy
excising or debulking the tumor, and decompressing the Indications
brain (i.e. ventriculoperitoneal shunt) if appropriate. • Radiosensitive tumors such as pineal germinomas,
lymphoma and leukemic deposits are usually treated with
Establish histologic diagnosis external beam brain irradiation. An optimal total dose of
• Stereotactic surgery under CT or MRI guidance for 50–60 Gy (5000–6000 rads or cGy) in 30 fractions over
deep-seated tumors, including some gliomas and many 5–6 weeks is conventional. The addition of a radiosensi-
lymphomas, or tizer during radiotherapy has not significantly improved
• Open craniotomy or burr hole: a preliminary diagnosis survival times. Doses >60 Gy (>6000 rads) are associated
can be made at the time of the operation based on smear with a high incidence of acute and short term adverse
and frozen section techniques, and a definitive diagnosis neurologic effects and later cerebral radionecrosis with
based on paraffin sections. no benefit in either median or longer term survival.
• Tumors unamenable to surgery such as multifocal
Complete removal lymphoma or diffuse brain stem glioma.
• Benign capsulated tumors, such as meningiomas, • Following surgery for glioma or brain metastases (see
pituitary tumors, acoustic neuromas and choroid plexus pp.364, 396).
papillomas, can often be completely excised but the • Multiple brain metastases: radiotherapy is the standard
benefits of complete removal need to be weighed against treatment, resulting in a median survival or 3–7 months,
the possible resultant neurologic deficit (e.g. with depending on prognostic factors.
meningiomas of the cavernous sinus). • Medulloblastoma in childhood: postoperative
• Cerebellar pilocytic astrocytoma in childhood can be craniospinal irradiation is given to reduce the risks of CSF
cured if removed totally. tumor seeding.
• Medulloblastoma and ependymoma which are
completely resected have the best long term results.
Brain Tumors 363

• Stereotactic radiosurgery (focused radiation given to a Secondary malignant neoplasia:


stereotactically localized area of the brain; e.g. proton • Secondary CNS tumors, particularly meningiomas and
beam gamma knife) for treatment of metastases and malignant gliomas.
recurrent glioma. Less likely to cause these adverse effects • Thyroid tumors.
of irradiation but preliminary experience for malignant • Bone, soft tissue and skin tumors.
glioma is disappointing.
New experimental approaches
Adverse effects Suicide genes, such as herpes simplex thymidine kinase (HS-
• Determined by the total dose given, the number of tk), have been transfected into tumor cells using a retroviral
fractions given, the inherent vulnerability of the exposed vector. The transfected cells are rendered sensitive to the
tissue, the amount of irradiation absorbed by the exposed cytotoxic effects of ganciclovir.
tissue, and the time that has elapsed since the irradiation.
• Acute reactions may occur within days of an excessive PROGNOSIS
dose (i.e. during treatment), sometimes as low as 30 Gy • Benign tumors: complete removal achieves a cure. Even
(3000 rads). Transient exacerbation of the neurologic with incomplete removal, prolonged survival is possible
deficit and headache, nausea and vomiting occur due to with repeated operations and adjuvant therapy.
cerebral edema, which responds to corticosteroid • Malignant tumors: prognosis is poor, despite surgery and
treatment, but higher doses can result in coma and radiotherapy; therefore, palliation of distressing
death. symptoms is often the goal of therapy.
• Early delayed reactions may occur rarely several weeks • Anaplastic astrocytoma: median survival time with
after irradiation. Focal neurologic deficits evolve radiotherapy and chemotherapy: 36–48 months.
progressively due to focal demyelination, which usually • Mixed anaplastic astrocytoma and glioblastoma: median
recovers. survival time with brain irradiation: 9–11 months; 10%
• Late delayed reactions may occur months or years after of patients with glioblastoma survive 2 years.
total doses of irradiation exceeding 50 Gy (5000 rads). • Secondary deposits from the bronchus, gastrointestinal
Epileptic seizures and focal neurologic deficits, including tract and melanoma have a worse prognosis than breast
optic nerve and chiasm dysfunction, evolve progressively or renal metastases to the brain.
due to focal brain necrosis and edema caused by
progressive occlusive radiation vasculopathy. The PROGNOSTIC FACTORS
condition can be very difficult to differentiate clinically Favorable prognostic factors for adult supratentorial
from tumor recurrence and may stabilize or progress. tumors
• Late reactions may occur years after even low doses of • Epileptic seizure as the initial presenting symptom.
radiotherapy and manifest as intracranial tumors: • Young age (<45 years).
schwannomas and meningiomas in particular, and • Absence of focal neurologic signs (i.e. hemiparesis).
gliomas to some extent. • Absence of mental signs (confusion, altered awareness,
personality change).
Late sequelae related to craniospinal irradiation • Absence of contrast enhancement on cranial CT scan.
Neuropsychologic: • Presence of cystic change on CT (a circular low density
• Diminished memory. area before enhancement, with clear cut margin).
• Diminished intellect. • Presence of diffuse low density on CT (diffuse, poorly
• Diminished academic achievement. demarcated low density without contrast enhancement).
• Emotional adjustment. • Presence of calcification on CT.
• Adaptic behavior.
N.B. Histologic grading of tumors may not be accurate
Neuroendocrine: in determining the prognosis. Furthermore, error can occur
• Growth hormone deficiency. when a small sample is taken for biopsy and the examined
• Hypothyroidism (decreased thyroid stimulating hormone tissue does not reflect the biology of the entire tumor,
[TSH]). particularly if features indicative of malignancy are missed.
• Hypogonadism (decreased follicle stimulating hormone
[FSH], decreased luteinizing hormone [LH]).

Skeletal:
• Diminished height due to vertebral body hypoplasia.

Cardiac:
• Cardiomyopathy.

Ocular:
• Cataract formation.
364 Tumors of the Central Nervous System

GLIOMAS About 80% of gliomas are ‘high grade’ (malignant,


anaplastic), and 20% ‘low grade’.

DEFINITION Glioblastoma multiforme


A glioma is any neuroepithelial tumor of the brain and arises Macroscopic
from glial cells such as astrocytes (astrocytoma and other • Sites: about half occupy more than one lobe or are
astroglial neoplasms), oligodendrocytes (oligodendro- bilateral, and 5% show multicentric foci of growth.
glioma) and ependymal cells (ependymomas). The term • Appearance: variegated: mottled gray, red, brown or
does not imply any degree of differentiation. orange, depending on the degree of necrosis and the
presence, degree and age of hemorrhage (441).
CLASSIFICATION OF GLIOMAS ACCORDING TO • Highly vascular.
NEUROEPITHELIAL CELLS OF ORIGIN • Mass effect.
• Astrocytes.
– Astrocytoma (25–30% of brain gliomas): Microscopic
Pilocytic astrocytoma. • Very cellular with pleomorphism of cells and
Low grade astrocytoma. hyperchromatism of nuclei.
Anaplastic astrocytoma. • Astrocytes with fibrils and astroblasts, tumor giant cells,
Pleomorphic xanthoastrocytoma. mitotic cells.
Subependymal giant cell astrocytoma. • Vascular proliferation with hyperplasia of endothelial cells
– Glioblastoma multiforme (50% of gliomas). of small vessels.
• Oligodendrocytes: • Necrosis, hemorrhage and thrombosis of vessels.
– Oligodendroglioma (10–20% of gliomas).
– Anaplastic oligodendroglioma. Growth
• Ependymal cells: • Arises from anaplasia of mature astrocytes.
– Ependymomas (5% of all brain gliomas, 8% of gliomas in • Highly malignant.
childhood). • May extend to the meningeal surface or the ventricular wall.
– Anaplastic ependymoma. Malignant cells, carried in the CSF may form distant foci on
– Myxopapillary ependymoma. spinal roots or cause widespread meningeal gliomatosis.
– Subependymoma. • Extraneural metastases are rare, and usually involve bone
• Choroid plexus tumors (papilloma, carcinoma). and lymph nodes after craniotomy.
• Colloid cysts.
• Mixed gliomas (oligoastrocytoma, anaplastic oligoastro- Anaplastic astrocytoma
cytoma). • Moderate hypercellularity and pleomorphism.
• Vascular proliferation.
EPIDEMIOLOGY
The most common primary brain tumors in adults. Astrocytoma (grades 1 and 2)
• Incidence: 6 (range 2–10) per 100 000 per year. Macroscopic
– Gender-specific incidence (per 100 000 per year): • Sites: anywhere in the CNS, particularly the cerebral
Males Females hemispheres (usually in adults, aged 20–40 years) and
Glioblastoma 2.8 1.8 the cerebellum (442), hypothalamus, optic nerve and
Astrocytoma 1.7 1.3 chiasm and pons (the latter of which are more common
Oligodendroglioma 0.15 0.10 in children and adolescents).
Ependymoma 0.15 0.15 • Appearance: solid, grayish white, firm, relatively avascular
tumor, almost indistinguishable from normal white
• Age: any age: matter. Calcium deposits may be present.
– Glioblastoma: peak incidence in middle adult life: mean • Often forms large cavities or pseudocysts.
age 56 years.
– Anaplastic astrocytoma: mean age 46 years. Microscopic
– Oligodendroglioma: any age: most often 20–40 years; • Well differentiated astrocytes of fibrillary type and less
early peak at 6–12 years. frequently plump gemistocytic (gemistos-filled) type.
– Ependymoma: peak incidence at 5 years of age: • Mildly hypercellular, with pleomorphism (443).
Supratentorial: any age. • No vascular proliferation or necrosis.
Infratentorial: 40% occur in the first decade of life; make • Glial fibrillary acidic protein.
up 5–10% of childhood tumors. • Many cerebral astrocytomas are mixed astrocytomas and
• Gender: M>F (2:1). glioblastomas.

PATHOLOGY Growth
• Malignant tumors of glial cells. • A slowly growing, infiltrative tumor.
• Tumors are graded in various ways. A simple and • May transform to a higher grade tumor.
reproducible method is based on four histologic features:
pleomorphism of cells and presence or absence of nuclear Oligodendroglioma
atypia, mitosis, endothelial/microvascular proliferation Many oligodendrogliomas have deletions of chromosomes 1p
and necrosis. Any two features classify a tumor as grade and 19q, and molecular changes such as these may prove to
3, and three or four features constitute a grade 4 tumor. be the defining criteria for this kind of tumor.
Gliomas 365

Macroscopic • May extend to the pial surface or ependymal wall, and


• Sites: commonly frontal lobe (40–70%), deep in white metastasize distally in the ventriculosubarachnoid spaces
matter; sometimes lateral ventricle; and rarely in other (accounts for about 10% of gliomas with meningeal
parts of the CNS. dissemination; less frequent than medulloblastoma and
• Appearance: multi-lobular, pink-gray, moderately firm, glioblastoma).
and relatively avascular. • Malignant degeneration (greater cellularity and numerous
• Tends to encapsulate and form calcium and small cysts. and abnormal mitoses) in about a third of cases.
• Little or no surrounding edema.
Ependymoma
Microscopic Macroscopic
• Oligodendrocytes: small, round nucleus and a halo of • Sites: most commonly the wall of the fourth ventricle (70%)
unsustained cytoplasm. Cell processes are few and stubby, (444); other sites include the lateral ventricles of the brain
seen only with silver carbonate stains. and the conus or filum terminale of the spinal canal.
• Calcification is common, mainly in relation to zones of • Appearance: gray-pink, cauliflower-like, firm tumors in
necrosis. the fourth ventricle; large (up to several centimetres in
• Myelin basic protein. diameter), gray-red, softer tumors in the lateral ventricles.
• Many oligodendrogliomas are mixed oligodendroglioma- • May be cystic.
astrocytomas. • Not encapsulated but well defined and homogeneous.
• More clearly demarcated from surrounding brain than
Growth astrocytomas.
• Slow.

441 442

441 Coronal section of the brain at autopsy showing a large 442 Autopsy specimen, section in the axial plane through the
hemorrhagic and necrotic glioblastoma multiforme in the temporal cerebellum and pons, showing a cystic cerebellar astrocytoma in an
lobe causing mass effect with compression of the lateral ventricle and adult. On CT scan (see 452), the mass is partly enhancing, partly cystic
shift of the midline. and has some calcification.

443 444

443 Histologic section of astrocytoma of the brain showing 444 Section through the medulla and floor of the fourth ventricle at
hypercellularity, pleomorphism of cells and mitoses (arrow). autopsy showing an ependymoma in the floor of the fourth ventricle
(arrows).
366 Tumors of the Central Nervous System

Microscopic • CT brain scan shows small ventricles and multifocal areas


• Cells tend to form canals (rosettes) or circular arrange- of low attenuation (despite the diffuse brain involvement
ments (pseudorosettes). by tumor).
• Some are densely cellular and anaplastic (epithelial ependy-
momas); others are more differentiated and form papillae. Choroid plexus papilloma
• Ciliary bodies. • Rare: one-fifth as common as ependymoma.
• Occur mainly in childhood: 75% in the first decade.
Growth • Arises from plexus epithelium in the lateral (50%) and
• Derived from differentiated ependymal cells lining the fourth (40%) ventricles commonly, and the third (10%)
ventricles of the brain and the central canal of the spinal ventricle occasionally.
cord. • Takes the form of a giant choroid plexus. May increase
• Grow into the ventricle or adjacent brain tissue, and CSF formation.
fourth ventricle ependymomas may extend through the • Benign usually.
foramina of Luschka and Magendie. • Occasionally there is carcinomatous transformation with
• Neuraxis dissemination if uncommon (<10%). CSF seeding and malignant meningitis.
• Presents with raised intracranial pressure (often
ETIOLOGY (see Brain tumors, p.357) aggravated acutely by hemorrhage) due to obstructive
Astrocytoma Primary genetic changes Secondary changes hydrocephalus or, less commonly, cerebellar or brainstem
Astrocytoma p53 mutation signs if the tumor is in the fourth ventricle.
Chromosome 22q loss • The CSF protein is raised.
Anaplastic p16 gene loss/inactivation E.g.VEGF, • Treatment is surgical: excision (which can be difficult)
astrocytoma Chromosome 19q loss proteinases and/or shunting.
Glioblastoma EGFR mutation of over-expression
multiforme p16 deletions Colloid cyst
Mutations in the gene for phosphatase • Rare.
and tensin homologues (PTEN) • Congenital.
Chromosome 10 loss • Site: anterior portion of third ventricle, between the
Overexpression of platelet-derived interventricular foramina and attached to the roof of the
growth factor A and its receptor, third ventricle (445); derived from ependymal cells.
platelet-derived growth receptor α • Size: usually small (1–4 cm [0.4–1.6 in] in diameter).
EGFR: epidermal growth factor receptor • Shape: oval or round, smooth external surface, filled with
VEGF: vascular endothelial growth factor gelatinous material containing a variety of mucopoly-
saccharides.
CLINICAL • Histologically benign.
• Epileptic seizures (partial or generalized) are the present- • Neurologically asymptomatic usually.
ing symptom in at least half of patients with oligodendro- • May present in adult life with symptoms of hydro-
glioma and low grade (grades 1 and 2) astrocytoma, and cephalus which may be acute (paroxysms of generalized
develop later in another 10–25% of such patients. headache that is markedly affected by head position, and
• Focal neurologic symptoms and signs occur referable to sometimes with loss of consciousness) or chronic
the site of the tumor (e.g. temporal lobe: altered (dementia and gait disturbance).
personality, mood, character, complex partial seizures; • CT scan shows a hyperdense cyst that may enhance with
frontal lobe: mild hemiparesis, word-finding difficulty; i.v. contrast (446, 447).
cerebellum: limb and gait ataxia). Usually the signs are • Differential diagnosis includes craniopharyngioma,
not confined to one lobe and it can be difficult enough choroid plexus papilloma, and ependymoma.
to localize the lesion to one region of a hemisphere. • Treat, if necessary, with ventriculo-peritoneal shunt or
• Diffuse encephalopathy (e.g. cognitive decline, headache), decompression of the cyst by aspiration under
evolving over a few weeks or months in about a third of stereotactic guidance.
patients with glioblastoma and anaplastic astrocytoma.
• Headache and other features of raised intracranial pressure. Optic nerve glioma (see p.372)
• Cranial or spinal radiculopathies due to meningeal
oligodendrogliosis. OTHER NEUROEPITHELIAL TUMORS
• The onset of symptoms is occasionally abrupt, due to Medulloblastoma
hemorrhage into the tumor (e.g. glioblastoma, • Accounts for about 20% of all childhood brain tumors
oligodendroglioma), or rapid expansion of edema. and 50% of cerebellar tumors in children.
• Symptoms have often been present for 3–6 months • Arises from primitive neuronal cells, usually in the
before the diagnosis is made. midline cerebellar vermis (448).
• May fill the cavity of the fourth ventricle (usually
RARE GLIOMAS posteroinferiorly) and cause hydrocephalus (449),
Gliomatosis cerebri infiltrate the floor of the fourth ventricle or adjacent
• Rare. cerebellar hemispheres or brainstem. Medulloblastoma
• Diffuse infiltration of an entire hemisphere or the entire brain. may seed through the cranial or particularly the spinal
• No discrete tumor mass. subarachnoid space, via the CSF, or metastasize to bone,
• No distinctive clinical syndrome; usually non-specific bone marrow or lymph nodes (rare).
cognitive decline, headache, seizures, and papilledema.
Gliomas 367

• Symptoms include raised intracranial pressure, cerebellar dysfunction, 445


or back pain with signs of spinal cord compression or nerve root
dysfunction.
• When presenting in older patients (20% occur in young adults), about
half are lateral within the cerebellar hemisphere. Calcification is seen in
20% (449), cystic change in 60–80%.
• Appears hyperdense on CT (449), may contain calcification and may
enhance moderately with contrast.
• On T1WI they are hypointense and on T2WI isointense. Midline
sagittal views are best for visualizing the origin from the superior velum
of the fourth ventricle. Contrast enhancement is moderate.
• Contrast MRI is the best method of detecting seedlings.
• Treatment is by surgery with gross total resection.
• Radiotherapy is comprised of craniospinal axis treatment to 36 Gy
(3600 rads), and 54 Gy (5400 rads) to the primary site. Neuro-
endocrine or neuropsychologic sequelae may be anticipated in younger
patients receiving craniospinal irradiation.
• Chemotherapy is effective in high risk patients.
• 60–80% of patients experience long term disease-free survival.
• The chance of survival is reduced in patients with CSF or neuraxis
dissemination or extra-axial disease.

445–447 Cranial CT scan in 446 447


the axial (446) and coronal
(447) planes, and autopsy
brain specimen cut in the
axial plane (445) showing a
midline colloid cyst in the
third ventricle in the region
of the foramen of Monro
(445) (arrows).

449
448 T1W MRI with contrast 448
of a medulloblastoma in a 2
year old.The lesion arises
posteroinferior to the fourth
ventricle.

449 CT scan without


contrast of the
medulloblastoma in 448.
Note the high density and
calcification.There is
secondary hydrocephalus.
368 Tumors of the Central Nervous System

Pineal cell tumors (pinealomas) • Enhancement may be diffuse or ring-like (455).


• Arise from the pineal gland.
• May compress the rostral dorsal midbrain tectum causing Oligodendroglioma
Parinaud’s syndrome (dilated pupils which do not react • Common sites are the frontal and temporal lobes and
to light but may continue to react to accommodation, within the ventricles.
supranuclear impairment of upward gaze and vergence, • Mass effect and low density on CT (unless calcification
and retractory nystagmus). or hemorrhage have occurred) are typical, and in the less
• Compression of the cerebral aqueduct leads to malignant forms white matter edema is absent.
hydrocephalus and raised intracranial pressure. • Calcification is frequent (456).
• Hemorrhage and cyst formation appear in 20%.
INVESTIGATIONS • Skull erosion may occur.
Brain imaging (CT and MRI scan) • Enhancement occurs in 50%.
Astrocytoma and glioblastoma • On MRI they appear hypointense on T1WI and
• The frontal and temporal lobes are common sites, but hyperintense on T2WI.
they may be supra-tentorial (450, 451) or infra-tentorial • The overall appearance depends on the mix of more
(452–454). malignant cells with the purer oligodendrocytic elements.
• Appear as mixed component lesions with mass effect,
white matter edema and rapid growth. Ependymoma
• The central necrosis of malignant glioblastoma is shown • In children, most common in the fourth ventricle (444,
as a low density area on CT. 457, 458).

450 451 450, 451 CT with contrast


(450) and T2W MRI (451)
of a left occipital low grade
astrocytoma. Note how
difficult it is to see on CT, and
the minimal mass effect.

452 453 452 CT scan with contrast


showing a cystic cerebellar
astrocytoma in an adult.The
mass is partly enhancing,
partly cystic and has some
calcification.The autopsy
specimen is shown in 442.

453, 454 CT with contrast


showing a brain stem glioma
(453) extending into the
posterior part of the corpus
callosum(454). Note the low
density in the brainstem and
the thickened low density of
the corpus callosum (arrows).
Gliomas 369

• In teenagers and adults, more common in the lateral ventricles or brain 454
parenchyma (20%).
• 40–50% contain calcification, typically punctate.
• Tend to spread by seeding throughout the subarachnoid space.
Contrast is essential to detect seed metastases.
• Frequently causes hydrocephalus.
• Enhances mildly with contrast.
• On MR they appear hypointense on T1WI and intermediate on T2WI.
• Ependymomas also occur in the spine, accounting for 50–60% of spinal
cord tumors in 20–40 year olds, and 25% of spinal cord tumors in
children (see Spinal cord tumors, pp).

CSF
• Pressure: increased in some cases.
• Cells: acellular or occasional pleocytosis of 10–100 cells/mm3 or more,
mostly lymphocytes, occasional blood.
• Protein: normal or mildly increased in low grade astrocytoma, increased
>1.0 g/l (100 mg/dl) in many cases of glioblastoma.
• Cytology: malignant cells may be identified.

454 CT with contrast 455 456


showing a brain stem glioma
extending into the posterior
part of the corpus callosum.
Note the low density in the
brainstem and the thickened
low density of the corpus
callosum (arrows). See also
453.

455 CT brain scan with


contrast of a primary tumor,
probably an astrocytoma.
Note the central necrosis,
thick rim of enhancement
and surrounding edema
(arrows).

456 CT scan with contrast


of a frontal oligodendro-
glioma. Note the calcification
(whiteness) and considerable 457 458
mass effect.The tumor has
reached this size partly
because of its position in the
relatively ‘silent’ frontal lobes.
There is relatively little
edema.

457, 458 CT brain scan


with contrast (457) and
PDW MRI (458) of an
ependymoma.There is a large
enhancing mass closely
related to the fourth
ventricle without calcification.
370 Tumors of the Central Nervous System

Pathology Chemotherapy is probably not curative in patients with


Tissue biopsy low-grade oligodendroglioma but it can produce sus-
Aims to establish the tissue diagnosis if there is clinical and tained remissions with durable clinical improvement.
CT or MRI brain scan evidence of a mass lesion and to • Eventually, most oligodendrogliomas, like astrocyomas,
exclude treatable differential diagnoses such as brain abscess, progress and become malignant.
lymphoma, meningioma, and tuberculoma. About 50% of
patients between 30 and 50 years of age who have a non- Ependymoma
enhancing lesion on CT suggestive of a low grade glioma turn • Median survival: 12 months.
out to be harboring a high grade malignant glioma. These • 5-year disease-free survival: 30–60%.
patients should undergo early biopsy or surgical resection for: • 10-year survival; 10–15%.
• Histology.
• Cytology. Prognosis is worse with young age, incomplete resection,
• Immunohistochemistry. metastases, brainstem invasion, or radiation therapy doses
• DNA index: a rapid estimate, using flow cytometry <45 Gy (<4500 rads).
techniques, of the DNA content of a tumor cell
compared with that of a normal diploid cell. It may Prognostic factors
identify patients who may benefit from more intensive Favorable
therapy for certain tumors (e.g. medulloblastoma). • Good clinical performance status (WHO): independent
in activities of daily living and self care: 80% chance of
Biochemical tests survival free of disability for 6 months.
Tumor markers in serum and CSF (e.g. alpha fetoprotein • Young age at diagnosis.
and beta-human chorionic gonadotrophin [βHCG]). • History of epileptic seizures as a presenting feature.
• Partial or complete surgical resection compared with
DIAGNOSIS biopsy alone.
Based on appropriate clinical features and imaging studies • Low tumor grade.
followed by tissue biopsy or surgery. • Absence of necrosis.
• Complete surgical resection.
PROGNOSIS • Treatment with radiotherapy.
Glioblastoma
• Median survival: Poor
– 14 weeks if steroids and maximally feasible surgical • Poor clinical performance: functionally disabled and
resection only. dependent (e.g. often confined to bed or chair) at the
– 35 weeks (i.e. an additional 5 months) with a combin- time of treatment.
ation of surgery and radiation therapy. Adjuvant chemo- • Increasing age.
therapy modestly increases survival at 1 year but this • Tumor histology (anaplastic astrocytoma or glioblastoma
disappears by 2 years. multiforme) and grade (high).
• 18-month post operative survival: 15%.
• 2-year survival after treatment: 5–10%. These data mean that patients older than 60 years of age
who are disabled due to glioblastoma multiforme will derive
Anaplastic astrocytoma little benefit from aggressive treatment whereas a 35 year
• Median survival: 27 months–3 years. old with minimal neurologic deficit and post-resection
• 18-month post operative survival: 62%. histology showing anaplastic astrocytoma has a high
• 2-year survival: 50%. probability of remaining well for 18 months or longer if
treated with the optimal schedule of high dose radiotherapy.
Astrocytoma
Mean survival after first symptom: TREATMENT
• Cerebral astrocytoma: 67 months. • A multidisciplinary neuro-oncology team approach
• Cerebellar astrocytoma: 89 months. (neurosurgeons, radiation therapists, neuro-oncologists,
• Cystic cerebellar astrocytoma: particularly benign. endocrinologists, rehabilitation teams, neuro-
Surgical excision of juvenile cerebellar astrocytoma leads psychologists, social workers, nurses) is required.
to a 10-year survival rate of >80%. Indeed, 10% survive • Indicated if the benefits of treatment (e.g. survival with
25 years after excision of the cyst. an acceptable quality of life) are likely to outweigh the
• Pilocytic astrocytomas of the cerebral hemispheres: gross risks and toxicity of treatment.
total removal of the tumor can improve 5-year survival • Determined by the age and functional status of the
to 85% and 10-year survival to about 80%. patient, and the site and pathology of the glioma. For
some gliomas such as optic gliomas (which are frequently
Oligodendroglioma associated with neurofibromatosis), thalamic glioma or
• Survival is variable, from weeks to many years, after brainstem gliomas, the site clearly restricts treatment
surgical confirmation of the histologic diagnosis median options. The site also influences biologic behavior. For
survival – 16 years. example, cerebellar gliomas behave differently from
• Recently recognized to be uniquely sensitive to chemo- supratentorial tumors with the same histology.
therapy: 75% of patients with malignant oligodenroglioma
now respond to treatment with a regimen of procabazine,
lomustine and vincristine, and 50% recover completely.
Gliomas 371

Glioblastoma and anaplastic astrocytoma • Two randomized, controlled trials have demonstrated
Surgery equivalence in survival and time to disease progression
• Only partial resection is possible due to the multicen- between patients with low-grade astrocytoma who
tricity and the diffusely infiltrative nature of the tumor. received focal radiotherapy at low dose (50.4 or 45.0 Gy
• Attempted gross total resection is associated with longer [5040 or 4500 rads]) at the time of diagnosis and those
survival and improved neurologic function. who received a high dose (64.8 or 59.4 Gy [6480 or
• Median survival is about 14 weeks if steroids and 5940 rads]). However, higher doses of radiation were
maximally feasible surgical resection only are given. associated with a higher incidence of fatigue, malaise,
insomnia, and poor emotional functioning, suggesting
Radiotherapy that lower doses are the superior treatment.
• A total of up to 60 Gy (6000 rads) given in fractions of • Immediate radiotherapy (after surgery or biopsy) is
1.8–2.0 Gy (180–200 rads) each weekday for 3–6 weeks associated with a significantly delayed time to disease
significantly prolongs survival. progression, compared with deferral of radiotherapy, but
• Median survival is about 35–38 weeks (i.e. an additional does not improve overall survival. Consequently, patients
5 months) if radiation therapy is combined with surgery. with low-grade astrocytoma who are neurologically
normal or whose seizures are controlled, whether or not
Chemotherapy they have undergone tumor resection, should be
• Various chemotherapy regimes have been used but none followed until there is evidence of tumor progession.
has consistently been shown to have a substantially • Some astrocytomas, such as those causing focal neurologic
favorable influence on outcome. Single agent nitrosurea signs (e.g. hemiparesis), require immediate intervention
therapy or BCNU (carmustine) or temozolomide is used (surgical debulking followed by radiotherapy to the
widely as adjunctive therapy after surgery and radiotherapy, involved field, with a total dose ≤54 Gy [≤5400 rads]).
or after glioma recurrence, with a small improvement in There is no indication for the routine use of chemotherapy
survival. A combination of drugs (e.g. procarbazine, in the treatment of astrocytomas.
lomustine, and vincristine [PCV]) can achieve somewhat
longer survival if there is an oligodendroglial component. Chemotherapy
• Relapse is invariably due to recurrence within 2 cm There is no indication for the routine use of chemotherapy
(0.8 in) of the original lesion. This is because malignant in the treatment of astrocytomas.
glioma cells have an inherently high chemoresistance
(and radioresistance) with a large capacity for DNA repair Oligodendroglioma
and because of failure of drug delivery. Recently recognized to be uniquely sensitive to chemotherapy.
• Therapeutic approaches designed to improve drug
delivery include supraophthalmic intracarotid arterial Surgery
infusion, osmotic opening of the blood–brain barrier, Excision of anaplastic oligodendrogliomas.
and the use of implantable polyanhydride polymers
impregnated with chemotherapeutic agents. Radiotherapy
• After surgery, for mixed oligodendrogliomas and
Astrocytoma astrocytomas (i.e. as for astrocytomas).
Surgery • Total dose of 54 Gy improves symptoms and probably
• For young patients (<35 years of age), extensive surgical prolongs survival.
resection generally leads to an improved outcome, if it can be • Not indicated for well-differentiated oligodendrogliomas
performed safely. They can be followed without administering (cf. Chemotherapy).
radiotherapy. If surgery is not performed, the patient’s pro-
gress should be followed closely. Subsequent indications for Chemotherapy
surgery include a need for a tissue diagnosis, clinical neuro- • Effective for anaplastic oligodendrogliomas: PCV.
logic deterioration that can be treated surgically, impending • Chemosensitivity linked to loss of heterozygosity at
herniation, hydrocephalus and uncontrollable seizures. chromosomes 1p and 19q.
• Older patients (>35 years ) should undergo early biopsy • Agents that appear to have some efficacy against recurrent
or surgical resection. The extent of surgical resection is oligodendroglial tumors include melphalan, thiotepa,
probably not as important as it is in younger patients, temozolomide, carboplatin, cisplatin, and etoposide.
partial excision (especially the cystic part) may preserve
survival free of dependency for several years. Ependymoma
Surgery
Radiotherapy Surgery (gross total resection) followed by radiotherapy.
• Improves survival. Controversies concerning radiation fields (local field versus
• Radiotherapy should be given to most patients older than craniospinal treatment) are ongoing.
35 years, independent of the extent of surgical resection,
because this is associated with a 5-year survival of 30% or more. Chemotherapy
• The radiation field should be limited to the tumor bed Chemotherapy may be of value in infants to defer radiation
with large margins; whole brain irradiation increases the therapy in patients with recurrent disease Chemotherapy
likelihood of late radiation damage. combined with radiotherapy may also be useful for cerebral
ependymoblastomas.
372 Tumors of the Central Nervous System

OPTIC NERVE GLIOMA • Meningioma (see p.374): medial sphenoid, olfactory


groove, intraorbital.
• Pituitary adenoma (see p.379).
DEFINITION • Diencephalic glioma: gliomas arising in the hypothalamus
Glial tumor (usually pilocytic astrocyoma) of the optic pathway. or thalamus.
• Hand–Schuller–Christian disease.
EPIDEMIOLOGY • Craniopharyngioma (suprasellar epidermoid cyst) (see
• Incidence: uncommon: about 1% of primary intracranial p.377).
tumors; about 2–5% of all brain tumors in childhood, about
5% (1.5–7.5%) of children with neurofibromatosis-1 (NF1). INVESTIGATIONS
• Age: 50% present before the age of 5 years, 90% before age CT brain scan
20. Chiasmal lesions are more frequent in adolescents than Coronal, sagittal and axial studies:
in children. • The optic nerve appears enlarged (fusiform or nodular) and
• Gender: F>M (2:1). tortuous.
• Calcification is rare.
PATHOLOGY • Enhancement is variable.
• The vast majority are pilocytic astrocytomas (histologically • There may be cystic components.
identical to pilocytic astrocytomas elsewhere in the CNS), • The optic canal may be enlarged (also visible on plain films
WHO grade 1 (459, 460); anaplastic astrocytoma occurs of the orbit) if the tumor extends that far back.
rarely. • The tumor may be bilateral and extend back into the chiasm.
• Two architectural forms:
– Diffuse expansion and obstruction of the optic nerve MRI scan
without extensive subarachnoid spread. • The appearance is similar to that on CT but the detail is
– Predominant infiltration of the subarachnoid space leaving more clearly seen.
the mildly involved nerve surrounded by a rim of tumor • The lesion appears isointense on T1WI (461) and iso- to
tissue. hyperintense on T2WI (462).
• Most are slowly growing and grow along optic pathways • It is impossible to separate the tumor from the nerve
(causing enlargement of the optic nerve [50%], optic chiasm radiologically (as opposed to meningiomas where it is often
[45%], or optic tract [5%]) and invade the subarachnoid possible to distinguish the tumor from the nerve).
space, causing hyperplasia of the overlying arachnoid mater, • MRI also defines the intracranial portion of the tumor better
known as arachnoid gliomatosis. Optic nerve gliomas may than CT.
also invade the anterior hypothalamus and cause obstruction • Magnetic resonance spectroscopy enables assessment of the
of the foramen of Monro and obstructive hydrocephalus. biochemical composition of focal areas within the tumor and
Tumor growth is generally limited by dura. other regions of the brain.

ETIOLOGY AND PATHOPHYSIOLOGY Visual evoked potentials


15–20% of optic gliomas occur in patients with neurofibro- Help establish and localize the lesion and establish the
matosis; 50–70% of optic gliomas in children are associated functional integrity of the visual system.
with neurofibromatosis; 15–25% (50–70% of children) are as-
sociated with neurofibromatosis (see p.147). Blood for DNA analysis
Mutation of the NF1 gene located on chromosome 17q11.2.
CLINICAL FEATURES
• Slowly progressive visual deterioration: dimness of vision Chiasmal biopsy
with decreased visual acuity and color vision; abnormal Contraindicated since it is the main cause of visual deterioration
pupillary function; constricted fields, followed by variable and is associated with significant but unexplained mortality.
visual field defects, depending on the location of the tumor,
that can be monocular, binocular, eccentric, centrocecal DIAGNOSIS
homonymous, heteronymous, and bitemporal, and The clinical diagnosis of an optic pathway tumor is confirmed
progressing to blindness; and optic atrophy with or without by CT or MRI scan, and the pathology by tissue biopsy.
papilledema. Papilledema with or without a field cut implies
involvement of the hypothalamus and obstructive TREATMENT
hydrocephalus whereas papillitis and central visual field loss • Depends on the nature of the symptoms, and the location
suggests an orbital lesion. and rate of growth of the tumor; there is no universal
• Ocular proptosis due to the orbital mass. approach to treatment
• Nystagmus, if chiasmatic involvement. • Children with symptomatic optic pathway glioma,
• Hypothalamic and pituitary dysfunction occur occasionally particularly if it involves the chiasm and either the optic
in gliomas involving the optic chiasm (e.g. accelerated linear nerves or the posterior regions of the visual pathway, should
growth due to precocious puberty; adiposity, polyuria, be closely followed, particularly for the development of
somnolence). precocious puberty (e.g. signs of premature secondary
• Secondary hydrocephalus. sexual characteristics, or accelerated linear growth rate,
detected by yearly measurements of height and weight
DIFFERENTIAL DIAGNOSIS plotted on standard pediatric growth charts) and evidence
• Optic neuritis (see p.489): sarcoidosis, multiple sclerosis. of reduced visual acuity, color vision or visual fields or
• Optic nerve hamartoma (NF1, see p.147). changes on slit lamp examination and fundoscopy.
Optic Nerve Glioma 373

• Treatment is indicated for progressive proptosis or loss of tumors may be quite extensive, large volumes of normal
vision, or neuroradiographic progression. young brain may be included in the radiation portal; there are
• Therapeutic options include surgery, radiation, chemo- significant potential neurocognitive and endocrinologic
therapy, or a combination of these modalities. sequelae of radiotherapy to consider before embarking on this
• A blind eye due to intraorbital glioma can be removed treatment, let alone the theoretical possibility of radiotherapy
surgically for cosmetic reasons or to prevent potentially inducing more malignant transformation of the tumor.
further extension of the tumor into the chiasm (which must • There are few data to support the efficacy of chemotherapy
be very rare). Surgery has a limited role in the treatment of in intraorbital glioma. The combination of carboplatinum
chiasmatic gliomas that may or may not also involve the and vincristine appears promising for extraorbital optic
optic nerve or the posterior portion of the visual pathway. pathway glioma but this therapy still remains experimental.
Otherwise, surgical excision is generally inappropriate
because the short term risks outweigh the longer term PROGNOSIS
benign course. • May vary from extreme indolence to rapid progression
• Radiotherapy should be reserved primarily for patients with but most are very slow-growing.
isolated intraorbital gliomas and residual useful vision, • Optic pathway gliomas in children with NF1 infrequently
progressive (either visual or radiographic) isolated optic nerve progress once the tumors have come to medical
gliomas (because tumor progression may be halted, at least attention: in one study, demonstrable tumor growth or
temporarily) or extraorbital (e.g. optic chiasm) optic pathway progression of visual disturbances occurred in only 3 of
glioma, but the long term efficacy for disease control is 26 children over a mean follow-up period of 4.2 years.
questionable. As many patients are children, and as these • 20-year survival rate: 40–50%.

459 460

459, 460 Histology of optic nerve glioma showing infiltration of the 462
optic nerve by glioblasts and astrocytes.

462 T2W axial MRI of the brain in NF1 showing areas of increased
signal in the basal ganglia bilaterally, worse on the right, also visible in
the periventricular white matter (arrows).The precise nature of these
is the subject of debate.

461 T1W MRI of the orbits showing an optic nerve glioma in NF1. Note
the thickened optic nerves bilaterally (arrows).

461
374 Tumors of the Central Nervous System

MENINGIOMA • Necrosis.
• Invasion of surrounding brain.

DEFINITION Histologic classification


A generally benign tumor, which originates from • Groups:
meningothelial cells of the dura mater or arachnoid. – Classic: meningothelial, syncytial, transitional, fibro-
blastic, psammomatous, angiomatous, microcystic,
EPIDEMIOLOGY secretory, clear cell, choroid, lymphoplasmacyte-rich
• Incidence: 7.8 per 100 000 per year (2 per 100 000 per and metaplastic variants.
year in symptomatic patients). Most are asymptomatic – Atypical.
tumors discovered incidentally at autopsy. 15–20% of all – Papillary.
primary intracranial tumors and the most common – Anaplastic.
benign brain tumor. • Grades:
• Age: 40–70 years old; highest incidence in seventh 1 Benign (the vast majority).
decade. 2 Atypical.
• Gender: F>M (2:1). 3 Anaplastic.
4 Sarcomatous.
PATHOLOGY
• Arise from the arachnoid cap cell in the arachnoid layer Cytogenic and molecular
of the meninges and almost always have a dural There are multiple deletions on chromosome 22 in nearly
attachment. all fibroblastic meningiomas and one-third of meningo-
• Rarely arise from arachnoidal cells within the choroid thelial meningiomas.
plexus, forming intraventricular meningiomas.
ETIOLOGY AND PATHOPHYSIOLOGY
Macroscopic • The cell of origin, the arachnoid cap cell, has a slow rate
• Firm, gray and sharply circumscribed; clearly demarcated of cell division.
from brain tissue (463). • Tumorigenesis is probably the result of exogenous or
• Takes the shape of the space in which it grows: may be endogenous factors acting alone or together.
flat and plaque-like, or round and lobulated.
• Sites of origin: Exogenous factors implicated in the development
– Olfactory groove: of meningiomas
Arise from arachnoidal cells along the cribriform plate in • Prior irradiation to the scalp or brain (definite):
the floor of the anterior cranial fossa. increased risk and earlier age of onset of meningioma.
May reach enormous size before coming to medical • Trauma (possible).
attention. • Viral infection (possible).
– Tuberculum sellae.
– Lesser wing of sphenoid bone (sphenoidal ridge): Endogenous factors implicated in the development
May grow medially and compress structures in the wall of meningiomas
of cavernous sinuses. • Hormones: progestins, androgens, glucocorticoids.
May grow anteriorly into the orbit. Meningiomas occur twice as often in women as in men,
May grow laterally into the temporal bone. they are more common in people with breast carcinoma
– Parasagittal surface of the frontal and parietal lobes (falx and they may enlarge during pregnancy. They express
meningioma). estrogen and progesterone receptors, and some may
– Sylvian region. express D1 dopamine receptors. Androgens may
– Superior surface of the cerebellum. regulate progesterone receptors in meningioma cells
– Cerebellopontine angle. and may induce synthesis of EGFR.
– Spinal canal. • Growth factors: platelet-derived growth factor and
• Extend to dural surface and often invade and erode epidermal growth factor.
cranial bones or excite an osteoblastic reaction; some give • Genetic:
rise to an exostosis on the external surface of the skull. – Loss of one chromosome 22q, which is also the
• Most are histologically benign, 5% are atypical and 2% molecular characteristic of NF2 (in nearly half of cases),
show frankly malignant, invasive qualities. or a partial deletion of chromosome 22q (in another
20% of cases).
Microscopic – Mutations in the NF2 gene in sporadic meningiomas.
Cells
• Uniform with round or elongated nuclei. CLINICAL FEATURES
• Visible cytoplasmic membrane. Small (<2.0 cm [0.8 in] diameter) surface
• Tend to encircle one another, forming whorls and meningiomas
psammoma bodies (464–466). Asymptomatic: do not compress neural tissue.

Histologic features suggesting malignant change


• Hypercellularity.
• Mitotic figures.
• Nuclear atypia.
Meningioma 375

Parasagittal fronto-parietal meningioma 463


• Partial seizures.
• Slowly progressive spastic paraparesis.
• Altered sensation of one leg and later both legs.
• ‘TIA-like’ presentations of sudden onset of leg ± arm
weakness or sensory change.

Olfactory groove meningioma


• Anosmia: ipsilateral (rarely reported by patient) or
bilateral, due to compression of the olfactory bulb.
• Blindness: ipsilateral or bilateral, often with ipsilateral
optic atrophy and contralateral papilledema (Foster
Kennedy syndrome).
• Frontal lobe syndrome later: abulia, confusion,
forgetfulness, inappropriate jocularity (Witzelsucht), due
to compression of the inferior frontal lobe.

Tuberculum sellae meningioma


• Visual failure: a slowly advancing bitemporal hemianopia
with a sella of normal size.
• Visual field defects are often asymmetric due to
involvement of the optic chiasm and optic nerve.
• No hypothalamic or pituitary deficits usually.

Sphenoidal wing meningioma (medial)


Loss of vision and optic atrophy ipsilaterally (optic nerve
compression at the optic foramen), sometimes with papilledema
in the contralateral eye (Foster Kennedy syndrome). 463 Axial section of brain showing a large, round, gray and vascular
left frontal meningioma (arrows). (Courtesy of Professor BA Kakulas,
Sphenoidal wing meningioma (lateral, near Royal Perth Hospital,Western Australia.)
superior orbital fissure)
• Exophthalmos ipsilaterally: slowly developing.
• Diplopia (compression of cranial nerves III, IV and VI).
• Upper facial sensory disturbance (compression of cranial
nerve V1).
• A family history may be present. 464

464–466 Histologic sections of a meningioma showing arachnoid cells


arranged in whorls and prominent vascularity. (Courtesy of Professor
BA Kakulas, Royal Perth Hospital,Western Australia.)

465 466
376 Tumors of the Central Nervous System

DIFFERENTIAL DIAGNOSIS TREATMENT


Tuberculum sellae meningioma Conservative
• Pituitary adenoma. Incidentally discovered meningiomas can be followed safely
• Aneurysm of the terminal carotid artery. with CT or MRI.

Sphenoid ridge meningioma Surgical excision


• Sarcoma arising from skull bone. • The mainstay of therapy.
• Orbitoethmoidal osteoma. • Effective for accessible surface tumors that can be totally
• Benign giant cell bone cyst. resected, such as meningiomas of the convexity dura, falx,
• Optic nerve tumors. lateral sphenoid wing, frontal base and posterior-fossa dura.
• Orbital angioma. A cuff of normal dura should be removed to reduce the
• Metastatic carcinoma. chance of marginal recurrence.
• Tumors that lie beneath the hypothalamus, along the
INVESTIGATIONS medial part of the sphenoid bone and parasellar region and
CT brain scan (467, 468) anterior to the brainstem are the most difficult to remove
• Meningiomas arise from any dural surface and push the surgically. They are inoperable if they invade adjacent bone.
brain away.
• They are usually rounded or lobulated, iso- or slightly hypo- Stereotaxic radiosurgery
dense lesions that have well-defined, sharply demarcated Appropriate for some well localized (i.e. not adjacent to the
contours and enhance strongly with i.v. contrast. optic nerve or other critical structures) and small (<3 cm
• There may be an enhancing dural ‘tail’ extending from the [<1.2 in] in diameter) meningiomas, particularly in the skull
tumor margin. base.
• White matter edema varies from none (usually) to extensive
(occasionally). Radiotherapy
• Thickening of the adjacent bone (hyperostosis) may be Fractionated, external beam, radiation therapy can delay
visible. regrowth or recurrence and is indicated if the tumor is
• Calcification is occasional though can be dense. inoperable, incompletely resected or histologically aggressive.
• Large dilated venous sinuses around the tumor may be
visible. Interstitial brachytherapy
• The mass effect is usually less than for a similar sized intrinsic • Used for recurrent benign and malignant meningiomas
tumor as the slow growth allows time for the normal brain that have failed to respond to standard therapies, are
to shift and adapt to the presence of the meningioma. amenable to repeat operation, and have a residual tumor
thickness that does not exceed a few millimetres.
MRI brain scan • Both stereotactic and open craniotomy techniques have
• Meningiomas are often isointense to gray matter on been used for placement of the radiation sources (e.g.
T1WI and T2WI, so if small may be overlooked. iodine-125).
• They enhance strongly with i.v. contrast (gadolinium).
• Their other features are as described for CT. Cytotoxic chemotherapy
Little success to date. Hydroxyurea appeared promising in
Cerebral angiography preliminary reports. But shown recently to be ineffective.
• There is usually enlargement of the meningeal arteries
(branches of the external carotid) which supply the Biologic therapies
meningioma. Experimental (e.g. hormone receptor antagonists).
• Some arterial supply may come from the internal carotid
arteries also. PROGNOSIS
• The venous drainage is usually to the intracranial dural • Slow rate of growth.
venous sinuses. • Not always curable; the rate of recurrence depends on
• Some centres offer particulate embolization of the site of the tumor, its biologic aggressiveness, and the
meningiomas prior to surgery to reduce the vascularity completeness of the resection.
and blood loss during tumor removal. • Complete excision is associated with permanent cure in
many cases; invasive growth leads to recurrence in about
CSF 2% of patients per year (i.e. 20% at 10 years).
Increased protein; may be very high (2.0–4.0 g/l • Subtotal resection and malignancy are associated with a
[200–400 mg/dl]) with olfactory groove meningioma. higher rate of recurrence: for benign meningiomas, the
5 year rate of survival free of progression is about 90%
DIAGNOSIS and at 10 years about 75%; whereas for malignant menin-
• Some meningiomas reach an enormous size before the giomas, 5 year survival free of progression is only about
diagnosis is established. 50%. More than 80% recur after partial resection.
• Diagnosis is suggested by contrast-enhanced CT and MRI, • Histology alone may be inadequate to characterize
and angiography, but is confirmed histologically. accurately the biologic behavior of meningiomas. Age is
an important independent prognostic factor for survival.
Craniopharyngioma 377

467 CRANIOPHARYNGIOMA

DEFINITION
A rare, congenital cystic tumor which arises from cell rests
(embryologic remnants of pharyngeal epithelium in
Rathke’s pouch) above the pituitary fossa and gives rise to
a suprasellar mass that compresses and infiltrates adjacent
local structures, causing endocrine or visual symptoms in
children and young adults.

EPIDEMIOLOGY
• Incidence: rare, 2–3% of all brain tumors.
• Age: children predominantly, but may present in adults
of all ages.
• Gender: M=F.

ETIOLOGY AND PATHOGENESIS


• Congenital.
• The tumor arises from cell rests (embryologic remnants
of Rathke’s pouch) above the pituitary fossa at the
junction of the infundibular stem and pituitary gland.

PATHOLOGY
• Usually the tumor lies above the sella turcica, depressing
the optic chiasm and extending up into the third
468 ventricle.
• Less often it is subdiaphragmatic, within the sella, where
it compresses the pituitary body and erodes one part of
the wall of the sella or a clinoid process, but seldom does
it balloon the sella like a pituitary adenoma.
• A cystic tumor, containing dark albuminous fluid,
cholesterol crystals, and calcium deposits; it is partly calci-
fied by the time it is 3–4 cm (1.2–1.5 in) in diameter.
• Infiltrates locally.
• The sella beneath the tumor becomes flattened and
enlarged.

CLINICAL FEATURES
Presents in children and young adults with any one or a
combination of the following syndromes:
• Hypothalamic dysfunction: hypopituitarism (growth
failure in children: delayed physical and mental
development, diabetes insipidus, adiposity, amenorrhea,
disturbed temperature regulation, sexual dysfunction).
• Progressive or intermittent optic nerve or chiasm
compression (visual loss).
• Obstructive hydrocephalus (raised intracranial pressure:
headaches, vomiting).
• Frontal lobe symptoms (mental dullness, confusion,
spastic leg weakness).
467, 468 CT brain scan pre- (467) and post (468) i.v. contrast. Pre-
contrast the meningioma appears similar to adjacent brain and is
causing relatively little displacement of the normal brain for its size. Post
i.v. contrast there is intense enhancement (whiteness) making the
tumor very obvious. Note the rounded shape, the flat surface against
the inner skull table where it arises from the dura, the small granular
calcifications (white spots), and the relative lack of white matter edema
and mass effect. Also see 715 and 716 for spinal meningioma.
378 Tumors of the Central Nervous System

INVESTIGATIONS DIFFERENTIAL DIAGNOSIS


Imaging • Optic nerve/chiasm tumor (see p.372).
• Skull x-ray and CT brain scan may reveal calcification in • Meningioma (see p.374)): medial sphenoid, olfactory
the region of the pituitary gland. groove.
• CT and MRI scan reveal a mass immediately superior to • Pituitary adenoma (see p.379).
or arising out of the pituitary fossa (469, 470). The mass • Chondroma.
may be at least 2 cm (0.8 in) in diameter. It may • Hypothalamic glioma.
compress the optic chiasm and indent the third ventricle. • Supraclinoid carotid artery aneurysm.
Large tumors may obstruct the foramen of Monro. • Choroid plexus papilloma.
Typical features include calcification, solid and cystic • Colloid cyst of the third ventricle.
elements, and enhancement with contrast, usually in a
young person (471, 472). The calcification and cystic
elements are better appreciated on CT but the tumor
definition is generally better seen on MRI (with
contrast). MRI T1W images may show increased signal
in the mass lesion due to the cholesterol content.

469 470

469, 470 Sagittal (469) and coronal (470) T1W MRI of the pituitary fossa after contrast injection showing an ovoid, 10 × 12 mm (0.4 × 0.5 in)
mass of homogeneous brain signal intensity (arrows) in the suprasellar cistern applied to the inferior surface of the optic chiasm, more on the right
than the left, and anterior to the infundibulum of the pituitary gland.The optic chiasm is mildly elevated on the right side.The pituitary gland
appears normal.The mass enhances mildly with contrast and is a craniopharyngioma.

471 472

471 T1W MRI (sagittal plane) of a craniopharyngioma (arrows) in a 472 T1W coronal MRI with contrast (same patient) shows the lesion
20 year old male. Note the mixed signal elements with cysts and enhances markedly and irregularly with a prominent cyst extending up
extension to the foramen of Monro. into the third ventricle. On MRI the calcification often associated with
these tumors is difficult to appreciate and CT may help.
Pituitary Tumors 379

TREATMENT PITUITARY TUMORS


Treatment is controversial, because the risks of complete
surgical resection (memory deficits, appetite and behavioral
disturbances and endocrine dysfunction) have to be weighed DEFINITION
against the potential growth of any remaining remnants. Pituitary tumors are mostly benign epithelial neoplasms that
result from mutation and subsequent clonal expansion of
Surgery single adenohypophyseal parenchymal cells.
• Surgical excision of all or part of the tumor (difficult to
remove completely). EPIDEMIOLOGY
• Corticosteroid therapy before and after surgery. • Incidence: 10–15% of intracranial neoplasms; occult
• Careful control of fluid and temperature balance post pituitary adenomas are found in up to 20% of random
operatively. post mortem examinations.
• Age: adults, mean age 60 years.
Radiation therapy • Gender: M≥F.
External-beam radiation if any residual tumor.
PATHOLOGY
Palliation • Adenoma (473–475): benign slowly growing tumors.
• Stereotactic aspiration. However, prolactinomas may rapidly expand during
• Radiation therapy. pregnancy and cause subacute onset of neurologic
• Ventricular shunting. dysfunction.
• Three-quarters secrete inappropriate amounts of pituitary
PROGNOSIS hormones and one-quarter are ‘non-functioning’.
Mortality: • Tumors are classified histologically on the basis of
• 5–10% when complete surgical resection is attempted immunoperoxidase stains to identify specific hormones.
(i.e. smaller tumors). The most common hypersecretory tumors of the
• Higher mortality when only partial removal is possible pituitary are prolactin secreting, followed by adenomas
(i.e. larger and more infiltrative). that secrete growth hormone, ACTH, and rarely
• 10-year survival was 76% for patients undergoing surgical gonadotropin (10%) and TSH (<10%).
resection and radiation and 17% for patients undergoing • Gonadotrophic pituitary adenomas are inefficient
resection alone in one uncontrolled series of 74 cases. producers and secretors of the gonadotroph hormones:
LH, FSH, and the α-subunit of the pituitary
glycoprotein hormones. Even when gonadotrophin
hypersecretion occurs, no distinctive clinical endocrine
473 phenotype is present.
• Non-secreting adenomas, which comprise about 20–25%
of all pituitary tumors, are usually chromophobe
adenomas.

473–475 Histologic sections of a pituitary tumor at low, medium and


high power showing pleomorphic cells.

474 475
380 Tumors of the Central Nervous System

ETIOLOGY AND PATHOPHYSIOLOGY Local pressure on the optic chiasm, optic nerves, or
Unknown. rarely optic tract
Visual field defects in one or both eyes:
CLINICAL FEATURES • Lateral visual field defects (classically bitemporal hemianopia).
Most patients present with symptoms and signs of: • Scotomas.
• Complete blindness.
Hypersecretion of one or occasionally several
pituitary hormones Visual field testing by confrontation is insensitive and should
Hyperprolactinemia (prolactinoma) be followed by formal analysis. Goldmann perimetry is highly
• Women (pre-menopausal): dependent on the operator whereas computerized field testing
– Infertility. with a field analyser (Allergan Humphrey) gives highly
– Amenorrhea, oligomenorrhea, or normal periods. reproducible results and takes about 15 minutes for each eye.
– Galactorrhea.
– Decreased libido. Local pressure on the cavernous sinus
– Vaginal dryness and dyspareunia. Cranial nerve palsies: III, IV, V, VI.
– Delayed menarche.
• Men: Local pressure on the temporal lobes
– Decreased libido. Temporal lobe epilepsy.
– Impotence sometimes.
– Galactorrhea unusually. Local pressure on the third ventricle
– Reduced growth of facial and body hair. Obstructive hydrocephalus.
– Small, soft testes.
– Apathy. Other presentations
– Weight gain. • Headaches: non-specific, inconstant, seldom severe,
relieved by analgesics frequently and almost immediately
Growth hormone excess (growth hormone-producing by somatostatin in some patients.
adenoma) • Asymptomatic pituitary mass detected by CT or MRI.
• Gigantism in children. • Pre-operative diabetes insipidus is extremely rare in
• Acromegaly in adults. primary pituitary disease and suggests involvement of the
hypothalamus or pituitary infarction.
ACTH (corticotropin) excess
• Rare. DIFFERENTIAL DIAGNOSIS
• Cushing’s disease (476). Suprasellar lesions
Craniopharyngiomas, optic gliomas, chondromas, hypothala-
Hyposecretion of pituitary hormones due mic gliomas, supraclinoid carotid artery aneurysms, choroid
to pituitary gland infarction, hemorrhage plexus papillomas, and colloid cysts of the third ventricle.
or local pressure
Hypopituitarism Intrasellar lesions
• Hypogonadism: Pituitary adenomas, arachnoid cysts, and rare tumors of the
– Women: neurohypophysis.
Oligomenorrhea or amenorrhea.
Reduced libido. Perisellar lesions
Dyspareunia. Meningiomas, metastases, dermoids, teratomas, arachnoid
– Men: cysts, and cholesteatomas may occur in any of several
Reduced libido and potency. locations around the sella.
Reduced facial and body hair.
Gonadal atrophy. Parasellar lesions
• Hypoprolactinemia: Cavernous carotid aneurysms, cavernous sinus thrombosis,
– Failure to start and maintain lactation in women. temporal lobe neoplasms, and gasserion ganglion neuromas.
– No defined clinical entity in men.
• Hypothyroidism: mild with inappropriately low TSH in Retrosellar lesions
an otherwise well patient. Chordoma, basilar artery aneurysm.
• Growth hormone deficiency:
– Poorly defined clinical entity in adults. Infrasellar lesions
– Reduced body muscle : fat ratio. Sphenoid sinus mucocele, carcinoma, granuloma, and other
• Hypoadrenalism: nasopharyngeal tumors.
– Tiredness and malaise, especially in the afternoon and
evening. Empty sella syndrome
– Postural hypotension, pallor, anorexia and nausea. The major cause of asymptomatic sellar enlargement:
– No subtle electrolyte changes as the adrenal zona • The suprasellar subarachnoid space (arachnoid mater and
glomerulosa is intact. CSF) extends/herniates into the sellar cavity through an
– Loss of secondary sexual hair in women. incompetent diaphragm, thus compressing and flattening
the pituitary gland inferiorly and posteriorly, stretching
the stalk, and eventually causing hormone disregulation.
Pituitary Tumors 381

• Primary empty sella: arises in the absence of any previous Hyperprolactinemia


known pituitary disease. It is most commonly seen in Hypothalamic and infundibular lesions (loss of inhibitory
middle-aged obese women and may be associated with control of prolactin secretion), renal failure, and drugs
raised intracranial pressure. (phenothaizines, butyrophenones, benzodiazepines,
• Secondary empty sella: may follow idiopathic intracranial reserpine, morphine, alpha methyldopa and isoniazid).
hypertension, spontaneous regressive changes of pituitary
adenomas, and surgery (i.e. the meninges and CSF INVESTIGATIONS
herniate downwards to fill the void created by pituitary Pituitary hormones
involution as a result of other pathology such as pituitary • Hormones are stable for several days in plasma at room
surgery, irradiation, hemorrhage or tumor necrosis). temperature except for ACTH, which must be collected
• Usually asymptomatic. on ice and separated as soon as possible.
• Symptoms may include headache, occasional mild • Serum prolactin levels >100 μg/l (>100 ng/ml) (normal
endocrine abnormalities, CSF rhinorrhea, and, rarely, <15 μg/l [<15 ng/ml]) are almost always due to tumor.
visual disturbances. These are more commonly symptoms Levels from 15–100 μg/l (15–100 ng/ml) may be due
of the cause than the empty sella itself. Hypopituitarism, to tumor but are more commonly due to other
diabetes insipidus, and visual field defects are much less disorders, particularly drugs. Distortion or damage of the
common with the empty sella syndrome than pituitary hypothalamus or pituitary stalk by a functionless pituitary
lesions. adenoma may result in modest hyperprolactinemia.
• These patients are generally investigated for suspected • Assay of ACTH from the right and left petrosal sinuses,
pituitary adenoma because of sellar enlargement and comparing the values with those of the superior vena
discovered on plain x-rays. A ballooned sella without cava can establish the pituitary as the source of excess
erosion is characteristic of the empty sella. Empty sella is ACTH and often localizes the side of the adenoma as
now diagnosed more easily with high resolution CT and well.
MRI.
• The evolution appears to be wholly independent of the CT and MRI brain scan
sellar dimensions and of the grade of the disease at • CT clearly shows calcified lesions such as
diagnosis. craniopharyngiomas.
• MRI shows surrounding soft tissue structures (e.g.
Pituitary apoplexy (see p.268) pituitary stalk, optic chiasm) and can be performed
Chiasmal hemorrhage, subarachnoid hemorrhage, repeatedly without risk of excessive exposure to x-rays.
meningitis. • MRI is more sensitive than CT for detecting
microadenomas (477).

476 477

477 MRI brain scan (coronal T1W image with i.v. contrast) showing a
microadenoma on the left side of the gland (arrows).

476 Abdominal and axillary cutaneous striae in a patient with


Cushing’s disease due to an ACTH-producing pituitary microadenoma.
382 Tumors of the Central Nervous System

Microadenomas • The differential diagnosis on imaging includes


• On CT (thin section coronal with i.v. contrast) meningiomas, craniopharyngiomas, large aneurysms,
microadenomas appear as low density areas within the metastases, granulomas.
gland. Other signs include deviation of the pituitary stalk • Angiography may be necessary prior to surgery and to
away from the side of the adenoma, focal erosion of the exclude an aneurysm; MR is not reliable enough to
floor of the fossa and upward bowing of the superior exclude an aneurysm as slow blood flow within it may
margin of the gland. mimic a solid lesion.
• On MRI (thin section coronal with i.v. contrast)
microadenomas are hypointense relative to the normal Carotid angiography
gland on T1WI and of variable intensity on T2WI. Often a wise prelude to surgery to exclude an aneurysm of
Dynamic scanning has been advocated for elusive the carotid siphon that may cause identical clinical and CT
adenomas. features to those of an adenoma.

Macroadenomas DIAGNOSIS
• On CT or MRI these appear as masses arising out of the Patients present with a typical history and examination and the
pituitary fossa which may elevate and compress the optic diagnosis is confirmed by CT or MRI scan of the pituitary, and
chiasm and anterior cerebral arteries (478–481), and blood tests.
invade the cavernous sinus and encase the internal carotid
arteries laterally. Enhancement is usual. There may be
cystic elements.

478 479

480 481

478–481 Axial proton density MRI (478), axial T2W MRI (479), coronal T1W MRI pre-contrast (480), and coronal T1W post contrast MRI (481)
images showing a pituitary adenoma in the sella extending superiorly to compress the inferior aspect of the optic chiasm and elevate the A1
segment of the anterior cerebral artery, and inferiorly to depress the floor of the sella (arrows).There is no extension into the cavernous sinuses.
Acoustic Neuroma 383

TREATMENT ACOUSTIC NEUROMA


• If panhypopituitarism is suspected, glucocorticoids should
be replaced before thyroid hormones.
• Trans-sphenoidal microsurgical resection is usually the first DEFINITION
line treatment for adenomas that hypersecrete or cause mass A histologically benign tumor of the vestibulocochlear nerve
effect. There should be no mortality, and any morbidity sheath, arising most frequently from the superior portion of
should be minor (about 10% of cases). The remission rate the vestibular nerve
after 5 years is about 80%. Extension into brain requires an
intracranial approach. Pre- and post operative endocrine EPIDEMIOLOGY
and neurologic evaluation is essential. Corticosteroid • Incidence: 1 per 100 000 per year; 0.8– 2.7% of autopsy
coverage should be provided during surgery. series.
• Prolactinomas are the major exception because they are • Age: adults.
usually treated with the dopamine agonist bromocriptine • Sex: M=F.
or cabergoline, which shrinks the tumor and decreases
prolactin secretion. Adverse effects include nausea and ANATOMY
hypotension (and cost). Surgery is an effective alternative, Cerebellopontine angle
particularly if pregnancy is desired or there is rapid visual • Apex: trigeminal nerve (V).
loss, with a 70–80% remission rate after 1 year. Post • Mid zone:
operative bromocriptine or cabergoline help control – Facial nerve (VII).
residual or recurrent tumor. Failing that, radiotherapy can – Vestibulocochlear nerve (VIII).
be considered. – Anterior inferior cerebellar artery.
• Growth-hormone-producing adenomas may be treated • Base:
with the somatostatin analogue octreotide (Sandostatin). – Glossopharyngeal nerve (IX).
• Standard fractionated supervoltage radiotherapy with – Vagus nerve (X).
20–25 treatments over 4–6 weeks may halve the low, long – Accessory nerve (XI).
term rate of tumor recurrence (from about 15% to 7%), but
causes adverse effects such as low grade neuronal damage, PATHOLOGY
radionecrosis, vaso-occlusive disease, and secondary tumors Site
of the CNS such as anaplastic meningiomas, and so it is less A tumor of Schwann cells (and not neurons), and most
often given as an adjunct to surgery. Furthermore, ready frequently occurs on the superior portion of the vestibular
access to MRI for regular post operative scans allows tumor nerve. Hence, it is doubly misnamed and should really be
recurrence to be diagnosed early so that further surgery can called a vestibular schwannoma.
be contemplated.
Growth
PROGNOSIS From its origin on the vestibular nerve, it grows gradually
Although residual cells in parasellar structures may account to fill the internal auditory meatus, compressing the
for local recurrences that follow seemingly complete surgical cochlear and facial nerves and ultimately compressing the
clearances, metastatic spread and direct macroscopic cerebellar peduncles, cerebellum, fourth ventricle, and
invasion of surrounding structures is rare. cranial nerves IX, X and XI (482).

ETIOLOGY
• Uncertain (sporadic) in most cases.
482 • Inherited as part of neurofibromatosis 2 in some cases (see
p.147): a familial syndrome, usually inherited as an
autosomal dominant trait due to a mutation on chromo-
some 22q1, and consisting of multiple cranial nerve
tumors, along with neuromas of the spinal nerve roots.

CLINICAL FEATURES
Depend on the site of origin of the tumor along the
acoustico-vestibular nerve bundle:

Internal auditory meatus origin


Otolaryngologic symptoms and signs
• Altered hearing (distorted, muffled hearing):
– Reduced hearing in whisper tests.
– Sensorineural hearing loss: Weber test lateralized to ‘good ear’.
– Tinnitus.
• Pressure in the ear.

482 Axial section of the pons and cerebellum showing a large


acoustic neuroma in the cerebellopontine angle (arrows). (Courtesy
of Professor BA Kakulas, Royal Perth Hospital,Western Australia.)
384 Tumors of the Central Nervous System

Cerebellopontine angle origin • They are rounded, may be very large and cause brainstem
Compression of nearby structures displacement.
• Atypical trigeminal neuralgia (V). • The main differential diagnosis is meningioma, which does
• Tic douloureux (V). not usually extend into the acoustic canal.
• Loss or reduction of corneal reflex.
• Facial numbness (V). Pure tone audiometry
• Hemifacial spasm. Sensorineural hearing loss.
• Progressive painless lower motor neuron facial weakness
(VII). Brainstem auditory evoked potentials
• Hoarse weak voice (X). Not usually necessary, but if so may reveal prolongation of
• Dysphagia (IX, X). waves I–III.

Compression of brainstem and raised intracranial DIAGNOSIS


pressure (hydrocephalus) The definitive investigation is gadolinium-enhanced MRI in a
• Headache. patient with a suggestive clinical history.
• Unsteadiness, clumsiness, poor balance.
• Vertigo. TREATMENT
• Spontaneous nystagmus*. • Treatment depends on a complex interplay of the needs
• Vomiting. and expectations of the patient, the size of the tumor and
• Hearing loss and/or tinnitus in the other ear. the local expertise.
• Altered mental state. • Treatments include expectant care with repeat scanning to
• Visual changes. assess tumor growth in elderly people and surgery with or
without radiotherapy.
*Nystagmus: • Surgical approaches and techniques vary but generally
Peripheral nystagmus Central nystagmus involve micro-surgical techniques, an ultrasonic surgical
Direction Fast phase in same direction, Usually changes aspirator, intraoperative monitoring of brainstem auditory
irrespective of direction direction evoked potentials and facial nerve function (by
of gaze electromyography), and access to intensive care facilities for
Horizontal or rotary/torsional May be bizarre immediate post operative care.
Never vertical Often vertical
Prognosis Decreasing severity Does not improve Mortality varies from 2–4% depending on the size of the
over 2 weeks significantly tumor. Other risks include deafness, facial palsy, vertigo, lower
cranial nerve palsy, brainstem damage, brainstem and cerebellar
DIFFERENTIAL DIAGNOSIS infarction.
• Trigeminal neuralgia (that fails to respond or responds The risks increase with the size of the tumor and decrease
erratically to standard treatment with carbamazepine). with the experience of the operating team. The chances of
• Eustachian tube obstruction. surgery preserving useful hearing are slight unless the tumor
• Cerebellopontine angle tumor: is small or medium sized; surgical resection of tumors <2.5 cm
– Acoustic neuroma: (75% of cases). (<1 in) in diameter carries a one in four to one in three chance
– Meningioma: (6% of cases). of preserving hearing. Surgical resection of an acoustic
– Cholesteatoma: (6% of cases): neuroma measuring <2 cm (<0.8 in) in diameter should
Epidermoid cyst. however provide an 80% chance of immediate preservation of
Arising from temporal bone. facial nerve function and a 95% chance of long term facial
– Glioma: (3% of cases). nerve function.
– Other types: (10% of cases):
Metastatic tumors. • Stereotactic radiotherapy can deliver highly localized levels
Osteomas. of radiation with minimal damage to surrounding
Osteogenic sarcomas. structures. Because of the morbidity of surgery, especially
Neuromas of trigeminal, facial or glossopharyngeal nerve. to the facial nerve and hearing, there is a move toward
Angiomas. stereotactic radiotherapy for tumors with a maximum
Papillomas of choroid plexus. diameter of <3 cm (<1.2 in), particularly in the elderly and
Teratomas. for those with bilateral acoustic neuromas.
Lipomas. • Combined surgery and radiotherapy. Surgery to debulk a
large tumor has a lower operative morbidity compared with
INVESTIGATIONS attempted total removal. Post operative stereotactic
CT or MRI brain radiotherapy in such cases may be a useful combination but
• MRI is the method of choice, though thin section CT with the long term effects remain to be evaluated.
i.v. contrast or air meatography are alternatives in patients
who cannot have or tolerate MRI. PROGNOSIS
• The standard MRI sequence is a T1W thin section Acoustic neuromas grow but the rate of growth seems to be
sequence through the petrous bones with i.v. contrast. slower in older people and they may not cause serious
• Acoustic neuromas show up as an enhancing mass, within symptoms; 0.8–2.7% of post mortem studies find an acoustic
the VIIIth nerve canal if very small (483), and extending neuroma that had not contributed to the death of the person.
into the cerebellopontine angle if larger (484).
Primary CNS Lymphoma 385

PRIMARY CNS LYMPHOMA PATHOLOGY


Macroscopic
• Multifocal mass deposits in the brain in two-thirds of cases.
DEFINITION • Solitary mass within the subcortical white matter in one-
A tumor of lymphocytes or lymphoblasts, usually B cells, third.
confined to the CNS. • Sites:
– Brain:
EPIDEMIOLOGY Supratentorial (75% of cases with brain lymphoma):
• 1–2% of cases of non-Hodgkin’s lymphoma. Periventricular lesions are frequent.
• 3% of all intracranial neoplasms. Corpus callosum (41%).
• Marked increase in incidence in the last decade as part of Deep central nuclei (33%).
the AIDS epidemic but also within the immuno- Infratentorial (20% of cases with brain lymphoma).
competent population (for unknown reasons). Diffuse leptomeningeal (5% of cases with brain lymphoma).
• Age: median: 57 years (non-AIDS); young adults (AIDS). Diffuse neuropil involvement: rare.
• Gender: M>F (1.5:1). – Eye: posterior segment (retina and vitreous): present in
25% of cases with intracranial lymphoma.
• At autopsy, parenchymal lesions are always in contact
483 with either the leptomeninges or the ependymal surface.

Microscopic
• The tumor cell of origin is the lymphocyte or lymphoblast
(B cell, usually diffuse, large cell subtype: >2/3, T cell:
<1/3; this ratio is different to systemic lymphoma).
• The fine reticulum and microgliacytes are interstitial and
derived from fibroblasts and microglia (histiocytes).
• The tumor is highly cellular, with little tendency to necrosis.
Mitotic figures are numerous. The nuclei are oval or bean-
shaped with scant cytoplasm. The stainability of reticulum
and microglial cells, the latter by silver carbonate, serves to
distinguish this tumor microscopically. B cell markers
identify the tumor cell type.
• Most tumors are high-grade immunoblastic or diffuse
large cell type but many are arrested at relatively mature
stages of differentiation.
• Although B cell immunophenotype predominates in
immunocompromised (e.g. AIDS) and immuno-
competent (e.g. non-AIDS) cases, the tumor cells that
483 MRI brain,T1W image with i.v. contrast showing a small left arise in immunocompromised patients are usually
intracanalicular acoustic neuroma (arrow). polyclonal, tending to have high-grade histologic
characteristics (immunoblastic and small, non-cleaved
cells) and contain Epstein–Barr virus (EBV) genomic
484 material in about 80% of cases; whereas immuno-
competent patients usually have monoclonal, large-cell
lymphomas of B cell origin with a low-grade histologic
appearance and no evidence of EBV infection.
• The lymphocytes express either kappa or lambda light-
chain immunoglobulin. Molecular studies have also
demonstrated consistent profiles of light-chain and
heavy-chain immunoglobulin gene rearrangements in
primary, recurrent, and metastatic CNS lymphomas.
• Regardless of cell type, a perivascular pattern pre-
dominates, with malignant cells surrounding blood
vessels in concentric layers (resembling the inflammatory
response of encephalitis).

N.B. Systemic non-Hodgkin’s lymphoma, arising in lymph


nodes and other somatic organs, spreads to the CNS in about
25–30% of cases, and tends to infiltrate the leptomeninges
(sparing the parenchyma), particularly in the spinal region
(and causing spinal cord compression), in contrast to PCNS
lymphoma which tends to originate in the parenchyma.

484 MRI brain,T1W image of a large acoustic neuroma projecting


into the cerebellopontine angle (arrow).
386 Tumors of the Central Nervous System

ETIOLOGY Secondary lymphoma tends to involve the leptomeninges


Predisposing factors and CSF and may be very difficult to see with any imaging
Immunosuppression (inherited or acquired) modality. The only clue may be hydrocephalus.
• Congenital immunodeficiencies.
• Immunoglobulin A deficiency. CSF
• Combined immunodeficiency syndrome. Almost always abnormal:
• Immunodeficiency associated with systemic lupus • Protein: typically >1.0 g/l (>100 mg/dl) but may be normal.
erythematosus (SLE) and rheumatoid arthritis (RA) and • Cells: typically a slight lymphocytic pleocytosis
organ-transplant recipients. (<100 lymphocytes/mm3) but sometimes as few as only
• Long term steroid treatment for SLE, sarcoidosis, RA, 2 cells/mm3.
and other autoimmune or inflammatory illnesses. • Cytology: identifies malignant tumor cells in only
• AIDS due to HIV infection (see p.301). 25–30% of cases (the parenchyma tends to be involved).
The diagnostic yield may be increased by: multiple
Infection with EBV CSF samples (cells may initially be misdiagnosed as
May have a role in pathogenesis. reactive atypical cells); immunocytochemical staining,
demonstrating a predominance of expression of kappa or
PATHOGENESIS lambda light-chain immunoglobulin in the atypical
• HIV or other viruses (e.g. EBV), growth factors, lymphoid cells; polymerase chain reaction.
aberrant oncogene or tumor-suppressor gene expression, • β2-microglobulin concentration (>160 nmol/l) in CSF
and factors that induce genetic instability or DNA is another potential adjunctive marker of leptomeningeal
damage or alter host or viral genome repair may lead to spread of CNS lymphoma.
cell hyperproliferation and clonal expansion.
• B cell hyperactivation is thought to contribute to the Tissue biopsy
development of lymphoma associated with HIV Stereotactic CT-guided brain biopsy
infection. Diagnostic features are present in about one-third to one-half
and non-specific abnormalities in about one-half of specimens.
CLINICAL FEATURES Pre-treatment with corticosteroids may foil diagnostic
• Gradual onset and evolution over several months. attempts because they have a ‘lymphopenic’ effect on
• Expanding intracranial mass: primary CNS lymphomas (i.e. deplete biopsy specimens of
– Personality/behavioral/cognitive change or progressive abnormal lymphomatous cells) and result in temporary
psychomotor retardation (24%). regression of the tumor mass and contrast enhancement on
– Cerebellar signs (21%). CT scan.
– Headache (15%). Interpretation of biopsy and CSF cytologic specimens
– Motor dysfunction (11%). should include evaluation of immunohistochemical markers
– Visual changes (8%): an ophthalmologic examination is to demonstrate the largely monomorphous and probably
mandatory, seeking lymphomatous involvement in the clonal nature of the neoplastic lymphomononuclear cell
posterior segment of the eye. infiltrates.
• Seizures (13%).
• Multiple cranial and peripheral nerve root palsies: symptoms DIAGNOSIS
and signs suggesting leptomeningeal involvement are Pathologic – stereotactic biopsy.
present in only about one-quarter of patients with lepto-
meningeal lymphoma; in most patients it is clinically silent. TREATMENT
• During the subsequent clinical course, hemiparesis occurs The treatment for primary CNS lymphoma depends on
in about half of patients, headache in about a third, whether it arises in a normal or immunocompromised host.
aphasia in a quarter and ataxia in a quarter. However, unlike other brain tumors, resection does not
have a therapeutic role. Chemotherapy is the first-line of
INVESTIGATIONS treatment (e.g. high dose methotrexate ± cranial
CT/MRI brain scan irradiation), and is associated with complete response rates
Lesions are isotense or hyperintense on CT and are equally of 50–80% in immunocompetent patients. Chemotherapy
visible on CT or MRI, though MRI may define the precise combined with radiotherapy is also very effective (median
extent and relationship to adjacent tissues more clearly survival >40 months) in immunocompetent patients, but
(485, 486). Lesions may be solitary (1/3) or multiple results in substantial delayed neurotoxic effects, particularly
(2/3) and tend to be located in the basal ganglia and other in patients older than 60 years of age.
deep gray matter nuclei, periventricular white matter and
less commonly adjacent to the meninges and in the Immunocompetent patients
posterior fossa. They usually show: Radiation therapy alone
• Enhancement with contrast (487). • Radiotherapy to total doses of 50–60 Gy (5000–6000 rads)
• Mass effect. produces high control rates in systemic non-Hodgkin’s
• Rapid shrinkage with steroids. lymphoma but, paradoxically, the same doses in primary
CNS lymphoma are associated with a 90% risk of local
The differential diagnosis includes other malignant relapse and, accordingly, poor long term survival.
tumors, and in immunosuppressed patients, toxoplasmosis, • The mean survival is about 1.5 years (median
progressive multifocal leukocencephalopathy (PML) and 13 months) in patients treated with radiotherapy alone.
other opportunistic infections.
Primary CNS Lymphoma 387

Combined chemotherapy and radiotherapy • Early experience with high-dose methotrexate-based


• Standard chemotherapy protocols for non-Hodgkin’s regimens with leukovorin rescue may produce a similar
lymphoma (e.g. CHOP [cyclophosphamide, doxorubicin, disease-free interval with minimal toxicity. This treatment
vincristine and prednisone-based therapy]) are not of achieves uniform CNS and CSF levels throughout the
benefit, probably because of poor penetration of the CNS neuroaxis and when given without radiation is only rarely
by the drugs. The blood–brain barrier is already disrupted associated with methotrexate leukoencephalopathy.
when primary CNS lymphoma is present but it repairs • Methotrexate therapy can be complicated by nephro-
rapidly as soon as the tumor responds to treatment. genic diabetes insipidus. To prevent crystallization of
• High-dose intravenous (intrathecal or intraventricular if methotrexate in the renal tubules and to promote its
positive CSF cytology) methotrexate, 1 g/m2 on days 1 renal excretion, aggressive hydration with intravenous
and 8, followed by radiotherapy, commencing on day 15, fluids and sodium bicarbonate and alkalinization of the
with a total dose of 45 Gy (4500 rads) in 25 fractions urine are mandatory. The sodium load, however, can
delivered to the whole brain over the next 38 days, and a induce polyuria and hyperosmolarity. Desmopressin
boost of 5.4 Gy (540 rads) in three fractions to the acetate can reduce urine flow and methotrexate
original disease site, improves 2-year survival to 70% and excretion but it increases the likelihood of toxicity.
overall mean survival to about 3.5 years. If CSF cytology
results are positive, cytosine arabinoside (cytarabine)
(60 mg) or methotrexate may be given via an intracerebral 485
ventricular reservoir. Intrathecal chemotherapy is restricted
to patients with positive CSF cytology because syn-
chronous intrathecal chemotherapy and cranial irradiation
are associated with an increased risk of neurotoxicity. To
minimize further late neurotoxicity no more chemo-
therapy is given after radiotherapy. Morbidity from radia-
tion encephalopathy can also occur in long term survivors.
• Other agents, such as high-dose cytosine arabinoside
(cytarabine) and cyclophosphamide, have also been used
alone or in combination.

485 T2W axial MRI brain scan of a patient with AIDS and a
lymphoma in the posterior corpus callosum (arrows). (Courtesy of
Professor J Best, Department of Medical Radiology, Royal Infirmary,
Edinburgh, UK.)

486 T2W axial MRI


of lymphoma in the 486 487
midbrain in a patient
with AIDS (arrows).
Note the increased
signal around the
aqueduct in the
posterior midbrain
(proven at autopsy).
(Courtesy of
Professor J Best,
Department of
Medical Radiology,
Royal Infirmary,
Edinburgh, UK.)

487 CT scan with


contrast shows
enhancement of the
white matter in a
‘rind’ around the
lateral ventricles
(arrows).This
appearance is typical
for lymphoma.
388 Tumors of the Central Nervous System

Surgery GERM CELL TUMORS OF THE CNS


Ineffective except in rare cases, because the tumors are
usually deep and multicentric.
DEFINITION
Immunocompromised patients Tumors of germ cell origin, which include the germinoma
• Treatment aims to reverse the immunosuppression, (the most common), teratoma, embryonal carcinoma, yolk-
which may lead to tumor regression. sac tumor, choriocarcinoma, and mixed types.
• In patients with AIDS-associated primary CNS
lymphoma, therapy is not possible and the prognosis is EPIDEMIOLOGY
very poor, with an average survival of only 2.5 months, • Incidence: uncommon.
despite the use of corticosteroids followed by cranial • Age: predominantly school-aged children, adolescents
irradiation. and young adults (5–30 years).
• Gender: M>F.
PROGNOSIS
The natural history differs from other non-Hodgkin’s PATHOLOGY
lymphomas in that dissemination outside the neuroaxis is • Analogous to germ cell tumors arising in the gonads.
uncommon; 90% of patients die of progressive local disease. • Tumors probably arise from totipotential germ cells,
which are capable of producing several different cell
Survival subtypes. Consequently, they are composed of more than
Radiation therapy alone one tumor type.
• Median survival: 13 (12–18) months if immuno-
competent, 3 months if AIDS-related primary CNS Sites
lymphoma. • Midline: neurohypophyseal and pineal regions: the most
• Only 5–10% enjoy long term survival, despite increased common site is the pineal region, followed by the base
doses of radiotherapy. of the brain in the vicinity of the pituitary gland (e.g.
• 10% of long term survivors suffer neurologic third ventricle, hypothalamus).
deterioration in the absence of recurrence, suggesting a • ‘Multifocal’ germinoma: a localized, directly infiltrating,
possible long term effect of radiotherapy. disease characterized by multiple lesions involving the
pineal region, hypophysis, cavernous sinus, optic chiasm,
Combined chemotherapy and radiotherapy third ventricle wall, or anterior half of the lateral ventricle.
• Median survival after methotrexate and radiation therapy: • Disseminated germinoma: a metastatic disease.
40 months.
• Survival beyond 2 years: 70%; beyond 5 years: 25%. Growth
• Rapid.
• Tumors spread by direct infiltration or along the CSF
pathways in the subarachnoid space, seeding cranial and
peripheral nerve roots.

Histology
• Identical to the testicular seminoma: typically biphasic,
being composed of two distinct populations of cells. The
larger germ cells have abundant cytoplasmic glycogen and
alkaline phosphatase activity. The stroma contains variable
numbers of lymphocytes which are both T and B cells.
• Germinoma:
– Macroscopic: a firm, discrete mass, usually arising in the
pineal region, usually reaches 3–4 cm (1.2–1.6 in) in
greatest diameter. Inferiorly, it compresses the superior
colliculi and sometimes the superior surface of the cere-
bellum, and narrows the aqueduct of Sylvius. Anteriorly,
it may extend into the third ventricle and compress the
hypothalamus. It may also arise in the floor of the third
ventricle (a suprasellar germinoma or ectopic pinealoma).
– Microscopic: large, spherical epithelial cells separated by
a network of reticular connective issue, which contains
many lymphocytes.
– Growth: germinomas preferentially infiltrate the pineal
region, hypophysis, cavernous sinus, optic chiasm and
pathways, third ventricle wall, or anterior half of the
lateral ventricle.
• Embryonal carcinoma: the most primitive and undifferen-
tiated example of this group of tumors; immunocyto-
chemistry demonstrates alpha-fetoprotein in tumor cells.
Germ Cell Tumors of the CNS 389

• Endodermal sinus tumors: these exhibit a tendency to INVESTIGATIONS


form structures resembling the yolk sac. Imaging
• Choriocarcinoma: a highly vascular tumor characterized by Epidermoid tumors
differentiation into trophoblastic tissue; prone to massive • May be intra- or extradural.
hemorrhage. Immunocytochemical methods readily • Intradural tumors typically arise in the cerebellopontine
demonstrate chorionic gonadotrophin in tumor tissue. angle cistern, the suprasellar or the prepontine cisterns,
• Teratomas: tumors with the ability to differentiate into or the pineal region.
elements from all three germ layers. Teratomas may • Intradural tumors (less common) tend to fill the space in
contain mature components such as hair, teeth, cartilage, which they arise in a rather insidious fashion.
muscle, and bronchial wall. These elements are arranged • Usually tumors appear as low density mass lesions on CT
in a haphazard, non-functional manner, but they can be (may mimic CSF unless closely inspected) which tend to
benign. Immature anaplastic elements may be found envelop normal structures, and do not enhance with
alongside benign elements, or they may make up the contrast.
entire tumor. Teratomas may contain other tumor types, • Tumors appear hypointense on T1W MRI (488) and
especially embryonal carcinoma. hyperintense on T2WI (similar to CSF) (489), but on
proton density imaging are inhomogeneous and
WHO classification of histology of germ cell hyperintense to CSF.
tumors of the CNS • Extradural epidermoid tumors (more common) arise in the
• Germinoma. diploic space, the petrous or temporal bones and look like
• Embryonal carcinoma. rounded, well defined bony lesions with sclerotic borders.
• Yolk-sac tumor (endodermal sinus tumor): produces
alpha-fetoprotein.
• Choriocarcinoma: produces the beta subunit of chorionic
gonadotrophin. 488
• Teratoma:
– Immature.
– Mature.
– With malignant transformation.
• Mixed germ cell tumors: numerous combinations of
diverse histology and prognosis.

ETIOLOGY AND PATHOGENESIS


Potential genetic predisposition
Associations with genetic disorders, phakomatosis, meta-
chronous and synchronous extracranial neoplasms.

CLINICAL FEATURES

Hypothalamic dysfunction
• Diabetes insipidus.
• Sleep disturbance (due to lack of melatonin nocturnal
rise in those with pineal germ cell tumors).
• Precocious puberty (boys with germinoma).

Midbrain compression by hydrocephalus or tumor 489


• Parinaud’s syndrome:
– Supranuclear paresis of upward gaze.
– Dilated pupils that react to accommodation but not to
light.
– Effects are due to compression of the tegmentum of the
upper midbrain by a dilated posterior part of the third
ventricle (hydrocephalus) rather than local pressure
effects of the tumor.
• Ataxia of the limbs.
• Chorea of the limbs.
• Spastic quadriparesis.

488, 489 T1W (488) and T2W sagittal (489) MRI of an epidermoid
of the planum sphenoidale.The lesion creeps along below the frontal
lobes (arrows) widening the space between the bone and the brain. It
does not enhance.
390 Tumors of the Central Nervous System

Dermoid tumors and teratomas DIFFERENTIAL DIAGNOSIS


• Tumors arise in similar sites to epidermoids except that Pineal region tumors
they favor the midline, whereas epidermoids favor sites Pineal parenchymal cell tumors (pinealoma)
away from the midline. • Pineocytoma.
• Tumors contain calcification, fat and bone as well as • Pineoblastoma.
cystic elements.
• On CT (490) they appear as mixed density lesions with Pineal germ cell tumors
calcification, fat and bony elements, but are better seen • Germinoma (atypical teratoma, resembling the
on MRI (491) seminoma of the testicle).
• On T1W MRI (492) the low signal of calcification or • Teratoma (with cellular derivatives of all three germ
bone and the high signal of fat suggest a dermoid. layers).
• Dermoids may rupture and CT or MRI may show fat • Glioma.
globules spread through the CSF.
Other tumors
Teratomas • Astrocytoma.
• Occur more often in males than females. • Glioblastoma.
• Tumors are common in the pineal region (they make up • Ependymoma.
one-third of pineal tumors) and the suprasellar cistern. • Meningioma.
• They may contain fat, bone, calcification, cysts and • Ganglioglioma.
dermal elements and may bleed.
• Enhancement is variable depending on the constituents. Cystic structures
• Teratomas make up one-third of pineal tumors (493, • Dermoid cyst.
494) mostly in males, and also in the suprasellar cistern. • Epidermoid cyst.
• Simple pineal cyst.
Blood tests
• β-subunit of HCG: may be increased in embryonal Pituitary region tumors
carcinoma, choriocarcinoma, and malignant teratoma. • Optic pathway astrocytoma.
• Alpha-fetoprotein can be detected in embryonal • Pituitary adenoma.
carcinoma and endodermal sinus tumors (embryonal • Craniopharyngioma.
tumor with histologic features similar to those found in
the visceral yolk sac). DIAGNOSIS
A definitive diagnosis predicting histology and malignancy
Pituitary function serum tests (if the tumor is in the cannot be achieved by CT or MRI alone; surgical biopsy or
pituitary region) resection is required to establish firmly an accurate
• Testosterone, sex hormone binding globulin and free histologic diagnosis that guides the extent of adjuvant
androgen index. therapy.
• Estradiol.
• FSH. PROGNOSIS
• LH. Prognosis is governed by the most malignant component in
• Thyroxine and TSH. the tumor.

CSF Good prognosis


• Cells: may reveal lymphocytes and tumor cells (which 10-year survival rate >85%:
have seeded the subarachnoid space). • Solitary pure germinoma.
• Alpha-fetoprotein. • Mature teratoma.
• β-subunit of HCG.
Intermediate prognosis
Biopsy • Germinoma with an elevated level of serum β-HCG.
Frameless stereotactic surgery is a safe and effective method • Extensive (>4 cm [1.6 in] diameter) or multifocal
of sampling deep intracranial tissues, carrying a 0–0.5% mor- germinoma.
bidity rate, but biopsy of a small specimen of a large tumor • Disseminated germinoma.
may lead to an erroneous pathologic diagnosis when a het- • Immature germinoma.
erogeneous tumor is encountered. It is particularly recom- • Mixed germ cell tumors consisting of germinoma with
mended for patients with multifocal or disseminated tumors. either mature or immature teratoma.
Open surgical resection provides a more accurate
histologic diagnosis, and thereby facilitates more appropriate Poor prognosis
and adequate treatment than stereotactic biopsy. • Teratoma with malignant transformation.
Furthermore, surgical resection or significant bulk reduction • Embryonal carcinoma.
of a solitary mass may enhance the efficiency of adjuvant • Yolk sac tumor.
therapy, particularly in the case of malignant germ cell • Choriocarcinoma.
tumor. • Mixed germ cell tumors including a component of
embryonal carcinoma, yolk sac tumor, choriocarcinoma,
or teratoma with malignant transformation.
Germ Cell Tumors of the CNS 391

490 491

490–492 CT (490),T2W axial (491) and T1W sagittal 492


(492) MRI of a dermoid tumor in the brainstem (unusual
site).The lesion is very difficult to see on CT apart from its
displacement of the IVth ventricle. Note the ‘tail’ of the
lesion visible on MRI (arrow) giving a clue as to its origin.

493

494

493, 494 Axial CT with contrast (493) and T1W sagittal MRI without contrast (494) of a pineal teratoma. Note the mass
in the pineal which enhances (arrows).
392 Tumors of the Central Nervous System

TREATMENT • Craniospinal radiotherapy with a high dose local boost,


Management is determined by the histologic and serologic and intensive combination chemotherapy (regimens
diagnosis and extent of disease. including ifosfamide, cisplatin [or carboplatin] and
etoposide) is used.
Pineal region tumors • High dose chemotherapy with autologous bone marrow
• Frequently non-germinomatous germ cell tumors. transplant or peripheral blood stem-cell support has not
• Huge masses should be biopsied. If malignant, been successful to date but may have a role in the salvage
neoadjuvant chemotherapy should be given before the setting of patients with relapsed or disseminated non-
second radical surgical resection. germinomatous germ cell tumors in the CNS.
• Smaller tumors can be debulked safely, and if malignant, • Daily oral etoposide cycles may be employed in patients
radically removed. with extracranial germ cell tumors who have failed to
• Shunting procedures for hydrocephalus, such as respond to first-line chemotherapy but have shown a
ventriculo-peritoneal shunt, are rarely necessary and may response to salvage therapy; and also as a maintenance
lead to fatal peritoneal metastases. chemotherapy in patients with poor prognosis non-
• Histologically verified pineal germinomas can be germinomatous germ cell tumors.
remarkably reduced in size only through a course of low- • Malignant teratomas should be surgically removed if
dose radiotherapy or chemotherapy alone (see possible because this can be curable, but its applicability
Germinomas, below). depends on the site and size of the tumor.

Neurohypophyseal and hypothalamic COMPLICATIONS OF TREATMENT AND


region tumors THEIR MANAGEMENT
• Preferentially infiltrate the optic pathways. Lack of neurohypophyseal and pituitary hormones
• Biopsy or partially remove when possible. • Infertility, hypogonadism, small stature.
• Caused by tumor invasion or high-dose radiotherapy.
Suprasellar tumor confined to the optic pathways: • Pituitary hormone replacement therapy is required by
• Usually germinomas (see below). most children.
• May require craniotomy for biopsy.
Lack of regulation of essential endocrine
Germinoma (solitary or multifocal) processes (e.g. sleep cycle)
• Highly radiosensitive and historically associated with a Oral melatonin may improve sleep disturbance in children
high surgical mortality. with pineal tumors.
• Radiotherapy to the localized field, via appropriate
irradiation ports, following active chemotherapy, can be
curable. A reduced dose of 24 Gy (2400 rads) in daily
fractions of 2 Gy (200 rads) or less, minimizes adverse
effects of radiotherapy such as cognitive retardation and VON HIPPEL–LINDAU DISEASE (VHL)
neuroendocrine deficiencies (growth hormone deficiency
occurs with radiation doses >24 Gy (2400 rads) to the
posterior hypothalamus). DEFINITION
• Prophylactic craniospinal or ventricle irradiation is not A rare autosomal dominantly inherited, and occasionally sporadic,
indicated because of associated morbidity in the growing neuroectodermal disorder that is characterized by a predisposition
child. to tumor development in the retina, cerebellum, spinal cord,
• Adjuvant pre-radiation chemotherapy, with a combina- pancreas and kidneys; and usually presents in early adulthood
tion including either cisplatin or carboplatin and with visual or neurologic symptoms due to a hemangiomatous
etoposide, may decrease the total dose of radiation malformation of the retina or an associated hemangioblastoma
necessary for inducing tumor resolution, and enhance of the cerebellum. It is one of the phakomatoses (disseminated
control in malignant germ cell tumors. Chemotherapy hereditary hamartomas), along with conditions such as
alone is insufficient, and is often associated with an early Sturge–Weber syndrome and von Recklinhausen’s disease. Dr
relapse of disease. Eugen von Hippel, a German ophthalmologist, first described
• Disseminated germinoma should be treated with the familial nature of retinal hemangioblastomas, and Dr Arvid
craniospinal irradiation. Lindau, a Swedish ophthalmologist, first recognized that
• If the initial treatment was 24 Gy (2400 rads) of local cerebellar and retinal hemangioblastomas are part of a larger
irradiation, recurrent germinoma can be treated with the ‘angiomatous lesion of the CNS’ and the condition is inherited.
same or a larger dose of radiotherapy, and the
craniospinal field can be selected. EPIDEMIOLOGY
• Prevalence: 1 in 40 000.
Malignant germ cell tumors • Age: mean age of onset 26 years; range 1–78 years:
Examples include mixed germ cell tumors, with numerous – Retinal hemangioblastoma: mean age at diagnosis 25
combinations of diverse histology and prognosis, such as a years (range 1–67 years).
mixed germ cell tumor composed of immature teratoma – Cerebellar hemangioblastoma: mean age at diagnosis
with embryonal carcinoma: 30 years (range 1–78 years).
• High incidence of subarachnoid dissemination and spinal – Renal cell carcinoma: mean age at diagnosis 37 years
metastases. (range 16–67 years).
• Gender: M=F.
von Hippel–Lindau Disease (VHL) 393

PATHOLOGY • The susceptible target organs in VHL disease can be


Capillary hemangioblastomas grouped as:
Comprising endothelial cells and pericytes of: – Cerebellar and retinal cells (resulting in heman-
• Retina (45–59% of patients), bilateral in half . gioblastomas).
• Cerebellum (44–83%), brainstem (up to 18%), spinal cord – Neural crest cells (resulting in pheochromocytomas,
(13–44%), or adrenal glands. Cerebellar hemangioblastoma paragangliomas, and possible islet cell tumors).
is the most common primary intrinsic tumor in the posterior – Glandular viscera (resulting in renal, pancreatic,
fossa in adults (10% of all cerebellar tumors). Up to 20% of epididymal and endolymphatic sac tumors).
cerebellar hemangioblastomas secrete erythropoietin
sufficient to cause symptomatic polycythemia. Spinal cord CLINICAL FEATURES
hemangioblastomas have a predilection for the conus • Symptoms usually do not become evident until late
medullaris and craniocervical junction, and are associated adolescence or adulthood.
with a syringomyelic lesion in >70% of cases. • Progressive visual loss:
– Presenting symptom in up to 50% of patients due to
Additional lesions enlargement of the retinal angioma (bilateral in half of
Present in some cases, including: cases), exudative or tractional retinal detachment,
• Angiomas and cysts of the liver, pancreas, and kidneys vitreous hemorrhage, macular edema or epiretinal
(59–63%). membrane formation causing macular distortion.
• Adenomas of the liver, epididymis and adrenals. – Ophthalmologic examination may reveal a decrease in
• Renal cell carcinoma (24–45% of patients, onset beyond age 20 visual acuity, a visual field defect, enlargement of the
years): secretes erythropoietin which can cause polycythemia. ‘feeder’ retinal vessels leading to a peripheral retinal
• Pheochromocytoma (7–18%): probably of neural crest origin, capillary hemangioma (495) and exudative changes
generally considered benign but can metastasize. (signs of lipid deposition) in the macula.
• Ectopic paraganglioma, histologically identical to pheochro- • Progressive ataxia of limb movements, speech
mocytoma, may arise in the carotid body, glomus jugulare, (dysarthria) and swallowing (dysphagia) due to cerebellar
periaortic tissues, spleen and kidney. hemangioblastoma, or symptoms of increased intracranial
• Pancreatic islet cell tumor: probably of neural crest origin, pressure such as headache, nausea, and vomiting:
more frequent in patients with pheochromocytoma; islet cell presenting symptoms in about 20% of patients.
tumors are capable of metastasizing and may become • Spinal pain, focal sensory and motor deficits from spinal
symptomatic with biliary obstruction or endocrinopathy. tumors.
• Papillary cystadenoma of the epididymis: (10–26% of men • Polycythemia due to production of erythropoietin by
with VHL), when bilateral are virtually pathognomonic of cerebellar hemangioblastomas and renal cell carcinomas.
VHL disease; histologically similar lesions are seen in the • Episodes of uncontrolled hypertension, perspiration,
broad ligament of females with VHL disease. headaches and anxiety attacks due to massive release of
catecholamines by pheochromocytoma.
ETIOLOGY AND PATHOPHYSIOLOGY
• Inherited: A thorough search for other manifestations of the disease
– Autosomal dominant inheritance. (see Pathology, above) is also required.
– High penetrance (i.e. a high proportion of people with
the gene develop the disease; penetrance is in excess of DIFFERENTIAL DIAGNOSIS
90% by 60 years of age). • Cerebellar tumor.
– Variable expression (i.e. the severity of the disease varies • Cerebellar degeneration.
substantially between affected patients, even within a • Multiple sclerosis.
family), although some families seem to have a
propensity for pheochromocytoma.
• The gene responsible for VHL is located on the short 495
arm of chromosome 3, 3p25-26, the same region that is
associated with renal cell carcinoma, and close to the
locus of the RAF-1 oncogene.
• The VHL disease gene is a tumor suppressor gene, a
gene whose normal function is to regulate cell growth.
When both copies of the gene are inactivated by means
of mutation or loss (e.g. recombination or somatic
deletion), cell growth is unchecked and tumors result.
• Patients with VHL disease inherit the germ line mutation
on the short arm of one allele of chromosome 3 from the
affected parent, and one normal allele from the
unaffected parent.
• A ‘two-hit hypothesis’ has been proposed to explain the
development of tumors: the abnormal germ line
mutation is present in all cells, but only those cells in
target organs that develop a second mutation in the 495 Ocular fundus showing a peripheral retinal capillary hemangioma
normal allele go on to develop tumors. with enlarged ‘feeder’ retinal vessels. (Courtesy of Dr AM Chancellor,
Tauranga, New Zealand.)
394 Tumors of the Central Nervous System

INVESTIGATIONS • Annual abdominal (renal, adrenal and pancreatic)


• Hemoglobin and packed cell volume (?polycythemia). ultrasound, with abdominal CT scan every 2–3 years,
• Fluorescein angiography. pending renal findings.
• CT or MRI brain scan (496, 497): the lesion(s) appears
cystic with a solid nodule against the wall (which may be Asymptomatic at-risk relative
very small and difficult to see without contrast) • Annual physical and neurologic examination, and blood
(498–500) but can also appear completely solid or pressure measurement.
cystic. Hydrocephalus is frequent. • Annual plasma normetanephrine (and metanephrine),
• Cerebral angiography (501): shows a vascular nodule plasma catecholamines (norepinephrine and epinephrine)
with an adjacent avascular mass comprising a tightly and 24-hour urine collection for catecholamines,
packed cluster of small vessels about 1–2 cm (0.4–0.8 in) metanephrines and vanillylmandelic acid (VMA) from
in diameter, with or without draining veins. age 10 years.
• MRI and angiography are good for demonstrating • Annual indirect ophthalmoscopy from age 3 years, with
additional small and possibly asymptomatic lesions. fluorescein angioscopy when necessary.
• Plasma and 24-hour urine catecholamine (vanillyl- • MRI of the brain and spine every 3 years from age 11 to
mandelic acid [VMA] and metadrenaline) analysis. age 60, then every 5 years.
• Abdominal ultrasound. • Annual abdominal ultrasound from age 11, with ab-
• Abdominal MR or CT scans to image the kidneys, dominal CT scan every 2–3 years, pending renal findings.
adrenals, liver and pancreas. Abdominal CT scan is more
sensitive than ultrasound for detecting small renal cysts PREDICTIVE TESTING
and carcinomas. The discovery of the specific gene means that a particular
• DNA analysis. family’s genetic mutations can be mapped, and reliable
preclinical diagnosis in close relatives of affected individuals
DIAGNOSIS is possible by analysing the individual’s DNA for the family’s
Clinical mutation. Skilled counselling based on full knowledge of
• Positive family history of retinal or CNS the disease, its natural history, and the impact of a positive
hemangioblastoma. and negative diagnosis on the individual and other family
And: members is essential.
• One hemangioblastoma or visceral lesion (renal tumor,
pancreatic cyst or tumor, pheochromocytoma, papillary TREATMENT
cystadenoma of the epididymis); renal cysts and • Small peripheral retinal angiomas (<3 mm [<0.1 in]) are
epididymal cysts are too frequent in the population to be best treated by argon laser photocoagulation, whilst
reliable markers of VHL disease. cryotherapy is used for larger peripheral tumors. Larger
Or: retinal lesions may require penetrating diathermy or local
• Negative family history of retinal or CNS resection. As treatment of small lesions is more successful
hemangioblastoma. there is further impetus to screen patients at risk regu-
And: larly. Treatment may increase the vascular leakage from
• Two or more hemangioblastomas. the tumor and produce an exudative retinal detachment.
Or: • Cerebellar hemangioblastoma should be removed
• Negative family history of retinal or CNS hemangioblastoma. surgically if possible: craniotomy with opening of the
And: cerebellar cyst and excision of the mural hemangio-
• One hemangioblastoma and a visceral lesion. matous nodule may be curative. Recurrences may occur.
• Renal cysts and carcinomas can be removed via a partial
Genetic nephrectomy if detected early. If bilateral nephrectomy
DNA analysis identifying a mutation or loss of the gene is necessary, a delay of 1 year is usually advocated
responsible for VHL disease on the short arm of between nephrectomy and transplantation to ensure that
chromosome 3, 3p25-26. no metastases will occur.
• Pheochromocytoma is usually treated with adrenalectomy.
SCREENING • Pancreatic islet cell tumors are treated with partial
The malignant potential of the visceral disease makes early pancreatectomy.
diagnosis mandatory.
PROGNOSIS
Asymptomatic affected patient • Vision: patients have >70% chance of retaining vision of
• Annual physical and neurologic examination, blood 6/18 or better with small retinal tumors, and 50%
pressure measurement, plasma normetanephrine (and chance with larger tumors (>4 mm [0.2 in]).
metanephrine), plasma catecholamines (norepinephrine and • Renal cell carcinoma develops eventually in most long
epinephrine) and 24-hour urine collection for catechola- term survivors. More unusual and less serious
mines, metanephrines and vanillylmandelic acid (VMA). complications include renal, pancreatic, and ependymal
• Annual indirect ophthalmoscopy, looking for signs of the cysts, although the malignant potential of these lesions,
peripheral lesions or the secondary exudative response in especially those in the kidneys, remain unclear.
the macular region, with fluorescein angioscopy when • Mean age at death is about 41 years, median 49 years.
deemed necessary, but not routinely. • Cause of death is renal cell carcinoma, and less
• MRI of the brain and spine every 3 years to age 60, then commonly cerebellar hemangioblastoma.
every 5 years.
von Hippel–Lindau Disease (VHL) 395

496, 497 MRI brain 496 497


scans.T1W image
sagittal plane (496),
showing capillary
hemangioblastomas
as low densities in
the dorsal medulla
and upper cervical
spinal cord (arrows);
T2W image axial
plane (497),
showing a capillary
hemangioblastoma
as a high density
mass in the dorsal
medulla and floor of
the fourth ventricle
(arrows). (Courtesy
of Dr AM
Chancellor,Tauranga,
New Zealand.)

498–500 T1W 498 499


midline sagittal (498)
and T2W axial (499)
MRI of a cerebellar
hemangioblastoma.
Note the lesion is
mainly cystic, but
there is a small
nodule near the
superior aspect
(arrow). Following
contrast on a T1W
coronal image (500),
the nodule is seen to
enhance (arrow).
This appearance is
typical of a cerebellar
hemangioblastoma.

500 501

501 Vertebral angiogram, lateral view, showing a cerebellar hemangio-


blastoma (arrows). (Courtesy of Dr AM Chancellor,Tauranga, New
Zealand.)
396 Tumors of the Central Nervous System

METASTASES TO THE CNS PATHOLOGY


Skull and dura metastases
Common sources
DEFINITION • Carcinoma of the breast and prostate.
Dissemination of tumor cells beyond their primary site to • Multiple myeloma.
the CNS.
Meningeal (craniospinal) metastases
EPIDEMIOLOGY About 5% of neurologic metastases; widespread dissemination
• Incidence: 4 (95% CI: 1–9) per 100 000 per year in one of tumor cells throughout the meninges and ventricles.
study and 14 (95% CI: 12–16) per 100 000 per year in
another study. The commonest cause of multiple Common sources
intracranial masses in the Western world, and the • Adenocarcinoma of the breast, lung, and gastrointestinal
commonest cause of a solitary cerebellar mass in patients tract.
over 40 years old; 20–40% of patients with cancer. • Carcinoma of the prostate.
• Age and gender: any age and either sex (depending of • Melanoma.
course on the nature of the primary malignancy). • Leukemia.

Brain metastases
• May be solitary (up to half of cases) or multiple.
• Metastases form a circumscribed mass, usually solid but
sometimes cystic.
• They may be hemorrhagic, particularly metastases from
melanoma and choriocarcinoma, and sometimes lung,
breast, thyroid, and kidney.
• They excite considerable vasogenic edema but little glial
reaction.

Common sources (in adults)


• Carcinoma of the lung (the source of 30% of brain
metastases; 35% of bronchial carcinomas metastasize to the
brain), breast (15% of brain metastases), gastrointestinal
502 tract (particularly colon and rectum), kidney and thyroid.
• Malignant melanoma (75% of melanomas metastasize to
the brain) (502).

Uncommon sources (in adults)


• Carcinoma of the gallbladder, liver, testicle (57% of
testicular tumors metastasize to the brain), uterus, ovary
and pancreas.
• Rhabdomyosarcoma.
• Ewing’s tumor.
• Lymphoma.
• Carcinoid.

Atypical sources
Carcinoma of the prostate, esophagus, oropharynx and skin
almost never metastasize to brain.

Complications
• Mass effect.
• Hemorrhage: renal, thyroid, choriocarcinoma,
melanoma, retinoblastoma, lung, breast; in fact, in
patients with recurrent or multiple unexplained
intracranial hemorrhages, think of hemorrhagic
metastases.
• Calcification: mucinous adenocarcinoma: colon,
stomach, ovary, breast; osteosarcoma, chondrosarcoma.

502 Multiple sections through the brain in the coronal plane showing
multiple areas of hemorrhage into metastases of malignant melanoma.
(Courtesy of Professor BA Kakulas, Royal Perth Hospital,Western
Australia.)
Metastases to the CNS 397

ETIOLOGY AND PATHOPHYSIOLOGY DIFFERENTIAL DIAGNOSIS


Metastases to the skull and dura • Primary brain tumor: glioblastoma multiforme: similar
Hematogenous usually, via the systemic circulation (e.g. clinical features.
carcinoma of the breast) or via Batson’s vertebral venous • Paraneoplastic syndromes (see p.401) not due to
plexus: a valveless venous system that runs the length of the compression or invasion of the nervous system by tumor.
vertebral column from the pelvic veins to the large venous • Brain abscess.
sinuses of the skull, bypassing the systemic circulation (e.g. • Cerebritis/encephalitis.
carcinoma of the prostate). • Subdural hematoma: occasionally a carcinoma
metastasizes to the subdural surface and compresses the
Metastases to the brain brain, mimicking a subdural hematoma.
Hematogenous usually, via the systemic circulation. • Stroke: hemorrhagic (e.g. into a tumor) or ischemic (e.g.
tumor embolism causing brain infarction).
CLINICAL FEATURES • General paralysis of the insane (see p.312): can mimic the
Metastases to the skull and dura diffuse cerebral disturbance seen in some patients with
• Skull convexity metastases: usually asymptomatic. metastases.
• Skull base metastases: usually cause dysfunction of the • Psychologic: if only headache and vomiting are present.
pituitary gland or the cranial nerve roots.
INVESTIGATIONS
Metastases to the craniospinal meninges CT brain scan
• Headache. If metastases are suspected, a contrast-enhanced CT scan is
• Backache. the first line investigation.
• Cranial and peripheral neuropathies or radiculopathies
(particularly the cauda equina). Bony (skull) metastases
• Dementia. These are readily recognized on CT (503) and bone scans
• Hydrocephalus. (504).

Metastases to the brain


Similar to glioblastoma multiforme (see p.364).

Syndromes
• Focal or multifocal neurologic dysfunction (e.g. seizures,
aphasia, focal weakness, ataxia), which is usually insidious
in onset and progressive; but can be abrupt in onset
following hemorrhage into a tumor, or embolism of
tumor or thrombus (marantic endocarditis) causing
cerebral infarction.
• Raised intracranial pressure (e.g. headache).
• Diffuse encephalopathy: headache, confusion, forget-
fulness, depression.

503 CT brain scan on 503 504


bone settings showing
multiple lucent lesions
in the skull vault
(arrows) due to
prostatic secondaries.

504 Nuclear bone


scan of a patient with
disseminated
carcinoma of the
breast showing
multiple hyperdense
areas of increased
tracer uptake due to
bony metastases in the
skull and spine.
398 Tumors of the Central Nervous System

Brain metastases Meningeal metastases


These are: • May be difficult to detect with imaging unless there are
• Iso- or hypodense to normal brain. discrete nodules.
• Located near the gray-white matter junction usually. • Hydrocephalus may be present.
• Show marked contrast enhancement (may be ring • Occasionally contrast enhancement of the meninges may
enhancement) (505, 506). be seen (MRI is more sensitive than CT for that)
• Mass effect. (507–510).
• Substantial white matter edema. • Parenchymal metastases are present in about 40% of cases.

505 506 505 CT brain scan


with contrast showing
multiple enhancing
peripheral nodules
(arrows) typical of
metastases.These
were from a primary
carcinoma of the lung.

506 CT brain scan


with contrast showing
multiple partially
calcified metastases
from a primary
carcinoma of the
colon.

507 508 507, 508 T1W sagittal MRI with


contrast (507) and T2W sagittal
MRI (508) showing multiple
enhancing nodules on the nerve
roots (arrows) in a patient with
malignant meningitis.
Metastases to the CNS 399

MRI brain scan DIAGNOSIS


• More sensitive to small lesions than CT (511, 512) and • CT and MRI brain scan, before and after the injection of
may demonstrate multiple lesions when CT only demon- contrast, are the most sensitive tests for the diagnosis of
strated one large one (thus helping to establish the diag- brain metastases.
nosis of metastases). • Surgery has an important role in establishing the
• Variable appearance of lesions, depending on the type and diagnosis (e.g. frameless stereotactic biopsy or resection),
presence of hemorrhage or calcification, but generally they even for patients known to have a primary cancer
appear as masses with edema that enhance. elsewhere because a new brain lesion in these patients
may not necessarily be a metastasis.
CSF
Meningeal metastases TREATMENT
• Pressure: normal or increased. • Aims to obtain local control of the brain tumor and to
• Cells: normal or lymphocytic pleocytosis, cytology preserve neurologic function.
(cytocentrifugation, Millipore filtering, cell typing). • Corticosteroids.
• Protein: normal or elevated.
• Glucose: normal or low.
• Biochemical markers of cancer: lactate dehydrogenase,
beta glucuronidase, β2-microglobulin and carcino-
embryonic antigen.

509 T1W axial MRI with 509 510


contrast of the brainstem
showing multiple enhancing
nodules. Note the peripheral
sites and the brainstem
encasement (arrows)
indicating that these are in
the leptomeninges. (Courtesy
of Dr D Collie, Dept of
Neuroradiology,Western
General Hospital, Edinburgh.)

510 CT brain scan with


contrast showing marked
enhancement of all the
meninges (arrows) in a patient
with malignant meningitis
secondary to neuroblastoma
metastases. (See 509.)

511 T1W axial MRI without


contrast through the upper
brainstem showing a small
mass at the left petrous apex 511 512
adjacent to the carotid
siphon (arrows).This was a
solitary metastasis from a
lung primary in a patient
presenting with pain in the
left side of the face.

512 T1W MRI with contrast


showing multiple tiny
enhancing areas (arrows)
including one in the left side of
the upper pons (arrowhead).
These were breast carcinoma
metastases and the patient
had complained of mild
headache and had a left Vth
nerve palsy.The CT scan had
been normal.
400 Tumors of the Central Nervous System

Single brain metastases • Median survival free of functional disability is


• If the primary extracranial tumor is under control and if 2.5 months.
the metastasis is surgically accessible: surgical resection is • Older age (>60 years) increases the hazard of dying by
performed followed by whole brain irradiation to a dose 2.7 times, compared with younger patients.
of at least 20 Gy (2000 rads) in five fractions (and up to
two fractions per day, each of 2 Gy [200 rads] to a dose Multiple metastases
of 40 Gy [4000 rads]). • Untreated: median survival is 1 month; nearly all die
• If the primary extracranial tumor has progressed during from the brain tumor.
the previous 3 months, radiotherapy alone is sufficient. • Steroid treatment: median survival is 2 months.
• If the metastasis is surgically inaccessible and small • Whole brain irradiation: median survival is 3–6 months.
(<3 cm): stereotactic biopsy is performed, followed by
whole brain irradiation or preferably, entry into a trial Meningeal metastases
comparing whole brain irradiation with radiosurgery and • Median survival is 5.8 months.
whole brain irradiation. Stereotactic radiosurgery delivers • Treatment rarely stabilizes neurologic symptoms for
a high dose of focused radiation to a defined volume. more than a few weeks.
The three principal sources of radiation are x-rays (linear
accelerator), γ-rays (Gamma Knife – cobalt 60), and Independent prognostic factors for
charged particles (proton beam). The linear accelerators improved survival
are the most commonly used. • Age <65 years.
• Karnofsky performance status (high). (The Karnofsky
Multiple brain metastases performance score is a clinical grading scale which assigns
• Whole brain irradiation: the standard treatment. It gives patients a score ranging from 10–100 according to their
significant palliation. Radiosensitive tumors include ability to perform daily tasks.)
lymphoma, germ cell tumors, and small cell lung cancer. • Control of primary cancer.
Relatively insensitive tumors include melanoma, renal cell • Metastatic spread limited to the brain.
carcinoma, and thyroid carcinoma.
• Surgery to relieve significant neurologic dysfunction from
a large surgically accessible lesion should be considered Table 38 Classification of paraneoplastic syndromes
in a patient whose primary cancer is under control and
who is able to withstand surgery. Clinical syndrome Some associated neoplasms
CNS
Recurrent brain metastases Encephalomyelitis: Small cell lung carcinoma
If stable primary disease and good performance status, – Limbic encephalitis
consider any of: – Brainstem encephalitis
• Re-operation. – Myoclonus-opsoclonus
• Re-irradiation. – Cerebellar degeneration
• Chemotherapy: for chemosensitive tumors such as small Subacute cortical cerebellar Breast, ovary, ?lymphoma
cell lung cancer, choriocarcinoma, and breast cancer. degeneration
• Brachytherapy: utilizes radioactive isotopes, usually
iridium 192 or iodine 125, directly implanted into the Opsoclonus Breast
tumor site by catheters placed stereotactically to deliver Necrotizing myelopathy Lymphoma (retroperitoneal)
a high dose or radiation to the target. Small cell lung carcinoma
• Stereotactic radiotherapy, preferably as part of a clinical trial.
Peripheral nervous system (PNS)
Meningeal metastases Subacute sensory neuronopathy Small cell lung carcinoma
• Radiation therapy to the symptomatic areas. Peripheral neuropathies associated
• Intraventricular methotrexate. with IgM paraproteins:
– Motor neuropathy Lymphomas, plasma cell
PROGNOSIS – Motor/sensory demyelinating dyscrasias
More than half of patients die from causes other than their neuropathy
brain metastases. – Axonal neuropathy
Single metastasis in patients with controlled Neuromuscular junction syndromes
extracranial cancer Lambert–Eaton syndrome: Small cell lung carcinoma
• Median survival is 7 months if treated with radiotherapy – With cerebellar degeneration Small cell lung carcinoma
alone, 10 months with surgical excision and 12 months Myasthenia gravis Thymoma
with surgical resection and radiotherapy. Muscle
• Median survival free of functional disability is 4 months Polymyositis-dermatomyositis Gynecologic, lung,
if treated with radiotherapy alone and 9 months if treated lymphoma
with surgical resection and radiotherapy.
N.B. Myasthenia gravis is not a bona fide paraneoplastic syndrome
Single metastasis in patients with progressive but there is a strong association with thymomas in middle-aged
extracranial cancer and elderly patients.
• Median survival is 5 months (with radiotherapy alone).
Paraneoplastic Syndromes 401

PARANEOPLASTIC SYNDROMES CLASSIFICATION OF PARANEOPLASTIC


SYNDROMES (see Table 38)

DEFINITION PATHOLOGY
A term used to describe a number of disorders of the The nervous system may be affected at any site: cerebral
nervous system which are manifestations of the remote cortex, limbic system, brainstem, cerebellum, spinal cord,
effects (predominantly autoimmune) of a systemic cancer, spinal ganglia, peripheral nerve, neuromuscular junction,
and are not ascribable to nervous system metastases or to and muscle, by different pathologies:
destruction of vital systemic organs by the tumor or • Inflammatory (513–515): limbic encephalitis, brainstem
treatment of the tumor. encephalitis, poliomyelitis-like syndrome, and posterior
Neurologic disorders that are clinically and pathologically root ganglionitis.
identical to paraneoplastic syndromes also occur in the • Vascular: hypercoagulability, venous thrombosis, non-
absence of cancer but it is the statistical relationship between bacterial or marantic endocarditis, intravascular
the presence of cancer and the specific neurologic disorder coagulopathies.
that defines a given neurologic syndrome as paraneoplastic. • Immunologic: myasthenic syndrome of Lambert–Eaton,
For example, among patients with Lambert–Eaton myasthenia gravis, polymyositis-dermatomyositis complex.
myasthenic syndrome (LEMS) about 60% have small cell lung
cancer as the underlying cause and 40% do not. LEMS is The pathology of subacute myelopathies and of sensory
exceptionally rare in the general population yet it affects about and sensori-motor neuropathies is less clear.
3% of patients with small cell lung cancer.
Cerebellar degeneration
EPIDEMIOLOGY • Diffuse degenerative changes in the cerebellar cortex
• Incidence: rare. (pallor of the Purkinje cells) and deep cerebellar nuclei.
• Age: adults, occasionally children. • Perivascular and meningeal clusters of inflammatory cells.
• Gender: M=F.

513 514

515

513 Axial section through the pons showing


multifocal areas of gliosis and neuronal loss due to
paraneoplastic encephalitis.

514, 515 Low (514) and high magnification (515) sections showing
neuronal loss, gliosis and perivascular cuffing due to paraneoplastic
encephalitis.
402 Tumors of the Central Nervous System

Limbic encephalomyelitis (e.g. Lambert–Eaton syndrome: antibodies to voltage-gated


This syndrome results in subtotal neuronal loss, calcium channels [VGCCs] of motor nerve terminals;
neuronophagia, astrocytic gliosis, microglial proliferation, myasthenia gravis: antibodies to nicotinic acetylcholine
and perivascular cuffing in the limbic structures, especially receptor of the motor endplate; each is associated with small
the hippocampal gyrus, Ammon’s horn and amygdala. cell lung cancer [SCLC], can be induced in passive transfer
experiments, and responds to therapies which reduce
Myelopathy antibody titres). SCLC cells appear to express VGCCs.
Spinal cord disorders associated with malignant tumors:
amyotrophic lateral sclerosis, subacute necrotizing Secretion of non-immune origin soluble factors by
myelopathy, and subacute motor neuropathy. the tumor (e.g. neurohormones, neurotransmitters)

Neuropathy CLINICAL FEATURES


• Subacute pure sensory neuropathy: the primary lesion is • The clinical manifestations precede (in about two-thirds
dorsal root ganglion cell degeneration associated with of patients) by months and rarely years, or accompany
secondary degeneration of the ascending tracts in the (in about one-third) the manifestations of the underlying
posterior columns and sensory fibers in peripheral nerves. cancer.
• Axonal or demyelinating peripheral neuropathy. • Onset: typically subacute, occasionally abrupt.
• Clinical course: rapid progression to a peak over a few
Lambert–Eaton myasthenic syndrome (LEMS) weeks to a few months followed by stabilization.
Reduced number of functional, voltage-gated calcium
channels (VGCCs) at motor nerve terminals results in Limbic encephalomyelitis
reduced release of acetylcholine from the presynaptic motor • Confusion, impaired recent memory (forgetfulness),
nerve terminals in this syndrome. hallucinations, depression, personality change, sleep
disturbances, and behavioral disorder (agitation, anxiety,
Dermatomyositis/polymyositis depression) occur.
Acute muscle necrosis: dermatomyositis is due to a humoral • Seizures, myoclonus, sensory ataxia and brainstem and
immune attack on muscle capillaries causing muscle fiber spinal cord signs may also occur.
infarcts and perifascicular inflammation and atrophy. • Occurs as a remote manifestation of SCLC or, less
commonly, transitional cell carcinoma of the bladder,
ETIOLOGY mediastinal teratoma, malignant thymoma, and testicular
Most common underlying cancers carcinoma.
• Small cell lung carcinoma.
• Breast carcinoma. Cerebellar degeneration
• Ovarian carcinoma. • Limb and gait ataxia, nystagmus and dysarthria, sometimes
• Lymphoma. with diplopia, vomiting and pyramidal signs are present.
• Carcinoma of the uterus, bowel and other viscera • May be the presenting feature of carcinoma of the lung,
account for a minority of cases. breast and ovary.
• Anti-neuronal antibodies are found in most cases,
PATHOPHYSIOLOGY particularly when associated with ovarian or other
Several mechanisms have been suggested as a possible gynecologic malignancies.
pathogenesis:
Opsoclonus-myoclonus-ataxia
Hypothesis Example • Presents as irregular, chaotic, rapid, jerky, dysconjugate
Autoimmune process LEMS movement of the eyes in all directions of gaze, myoclonic
jerks and ataxia.
Toxin secreted by tumor ACTH → Cushing’s syndrome • It is a rare complication of neuroblastoma in childhood
PTHRP → hypercalcemia and even rarer in adults with cancer.

Competition for essential substrate Utilization of glucose by Myelopathy


sarcomas → hypoglycemia • Presents as paraparesis, sometimes associated with spinal
Carcinoid tumors compete myoclonus and spinal muscular atrophy with muscle
with brain for tryptophan → fasciculations.
pellagra-like syndrome • A rare complication of lymphoma and small cell lung cancer.

Opportunistic infection Papovavirus → progressive Peripheral neuropathy


multifocal leukoencephalopathy • Subacute pure sensory neuropathy (dorsal-root ganglionitis).
• Sensori-motor neuropathy:
PTHRP: Parathyroid hormone-related protein. – Four times more common than subacute pure sensory
neuropathy.
Immune-mediated – Associated with cancer of the lung, stomach, colon,
Most paraneoplastic syndromes are autoimmune (either breast, ovary, pancreas and testis; lymphoma and multiple
humoral or cell-mediated) and result from the production of myeloma.
antibodies directed against tumor-associated antigens that – Usually a distal, symmetric sensorimotor axonal
cross-react with important elements of the nervous system polyneuropathy.
Paraneoplastic Syndromes 403

– Occasionally, mimics acute Guillain–Barré syndrome or Nutritional deficiencies


relapsing chronic inflammatory demyelinating Syndromes result from vitamin B1, B12 and folate
neuropathy, particularly in patients with seminoma. deficiencies due to anorexia, vomiting, diarrhea, dysphagia
– Associated with the presence of anti-neuronal nuclear and gastrointestinal resections.
antibodies.
– Anti-Hu-antibody-associated encephalomyelitis with Intracranial hemorrhage
sensory neuropathy most commonly features sensory Thrombocytopenia, liver failure, hemorrhagic secondary
symptoms such as paresthesias, painful neuropathy, deposits can cause syndromes.
motor weakness, and diplopia.
– May respond to plasmapheresis or steroids. Marantic (non-infectious or non-bacterial
• Multiple mononeuropathy. thrombotic) endocarditis
• Entrapment and pressure neuropathies due to cachexia. Small, sterile (non-infected) thrombi on heart valves,
• Nutritional neuropathies. particularly mitral valve, give rise to multiple embolic cerebral
(and renal, splenic, gastrointestinal) infarcts. Blood cultures are
LEMS (see p.664) usually sterile and echocardiography may be normal.
Associated with SCLC in about 60% of cases; about 40% of Arteriography may reveal aneurysmal dilatation of distal vessels.
have no evidence of cancer but often have associated
autoimmune diseases, such as vitiligo, pernicious anemia and Cerebral vein and dural sinus thrombosis
thyroid disease. Procoagulant state associated with malignancy.
Myasthenic syndrome:
• Proximal muscle fatiguability and weakness (which Disseminated intravascular coagulation
increases with exertion). The syndromes of intravascular coagulopathy, dural venous
• Muscle wasting. sinus thrombosis and marantic endocarditis are all thought to
• Depressed deep tendon reflexes that increase after muscle be related to a hypercoagulable state associated with elevated
activation. levels of fibrinogen, increased prothrombin and activated
• Symptoms of autonomic dysfunction, such as dry mouth, partial thromboplastin times, and accelerated turnover of
impotence and constipation. fibrinogen and platelets. Also, some chemotherapeutic agents,
such as asparaginase and corticosteroids, alter hemostasis.
Disorders of muscle
• Cachectic myopathy: significant muscle wasting is present DIFFERENTIAL DIAGNOSIS
(type II fiber atrophy) but power is usually retained. The If not known to have cancer
cause is unknown. • Degenerative diseases with a chronic and progressive
• Proximal muscle weakness: present in patients with overt course: the rapid development of neurologic disability
or occult malignancy of the lung, breast, and gastro- over a few weeks to a few months followed by
intestinal tract, and is usually characterized by atrophy of stabilization differentiates paraneoplastic syndromes from
type II more than type I fibers. EMG may show more chronic and progressive degenerative diseases.
myopathic motor unit potentials. • Stroke: occasionally, the onset of paraneoplastic
• Polymyositis/dermatomyositis (see pp.681, 684): syndromes can be so abrupt that the disorder can be
– About 20% of patients have associated cancer; usually confused with a stroke.
lung, colon, ovary or breast. The association with cancer
is greatest in middle-aged women. If known to have cancer
– Subacute proximal muscle weakness, sometimes with • Previous therapy:
associated dysphagia, voice change and respiratory – Radiation.
muscle weakness is characteristic. – Chemotherapy.
– Muscle pain is not severe and the skin rash is sometimes – Immunotherapy.
very trivial. • Metastases.
• Metabolic disturbance:
Secondary non-metastatic complications of – Renal failure.
malignancy – Liver failure.
Metabolic/endocrine • Nutritional deficiency:
• Hyperviscosity syndrome. – Wernicke–Korsakoff syndrome.
• Hyponatremia due to syndrome of inappropriate – Cancer cachexia.
antidiuretic hormone (SIADH) secretion. • Opportunistic infection (lymphoma, immunosuppression):
• Adrenal failure due to metastases to adrenal glands. – PML.
• Cushing’s syndrome due to aberrant ACTH secretion. – Cytomegalovirus.
• Renal failure. – Herpes virus.
• Hypoglycemia. • Vascular disorders:
• Hypercalcemia. – Hypercoagulability.
• Carcinoid syndrome. – Hypocoagulability.
• Non-cancer related:
Opportunistic infections – Alcohol.
These occur particularly in patients with lymphomas and – Diabetes.
undergoing immunosuppressive treatment.
404 Tumors of the Central Nervous System

INVESTIGATIONS • Electromyography: features of neuropathy or myopathy


Investigations are indicated if neurologic symptoms are may be present.
typical of a paraneoplastic syndrome (typical syndrome,
subacute onset, and rapid evolution). Sural nerve biopsy
Shows a severe depletion of predominantly large myelinated
Anti-neuronal antibodies fibers, due to a neuronopathy, in pure sensory neuropathy.

Anti-Yo antibody (also called ‘type 1 anti-Purkinje cell If not known to have cancer, search for cancer
antibody’ [PCA-1], ‘APCA-1’ or ‘type 1 antibody’) • Liver function tests, and abdominal ultrasound if indicated.
• Polyclonal antibodies, bind to cytoplasm of Purkinje cells • Pelvic examination (rectal and vaginal), CT, ultrasound.
and proximal axon and dendrites, and fix complement. • Chest x-ray, CT scan.
• They are present in most cases of paraneoplastic subacute • Mammograms.
cerebellar syndrome, dysarthria and ataxia, particularly • Testis examination, ultrasound.
women with breast, ovarian or other gynecologic • Lymph node examination.
malignancies, and less commonly in SCLC and lymphoma. • Stool guaiac (i.e. blood in feces), and endoscopy if
• The lack of detectable anti-Yo antibodies in some indicated.
patients suggests that the antibodies, per se, may not be • Tumor markers in blood:
the cause but may reflect an immune response to an – CEA (cf. gastrointestinal cancer; carcino-embryonic
epitope. Patients with seronegative paraneoplastic antigen).
cerebellar degeneration have a high frequency of other – PSA (cf. prostatic cancer; prostatic specific antigen).
autoimmune paraneoplastic syndromes (such as – CA-125 (cf. ovarian cancer).
Lambert–Eaton syndrome). – BRCA1 (cf. breast cancer).

Anti-Hu antibody (also called ‘type 1 anti-neuronal nuclear If known to have cancer
antibody’ (ANNA-1), or ‘type 1Ia antibody’) • Search for metastases:
• Antibody binds to nuclei of neurons of central and – MRI of symptomatic area(s).
peripheral nervous system, sparing nucleoli. – CSF for malignant cells.
• The antibody is present in paraneoplastic encephalomyelitis, • Exclude metabolic problems:
sensory neuronopathy, and autonomic neuropathy. – Serum creatine (renal failure).
• It is mainly associated with SCLC, but also with – Liver function tests (liver failure).
neuroblastoma, and rarely non-SCLC, prostate cancer,
and seminoma. DIAGNOSIS
Although the clinical features precede the diagnosis of the
Anti-Ri antibody (also called ‘type 2 anti-neuronal nuclear underlying neoplasm in more than half of cases, diagnosis
antibody’ (ANNA-2), or ‘type 1Ib antibody’) during life can be difficult because clinical symptoms and
• Antibody binds to CNS but not peripheral nervous findings on ancillary investigations are not specific.
system neuronal nuclei, sparing nucleoli.
• It is present in paraneoplastic opsoclonus, ataxia, Diagnostic clues
nystagmus, dizziness, dysarthria. • Associated statistically with cancer (the neurologic
• The antibody is most frequently associated with breast disorder is known to occur with increased frequency in
cancer (total number of cases described is still small), and patients with cancer).
SCLC (one case). • Subacute onset (the neurologic disorder develops acutely
or subacutely and generally stabilizes after a few months).
Calcium channel autoantibodies • Severe neurologic disability.
Detected in about 30% of patients with LEMS. • CSF pleocytosis and increased IgG index (intrathecal
synthesis of IgG) is often present if the CNS is involved.
CSF • One neural cell type or structure is often affected (e.g.
• Cells: nil (if late diagnosis). Purkinje cells in PCD, cholinergic synapse in LEMS).
• Protein: modestly elevated. • Specific autoantibodies in serum or CSF (anti-Hu, anti-
• IgG and IgG index: elevated. Yo, anti-Ri).
• Oligoclonal bands: may be present.
• Anti-neuronal antibodies: may be present. Diagnostic criteria
In a given patient a reasonably secure diagnosis can be made
Nerve conduction studies and electromyography in the presence of a ‘typical’ paraneoplastic neurologic
• Sensory nerve action potentials (SNAPs): reduced or syndrome and:
absent in pure sensory neuropathy. • An appropriate underlying cancer (see Table 38, p.401).
• Compound muscle action potentials (CMAPs): low However, although paraneoplastic syndromes are usually
amplitude in LEMS. associated with specific cancers, any cancer can cause any
• Nerve conduction velocities (NCV): normal motor NCV paraneoplastic syndrome. In addition, the presence of
in pure sensory neuropathy; synaptic transmission cancer and a neurologic syndrome may be coincidental
(repetitive nerve stimulation): dramatic increase in and not causal. For example, about 2% of people
CMAP amplitude (by 2–20 times) following supra- diagnosed with motor neuron disease have, or develop
maximal stimulation of the motor nerve at rates of lung cancer within 2 years.
>40 Hz in LEMS.
Paraneoplastic Syndromes 405

• A well characterized antineuronal antibody in the serum Interfere with auto-reactive T lymphocyte activation
that has been associated with cancer, such as anti-Yo • Inhibit MHC recognition with anti-MHC monoclonal
antibody, which virtually establishes the diagnosis of antibodies.
cancer, particularly a gynecologic cancer, or anti-Hu • Compete for antigen binding with ‘design’ peptides.
antibody which virtually establishes the diagnosis of small • Inhibit T cell activation.
cell lung cancer. • Anti-CD4.
• Resolution of the syndrome when a cancer is discovered • Anti-TCR.
and adequately treated. However, most paraneoplastic
syndromes do not respond to treatment of the Induce tolerance: feed antigen
underlying neoplasm.
Suppress inflammation
TREATMENT • Corticosteroids.
Usually unsatisfactory; most patients fail to respond to either • Non-steroidal anti-inflammatory drugs.
treatment of tumor or suppression of immunity. The major
problems are that the pathogenesis is not fully understood, PROGNOSIS
the disorders cause neuronal destruction (so treatments Except on rare occasions, treatment of the associated cancer
need to be given early before the syndrome has evolved) and does not alter the clinical course.
the disease is uncommon for evaluation of treatments in
large controlled trials.

Treat the underlying tumor and remove


the antigen
• Surgery.
• Radiation.
• Chemotherapy.

Treat the paraneoplastic syndrome


Suppress humoral immunity
B-cells, antibody:
• Plasma exchange:
– Effective for LEMS.
– Fails to remove antibody from the CSF.
– Not effective for cerebellar degeneration, seropositive or
seronegative.
• Intravenous immunoglobulin. ?Effective for LEMS,
subacute cerebellar syndrome.
• Protein A immunoadsorption.
• Radiation of lymph nodes, brain.
• Immunosuppressive drugs (corticosteroids, azathioprine,
cyclosporine A, cyclophosphamide, chlorambucil,
methotrexate, tacrolimus [FK506]).

3,4-Diaminopyridine (up to 100 mg/day in divided


doses) is indicated in all patients with LEMS, with the
addition of prednisolone and azathioprine in non-smoking
patients who have severe symptoms and no evidence of
tumor. If severe symptoms develop in patients with SCLC
or smokers, then prednisone ± plasma exchange should be
considered. After control of symptoms the dosage can be
reduced to the most effective minimal dose.

Suppress cytotoxic T cells


CD8:
• Irradiation.
• Steroids.
• Cyclosporine.

Suppress helper T cells


CD4:
• Anti-CD4 antibodies.

Increase suppressor T cell function


• β-interferon.
406 Tumors of the Central Nervous System

Perry JR, Louis DN, Cairncross G (1999) Current VON HIPPEL–LINDAU DISEASE
FURTHER READING treatment of oligodendrogliomas. Arch. Neu- Burn DJ, Bates D (1998) Neurology and the kid-
rol., 56: 434–436. ney. J. Neurol. Neurosurg. Psychiatry, 65:
810–821.
Prognosis and prognostic factors Eisenhofer G, Lenders JWM, Linehan WM, et al.
BRAIN TUMORS
Davies E, Clarke C, Hopkins A (1996) Malignant (1999) Plasma normetanephrine and
General
cerebral glioma-I: Survival, disability, and mor- metanephrine for detecting pheochromocy-
De Angelis LM (2001) Brain tumors. N. Engl. J.
bidity after radiotherapy. BMJ, 313: toma in von Hippel-Lindau disease and mul-
Med., 344: 114–123.
1507–1512. tiple endocrine neoplasia type 2. N. Engl. J.
Hess KR (1999) Extent of resection as a prog- Med., 340: 1872–1879.
Classification nostic variable in the treatment of gliomas. J. Gläsker S, Bender BU, Apel TW, et al. (1999) The
Kleihues P, Cavenee WK (eds) (2000) Pathology Neuroncol., 42: 227–231.Nieder C, Petersen impact of molecular genetic analysis of the
and Genetics of Tumors of the Nervous System. S, Petersen C, et al. (2000) The challenge of VHL gene in patients with haemangioblas-
World Health Organisation Classification of p53 as prognostic and predictive factor in tomas of the central nervous system. J. Neu-
Tumors. IARC Press, Lyon. gliomas. Cancer Treatment Reviews 26: rol. Neurosurg. Psychiatry, 67: 758–762.
Smirniotopoulos JG (1999) The new WHO clas- 67–73. Gläsker S, Bender BU, Apel TW, et al. (2001) Re-
sification of brain tumors. Neuroimaging Clin. Olson JD, Riedel E, DeAngelis LM (2000) Long- consideration of biallelic inactivation of the
N. Am., 9(4): 595–613. term outcome of low-grade oligoden- VHL tumour suppressor gene in heman-
droglioma and mixed glioma. Neurology, 54: gioblastomas of the central nervous system. J.
Epidemiology 1442–1448. Neurol. Neurosurg. Psychiatry, 70: 644–648.
Surawicz TS, McCarthy BJ, Kupelian V, Jukich PJ, Smith JS, Perry A, Borell TJ, et al. (2000) Alter- Maher ER, Kaelin WG Jr (1997) von Hippel-Lin-
Bruner JM, Davis FG (1999) Descriptive epi- ations of chromosome arms 1p and 19q as dau disease. Medicine, 76: 381–391.
demiology of primary brain and CNS tumors: predictors of survival in oligodendrogliomas,
Results from the Central Brain Tumor Reg- astrocytomas, and mixed oligoastrocyomas. J. METASTASES TO THE CNS
istry of the United States, 1990–1994. Neu- Clin. Oncol., 18: 636–645. Karnofsky DA, Burchenal JH, Armistead GC Jr, et
rooncology , 1: 14–25. Wong ET, Hess KR, Gleason MJ, et al. (1999) al. (1951) Triethylene melamine in the treat-
Outcomes and prognostic factors in recurrent ment of neoplastic disease. Arch. Intern. Med.,
glioma patients enrolled in phase II clinical tri- 87: 477–516.
Etiology als. J. Clin. Oncol., 17: 2572–2578.
Hill JR, Kuriyama N, Kuriyama H, Israel MA PARANEOPLASTIC SYNDROMES
(1999) Molecular genetics of brain tumors. OPTIC NERVE GLIOMA Dalmau JO, Posner JB (1999) Paraneoplastic syn-
Arch. Neurol., 56: 439–441. Listernick R, Louis DN, Packer RJ, Gutmann DH dromes. Arch. Neurol., 56: 405–408.
(1997) Optic pathway gliomas in children Darnell RB (1999) The importance of defining
Imaging with neurofibromatosis 1: Consensus state- the paraneoplastic disorders. N. Engl. J. Med.,
Ricci PE (1999) Imaging of adult brain tumors. ment from the NF1 Optic Pathway Glioma 340: 1831–1833.
Neuroimaging Clin. N. Am., 9(4): 651–669. Task Force. Ann. Neurol., 41: 143–149. Smitt PS, Kinoshita A, Leeuw BD, et al. (2000)
Treatment Paraneoplastic cerebellar ataxia due to au-
MENINGIOMA toantibodies against a glutamate receptor. N.
Bampoe J, Bernstein M (1998) Advances in ra- Go RS, Taylor BV, Kimmel DW (1998) The nat-
diotherapy of brain tumors: radiobiology ver- Engl. J. Med., 342: 21–27.
ural history of asymptomatic meningiomas in Trivedi R, Mundanthanam G, Amyes E, et al.
sus reality. J. Clin. Neuroscience, 5: 5–14. Olmsted County, Minnesota, Neurology, 51:
Brada M, Cruickshank G (1999) Radiosurgery for (2000) Autoantibody screening in subacute
1718–1720. cerebellar ataxia. Lancet, 356: 565–566.
brain tumors. BMJ, 318: 411–412. Kondziolka D., Levy EI, Niranjan A, Flickinger
Fathallah-Shaykh H (1999) New molecular strate- JC, Lunsford LD (1999) Long-term out-
gies to cure brain tumors. Arch. Neurol., 56: comes after meningioma radiosurgery: Physi-
449–453. cian and patient perspectives. J. Neurosurg.,
Shapiro WR (1999) Current therapy for brain tu- 91: 44–50.
mors. Back to the future. Arch. Neurol., 56: McDermott MW (1996) Current treatment of
429–432. meningiomas. Curr. Opin. Neurol., 9:
Palliation 409–413.
Guidance prepared by a working group of the eth- Niiro M, Yatsushiro K, Nakamura K, et al. (2000)
ical issues in medicine committee of the Royal Natural history of elderly patients with asymp-
College of Physicians (2000) Principles of pain tomatic meningiomas. J. Neurol. Neurosurg.
control in palliative care for adults. J. R. Coll. Psychiatry, 68: 25–28.
Physicians Lond., 34: 350–352. Schrell UMH, Rittig MG, Koch U, Marschalek R,
Kissane DW, Spruyt O, Aranda S (2000) Palliative Anders M (1996) Hydroxyurea for treatment
care – new approaches to the problem of suf- of unresectable meningiomas. Lancet, 348:
fering. Aust. NZ. J. Med., 30: 377–384. 888–889.

GLIOMAS PITUITARY TUMORS


Cairncross JG (2000) Understanding low-grade Anderson JR, Antoun N, Burnet N, Chatterjee K,
glioma. A decade of progress. Neurology, 54: Edwards O, Pickard JD, N Sarkies N (1999)
1402–1403. Neurology of the pituitary gland. J. Neurol.
Bigner SH, Rasheed K, Wiltshire RM, McLendon Neurosurg. Psychiatry, 66: 703–721.
R (1999) Morphological and molecular ge- Bevan JS (1999) Management of pituitary tumors.
netic aspects of oligodendroglial neoplasms. BMJ, 318: 1226–1227.
Neurooncology, 1: 52–60. Levy A, Lightman SL (1994) Diagnosis and man-
agement of pituitary tumors. BMJ, 308:
Management 1087–1091.
Batchelor T (2000) Temozolomide for malignant
brain tumors. Lancet, 355: 1115–1116. ACOUSTIC NEUROMA
Karim ABMF, Maat B, Hatlevoll R, et al. (1996) Wright A, Bradford R (1995) Management of
A randomised trial on dose-response in radia- acoustic neuroma. BMJ, 311; 1141–1144.
tion therapy of low-grade cerebral glioma. Eu- PRIMARY CNS LYMPHOMAS
ropean Organisation for Research and Treat- Abrey LE (2000) Primary central nervous system
ment of Cancer (EORTC) Study 22844. Int. lymphoma. The Neurologist 6: 245–254.
J. Radiat. Oncol. Biol. Pins., 36: 549–556. DeAngelis LM (1999) Primary central nervous
Kaye AH, Walker DG (2000) Low grade astrocy- system lymphoma. J. Neurol. Neurosurg. Psy-
tomas: controversies in management. J. Clin. chiatry, 66: 699–701.
Neuroscience, 7: 475–483.
Chapter Fourteen 407

Degenerative
Diseases of the
Nervous System
ALZHEIMER’S DISEASE (AD) 516

DEFINITION
A clinico-pathologic entity comprising clinical evidence of
dementia (sufficient to interfere with social and work
function) and histopathologic evidence of neurofibrillary
tangles and senile plaques in the brain.

EPIDEMIOLOGY
The most common cause of dementia, accounting for
almost half of cases.
• Incidence: 123 per 100 000 population per year.
• Prevalence: increases exponentially with age, roughly
doubling every 5 years from about 1% of the population
at age 65 to about 16–20% at age 85 years.
• Age:
– Predominantly a disease of old age but not an invariable
accompaniment of ageing.
– About 5% of cases are below retirement age.
– Seldom presents before 40 years of age except in Down’s
syndrome.
• Gender: M=F.

PATHOLOGY
Macroscopic
Brain atrophy (516, 517) is present: narrowed cerebral
convolutions and widened cerebral sulci, enlarged third and 516 Surface of the brain of showing prominent cerebral sulci due to
lateral ventricles (hydrocephalus ex vacuo). cerebral atrophy.

517

517 Coronal section of the brain of a patient with Alzheimer’s disease


showing considerable atrophy of the brain and dilatation of the
ventricles due to the severe brain atrophy (hydrocephalus ex vacuo).
408 Degenerative Diseases of the Nervous System

Microscopic which project to the cortex and hippocampus. A family of


Argyrophilic senile (neuritic) plaques (518–521) neurotrophic factors (small proteins), of which the
• Plaques are large extracellular lesions in which the archetypal neurotrophin is nerve growth factor (NGF), are
principal component of the core of the plaque is the important (protective) for the survival of cholinergic cells.
insoluble 40–42/43 amino acid peptide called β-amyloid • Diminution of monoaminergic neurons and
protein which is a minor breakdown product of amyloid noradrenergic, GABAergic and serotonergic functions
precursor protein (APP). occur in affected neocortex.
• Plaques result from the accumulation of several proteins • Decreased neuropeptide transmitters: substance P,
and an inflammatory reaction around deposits of somatostatin, cholecystokinin.
β-amyloid.
• Degenerating nerve terminals in the plaques also contain ETIOLOGY
the microtubular protein tau. • Most cases are probably polygenic (caused by the action
• Amyloid burden and the number of plaques do not of several different genes), and the penetrance of disease
correlate with the severity of the disease. is influenced by age and environmental factors (e.g.
possibly herpes simplex virus type 1 infection in the brain).
Neurofibrillary tangles (522–524) • The concordance rate is higher (1.2–2.7) in mono-
• Tangles are intracellular lesions principally composed of zygotic (MZ) and dizygotic twin pairs, suggesting the
aggregations of an abnormal form of a cytoskeletal- contribution of genetic factors, but MZ twins are not
associated microtubular protein, tau, which is hyperphos- fully concordant, implicating non-genetic and environ-
phorylated. The aggregated neurofibrils are visualized as mental factors are involved.
paired helical filaments on electron microscopy. • Genetic factors appear to lead to quantitatively worse
• They are present, together with plaques, in the disease but not to a qualitatively different pattern of brain
projection neurons of the limbic and association areas of involvement. β-amyloid deposition seems to be the
the cerebral cortex, particularly the parietal cortex and neuropathologic factor most strongly influenced by
hippocampus, especially the CA1 zone and the genetic factors.
entorhinal cortex, subiculum, and transitional cortex of
the hippocampus.
• The number of tangles increases with the duration and 518
severity of disease.
• Tangles are not specific for AD however, they are also
found in other conditions such as dementia pugilistica,
subacute sclerosing panencephalitis, and tuberous sclerosis.

Dystrophic neurites

Loss of neocortical neurons (>40%)


Loss is widespread (hippocampus, entorhinal cortex,
association areas of neocortex, and nucleus basalis of
Meynert [the substantia innominata] and locus ceruleus)
and predominantly a loss of cholinergic, noradrenergic, and
dopaminergic neurons. N.B. There is no loss of neocortical
neurons in the course of normal ageing.
518 Microscopic section of cerebral cortex, cresyl violet stain, showing
Loss of neuronal synapses neuronal loss and an extracellular senile or ‘neuritic’ plaque containing a
• Assessed using antibodies to synaptic proteins such as homogeneous central core of amyloid (arrow).
synaptophysin.
• The degree of synaptic loss is the best correlate with the
severity of dementia. 519
N.B. It is not known if senile plaques or neurofibrillary
tangles come first, or which is the primary factor causing
AD, but as these cellular changes progress, neurons are lost.

Associated pathologic states (525)


• Concomitant Parkinson’s disease changes (nigral
degeneration and Lewy bodies at various sites) are
present in about 20–30% of AD autopsies.
• Diffuse Lewy body disease (see p.430).
• ‘Punch-drunk’ syndrome, or ‘dementia pugilistica’:
neurofibrillary changes in boxers.

Chemical pathology
• A cholinergic deficit secondary to degeneration of 519 Microscopic section of cerebral cortex, Cajal stain, showing two
subcortical neurons (e.g. in the basal nucleus of Meynert) senile or ‘neuritic’ plaques (arrows).
Alzheimer’s Disease (AD) 409

520 521

520 Microscopic section of cerebral cortex, silver stain, showing an 521 Amyloid in the walls of small blood vessels near senile plaques
extracellular senile or ‘neuritic’ plaque as a deposit of amorphous (amyloid or congophilic angiopathy).
material that contains a central core of amyloid, surrounded by
numerous short fibrils (resembling a bird’s nest) that represent
products of degenerated nerve terminals, mainly dendritic, containing
lysosomes, abnormal mitochondria, and often twisted tubules.

522 523

524 525

522–524 Microscopic section of brain showing neurofibrillary tangles 525 Microscopic section of brain showing granulovacuolar
as thick, fiber-like strands of silver-staining material, often in the form of degeneration of neurons in the pyramidal layer of the hippocampus.
loops, coils or tangled masses, in the nerve cell cytoplasm.
410 Degenerative Diseases of the Nervous System

Sporadic (most cases of AD) – ApoE is produced predominantly in astrocytes and is


Negative family history (50–75% of cases) carried by the low-density lipoprotein receptor into
• Associations with advanced maternal age, head trauma, neurons, where it binds to neurofibrillary tangles.
history of depression, and the coexistence of herpes – ApoE exists in three major isoforms (E2, E3, and E4)
simplex virus type 1 in the brain and carriage of an encoded by three alleles (ε2, ε3 and ε4) of the ApoE gene.
apolipoprotein E (ApoE) ε4 allele. The three variants of ApoE vary in their affinity for β-
• Postmenopausal estrogen replacement therapy has been amyloid. The ApoE4 variant increases the rate of
associated with reduced risk and delayed onset of AD in deposition of fibrillar β-amyloid, resulting in the β-pleated
elderly women. Estrogen is linked with preservation of sheets that stain with birefringent stains for amyloid.
cholinergic neurons, increased secretase metabolism of – In the general population, 75–85% have the ε3 allele,
the APP and interaction with ApoE. 10–15% have ε4, and 5–10% have ε2. In AD, ε4 is over-
• The above associations have not been established as represented (40% [32–58%]) and ε2 is under-represented
causal, however (see Risk factors, below). (4%).
– Up to 30–40% of the risk for late-onset AD is
Positive family history in one or more first-degree attributable to alleles at the ApoE locus.
relatives (25–50% of cases) – The ε4 allele is associated with a threefold
• ApoE gene on chromosome 19q encodes ApoE. (heterozygotes) to eightfold (homozygotes) increased
Although no mutations have been found in ApoE, one risk of sporadic and familial late-onset AD. However,
of its three genetic variants, ε4, substantially increases the even among homozygotes for ε4, only about 50%
risk of AD. This is, therefore, a risk factor or develop dementia by age 90 years.
‘susceptibility’ gene (see below) rather than a causative – The ε2 allele may be protective.
gene. The onset of AD is 10–20 years earlier in – Although ApoE4 is an important risk factor for AD, it is
individuals who are homozygous for the ApoEε4 allele neither essential for the development of the disease, nor
than in those with the ε2 or ε3 alleles, and it is 5–10 specific for the disease. Hence, testing for ApoE
years earlier in individuals who are heterozygous for ε4. genotype is not advised.
• Presenilin-1 (PS-1) gene on chromosome 14: about 5% • Down’s syndrome.
(and up to 20%) of AD in the white population may be
attributed to homozygosity at this locus. Possible
• Ethnic group: AD could be less common in some non-
Familial variants of AD (rare) white groups (Asians, Black Africans, Native Americans)
Autosomal dominant inheritance of mutations in one of the due to unknown genetic or environmental differences.
three known causative genes; tends to present clinically at a • Head injury: neuronal injury may trigger the deposition
younger age (40–60 years): of β-amyloid.
• Amyloid precursor protein gene on the long arm of • Lower educational attainment and socioeconomic status.
chromosome 21 which encodes APP (<1% of all early- • Herpes simplex virus type 1 infection of the brain.
onset cases). • Aluminum in drinking water: aluminum is neurotoxic
• PS-1 gene on chromosome 14q (50% of early-onset and may been found in the brain in people with AD, but
cases): >50 mutations in the gene for PS-1 have been the aluminum hypothesis remains controversial.
identified in patients with early-onset AD. • Susceptibility loci on chromosomes 9 and 10. The locus
• Presenilin-2 (PS-2) gene on chromosome 1 (15–20% of on chromosome 10 probably modifies risk for AD by
cases). modulating long β-amyloid-42 levels.
• High blood pressure and other vascular risk factors.
Mutations in all these genes result in a shift in the • High plasma homocysteine.
metabolism of APP to more of a 42–43 amino acid form of
β-amyloid (‘long Aβ’), as distinguished from the more Possible protective factors
prevalent species of β-amyloid that has just 40 residues • Anti-inflammatory drugs: AD may involve inflammatory
(‘short Aβ’). mechanisms.
• Estrogen replacement therapy.
Genetic classification of Alzheimer’s disease (Table 39) • Education and premorbid intelligence: greater cognitive
reserve may allow better compensation for disease pathology.
Down’s syndrome
An extra chromosome 21 (trisomy 21) and thus extra copy PATHOGENESIS
of the β-amyloid precursor gene on chromosome 21 leads The amyloid hypothesis
to β-amyloid deposition and the brain changes of AD before The β-amyloid gene encodes a large protein, APP, which is a
age 40, but not dementia until much older. transmembrane protein, with the β-amyloid motif extending
from the exterior to half-way through the cell membrane. The
Risk factors for AD APP gives rise to β-amyloid, a fragment of 40–42 amino acids.
Definite There are at least three different forms of β-amyloid, depending
• Old age (accumulation of β-amyloid in the brain with age). on the site of RNA splicing. Seven different mutations in the
• Family history of dementia (having a first-degree relative gene for the APP have been found, all of which increase the
with AD increases the risk around 3.5 times, perhaps production of β-amyloid 1–42 (β-A4) leading to fibrillar
because vulnerability genes are shared by family members). aggregation toxic to neurons. The amyloid hypothesis is that
• ApoE allele status. The ApoE gene may be involved in mutations in the APP and presenilin genes are associated with
neuronal repair: increased cellular production of β-A4, which is toxic to
Alzheimer’s Disease (AD) 411

neurons. The ApoE ε4 allele does not increase production mislay items, get lost when alone, or fail to recognize people
of β-A4, but promotes an increase in the rate of β-amyloid (i.e. they may have anosognosia). Changes in family roles are
deposition as compared with Apoε3. AD may be a restricted also important clues; a person who normally deals with family
form of cerebral amyloidosis, in which β-amyloid fibril finances for example may have recently had to abrogate the
deposition is essential, but not necessarily the sole cause, and responsibility because of failing attention and memory.
that neurofibrillary tangles are caused by damage from Psychologic, medical, psychiatric, neurologic and social
aggregated β-amyloid. factors that may contribute to a patient’s intellectual decline
should also be evaluated. In many cases it is quite clear from
CLINICAL FEATURES the history and examination that dementia is present, but
• Insidious onset. in others a more formal assessment is necessary by means of
• Memory decline/forgetfulness: a formal neuropsychologic evaluation.
– Defects in learning/encoding new verbal and visual-
spatial information are apparent. Impaired antegrade DIFFERENTIAL DIAGNOSIS
episodic memory is the earliest neuropsychologic deficit, Dementia
and is due to plaques and tangles in the transentorhinal • Alzheimer’s disease: 50–55%.
region, which deafferent the hippocampal complex. • Vascular dementia (see p.261): 15–20%.
Elaborate encoding strategies are ineffective whereas they • Diffuse Lewy body disease (see p.430): 15–25%.
substantially improve learning in normal elderly people. • Parkinson’s disease (see p.420): 5–10%.
– Defects in retrieving information with minimal cueing is • Brain injury: alcohol, head trauma: 5%.
present (episodic or autobiographical memory is pre- • Other causes: 5%:
dominantly affected, with early loss of memory for – Normal pressure hydrocephalus (see p.473).
everyday events). – Intracranial mass lesion: frontal or temporal lobe tumor,
– There is relative preservation of immediate short term chronic subdural hematoma.
memory (e.g. digit span) early on. – Metabolic/toxic: chronic drug intoxication (e.g. alcohol,
• Focal ‘cortical’ neurologic signs such as dysphasia, barbiturates, sedatives), chronic hepatic encephalopathy.
dyscalculia, dysgraphia (dysphasic or dyspraxic), visual- – Endocrine: hypothyroidism, Cushing’s syndrome.
spatial-perceptual dysfunction, ideomotor and dressing – Autoimmune: SLE.
dyspraxia, agnosia for objects or faces, and sensory inat- – Nutritional: vitamin B12 deficiency (see p.463);
tention occasionally mark the onset of the disease. Other- Wernicke– Korsakoff syndrome (see p.460).
wise, focal cortical dysfunction becomes apparent later. – Syphilis (general paresis of the insane) (see p.312); HIV
• Paranoia and other personality or behavioral changes may (see p.301).
be present. – Pick’s disease and other frontal lobe dementias (see
• Myoclonic jerks and occasionally seizures occur in some p.414).
patients. – Huntington’s disease (see p.417): subcortical dementia.
• Muscular rigidity and signs of corticospinal tract – Progressive supranuclear palsy (see p.432): subcortical
dysfunction may occur in the late stages. dementia.
• Gait disorder: short-stepped, slower gait and poor – Creutzfeldt–Jakob disease (see p.330): myoclonus and
balance often appear late in the disorder. rapidly progressive dementia.
• Primitive reflexes and other frontal release signs. • Multiple causes (i.e. combinations of the above): 10–15%.
• Urinary incontinence.
• Akinesia and mutism.

The Mini-Mental State Exam


(MMSE) evaluates a spectrum of Table 39 Genetic classification of Alzheimer’s disease
cognitive functions but is only a Type Chromosome Gene Age at onset % of cases
screening test and not necessarily
diagnostic, since an individual with a Early-onset familial AD 21q21 APP 45–64 <0.1% (<1% of
previous high level of intellectual early-onset cases)
ability may be demented but function Early-onset familial AD 14q24.3 S182 40s mean 1–2% (50% of early
at a level above that used as a cut-off PS-1 (28–62) -onset familial AD)
on the MMSE scale. Similarly, a score Early-onset familial AD 1q31-q42 STM2 50s mean <0.1% (15–20% of
within the range that is usually indica- PS-2 (42–68+) early-onset cases)
tive of dementia may be caused by
factors such as dysphasia or depression. Late-onset 12 α2-macro >65 30%
The MMSE should be combined with Late-onset familial AD 19q13 ApoE >60 40% (32–58%)
complementary tools such as the and sporadic; ApoE ε4
Functional assessment staging test allele associated
(FAST) which consists of seven stages Late-onset familial; ? ? >75 10–40%
incorporating 16 substages. and sporadic; not
The spouse, family and friends ApoE allele associated
should be interviewed. Patients with
APP: amyloid precursor protein; α2-macro: alpha2-macroglobulin mutation; ApoE:
dementia may be unaware of difficulties
apolipoprotein E; PS-1: presenilin-1; PS-2: presenilin-2
at work and any tendency to be for-
getful of appointments and names,
412 Degenerative Diseases of the Nervous System

Familial dementia • The most relevant issue is predicting the development of


• Huntington’s disease. disease in cognitively unimpaired relatives of a proband.
• Familial prion diseases. Detection of one of the known APP, PS-1, or PS-2 muta-
• Hereditary cerebral hemorrhages with amyloidosis, tions in an unaffected relative predicts development of the
Dutch type. disease with almost 100% probability, but the accuracy of
• Chromosome 17- and chromosome 3-linked forms of the age of onset is less certain; it is within 2.5 SD from
frontal lobe dementia. the mean age of onset for the family with APP mutations.
• Cerebral autosomal dominant arteriopathy with
subcortical infarcts and leukoencephalopathy (CADASIL). Late-onset AD with a positive family history
A routine search for the known mutations seems limited.
Pseudo-dementia
• Depression: may cause a pseudodementia and may also Sporadic AD
commonly accompany the earlier stages of AD. In • The ε4 allele of the ApoE gene is more common in
general, patients with cognitive impairment due to AD patients with sporadic late-onset AD (40%) than in
underestimate the severity of their cognitive deficit, in elderly controls (10–15%).
contrast to those with anxiety or depression. • ApoE genotyping should be used only as a diagnostic test
• Drug intoxications: anticholinergics. in patients who are clinically diagnosed with dementia and
• Age-related cognitive impairment in ‘normal’ aged persons. the cause of the dementia is unknown, but is probably AD.
Among people over 60 years of age who have dementia, the
INVESTIGATIONS pre-test probability (or prevalence) of AD is about 66%.
• Full blood count and film. Additional diagnostic tests aim to identify other causes of
• Thyroid function tests. dementia and improve the post-test probability of AD; a
• VDRL/RPR, TPHA. cranial CT scan might identify a tumor, a B12 test may
• Serum vitamin B12. disclose vitamin B12 deficiency, and an ApoE4/E4 genotype
• Antinuclear antibodies. increases the probability that the correct diagnosis is AD to
• HIV serology. about 94%. If the patient has ApoE3/E4, the probability
• Cranial CT or MRI scan: increases from about 66% to 81%. If the patient has
– The main findings are diffuse cortical atrophy (most ApoE2/E3, the probability of AD is reduced by half; i.e.
prominent in the temporal lobes) and large ventricles, there is double the possibility that there may be another
sulci and fissures (due to loss of brain substance). cause for the dementia, particularly if the age of onset is
– Atrophy is more prominent and rapid than in age- 50–70 years, since the age of onset of AD is later with the
matched normal controls. The degree of temporal ApoEε2 allele. Detecting an ε4 allele in a demented patient
atrophy can be measured on CT (526) or MR, but the may therefore increase the likelihood of the diagnosis of AD.
findings are non-specific. • ApoE genotyping is not useful as a predictive test for AD
– The main reason for cross-sectional imaging is to exclude in an asymptomatic individual: the population at highest
a treatable cause of dementia (527). risk of developing AD (ApoEε4/ε4 genotype) is only 2%
• PET or SPECT scan shows reduced cerebral blood flow in of the normal population and, even for this small group,
the posterior temporoparietal regions. PET may also demon- the period of risk extends over five decades (50s–90s)
strate reduced cerebral oxygen utilization in these areas. and some may not develop AD at all.
• EEG: normal or shows generalized slow wave activity.
• CSF: DIAGNOSIS
– Cells: normal. • During life, the diagnosis is largely one of exclusion of
– Tau levels: increased. other causes of progressive dementia. A diagnosis of
– β-A4 levels: reduced, possibly because the β-A4 is probable AD can be made if dementia is manifest by
deposited in plaques rather than remaining in solution. deficits in at least two areas of cognitive ability, including
High levels indicate that AD is not likely. memory, in a person showing progressive deterioration,
• Tropicamide eye drops: hypersensitivity of the pupillary whose activities of daily living are also significantly
response in AD to a solution of dilute topical tropica- impaired, and in whom there is no evidence of an
mide has been reported and awaits further evaluation. alternative cause for the symptoms. The probability is
strengthened if the pattern of cognitive deficit is typical
Genetic testing of AD: early amnesia followed by apraxia and aphasia and
• In the individual with cognitive impairment, genetic so on. The accuracy of such a diagnostic approach is
testing can be used to increase diagnostic accuracy and about 80–90%. The addition of neuroimaging and
detect transmissibility. SPECT scanning may improve this.
• In the individual without cognitive impairment, genetic • The histologic diagnosis relies on quantitative and
testing can be used to predict disease development and qualitative features; both of the commonly used
the approximate age of onset. diagnostic criteria depend on a plaque count.
• The overall very low prevalence of genetic defects in AD
militates against indiscriminate screening for APP, PS-1 TREATMENT
and PS-2 mutations in clinical practice. At present, treatment is symptomatic and not curative.

Early-onset familial (autosomal dominant) AD Dementia


• Transmissibility of the disease is often clear from a very • Life history booklet (memory book).
strong family history in patients with early-onset dementia. • Instruction tapes (memory tape).
Alzheimer’s Disease (AD) 413

Acetylcholinesterase inhibitors alleviate some of these adverse effects. Use with caution in
• Acetylcholinesterase inhibitors (donepezil, rivastigmine and patients with supraventricular conduction abnormalities,
galantamine) are used as a specific symptomatic treatment peptic ulcers and obstructive airways disease.
of cognition and behavior in mild to moderate dementia of • Rivastigmine: a pseudo-irreversible acetyl- and butyryl-
Alzheimer’s type, rather than dementia in general. cholinesterase inhibitor which is selective for the CNS and
• They inhibit acetylcholinesterase and thus decrease has regional selectivity within the brain for the cortex and
acetylcholine breakdown in the synaptic cleft. hippocampus. Treating 8 patients with rivastigmine (6–12
• They do not affect the underlying disease process that mg/day) produces a four-point improvement on the
causes loss of neurons and synapses and leads to ADAS-Cog scale in one patient, and significant improve-
intellectual and functional deterioration. ment in global outcome and function. Dosage: initially 1.5
• All have broadly similar efficacy, equivalent to about mg orally twice daily for a minimun of 2 weeks, increasing
6–12 months delay in the course of the disease after by 1.5 mg twice daily every 4 weeks if well tolerated.
30 weeks treatment in about two-thirds of patients with Maintain patient on highest well-tolerated dose up to a
mild to moderate AD. maximum of 6 mg twice daily. Adverse effects include
• Tacrine (tetrahydroaminoacridine): the first drug nausea (47%), vomiting (31%), diarrhea (19%), headaches
approved for the treatment of AD in the USA in 1993, (17%), dizziness (21%), and abdominal pain (13%) with
but withdrawn in 2000 because of poor gastrointestinal 6–12 mg/day. These are usually transient and minimized
tolerance (peripheral cholinergic symptoms [nausea, by gradual titration and taking the drug with food.
vomiting, diarrhea] and hepatotoxicity [rises in serum • Galantamine: reversibly and competitively inhibits
transaminases]). acetylcholinesterase and enhances the response of nicotinic
• Donepezil: approved in the USA and UK in 1997, and the receptors to acetylcholine. Clinical trials have shown that
first drug to be licensed in the UK for AD. Easily galantamine in maintenance doses of 24 or 32 mg per day
administered. Treating 4–6 patients with donepezil significantly improves cognitive function and slows the
10 mg/day results in a four-point improvement on the progression of functional decline over 6 months relative to
ADAS-Cog scale in one patient, and benefits in functional placebo. Tablet sizes of galantamine are 4 mg and 8 mg,
preservation. Donepezil has a favorable adverse effect with a liquid preparation (4 mg/ml) which is useful for
profile, devoid of hepatotoxicity and with improved titration. Dose can be titrated from 4 mg twice daily to
gastrointestinal tolerance, and simplified compliance, 8 mg twice daily over 4 weeks according to tolerance and
prescribing and monitoring. Begin at a dose of 5 mg once benefit, but start with 4 mg daily in patients with hepatic
daily (clinically effective dose), taken at night (unless impairment. Galantamine is contraindicated in patients with
insomnia), for the first 4–6 weeks. The dose can be severe renal impairment, but no dose adjustment is required
increased to 10 mg once daily if the lower dose is well for mild to moderate renal impairment (creatinine clearance
tolerated. Nausea (17% of patients), and diarrhea (17%), rate >9 ml/min). It is well tolerated. With 8 mg daily, most
and vomiting (10%) are the most common adverse effects. adverse effects occur in the first 4 weeks: nausea (6%),
They are predictable (i.e. cholinergic) and generally vomiting (4%), diarrhea (5%), anorexia (6%), agitation (15%.
transient, occurring on initiation or up-titration of the • If used, cholinesterase inhibitors should be given within
drug. Other symptoms may include headache, fatigue the first 5 years of the disease, while there are more
(8%), insomnia, dizziness (8%), muscle cramps (8%), remaining cholinergic neurons, and while the patient is
agitation, hallucinations, unpleasant dreams, and urinary still functioning and independent.
urgency. Dose reduction or morning administration may

526 CT brain scan of 526 527


patient aged 64 with
dementia attributed to
Alzheimer’s disease.The scan
has been done to
demonstrate the temporal
lobes preferentially by angling
the gantry along the long axis
of the temporal lobes, hence
the odd appearance. Note
the very atrophied medial
temporal lobes (arrows).

527 CT scan at the level of


the lateral ventricles in the
same patient.There is
atrophy, but not much in the
way of vascular disease,
therefore the likely diagnosis
is Alzheimer’s disease.
414 Degenerative Diseases of the Nervous System

Possible neuroprotective agents CLINICAL COURSE AND PROGNOSIS


• Selegiline (10 mg/day): a monoamine oxidase B • When the patient first presents they often have only a
inhibitor that also facilitates catecholaminergic activity. relatively circumscribed cognitive impairment, typically
• Alpha-tocopherol (vitamin E, 2000 IU a day): an a memory deficit. As the disease progresses, cognitive
antioxidant that traps oxygen free radicals. It may be impairment becomes generalized.
protective against AD. Adverse effects (diarrhea) are • Average survival after diagnosis is 5–10 years.
infrequent. Vitamin E may interact with warfarin and
lead to bleeding problems.
• Lamotrigine 300 mg/day.
• Estrogen.
• Folic acid. FRONTOTEMPORAL DEMENTIA
• Ginkgo biloba. ASSOCIATED WITH MUTATION IN
TAU (PICK’S DISEASE, LOBAR
Possible predictors of response to drug treatment ATROPHY)
ApoE genotyping.

Depression DEFINITION
• Can coexist with AD and should be treated appropriately Frontotemporal dementia (FTD) is a term used to describe
(i.e. supportive counselling and, if necessary, antidepres- patients with one of three major clinical syndromes (frontal
sant drug therapy). variant FTD [dementia of frontal type], semantic dementia
• Selective serotonin reuptake inhibitors (SSRIs) are a [progressive fluent aphasia], and progressive nonfluent apha-
good choice because of their relatively short half-lives, sia), progressive focal atrophy of the frontal and/or tempo-
and minimal anticholinergic, adrenergic and histaminic ral lobes, and, in some patients, mutations in the gene for
adverse effects; tricyclic antidepressants often aggravate the microtubule binding protein tau, and characteristic tau-
the situation due to their anticholinergic action. positive inclusions in affected neurons.
Pick’s disease is one cause of localized cerebral atrophy, or
Behavioral disturbance ‘lobar atrophy’ and frontotemporal dementia. Pick’s disease
• Identify the specific problem behavior, document relevant was a nosologic entity or syndrome complex, first described
antecedents and consequences, and search for any medical by Arnold Pick of Prague in 1892, characterized clinically by
illness, physical symptoms or iatrogenic factors (drug dementia, personality changes, speech disturbances,
adverse effects or interactions) which may be contributory. inattentiveness, and occasionally also extrapyramidal
• Pharmacologic treatment of behavioral symptoms should phenomena, and pathologically by circumscribed atrophy of
be reserved for drug-responsive symptoms that are causing the gray and white matter of the frontal and/or temporal
at least moderate distress to the patient or caregivers. lobes predominantly; but any region of the brain may be
• Thioridazine or fluoxetine, beginning in low dose and involved. Later, Alzheimer (1911) and Altman (1923) drew
increased slowly, may be effective. Haloperidol is a potent attention to the underlying pathology of ‘ballooned cells’
antipsychotic with minimal anticholinergic action in low (Pick cells), neuronal argyrophilic inclusions (Pick bodies) and
dose (0.5 mg once or twice daily). the absence of fibrillary tangles and senile plaques.
• Newer antipsychotic drugs such as risperidone and
olanzapine appear to be at least as effective as EPIDEMIOLOGY
conventional neuroleptics but with fewer undesirable • Incidence: not uncommon, but frontotemporal
adverse effects, particularly on the extrapyramidal system. dementias account for up to 10% of all cases of dementia,
and even more in the presenium.
Prevention • Age: middle-aged and elderly; usually mid-50s.
• Recent advances in understanding AD in cases with a • Gender: F=M.
more straightforward pathogenesis, such as Down’s
syndrome or APP gene mutations, give rise to the hope PATHOLOGY
that rationally based preventive therapy aimed at A primary tauopathy.
reducing β-A4 production or aggregation into amyloid
may one day replace current symptomatic treatments, Macroscopic (528, 529)
which are of minor and probably temporary efficacy. • Atrophy:
• Vitamin E may be protective against AD, and therapy – Frontal and/or temporal lobes predominantly, caused by
with 1000 IU twice daily may be considered. neuronal loss in the cerebral cortex and amygdala, with
• Estrogen replacement therapy may reduce the risk and less marked changes in the hippocampus.
delay the onset of AD in postmenopausal women, but – Parietal lobes less frequently affected.
this was not found in a recent randomized trial. – Gyri of affected lobes: paper thin (thinned cortical
• There is also insufficient evidence to support the use of ribbon), grayish appearance.
antioxidant agents (other than vitamin E perhaps), anti- – White matter of affected lobes.
inflammatory agents, monoamine oxidase B inhibitors, – Corpus callosum.
folic acid, or antihypertensive drugs. – Anterior commissure.
• Antiviral and anti-inflammatory agents to prevent viral – Caudate nucleus.
reactivation, and the use of vaccines against herpes simplex – Thalamus, subthalamic nucleus, substantia nigra, and
virus type 1 in infancy, may also have promise if an globus pallidus sometimes.
infective component to the etiology of AD is confirmed. • Enlarged ventricles.
Frontotemporal Dementia Associated with Mutation in Tau (Pick’s Disease, Lobar Atrophy) 415

• Thickened overlying pia-arachnoid. There are three common clinical presentations: (1) Frontal
• Relative sparing of the pre- and postcentral, superior variant FTD (dementia of the frontal type) – a frontal
temporal, and occipital convolutions. dysexecutive syndrome with social conduct disorders and
disinhibition. There is orbitobasal frontal atrophy. (2)
Microscopic (530, 531) Temporal lobe variant FTD (semantic dementia) – a
• Loss of neurons, most marked in the first three cortical progressive fluent aphasia with impairment of semantic
layers. verbal memory. There is asymmetric anterolateral temporal
• Loss of medullated fibers in white matter beneath atrophy with relative sparing of the hippocampal formation,
atrophic cortex (probably due to neuronal loss). typically worse on the left side. (3) Progressive nonfluent
• Astrocytic gliosis of cortex and subcortical white matter. aphasia – there is left peri-sylvian atrophy.
• Mild granulovacuolar degeneration of neurons in the
hippocampus (525). 528
• The tau protein is the major component of
neurofibrillary tangles and other pathologic features of
frontotemporal dementia; tau positive inclusions are
found in neurons and glial cells. The different tau
isoforms are deposited in characteristic patterns in
different diseases (e.g. the tau deposition of AD includes
all 6 isoforms, whereas the tau deposited in Pick’s disease
consists of only 3-repeat tau isoforms.
• Pick cells: swollen (‘ballooning’), achromatic surviving
neurons of frontal cortex (531).
• Pick bodies: argyrophilic straight fibrils within the
cytoplasm of some surviving neurons, predominantly in
the cortex of the medial temporal lobes, and particularly
in atrophic hippocampi. They are both tau- and 529
ubiquitin-positive.
• Senile plaques are not present.

ETIOLOGY AND PATHOPHYSIOLOGY


• Usually sporadic and of unknown etiology.
• A subgroup of these disorders are familial, due to mutations
in the tau gene. They are probably transmitted as an
autosomal dominant trait with polygenic modification.

CLINICAL FEATURES
• Gradual onset, frequently before the age of 65 years.

528 Lateral view of the brain showing severe circumscribed atrophy


of the left frontal and temporal lobes.

529 Asymmetric section through the brain in the axial plane at the
level of the thalami showing atrophy of the frontal lobes.

530, 531 Histologic section of brain showing loss of neurons,


astrocytic gliosis and swollen (‘ballooning’) surviving neurons.

530 531
416 Degenerative Diseases of the Nervous System

Frontal lobe dysfunction • Creutzfeldt–Jakob disease: a rapidly progressive dementia


• Abulia. that may be indistinguishable from that seen in Pick’s
• Apathy. disease.
• Behavioral change: disinhibited and inappropriate, • Gerstmann–Straussler–Scheinker syndrome: dementia
stereotyped and perseverative behavior. and cerebellar ataxia or extrapyramidal dysfunction.
• Personality change: varying from emotionally dull, inert, • Corticobasal degeneration: language disturbances and
lacking initiative, spontaneity and impulse, and taciturn, to frontal lobe-type behavior may be seen but there is
emotional, social and sexual indifference or disinhibition. usually evidence of limb dystonia, ideomotor dyspraxia,
• Inattention. myoclonus, and anasymmetric akinetic-rigid syndrome
• Loss of judgement and insight. with late-onset of gait or balance disturbances.
• Difficulty planning. • Depression: the profound apathy of frontal lobe dementia
• Variable memory loss. may be erroneously attributed to depression. However,
• Motor perseveration. the absence of neurovegetative symptoms and depressive
• Speech reduction: decreased fluency followed by ideation (e.g. guilt, worthlessness) should be clear.
echolalia and mutism. • Mania: manic symptoms are reported in about one-third
• Hyperorality. of cases.
• Prominent grasp and suck reflexes. • Schizophrenia: persecutory ideas and bizarre, hypo-
• Unable to perform sequences of motor tasks. chondriacal delusions may occur in some cases and
• Unable to cope with unaccustomed problems. suggest schizophrenia.
• Deterioration of social and work habits. • Obsessive compulsive disorder: compulsive hoarding and
• Gait impairment. ritualized routines occur in up to one-quarter of cases of
Pick’s disease, but primary obsessive compulsive disorder
Temporal lobe dysfunction usually occurs in young people.
• Dysphasia: receptive (slow comprehension), jargon • Primary progressive aphasia:
aphasia, dysnomia. – Linguistic disturbance varying from agrammatic, non-
• Forgetfulness. fluent speech to fluent aphasia with comprehension
• Disorientation in time and place. deficits, due to focal cerebral atrophy in the perisylvian
• Behavioral change: lighthearted, anxious or happy, region of the dominant hemisphere.
physically active and on the move constantly, talkative, – Social conduct, judgement, insight and memory are
occupied with trivia, and attentive to all passing incidents. preserved, as well as the capacity to develop strategies to
• Bulimia. circumvent any impairments.
• Altered sexual behavior. – It is uncertain whether this disorder is a prodrome of
• Apraxia involving articulatory, buccofacial, limb and AD, a discrete disorder without progression to global
truncal movements. dementia, or part of a spectrum involving both. Long
term follow-up studies are required.
Exceptionally
• Cerebellar ataxia. INVESTIGATIONS
• Extrapyramidal syndrome: shuffling gait, rigidity, • Neuropsychologic evaluation: frontal or temporal lobe
pseudocontractures of limbs. dysfunction.
• CT or MRI brain: striking atrophy of the cortex and
DIFFERENTIAL DIAGNOSIS white matter of the frontal and/or temporal lobes.
• AD: • Dynamic/ functional neuroimaging, such as SPECT and
– May coexist with Pick’s disease. PET, is often more informative than static techniques
– Frontal lobe presentations of AD (e.g. behavioral such as CT and MRI, and shows an anterior perfusion
disturbance) are unusual, particularly before 65 years. deficit, in contrast to AD which usually shows posterior
– Atrophy is relatively mild and diffuse. perfusion deficits.
– Paired helical filaments, not straight fibrils of Pick bodies. • EEG: normal.
– More severe granulovacuolar degeneration of neurons.
• Vascular dementia: e.g. Binswanger’s disease: DIAGNOSIS
– May share clinical features with frontal lobe dementia • Neuropathologic.
syndromes, such as irritability, jocularity, hyperactivity, • The best clinical predictors for the early diagnosis include
and mood fluctuation. ‘frontal dementia’, early ‘cortical’ dementia with severe
– Neurologic signs are common. frontal lobe disturbances, absence of apraxia, and absence
– CT scan usually reveals diffuse or multifocal white matter of gait disturbance at onset.
disease.
• Dementia with Lewy bodies (see p.430). TREATMENT
• Alcoholic brain damage: No specific treatment.
– May affect the frontal lobes.
– Patients with Pick’s disease may also abuse alcohol, CLINICAL COURSE
however. Progressive.
• Huntington’s disease:
– Involuntary choreiform movements are usually, but not PROGNOSIS
always, present. The average time from diagnosis to death is about 8 years,
– Positive family history. i.e. longer than in AD.
Huntington’s Disease (HD) 417

HUNTINGTON’S DISEASE (HD) • Ventricular dilatation, most marked in the frontal horns
of the lateral ventricles.

DEFINITION Microscopic (534, 535)


An autosomal dominant inherited disorder characterized by • Loss of neurons occurs first in the striatum (putamen and
progressive involuntary choreiform movements, cognitive caudate nucleus), and later to a lesser extent throughout
decline, and emotional and psychiatric disturbance, due to the brain in most of the gray matter of the cerebral
severe neuronal loss, initially in the neostriatum and later in hemispheres and cerebellum.
the cerebral cortex, as a result of an increase in the number • Neuronal loss is predominantly of medium sized spiny
of trinucleotide CAG repeats in the HD gene on neurons which make up 80% of the neurons in the
chromosome 4p16.3 that encodes the protein huntingtin. striatum; larger neurons are spared.
• Neurons in the striatum and cerebral cortex have
EPIDEMIOLOGY intranuclear inclusions of huntingtin and ubiquitin.
• Prevalence: 1 per 10 000; all races. • Reactive gliosis occurs with prominent characteristic
• Age of onset: average: 35–42 years, but may start in astrocytes in the basal ganglia.
childhood (juvenile or Westphal variant) or old age. • Biochemically, GABA, acetylcholine, substance P, and dyno-
• Gender: M=F. phin are decreased in the striatum. The dopaminergic nigro-
striatal pathway and dopamine concentrations are preserved.
PATHOLOGY • An imbalance between GABA, acetylcholine, and dopa-
Macroscopic mine may account for the involuntary choreiform
• Atrophy of the caudate nuclei and frontal lobes (532, 533). movements.

532 533

532 Coronal section through the frontal lobe of one cerebral 533 Brain at post mortem of a patient with severe Huntington’s
hemisphere from a patient with advanced Huntington’s disease (left) disease showing atrophy of the caudate nuclei and frontal lobes
and a person with a normal brain (right) showing severe atrophy of bilaterally.
the caudate nucleus and frontal lobe with dilatation of the frontal horn
of the lateral ventricle of the brain of the patient with Huntington’s
disease on the left.

534 535

534, 535 Normal cerebral cortex (534) and cerebral cortex of frontal lobe in Huntington’s disease (535).
418 Degenerative Diseases of the Nervous System

ETIOLOGY AND PATHOPHYSIOLOGY Emotional/psychiatric disturbance


• Autosomal dominant inheritance with high penetrance. Many patients develop depression and suicidal thoughts.
• Gene defect: an abnormally expanded and unstable
polymorphic CAG trinucleotide repeat (37–121 copies; Oculomotor abnormalities
normal range 11–34, median 19) in the HD (initially • Fixational instability.
called IT-15) gene on the short arm of chromosome 4 in • Saccadic pursuit.
the 4p16.3 region. • Difficulty initiating saccades.
• The CAG repeat of the HD gene encodes a • Slow, hypometric saccades.
polyglutamine stretch proportional to the number of
triplets.The mutant protein product of the HD gene is Family history of HD
an elongated protein with 40–150 glutamine residues, Not always known or volunteered.
called huntingtin. It fails to show any homologue to
known proteins. The transcript is cytoplasmic and widely SPECIAL FORMS
expressed in all cell types, but is most severe in the Juvenile Westphal variant
striatum, hippocampus, cerebellar granular cell layer and • Onset in childhood and adolescence.
Purkinje cells of the brain. Translation of the mutant • Usually associated with paternally inherited mutations
huntingtin protein appears to be necessary for patho- with large numbers of trinucleotide repeats.
genesis. However, the brain pathology of HD cannot be • Rigidity develops in the trunk and proximal limb muscles
accounted for by the pattern of expression of huntingtin and spreads to involve all muscle groups.
and its function is not known. Theories include: • Progressive dementia, mask-like faces and bilateral
(1) Loss of function: unlikely with triplet repeats. increased reflexes and extensor plantar responses.
Dominant-negative effect: a mutated gene from one • Seizures.
chromosome interacts with subunits of a normal protein • Death within a few years of onset.
to inactivate it.
(2) Gain of function: the expansion allows a protein to DIFFERENTIAL DIAGNOSIS
do what it normally does but also a new additional and Chorea (see p.122)
pathologic function, possibly unrelated to the function Hereditary
of the normal protein. Huntingtin may induce defective • Autosomal dominant spinocerebellar ataxia.
mitochondrial function, resulting in impaired oxidative • Benign familial chorea: dominant inheritance, childhood-
phosphorylation and cell death by excitotoxicity. onset, not severe.
• The number of CAG repeats correlate inversely with the • Paroxysmal choreoathetosis.
age of onset of the illness, and directly with severity and, • Dentato-rubro-pallido-luysian atrophy (DRPLA).
to some extent, with neuropathology and clinical features. • Wilson’s disease.
• The biochemical basis of the disease is unknown but may • Neuroacanthocytosis.
be a defect in energy metabolism. In HD brain there is
evidence of glucose hypometabolism, increased lactate Other
concentrations, and a decrease in activities of • Systemic lupus erythematosus.
predominantly complexes II and III of the mitochondrial • Polycythemia rubra vera.
respiratory chain in the caudate nucleus. Apoptotic cell • Thyrotoxicosis.
death appears to occur. • Drugs: levodopa, amphetamine-like drugs, lithium,
carbamazepine, phenytoin.
CLINICAL FEATURES • Rheumatic (Sydenham’s) chorea.
About half of patients first present with a movement • Pregnancy.
disorder and the other half with an emotional, cognitive, • Oral contraceptives.
behavioral or psychiatric disturbance. • Anoxic encephalopathy.
Homozygous individuals are not phenotypically different • Post-infectious encephalomyelitis.
from heterozygous individuals. • Viral encephalitis.
• Stroke.
Involuntary, irregular, predominantly
choreiform, movement disorder Other movement disorders
• Initial symptoms: commonly a sense of restlessness or • Tardive dyskinesia: dopamine receptor-blocking drugs
being fidgety. (e.g. phenothiazines, neuroleptics).
• Chorea of the fingers and wrists, and later the more • Idiopathic orofacial dyskinesia.
proximal muscle groups of the upper limbs, the face and • Hemiballismus.
tongue (causing dysarthria and dysphagia) and the legs,
causing the gait to become unsteady and assume a bizarre, Dementia
bouncing quality due to irregular involuntary movements. • AD.
• Other movement disorders, such as dystonia and • Vascular dementia.
myoclonus, may occur. • Parkinson’s disease.
• Pick’s disease.
Slowly progressive subcortical dementia • Creutzfeldt–Jakob disease.
Initially manifests as forgetfulness, slowness of central process- • Vitamin B12 deficiency.
ing, diminished attention and concentration and reduced • Hypothyroidism.
insight and judgement. May not be prominent in the elderly. • Neurosyphilis.
Huntington’s Disease (HD) 419

• HIV infection. 536


• Non-convulsive status epilepticus.

Depression

INVESTIGATIONS
Predictive testing
• Refer to a specialist Huntington’s disease predictive
testing team for assessment and intervention by
neurologist, geneticist, psychiatrist, psychologist, and
social worker. The ‘patient’ being tested needs to have
no clinical features of HD (otherwise they are embarking
on a diagnostic test), to be psychologically equipped to
deal with the news of a positive or negative blood test
result, and to understand the implications of the result
on other members in the family. This also needs to be
appreciated and anticipated by the team.
• Blood DNA analysis: expanded CAG trinucleotide repeat
(>37 repeats) in the gene on chromosome 4p16.3.

Differential diagnosis
• Cranial CT or MRI scan: to exclude intracranial
structural lesion, diffuse cerebral atrophy or a multi- 537
infarct state. In moderate to severely affected HD
patients, there is flattening (atrophy) of the head of the
caudate nuclei, and atrophy of the frontal lobe with
widening of the cortical sulci anteriorly and
dilated/rounded frontal horns of the lateral ventricles
(536–538). Increased signal on T2W MRI has been
described in the basal ganglia.
• Full blood count and blood film looking for
acanthocytes.
• Thyroid function tests.
• VDRL/RPR, TPHA.
• Copper and ceruloplasmin.
• Antinuclear antibody.
• Blood DNA analysis: expanded CAG trinucleotide repeat
in a gene on chromosome 4p16.3 in HD.

DIAGNOSIS
Positive family history (if available), appropriate clinical
findings, and blood DNA analysis reveals expanded CAG
trinucleotide repeat in the gene on chromosome 4p16.3.

538

536 CT scan from a patient with very early symptoms of Huntington’s


disease.The small caudate nuclei are outlined in white on one side.
Note the prominent anterior horns of the lateral ventricles secondary
to the caudate atrophy, but the rest of the brain appears relatively
normal.

537 CT scan from an advanced case of Huntington’s disease. Note


the generalized cerebral atrophy and particularly small caudate nuclei
(outlined in white).

538 Cranial CT scan, axial plane, showing dilatation of the sulci of the
frontal lobes and of the frontal horns of the lateral ventricles due to
atrophy of the frontal lobes and caudate nuclei in a patient with
advanced Huntington’s disease.
420 Degenerative Diseases of the Nervous System

TREATMENT PARKINSON’S DISEASE (PD)


• No treatment has been shown to be effective in slowing
the course of the neurologic deterioration, although
there is a preliminary report of functional, motor and DEFINITION
cognitive impairment in five patients after human fetal A slowly progressive, age-related, degenerative disorder of
neural allografts into the right striatum, then after a year, the CNS, characterized clinically by tremor, bradykinesia,
the left striatum. rigidity, and disturbed postural reflexes (parkinsonism) and
• Coenzyme Q10 (ubidecarenone), an essential cofactor of pathologically by loss of dopaminergic cells in the pars
the electron transport chain, reduces cortical lactate compacta of the substantia nigra, with typical neuronal
concentrations if given orally in a dose of 360 mg/day inclusions known as Lewy bodies. It is named in honor of
for >2 months. If correction of lactate production has a James Parkinson who, in 1817 wrote a classic monograph
favorable effect on symptoms or disease progression, this An Essay on a Shaking Palsy.
may be a possible therapy. Ubiquinone (coenzyme Q) is
an integral part of the functioning respiratory chain. It EPIDEMIOLOGY
serves to shuttle electrons between complexes I and II • Incidence: 20 (95% CI: 12–27) per 100 000 per annum.
and complex III and functions as an antioxidant and as Incidence increases progressively with advancing age.
a respiratory chain activator. • Prevalence: prevalence of probable and possible PD
combined: 128 (95% CI: 109–150) per 100 000 popu-
General lation (crude prevalence), and 168 (95% CI: 142–195)
Information and support for patients and families about the per 100 000 population (age adjusted prevalence). It
disease and its implications. occurs in every racial and cultural group.
• Age-specific prevalence: 60–69 years: 342 per 100 000
Symptomatic population; 70–79 years: 961 per 100 000; ≥80 years:
Chorea 1265 per 100 000 population.
• Haloperidol: beginning with 0.5 mg twice daily and • Age: mean age of onset 60–65 years; onset <40 years of
increasing slowly until an adequate response or to 20 mg age in about 8% of patients.
per day is probably the most effective treatment. • Gender: M≥F: age adjusted prevalence: 171 per 100 000
• Tetrabenazine 50 mg three times daily. men, 164 per 100 000 women.
• Chlorpromazine (thioridazine), beginning with 10 mg
three times daily and increasing until control of PATHOLOGY
movements up to 150 mg per day. • Loss (>50%) of melanin-containing, pigmented, dopa-
• Olanzapine 5–10 mg daily combined with valproate (a minergic neurons in the substantia nigra, locus ceruleus
mood stabilizer) 125 mg twice daily to 500 mg three and dorsal motor nucleus of the vagus nerve (particularly
times daily. the ventrolateral substantia nigra pars compacta which
• Reserpine beginning 1 mg per day and increasing slowly projects to the posterior putamen, with less involvement
to as high as 15 mg per day. of the medial tegmental pigmented neurons that project
• Others: amantadine, bromocriptine, thiopropazate, to the caudate nucleus) (539–544).
lithium. • Eosinophilic intracellular cytoplasmic inclusions, known
as Lewy bodies (which contain phosphorylated
Depression neurofilaments, ubiquitin, phospholipids and other
• Amitriptyline or imipramine beginning with 50 mg at cytoskeletal components) in the brainstem and other
night and increasing until effective, or until adverse parts of the brain (545, 546).
effects intervene. • Dopamine deficiency (>80%) in the nigrostriatal pathway
• SSRIs. and relative hyperactivity of striatal (putamen and caudate
nucleus) cholinergic activity. Clinical features don’t
Psychosis emerge until 60–80% of nigral neurons and striatal
Antipsychotic medication, as appropriate. dopamine are lost.

PROGNOSIS
• Slowly but inexorably progressive, leading to death on
average about 15–20 years after symptom onset. A few 539
patients have a more chronic course and survive beyond
20 years.
• The progression of the disease is slow in patients with a
late age of onset of symptoms.

539 Section through the mid-brain showing normal pigmentation of


the substantia nigra (arrows).
Parkinson’s Disease (PD) 421

540 Section through the pons 540 541


showing normal pigmentation of the
locus ceruleus in the tegmentum
immediately anterolateral to the
fourth ventricle (arrows). (Courtesy
of Professor BA Kakulas, Royal Perth
Hospital,Western Australia.)

541 Axial sections through the mid


brain (above) and pons (below) of a
patient with Parkinson’s disease
showing lack of pigmentation in the
substantia nigra and the locus
ceruleus due to loss of melanin-
containing pigmented dopaminergic
neurons.

542 543

542, 543 Microscopic sections of the substantia nigra at low 544


magnification power showing normal melanin-containing, pigmented,
dopaminergic neurons (542), and reduction in the number of normal
melanin-containing, pigmented, dopaminergic neurons (543).

544–546 Microscopic sections of the substantia nigra at high


magnification power showing normal melanin-containing, pigmented,
dopaminergic neurons (544), and reduction in the number of normal
melanin-containing, pigmented, dopaminergic neurons, and the
presence of an eosinophilic intracellular cytoplasmic inclusions (Lewy
bodies) in the neurons (arrow) (545, 546).

545 546
422 Degenerative Diseases of the Nervous System

PATHOPHYSIOLOGY component of Lewy bodies.


The dopamine in the basal ganglia participates in a complex – Ubiquitin carboxy-terminal hydrolase L1 (UCH-L1): a
circuit of both excitatory and inhibitory pathways that are deubiquitinating enzyme.
part of a loop that connects the cortex to the thalamus via • Prevalence of PD in first-degree relatives is 1.3–2.1%, which
the basal ganglia and back to the frontal cortex and serves is about double what is expected. The lifetime risk of PD
to modulate the motor system. The pathophysiologic in first-degree relatives of sporadic cases is as high as 17%.
hallmark of parkinsonism is hyperactivity in the subthalamic • For the vast majority of patients (98% or more) who have
nucleus (STN) and medial/internal globus pallidus (GPm). no family history, there may be no genetic contribution
Loss of dopaminergic neurons in the substantia nigra pars or several genes may be responsible.
compacta (SNc) results in dopamine deficiency in the
nigrostriatal pathway, which reduces the normal inhibition of Environmental toxins
the nigrostriatal pathway on GABA-enkephalin neurons in the • N-methyl-4-phenyl-1,2,3,6,-tetrahydropyridine (MPTP),
putamen. This increases the activity in the GABA/enkepha- a synthetic opiate derivative, causes a form of parkinsonism
linergic putaminal neurons that project to, and inhibit, the that strongly resembles PD both clinically and in response
lateral globus pallidus (GPl). The GPl, which sends GABAergic to levodopa. However, a widely distributed equivalent of
inhibitory projections to the STN and the GPm, is now MPTP has not been discovered as yet.
inhibited and so the inhibitory tone of GPl on the STN and • Only a few cases of parkinsonism induced by other toxins
GPm is reduced. The STN, which exerts a powerful excitatory (e.g. pesticides, herbicides, solvents, mercury) have been
drive on to the GPm and substantia nigra reticulata (SNr), now documented.
increases its activity well above normal to excite the GPm/SNr. • PD is more common in non-smokers and rural dwellers
The GPm and SNr are the major output nuclei of the basal but these may be epiphenomena.
ganglia, and finally project, via inhibitory GABAergic pathways,
to the ventrolateral thalamus (VL) on their route to the pre- Infection
motor cortices. The increased inhibitory GABAergic outflow • Epidemics of Von Economo’s encephalitis (encephalitis
from neurons in the GPm/SNr leads to increased inhibition of lethargica) swept Europe in the early 1920s and some
thalamocortical projection neurons and decreased activation of patients developed progressive parkinsonism but it was
the precentral motor fields, resulting in bradykinesia. Excessive not identical to PD and post-encephalitic parkinsonism
tonic discharges are not the only physiologic abnormality of the is now very rare.
basal ganglia in PD; phasic oscillations in neuronal firing appear • Influenza and whooping cough infection have been
to be responsible for tremor. associated with an increased risk of PD in two ecologic
studies but have not been confirmed.
Pathogenesis of cell death
Unknown but probably related to a cascade of events involving: CLINICAL FEATURES
• Oxidative stress. Insidious onset:
• Mitochondrial dysfunction. • Tremor*: at rest, 4–5 Hz, rhythmic, involves hand (‘pill-
• Excitotoxicity. rolling’) ± leg, voice, jaw.
• A rise in intracellular free calcium. • Bradykinesia: diminished rate and range of movements
(e.g. impassive face [hypomimia] (547), reduced finger
ETIOLOGY tapping and arm swing).
Unknown. PD may be due to a complex interaction among • Rigidity: passively move the wrist and elbow joints through
genetic and environmental factors that may differ in their full range and note a uniform increased resistance to
individuals, or alternatively, it may not be one condition passive muscle stretch (‘lead-pipe’ rigidity), with or
with a single cause at all, but a common clinical without a superimposed jerky ‘cogwheel’ character due to
manifestation of different types of insults to the substantia a superimposed tremor that rhythmically interrupts tone.
nigra (e.g. hereditary, toxic, infectious). • Postural instability: impaired equilibrium reactions/right-
ing reflexes: slow to correct balance and tendency to fall
Heredity/genetic susceptibility backwards (retropulsion) or totter forwards (festination).
• 5–10% of patients with PD have a positive family history • Gait disturbances: stooped posture (548), arms flexed at
of a similar disease. the side, narrow stance base, reduced or absent swinging
• PD is inherited in perhaps 1–2% of cases (both autosomal of one arm initially, followed by shortening of stride
dominant and recessive inheritance are known). length (short, shuffling steps), difficulty initiating gait,
• Three genes for PD have been characterized among stiffening of the trunk so that when the patient turns, the
families in which PD is clearly inherited as a mendelian trait: whole body moves in one mass (en bloc), stooped
– α synuclein gene mutations (Park 1): a single amino acid posture, festination and freezing. The gait has a narrow
substitution in the gene on chromosome 4q21-q23 has base, irrespective of the severity of the disease. In
been shown to segregate with PD in a large Italian contrast, patients with ‘lower-half’ parkinsonism due to
kindred with autosomal dominant inheritance. diffuse cerebrovascular disease stand erect in a ‘stiff ’
α synuclein is a major component of the Lewy body. military posture, with the shoulders back, arms extended,
– Parkin gene mutations (Park 2): various deletion and on a wide stance base, but also walk with a short
mutations and point mutations in the parkin gene cause shuffling stepping pattern and freezing of locomotion.
autosomal recessive PD. The parkin gene product, parkin • Asymmetry*: onset is usually unilateral, becoming
protein, is a ubiquitin protein ligase (E3), a component bilateral after a few years.
of the ubiquitin system, which is an important ATP • A good response to levodopa therapy*.
dependent protein degradation machine, and a *Best predictors of PD.
Parkinson’s Disease (PD) 423

Additional features • Dyskinesias: writhing, swinging movements of the limbs


• Many other features are due to the core features. For and trunks. Typically occur in patients with advanced
example, infrequent blinking, facial immobility, soft disease and prominent motor fluctuations and caused by
voice, the ‘reptilian stare’, saccadic ocular pursuit, excess levodopa, but may also occur with dopamine
hypometric ocular saccades, drooling, micrographia, agonists. Dystonic dyskinesia (sustained twisting
flexed body posture at the trunk, neck, elbows and knees; movements) may be painful and necessitate reduction of
joint and muscle pain (e.g. frozen shoulder), and possibly levodopa dosage.
bursitis are a result of muscle rigidity and bradykinesia. • A positive family history of PD is present in 4–16% of
• Pain and sensory phenomena are common. Deep cases.
cramping sensations in the limbs may be a primary
symptom or related to levodopa medication. Superficial The glabella tap sign (tapping the forehead repeatedly
burning dysesthesiae also may occur. with a finger fails to inhibit eye-blinking) is neither sensitive
• Anxiety and depression (30% of cases). nor specific. The deep tendon reflexes remain preserved and
• Forgetfulness and dementia (30% of cases): must be the plantar responses flexor.
distinguished from depression, physical slowness, and
adverse effects of drug treatment. DIFFERENTIAL DIAGNOSIS
• Confusion and hallucinations: usually due to Benign essential tremor (see p.119)
antiparkinsonian medications. Upper limb tremor which is worse with posture and action
• Autonomic dysfunction: postural hypotension, urgency (on attempted writing, the tremor is exacerbated and the
of micturition, erectile impotence, excessive sweating, a script becomes enlarged and irregular whereas in PD, the
feeling of incomplete bladder emptying and constipation. tremor usually abates and the writing becomes smaller as the
script progresses across the page [micrographia]), there is a
positive family history, and the tremor responds to alcohol.

547 548

547 Typical expressionless facies of Parkinson’s disease. 548 Flexed posture and difficulty initiating gait,
with small shuffling steps and diminished arm swing
due to Parkinson’s disease.
424 Degenerative Diseases of the Nervous System

Secondary parkinsonism INVESTIGATIONS


Disease of the basal ganglia and cortical-subcortical Indicated if parkinsonism is atypical for idiopathic PD:
connections: • Onset before the age of 40 years.
• Inherited: • Bilateral symptoms and signs at onset.
– Wilson’s disease (see p.157). • Symmetrical disease.
– Huntington’s disease (see p.417). • Early, severe autonomic failure.
– Levodopa-responsive dystonia-parkinsonism (occurs in • Early involvement of speech and balance:
childhood). – Early dysphagia.
• Recurrent head trauma. – Early postural instability and falls.
• Vascular parkinsonism (‘lower-half parkinsonism’)*: – Early dementia.
ischemic or hemorrhagic stroke (often multiple): sudden • Absence of resting tremor.
onset, pyramidal deficits, clasp-knife spasticity, flexor • Oculomotor (restricted eye movements due to
spasms, erect posture, arms extended, wide stance base, supranuclear gaze palsy), cerebellar or pyramidal tract
dementia, stepwise progression. signs (pseudobulbar palsy).
• Infection: encephalitis lethargica followed a worldwide • Peripheral neuropathy.
flu epidemic in 1918, AIDS in the current era. • Poor initial response to adequate doses of levodopa.
• Tumor of the basal ganglia and cortical-subcortical • Family history of a movement disorder.
connections. • Rapid or stepwise progression.
• Normal pressure hydrocephalus (see p.473): urinary
incontinence, greater involvement of the legs than arms Serum copper and ceruloplasmin
(‘lower-half parkinsonism’), early dementia. In young patients to exclude Wilson’s disease.
• Drug-induced parkinsonism*: dopamine antagonists
(history of exposure, improvement after withdrawal, but Twenty-four hour urinary copper excretion
may take months): In young patients to exclude Wilson’s disease.
– Dopamine receptor blockers:
– Antipsychotic agents (haloperidol, chlorpromazine, CT brain scan
fluphenazine, pericyazine and so on). • To exclude hydrocephalus, cerebral infarction or
– Anti-emetic agents (metoclopramide, prochlorperazine). hemorrhage, and a structural lesion such as AVM or
– Dopamine storage and transport inhibitors: tumor (usually convexity meningioma causing
– Reserpine, tetrabenazine. contralateral hemiparkinsonism).
– Methyldopa. • Usually shows non-specific generalized atrophy of the
– Amiodarone. brain (549).
– Calcium channel blockers.
• Toxins: MRI brain scan
– Carbon monoxide. • May show generalized atrophy.
– Methyl bromide. • Prominent iron deposition (dark signal on T2WI) has
– Manganese poisoning. been described in severe forms of the disease.
– N-methyl-4-phenyl-1,2,3,6,-tetrahydropyridine (MPTP). • Some narrowing of part of the substantia nigra has been
• Degenerative: demonstrated in some cases but both this and the
– Progressive supranuclear palsy (see p.432): early falling, previous feature are very non-specific and hard to spot.
eye movement abnormalities, appreciably more • A combination of putamenal hypointensity and
tone/rigidity in the neck than the limbs (axial rigidity). brainstem atrophy is a consistent finding in Parkinson-
– Multiple systems atrophy (see p.435): early postural plus syndromes and virtually excludes PD.
hypotension, other autonomic disturbance, cerebellar • The main problem with imaging of PD patients is their
ataxia. inability to keep still.
– Alzheimer’s disease (see p.407): early prominent
dementia, particularly with dysphasia and dyspraxia. Positron emission tomography
– Hallevorden–Spatz disease. PET with 18F-fluorodopa is a form of metabolic imaging
– Corticobasal degeneration: rare, progressive, asymmetri- which may indirectly provide a quantitative assessment of
cal akinetic rigid syndrome, usually begins with a presynaptic nigrostriatal dopaminergic function. PET and
unilateral jerky, tremulous, akinetic, rigid and apraxic 18F-fluoro-deoxyglucose may provide an assessment of

limb held in a fixed dystonic posture (e.g. wrist and regional metabolic rates of glucose.
thumb flexed) and displaying the alien limb syndrome.
The hand is severely apraxic, making it functionally Apomorphine test
useless. No weakness or rest tremor. No response to The response of the patient to levodopa therapy can usually
levodopa. be predicted by the apomorphine test (somewhat akin to
• Psychogenic: rare, sudden onset, precipitating factor(s), the edrophonium test for myasthenia gravis):
tremor at rest and with action, no cogwheeling, no • Pre-treat the patient with domperidone 20 mg 8 hourly
fatiguing (decrementing amplitude of movements). for 24 hours before the first dose.
• Depression*. • Measure motor function at baseline:
– Alternate, unilateral hand-tapping on two points 20
*Common. cm(8 in) apart for 30 seconds (Hughes AJ, et al., Lancet,
1990; 336: 32–34.).
– Time taken to walk 12 m (39 ft).
Parkinson’s Disease (PD) 425

– Clinical assessment of tremor and dyskinesia (0=nil, • The patient responds to levodopa.
1=mild, 2=moderate, 3=severe) scoring on a modified • The course is progressive.
Webster disability scale to assess 12 features of PD
(Kempster PA, et al., J. Neurol. Neurosurg. Psychiatry, However, the clinical diagnosis is incorrect in up to 25%
1989; 52: 718–723). of patients, particularly early on in the clinical course. Clues
• Apomorphine 1.5 mg (0.15 ml) subcutaneously (or to an alternative diagnosis are the presence of additional
50 μg/kg subcutaneously) non-parkinsonian features and a partial or absent response
• Observe and measure motor function 30 minutes later. to levodopa and dopamine agonists (see below).
• Motor response is judged to be positive if 2 or more of
the following are seen: Clinical features raising doubts about the diagnosis of
– >15% increase in tapping score over 30 seconds. idiopathic PD
– >25% improvement in walking time. • Early falls and instability.
– At least 2 points improvement of tremor score. • Severe dysarthria.
– An improvement of Webster’s score of 3 or more (out of • Myoclonic jerks.
36). • Pyramidal signs.
• If no or poor response, a second dose of 3 mg • Cerebellar signs.
apomorphine (0.3 ml) is given 40 minutes after the first • Autonomic failure.
dose, and patient observed for a further 30 minutes. • Poor or absent response to levodopa.
• The dose is increased in an incremental fashion every 40 • Pains not relieved by levodopa.
minutes and the patient observed. If required, the third • Rapid clinical deterioration despite dopaminergic
dose is 5 mg s.c., and the fourth dose is 7 mg s.c. If no treatment.
response to 7 mg, the patient is deemed a non- • Lack of typical levodopa induced dyskinesias.
responder. If a mild response to 7 mg occurs, try 10 mg • Atypical levodopa induced dyskinesias (e.g. torticollis,
(1.0 ml) as a maximum dose. antecollis, sustained dystonic spasm of facial muscle).
• 95% of cases of PD improve.
• 25% of cases of Parkinson-plus syndromes improve. TREATMENT
• Severe drowsiness during the test may occur in patients • Most patients are best managed at home (i.e. as an
with Parkinson-plus syndromes but not in patients with outpatient). An hour-by-hour diary of presence and
PD. severity of parkinsonian symptoms and dyskinesias can be
helpful.
DIAGNOSIS • Patients should be encouraged to keep as active as
A clinical diagnosis, which can be made when: possible.
• There are two of the four cardinal clinical features of • Beware of, and treat, concurrent symptoms such as pain
parkinsonism: tremor (present in 80%), bradykinesia, (e.g. with tricyclic antidepressants), anxiety (e.g. with
rigidity, and disturbed postural reflexes. benzodiazepines), and depression (with antidepressants
• There is no alternative cause for the parkinsonism. or ECT).
• The mainstay of antiparkinsonian therapy is dopamine
replacement.
549 • There is no compelling evidence to suggest that early
medical treatment of PD affects the progression of the
disease. There is some evidence that treatment-related
adverse effects may be caused partly by the duration of
drug treatment. It is therefore best to delay treatment
until the patient begins to suffer disability and handicap.
Compelling indications for starting symptomatic
treatment are when employment is in jeopardy and when
falling becomes a risk; otherwise it is a decision based
upon the patient’s personal needs and whether the
symptoms bother the patient sufficiently to make it
worthwhile to pay for the tablets (if they have to) and
take them daily.
• Commence medical therapy at a low dose to minimize
risk of adverse effects such as mental confusion,
particularly in the elderly.

Neurotransmitter replacement therapy


Levodopa
• The most effective drug for PD; it improves most
features.
• A naturally occurring, large, neutral amino acid, most of
549 CT scan from a typical patient with Parkinson’s disease (or with which is metabolized by catechol-O-methyltransferase
head tremor/titubation).The scan is probably normal but any view of (COMT) to form an inactive metabolite, and some of
the brain detail is completely obscured by the motion artefact from which is decarboxylated by an aromatic amino acid
the patient’s tremor! decarboxylase to form dopamine.
426 Degenerative Diseases of the Nervous System

• Usually it is combined with a peripheral decarboxylase Anticholinergics


inhibitor (carbidopa [Sinemet] or benserazide • Benzhexol (Artane), benztropine and trihexyphenidyl have
[Madopar]), that does not cross the blood–brain barrier, a role in young patients with early PD and prominent or
to minimize production of dopamine in the systemic refractory resting (alternating) tremor.
(peripheral) circulation and help prevent adverse effects • Benefit is modest.
such as nausea and vomiting. • Begin with benzhexol or benztropine in a low dose of
1 mg (half a 2 mg tablet) once daily (even lower in the
Sinemet: 100 mg levodopa + 10 mg carbidopa elderly, in whom they may cause a confusional state) and,
100 mg levodopa + 25 mg carbidopa if tolerated but not effective, increase slowly to 2 mg daily,
250 mg levodopa + 25 mg carbidopa 2 mg twice daily, up to a maximum of 5 mg three times
Madopar: 50 mg levodopa + 12.5 mg benserazide daily.
100 mg levodopa + 25 mg benserazide • Adverse effects include dry mouth, mental confusion,
200 mg levodopa + 50 mg benserazide hallucinations, blurred vision, and difficulty initiating
micturition and urinary retention.
Early in the disease, low dose levodopa, 100 mg (or even • Contraindicated in glaucoma.
50 mg), in combination with decarboxylase inhibitor, 25 mg • Avoid if possible, or use with caution, in the elderly
(or even 12.5 mg), taken three to four times daily controls because of the high incidence of confusion and only
most patient’s symptoms very well (the ‘honeymoon period’ modest antiparkinsonian benefit.
of a few years). If not effective but tolerated, increase the dose • Discontinue gradually; acute withdrawal of anticholinergics
slowly to an effective dose (that improves function), which is may be associated with dramatic worsening of
commonly about 500–600 mg per day, and which does not parkinsonism.
cause adverse effects such as confusion and involuntary
movements. Failure to evoke substantial benefit should lead Amantadine
to a re-evaluation of the diagnosis. • Anticholinergic (antimuscarinic) and weak dopamine
In the early stages of PD, the response to levodopa is agonist activity: releases dopamine from body stores. It
sustained, despite the relatively short half-life of levodopa may also work as a glutamate antagonist.
(about 90 minutes). Patients are often able to miss doses • May have a mild and temporary (up to 1 year)
without any deterioration in clinical response. As the disease antiparkinsonian effect in early stages of the disease and
progresses, with continued loss of dopaminergic neurons in may reduce dyskinesias in patients with motor fluctuations.
the substantia nigra, the duration of benefit following a single • Begin with 100 mg capsule in the morning and, if
dose of levodopa diminishes, eventually mirroring the drug’s necessary add another at mid-day.
plasma concentration curve. This phenomenon is known as • Lower dose in renal impairment; its elimination depends
‘end of dose failure’ or ‘wearing off’ and is characterized by on renal clearance.
increasing bradykinesia and tremor in the hour or two before • Adverse effects include skin mottling (livedo reticularis) in
the next dose of levodopa is due. These predictable motor half of patients, inflamed, swollen legs (erythromelalgia),
fluctuations are best managed by aiming for relatively constant and anticholinergic effects.
levels of levodopa by reducing the time interval between each
dose and prescribing more frequent, and sometimes smaller, Synthetic directly acting dopamine receptor agonists
doses of levodopa, keeping the total daily intake of levodopa Advantages (theoretical at least) over levodopa
to 600–800 mg. Alternatively, slow-release levodopa • Do not require biologic conversion to an active agent and
preparations or dopamine agonists (see below) may be used. therefore are not dependent on the presence of residual
Subsequently, the patient may develop unpredictable dopaminergic neurons or a pool of decarboxylase enzyme.
motor fluctuations that are independent of plasma levodopa • Long half-life which helps to smooth out motor
concentration and are attributed to postsynaptic changes in fluctuations and reduce ‘on-off’ phenomena.
the dopamine receptors and second messengers. Recurrent • Less likely to cause dyskinesias.
swings between dyskinetic adverse effects of levodopa (‘on’) • Lack of competition for absorption into the brain.
and severe bradykinesia (‘off’) may occur, as may sudden • Potential to stimulate selectively a subset of dopamine
episodes of ‘freezing’. Individual levodopa doses may fail to receptors.
provide any benefit at all, known as ‘no-on’ phenomenon. • Lack of potentially harmful oxidative metabolites.
The addition of a dopamine agonist such as bromocriptine • Fewer long term adverse effects.
(up to 20 mg/day, starting gently with 1.25 mg bd) or
pergolide or lisuride, 2 mg) may be helpful, or prescribing a Disadvantages over levodopa
slow release form of levodopa. Controlled release levodopa • Less potent and therefore less antiparkinsonian effect.
for patients with motor fluctuations results in fewer off • More likely to cause confusion: use cautiously in the
periods than with standard levodopa, a 5% increase in ‘on’ elderly because of possible acute psychotic reactions.
time and a reduction in end of dose dystonia but an increase • May cause vasospasm, ankle swelling, pulmonary edema
in dyskinesia. and (rarely) pleuropulmonary or retroperitoneal fibrosis
Increasing the dose of levodopa to a maximum of 1–1.5 (perform a chest x-ray before starting treatment).
g daily (in 4–8 divided doses to smooth fluctuations in blood
levels) runs the risk of causing adverse effects which include Bromocriptine
nausea, postural hypotension, neuropsychiatric problems • A D2 dopamine receptor agonist with weak D1
(mental confusion and hallucinations), and involuntary antagonistic effects.
movements of the mouth, tongue and limbs (peak dose • Acts for about 3–5 hours.
dyskinesia).
Parkinson’s Disease (PD) 427

• Can be used as the sole agent in some cases or more • May also be given sublingually, and as a lozenge taken
commonly, as adjuvant therapy with levodopa. orally.
• Available as 2.5 mg tablets, or 5 mg and 10 mg capsules. • Equivalent potency to levodopa.
• Begin at a low dose 2.5 mg bd and increase slowly as • Benefit occurs within 5–15 min, lasting 40–90 min.
required and tolerated to a typical dose of 5 mg tds/qid • Adverse effects similar to those from levodopa can occur,
and a maximum dose of about 30–40 mg/day. Higher with the addition of yawning, drowsiness, and local skin
doses (40–60 mg per day) tend to cause mental reactions or abscesses at injection sites.
confusion and postural hypotension. • Oral domperidone is often given before each dose to
• Adverse effects similar to levodopa can occur, but it is prevent nausea.
more likely to cause confusion and commonly causes
initial nausea because it acts centrally as well a Selective inhibition of catechol-O-methyltransferase
peripherally, and stimulates dopamine receptors in the (COMT) in the periphery
vomiting center in the medulla. Entacapone
• Increases availability of levodopa for transport across the
Pergolide blood–brain barrier by reducing peripheral levodopa meta-
• A combined D1 and D2 dopamine receptor agonist. bolism, thus extending the duration of action of each levo-
• Has a long motor benefit (4–6 hours). dopa dose by about 30–50 minutes, irrespective of
• Use about one-tenth the dose of bromocriptine: begin whether a standard or slow release form of levodopa is
with 50 µg orally, twice daily, gradually increasing if used.
necessary up to a maximum of 1.5 mg orally, twice daily. • Not indicated for early untreated PD, but indicated for
PD with motor fluctuations.
Lisuride • It must be used in conjunction with levodopa and a
• D2 dopamine receptor agonist. peripheral decarboxylase inhibitor.
• Use about one-tenth the dose of bromocriptine. • Adverse effects include dyskinesias and other
dopaminergic adverse effects, and require a reduction in
Cabergoline levodopa dose.
• Dose 3–5 mg/day.
• For patients with early untreated PD, monotherapy is Selective inhibition of the major catabolic
comparable to levodopa in terms of efficacy and delays enzyme of dopamine in the brain, monoamine
motor fluctuations compared to levodopa. oxidase type b (MAO-B)
• For patients with motor fluctuations, carbergoline is Selegiline HCl (deprenyl)
comparable to bromocriptine and pergolide and increases • Selectively inhibits MAO-B, one of the enzymes that
‘on’ time by about 10%, reduces ‘off’ time by about 17%, catabolizes dopamine in the brain, and thereby retards
increases motor scores by about 37%. the breakdown of dopamine and increases the duration
of action of dopamine.
Pramipexole • Does not inhibit MAO-A, so there is not the
• Dose 3–5 mg/day. requirement for dietary and drug restriction as there is
• For patients with early untreated PD, monotherapy for patients taking MAO-A inhibitors. However, dietary
improves motor scores by 20–30% compared with restrictions are required when used with moclobemide
placebo. (a reversible MAO-A inhibitor) due to increased
• For patients with motor fluctuations, pramipexole tyramine sensitivity.
increases motor and ADL scores by 20–25%, and reduces • Metabolized to desmethyldeprenyl, methylamphetamine
‘off ’ time by about 30% compared with placebo; and and amphetamine in the (–) form.
increases motor scores by 12% and ADL scores by 4% • Does not usually exert a significant symptomatic effect
compared to bromocriptine. when given alone.
• 5 mg tablets, taken in the morning and, if necessary, at
Ropinirole midday to a maximum of 10 mg daily.
• Dose 10–20 mg/day. • When taken with levodopa it slightly improves the
• For patients with early untreated PD, monotherapy duration of the levodopa effect and can smooth out early
increases motor scores by 24% compared with placebo, wearing off, but it can also provoke or worsen dyskinesias
and controls symptoms in about 30% of patients. After 5 and psychiatric adverse effects.
years monotherapy, patients have fewer dyskinesias than • Adverse effects include nausea, insomnia, musculoskeletal
if taking levodopa. injuries, non-threatening cardiac arrhythmias, and
• For patients with motor fluctuations, ropinirole reduces elevations in liver enzyme levels.
‘off’ time by 20%, and enables the levodopa dose to be • Avoid concurrent use with fluoxetine and pethidine.
reduced by about 30%. • Controversy as to whether it protects dopaminergic
neurons and slows the progression of PD; the presumed
Apomorphine mechanism may be that it reduces oxygen free radical
• A combined D1 and D2 dopamine receptor agonist. formation generated by the MAO-B oxidation of
• Used in patients with motor fluctuations for rapid relief dopamine, and prevents the activation of exogenous
from sudden ‘off’ periods. neurotoxins. Those who believe this often use it as an
• Given by s.c. injection (0.2–5 mg), either intermittently initial treatment.
or by continuous infusion, for ‘off’ periods. • Controversy also as to whether it may be associated with
excess mortality.
428 Degenerative Diseases of the Nervous System

Problems with medical therapy • Related to a reduction in the PD brain’s capacity to store
Nausea dopamine (because of progressive reduction in number
• Start with low doses of levodopa/dopamine agonists, of nerve terminals capable of storing dopamine), leading
such as half a tablet of the smallest dose, and increase by to a reduced capacity to buffer fluctuations in the plasma
half a tablet every third day, because the patient’s initial levodopa concentration, and a translating of fluctuating
tolerance is poor. plasma levels of levodopa into fluctuating striatal
• Take the medication 30 minutes after food. dopamine levels and motor response. Concurrently, there
• If nausea persists, commence the peripherally-acting anti- is an increasing dependency on exogenously administered
emetic drug domperidone, 10 mg, to be taken half an levodopa to provide dopamine for stimulation of striatal
hour before each dose of antiparkinsonian medication receptors. Accordingly, factors that interfere with
(i.e. with food, followed by antiparkinsonian drug 30 levodopa absorption, such as dietary protein or
minutes later). Avoid other anti-emetics which are alterations in gastrointestinal transit time, can lead to
dopamine antagonists, such as metoclopramide and ‘off’ episodes.
prochlorperazine. • Can be controlled in the early stages by:
– Strategies that enhance levodopa absorption in the brain
Postural hypotension (e.g. reschedule protein intake, low protein diet [to avoid
• Minimize dose of antiparkinsonian drugs (particularly competition of neutral amino acids with L-dopa
dopamine agonists, selegeline). absorption and entry into the brain]).
• Take (lower dose) antiparkinsonian drugs after meals. – Manipulation of the levodopa dose or use of the
• Elastic support/compression stockings (poorly tolerated). sustained release formulations of levodopa.
• Small frequent meals. – Entacapone, a COMT inhibitor that prolongs plasma
• Head-up tilt of the bed at night. half-life of levodopa.
• Caffeine, pseudoephedrine, indomethacin, domperidone. – Dopamine agonists.
• Fludrocortisone. • In advanced stages of the disease, fluctuations are
• Midodrine. difficult to treat and patients frequently cycle between
‘on’ periods complicated by dyskinesia and ‘off’ periods
Motor fluctuations (see Table 40) in which they are frozen and akinetic.
• After about 5 years of treatment, about half (30–80%) of
patients develop motor fluctuations. Dyskinesias (abnormal involuntary movements)
• Motor fluctuations consist of variations in response to a • Tend to develop in younger PD patients.
single dose of levodopa; i.e. swings between parkinson- • Do not occur in normal individuals.
ism and dyskinesias. • Usually choreiform (dance-like).
• Patients oscillate between periods in which they respond • May be dystonic (sustained and often painful muscle
to the drug (‘on’ periods) and periods in which they do contractions) or myoclonic (sudden jerks).
not (‘off’ periods). • In the extreme, may be more disabling than the
parkinsonism itself.

Table 40 Management of motor fluctuations and dyskinesias


Motor fluctuations Dyskinesias
‘Wearing-off ’, end-of-dose deterioration Peak-dose dyskinesia
Take levodopa 30 minutes before meals Reduce total levodopa dose
Smaller, more frequent doses of levodopa Add dopamine agonist to maintain motor response
Add dopamine agonist
Add COMT inhibitor (e.g. entacapone) Early morning dystonia
Add MAOI (e.g. selegilene) Slow-release levodopa at night
Long-acting dopamine agonist at night
Delayed ‘on’ response to levodopa Antispasmodic (e.g. baclofen)
Take levodopa 30 minutes before meals
Low protein diet End-of-dose dystonia
Antacids, gastric prokinetic agents (cisapride) Slow-release levodopa
Long-acting dopamine agonist
Sudden ‘off ’ periods
Liquid levodopa ‘Off ’ period dystonia
Subcutaneous or sublingual apomorphine Liquid levodopa
Low protein diet Subcutaneous or sublingual apomorphine

‘On-off ’ phenomena Diphasic dyskinesia (beginning and end of dose)


Higher, less frequent doses of levodopa Higher, less frequent doses of levodopa
Dopamine agonist with levodopa Pallidotomy if refractory to drug manipulation
Apomorphine infusion
Parkinson’s Disease (PD) 429

• May be monophasic (peak-dose dyskinesias), occurring at Surgery


the time of maximal clinical improvement; and biphasic Surgical treatments for PD are now enjoying a renaissance
(onset- and end-of-dose dyskinesias), occurring at time of with the development of models of basal ganglia circuitry,
disappearance and reappearance of parkinsonian symptoms. refinements of stereotactic surgery (due to better stereotactic
• Cause is unknown; may be related to upregulation of frames, imaging by high resolution brain CT and MRI, and
dopamine receptors or post-synaptic changes associated intraoperative electrophysiologic microelectrode assessment)
with both PD and exposure to levodopa. and the limitations of existing medical therapy.
• Monophasic dyskinesias can be reduced by decreasing and The main surgical ‘targets’ in the brain are the internal
spreading the daily doses of antiparkinsonian medication, but (medial or posteroventral) globus pallidus (GPi) and the
this may preclude a satisfactory antiparkinsonian response subthalamic nucleus. The main surgical strategies are the
and lead to the emergence of motor fluctuations in the form creation of lesions (i.e. destroying brain tissue) or high-
of end-of-dose akinesia and ‘on-off’ phenomena. frequency deep-brain stimulation (DBS) via stimulating
• Biphasic dyskinesias can be alleviated by increasing the electrodes (i.e. stimulating brain tissue).
daily dose of antiparkinsonian medication to maintain
constant high plasma levels of medication (e.g. Pallidotomy
levodopa), but this may produce chaotic dyskinesias and Lesions in the posteroventral portion of the medial/internal
severe psychiatric disorders. globus pallidus (GPm) presumably reduce the inhibitory
• Dyskinesias may also be alleviated to some extent by output from the medial globus pallidus and thereby improve
coadministering various agents such as anticholinergics, many of the ‘off’ period symptoms of PD (tremor, rigidity,
benzodiazepines, serotonin antagonists (e.g. fluoxetine), and bradykinesia on the side contralateral to the surgery and
beta-blockers, low dose clozapine (50 mg) (a some elements of gait). However, they do not improve the
dibenzodiazepine derivative that blocks D1, D2 and D4 patient’s level of function when ‘on’ except for elimination of
dopamine receptors) and riluzole (an inhibitor of peak-dose levodopa-induced dyskinesias. Midline symptoms,
glutamatergic transmission in the CNS). such as postural instability and abnormal gait, also improve
less. Any improvements are immediate and sustained for at
Neuropsychiatric problems (confusion, hallucinations, least 6 months.
delusions, psychosis) Complications include homonymous hemianopia (up to
• More frequent in older patients and those with advanced PD. 14%), facial paresis (up to 51%), and hemiparesis (up to 4%).
• Can be produced by all antiparkinsonian drugs, but Because the optic tract is in intimate relation to the surgical
levodopa is the least likely to do so. target in the ventral GPm, it is essential that the optic and
corticospinal tracts are identified by microelectrode recording
Can be minimized or treated by: and stimulation, not only to avoid injury but also for precise
• Eliminating unnecessary psychoactive or sedative placement of the lesion at the most ventral, medial and
medications. posterior aspect of the nucleus. The optimum size and site
• Withdrawing anticholinergics, amantadine and selegiline, of the lesion within the GPm remain to be determined. In
and then ergot-based dopamine agonists if necessary, addition we do not know whether lesions in the GPm proper
restricting antiparkinsonian therapy to levodopa-carbidopa. and in the pallidofugal outflow axons are equivalent. Medical
• Using the lowest dose of antiparkinsonian medication therapy is usually still required after pallidotomy although
that will provide a satisfactory clinical response. the response becomes smoother and more predictable.
• Clozapine, an atypical neuroleptic, when given in small Indications: relatively young (<60 years), cognitively
doses (starting at 12.5 mg daily and seldom increasing intact PD patients who maintain a response to levodopa but
above 100 mg daily) to patients without dementia who who experience disabling off periods (akinesia/rigidity) or
are experiencing hallucinations, may help control the troublesome levodopa-induced dyskinesias, particularly if
psychotic features and permit higher doses of the signs are asymmetric.
levodopa–carbidopa to be used, but weekly blood counts
are required because of the risk of agranulocytosis. Subthalamotomy
• Other ‘atypical’ antipsychotic agents: olanzapine, quetiapine. Lesions may improve motor signs but at present carry an
• In patients with dementia, it may not be possible to unacceptable risk of inducing hemiballismus or causing
control parkinsonism without adversely affecting mental midbrain hemorrhage due to its high vascularity.
function.
Thalamotomy
Depression • Effective predominantly for medically intractable tremor.
• May be an inherent component of parkinsonism or a • A thermally induced lesion of the ventrointermediate
reaction to having a chronic progressive neurodegenera- nucleus (Vim) of the thalamus improves contralateral
tive disorder. tremor in about 90% of patients.
• Treatment for PD should be the first consideration and • Recurrence of tremor occurs in about 5–10% of cases,
may itself improve the depression. usually within the first 3 months.
• Tricyclic antidepressants have anticholinergic properties • Lesioning the ventralis oralis anterior/posterior (rostral
that may improve PD features in the early stages but may to the Vim) may improve rigidity of the contralateral
aggravate mental function in patients with more limbs.
advanced disease. • Risks of this procedure are those of passing a needle
• SSRIs that are free of anticholinergic effects, such as through the brain (intracerebral hemorrhage) and
fluoxetine or sertraline, are probably preferable but may thermolytic lesioning of structures adjacent to the target
interfere with the antiparkinsonian effect of levodopa. site. Mortality varies from 0.4–6%.
430 Degenerative Diseases of the Nervous System

Deep brain stimulation (DBS) DIFFUSE LEWY BODY DISEASE (DLBD)


• High frequency DBS (about 150 cycles per second)
functionally inhibits neuronal activity in specific brain
targets without the need to make a lesion. By connecting DEFINITION
an implanted stimulating electrode (diameter of 1.3 mm A syndrome, first reported in 1961, and still in the process
[0.05 in] and length of 12 mm 0.5 in]) to a sub- of being defined clinically and neuropathologically, that is
cutaneous pacemaker, target sites can be continuously characterized clinically by:
stimulated. • Fluctuating visual hallucinations and delusions.
• Stimulation of Vim of the thalamus effectively and safely • Parkinsonism (muscle rigidity and bradykinesia)
suppresses tremor in >80% of patients, with benefits • Progressive dementia.
sustained for >5 years in some patients. • A poor tolerance of neuroleptic drugs.
• Chronic bilateral stimulation of the STN may
dramatically improve all cardinal features of PD and DLBD is defined pathologically by diffuse cortical Lewy bodies.
medication requirements but hemiballism may emerge.
• Implantation of DBS into the GPm instead of the STN EPIDEMIOLOGY
is being investigated. • Incidence: not uncommon: up to 20% of cases of dementia.
• The advantages of DBS over thermolytic lesioning are • Age: elderly.
that it is reversible and causes minimal or no damage to • Gender: M=F.
the brain. However, it requires two surgical procedures
(one for targeting and another for definitive electrode PATHOLOGY
internalization), the life of the battery is limited to a few Essential for the diagnosis of DLBD
years, and tolerance to the therapeutic efficacy may • Lewy bodies in the neocortex (temporal>frontal=parietal),
develop. limbic cortex (cingulate, entorhinal, amygdala), subcortical
nuclei, and brainstem (as opposed to nigrostriatal and brain-
Transplantation/cell implantation stem Lewy bodies associated with Parkinson’s disease).
Strategies are based on the notion that dopaminergic cells • Lewy bodies are intracytoplasmic, spherical, eosinophilic,
implanted in the striatum can compensate for degenerating neuronal inclusion bodies composed of abnormally
nigral neurons. More than 150 patients have undergone ubiquinated neurofilament proteins (550). They are
fetal nigral cell implantation with inconsistent clinical results; immunoreactive to ubiquitin (hence are more readily
some patients have shown benefit and adverse effects have visualized with anti-ubiquitin immunocytochemical
not been a major problem. The role of fetal nigral detection), and are surrogate markers of neuronal loss.
transplantation as a treatment for PD needs to be better α-synuclein immunocytochemistry is potentially the most
defined by controlled trials. sensitive and specific technique for detecting and
quantifying Lewy bodies and Lewy neurites.
PROGNOSIS
• Slowly progressive over several years. Associated but not essential for the diagnosis of DLBD
• Symptoms are usually well controlled for the first few • Lewy-related neurites: found by means of ubiquitin
years of treatment. staining in the hippocampus (CA2/3 region), amygdala,
• About half of patients experience significant nucleus basalis of Meynert, dorsal vagal nucleus, and
complications of therapy after 5 years. other brainstem nuclei. They are a neurofilament
• Unless there is an intercurrent cause of death, patients abnormality in which the proteins are present as a diffuse
eventually become bedbound and die of septicemia due aggregate that does not contain crystallin.
to pneumonia or urinary tract infection, or pulmonary • Plaques (all morphologic types). Senile neuritic plaques,
embolism. often in similar numbers to those found in Alzheimer’s
• The average duration of the disease from diagnosis to disease, and β-amyloid deposition are common.
death is about 13 years. • Neocortical neurofibrillary tangles are few or absent.
• Regional neuronal loss occurs, particularly in brainstem (sub-
Prognostic factors stantia nigra and locus ceruleus) and nucleus basalis of
• Gait disturbance at presentation tends to indicate more Meynert. There is no evidence of significant neuronal loss in
severe disease and a faster rate of progression. the hippocampus and medial temporal and frontal cortices.
• Tremor-dominant parkinsonism tends to have a more • Microvacuolation (spongiform change) and synapse loss.
benign course. • Neurochemical abnormalities and neurotransmitter deficits.
• Onset of PD after the age of 60 years is associated with
a greater likelihood of developing dementia, while ETIOLOGY
younger patients have a greater risk of developing adverse Unknown.
effects associated with using levodopa long term.
CLINICAL FEATURES
The clinical presentation is typically delirium-like, with
fluctuating confusion, attentional deficits, and psychiatric
symptoms, particularly visual hallucinations. Rigid-akinetic
parkinsonism (usually mild), intermittent loss of con-
sciousness, and falls are also common.
• Consciousness: recurrent falls and/or transient clouding,
or loss, of consciousness occur.
Diffuse Lewy Body Disease (DLBD) 431

• Cognition: Repeated falls, syncope, and transient loss of


– Cognitive impairment is present, affecting memory, lan- consciousness
guage, visuospatial ability, praxis, and reasoning skills (e.g. Transient ischemic attacks of the brain (see p.186).
early prominent attention deficits, disproportionate diffi-
culties with problem solving and visual-spatial-perceptual Delusions and hallucinations
function). Cognitive impairment is persistent and rapidly • Complex partial seizures.
progressive but characterized by pronounced fluctuation, • Delusional disorder (late paraphrenia).
varying between lucid intervals and episodic confusion.
– Hallucinations: usually persistent, well-formed, visual N.B. Neuroleptic drugs, which are commonly the first choice
hallucinations, and accompanied by secondary paranoid of medication for psychiatric symptoms and behavioral dis-
delusions. Auditory hallucinations may occur. turbances in dementia, commonly induce severe sensitivity reac-
• Extrapyramidal syndrome: tion in DLBD patients, exacerbating motor and mental disability.
– Mild extrapyramidal features occur in some patients at
presentation but more commonly occur later or after INVESTIGATIONS
treatment with neuroleptic drugs (see below). • Neuropsychologic testing: global cognitive dysfunction
– Unusually severe extrapyramidal symptoms or sedation occur with marked impairment of tests sensitive to frontal lobe
after administration of standard doses of neuroleptic agents dysfunction.
(patients are ‘exquisitely’ sensitive to neuroleptic agents). • CT brain scan: normal or generalized cortical atrophy.
• Urinary incontinence: this may precede, but more • Blood: ApoEε4 and debrisoquine oxidase C4P2D6B:
commonly soon follows, the onset of cognitive decline. increased frequency in DLBD.
• SPECT: reduced cerebral blood flow, similar pattern to
DIFFERENTIAL DIAGNOSIS Alzheimer’s disease (see p.412).
Cognitive decline • EEG: early generalized slowing of the background rhythm.
• Alzheimer’s disease (see p.407): behavioral/psychiatric
symptoms and urinary incontinence do not tend to occur DIAGNOSIS
in early Alzheimer’s disease. The dementia does not show • Pathologic: at post mortem.
marked fluctuation and accompanies significant neuronal • Clinical diagnosis: consensus criteria (McKeith, et al.
loss in the hippocampus and medial temporal and frontal [1996, 1999]):
cortices. Neurofibrillary tangles and senile neuritic 1 Progressive cognitive decline of sufficient magnitude to
plaques are common. interfere with normal social or occupational function is
• Parkinson’s disease (see p.420) with or without the central feature required. Prominent or persistent
dementia: Lewy bodies are predominantly subcortical in memory impairment may not necessarily occur in the
location (substantia nigra, locus ceruleus, substantia early stages but is usually evident with progression.
innominata, and dorsal motor nucleus of the vagus Deficits on tests of attention and of frontal-subcortical
nerve), in contrast to DLBD where immunocyto- skills and visuospatial ability may be especially prominent.
chemically similar Lewy bodies are more widespread and 2 Two of the following core features are essential for a
found in the neocortex as well as brainstem neurons. diagnosis of probable DLBD, and one is essential for
• Vascular dementia (see p.261). possible DLBD:
• Progressive supranuclear palsy (see p.432). a Fluctuating cognition with pronounced variations in
• Normal pressure hydrocephalus (see p.473). attention and alertness.
• Creutzfeldt–Jakob disease (see p.330): rapidly progres- b Recurrent visual hallucinations that are typically well
sive cognitive decline and myoclonus. formed and detailed.
c Spontaneous motor features of parkinsonism.
Fluctuating cognitive function 3 Features supportive of the diagnosis are:
Delirium due to drug toxicity (particularly anticholinergics or a Repeated falls.
catecholaminergics) or intercurrent illness. b Syncope.
c Transient loss of consciousness.
d Neuroleptic sensitivity.
550 e Systematized delusions.
f Hallucinations in other modalities.
4 A diagnosis of DLBD is less likely in the presence of:
a Stroke, evident as focal neurologic signs or on brain imaging.
b Evidence of any physical illness or other brain disorder
sufficient to account for the clinical picture.

TREATMENT
• No response to a trial of levodopa.
• Some may respond to cholinesterase inhibitors (e.g.
rivastigmine).

PROGNOSIS
Progressive deterioration occurs over the next 2–5 years,
with increasing parkinsonism, cognitive decline and
550 A round eosinophilic (pink) Lewy body in the cytoplasm of a neuron. psychiatric symptoms.
432 Degenerative Diseases of the Nervous System

PROGRESSIVE SUPRANUCLEAR – Axial (neck and trunk) dystonia and rigidity and
PALSY (PSP) symmetric bradykinesia (552).
Retrocollis or dystonic arm.
Unsteady gait (wide-based, shuffling, the patient moves
DEFINITION ‘en bloc’).
A neurodegenerative disease of the basal ganglia and Postural instability.
brainstem, otherwise known as the Steele–Richardson– – Sudden falls.
Olszewski syndrome, which presents with a disturbance of • Supranuclear ophthalmoparesis, initially involving vertical
balance and downward gaze, and L-DOPA-unresponsive (particularly downgaze) and subsequently horizontal eye
parkinsonism, and subsequently causes progressive movements. The disproportionate hypometria of vertical
dysphagia and dysarthria and death from complications of compared with horizontal sacccades produces a curved
immobility and aspiration. course of oblique saccades. In some patients in whom full
vertical excursions are present, vertical saccades can only
EPIDEMIOLOGY be accomplished by moving the eyes in a lateral arc
• Prevalence: 6.4 per 100 000 (five probable and one instead of strictly vertically, in the mid line. The slowing
possible case) (95% CI: 2.3–10.6). of vertical saccades probably reflects impaired function
• Age of onset: 40–60 years of age. of burst neurons in the rostral interstitial nucleus of the
• Gender: M≥F. medial longitudinal fasciculus (riMLF). As a consequence
of the downgaze paresis, patients have difficulty reading
PATHOLOGY and walking downstairs.
• Neurofibrillary degeneration is the cellular hallmark of • Mild subcortical dementia characterized by slowness of
the disease. central processing time.
• Main lesions are in the SNc, SNr, the globus pallidus, the • Pseudobulbar palsy:
STN and the midbrain and pontine reticular formation. – Dysarthria (spastic: voice has a strained, harsh quality).
• Loss of neurons and gliosis occurs in the periaqueductal – Dysphagia.
gray matter, the superior colliculus, subthalamic nucleus – Emotionalism.
of Luys, red nucleus, pallidum, dentate nucleus, pretectal – Spasticity of lower and (less so) upper limbs.
and vestibular nuclei and to some extent in the – Hyperreflexia and extensor plantar responses.
oculomotor nucleus (551). Surviving neurons in these – Bladder and bowel dysfunction in end stage disease.
areas contain neurofibrillary tangles. The cerebral and • Frontal lobe signs (bradyphrenia, perseveration, primitive
cerebellar cortices are usually spared. reflexes: forced grasping, pout and palmo-mental reflex;
• Atrophy of the brainstem occurs causing dilatation of the imitation and utilization behavior).
third ventricle and cerebral aqueduct, thinning of the • Stuttering speech, torticollis and blepharospasm may
midbrain tegmentum (midbrain diameter <17 mm occur.
[<0.7 in]), and dilatation of the fourth ventricle. • Segmental dystonia or myoclonus may occur.
• There is decreased pigment in the substantia nigra and • Nocturnal disturbances: prolonged latency of sleep onset,
locus ceruleus. prolonged wakefulness, frequent early morning
awakenings, reduced total sleep time.
Molecular pathology • No family history.
• Neuronal degeneration is associated with deposition of
hyperphosphorylated tau protein as neurofibrillary
tangles (similar to corticobasal degeneration, post- 551
encephalitic parkinsonism, post-traumatic parkinsonism,
some forms of frontotemporal dementia, and the
parkinsonism dementia complex of Guam).
• Tau neurofibrillary tangles appear on light microscopy most
commonly as globose tangles and on electron microscopy
as straight filaments with a diameter of 15–18 nm.

ETIOLOGY AND PATHOPHYSIOLOGY


• Unknown.
• PSP is considered a sporadic disorder but a few cases
show autosomal dominant inheritance.
• A genetically determined alteration in the microtubule-
binding protein τ (tau) may be responsible for neuronal
degeneration.

CLINICAL FEATURES
Typical 551 Axial section through the midbrain of a patient with progressive
• Insidious onset of a progressive, symmetric (but may be supranuclear palsy showing midbrain atrophy with dilatation of the
asymmetric) parkinsonian syndrome, unresponsive to cerebral aqueduct due to loss of neurons and gliosis in the
levodopa, characterized by: periaqueductal gray matter, the superior colliculus, substantia nigra,
– Staring, non-blinking, wide eyed (lid retracted) facies subthalamic nucleus of Luys, red nucleus and to some extent in the
(‘reptilian stare’). oculomotor nucleus.
Progressive Supranuclear Palsy (PSP) 433

Atypical findings that do not exclude the diagnosis – Tremor is usually present; in PSP tremor is either absent,
• Appendicular (limb) more than axial rigidity. or not pronounced and of low amplitude.
• Narrow-based gait. – Responds to levodopa usually.
• Mild rest tremor. • Diffuse Lewy body disease.
• Upper limb apraxia. • Hydrocephalus.
• Upper limb ataxia. • Frontotemporal dementia with parkinsonism linked to
• Myoclonus. chromosome 17.
• Chorea. • Prion diseases (Creutzfeldt–Jakob disease, progressive
• Respiratory disturbance. subcortical gliosis).
• Whipple’s disease.
Absence of • Niemann–Pick disease type C.
• Unilateral presentation or pronounced asymmetry. • Vascular pseudo-parkinsonism.
• Early and prominent dysautonomia, particularly postural • Compressive midbrain syndromes (Parinaud syndrome),
hypotension. e.g. pinealoma, glioma.
• Prominent polyneuropathy. • Neurosyphilis.
• Pronounced rest tremor. • Corticobasal degeneration.
• Discriminative (‘cortical’) sensory loss.
• Alien limb sign. Corticobasal degeneration is a rare, sporadic, progressive
extrapyramidal degenerative disease of unknown etiology
DIFFERENTIAL DIAGNOSIS which is characterized by large pale ballooned neurons in
Supranuclear vertical gaze paresis the basal ganglia and the motor and pre-motor cortex. It
• Normal ageing. presents as an asymmetric akinetic rigid syndrome that
• Idiopathic Parkinson’s disease (see p.420): usually begins with a dystonic posturing of the hand with
– Asymmetric in onset. the wrist flexed and the thumb flexed across the palm. The
– Facies not so immobile and without lid retraction. hand becomes functionally useless because of a severe
– Blink rate not so reduced until advanced disease. apraxia rather than any weakness. There is no tremor at rest
– Absence of supranuclear ophthalmoparesis, particularly but attempted movement may evoke episodes of fine
of downgaze. myoclonus in the forearm flexor muscles that can be
– Absence of early pronounced gait imbalance. misinterpreted as an action tremor. Later the ipsilateral foot
– Absence of signs of pseudobulbar palsy (such as harsh, becomes involved followed by the contralateral limbs.
strained voice quality of spastic dysarthria) until late, if at all. However, symptoms and presentations vary, including
– Rigidity and bradykinesia are more appendicular than postural instability, athetosis, focal stimulus-sensitive
axial. myoclonus, action tremor, cortical sensory loss, supranuclear
gaze palsy, Babinski signs, and pseudobulbar palsy.
Behavioral manifestations include frontal-lobe-type behavior
552 Photograph of 552 and language disturbances. Dysarthria is a late sign. The
a man with intellect is usually preserved. No response to levodopa.
progressive
supranuclear palsy Other akinetic-rigid syndromes with
manifesting axial parkinsonian features
rigidity, downgaze • Idiopathic Parkinson’s disease (see above and p.420).
paresis, and • Multiple system atrophy.
bradykinesia. Here he • Cortico-basal degeneration.
tries to look down at • Pallidal, pallidoluysian, and pallidoluysonigral degenera-
the white handle of tions; dentato-rubro-pallido-luysian atrophy.
the inverted walking • Adult-onset Hallervorden–Spatz disease (see p.162): a
stick to help him very rare, sometimes familial, condition of progressive
initiate the first step. rigidity, bradykinesia, dystonia, dysarthria, and dementia
in childhood, or occasionally adulthood. Epileptic
seizures, chorea, cerebellar ataxia, muscle atrophy and
retinitis pigmentosa may also occur. Iron-containing
pigment deposited in the substantia nigra and globus
pallidus can be imaged by MRI.
• Huntington’s disease (see p.417).
• Wilson’s disease (see p.157).
• Cerebrovascular disease:
– Top of the basilar syndrome causing midbrain and
diencephalic infarction.
– Multiple infarcts in brainstem and basal ganglia.
• Subcortical gliosis.
• Prion disease.
434 Degenerative Diseases of the Nervous System

Subcortical dementia • Alien hand syndrome, cortical sensory deficits, severe


• Diffuse Lewy body disease (see p.430). limb apraxia, severe asymmetric bradykinesia, or focal
• Advanced Alzheimer’s disease (see p.407). frontal or temporoparietal atrophy, suggesting cortico-
• Vascular dementia (see p.261). basal degeneration.
• Neuroradiologic evidence of relevant structural
INVESTIGATIONS abnormality (infarcts in basal ganglia or brainstem, lobar
Cranial CT scan atrophy).
• May show enlargement of the third ventricle and • Ocular-masticatory myorhythmia, suggesting Whipple’s
interpeduncular cistern due to atrophy of the midbrain. disease (rule out by polymerase chain reaction [PCR], if
• It is performed to exclude structural lesions, indicated).
hydrocephalus and multi-infarct states which may • Disease course of <1 year or EEG abnormalities sug-
produce clinical findings similar to PSP. gestive of Creutzfeldt–Jakob disease.
• In about one-third of patients, the diagnosis is not made
MRI brain with certainty during life.
• Various signs of brainstem atrophy such as reduction in
size of part of the substantia nigra (as in Parkinson’s TREATMENT
disease) may be seen. Symptomatic
• A high signal in the periaqueductal gray matter is • Antiparkinsonian agents such as levodopa, bromocriptine
present. and amantadine may reduce the symptoms in some cases
• Signal hypointensity within the putamen (as in other but a sustained response is rare.
parkinsonian syndromes) is seen. • Zolpidem, a short-acting hypnotic drug which is
GABAergic and a selective agonist of the benzodiazepine
DIAGNOSIS subtype receptor BZ1, may ameliorate motor symptoms
Clinical diagnostic criteria when used as a single 5 mg or 10 mg dose.
Possible PSP • Cholinergic treatments such as intravenous
• Onset of symptoms at 40 years of age or later. physostigmine may improve cerebral metabolism and
• Either vertical supranuclear gaze palsy or slowing of some measures of neuropsychometric and oculomotor
vertical saccades. performance.
• Prominent postural instability with falls in the first year • Speech and swallowing assessment and management.
of symptoms, and no evidence of other diseases that • Physiotherapy.
could explain the symptoms. • Occupational therapy.

Probable PSP PROGNOSIS


• The presence of a gradually progressive disorder. • Progressive clinical course leading to immobility and
• Vertical supranuclear gaze palsy. anarthria. Pneumonia is the most common immediate
• Other features of possible PSP or supportive criteria: cause of death.
– Frontal/subcortical cognitive dysfunction. • Median survival is about 5 (2–17) years.
– Axial rigidity. • Predictors of a shorter survival time: onset of falls during
– Pseudobulbar dysphagia and dysarthria. the first year, early dysphagia, incontinence.
– Blepharospasm/apraxia of eyelid opening.

Definite PSP
• A history of probable or possible PSP.
• Histopathologic evidence of typical PSP.

Exclusion criteria for possible or probable PSP


• Recent history of encephalitis or oculogyric crisis,
suggesting postencephalitic parkinsonism.
• Severe asymmetry or asymmetric onset of parkinsonian
symptoms (bradykinesia) and/or tremor-dominant
disease, and/or marked and prolonged levodopa benefit,
suggesting Parkinson’s disease.
• Hallucinations, especially if of early-onset or unrelated to
dopaminergic therapy, or delusions unrelated to therapy,
suggesting Lewy body dementia.
• Cortical dementia of Alzheimer’s type (amnesia and
aphasia or agnosia).
• Prominent cerebellar symptoms or unexplained
dysautonomia (early, prominent urinary incontinence or
marked postural hypotension), suggesting multiple
system atrophy.
Multiple Systems Atrophy (MSA) 435

MULTIPLE SYSTEMS ATROPHY (MSA) corticobasal degeneration, progressive supranuclear palsy,


sporadic OPCA, SCA1 with multiple system degeneration, and
chromosome 17-linked dementia. This is analogous to the
DEFINITION situation vis-à-vis Lewy bodies in idiopathic Parkinson’s disease.
A sporadic, adult-onset progressive, degenerative disease of
unknown etiology which is clinically protean, characterized ETIOLOGY AND PATHOPHYSIOLOGY
by autonomic dysfunction, parkinsonism, and ataxia in any Unknown.
combination, but a unifying cellular pathology featuring
glial cytoplasmic inclusions. CLINICAL FEATURES
Any combination of symptoms and signs of dysfunction of the:
TERMINOLOGY • Cerebellum: dysarthria, ataxia (about 50% of patients).
The term MSA was coined in 1969 by Graham and Oppen- • Extrapyramidal system: atypical parkinsonism, poorly
heimer in a report of a patient who developed autonomic responsive to levodopa therapy in most patients.
failure followed by cerebellar and then pyramidal signs and • Pyramidal system: brisk reflexes, spastic quadriparesis
died aged 62 years, just over 4 months after disease onset. (about 50% of patients).
Autopsy revealed cell loss and gliosis (without Lewy bodies) • Autonomic nervous system: postural hypotension,
in the substantia nigra and striatum, olives, pons, cerebellum, impotence and urinary incontinence (almost all patients).
and intermediolateral cells columns of the spinal cord. This
established the notion of pathology in clinically relevant sites Most patients present with one of the three syndromes
being clinically silent. The term was proposed as a shorthand listed below (OPCA, SDS, and SND) but progress so that
to cover many cases described under different titles that they usually end up having features of all three.
overlapped with each other (e.g. sporadic olivoponto-
cerebellar atrophy [OPCA], the Shy–Drager syndrome PREVIOUSLY DEFINED SUBGROUPS
[SDS], striatonigral degeneration [SND]) and which were Sporadic adult-onset OPCA
felt to be variants of the same disorder. • Part of MSA and different from the inherited form.
MSA is now considered a well defined clinicopathologic • Cerebellar signs are almost always accompanied by
entity with protean manifestations, rather than three autonomic failure (e.g. urinary incontinence). As the
different diseases (OPCA, SDS, SND) with some clinical disease progresses, parkinsonism, which is unresponsive
features in common. It is one of the multisystem to levodopa, usually also develops.
degenerations, which is an umbrella term to describe a large • Brisk reflexes.
number of inherited and sporadic degenerative neurologic • No family history.
diseases involving multiple areas of the brain, such as
Huntington’s disease, inherited OPCA, Machado–Joseph Patients with inherited OPCA are not considered to have
disease, and progressive supranuclear palsy. MSA. They present younger, and have less autonomic signs
and parkinsonian features.
EPIDEMIOLOGY
• Prevalence: 4.4 per 100 000 (two probably and two Shy–Drager syndrome (SDS)
possible cases) (95% CI: 1.2–7.6). In 1960, Shy and Drager reported two men with the clinical
• Age: middle-aged and elderly; mean age of onset: and pathologic features of MSA:
54 years (range 31–78 years). • Progressive, primary orthostatic hypotension due to
• Gender: M=F. autonomic failure due to loss of cells in the
intermediolateral column of the spinal cord.
PATHOLOGY • Neurologic signs of extrapyramidal, pyramidal and
Cell loss and gliosis of varying degrees and proportions cerebellar dysfunction.
occurs, without Lewy bodies (unless they are incidental), in • Cell loss and gliosis in the striatum, substantia nigra,
the striatum (particularly the putamen), substantia nigra, cerebellum, pons, olives, and intermediolateral cell columns.
locus ceruleus, inferior olives, pontine nuclei, and cerebellar
Purkinje cells, as well as the intermediolateral cell columns 553
and Onuf ’s nucleus of the spinal cord. Other, more
widespread, pathologic changes may also be present.
Five cellular features are present: glial cytoplasmic inclu-
sions (GCIs), neuronal cytoplasmic inclusions, neuronal nu-
clear inclusions, glial nuclear inclusions, and neuropil threads.
The most characteristic cellular pathology is the argyrophilic
GCI in oligodendroglia (553). They are widely distributed,
more common in white matter than gray matter, more
common in motor fibers than sensory fibers, and may be flame
or sickle-shaped. Ultrastructurally, they are randomly arranged
tubules or filaments, of diameter 20–40 nm, and associated
with granular material. GCIs contain α-synuclein, a synaptic
protein also found in Lewy bodies in Parkinson’s disease. They
are present in the brains of all cases of MSA (i.e. 100% 553 Immunocytochemistry for ubiquitin shows positively-stained
sensitivity) but not all brains containing GCIs are from patients (yellow) glial cytoplasmic inclusions in oligodendrocytes in the pons.
with MSA (i.e. <100% specificity); they are also found in Magnification ×400.
436 Degenerative Diseases of the Nervous System

It is now inappropriate to use the term SDS to describe – Combined with other features such as parkinsonism, pyra-
parkinsonism with autonomic failure. midal and cerebellar signs, as may be the case with MSA.
• Secondary:
Striatonigral degeneration (SND) – Addison’s disease.
In 1964, Adams et al. described three patients with – Amyloidosis.
parkinsonism and brisk reflexes, two of whom had autonomic – Diabetes.
failure, one of whom had extensor plantar responses, and one – Drugs.
had cerebellar signs. In all three, autopsy showed evidence of
striatal neuronal loss, demyelination, and gliosis, principally Cerebellar ataxia
in the putamen and substantia nigra, but also in the olives and • Inherited spinocerebellar ataxia (see p.437).
the cerebellum (and the pons in one case). Incidental nigral • Multiple sclerosis.
Lewy bodies were found in one patient. • Posterior fossa/diencephalic arteriovenous malform-
The common clinical features are: ation/tumor.
• Parkinsonism. • Drug toxicity.
• Autonomic failure.
• Late cerebellar signs. Pyramidal signs
• Parasagittal meningioma.
Relative quantification of clinical features: • Multi-infarct state.
Parkinsonism Autonomic Ataxia
OPCA + ++ +++
SDS +++ ++ + 554
SND +++ ++ +

DIFFERENTIAL DIAGNOSIS
Parkinsonism
Parkinson’s disease (see p.420)
• Most MSA patients are initially diagnosed as having
idiopathic PD, and one-third retain this erroneous
diagnosis until they die.
• Between 4% and 22% (mean 8%) of brains in
parkinsonian brain banks are found to have MSA.
• The clue is that idiopathic PD causes parkinsonism with
or without autonomic failure but it does not give rise to
pyramidal or cerebellar signs.

Lewy body disease (see p.430)


Lewy body disease can cause parkinsonism with autonomic
failure but not pyramidal or cerebellar signs. 555
Progressive supranuclear palsy (see p.432)

Corticobasal degeneration (see p.433)

Drugs
Neuroleptics.

Wilson’s disease (see p.157)

Autonomic failure
• Primary:
– Pure autonomic failure (PAF) (isolated): a syndrome that
has several causes. It may be due to the rare dopamine-
beta-hydroxylase deficiency or may persist in isolated form
and be revealed at autopsy to be related to the pathology
of Lewy body disease or MSA. So, with the passage of
time, an initial syndrome of pure autonomic failure may
mature into clinical idiopathic Parkinson’s disease, Lewy
body disease, or MSA. Helpful distinguishing tests are
plasma norepinephrine concentrations in the supine 554, 555 T1W midline sagittal (554) and T2W axial (555) MRI of the
resting position (normal in MSA, low in PAF), and brain in a patient with a clinical picture of late-onset cerebellar ataxia
plasma arginine vasopressin in upright tilt (very little and additional extrapyramidal symptoms due to MSA (predominantly
increase in AVP in patients with MSA for the degree of olivopontocerebellar degeneration syndrome). Note the shrunken
hypotension, marked rise in AVP in patients with PAF for brainstem and the cerebellar atrophy (arrows).The supratentorial brain
the same degree of hypotension). appears relatively normal.
Autosomal Dominant Cerebellar Ataxias 437

• Normal pressure hydrocephalus. • Impotence may be treated with sildenafil, intracavernosal


• Cervical spondylitic myelopathy. injections of papaverine, prostaglandin E, or implantation
• Amyotrophic lateral sclerosis. of a penile prosthesis.
• Urinary frequency may be reduced by anticholinergics,
INVESTIGATIONS but they may precipitate urinary retention.
CT brain scan • Urinary retention with overflow may require bethanechol
CT is used to exclude some of the differential diagnoses and chloride, a cholinergic muscarinic agonist, intermittent
to identify atrophy of the cerebellum and brainstem, self-catheterization, or an indwelling catheter.
particularly the pons, inferior olives, vermis and cerebellar • Constipation may improve with a high fibre diet and
peduncles. bulk laxatives, or suppositories or enemas.

MRI brain PROGNOSIS


• Atrophy of the brain, particularly the pons, inferior olives, • MSA has a more aggressive course than idiopathic PD,
vermis and cerebellar peduncles (554, 555) is seen. and in most cases significantly shortens life.
• Abnormally low signal intensity may be noted in the • Median survival is about 6 years (range 0.5–24 years).
putamen compared with the globus pallidus on T2W • Older age of onset is associated with shorter survival.
images due the excess iron deposition.
• Linear signal changes along the outer/lateral margin of
the putamen, manifesting as slit-like hyperintensities (on
T2W and proton density sequences and hypodensities on
T1W sequences): a useful MRI feature to help AUTOSOMAL DOMINANT
differentiate between Parkinson’s disease and MSA CEREBELLAR ATAXIAS
predominantly affecting the extrapyramidal system.

PET DEFINITION
• 18F-fluorodeoxyglucose PET: decreased glucose A heterogeneous group of dominantly inherited late-onset
metabolism is found in cerebellum, thalamus, putamen clinical phenotypes that include cerebellar ataxia, nystagmus,
and cortex (i.e. forebrain glucose metabolic defects as dysarthria, dysmetria, intention tremor and ophthalmoparesis,
well as cerebellar). resulting from neuronal degeneration in the cerebellum and
• 11C-diprenorphine: decreased putamenal uptake. cranial nerve nuclei. There may also be varying degrees of
• 18F-fluorodopa: decreased putamenal uptake (as in all dysfunction of the basal ganglia, brainstem, spinal cord, optic
parkinsonian syndromes). nerve, retina and peripheral nerves, resulting in visual loss,
parkinsonism, hyperreflexia and spasticity.
Cardiovascular autonomic function tests
• Plasma norepinephrine concentrations – normal or EPIDEMIOLOGY
slightly raised. • Prevalence: about 1 per 100 000 throughout the world.
• Age: adult onset, after age 20 years.
Electrophysiological studies
• Monitoring of individual motor units from the striated CLASSIFICATION
component of the urethral sphincter shows features Clinical, pathologic and genotypic
consistent with loss of anterior horn cells in Onuf ’s Early classifications emphasized the clinical features, leading
nucleus in the sacral cord: specificity 92%, sensitivity 62%. to designations such as ‘Holmes’ ataxia’ and ‘Marie’s ataxia’.
The pathology was then emphasized and the disorders were
DIAGNOSIS given names such as olivocerebellar atrophy, olivoponto-
• Clinical: progressive symptoms and signs of dysfunction cerebellar atrophy, and cerebellar cortical atrophy. The clinical
of at least two of the cerebellar, extrapyramidal and and pathologic features were then correlated with inheritance
autonomic systems. patterns, and Harding’s classification, including autosomal
• Pathological: see above. dominant cerebellar ataxia types I, II, and II, was widely used.
Recently, knowledge of genotypes has been correlated with
TREATMENT clinical phenotype and pathology, and previous classifications
• The response to levodopa is usually transient, poor, waning, have been refined . Each aspect of the classification has a role;
or absent, because the striatum degenerates. In contrast, in genotypes are likely to provide the firmest basis for genetic
idiopathic PD, the nigra degenerates but the striatum is counselling and for investigations into pathogenesis, while
normal, which is the reason that dopamine replacement clinical and pathologic information may be more relevant to
therapy is so effective. The responsiveness (or lack of) to prognosis and management.
levodopa is therefore a clinical clue to the diagnosis.
• Orthostatic hypotension can often be managed Disorders of trinucleotide repeats
successfully with head-up tilt of the bed at night, elastic The spinocerebellar ataxias comprise five of the eight
support stockings, fludrocortisone, and desmopressin neurologic disorders caused by an increase in the number
(DDAVP). In addition, subcutaneous octreotide inhibits of CAG repeats that encode expanded sequences of
release of vasodilator peptides, and oral vasoconstrictors glutamine residues. The other three are spinal and bulbar
that may be effective include midodrine (a peripherally muscular atrophy, Huntington’s disease, and DRPLA.
acting α1-agonist), epedrine, and phenylpropanolamine. These disorders are characterized by autosomal dominant
or X-linked inheritance, onset in midlife, a progressive
438 Degenerative Diseases of the Nervous System

course, anticipation (a tendency toward earlier onset in of the (CAG) repeat and earlier age of onset
successive generations), preponderance of unstable repeats (anticipation). The repeat number increases with paternal
from the paternal chromosome, and correlation of the transmission of disease (imprinting). The proteins
number of CAG repeats with the severity of disease and the encoded by each mutation all have an increased number
age at onset. The abnormal proteins in each disorder are of glutamine residues. The resulting proteins are not
expressed in a wide range of tissues and are not limited to homologous, and the functions of these so-called ataxins
the affected brain regions. are unknown. The neuronal selectivity that characterizes
each subtype of spinocerebellar ataxia has been correlated
PATHOLOGY with the degree of accumulation of intraneuronal
Microscopic ubiquinated components containing fragments of the
Loss of neurons and degenerative changes in various respective protein. Non-degraded fragments of the
combinations of sites that include: glutamine repeat have a key role in the selectivity of
• Pontine and olivary nuclei. neuronal death.
• Cerebellar dentate nuclei. • The SCA 6 gene, located on chromosome 19q13, encodes
• Cerebellar Purkinje cells. the α1A-voltage-dependent calcium channel subunit.
• Basal ganglia (substantia nigra, subthalamic nuclei, red Intriguingly, point mutations in the SCA 6 gene may cause
nuclei). familial hemiplegic migraine and episodic ataxia.
• Spinal cord (Clarke’s columns, spinocerebellar tracts,
anterior horn cells). CLINICAL FEATURES
• Peripheral nerves (dorsal root ganglia). • Family history of similarly affected members.
• Slow, gradual onset.
For example, in Machado–Joseph disease (MJD), SCA 3,
there is sparing of the inferior olivary nuclei and Purkinje cells Phenotypically heterogeneous:
but substantial involvement of the dentate nucleus and • Cerebellar ataxia, dysarthria, dysmetria, and intention
substantia nigra. In SCA 1 and SCA 3, intranuclear inclusions tremor ±:
are found in neurons that die. The formation of intranuclear – Supranuclear ophthalmoplegia.
inclusions involves the ubiquitin-proteasome complex. – Optic atrophy.
– Pigmentatory retinopathy.
Macroscopic – Dementia.
Atrophy of the brainstem and cerebellum (556, 557). – Extrapyramidal dysfunction (see Table 41).

ETIOLOGY AND PATHOPHYSIOLOGY The clinical syndrome may vary remarkably, even within the
• Autosomal dominant inheritance. same disease and members of the same family. For example,
• Gene mutations on chromosomes 3,6,11,12,14,16,19 there are three phenotypes of MJD (SCA 3). Cerebellar ataxia
identified to date (Table 41). Five genotypes that involve and ophthalmoplegia are common to all types of MJD. Facial
increases in the number of CAG repeats have been and lingual fasciculations, and staring due to lid retraction are
identified. There is a direct correlation between the size other uncommon but helpful diagnostic features.

556 557 Brainstem from a patient with autosomal dominant cerebellar


ataxia (left) and a normal brainstem (right), showing the severe atrophy
of the midbrain, pons and medulla in the ataxic patient (left).The
brainstems have been disconnected from the brains and spinal cords
by cuts through the cerebellar peduncles, midbrain and medulla.

556 Ventral surface of the brain showing severe atrophy of the


medulla, pons and cerebellum.

557
Autosomal Dominant Cerebellar Ataxias 439

Type 1 MJD • Middle age (40 years, mean).


• Early symptom onset, typically around 25 years. • Ataxia and ophthalmoplegia with or without pyramidal signs.
• Pyramidal signs.
• Dystonic postures. Type 3 MJD
• Later onset (47 years, mean).
Type 2 MJD • Amyotrophy prominent.
• Most common type. • Slow progression.

Table 41 Classification of autosomal dominant cerebellar ataxias


Name Gene locus Type of Pathology Phenotype
and protein mutation
SCA 1 6p22-p23 CAG repeats Purkinje cells Ataxia, dysarthria
(ADCA type 1) Ataxin 1* 41–81 Pontine nuclei Ophthalmoparesis
(normal 25–36) Inferior olivary nuclei Optic atrophy
Dysphagia, Amyotrophy
Pyramidal signs
Extrapyramidal signs
SCA 2 12q22-24 CAG repeats Purkinje cells Ataxia, dysarthria
(ADCA type 1) Ataxin 2* 35–59 Basis pontis Slow saccades
(normal 15–24) Inferior olivary nuclei Ophthalmoplegia
Peripheral neuropathy
Minimal pyramidal and extrapyramidal signs
Dementia (rarely)
SCA 3 14q24.3-q31 CAG repeats Substantia nigra Ataxia,
(Machado– Ataxin 3* 62–82 Subthalamic nuclei Ophthalmoparesis
Joseph (normal 13–36) Pontine nuclei Variable pyramidal, extrapyramidal and
disease) Dentate nuclei amyotrophic signs
(ADCA type 1) Clarke’s columns Cranial nerve deficits
Spinocerebellar tracts Exophthalmos
Anterior horn cells
Posterior root ganglia
SCA 4 16q24-ter Unknown Unknown Ataxia (late onset)
(ADCA type 1) Eye movements normal
Pyramidal signs
Sensory axonal neuropathy
SCA 5 Chromosome 11, Unknown Unknown Ataxia (late onset)
(ADCA type III) centromeric region Dysarthria
Pyramidal signs in young-onset patients
Benign course relatively
SCA 6 19q13 CAG Cerebellar atrophy Slowly progressive ataxia
(ADCA type III) α1A Ca channel 21–27 repeats Relative sparing of Dysarthria, nystagmus
Ataxin 6* (normal 4–16) brainstem Limited vertical and horizontal gaze
Hyporeflexia
Loss of proprioception
SCA 7 3p14-21.1 CAG Unknown Ataxia
(ADCA type II) Ataxin 7* 37–130 repeats Dysarthria
(normal 7–35) Pigmentary retinal degeneration
Macular dystrophy
Blindness
SCA 8 13q21 CTG Ataxia, dysarthria nystagmus, spasticity,
(ADCA type III) (untranslated) expansion diminished vibration sense
(untranslated)
SCA 12 CAG
Dentatorubro- 12p12-ter CAG repeats Dentate nucleus Ataxia
pallido-luysian Atrophin 1* 49–85 Red nucleus Choreoathetosis
atrophy (normal 7–255) Dystonia
(DRPLA) Myoclonus
Epileptic seizures
Dementia
ADCA: autosomal dominant cerebellar ataxia; SCA: spinocerebellar atrophy subtype; q: long arm of chromosome; *Function of protein is unknown.
440 Degenerative Diseases of the Nervous System

DIFFERENTIAL DIAGNOSIS • Other spinocerebellar degenerations (see Friedreich’s


• Structural cerebellar, brainstem or spinal cord lesion ataxia, p.441).
(tumor, arteriovenous malformation). • Progressive supranuclear palsy (dementia and supra-
• Alcoholic cerebellar degeneration. nuclear ophthalmoplegia).
• Multiple sclerosis.
• Hypothyroidism. INVESTIGATIONS
• Neurosyphilis. • Blood DNA analysis using PCR for gene mutations,
• Subacute combined degeneration of the spinal cord commonly CAG repeat expansions, on chromosomes
(sensory, not cerebellar, ataxia). 3,6,11,12,14,16,19.
• Wilson’s disease. • Cranial CT or, preferably, MRI scan of the brain and
• Mitochondrial cytopathy. craniocervical junction: pronounced cerebellar atrophy,
• Paraneoplastic cerebellar degeneration: usually a subacute particularly affecting the superior vermis (558–560).
ataxic syndrome. • Full blood count and film.
• Iatrogenic cadaveric human growth hormone-induced • Urea and electrolytes, plasma cortisol, very long chain
Creutzfeldt–Jakob disease: a drug history of hormonal fatty acids (in males, adrenoleukodystrophy).
therapy should be taken from all young well-muscled, or • Thyroid function tests.
previously well-muscled, patients and top athletes with • Liver function tests (alcoholic cerebellar degeneration).
cerebellar signs. • VDRL, TPHA.
• Idiopathic late-onset cerebellar ataxia: age of onset 25–70 • Vitamin B12 and vitamin E.
years (onset after 55 years: often a relatively pure midline • Plasma lactate and pyruvate, mitochondrial DNA analysis
cerebellar syndrome with marked gait ataxia, mild (mitochondrial cytopathy).
appendicular ataxia). • Plasma copper, ceruloplasmin (Wilson’s disease).

558 559

560

558–560 T1W midline sagittal (558) and T2W axial (559, 560) MRI of the brain in a patient with autosomal dominantly inherited
cerebellar ataxia due to a mutation of the gene for spinocerebellar ataxia 1 on chromosome 6 causing expansion of the trinucleotide
CAG repeat. Scans show pronounced atrophy of the medulla, pons and cerebellum, particularly affecting the superior vermis.
Friedreich’s Ataxia 441

• Alpha fetoprotein (ataxia telangiectasia). FRIEDREICH’S ATAXIA


• Antipurkinje cell antibodies (paraneoplastic syndrome).
• EKG, and if abnormal, echocardiograph.
• Nerve conduction studies and EMG. DEFINITION
• Visual evoked potentials and somatosensory evoked An autosomal recessively inherited disease caused by a large
potentials. increase in the number of trinucleotide GAA repeats within the
first intron of the FRDA gene (X25) on the proximal long arm
DIAGNOSIS of chromosome 9. This results in decreased expression of the
Clinical syndrome of cerebellar ataxia, positive family target protein frataxin, and dysfunction of the central and
history, and positive DNA analysis. peripheral nervous systems and the heart. Clinically, Friedreich’s
ataxia is characterized by the onset before age 25 years of
TREATMENT progressive limb and gait ataxia, cerebellar dysarthria, depressed
Symptomatic rather than curative: deep tendon reflexes, pyramidal signs, distal vibration and
• Cholinergic agents: physostigmine, lecithin, and choline proprioceptive sensory loss, axonal sensory neuropathy and
chloride. often skeletal deformities and hypertrophic cardiomyopathy.
• GABAergic drugs: baclofen and sodium valproate.
• Serotonergic compounds: EPIDEMIOLOGY
– L-5-hydroxytryptophan combined with a peripheral • Prevalence: 2 per 100 000; the most common form of
decarboxylase inhibitor. hereditary ataxia.
– Buspirone hydrochloride, a serotonin (5-hydroxy- • Carrier frequency: 1 in 120 in European populations.
tryptamine1A) agonist, 20 mg/day in four divided doses • Age: adolescence and early adult life. Onset usually
and increased by 5–10 mg/day up to 60 mg/day. Adverse occurs at 8–15 years.
effects include transient light-headedness and nausea.
PATHOLOGY
PROGNOSIS Nervous system
Gradually progressive. • Dorsal root ganglia: degeneration/loss of large sensory
neurons.
• Dying back of axons in:
– Large myelinated sensory nerve fibers in peripheral
561 nerves.
– Posterior columns of the spinal cord.
– Nucleus gracilis and cuneatus, and the medial lemniscus.
– Dorsal and ventral spinocerebellar tracts.
• Corticospinal tracts: demyelination, with increasing
involvement caudally.
• Cerebellum:
– Loss of Purkinje cells (561, 562).
– Degeneration of dentate nucleus.
– Axonal loss and demyelination of superior cerebellar
peduncles.

Heart
Degeneration leading to hypertrophy and diffuse fibrosis.

Pancreas
562 Degeneration, giving rise to:
• Diabetes mellitus in about 10% of patients.
• Carbohydrate intolerance in an additional 20%.
• A reduced insulin response to arginine stimulation in all
patients.

ETIOLOGY AND PATHOPHYSIOLOGY


Inheritance
Autosomal recessive.

Gene mutation
Friedrich’s ataxia is due to a mutation in the FRDA gene
(X25), which is located on the proximal long arm of
chromosome 9 (9q13-q21.1). It has five exons spread over
40 kb that encode a novel 210-amino acid protein, named
561, 562 Normal cerebellar cortex, H&E stain, with plentiful ‘frataxin’. More than 95% of patients with classic
Purkinje cells (561, arrowheads), and higher magnification view of Friedreich’s ataxia are homozygous for the increase in GAA
the cerebellar cortex of a patient with Freidreich’s ataxia showing repeats, but a few have a combination of an increase in GAA
Purkinje cell loss (562). repeats in one allele and a point mutation in the other allele,
442 Degenerative Diseases of the Nervous System

confirming that Friedreich’s ataxia is a loss-of-function Gait


disorder. Larger GAA expansions correlate with earlier age Spastic, ataxic (cerebellar and sensory) gait.
of onset and shorter times to loss of ambulation. Friedrich’s
ataxia is the only known example of a triplet repeat DIFFERENTIAL DIAGNOSIS
expansion causing an autosomal recessive disease. Inherited ataxias with known metabolic defects
Ataxia with isolated vitamin E (α-tocopherol) deficiency
Gene expression • Inherited: autosomal recessive: frame-shift mutations in
The expression of the FRDA gene (X25) is not ubiquitous the gene encoding α-tocopherol-transfer protein (α-
but is restricted to the sites of degeneration. The mechanism TTP) on chromosome 8q (13.1-13.3).
by which the intronic triplet repeat mutations leads to • Acquired fat malabsorption syndromes:
cellular degeneration remains unclear. Frataxin appears to – Abeta- and hypobeta-lipoproteinemia (Bassen–Korns-
be a nuclear-encoded mitochondrial protein important for weig syndrome).
normal production of cellular energy. Reduced frataxin (loss – Cholestatic liver disease.
of function) in spinal cord, heart, and pancreas has been – Cystic fibrosis.
postulated as the primary cause of neuronal degeneration, – Short bowel syndrome.
cardiomyopathy and increased risk of diabetes, perhaps by – Onset in second to sixth decade of life.
resulting in abnormal accumulation of iron in mitochondria. Progressive ataxia, dysarthria areflexia, extensor plantar
responses, and proprioceptive loss ± ophthalmoplegia,
CLINICAL FEATURES dystonic posturing of hands/feet, bradykinesia, tongue
The clinical spectrum is broader than previously recognized. fasciculations and pigmentary retinal degeneration. Low
serum vitamin E (11.7 µmol/l [<5 µg/ml]).
HISTORY Low serum lipid concentrations, particularly cholesterol.
Symptoms of incoordination and ataxia of the lower limbs Oral vitamin E (800–3500 mg/day [800–3500 IU])
begin in the early ‘teen’ years and progress to involve the may cause improvement or retard progression.
upper limbs and cranial musculature, so that by the age of
25 years almost all patients have some well established Hexosaminidase A deficiency (GM2 gangliosidosis)
neurologic signs. • Onset in adolescence or early adult life (see p.172).
• Ataxia, tremor, supranuclear ophthalmoplegia, facial grimac-
PHYSICAL EXAMINATION ing, dystonia and proximal neurogenic muscle weakness.
General
• Kyphoscoliosis (may affect posture and pulmonary function). Cholestanolosis
• Foot deformity (pes cavus and extension of the meta- • Autosomal recessive inheritance.
tarsophalangeal joints in about 90% of patients). • Defective bile salt metabolism.
• Hypertrophic cardiomyopathy. • Onset in second decade of life.
• Ataxia, dementia, spasticity, peripheral neuropathy,
Cranial nerves cataract, tendon xanthomata.
• Reduced visual acuity. • Chenodeoxycholic acid (chenodiol) treatment may
• Optic atrophy. improve neurological function.
• Eye movements:
– Square wave jerks at fixation. Leukodystrophies
– Saccadic intrusion upon ocular pursuit (jerky pursuit). • Metachromatic leukodystrophy (arylsulfatase A deficiency).
– Gaze-evoked nystagmus. • Late-onset globoid cell leukodystrophy.
– Reduced gain of vestibulo-ocular reflex. • Adrenoleukomyeloneuropathy: a phenotypic variant of
• Speech: slurred, slow, staccato, and explosive (ataxic dysarthria). adrenoleukodystrophy.

Limbs Other
• Wasting of the intrinsic hand and distal lower leg muscles. • Mitochondrial cytopathy.
• Weakness (pyramidal) of the legs (paraparesis). • Wilson’s disease.
• Ataxia of the limbs, speech and eye movements: • Ceroid lipofuscinosis.
– Bilateral intention tremor. • Sialidosis.
– Dysmetria (overshoot). • Ataxia telangiectasia.
– Dysdiadochokinesia (poor coordination of rapid alternating • Niemann–Pick disease type C (juvenile dystonic lipidosis):
movements). – Ataxia, supranuclear gaze palsy and psychosis.
• Absent deep tendon reflexes (although they may be – Sphingomyelinase activity is normal.
retained) due to axonal degeneration of afferent fibers. This – Foamy storage cells in bone marrow.
is reflected neurophysiologically by absence of SNAPs and
loss of the H-reflex. Autosomal dominant cerebellar ataxias (see p.437)
• Extensor plantar responses.
• Impaired touch, pain and temperature sensations in the feet Early onset ataxias of unknown etiology
and distal lower limbs is unusual but found in a small Early onset cerebellar ataxia with retained tendon reflexes
fraction of patients. • About one-quarter as common as Friedreich’s ataxia.
• Impaired vibration sense in the feet and hands. • Optic atrophy, severe skeletal deformity, and cardiac
• Impaired joint position sense in the distal lower limbs and involvement do not occur.
hands. • Deep tendon reflexes are normal or increased.
Friedreich’s Ataxia 443

• Gait may have a spastic component. Subacute combined degeneration of the spinal cord
• Prognosis is worse than Friedreich’s ataxia. • Ataxia is predominantly sensory rather than cerebellar.
• Associated features include: • Low serum vitamin B12 level.
– Hypogonadism. • Antibodies to intrinsic factor and gastric parietal cell may
– Myoclonus. be present.
– Pigmentary retinopathy.
– Optic atrophy ± mental retardation (including Behr’s INVESTIGATIONS
syndrome). Diagnosis
– Cataract and mental retardation (Marinesco–Sjogren • EKG and echocardiography: many have obstructive
syndrome). hypertrophic cardiomyopathy.
– Childhood deafness. • Pulmonary function tests: may deteriorate due to
– Congenital deafness; extrapyramidal features. kyphoscoliosis.
• Nerve conduction studies:
Congenital deformities (exclude with MRI scan of – Absent sensory nerve action potentials.
the cranio-cervical junction) – Prolonged sensory conduction velocities.
• Arnold–Chiari malformation. – Loss of H-reflex indicative of afferent axonal neuropathy.
• Platybasia. • Somatosensory evoked potentials: absence or abnormalities
• Odontoid compression. of cortical responses to peroneal or tibial nerve stimulation.
• Electronystagmography: fixational instability with square
Multiple sclerosis wave jerks.
• Relapsing and remitting course is usually in young-onset • MRI scan of brain and craniocervical junction: non-
cases. specific atrophy of the cerebellum (563, 564), the
• Bladder involvement is common. cervicomedullary junction and upper cervical spinal cord
• Sensory loss is usually patchy. may be present.
• CSF usually shows elevated protein and IgG and • Molecular DNA analysis for point mutation in X25, or
oligoclonal bands. unstable GAA trinucleotide expansion in the first X25
• MRI brain usually shows multiple lesions in intron, on the proximal long arm of chromosome 9
periventricular white matter, corpus callosum and (9q13-q21.1): useful for diagnosis, determination of
adjacent to the temporal horn of the lateral ventricles. prognosis and genetic counselling.

Structural spinal cord lesion Differential diagnosis


Spinal cord tumor or arteriovenous malformation • Full blood count and film.
• Pain, particularly nerve root pain, is common. • Glucose.
• Progressive spasticity below the level of the lesion. • Lipids.
• Progressive urgency of micturition. • RPR or VDRL, TPHA.
• Sensory level. • Thyroid function tests.
• MRI (± spinal angiography) of spinal cord discloses a • Vitamins B12 and E.
focal lesion. • Copper, ceruloplasmin.
• Cortisol and long chain fatty acids (L C26:C22, C24:C22
Syphilitic pachymeningitis ratio) if spastic paraparesis, axonal neuropathy, male
• Rare. (adrenoleukodystrophy).
• CSF pleocytosis, raised protein and positive VDRL, • Alpha fetoprotein (ataxia telangiectasia).
TPHA, FTA. • DNA analysis for SCA mutations (see p.439).

563, 564 T1W midline 563 564


sagittal (563) and T2W axial
(564) MRI of the brain in a
patient with Freidreich’s
ataxia. Note the marked
cerebellar atrophy (arrows),
but that the rest of the brain
including the brainstem is
normal. (Compare with 553,
multiple systems atrophy.)
444 Degenerative Diseases of the Nervous System

Differential diagnosis (continued) ATAXIA TELANGIECTASIA


• Hexosaminidase: if vertical gaze palsy, dystonia, (LOUIS–BAR SYNDROME)
neurogenic weakness.
• Arylsulfatase A (metachromatic leukodystrophy).
• Galactocerebrosidase: if dementia, psychiatric problems, DEFINITION
optic atrophy, demyelinating neuropathy, radiologic A rare autosomal recessive disorder characterized by onset
evidence of white matter disease. of ataxia in early childhood and subsequent progressive
• Plasma lactate and pyruvate, muscle biopsy and blood for neuromotor degeneration, usually resulting in dependence
mitochondrial DNA analysis: if short stature, myoclonus, on a wheelchair by 10 years of age.
retinopathy, dementia, stroke-like episodes, and
fatiguable weakness. EPIDEMIOLOGY
• Cholestanol: if cataract, tendinous swellings. • Incidence: rare.
• Gonadotrophins: if hypogonadism. • Age: early childhood.
• Ammonia/amino acids: if fluctuating course, mental • Gender: M=F.
retardation.
• Antipurkinje cell antibodies: if subacute course (para- PATHOLOGY
neoplastic). Nervous system
• Bone marrow examination for sea blue histiocytes • Cerebellar cortex: severe degeneration: extensive loss of
(Niemann–Pick disease type C): if vertical gaze palsy, Purkinje cells.
epileptic seizures, extrapyramidal signs, dementia. • Brain and spinal cord white matter: vascular
abnormalities, like the mucocutaneous ones, are scattered
DIAGNOSIS diffusely in a few cases.
• Progressive limb and gait ataxia developing before the • Substantia nigra and locus ceruleus: loss of pigmented
age of 25 years. cells may be present, and cytoplasmic inclusions (Lewy
• Absent deep tendon reflexes. bodies) may be present in the cells that remain.
• Electrophysiologic evidence of axonal sensory neuropathy. • Anterior horn cells: loss at all levels of the spinal cord.
• Point mutation in X25, or unstable GAA trinucleotide • Posterior columns: loss of myelinated fibers.
expansion in the first X25 intron, on the proximal long • Spinocerebellar tracts: loss of myelinated fibers.
arm of chromosome 9 (9q13-q21.1). • Sympathetic ganglia cells: degeneration.
• Dorsal nerve root ganglion neurons: intranuclear
TREATMENT inclusions and bizarre nuclear formations may be found
No specific treatment is available to slow the progression of in the satellite cells (amphicytes).
disease. • Posterior nerve roots: degeneration.
• Peripheral nerves: loss of myelinated fibers.
Symptomatic treatment
• Spasticity (e.g. baclofen, botulinum toxin). Thymus
• Ataxia (e.g. clonazepam). Hypoplasia.
• Physiotherapy.
• Occupational therapy. Lymph nodes
• Podiatry. Loss of follicles.
• Speech therapy.
• Social work. ETIOLOGY AND PATHOPHYSIOLOGY
• Autosomal recessive inheritance.
Cardiologic consultation • Defective gene: ATM, mapped to chromosome 11q22-
• Hypertrophic cardiomyopathy. 23, which encodes a protein belonging to the
• Pulmonary function. superfamily of phosphatidylinositol-3´ kinases.
• Breaks in chromosome 14 and translocations.
Orthopedic spinal surgery (Harrington rod) • Decreased synthesis of immunoglobulins.
For scoliosis. • Defective repair of DNA.
• ATM homozygotes are hypersensitive to ionizing
Genetic testing radiation and radiomimetic drugs.
Testing of members of the family of an affected person with
vitamin E deficiency may identify couples heterozygous for CLINICAL FEATURES
the α-TTP mutation, which in turn will lead to the early 1–2 years of age
identification of any homozygous children, so that vitamin Onset with the acquisition of walking; ataxic-dyskinetic
E therapy can be initiated before the onset of ataxia. syndrome: awkward, unsteady gait.

PROGNOSIS 4–5 years of age


• Progressive deterioration. • Telangiectases: subpapillary venous plexuses, most
• Most patients are unable to walk independently and evident in the outer parts of the bulbar conjunctivae
safely >5–10 years after the onset of symptoms, so almost (565, 566), over the ears, on exposed parts of the neck
all are confined to a wheelchair by their late 20s. (567, 568), on the bridge of the nose and cheeks in a
• Death usually occurs 10–25 years after symptoms onset (in butterfly pattern, and in the flexor creases of the forearms
40s and 50s), usually due to cardio-pulmonary complications. (569, 570).
Ataxia Telangiectasia (Louis–Bar Syndrome) 445

565 566

565, 566 Conjunctival telangiectases, most evident in the outer parts of the bulbar conjunctivae, in a young man with ataxia telangiectasia.

567 568

567, 568 Telangiectases on exposed parts of the neck and back.

569 570

569, 570 Telangiectases in the flexor crease of the right forearm.

• Ocular pursuit: jerky due to interruption by saccadic


intrusions.
• Saccades: slow and long latency.
• Apraxia for voluntary horizontal gaze (the head, not the
eyes, turn on attempting to look to the side).
• Loss of optokinetic nystagmus.
• Limb ataxia and dysarthric speech.
• Choreoathetosis.
• Grimacing.
446 Degenerative Diseases of the Nervous System

9–10 years of age INVESTIGATIONS


• Mild intellectual decline. • Lymphopenia.
• Mild polyneuropathy: hyporeflexia. • Immunoglobulins: IgA, IgE and isotypes, IgG2, IgG4:
• Growth retardation. reduced or absent.
• Failure of delayed hypersensitivity reactions.
10–20 years of age • Abnormal humoral and cell-mediated immunity.
• Progressive decline. • Alpha fetoprotein: elevated serum levels in almost all patients.
• Premature ageing. • CT or MRI brain scan may demonstrate cerebellar
• Recurrent pulmonary and sinus infections due to atrophy, and occasionally intracranial vascular malform-
immunologic abnormalities. ations may be present.
• Death due to intercurrent bronchopulmonary infection
or neoplasia, usually lymphoma, less often glioma. DIAGNOSIS
• Clinical features consistent with diagnosis.
DIFFERENTIAL DIAGNOSIS • Abnormal humoral and cell-mediated immunity.
• Metachromatic leukodystrophy: reduced aryl sulfatase A • Raised serum levels of alpha fetoprotein.
in urine.
• Neuroaxonal dystrophy (degeneration): characteristic TREATMENT
spheroids within axons upon electron microscopy of skin • Supportive.
and conjunctival nerves. • Prophylaxis of recurrent infections due to immune defect.
• Niemann–Pick disease: reduced sphingomyelinase in
leukocytes and cultured fibroblasts. PROGNOSIS
• GM1 gangliosidosis: deficiency of β-galactosidase activity • Poor.
in leukocytes and cultured fibroblasts. • Median age at death is 20 years.
• Neuronal ceroid lipofuscinosis: inclusions (translucent
vacuoles) in lymphocytes and azurophilic granules in
neutrophils.
• Abeta-lipoproteinemia (Basses–Kornzweig acanthocyto-
sis): thorny red blood cells (acanthocytes), reduced
serum low density lipoproteins.
• Friedreich’s ataxia.
Further Reading 447

Mohs RC, Doody RS, Morris JC, et al. (2001) A Van Swieten JC, Stevens M, Ross SM, et al.
FURTHER READING 1-year placebo-controlled preservation of func- (1999) Phenotypic variation in hereditary fron-
tion survival study of donepezil in AD patients. totemporal dementia with tau mutations. Ann.
Neurology, 57: 481–488. Neurol., 46: 617–626.
ALZHEIMER’S DISEASE Mulnard RA, Cotman CW, Kawas C, et al. (2000)
Estrogen replacement therapy for treatment of HUNTINGTON’S DISEASE
Pathogenesis
mild to moderate Alzheimer disease. A ran- Bachoud-Lévi A–C, Rémy P, Nguyen J-P, et al.
Breteler MM (2000) Vascular risk factors for
domised controlled trial. JAMA, 283: (2000) Motor and cognitive improvements in
Alzheimer’s disease: an epidemiological per-
1007–1015. patients with Huntington’s disease after neural
spective. Neurobiol. Aging, 21: 153–160.
Oken BS, Storzbach DM, Kaye JA (1998) The ef- transplantation. Lancet, 356: 1975–1979.
Craddock N, Lendon C (1998) New susceptibili-
ficacy of Ginkgo biloba on cognitive function in Grove VE Jr, Quintanilla J, DeVaney GT (2000)
ty gene for Alzheimer’s disease on chromosome
Alzheimer’s disease. Arch. Neurol., 55: Improvement of Huntington’s disease with
12. Lancet, 352: 1720–1721.
1409–1415. olanzapine and valproate. N. Engl. J. Med.,
Martin JB (1999) Molecular basis of the neu-
Rockwood K, Mintzer J, Truyen L, et al. (2001) 343: 973–974.
rodegenerative disorders. N. Engl. J. Med.,
Effects of a flexible galatamine dose in Gutekunst C-A, Norflus F, Hersch SM (2000)
340: 1970–1980.
Alzheimer’s disease: a randomised, controlled Recent advances in Huntington’s disease. Curr.
Myers AJ, Goate AM (2001) The genetics of late-
trial. J. Neurol. Neurosurg. Psychiatry, 71: Opin. Neurol., 13: 445–450.
onset Alzheimer’s disease. Curr. Opin. Neurol.,
589–595. Hayden MR (2000) Predictive testing for Hunt-
14: 433–440.
Rösler M, Anand R, Cicin-Sain A, et al. (1999) ington’s disease: the calm after the storm.
Selkoe DJ (2000) The origins of Alzheimer’s dis-
Efficacy and safety of rivastigmine in patients Lancet, 356: 1944–1945.
ease. A is for amyloid. JAMA, 283:
with Alzheimer’s disease: international ran- Kuemmerle S, Gutekunst C-A, Klein A, et al.
1615–1617.
domised controlled trial. BMJ, 318: 633–640. (1999) Huntington aggregates may not predict
Terry RD (2000) Where in the brain does
Wilcock GK, Lilienfeld S, Gaens E, on behalf of neuronal death in Huntington’s disease. Ann.
Alzheimer’s disease begin? Ann. Neurol., 47:
the International-1 Study Group (2000) Effi- Neurol., 46: 842–849.
421.
cacy and safety of galantamine in patients with Lindvall O, Björklund A (2000) First step towards
Classification mild to moderate Alzheimer’s disease. Multi- cell therapy for Huntington’s disease. Lancet,
Neary D (1999) Classification of the dementias. centre randomised controlled trial. BMJ, 321: 356: 1945–1946.
Rev. Clin. Gerontology, 9: 55–64. 1445–1449. Meiser B, Dunn S (2000) Psychological impact of
genetic testing for Huntington’s disease: An
Diagnosis update of the literature. J. Neurol. Neurosurg.
Dufouil C, Clayton D, Brayne C, et al. (2000) Driving Psychiatry, 69: 574–578.
Population norms for the MMSE in the very Dubinsky RM, Stein AC, Lyons K (2000) Practice
old: estimates based on longitudinal data. Neu- parameter: risk of driving and Alzheimer’s dis- PARKINSON’S DISEASE
rology, 55: 1609–1619. ease (an evidence-based review). Neurology, 54: General
Mathuranath PS, Nestor PJ, Berrios GE, et al. 2205–2211. Schapira AHV (1999) Parkinson’s disease. BMJ,
(2000) A brief cognitive test battery to differen- 318: 311–314.
FRONTOTEMPORAL DEMENTIA
tiate alzheimer’s disease and frontotemporal de- ASSOCIATED WITH MUTATION IN Epidemiology
mentia, Neurology, 55: 1613–1620 TAU (PICK’S DISEASE, LOBAR ATROPHY) Schrag A, Ben-Shlomo Y, Quinn NP (2000) Cross
Perry RJ, Hodges JR (2000) Differentiating General sectional prevalence survey of idiopathic Parkin-
frontal and temporal variant frontotemporal de- Lippa CF, Zhukareva V, Kawari T, et al. (2000) son’s disease and parkinsonism in London.
mentia from Alzheimer’s disease. Neurology, Frontotemporal dementia with novel tau BMJ, 321: 21–22.
54: 2277–2284. pathology and a GLU342 VAL tau mutation.
Richards SS, Hendrie HC (1999) Diagnosis, man- Ann. Neurol., 48: 850–858. Etiology
agement, and treatment of Alzheimer disease. Pasquier F, Delacourte A (1998) Non-Alzheimer Gerlach M, Koutsilieri E, Riederer P (1998) N-
Arch. Intern. Med., 159: 789–798. degenerative dementias. Curr. Opin. Neurol., methyl-(R)-salsolinol and its relevance to
Rossor MN, Fox NC (2000) Mere forgetfulness 11: 417–427. Parkinson’s disease. Lancet, 351: 850–851.
or early Alzheimer’s disease? Ann. Neurol., 47: Pickering-Brown S, Baker M, Yen S-H, et al. Jarman P, Wood N (1999) Parkinson’s disease ge-
419–420. (2000) Pick’s disease is associated with muta- netics comes of age. BMJ, 318: 1641–1642.
Snowden JS (1999) Neuropsychological evaluation tions in the tau gene. Ann. Neurol., 48: Mizuno Y, Hattori N, Mori H, et al. (2001)
and the diagnosis and differential diagnosis of 859–867. Parkin and Parkinson’s disease. Curr. Opin.
dementia. Rev. Clin. Gerontology, 9: 65–72. Rossor MN (2001) Pick’s disease: a clinical Neurol., 14: 477–482.
overview. Neurology, 56 (Suppl 4): S3–S5. Sveinbjörnsdóttir S, Hicks AA, Jónsson T, et al..
Treatment
(2000) Familial aggregation of Parkinson’s dis-
Birks JS, Grimley Evans J, Iakovidou V, Tsolaki M Pathogenesis ease in Iceland. N. Engl. J. Med., 343:
(2001) Rivastigmine for Alzheimer’s disease. In Delacourte A, Buée L (2000) Tau pathology: a 1765–1770.
Cochrane library, 4. Oxford: Update software. marker of neurodegenerative disorders. Curr. Terreni L, Calabrese E, Calella AM, et al. (2001)
Brauner DJ, Muir JC, Sachs GA (2000) Treating Opin. Neurol., 13: 371–376. New mutation (R42P) of the Parkin gene in
nondementia illnesses in patients with demen- Heutink P, Stevens M, Rizzu P, et al. (1997) the ubiquitinlike domain associated with
tia. JAMA, 283: 3230–3235. Hereditary frontotemporal dementia is linked Parkinsonism. Neurology, 56: 463–466.
Brodaty H, Ames D, Boundy KL, et al. (2001) to chromosome 17q21-q22: A genetic and
Pharmacological treatment of cognitive deficits clinicopathological study of three Dutch fami- Pathology
in Alzheimer’s disease. MJA, 175: 324–329. lies. Ann. Neurol., 41: 150–159. Porritt MJ, Batchelor PE, Hughes AJ, et al.
Dinusson J, Knopman D (2000) Pharmacoeco- Hutton M (2000) ‘Missing’ tau mutation identi- (2000) New dopaminergic neurons in Parkin-
nomics of Alzheimer’s disease. The Neurologist, fied. Ann. Neurol., 46: 417–418. son’s disease striatum. Lancet, 356: 44–45.
6: 116–125. Porkaj P, Bird TD, Wijsman E, et al. (1998) Tau
Emilien G, Beyreuther K, Masters CL, Maloteaux Diagnosis
is a candidate gene for chromosome 17 fron-
J-M (2000) Prospects of pharmacological in- Clarke CE, Davies P (2000) Systematic review of
totemporal dementia. Ann. Neurol., 43:
tervention in Alzheimer disease. Arch. Neurol., acute levodopa and apomorphine challenge
815–825. [Erratum, Ann. Neurol., 1998; 44:
57: 454–459. tests in the diagnosis of idiopathic Parkinson’s
428.]
Flicker L (1999) Acetylcholinesterase inhibitors disease. J. Neurol. Neurosurg. Psychiatry, 69:
Wilhelmsen KC (1997) Frontotemporal dementia
for Alzheimer’s disease. BMJ, 318: 615–616. 590–594.
is on the MAPt. Ann. Neurol., 41: 139–140.
Henderson VW, Paganini-Hill A, Miller BL, et al. Gelb DJ, Oliver E, Gilman S (1999) Diagnostic
(2000) Estrogen for Alzheimer’s disease in Clinical criteria for Parkinson disease. Arch. Neurol., 56:
women. Randomised, double-blind, placebo- Hodges JR (2001) Frontotemporal dementia 33–39.
controlled trial. Neurology, 54: 295–301. (Pick’s disease): clinical features and assessment. Jankovic J, Rajput AH, McDermott MP, et al.
Levey AI (2000) Immunization for Alzheimer’s Neurology, 56 (Suppl 4): S6–S10. (2000) The evolution of diagnosis in early
disease: a shot in the arm or a whiff? Ann. Neu- Mathuranath PS, Xuereb JH, Bak T, Hodges J Parkinson disease. Arch. Neurol., 57: 369–372.
rol., 48: 553–555. (2000) Corticobasal ganglionic degeneration
Mayeux R, Sano M (1999) Treatment of and/or frontotemporal dementia? A report of
Alzheimer’s disease. N. Engl. J. Med., 341: two overlap cases and review of the literature.
1670–1679. J. Neurol. Neurosurg. Psychiatry, 68: 304–312.
448 Degenerative Diseases of the Nervous System

Clinical Rascol O, Brooks DJ, Korczyn AD, et al. (2000) Tu PH, Galvin JE, Baba M, et al. (1998) Glial cy-
Korczyn AD (2001) Hallucinations in Parkinson’s A five-year study of the incidence of dyskinesia toplasmic inclusions in white matter oligoden-
disease. Lancet, 358: 1031–1032. in patients with early Parkinson’s disease who drocytes of multiple system atrophy brains con-
Winikates J, Jankovic J (1999) Clinical correlates were treated with ropinirole or levodopa. N. taining insoluble alpha-synuclein. Ann. Neurol.,
of vascular parkinsonism. Arch. Neurol., 56: Engl. J. Med., 342: 1484–1491. 44: 415–422.
98–102.
DIFFUSE LEWY BODY DISEASE AUTOSOMAL DOMINANT CEREBELLAR
Treatment Galasko D (1999) A clinical approach to demen- ATAXIAS
De Bie RMA, de Haan RJ, Nijssen PCG, et al. tia with Lewy bodies. The Neurologist, 5: Devos D, Schraen-Maschke S, Vuillaume I, et al.
(1999) Unilateral pallidotomy in Parkinson’s 247–257. (2001) Clinical features and genetic analysis of
disease: a randomized, single-blind, multicen- McKeith I, Del Ser T, Spano PF, et al. (2000) Ef- a new form of spinocerebellar ataxia. Neurolo-
tre trial. Lancet, 354: 1665–1669. ficacy of rivastigmine in dementia with Lewy gy, 56: 234–238.
The Deep-brain Stimulation for Parkinson’s Dis- bodies: a randomised, double-blind, placebo- Durr A, Brice A (2000) Clinical and genetic as-
ease Study Group (2001) Deep-brain stimula- controlled international study. Lancet, 356: pects of spinocerebellar degeneration. Curr.
tion of the subthalamic nucleus or the pars in- 2031–2036. Opin. Neurol., 13: 407–413.
terna of the globus pallidus in Parkinson’s dis- McKeith IG, Galasko D, Kosaka K, et al., for the Evidente VGH, Gwinn-Hardy KA, Caviness JN,
ease. N. Engl. J. Med., 345: 956–963. Consortium on Dementia with Lewy Bodies Gilman S (2000) Hereditary ataxias. Mayo
Freed CR, Greene PE, Breeze RE, et al. (2001) (1996) Consensus guidelines for the clinical Clin. Proc., 75: 475–490.
Transplantation of embryonic dopamine neu- and pathologic diagnosis of dementia with Gosalakkal JA (2001) Ataxias of childhood. The
rons for severe Parkinson’s disease. N. Engl. J. Lewy bodies (DLB): Report of the consortium Neurologist, 7: 300–306.
Med., 344: 710–719. on DLB international workshop. Neurology, Hurko O (1997) Recent advances in heritable
Friedman JH, Fernandez HH (2000) The non- 47: 1113–1124. ataxias. Ann. Neurol., 41: 4–6.
motor problems of Parkinson’s disease. The McKeith IG, O’Brien JT, Ballard C (1999) Diag- Jen J (2001) Atana and calcium channels. What a
Neurologist 6: 18–27. nosing dementia with Lewy bodies. Lancet, headache? Arch. Neurol., 58: 179–180.
Hely MA, Fung VSC, Morris JGL (2000) Treat- 354: 1227–1228. Nagaoka U, Takashima M, Ishikawa K, et al.
ment of Parkinson’s disease. J. Clin. Neurosci., McKeith IG, Perry EK, Perry RH, for the Con- (2000) A gene on SCA4 locus causes domi-
7(6): 484–494. sortium on Dementia with Lewy Bodies (1999) nantly inherited pure cerebellar ataxia. Neurol-
Kieburtz K, Hubble J (2000) Benefits of comt in- Report of the second dementia with Lewy body ogy, 54: 1971–1975
hibitors in levodopa-treated Parkinsonian pa- international workshop. Diagnosis and treat-
tients. Results of clinical trials. Neurology, 55 ment. Neurology, 53: 902–905. FRIEDREICH’S ATAXIA
(Suppl 4): S42–S45. Evidente VGH, Gwinn-Hardy KA, Caviness JN,
Marsh L, Dawson TM (2000) Treatment of early PROGRESSIVE SUPRANUCLEAR PALSY Gilman S (2000) Hereditary ataxias. Mayo
Parkinson’s disease. BMJ, 321: 1–2. Daniele A, Moro W, Bentivoglio AR (1999) Clin. Proc., 75: 475–490.
Munchau A, Bhatia KP (2000) Pharmacological Zolpidem in progressive supranuclear palsy. N. Klockgether T (2000) Recent advances in degen-
treatment of Parkinson’s disease. Postgrad. Med. Engl. J. Med., 341: 543–544. erative ataxias. Curr. Opin. Neurol., 13:
J., 76: 602–610. Golbe LI (2000) Progressive supranuclear palsy in 451–455.
Parkinson Study Group (2000) Pramipexole vs the molecular age. Lancet, 356: 870–871. Pandolfo M (1999) Molecular pathogenesis of
levodopa as initial treatment for Parkinson Dis- Morris HR, Wood NW, Lees AJ (1999) Progres- Friedreich’s ataxia. Arch. Neurol., 56:
ease. JAMA, 284: 1931–1938. sive supranuclear palsy (Steele-Richardson-Ol- 1201–1208.
Quinn N (1999) Progress in functional neuro- szewski disease). Postgrad. Med. J., 75: Sherer T, Greenamyre JT (2000) A therapeutic
surgery for Parkinson’s disease. Lancet, 354: 579–584. target and biomarker in Friedreich’s ataxia.
1658–1659. Schrag A, Ben-Shlomo Y, Quinn NP (1999) Neurology, 55: 1600–1601.
Schuurman PR, Bosch A, Bossuyt PMM, et al. Prevalence of progressive supranuclear palsy
and multiple system atrophy: a cross-sectional ATAXIA TELANGIECTASIA
(2000) A comparison of continuous thalamic Lewis RF, Lederman HM, Crawford TO (1999)
stimulation and thalamotomy for suppression study. Lancet, 354: 1771–1775.
Ocular motor abnormalities in ataxia telangiec-
of severe tremor. N. Engl. J. Med., 342: MULTIPLE SYSTEM ATROPHY tasia. Ann. Neurol., 46: 287–295.
461–468. Adams RD, Van Bogaert L, Van der Eecken H.
Tanner CM (2000) Dopamine antagonists in early (1964) Striato-nigral degeneration. J. Neu-
therapy for Parkinson Disease. Promise and ropathol. Exp. Neurol., 23: 584–608.
Problems. JAMA, 284: 1971–1973. Ben-Shlomo Y, Wenning GK, Tison F, Quinn NP
Complications (1997) Survival of patients with pathologically
Agid Y, Chase T, Marsden D (1998) Adverse re- proven multiple system atrophy: A meta-analy-
actions to levodopa: drug toxicity or progres- sis. Neurology, 48: 384–393.
sion of disease? Lancet, 351: 851–852. Consensus Committee of the American Auto-
Allain H, Schuck S, Mauduit N (2000) Depres- nomic Society and the American Academy of
sion in Parkinson’s disease. BMJ, 320: Neurology (1996) Consensus statement on the
1287–1288. definition of orthostatic hypotension, pure au-
Chaudhuri KR, Clough C (1998) Subcutaneous tonomic failure, and multiple system atrophy.
apomorphine in Parkinson’s disease. BMJ, 316: Neurology, 46: 1470.
641. Gilman S, Quinn NP (1996) The relationship of
Donnan PT, Steinke DT, Stubbings C, et al. multiple system atrophy to sporadic olivopon-
(2001) Selegiline and mortality in subjects with tocerebellar atrophy and other forms of late-
Parkinson’s disease. A longitudinal community onset cerebellar atrophy. Neurology, 46:
study. Neurology, 55: 1785–1789. 1197–1199.
Fahn S (2000) The spectrum of levodopa-induced Graham JG, Oppenheimer DR (1969) Orthosta-
dyskinesias. Ann. Neurol., 47 (Suppl 1): tic hypotension and nicotine sensitivity in a case
S2–S11. of multiple system atrophy. J. Neurol. Neuro-
Ferreira JJ, Rascol O (2000) Prevention and ther- surg. Psychiatry, 32: 28–34.
apeutic strategies for levodopa-induced dyski- Rehman HU (2001) Multiple system atrophy.
nesias in Parkinson’s disease. Curr. Opin. Neu- Postgrad. Med. J., 77: 379–382.
rol., 13: 431–436. Schrag A, Ben-Shlomo Y, Quinn NP (1999)
The Parkinson Study Group (1999) Low-dose Prevalence of progressive supranuclear palsy
clozapine for the treatment of drug-induced and multiple system atrophy: a cross-sectional
psychosis in Parkinson’s disease. N. Engl. J. study. Lancet, 354: 1771–1775.
Med., 340: 757–763. Shy GM, Drager GA (1960) A neurologic syn-
Rascol O (2000) Medical treatment of levodopa- drome associated with orthostatic hypotension.
induced dyskinesias. Ann. Neurol., 47 (Suppl Arch. Neurol., 2: 511–527.
1): S179–S188.
Chapter Fifteen 449

Acquired Metabolic
Diseases of the
Nervous System
HYPOXIC ENCEPHALOPATHY 571

DEFINITION
Brain dysfunction caused by a lack of oxygen to the brain as
a result of failure of the circulation or respiration.

EPIDEMIOLOGY
• Incidence: uncommon.
• Age: middle-aged and elderly.
• Gender: either sex.

PATHOLOGY
Acute, global brain hypoxia (571–573)
• Laminar cortical necrosis (571): extensive, multifocal or
diffuse, and almost invariably involving the hippocampus. 572
• Scattered small areas of infarction or neuronal loss may
be present in the deep forebrain nuclei, hypothalamus or
brainstem. Sometimes, relatively selective thalamic
necrosis occurs.
• Most, if not all, of the gray matter of the cerebral,
cerebellar and brainstem structures, and even the spinal
cord is severely damaged if the hypoxia is severe enough
and prolonged.

573

571 Section of the cerebral cortex showing laminar necrosis due to


cerebral hypoxia (arrow). (Courtesy of Professor BA Kakulas, Royal Perth
Hospital,Western Australia.)

572 Hypoxic neurons in the brain (arrows): swollen, more eosinophilic,


indistinct outline, faint-staining nuclei, and loss of Nissl substance.

573 Hypoxic, eosinophilic purkinje cells in the cerebellum (arrows).


450 Acquired Metabolic Diseases of the Nervous System

Delayed post-anoxic encephalopathy • The most vulnerable neurons to mild hypoxia are the
• Demyelination throughout the subcortical white matter pyramidal neurons in the CA1 and CA4 zones of the hip-
of the cerebral hemispheres. pocampus, followed by neurons in the cerebellum, striatum
• Necrosis of the globus pallidus. and neocortex. Consequently, cerebral anoxia manifests itself
as clinical features of dysfunction of these neurons (i.e.
N.B. The above features can occur in any patient with amnesia, bradykinesia, rigidity, dystonia, choreoathetosis,
hypoxia and are not predictors of delayed anoxic and bilateral corticospinal tract involvement).
encephalopathy.
CLINICAL FEATURES
ETIOLOGY Mild hypoxia (e.g. mountaineering, chronic low level
Reduced oxygen saturation of hemoglobin carbon monoxide poisoning)
Suffocation • Inattentiveness, irritability, fatigue, headache, nausea and
• Aspiration of blood or vomitus. mild confusion.
• Compression of the trachea by hemorrhage or a surgical • Poor judgement.
pack. • Motor incoordination.
• Obstruction of the trachea by a foreign body.
• Drowning. No lasting clinical effects other than slight decline in visual
• Status asthmaticus. and verbal long term memory and mild aphasic errors in some
• Strangulation. people. Degrees of hypoxia that at no time abolish consciousness
• Altitude sickness. rarely, if ever, cause permanent damage to the nervous system.

Carbon monoxide poisoning (574) Moderate hypoxia


Carbon monoxide binds to hemoglobin 200 times more After a short period of coma, consciousness is regained and
avidly than oxygen: some patients pass quickly through an acute post-hypoxic
• Car exhaust fumes. phase characterized by variable degrees of confusion, visual
• Fumes from incomplete combustion of fossil fuels, agnosia, or any one of several types of movement disorder (e.g.
particularly in poorly ventilated heating systems. action or intention myoclonus, extrapyramidal rigidity, choreo-
• Household gas where natural gas is not used. athetosis) and proceed to make a complete recovery. Others,
• Metabolic conversion of methylene chloride that is found however, are left with permanent neurologic sequelae of
in paint strippers. various combinations of one of the post-hypoxic syndromes:
• Persistent coma or stupor (see below).
Respiratory paralysis • Visual agnosia.
• Guillain–Barré syndrome. • Dementia with or without extrapyramidal signs.
• Poliomyelitis. • Extrapyramidal (parkinsonian) syndrome with or without
• Diffuse CNS disease (trauma, vascular disease, meta- cognitive impairment.
bolic/toxic encephalopathy, including drug overdose). • Choreoathetosis.
• Cerebellar ataxia.
Reduced cerebral perfusion • Action or intention myoclonus.
Shock • Korsakoff amnesic state.
• Cardiac arrest during inhalation or spinal anesthesia. • Seizures may or may not persist.
• Myocardial infarction.
• Massive systemic hemorrhage.
• Septicemia. 574
Delayed post-anoxic demyelination
Relative arylsulfatase A predisposes susceptible individuals
to delayed post-hypoxic leukoencephalopathy and implicates
lactic acidosis in its pathogenesis.

PATHOPHYSIOLOGY
• Although the brain only weighs 1300–1400 g (46–49 oz)
(2% of total adult body weight), it consumes about 20%
of the total oxygen consumption of the body at rest.
• If the brain is deprived of any oxygen for longer than
about 5 minutes, some brain cells become permanently
damaged.
• The persistence and severity of the neurologic damage
reflect the site of ischemia, the duration and severity of
the anoxic insult and the metabolic demands of the cells
(e.g. hypothermia lowers metabolism and prolongs the 574 Cross section of the brain in the coronal plane at post mortem
tolerable period of hypoxia). showing a diffuse pink coloration due to vasodilatation induced by
• The brain cells which are most vulnerable to hypoxia are cerebral anoxia in a patient who died from carbon monoxide
neurons, followed in order of decreasing sensitivity by poisoning. (Courtesy of Professor BA Kakulas, Royal Perth Hospital,
oligodendroglia, astrocytes, and endothelial cells. Western Australia.)
Hypoxic Encephalopathy 451

Severe but not sustained hypoxia SPECIAL FORMS


Initially patients are comatose with eyes slightly divergent Delayed post-anoxic encephalopathy
and motionless, pupils reactive, limbs inert and flaccid or • Encephalopathy usually manifesting 1–4 weeks after
rigid, and tendon reflexes that are diminished. acute anoxic exposure, though the interval may be as
Within a few minutes of restoring cardiac action and short as 1 day and as late as 7 weeks.
breathing, generalized convulsions and muscle twitches • Epidemiology: rare; middle-aged and elderly are usually
(myoclonus) may occur. Coma and decerebrate postures affected.
persist, or the latter occur in response to painful stimuli, and • Etiology: most commonly reported with carbon
bilateral extensor plantar responses can be elicited. monoxide exposure.
After about 1 week or more, the eyes open, initially in • Pathogenesis: uncertain. Pseudo-deficiency of arylsulfa-
response to pain and later spontaneously, the eyelids blink tase A, and reversible alteration of CSF γ-aminobutyric
in response to any threat to the eye, and the eyes rove acid (GABA) and dopamine concentrations following
around inattentively. The patient is unaware of the recovery, may have pathogenic implications.
surroundings and may manifest chewing movements and • Pathology: characteristic subcortical white matter
grinding of teeth, and groan and grunt. Swallowing is demyelination with necrosis of globus pallidus.
possible. Body posture may be decorticate or decerebrate. • Clinical: most patients have severe anoxia with deep
Primitive reflexes such as pouting and sucking reflexes, grasp coma when found but regain consciousness within
reflex, and withdrawal reflexes to pain are present. Plantar 24 hours and resume full activity in 4–5 days. One to
responses are extensor. The individual may survive in this four weeks later, the clinical triad develops of mental
vegetative state (mute, unresponsive, and unaware of the deterioration (apathy, confusion, irritability, and occa-
environment) for weeks, months or years (see p.52). sionally agitation or mania), sphincteric incontinence and
gait disturbances consequent to frontal lobe and basal
Vegetative state ganglia dysfunction. Examination reveals dementia,
• No awareness of self or environment and an inability to pseudo-bulbar palsy and parkinsonism, and other frontal
interact with others. lobe signs of grasp reflex, glabella tap and retropulsion,
• No sustained, reproducible, purposeful, or voluntary and extrapyramidal signs of short steppage gait, masked
behavioral responses to visual, auditory, tactile, or face and rigidity.
noxious stimuli. • Differential diagnosis: many cases are mistakenly
• No language comprehension or expression. diagnosed as suffering from psychiatric disturbances.
• Intermittent wakefulness manifested by sleep–wake • Imaging: CT and MRI show bilaterally symmetric
cycles. extensive hemispheric subcortical demyelination.
• Sufficiently preserved hypothalamic and brainstem Symmetric lesions in the region of the globus pallidus
autonomic functions to permit survival with medical and suggest a poor outcome.
nursing care. • Treatment and prognosis: no specific therapy known but
• Bowel and bladder incontinence. the majority (50–75%) recover spontaneously. Some
• Variable preserved cranial nerve (pupillary, oculocephalic, patients experience persisting late sequelae including
corneal, vestibulo-ocular, gag), and spinal reflexes. memory disturbances and parkinsonism; in others the
neurologic syndrome progresses with weakness, shuffling
Severe and sustained hypoxia (e.g. cardiac arrest) gait, diffuse rigidity and spasticity, incontinence, coma
Brain death (see p.51) and death after 1–2 weeks; and rarely, the hypoxic
A clinical diagnosis of irreversible loss of function of the episode is followed by a slow deterioration over weeks to
brain when the cause is known, characterized by: months until the patient is mute, rigid, and helpless.
• Coma: unaware and unresponsiveness to pain in all
extremities. DIFFERENTIAL DIAGNOSIS
• Absence of brainstem reflexes: • Anesthesia.
– Pupils: midposition (4 mm [0.2 in]) to dilated (9 mm • Drug intoxication.
[0.4 in]); no response to bright light. • Hypothermia.
– Ocular movement: no oculocephalic reflex; no deviation • Psychiatric disturbance.
of the eyes within 1 minute of irrigating each ear with
50 ml cold water (wait at least 5 minutes between testing INVESTIGATIONS
on each side). Blood
– Facial sensation and motor response: no corneal reflex; Carboxyhemoglobin level (<10% is not symptomatic).
no jaw reflex; no grimacing to deep pressure on nail bed,
supraorbital ridge, or temperomandibular joint. EEG
– Pharyngeal and tracheal reflexes: no response after May be normal or show a spectrum of diffuse generalized
stimulation of posterior pharynx with tongue blade; no polymorphic theta or delta activity which may be of very low
cough response to bronchial suctioning. voltage and not attenuated by sensory stimulation. The
• Apnea. EEG may become isoelectric. Epileptiform activity is
unusual.
Cardiac action and blood pressure can be maintained but
natural respiration cannot be sustained. The EEG shows no
electric activity.
452 Acquired Metabolic Diseases of the Nervous System

CT brain HEPATIC ENCEPHALOPATHY (HE)


• May show diffuse white matter low density, low density
in the globus pallidus, and parenchymal atrophy.
• Carbon monoxide poisoning produces a similar picture DEFINITION
but with more basal ganglia involvement, seen as A clinical neuropsychiatric syndrome characterized by
symmetric low density. abnormal mental status occurring in patients with severe
acute, subacute, or chronic hepatocellular insufficiency.
MRI brain
• Increased signal throughout the white matter on T2W CLASSIFICATION
imaging and PDW imaging. Porto-systemic encephalopathy
• Carbon monoxide poisoning produces a similar picture Encephalopathy associated with increased porto-systemic
but with more basal ganglia involvement, seen as shunting in the absence of unequivocal evidence of
symmetric increased signal in the globus pallidus (575). hepatocellular insufficiency.

DIAGNOSIS Subclinical hepatic encephalopathy


Hypoxic encephalopathy Abnormal brain function, as detected by psychometric or
A history of a hypoxic episode with evidence of a cardiac electrophysiologic tests, but a normal ‘routine’ neurologic
arrest, sustained blood pressure below 9.3 kPa (70 mmHg) examination, in a patient with chronic liver disease (e.g.
systolic, reduced oxygenation of arterial blood, or carbon cirrhosis). The abnormal brain function is reversible with
monoxide poisoning (cherry red color of skin) and the treatment for hepatic encephalopathy.
typical clinical sequence of events as outlined above aids the
diagnosis. Fulminant hepatic failure and subfulminant
(or late-onset) hepatic failure
Carbon monoxide poisoning Acute liver failure complicated by hepatic encephalopathy
The diagnosis is made by measuring the blood within one to several weeks of the development of jaundice
carboxyhemoglobin level (<10% is not symptomatic) in or the first evidence of liver disease.
patients with any of the above symptoms from chronic
headache to coma, if unexplained. EPIDEMIOLOGY
• Prevalence: increasingly common disorder in most major
TREATMENT hospitals since the advent of many new hepatotoxic drugs
• Prevention of a critical degree of hypoxic injury. and the increased consumption of alcohol by patients
• Carbon monoxide poisoning: hyperbaric oxygen may with pre-existing hepatic dysfunction. The prevalence of
restore consciousness more rapidly and completely than subclinical HE in cirrhosis patients ranges from 30–84%.
normobaric oxygen. • Age: usually middle-aged and elderly.
• Treat seizures as appropriate, if they occur (see Epilepsy, • Gender: M=F.
pp.65, 74).

PROGNOSIS
• Degrees of hypoxia that at no time abolish consciousness 575
rarely, if ever, cause permanent damage to the nervous
system.
• Hypoxic patients who demonstrate intact brainstem
function (as indicated by normal pupillary light and
ciliospinal responses and intact reflex (doll’s-head) eye
movements with the oculocephalic maneuver and
oculovestibular reflexes) have a good prognosis for
recovery of consciousness and perhaps all of their
faculties. On the other hand, absence of brainstem
reflexes, particularly absence of pupillary response to
light, implies a hopeless prognosis.
• The presence of deep coma with fixed dilated pupils and
paralysis of eye movement for 24–48 hours, along with
absence of motor responses to painful stimuli and
marked slowing of the EEG, usually signify irreversible
brain damage (in the absence of intoxication).

575 PDW axial MRI through the basal ganglia showing increased signal
(arrows) typical of carbon monoxide poisoning. (Courtesy of Dr R Sellar,
Department of Neuroradiology,Western General Hospital, Edinburgh, UK.)
Hepatic Encephalopathy (HE) 453

PATHOLOGY Drugs and toxins


Porto-systemic encephalopathy associated with • Acetaminophen (paracetamol) overdose.
chronic liver disease • Poisoning with carbon tetrachloride, natural products
• Large Alzheimer type 2 astrocytes (astrocytes containing such as certain mushrooms or medicinal herbs, and the
characteristic glycogen inclusions with periodic-acid recreational drug methylenedioxymethamphetamine
Schiff [PAS] staining) are the cardinal neuropathologic (‘ecstasy’).
feature of porto-systemic encephalopathy. • Hypersensitivity reactions to drugs (e.g. halothane,
• Astrocytic rather than neuronal changes: diffuse increase antidepressants, antithyroid drugs, non-steroidal anti-
in the number and size of the protoplasmic astrocytes in inflammatory drugs, and antituberculous drugs).
the deep layers of the cerebral cortex, lenticular nucleus,
thalamus, substantia nigra, cerebellar cortex, and red, Ischemia
dentate, and pontine nuclei, with little or no visible • Cardiogenic shock.
alteration in the nerve cells or other parenchymal • Acute occlusion of suprahepatic veins.
elements. The degree of this glial abnormality is roughly
proportional to the intensity and duration of the Miscellaneous
neurologic disorder. • Wilson’s disease, sometimes associated with hemolytic
• Reduced number of pallidal D2 post-synaptic receptors anemia.
by about 50%. • Reye’s syndrome.
• Increased 5-hydroxytryptophan receptors in the • Acute fatty liver of pregnancy.
hippocampus.
PATHOGENESIS
Acute fulminant liver failure Unclear but it is hepatic insufficiency and not the cause of
• Cytotoxic brain edema (blood–brain barrier intact), the liver disease which is important in causing the
raised intracranial pressure, herniation. neuropsychiatric clinical features.
• Loss of non-NMDA receptors in the brain.

ETIOLOGY
• Acute hepatic encephalopathy in a patient with liver
disease (e.g. cirrhosis) is usually associated with a clearly
identifiable precipitating factor and usually resolves when
the precipitating factor is removed or corrected. Failure Table 42 Precipitating factors for hepatic encephalopathy
to find a precipitating factor may imply a decrease in
overall hepatocellular function. Precipitant Possible mechanism
• Chronic hepatic encephalopathy usually occurs in a
patient with cirrhosis and substantial porto-systemic Dietary protein excess Increased ammonia
shunting, who has hepatic encephalopathy that is Constipation (anorexia, production
persistent or episodic, with or without complete fluid restriction)
resolution of encephalopathy between episodes. Gastrointestinal hemorrhage
Infection
Porto-systemic venous shunting associated with Uremia
chronic liver disease Hypokalemia
• Predisposing factors: hypoxia, hypokalemia, metabolic
alkalosis, electrolyte depletion, excessive diuresis, and use Systemic alkalosis Increased diffusion of ammonia
of sedative-hypnotic drugs. across blood–brain barrier
• Precipitating factors (see Table 42).
Dehydration Reduced metabolism of toxins
Acute liver failure Hypotension because of hepatic hypoxia
Infection Hypoxemia
• Hepatitis viruses: most commonly hepatitis B virus, but Anemia
also hepatitis D and E viruses, and rarely hepatitis A, C
and G viruses. Benzodiazepine use Activation of central GABA-
• Non-hepatotropic viruses: herpes virus, varicella-zoster benzodiazepine receptors
virus, Epstein–Barr virus, cytomegalovirus, paramyxo-
virus, and others. Other psychoactive Compounding of CNS
drug use depressant effect

Porto-systemic shunts Reduced hepatic metabolism of


toxins because of diversion of
portal blood

Progressive hepatic Reduced hepatic metabolism of


parenchymal damage toxins because of reduced
Development of hepatoma functional reserve
454 Acquired Metabolic Diseases of the Nervous System

Possible mechanisms Abnormal mental status


• Accumulation in brain extracellular fluid of • Mild restlessness, irritability and perhaps dysarthria are
unmetabolized gut-derived neurotoxins (e.g. ammonia, seen initially. In some patients the syndrome does not
see below) as a result of poor hepatic function and porto- evolve beyond the stage of mild mental dullness and
systemic shunting. confusion, with asterixis and EEG changes. These
• Functional disturbance of the blood–brain barrier. relatively mild forms must be differentiated from other
• Alteration in neurotransmission: acute confusional states. In other patients with an
– Decreased neuroexcitation (glutamate, aspartate, extensive porto-systemic collateral circulation, a disorder
dopamine, catecholamines). of mood, personality and intellect may be protracted over
– Increased neuroinhibition (GABA, endogenous many months or even years, and be associated with
benzodiazepines, serotonin). dietary protein intake.
– Activation of central GABA-benzodiazepine receptors by • Confusion, clouding of the sensorium, drowsiness,
ligands of endogenous origin. constructional apraxia, ataxia, bradykinesia, rigidity, and
– Production of false neurotransmitters. asterixis follow, and then coma with extensor plantar
• Reduced energy metabolism in the brain: responses bilaterally.
– Disturbed activity of Na+/K+-ATPase.
– Decreased activity of urea-cycle enzymes (due to zinc Asterixis
deficiency). Elicited by having the patient extend his or her arms, with
• Deposition of manganese in the basal ganglia. dorsiflexion of the wrist, and watching for the characteristic
‘flap’ due to an inability to maintain the posture; a form of
Neurotoxins negative myoclonus. If the patient is comatose, the flap
Ammonia response can be elicited by pressing on the finger tips,
• In patients with hepatic failure, ammonia (which is causing dorsiflexion of the hand. If the patient can only
produced in the gut by the action of colonic bacteria and grasp the examiner’s fingers, asterixis can be demonstrated
mucosal enzymes on dietary protein) is not completely by the involuntary clenching and unclenching of the fist.
metabolized to urea through the urea cycle so it enters
the systemic circulation and the brain. Signs of advanced liver disease
• Ammonia uptake in the brain is increased, where it is Jaundice, fetor hepaticus, hepatomegaly (or small liver size),
normally detoxified in the brain by astrocytes. and ascites (576).
• Ammonia has a direct post-synaptic effect on excitatory
and inhibitory post-synaptic potentials. Signs of comorbidities
• Expression of the astrocyte glutamate transporter gene, Comorbidities such as alcoholism, electrolyte disturbance,
GLT-1, is reduced in acute liver failure resulting in hypoglycemia, renal failure, hypotension due to
reduced uptake of glutamate into synaptosomes gastrointestinal bleeding (e.g. boundary zone infarction),
(astrocytes in particular) and a rise in glutamate in the infection, and hemostatic failure may complicate or mask
extracellular fluid. the clinical features of hepatic encephalopathy, and
• Excessive ammonia reacts, in the presence of glutamine development of metabolic or infectious complications.
synthetase, with the excess glutamic acid (glutamate) in
the extracellular fluid to form glutamine. DIFFERENTIAL DIAGNOSIS
• Ammonia also affects cerebral energy metabolism by Metabolic encephalopathy
inhibiting alpha ketoglutarate dehydrogenase and • Hypoglycemia or hyperglycemia.
combining with alpha ketoglutarate in the citric acid • Electrolyte imbalance: hypernatremia, hyponatremia.
cycle to form glutamic acid, which then combines with • Hypoxia.
more ammonia to form more glutamine. The effect of • Hypercapnia: carbon dioxide narcosis.
ammonia on glutamine function also has an effect on • Uremia: renal failure.
tryptophan uptake by the brain. • Ketoacidosis.
• Wilson’s disease.
Manganese • Liver transplantation may be complicated within the first
Excess (2–7 fold) manganese is deposited in the pallidum in few weeks by an encephalopathy (headache, confusion,
patients dying in hepatic coma. seizures, cortical blindness, coma) due to electrolyte
imbalance, hypocalcemia, hypomagnesemia, cyclosporin,
CLINICAL FEATURES air embolism, intracerebral hemorrhage and hypotension.
The nature and severity of the symptoms depend on the Opportunistic infections may occur later.
acuteness and extent of the liver failure and on the • Hyperammonemia: other causes include ornithine
development of metabolic or infectious complications. transcarbamylase deficiency, amino and organic acidurias,
Consequently, the clinical manifestations range from subtle arginosuccinic aciduria, citrullinemia, argininemia,
abnormalities detectable only on psychometric testing to carbamylphosphate synthetase deficiency, lysinuric
deep coma. protein intolerance, Reye’s syndrome, sodium valproate,
systemic carnitine deficiency, asparaginase toxicity.
Hepatic Encephalopathy (HE) 455

Toxic encephalopathy INVESTIGATIONS


• Alcohol: Blood
– Acute intoxication. • Liver function tests: hyperbilirubinemia, high
– Withdrawal syndrome: delirium tremens (rapid postural aminotransferase levels.
and action tremor, cortical excitation rather than cortical • Plasma ammonia: elevated usually but not consistently.
inhibition). Hyperammonemia is not invariable because blood
– Wernicke–Korsakoff syndrome. ammonia levels depend on both the rate of production
• Sedative/hypnotic/psychoactive drug intoxication. and metabolism.
• Salicylates. • Coagulation factors: very low values.
• Heavy metals. • Viral serology.

Intracranial lesions Differential diagnosis


• Intracranial hemorrhage: subdural hematoma. • Urea and electrolytes.
• Meningo-encephalitis: e.g. herpes simplex encephalitis. • Glucose: hypoglycemia.
• Creutzfeldt–Jakob disease. • Alcohol: acute intoxication.
• Brain infarction. • Erythrocyte transketolase activity: Wernicke–Korsakoff
• Brain abscess. syndrome.
• Brain tumor. • Toxicology: salicylates, heavy metals, psychoactive drugs.
• Head trauma.
• Epilepsy (e.g. non-convulsive status epilepticus) or EEG
postictal encephalopathy. • Initially the EEG shows non-specific generalized slowing
of the background rhythm.
Neuropsychiatric disorders • Later, it becomes dominated by trains of high amplitude,
Dementia/pseudodepression: in some patients with an bisynchronous, slow waves of delta frequency (1.5–3 Hz)
extensive porto-systemic collateral circulation, a disorder of or frontal-dominant, triphasic waves with a phase lag
mood, personality and intellect may be protracted over from front to back, before flattening in late coma (577).
many months or even years, and be associated with dietary These abnormalities are characteristic of, but not specific
protein intake. for, the disorder, since other metabolic encephalopathies
can cause similar abnormalities.

CT brain
To exclude a structural intracranial lesion if suspected (e.g.
hemorrhage).

576 577

576 Abdomen of a man with hepatic encephalopathy and portal 577 EEG, 8 channels, showing trains of medium to high amplitude
hypertension showing diminished body hair and dilated superficial bisynchronous, frontal-dominant, triphasic waves with a frequency of
abdominal veins (arrow) shunting blood from the portal to the systemic 1.5–2.5 Hz and phase lag from front to back.Triphasic waves are often
circulation. Ascites was also present. seen in patients with an acute deterioration in conscious state due to
metabolic disorders such as hepatic or renal failure, or cerebral anoxia.
Atypical triphasic waves may rarely be seen in herpes simplex
encephalitis, drug withdrawal, Creutzfeldt–Jakob disease, or following
head trauma.
456 Acquired Metabolic Diseases of the Nervous System

MRI brain by preventing surges in intracranial pressure related to


• Increased signal in the basal ganglia (particularly pallidum psychomotor agitation. The cerebral edema is treated with
but midbrain may also be affected) on T1WI (578, 579). mannitol given in repeated bolus injections of 0.5 g/kg body
• The substances which may cause an increased signal on weight over a period of 10 minutes, and acetylcysteine can
T1WI include manganese, melanin, calcium, and blood. be given by continuous infusion to improve cerebral blood
The differential diagnosis of increased signal in the basal flow and metabolic rate for oxygen in grade 4 patients.
ganglia on T1WI includes hyper-, hypo-, pseudohypo- and Hepatic encephalopathy due to the much more common
pseudopseudohypoparathyroidism (calcium), hyper- chronic liver disease can be managed quite differently
alimentation, Hallevorden–Spatz disease, carbon because it is usually due to a clinically apparent precipitating
monoxide poisoning, hemorrhage and neurofibromatosis. event (or combination of events), or to the spontaneous
development of porto-systemic shunting, and because
CSF cerebral edema is much less frequent.
Usually normal apart from elevated levels of glutamine, the
end product of cerebral ammonia metabolism. General
• Nasogastric tube.
Abdominal ultrasound • Central venous line.
Size and echostructure of liver. • Indwelling urinary catheter.
• Monitor intracranial pressure (extradural monitor) if
Liver biopsy (transjugular route) encephalopathy reaches grades 3 or 4.
• Avoid maneuvers that increase intracranial pressure (such
as sensorial stimuli).
DIAGNOSIS (Table 43) • Control restlessness.
There are two components to making the diagnosis: • Hydrate carefully.
• To determine that encephalopathy (subclinical or overt) • Elevate patient’s head 20–30°.
is present. • Avoid (or use with extreme caution) drugs such as
• To establish hepatocellular insufficiency and increased sedatives, phenytoin, phenobarbitone (phenobarbital),
portal-systemic shunting. valproate, and dantrolene because drug metabolism and
protein binding are frequently disturbed in liver disease.
The diagnosis is based on the history, physical • Avoid/be careful with benzodiazepines because plasma
examination and laboratory findings. half-life of benzodiazepines is increased four- to fivefold
in hepatic failure, so substantial toxicity may ensue (e.g.
TREATMENT when used for endoscopy or nocturnal sedation).
Treatment is determined by the underlying cause. • Avoid hypoglycemia (monitor blood glucose).
Hepatic encephalopathy due to the rare condition of
acute liver failure is rapid in onset and progression, almost Specific
always complicated by cerebral edema in the later stages, and • Remove the underlying cause:
has a poor prognosis. Orthotopic liver transplantation should – N-acetylcysteine for early paracetamol overdose.
therefore be considered in these patients and meanwhile – Forced diuresis and activated charcoal for mushroom
those who are grade 3 or 4 should be electively sedated with poisoning.
fentanyl, paralysed with atracurium, and ventilated to protect – Acyclovir for herpes virus infection.
the airway and facilitate the management of cerebral edema – Surgery for acute hepatic-vein occlusion.

578 579 578, 579 T1W axial


(578) and T1W sagittal
(579) MRI of the brain
in a patient with hepatic
encephalopathy. Note
the increased signal in
the basal ganglia
(arrows). (Courtesy
of Dr R Gibson, Depart-
ment of Neuroradiology,
Western General
Hospital, Edinburgh, UK.)
Central Pontine Myelinosis 457

• Lower blood ammonia levels: CENTRAL PONTINE MYELINOLYSIS


– Reduce ammonia production (restrict intake of dietary
protein and inhibit urease-producing colonic bacteria).
Low protein diet (20 g a day initially, increasing gradually DEFINITION
later). A disorder characterized by rapidly developing quadriparesis
Vegetable, rather than animal, protein diet. and a large, symmetric, demyelinating lesion of the greater
Carbohydrate enemas. part of the basis pontis. It may also affect extrapontine brain
Oral non-absorbable disaccharides: lactulose or lactisol areas.
(lactose 100 g daily in lactase deficiency): 30–60 g/day
to produce 2–4 soft stools daily. EPIDEMIOLOGY
Oral antibiotics: neomycin 6 g daily, metronidazole • Prevalence: up to 0.25% of autopsies.
800 mg daily, or rifaximin 1200 mg daily. • Age: any age; reported in children (particularly those
Oral Enterococcus faecium. with severe burns) as well as adults.
– Increase ammonia metabolism: sodium benzoate 10 g • Gender: M=F.
daily; ornithine aspartate 9 g tds; phenylacetate.
• For patients who have previously been treated with PATHOLOGY
benzodiazepines (e.g. for treatment of behavioral Demyelination occurs in the centre of the pons (580) and
disturbance caused by hepatic encephalopathy), the occasionally other parts of the brainstem.
administration of a benzodiazepine antagonist, such as
flumazenil, may improve conscious level.
• Consider pharmacologic manipulation of glutaminergic
and monoaminergic systems (experimental).
• If intracranial pressure exceeds 2.7–4 kPa
(20–30 mmHg), apply hyperventilation (paCO2
3.3–4 kPa [25–30 mmHg]), then mannitol 0.5 g/kg in
bolus; then hemodialysis or venovenous hemofiltration;
then barbiturate coma. 580
• Orthotopic liver transplantation: acute liver failure;
severe, refractory hepatic encephalopathy.

CLINICAL COURSE AND PROGNOSIS


• The encephalopathy usually evolves over a period of days
to weeks and, if untreated, often terminates fatally.
Sometimes the symptoms spontaneously regress
completely or partially and may fluctuate in severity for
several weeks or months.
• Most manifestations are reversible with medical treatment.
• Some patients have progressive debilitating neurologic
syndromes such as dementia, spastic paraparesis,
cerebellar degeneration, and extrapyramidal movement
disorders that are associated with structural abnormalities
of the CNS and were hitherto regarded as irreversible, 580 Transverse section of the pons showing demyelination of
but now may gradually improve after successful corticospinal tract fibers in the center of the basis pontis (arrows)
orthotopic liver transplantation. with preserved neurons in the pontine nuclei and no evidence of
inflammation.

Table 43 Grading system based on clinical and EEG features


Grade Conscious level Personality and intellect Neurologic signs EEG abnormalities
0 Normal Normal None None
Subclinical Normal Normal Only on psychometric testing None
1 Inverted sleep Forgetfulness, mild confusion, Tremor, apraxia, incoordination, Triphasic waves
pattern, restlessness agitation, irritability impaired handwriting
2 Lethargy, slow Disorientation in time, Asterixis, dysarthria, ataxia, Triphasic waves
responses amnesia, disinhibited, hypoactive reflexes
inappropriate behavior
3 Somnolent but Disorientated in place, Asterixis, rigidity, hyperactive Triphasic waves
rousable, confusion aggressive behavior reflexes, Babinski signs
4 Coma None Decerebration Delta activity
458 Acquired Metabolic Diseases of the Nervous System

Macroscopic • Dehydration and electrolyte disturbances particularly


• Grayish discoloration and fine granularity in the centre hyponatremia.
of the basis pontis. • Acute hemorrhagic pancreatitis.
• The lesion may be only a few millimetres in diameter or • Pellagra.
it may occupy almost the entire basis pontis.
• The lesion may extend posteriorly to involve the medial Variable clinical illness
lemnisci and, in most advanced cases, other tegmental • Subacute onset over several days, usually after correction
structures as well; superiorly to encroach on the of hyponatremia, of:
midbrain; inferiorly to the pontomedullary junction; and – Mental changes ranging from mild confusion to coma.
anteriorly, but there is always a rim of intact myelin – Pseudobulbar palsy.
between the lesion and the surface of the pons. – Flaccid or spastic quadriparesis.
• Particularly extensive pontine lesions may be associated – Extensor plantar responses bilaterally.
with identical myelinolytic foci symmetrically distributed – Locked-in syndrome (see p.55): a de-efferented state
in the thalamus, subthalamic nucleus, striatum, internal whereby the patient cannot speak or move the limbs but
capsule, amygdaloid nuclei, lateral geniculate body, white can often move the eyes (particularly in the vertical
matter of the cerebellar foliae, and deep layers of the plane) and blink, in order to try and communicate.
cerebral cortex and subjacent white matter (‘extrapontine Awareness and consciousness is preserved because the
myelinolysis’). brainstem tegmentum, including the reticular formation,
are spared.
Microscopic – Conjugate eye movements may be limited and
• Destruction of medullated sheaths throughout the lesion, nystagmus may be present.
with relative sparing of the axis cylinders and intactness – Pupillary reflexes, eye movements, corneal reflexes, and
of the nerve cells of the pontine nuclei. These changes facial sensation are spared.
always begin and are most severe in the geometric centre • Absent or transient mild neurologic symptoms and signs
of the lesion, where they may proceed to frank necrosis may be all that are seen if the pontine lesion is small,
of tissue. extending only 2–3 mm on either side of the median
• Reactive phagocytes and glial cells are present through- raphe and involving only a small portion of the
out the demyelinative focus, but no oligodendrocytes or corticopontine or pontocerebellar fibers.
signs of inflammation are present. • Coma may be present due to underlying metabolic or
other associated disease which obscures the presence of
ETIOLOGY AND PATHOPHYSIOLOGY central pontine myelinolysis.
• Sporadic.
• Specific regions of the brain, such as the centres of the Variants
basis pontis, have a special susceptibility to some acute • Confusion, stupor, and cerebellar ataxia, without
metabolic fault (possibly to rapid correction or to over- corticospinal tract signs, due to brainstem tegmentum
correction of hyponatremia, and possibly hyper- and cerebellar lesions.
osmolality), analogous perhaps to the selective vul- • ‘Locked-in’ syndrome due to large symmetric lesions of
nerability of the corpus callosum and anterior com- the frontal cortex and underlying white matter.
missure in Marchiafava–Bignami disease.
• Myelinolysis is more likely to occur after the treatment DIFFERENTIAL DIAGNOSIS
of chronic rather than acute hyponatremia and is more Stroke due to brainstem infarction
likely to occur with a rapid rate of correction. • Sudden onset or step-like progression.
• The exact pathogenesis has not been determined. • History of vascular risk factors (atheroma) or neck
trauma (dissection).
Risk factors • Asymmetry of long tract signs.
• Hyponatremia. • Caused by basilar artery occlusion.
• Over-rapid correction of severe hyponatremia • More extensive involvement of tegmental structures of
(<130 mmol/l [<130 mEq/l]) to normal or higher than pons as well as midbrain and thalamus.
normal levels of serum sodium.
• Extreme serum hyperosmolality. Multiple sclerosis
• Nutritional deficiency. Massive pontine demyelination in acute or chronic relapsing
• Serious, often life-threatening, medical illness. MS may rarely produce a pure basis pontis syndrome but
other features of this disease provide the clue to the correct
CLINICAL FEATURES diagnosis (see p.340).
Predisposing illness
Patients are usually already very ill: Post-infectious encephalomyelitis (see p.292)
• Vomiting or comatose chronic alcoholics.
• Cachectic patients from a variety of causes. Hyponatremic encephalopathy
• Malnourished patients. A generalized encephalopathy without localizing motor
• Chronic renal failure treated by dialysis. signs. Usually there is a distinct interval between the
• Hepatic failure. occurrence of a generalized hyponatremic encephalopathy
• Advanced lymphoma. initially which improves with elevation of the sodium level
• Carcinoma. and then, about 2–3 days after hyponatremia is corrected,
• Severe bacterial infection. the neurologic syndrome of myelinolysis evolves. However,
Further Reading 459

sometimes the hyponatremic encephalopathy does not TREATMENT


improve before focal myelinolytic symptoms emerge and it • Supportive (be aware that ‘locked-in’ patients can see,
can be difficult to recognize that two separate disease hear and feel everything [including pain and itch] but are
processes are occurring sequentially. incredibly frustrated that they cannot move).
• Active treatment of the underlying medical illness.
Psychiatric illness • Prevention of complications of immobility: pneumonia,
The common initial symptoms of myelinolysis (mutism, deep vein thrombosis, contractures, urinary tract
dysarthria, lethargy and affective changes) may be mistaken infection.
for psychiatric/psychologic illness. • Rehabilitation: physiotherapy, swallowing and speech
therapy, occupational therapy, psychologic support and
INVESTIGATIONS therapy.
• Full blood count: infection.
• Urea and electrolytes: hyponatremia, renal failure. PREVENTION
• Liver function tests: chronic alcoholism, liver failure. • Optimal management of hyponatremia patients involves
• Amylase: pancreatitis. weighing the risk for illness and death from untreated
• CT brain scan: there may be low density throughout the hyponatremia against the risk for myelinolysis due to
pons, basal ganglia, hemispheric white matter, without correction of hyponatremia.
mass effect or enhancement. • Chronic hyponatremia should be corrected by no more
• MRI brain scan: more sensitive than CT. Increased signal than 10 mmol/l (10 mEq/l) in any 24-hour period.
is visible in the pons with a thin rim of normal signal
around the margins of the pons and possible sparing of CLINICAL COURSE AND PROGNOSIS
the corticospinal tracts which leaves two rounded normal • Outcome varies: some die; many make a gradual but
signal areas within the high signal of the pons. There may partial recovery with residual sequelae that include bulbar
also be increased signal in the thalami, putamen, caudate dysfunction, spastic quadriparesis, movement disorders,
nuclei, amygdala, and hemispheric white matter. behavioral changes, and alterations in cognition, while
• Brainstem auditory evoked potentials. others recover completely.
• Some patients remain in a state of mutism and paralysis
DIAGNOSIS with relatively intact sensation and comprehension, i.e.
• Clinical: gravely ill patient with a general medical disease the ‘locked-in’ syndrome (see p.515).
who develops a quadriplegia, pseudobulbar palsy and
locked-in syndrome over a few days.
• Radiologic: lesion in the center of the basis pontis
consistent with demyelination.

FURTHER READING HEPATIC ENCEPHALOPATHY (HE) CENTRAL PONTINE MYELINOLYSIS


Jones EA, Weissenborn K (1997) Neurology and Chalela J, Kattah J (1999) Catatonia due to cen-
the liver. J. Neurol. Neurosurg. Psychiatry, 63: tral pontine and extrapontine myelinolysis:
279–293. case report. J. Neurol. Neurosurg. Psychiatry,
HYPOXIC ENCEPHALOPATHY Jalan R, Hayes PC (1997) Hepatic encephalopa- 67: 692–693.
Chalela JA, Wolf RL, Maldjian JA, et al. (2001) thy and ascites. Lancet, 350: 1309–1315. Karp BI, Laureno R (2000) Central pontine and
MRI identification of early white matter injury Mas A, Rodes J (1997) Fulminant hepatic failure. extrapontine myelinolysis after correction of
in anoxic-ischaemic encephalopathy. Neurolo- Lancet, 349: 1081–1085. hyponatraemia. The Neurologist, 6: 255–266.
gy, 56: 481–485. Lui HF, Jalan R, Hayes PC (1998) Hepatic en- Laureno R, Karp BI (1997) Myelinolysis after cor-
Gottfried JA, Mayer SA, Shungu DC, et al. cephalopathy: pathogenesis, diagnosis and rection of hyponatraemia. Ann. Intern. Med.,
(1997) Delayed posthypoxic demyelination. management. Proc. R. Coll. Physicians Edin., 126: 57–62.
Association with arylsulfatase A deficiency and 28: 111–118.
lactic acidosis on proton MR spectroscopy. Riordan SM, Williams R (1997) Treatment of he-
Neurology, 49: 1400–1404. patic encephalopathy. N. Engl. J. Med., 337:
Snyder BD, Tabbaa MA (1988) Assessment and 473–479.
treatment of neurological dysfunction after
cardiac arrest. Stroke, 19: 269–273.
460 Chapter Sixteen

Nutritional Deficiency
and the
Nervous System
WERNICKE–KORSAKOFF SYNDROME Wernicke’s disease), but there is greater cell loss in the
anterior principal nucleus of the thalamus in Korsakoff ’s
psychosis.
DEFINITION
Wernicke’s disease (thiamine deficient encephalopathy) is a ETIOLOGY
disorder characterized by rather abrupt onset of any Thiamine deficiency
combination of nystagmus, gait ataxia, conjugate gaze palsy • Chronic alcoholism (malnutrition and impaired activity
and mental confusion in association with nutritional of thiamine-dependent enzymes).
deficiency, especially due to alcoholism. • Malnutrition.
Korsakoff’s psychosis is a mental disorder, also associated • Deliberate starvation:
with alcoholism and malnutrition, in which retentive – Anorexia nervosa.
memory is enormously impaired due to a defect in learning – Hunger strike.
and memory, in an otherwise responsive patient. The – Obesity treatment (vertical banded gastroplasty).
Wernicke–Korsakoff syndrome is a symptom complex • Intractable vomiting:
comprising the manifestations of both Wernicke’s disease – Gastric carcinoma.
and the Korsakoff amnesic state. – Pyloric obstruction.
– Hyperemesis gravidarum.
EPIDEMIOLOGY • Prolonged intravenous feeding.
• Prevalence (from autopsy studies): 0.8–2.8% of autopsies; • Renal dialysis.
15% in psychiatric in-patients, 24% in homeless men. • Acquired immunodeficiency syndrome.
• Age: adults.
• Gender: M>F. Magnesium depletion
Magnesium is an essential cofactor in the conversion of
PATHOLOGY thiamine into active diphosphate and triphosphate esters:
Wernicke’s disease • Diuretics: frusemide (furosemide).
Macroscopic • Alcohol.
Loss of brain tissue (lower brain weight, increased • Hyperemesis gravidarum.
ventricular volume and pericerebral space) occurs, mainly in • Diarrhea: Crohn’s disease, gluten enteropathy.
white matter.
PATHOGENESIS
Microscopic • Absorption of thiamine is impaired by both malnutrition
• Neuronal loss, proliferative symmetric vasculopathy, and and alcohol.
gliosis occur in the mamillary bodies (medial mamillary • Liver disease that leads to reduced body stores and
nucleus, diagnostic); thalamus (medial dorsal nucleus); impaired metabolism of thiamine often compounds the
hypothalamus; around the third ventricle, aqueduct in situation and may also enhance the toxic effect of alcohol
the mid brain and floor of the fourth ventricle; brainstem on the brain. Nevertheless, about a third of substantial
and sometimes the cerebellum (581–583). There is little alcohol abusers seem to be resistant to developing the
or no specific pathology in the locus ceruleus, Wernicke–Korsakoff syndrome.
hippocampus and cerebral cortex. • Individual differences in thiamine enzyme systems, such
• Acute Wernicke’s disease is characterized by small areas as different levels of affinity between thiamine
of petechial hemorrhage and marked vascular endothelial pyrophosphate, which acts as coenzyme, and trans-
proliferation in the same sites listed above (i.e. mamillary ketolase, an enzyme concerned with brain glucose
bodies and so on). metabolism, may predispose certain individuals to the
Wernicke–Korsakoff syndrome. If so, they are likely to
Korsakoff’s psychosis be extragenetic as there is little evidence of an inborn
Cell loss and gliosis occur in the medial mammillary nucleus transketolase abnormality.
and mediodorsal nucleus of the thalamus (similar to
Wernicke–Korsakoff Syndrome 461

CLINICAL FEATURES • Retinal hemorrhages occur occasionally.


Wernicke’s disease • Papilledema rarely.
Subacute onset over hours to days of one or, more often, • Alcoholic patients: peripheral neuropathy in >80% of
various combinations of: patients, dementia, cerebellar ataxia, macrocytosis and
• Ataxia of stance and gait. abnormalities of liver function.
• Ophthalmoplegia: supranuclear horizontal and/or
vertical gaze palsies, internuclear ophthalmoplegia, and Korsakoff amnestic confabulatory state
lateral rectus palsies, often causing diplopia. (Korsakoff’s psychosis)
• Nystagmus: horizontal and/or vertical. A permanent gap in memory characterized by:
• Mental state disturbance: a global confusional state causing • Defect in learning (anterograde amnesia), due to
apathy, inattention, disorientation, minimal spontaneous defective encoding at the time of original learning rather
speech, forgetfulness, drowsiness, and even coma. than exclusively a defect in the retrieval mechanism.
• Loss of short term verbal and non-verbal memory
Other features may include: (retrograde amnesia); immediate memory is preserved
• Stupor or coma as the initial manifestation. and long term memory appears to be maintained
• Signs of alcohol withdrawal: agitation, hallucinations, through multifocal networks.
confusion, and autonomic hyperactivity. • Confabulation and confusion
• Hypothermia.
• Postural hypotension (autonomic neuropathy). The learning defect can render patients incapable of all
• Cardiovascular dysfunction (tachycardia, exertional but the most habitual tasks.
dyspnea, minor EKG abnormalities).
• Impaired capacity to discriminate between odors (in the DIFFERENTIAL DIAGNOSIS
chronic stage of the disease due to a lesion of the medial Wernicke’s disease
dorsal nucleus of the thalamus). • Other causes of confusional states and coma (see p.44).
• Pupil abnormalities: miosis and non-reacting pupils can • Hepatic encephalopathy.
occur. • Brainstem/cerebellar/diencephalic stroke.
• Ptosis.

581 582

583

581, 582 Brain, coronal sections showing petechial


hemorrhages in the mamillary bodies (arrows) and medial
dorsal thalamus (arrowheads) of a patient with acute
Wernicke’s encephalopathy.
583 Axial section through the medulla showing a proliferative
vasculopathy and petechial hemorrhages (arrow) in the floor of the
fourth ventricle of a patient with acute Wernicke’s encephalopathy.
462 Nutritional Deficiency and the Nervous System

Korsakoff amnestic confabulatory state TREATMENT


• Third ventricle tumors. • Thiamine 100 mg intravenously or Parentrovite forte,
• Temporal lobe infarction or surgical resection. 10 ml i.v. (contains 250 mg thiamine), immediately and
• Herpes simplex encephalitis. then daily for 7 days because intestinal absorption is
• Hypoxic encephalopathy. usually impaired in alcoholics. The thiamine solution
• Alzheimer’s disease. should be fresh, since it can be inactivated by heat.
Thiamine prevents the progression of the disease and
INVESTIGATIONS reverses the brain abnormalities that have not resulted in
Blood fixed structural changes. Hypomagnesemia can
Red cell transketolase (reduced markedly) and thiamine compromise the treatment response and replacement
(reduced), and plasma glucose and magnesium levels. therapy should be considered.
Transketolase is one of the enzymes of the hexose • Oral vitamin maintenance, providing 10–50 mg thiamine
monophosphate shunt and requires thiamine pyrophosphate daily until the patient is no longer at risk (usually for
as a cofactor. several months).
• Oral chelated magnesium 500 mg daily (if depleted).
CT brain scan • Balanced diet.
CT may show widening of the third ventricle and • Alcohol abstinence.
interhemispheric fissures, and other features of long • Psychotherapy and educational efforts: ineffective in
standing alcohol abuse (generalized cerebral atrophy plus arresting the cognitive decline without abstinence.
more pronounced vermian and cerebellar atrophy). • Clonidine and fluvoxamine have been reported to be
effective in improving function, suggesting dysfunction
MRI brain scan in noradrenergic and serotonergic systems respectively,
MRI may show areas of increased signal in the but remain experimental.
periaqueductal gray matter of the midbrain and medial
portions of the thalami, atrophy of the mamillary bodies, PREVENTION
and dilatation of the third ventricle. • At risk patients should be given vitamin supplements.
• Comatose patients should be treated with thiamine or
DIAGNOSIS Parentrovite forte, especially if no obvious cause is
Clinical diagnostic criteria recognized.
• Oculomotor abnormalities: ophthalmoplegia and/or • At-risk patients should never be given a glucose load
nystagmus. (intravenous dextrose, nasogastric feeding) without
• Cerebellar dysfunction. vitamin supplements because this may precipitate
• Altered mental state (e.g. confusion) or mild memory Wernicke’s syndrome.
impairment.
• Dietary deficiencies. CLINICAL COURSE
• History of alcohol consumption. After thiamine treatment, the ophthalmoplegia begins to
• Red blood cell transketolase assay, with a positive improve within hours to days and nystagmus, ataxia and
thiamine pyrophosphate effect of >40%. confusion within days to weeks. The amnesic symptoms may
• MRI brain: medial thalamic and periaqueductal lesions. recover slowly over 1 year or so and incompletely. Recovery
• Clinical response to thiamine. of cognitive function depends on age and continuous
abstinence from alcohol.
The diagnosis of Wernicke–Korsakoff syndrome is missed
in about 25% of cases if brains are not examined PROGNOSIS
microscopically. This is commonly because the history of • Depends on the stage of the disease and prompt
symptom onset may be unclear, due to recent alcohol institution of thiamine treatment.
intoxication or the presence of Korsakoff psychosis. If • Can be fatal if untreated: mortality rate is 10–20%,
clinically suspected, however, the diagnosis can be mainly due to pulmonary infection, septicemia,
confirmed by showing a marked reduction in blood decompensated liver disease, and an irreversible stage of
transketolase activity and a striking restoration toward thiamine deficiency.
normal of the transketolase and the clinical features even • Residual nystagmus and ataxia in 60% of patients.
within hours of the administration of thiamine; completely • Residual chronic memory disorder in 80% of patients.
normal values of transketolase are usually attained within 24
hours. Failure of the ocular palsies to respond to thiamine
within a few days should raise doubts about the diagnosis
of Wernicke disease.
Vitamin B12 Deficiency 463

VITAMIN B12 DEFICIENCY PATHOPHYSIOLOGY


• Vitamin B12 deficiency leads to a rise in serum levels of
homocysteine and methylmalonic acid.
DEFINITION • The lack of methionine (produced from homocysteine),
Vitamin B12 deficiency is a nutritional disorder of the leads to a lack of S-adenosylmethionine, which is
nervous system that may be characterized by a symmetric, required for all methylation reactions, including myelin
distal, predominantly sensory peripheral neuropathy due to phospholipids. It is puzzling, however, that neurologic
axonal degeneration, autonomic neuropathy, subacute complications are rare in folate deficiency, even though
combined degeneration of the spinal cord (or combined methionine synthase also requires folate as a cosubstrate.
systems disease), optic neuropathy, dementia and other • Accumulation of methylmalonate and propionate may
disturbances of higher mental function. provide abnormal substrates for fatty acid synthesis,
resulting in abnormal odd-number carbon and branched-
EPIDEMIOLOGY chain fatty acids, which may be incorporated into the
• Prevalence: myelin sheath.
– Pernicious anemia: 1–3% over age 65 years. • Nitric oxide is a potent oxidizing agent and irreversibly
– Serum vitamin B12 levels <150 pmol/l (<200 pg/ml): oxidizes the cobalt core of vitamin B12, rendering
7–16% of the elderly population. methylcobalamin inactive. This inhibits the conversion
– Serum vitamin B12 levels <225 pmol/l (<300 pg/ml) of homocysteine to methionine, thus reducing the supply
and elevated levels of metabolites of homocysteine and of S-adenosylmethionine. In people with normal stores
methylmalonic acid: 21% over age 65 years. of vitamin B12, there may sufficient quantities of
• Age: mean age at diagnosis is 60 years. unoxidized vitamin B12 to maintain enzyme function for
– Caucasians: increases in frequency with increasing age, longer periods but in patients with compromised stores,
peaking after age 65 years. even brief exposure to nitrous oxide may be sufficient to
– Hispanics and Africans: younger age of onset, particularly precipitate a vitamin B12 deficiency syndrome.
among women.
• Gender: F>M (1.5:1). ETIOLOGY
Vitamin B12 deficiency
PHYSIOLOGY • Genetic: defect of methylmalonyl CoA mutase.
• The total body store of vitamin B12 is 2–5 mg, half of • Dietary deficiency (low intake): vegetarianism.
which is stored in the liver. • Malabsorption states:
• The body needs about 1–6 µg/day (the average diet – Pernicious anemia. Lack of intrinsic factor due to
contains about 7–30 µg per day). autoimmune gastric parietal cell dysfunction.
• Vitamin B12 is released from dietary food protein in the – Gastric achlorhydria.
stomach and passes into the duodenum and jejunum – Gastric resection.
where it is bound to intrinsic factor, which is produced – Chronic pancreatic disease.
by gastric parietal cells (along with HCl). The vitamin – Competition for intraluminal vitamin B12:
B12-intrinsic factor complex attaches to specific receptors Bacterial overgrowth in ‘blind loops’, anastomoses, and
in the terminal ileal mucosa, where it is internalized and diverticula.
degraded. Vitamin B12 enters the portal circulation, Cobalamin-metabolizing fish tapeworm (Diphyllobothrium
where some binds to transcobalamin II. The vitamin B12- latum) infestation.
transcobalamin II complex is taken up by cells and – Terminal ileum disease.
degraded in lysosomes, releasing vitamin B12 for Crohn’s disease.
conversion to methylcobalamin or adenosylcobalamin. Resection.
• Methylcobalamin is a cofactor of methionine synthase, – Nitric oxide exposure (anesthesia)
which catalyses the reaction of homocysteine and • Increased requirements: pregnancy.
methyltetrahydrofolate to produce methionine and • Low utilization: enzyme deficiency.
tetrahydrofolate. Methionine is further metabolized to S-
adenosylmethionine, which is necessary for methylation
of myelin sheath products. Tetrahydrofolate is the
precursor required for purine and pyrimidine synthesis.
Adenosylcobalamin catalyses the conversion of L-
methylmalonyl CoA to succinyl CoA in the mitochondria.
• The dietary source of vitamin B12 is from animals only.
It is tightly conserved through the enterohepatic
circulation, so it takes 2–5 years to develop vitamin B12
deficiency from malabsorption, and up to 10–20 years
for dietary deficiency from strict vegetarianism.
464 Nutritional Deficiency and the Nervous System

PATHOLOGY Neurologic
• Predominantly sensory, distal, symmetric, peripheral • Altered higher mental function: depression, hallucin-
neuropathy is seen, characterized by axonal degeneration ations, frank psychosis (so-called megaloblastic madness),
with or without demyelination. paranoia, violent behavior, personality changes, mental
• Subacute combined degeneration of the spinal cord (or confusion and dementia occur in about 10% of cases.
combined systems disease) occurs with degeneration of • Optic atrophy, with bilateral centrocecal scotomas and
the white matter of the posterior columns and lateral visual failure is rare.
corticospinal tracts (584). • Spastic paraparesis due to corticospinal tract lesions
• Optic neuropathy: white matter degeneration and occurs. Lower limb weakness may also be due to
occasional foci of spongy degeneration in optic nerves accompanying peripheral neuropathy; lower motor
and chiasm. neuron signs (e.g. wasting, weakness, areflexia) only
• Brain: white matter degeneration. become apparent when neuropathy is severe.
• Autonomic neuropathy: degeneration of small unmye- • Reflexes may be increased (with clonus) or decreased
linated axons. depending on the degree of corticospinal tract and
peripheral nerve involvement.
CLINICAL FEATURES • Plantar responses are often extensor because of
About three-quarters of patients present with neurologic corticospinal tract degeneration.
symptoms and about one-quarter with non-neurologic • Incoordination may be marked because of
symptoms that result from defective DNA synthesis in spinocerebellar degeneration (cerebellar ataxia) or
rapidly dividing cells of the bone marrow and gastro- proprioceptive loss (sensory ataxia).
intestinal mucosa. • Loss of vibration and joint position sense in the feet are
seen due to degeneration of the posterior columns, and
HISTORY loss of pain and temperature sensation distally in the
Neurologic symptoms limbs in a ‘glove and stocking’ distribution due to
• Paresthesiae (tingling sensations) and numbness in the peripheral neuropathy.
feet and later the fingers. • Rhomberg’s sign may be positive due to impaired
• Lhermitte’s symptom occasionally. proprioception as a result of degeneration of large
• Impaired memory. diameter fibers in peripheral nerves and posterior
• Anosmia. columns.
• Reduced visual acuity. • Broad-based gait.
• Diminished taste.
• Impaired manual dexterity. N.B. Neuropathy alone is found in about one-quarter of
• Lower limb weakness ( corticospinal tract lesions and patients, myelopathy alone in about 12%, and combined
peripheral neuropathy). myelopathy and neuropathy in about 40%. Paresthesiae,
• Unsteady gait. vibratory loss and ataxia may be caused by both neuropathic
• Impotence. and myelopathic lesions.
• Incontinence of urine or feces.

Non-neurologic symptoms
• Weakness.
• Tiredness.
• Faints (syncope).
• Palpitations.
• Headache.
• Sore tongue.
• Diarrhea. 584
• Bowel disturbances.
• Family history of anemia requiring monthly ‘vitamin
shots’.

Recent exposure to nitric oxide

PHYSICAL EXAMINATION
General
• Pallor of mucous membrane (anemia) (585).
• Lemon-yellow skin.
• Glossitis (smooth red tongue) (586).
• Premature graying of hair.
• Orthostatic hypotension.
584 Axial section of spinal cord showing tissue destruction and
fibrous gliosis in posterior columns (arrowheads) and lateral and
anterior (arrows) corticospinal tracts of a patient with longstanding
subacute combined degeneration of the spinal cord. (Courtesy of
Professor BA Kakulas, Royal Perth Hospital,Western Australia.)
Vitamin B12 Deficiency 465

DIFFERENTIAL DIAGNOSIS Low serum vitamin B12 level


Myeloneuropathy • Vitamin B12 deficiency.
• Syphilis. • Abnormalities of vitamin B12 metabolism: folate
• Friedreich’s ataxia. deficiency/antagonists.
• Adrenomyeloneuropathy. • Normal individuals with no tissue deficiency of vitamin
• Cervical spondylitic myeloradiculopathy. B12: second and third trimester of pregnancy.
• Multiple sclerosis and concurrent peripheral neuropathy
(e.g. alcoholic, diabetic). INVESTIGATIONS
• Chronic exposure to nitric oxide. Nitric oxide interdicts Vitamin B12 deficiency or not?
vitamin B12 metabolism. • Blood:
• Acute exposure to nitric oxide in patients with – Full blood count and blood film: may be normal.
unsuspected vitamin B12 deficiency (because of normal – Anemia or pancytopenia.
vitamin B12 levels); these patients may develop an acute – Macrocytosis (raised mean corpuscular volume).
vitamin B12 deficiency syndrome after a single exposure – Neutrophil hypersegmentation (may be a sensitive
to nitric oxide during a surgical procedure. marker of vitamin B12 deficiency, even in the absence of
• Celiac disease with vitamin E deficiency: sensory anemia or macrocytosis).
neuropathy, areflexia, confusion, dementia, cerebellar • Serum vitamin B12 level: <150 pmol/l (<200 pg/ml)
ataxia, myopathy, weight loss, diarrhea, steatorrhea. (radioassay method) in 90–95%, 150–225 pmol/l
(200–300 pg/ml) in 5–10%, and >225 pmol/l
Macrocytosis (>300 pg/ml) in 0.1–1% of patients.
• Alcoholism. • Serum homocysteine and methylmalonic acid levels
• Hypothyroidism. (metabolites of the B12 pathway): raised, but non-
• Liver disease. specific: also elevated in chronic renal disease, several
• Reticulocytosis. inborn errors of metabolism, hypothyroidism, folate
• Drugs, cytotoxics. deficiency and pyridoxine deficiency.
• Pregnancy. • Bone marrow: megaloblastic (can be masked by
• Myelodysplasia, myeloma, aplastic anemia. concurrent iron deficiency).
• Inborn errors of cobalamin metabolism causing
homocystinuria and methylmalonic aciduria.

585 586

585 Pale appearance of the skin and mucous membranes of a patient


with pernicious anemia. (Courtesy of Dr AM Chancellor,Tauranga, New
Zealand.)

586 Smooth, atrophic tongue of a patient with pernicious anemia.


(Courtesy of Dr AM Chancellor,Tauranga, New Zealand.)
466 Nutritional Deficiency and the Nervous System

Cause of vitamin B12 deficiency TREATMENT


• Antiparietal cell antibody: sensitive (present in 90% of • Remove/treat the underlying cause.
people with pernicious anemia [PA]) but not specific • Replenish vitamin B12 stores with intramuscular injections
(also present in 5% of 30 year olds, 10% of the of cyanocobalamin 100–1000 µg daily for 5 days, then
population over age 70, 15% of elderly women, and 60% • Maintain vitamin B12 stores with:
of people with simple atrophic gastritis). – 100–1000 µg cyanocobalamin intramuscularly each
• Anti-intrinsic factor antibody: month, or
– Insensitive (present in only 50–60% of people with PA) – 500–1000 µg cyanocobalamin orally each day, as a
but is highly specific for PA. supplement, until the underlying cause is removed.
– If anti-intrinsic factor antibody is positive (and serum
vitamin B12 level <150 pmol/l (<200 pg/ml) or The rationale for oral supplementation (e.g. 1000 µg/day)
homocysteine and methylmalonic acid levels are is that 1% of all ingested vitamin B12 (e.g. 10 µg, more than
elevated), the diagnosis of PA is confirmed and a the average daily requirement [1–6 µg/day]) may be
Schilling test is not necessary. absorbed by passive diffusion. This means that vitamin B12
• Serum gastrin level: indicated if anti-intrinsic factor is requirements may be satisfied with oral therapy, even in
negative (and serum vitamin B12 levels <150 pmol/l patients with PA, provided the dose is sufficient. So, oral
[<200 pg/ml] or homocysteine and methylmalonic acid replacement, and even sublingual therapy, are alternatives for
levels are elevated), to help establish the presence of patients who cannot tolerate intramuscular injections or for
achlorhydria, which is almost invariably associated with whom they are impractical.
PA. The therapeutic response to B12 should be a reticulocytosis
• Schilling test: to determine (1) if there is vitamin B12 and normalization of the blood count changes (unless
malabsorption in patients with proven B12 deficiency concomitant deficiencies or diseases are present). The
without a known cause (e.g. diet, gastrectomy, drugs), hemoglobin will rise by 10 g/l per week on average. The bone
and (2) the cause of the B12 malabsorption. marrow changes also resolve rapidly, within 48 hours of
– Conducted with and without intrinsic factor (IF). commencing therapy.
– Radioactive B12 alone is given by mouth (part I). If it is
not detected in the urine, the result is abnormal PROGNOSIS
indicating that radioactive B12 is not absorbed from the The most important factor influencing the neurologic
gut. The test is repeated with radioactive B12 bound to response to treatment is the duration of symptoms before
IF (part II). Radioactive B12 is now detected in the urine treatment is started. If treatment is given early enough, it
of patients with PA, due to a lack of intrinsic factor, may not only prevent progression but also reverse some
whereas patients with malabsorption of B12 from the neurologic symptoms and signs besides paresthesia in the
terminal ileum still fail to show any radioactive B12 in the feet and optic atrophy.
urine.

Myeloneuropathy or not, and type?


• Nerve conduction studies: reduced or absent sensory
potentials (axonal degeneration) and decreased motor
conduction velocities (demyelination).
• Electromyography (EMG): spontaneous fibrillations in
distal muscles if denervated.
• Sural nerve biopsy: axonal degeneration with or without
demyelination.
• MRI of the brain and spine (587–589): may be used to
exclude structural causes of the patient’s symptoms. In
advanced cases, T1W MR images of the spine may show
swelling of the upper cervical spinal cord and decreased
signal in the posterior columns of the spinal cord,
extending up to the brainstem. T2W images may show
increased signal in the posterior columns. T2W MRI
images of the brain may show confluent high density
changes in the white matter of the tempero-parieto-
occipital lobes bilaterally corresponding to gliosis and
edema with vacuolization of axonal sheath myelin.

DIAGNOSIS
Typical neurologic syndrome (subacute combined
degeneration of the spinal cord, neuropathy), macrocytic
anemia, low serum vitamin B12 level, and biochemical
evidence of vitamin B12 deficiency: elevated methylmalonic
acid and homocysteine levels. A normal vitamin B12 assay
does not fully exclude vitamin B12 deficiency.
Further Reading 467

587 588

589
587–589 T2W axial cervical spine (587),T2W axial brain (588), and
T2W midline sagittal cervical spine (589) MRI in a patient with
pernicious anemia and subacute combined degeneration of the cord.
Note the increased signal throughout the sensory and motor long
tracts in the brainstem and the posterior columns of the cord
(arrows).

FURTHER READING Thomas PK (1998) Subacute combined degener- Treatment


ation. J. Neurol. Neurosurg. Psychiatry, 65: Delpre G, Stark P, Niv Y (1999) Sublingual ther-
807. apy for cobalamin deficiency as an alternative
to oral and parenteral cobalamin supplemen-
WERNICKE–KORSAKOFF SYNDROME Etiology tation. Lancet, 354: 740–741.
McLean J, Manchip S (1999) Wernicke’s en- Mayall M (1999) Vitamin B12 deficiency and ni- Elia M (1998) Oral or parenteral therapy for vita-
cephalopathy induced by magnesium deple- trous oxide. Lancet, 353: 1529. min B12 deficiency? Lancet, 352: 1721–1722.
tion. Lancet, 353: 1768. Rosener M, Dichgans J (1996) Severe combined Kuzminski AM, Del Giacco EJ, Allen RH, et al.
Zubaran C, Fernandes JG, Rodnight R (1997) degeneration of the spinal cord after nitrous (1998) Effective treatment of cobalamin defi-
Wernicke–Korsakoff syndrome. Postgrad. Med. oxide anaesthesia in a vegetarian. J. Neurol. ciency with oral cobalamin. Blood, 98:
J., 73: 27–31. Neurosurg. Psychiatry, 60: 354. 1191–1198.
VITAMIN B12 DEFICIENCY Diagnosis
General Green R, Kinsella LJ (1995) Current concepts in
Hemmer B, Glocker FX, Schumacher M, et al. the diagnosis of cobalamin deficiency. Neurol-
(1998) Subacute combined degeneration: clin- ogy, 45: 1435–1440.
ical, electrophysiological, and magnetic reso-
nance imaging findings. J. Neurol. Neurosurg.
Psychiatry, 65: 822–827.
468 Chapter Seventeen

Disorders of the
CSF Circulation
HYDROCEPHALUS • The primary function of the CSF is to provide
buoyancy and allow the brain to float, protecting it
from repetitive trauma whenever the head moves.
DEFINITION Movement of CSF through the foramen magnum
Hydrocephalus is defined as an increase in volume of the compensates for the changes that occur in cerebral
CSF in association with dilatation of the cerebral ventricles. blood volume with each heart beat.

EPIDEMIOLOGY ETIOLOGY
• Incidence: Overproduction of CSF
– Neonatal hydrocephalus: 1/1000 births. Rare, if ever (choroid plexus papilloma usually obstructs).
– Adult hydrocephalus: depends on the incidence of preci-
pitating causes (e.g. meningitis, subarachnoid hemorr- Obstruction to CSF outflow within the ventricles
hage); 1–2% of patients presenting to dementia clinics. (obstructive hydrocephalus)
– In the UK: 3000 shunt operations annually (1500 new, Congenital
1500 recurrent). • Aqueduct stenosis, with or without gliosis, ‘forking’ (in
• Prevalence (lifetime): 0.1 (95% CI: 0.01–0.3) per 1000. which the aqueduct is replaced by a number of small,
• Age: any age. inefficient channels), septum formation, or atresia. May
• Gender: either sex. be caused by mumps and possibly other intra-uterine
infections.
PHYSIOLOGY OF CSF CIRCULATION • Dandy–Walker syndrome (atresia of the foramina of
• CSF contains little protein; bicarbonate is its only buffer. Luschka and Magendie associated with failure of
• The total volume of CSF in the cranial and spinal cavity development of the vermis of the cerebellum).
of the adult is about 150 ml, of which about 30 ml is in • Hind brain abnormalities, spina bifida.
the ventricular system. • Vein of Galen aneurysm.
• About 70% of CSF is actively secreted by the cells of the
choroid plexus of the lateral, third and fourth ventricles Space-occupying lesions
and the remaining 30% comes from the brain’s capillary • Acquired aqueduct stenosis.
bed and from metabolic water production. • Colloid and arachnoid cysts.
• CSF is produced at a rate of 0.35 ml/min (500 ml/day), • Thalamic glioma.
which is equivalent to a turnover of total CSF every 6 • Intraventricular tumors.
hours. • Tentorial herniation.
• The CSF flows slowly through the ventricular system and • Posterior fossa tumor, hemorrhage, infarct.
the foramina of Luschka and Magendie into the • Colloid cyst or tumor of the third ventricle.
subarachnoid space and then up and over the cerebral • Basilar artery dolichoectasia.
convexities. Some of the fluid flows into the central canal
of the spinal cord. Ventricular hemorrhage
• Circulation of the CSF is achieved by arterial pulsation • Prematurity.
in the brain and choroid plexus, and by changes in • Aneurysm, arteriovenous malformation.
posture, exercise and coughing, creating a pressure
differential between the newly formed CSF and its site Defective flow in the subarachnoid space
of drainage (a hydrostatic gradient). (communicating hydrocephalus)
• The CSF drains passively from the subarachnoid space into • Meningitis:
the venous system (principally the superior sagittal sinus) – Pyogenic.
via the arachnoid granulations in the cranial and spinal – Tuberculous.
compartments (open channels that connect the – Fungal.
subarachnoid side to the venous side of the arachnoid villi). – Neoplastic.
• Subarachnoid hemorrhage.
• Hyperviscosity of CSF due to high CSF protein (e.g.
spinal neurofibroma).
Hydrocephalus 469

Defective absorption of CSF at the arachnoid Older children and adults (skull sutures
granulations have begun to fuse)
• Congenital deficiency of arachnoid granulations (rare). Raised intracranial pressure causing
• Raised cerebral venous sinus pressure due to venous sinus • Headache, often worse in the early morning.
thrombosis (controversial). • Nausea and vomiting.
• Blurred vision, occasionally.
Idiopathic • Diplopia, due to VIth nerve palsy may occur.
Particularly in the elderly (see Normal pressure • Papilledema sometimes; its absence does not exclude
hydrocephalus, p.473). raised intracranial pressure.
• Optic atrophy with progressive visual loss and poor
Loss of brain tissue (hydrocephalus ex-vacuo) pupillary reaction to light: a late complication of chronic
Accumulation of CSF in the ventricular system and cerebral papilledema due to raised intracranial pressure.
sulci due to loss of brain tissue as a result of atrophy, • Upgaze paresis.
infarction and so on. • Increasing unsteadiness of gait occurs, culminating in
frequent falls due to a combination of ataxia, spasticity
PATHOPHYSIOLOGY and dyspraxia of gait.
Obstruction to CSF flow raises intraventricular pressure • Urgency of micturition and eventual incontinence.
which reverses the direction of flow of CSF from the • Personality change and behavioral disturbance.
ventricles through the ependyma and into the • Dementia.
periventricular brain parenchyma causing periventricular • Eventually, depressed consciousness.
edema. It also reduces periventricular cerebral blood flow
which, if not treated, results in periventricular demyelination Symptoms and signs of the primary obstructive lesion
and gliosis. For example, cerebellar signs of a posterior fossa tumor.

CLINICAL FEATURES Elderly


Infancy/early childhood Normal pressure hydrocephalus (NPH) syndrome
• Asymptomatic, or symptoms of irritability, poor appetite, This is characterized clinically by Hakim’s triad of gait
vomiting, and poor head control. apraxia, urinary incontinence and progressive dementia.
• Rapid head enlargement (crossing percentile lines) with Difficulty initiating movement, such as getting out of a
distortion of the normal proportions of the cranial cavity. chair, is characteristic (see p.473).
• Prominent scalp veins.
• Open, enlarged, tense fontanelle. DIFFERENTIAL DIAGNOSIS
• Delayed fusion of sutures. Infancy/early childhood
• ‘Cracked-pot’ tympanitic sound produced by percussion Enlarged head
of the thinned skull (late). • Achondroplasia.
• Brilliant transillumination of the skull (late sign). • Rickets.
• Delayed motor skills and milestones but alert. • Subdural hematoma.
• Papilledema (unusual in infants). • Megalencephaly (brain of excessive size and weight).
• Upward gaze and horizontal gaze paresis (‘setting sun’
eyes) occur, due to pressure of dilated third ventricle on Transillumination of the head
the posterior commissure in the tectum of the midbrain. Localized brain cysts.
• Paralysis or spasm of convergence.
• Spastic paraparesis with lower limb hyperreflexia and Elderly
extensor plantar responses: the corticospinal tracts • Alzheimer’s disease.
emanating from the parasagittal region of the motor • Vascular (multi-infarct) dementia.
cortex and destined for the legs have a long course • Binswanger’s disease.
around the lateral ventricles and tend to be most • Parkinson’s disease.
compressed and stretched by enlarged lateral ventricles. • Progressive supranuclear palsy.
• Multiple sclerosis.
N.B. Head circumference measurement should be
compared with normal values on a standard head-size chart. INVESTIGATIONS
Serial measurements which show a crossing of percentiles Imaging
are important signs. The role of imaging is to:
Transillumination of the skull with a torch held against • Exclude other causes of the patient’s symptoms.
the baby’s head in a dark room reveals increased redness • Try to decide whether the hydrocephalus is obstructive
around the torch light in the presence of an excessive (communicating or non-communicating) or non-
amount of fluid beneath the torch light. obstructive.
• Follow-up patients being managed conservatively to
check for the need for a shunt in future and in those who
have been shunted to check for shunt failure.
470 Disorders of the CSF Circulation

If obstructive non-communicating hydrocephalus is Other imaging methods


diagnosed, it may be possible to identify the cause such as • Transfontanelle ultrasonography/echo-encephalography
a third ventricular colloid cyst, posterior fossa tumor or are safe, non-invasive methods of imaging and moni-
aqueduct stenosis (590, 591). Communicating hydro- toring lateral ventricular size in infants and young
cephalus can be identified when all the ventricles appear children.
large but the basal cisterns are still patent (592–597). Non- • Skull x-ray: children <10 years: large head and widened
communicating hydrocephalus is present when the lateral ± skull sutures; older children and adults (closed skull
the third ventricle are much larger than the fourth ventricle. sutures): erosion of the dorsum sellae and clinoid
Meningitis and subarachnoid hemorrhage may cause either processes (raised ICP).
communicating or non-communicating hydrocephalus. • Radioactive cisternography, which is performed by
injecting a radiopharmaceutic into the lumbar sub-
CT and MRI brain scan arachnoid space, shows prolonged retention of the
In general, CT scan is satisfactory. In obstructive radioactive material within the ventricular system and
hydrocephalus it shows ventricular dilatation above the site impaired movement of radioactive material over the
of obstruction and possibly the causative lesion (e.g. tumor). cerebral convexities.
If there is no obstruction to CSF flow then all ventricles will
be dilated. The cortical sulci are usually compressed due to CSF
the rise in intracranial pressure. MRI is required, however, CSF pressure is usually elevated, but may be normal in so-
in particular circumstances such as the diagnosis of aqueduct called NPH (see p.473). The appearance, cell count, protein
stenosis, posterior fossa lesions, and malignant meningitis, and glucose are commonly normal, but may be abnormal in
to name but a few. For example, MRI of aqueduct stenosis the presence of an underlying causal lesion (e.g. tumor,
shows disproportionate dilatation of the lateral and third meningitis).
ventricles and a small fourth ventricle. On midline sagittal
MRI the aqueduct appears small. Bands or webs may be Intracranial pressure monitoring
visible in the aqueduct causing obstruction. Normally the A saline-filled catheter or catheter-tipped transducer inserted
CSF flows through the aqueduct fast enough to result in a into the lateral ventricle, brain or subdural space records a
flow void on MRI, so the absence of a flow void (on pulsatile pressure of 0–1.3 kPa (0–10 mmHg) relative to the
carefully chosen sequences) is good corroborative evidence foramen of Monro when the patient is lying flat. As a mass
of aqueduct stenosis. or the ventricles enlarge within the skull, the mean
Usually, the temporal horns of the lateral ventricles are the intracranial pressure rises and spontaneous periodic waves
first to dilate (this is a useful sign after subarachnoid hemorr- become more pronounced, particularly during rapid eye
hage as an early warning of possible complicating hydro- movement sleep.
cephalus). Note that the fourth ventricle is the last to dilate, Non-invasive techniques include tympanic membrane
so may appear small in communicating hydrocephalus simply displacement and transcranial doppler studies of the
because it has not yet had time to dilate. Therefore it can be pulsatility index.
very difficult to decide which sort of hydrocephalus is present
on any imaging modality in some cases.
Periventricular white matter lucencies on CT and high
signal on T2WI are common due to transependymal
seepage of CSF usually sited around the angles of the lateral
ventricles when due to hydrocephalus.

590 591 590, 591 CT brain


scan at the level of the
lateral ventricles (590)
and T1W midline
sagittal MR (591) in a
patient with aqueduct
stenosis. Note the
large ventricles
(obviously longstanding
because there is no
periventricular edema)
with typical shape
(590), the narrowed
inferior aqueduct
(591) (arrowhead)
and the small fourth
ventricle (arrow).
Hydrocephalus 471

592 593

592–594 Communicating
hydrocephalus. Cranial CT
scans showing dilatation of
the fourth ventricle
(arrowhead) and temporal
horns of the lateral ventricles
(592, arrows), and lateral
ventricles (593, arrows); and
narrowing of the cortical sulci
at the vertex of the brain
(594, arrows).

594 595
595 MRI brain scan,T1W
image, sagittal plane,
showing dilated ventricular
system without focal
obstructive lesion.

596 MRI brain scan,T2W 596 597


image, axial plane, showing
dilated lateral ventricles.

597 CT brain scan showing


marked dilatation of the
lateral ventricles in a patient
with communicating
hydrocephalus of unknown
cause.
472 Disorders of the CSF Circulation

Cognitive function testing Shunt complications


• Mini-mental state examination and other mental • Shunt failure: 80% of shunts fail within 12 years. The
screening tests are often insensitive to slight ‘subcortical’ highest risk of underdrainage is in the first year (30%),
cognitive impairment. usually due to gradual occlusion of the ventricular
• Psychometric tests are usually needed, particularly to detect catheters by choroid plexus. A CT brain scan is the best
frontal lobe dysfunction. investigation for suspected underdrainage to compare
with earlier scans.
TREATMENT • Shunt occlusion/blockage causing raised intracranial
Indication pressure.
Neurologically symptomatic, progressive ventriculomegaly. • Overdrainage: particularly in tall patients. May produce
The presence of ventriculomegaly in the absence of any slit ventricle syndrome, subdural hematomas, encysted
supporting clinical features does not mean there is active fourth ventricles or post-shunt craniosynostosis.
hydrocephalus. • Infection of the valve: 70% occur within 1 month of
insertion, often due to contamination at the time of
Treat underlying cause, if possible insertion with Staphylococcus epidermidis. Commonly
Surgically excise the primary lesion if possible (i.e. papilloma of presents as intermittent pyrexia with malaise and signs of
choroid plexus, or third ventricular or posterior fossa tumor). shunt obstruction. Occasionally there is cellulitis along
the shunt tract or signs of meningitis. Diagnose by
Symptomatic treatment aspirating CSF from the shunt reservoir. Intrathecal
Reduce CSF production vancomycin may eradicate the infection but often the
Carbonic anhydrase inhibitors (with or without entire shunt system has to be removed, followed by a
corticosteroids or diuretics). period of external ventricular drainage before a new
system can be re-inserted. The morbidity/mortality rate
Facilitate CSF drainage (ventricular decompression with is 30–40% and survivors risk neurologic deficit.
diversion of CSF flow) • Subdural hematoma.
• Transient hydrocephalus (e.g. after subarachnoid • Low pressure headache.
hemorrhage): ventricular or lumbar catheter for • Epileptic seizures.
temporary CSF diversion. • Shunt nephritis (an immunologically mediated disorder
• Non-communicating hydrocephalus with patent due to secondary Staphylococcus albus infection).
subarachnoid space: transventricular, endoscopic third • Pulmonary thromboembolism with ventriculoatrial shunts.
ventriculostomy, with puncture of the floor of the third
ventricle and drainage of CSF into the basal cisterns. PROGNOSIS
• Intraventricular or large arachnoid cysts: endoscopic • Depends on the underlying condition.
communication of cyst with normal CSF pathways. • Untreated cases of infantile hydrocephalus develop
• Infantile hydrocephalus: ventriculoperitoneal or necrosis of the scalp with leakage of CSF, infection, and
ventriculoatrial shunt (benefits about 80% of cases): CSF death. A minority survive to childhood with arrested
is drained from the frontal horn of the lateral ventricle hydrocephalus characterized by enlargement of the head,
(where there is no choroid to block the catheter) via a some degree of intellectual handicap, limb spasticity, and
ventricular catheter (a pressure valve with reservoir impaired bladder function.
mechanism and a distal catheter made of non-reactive • 70% of infants with treated non-tumoral hydrocephalus
tubing) into the peritoneal or atrial cavity. maintain a normal IQ and attend a normal school.
• Communicating hydrocephalus (some patients): Children with infantile hydrocephalus should therefore
lumboperitoneal shunt: drains fluid from the lumbar undergo a shunting procedure (because they will
subarachnoid space to the peritoneal cavity. These shunts benefit).
usually work well for a few months only and, where they
are successful, may be associated with symptomatic
secondary descent of the cerebellar tonsils. Insertion of
a separate ventricular access device at the time of shunt-
ing facilitates investigation should complications arise.
• Normal pressure hydrocephalus (see p.473):
– Removal of 50 ml of CSF may result in improvement in
gait and predict a favorable response to a shunting
procedure.
– Repeated lumbar punctures (LP), removing 15–20 ml
CSF, may give rise to transient improvement in the early
stages of the disease.
– Ventriculoperitoneal shunt may produce dramatic
improvement in some cases. Improvement in post
operative cognitive function is more likely if there was a
known cause, and short history.
Normal Pressure Hydrocephalus (NPH) 473

NORMAL PRESSURE CLINICAL FEATURES


HYDROCEPHALUS (NPH) Classic triad of:
• Slowly progressive gait disorder.
• Impairment of mental function.
DEFINITION • Urinary incontinence.
A chronic hydrodynamic hydrocephalus that occurs in adults
and is associated with a delay in the circulation or absorption Gait disturbance
of CSF and progressive neurologic deficit comprising The first, and in some cases only, apparent symptom and
Hakin’s triad of gait apraxia, urinary incontinence, and sign:
progressive dementia. First described in 1965. • Difficulty in initiating walking: feet feel ‘glued to the
floor’, also referred to as a ‘magnetic gait’ or gait apraxia.
EPIDEMIOLOGY • Postural instability.
• Prevalence: about 1 per 20 000 population. About 0–6% • A short-stepped shuffling gait in more advances stages,
of cases of dementia. often described as ‘gait apraxia’ but this term is perhaps
• Age: middle-aged and elderly. inappropriate in NPH because patients with NPH may
• Gender: M=F. execute nearly intact walking movements when minimally
supported or when lying down.
PATHOLOGY • Hyperreflexia (mainly in the lower limbs) and extensor
• Impaired CSF flow within the ventricles or in the plantar responses may be present but spasticity is not a
subarachnoid space (arachnoid villi) due to a variety of feature (tone is usually normal) and objective motor
possible causes (see below). weakness is seldom present (despite complaints of
• There is no pressure gradient between the ventricles and weakness and tiredness).
the cerebral convexity.
Causes
ETIOLOGY AND PATHOPHYSIOLOGY • Stretching or destruction of the paraventricular
Uncertain. The following are hypotheses: corticospinal fibers.
• Disconnection of basal ganglia from the frontal cortex.
Impairment of CSF flow within the ventricles or the • Uninhibited antigravity reflexes and co-contraction of
subarachnoid space agonists and antagonists during walking.
• Non-communicating NPH: partial obstruction to CSF
flow (e.g. aqueduct stenosis). Progressive mental impairment
• Communicating NPH: impairment of CSF flow distal to ‘Subcortical’ dementia
the fourth ventricle, most often at the level of the basal • Forgetfulness.
cisterns (‘cisternal block’) or arachnoid villi. • Inertia.
• Inattention.
Causes • Decreased speed of processing complex information.
• Idiopathic (at least 30%, probably about 50%). • Impaired ability to manipulate acquired knowledge.
• Subarachnoid hemorrhage. • Delayed recall is severely impaired but delayed recog-
• Meningitis. nition is only relatively mildly affected or even normal.
• Head trauma.
• Intracranial surgery. Cause
• Paget’s disease of the skull. • Compromise of the functional integrity of the frontal
• Mucopolysaccharidosis of the meninges. lobes and its connections.
• Unlikely to be caused by enlarged temporal horns and
N.B. Idiopathic cases have traditionally been attributed hippocampal dysfunction.
to defective CSF absorption through the arachnoid villi, but
evidence is lacking. More recently, the theory of a vascular Urinary urgency and incontinence
leukoencephalopathy has been proposed, with the concept • Urgency of micturition: almost always present.
that white matter lesions reduce periventricular tissue • Urinary incontinence: a late sign.
strength and elastic properties and thus predispose the
ventricles to dilate under CSF pulse pressure. Systemic Cause
hypertension is frequently associated with NPH and is Damaged periventricular pathways to the sacral bladder
thought to lead to ventricular enlargement because of center, resulting in decreased inhibition of bladder contrac-
decreased CSF absorption due to increased superior sagittal tions and hyperreflexia and instability of the bladder
sinus venous pressure and because of increased detrusor muscle, without concomitant defective sphincter
intraventricular pulse pressure from the choroid plexus. control. Urinary incontinence is not due to a so-called
Other possibilities include congenital hydrocephalus ‘incontinence sans gene’ except in the most severe forms,
becoming symptomatic in elderly persons and increasing age. when bladder hyperreflexia is associated with lack of concern
for micturition, due to severe frontal lobe dysfunction.
Episodes of slightly raised CSF pressure Clinical features are probably due to hydrocephalic
compression and stretching of the periventricular arterioles
and venules, leading to a state of ‘misery perfusion’
characterized by diminished periventricular blood flow that
is sufficient to result in axonal dysfunction but not enough
474 Disorders of the CSF Circulation

initially to cause irreversible loss of cellular integrity. Cauda equina syndrome


Prolonged periventricular ischemia eventually results in • Can produce gait instability, and bowel, bladder and
myelin degeneration and irreversible axonal loss, possibly sexual dysfunction, but usually there are lower motor
explaining why some patients with NPH do not improve neuron signs in the lower limbs (muscle wasting and
after a shunt. Narrowed periventricular arterioles due to weakness, and hyporeflexia or areflexia) and sensory loss
hyaline vessel wall degeneration in hypertensive patients (usually in a radicular distribution) in the lower limbs.
might be a predisposing factor, and explain the high • Parkinsonism is not present.
prevalence of vascular risk factors in NPH.
INVESTIGATIONS
DIFFERENTIAL DIAGNOSIS Neuroimaging with CT and MRI brain scan
Alzheimer’s disease • Imaging shows a communicating (non-obstructive)
• May cause a parkinsonian syndrome, and disorder of gait hydrocephalus with large ventricles, particularly the
and balance but usually late in its course when dementia is temporal horns, out of proportion to any degree of
severe. cortical atrophy (598–600).
• Cardinal features are memory deficits (particularly encoding • Frontal and occipital periventricular hypodensity,
deficits and impaired recognition) with or without aphasia, consistent with transependymal CSF absorption, may
apraxia, and agnosia. occur but are uncommon in NPH.
• MRI reveals atrophy of the hippocampal body as measured • Accentuation of the flow void in the aqueduct of Sylvius
by a reduction in its mean cross-sectional volume. is seen on MRI. N.B.T2W MRI images may show a
‘CSF flow voiding sign’, which consists of a decreased
Multi-infarct vascular dementia MRI signal, mainly in the aqueduct, which correlates
• Vascular risk factors are prevalent. with the velocity of pulsatile CSF flow. An increased
• Stepwise decline in function. aqueduct CSF flow velocity may be associated with
• CT or MRI brain scan shows multiple infarcts or shunt-responsive communicating NPH but there is no
strategically located infarcts or hemorrhages. good correlation between a pronounced CSF flow
voiding sign and NPH, and successful shunts do not
Subcortical arteriosclerotic encephalopathy (SAE) necessarily reduce this sign.
• One cause of vascular dementia. • Upward bowing of the corpus callosum and ballooning
• The clinical picture may be similar to NPH but often of the third ventricular recess are also seen on sagittal
hypertension and other vascular risk factors are present. MRI.
• Periventricular lucencies on cranial CT scan are usually • Unfortunately in practice it is very often difficult on
diffuse (around frontal and occipital horns) but in both SAE imaging to sort out the elderly atrophic brain from the
and NPH periventricular lucencies may be selectively located dilated ventricles of NPH.
around the frontal horns. • MRI can be used to measure the mean cross-sectional
volume of the hippocampal body in patients with
Obstructive hydrocephalus substantial global cognitive impairment to help dif-
• Headache. ferentiate Alzheimer’s disease (in which the hippocampal
• Papilledema may be present. body is atrophic) from NPH (in which it is not).
• CT or MRI brain scan evidence of a lesion obstructing CSF • An isotope study using indium 111-DTPA introduced
outflow and dilated ventricles rostral to the obstruction and into the CSF via an LP may help; in NPH and
normal sized ventricles caudal to it. communicating hydrocephalus the isotope is said to
reflux into the ventricles and does not accumulate over
Multiple systems atrophy the convexities by 24–48 hours after the LP.
A combination of parkinsonism, autonomic dysfunction,
cerebellar ataxia and corticospinal tract signs. Lumbar puncture (LP)
• To confirm the CSF pressure is normal.
Parkinson’s disease • To remove up to 30–50 ml of CSF (enough fluid to
• Usually manifests with asymmetric resting tremor, rigidity, decrease the opening pressure by about 50%) to see if a
bradykinesia, postural instability and shuffling gait. transient or, in rare cases, prolonged clinical improve-
• Symmetric involvement of the legs, early abnormalities of ment occurs. This can be assessed in various ways, such
bladder and sexual function, and a lack of response to as measuring pre-LP and post-LP gait pattern and time
levodopa are uncommon (but common in NPH). to walk 10 m (33 ft), and measuring psychometric
function. The ‘CSF tap test’ is easy, rapid and inexpensive
Vitamin B12 deficiency but its predictive accuracy is limited by the high rate of
May cause cognitive decline, gait instability and recurrent falls false negative results.
but usually there are optic atrophy and signs of a distal symmetri- • Continuous external lumbar drainage of about
c sensory neuropathy as well as corticospinal tract signs. 150–200 ml daily for 2–5 days may more accurately
predict the outcome of a shunt, even in cases of
Cervical myelopathy ‘negative’ CSF tap test. This test is technically simple but
May cause leg (and arm) spasticity and bladder and bowel complications such as radicular inflammation and
dysfunction but usually there is leg weakness and a motor meningitis may occur and experience with it is small. In
and sensory level. addition, it may not allow for the detection of delayed
improvement after several weeks or months unless
prolonged follow-up takes place.
Normal Pressure Hydrocephalus (NPH) 475

Cisternography Intracranial pressure monitoring and


CT cisternography, in which the isotope indium 111-DTPA hydrodynamic tests
is introduced into the CSF via an LP, can be used to assess • Continuous intracranial pressure monitoring: may show
CSF circulation and may help differentiate Alzheimer’s disease physiologic CSF pressure oscillations with a frequency of
from NPH; in NPH and communicating hydrocephalus the 0.5–2 per minute (B waves). It has been suggested that
isotope is said to reflux into the ventricles (i.e. ‘reversed CSF the occurrence of B waves is much increased in shunt-
flow’) and does not accumulate over the convexities by 24–48 responsive NPH, sometimes exceeding 50% of the
hours after the LP. This is a popular but unreliable predictive observation time, but there can be technical problems
test; the results do not seem to add to the diagnostic accuracy with this test and the results remain questionable.
of combined clinical and CT criteria. • Lumbar CSF infusion tests: measure the resistance of
CSF outflow (Rout) by lumbar or ventricular infusion of
Neuropsychologic assessment artificial CSF, and conductance of CSF (Cout, the
• Mini-mental state examination and other brief mental reciprocal of Rout), by measuring CSF reabsorption
screening tests may not be sensitive enough to detect through constant lumboventricular or ventriculo-
slight cognitive impairment of the ‘subcortical’ type. ventricular CSF infusions at different CSF pressures; tests
These are the very patients who may have the best have produced variable results.
surgical prognosis.
• Psychometric tests that include tests of frontal lobe
function, such as the trail making test and the Stroop
test, will reveal abnormalities even in the early stages of
the syndrome, and serial measures at intervals can be very
sensitive to a subtle decline, such as in cases where shunt 598
dysfunction is suspected. The test profile may not
differentiate NPH from other subcortical dementias but
it can distinguish them from Alzheimer’s disease by the
absence of cortical dysfunction and the presence of a
‘frontal’ pattern of mental deficit.

Cerebral blood flow (CBF) and metabolism measures


• Single photon emission computed tomography
(SPECT).
• Transcranial doppler (TCD) of the middle cerebral artery.
• Positron emission tomography (PET).

These methods may show decreased CBF and


metabolism, most pronounced in the frontal and
periventricular areas. However, there is no good correlation
between changes in CBF and outcome after CSF diversion.
Perhaps the techniques for measuring CBF are not sensitive
enough to detect subtle changes in the frontal
periventricular areas.

598–600 Plain CT 599 600


brain scan, axial slices,
showing dilatation of
the temporal horns
(598), the third
ventricle (599) and
the lateral ventricles
(600) with
periventricular lucency
and less prominent
dilatation of the
cortical sulci in a
patient with normal
pressure
hydrocephalus.
476 Disorders of the CSF Circulation

In most studies of the accuracy of ancillary tests for • Mild or moderate mental impairment, and not dementia.
predicting the outcome after shunting there has been a lack of • Hydrodynamic hydrocephalus suggested by cranial CT or
assessment of the pre-test probability of shunt responsiveness MRI (e.g. a CSF flow void on MRI).
based on well established clinical, CT and MRI data. • Positive CSF tap test: improvement in gait or psychometric
function after draining 30–50 ml of CSF.
EMG • 50–70% of patients with these features will do well after
• EMG may be helpful in cases in which gait impairment surgery.
is associated with prominent peripheral nerve or lower
motor neuron signs (e.g. wasting, fasciculations and Predictors of shunt non-responsiveness
weakness of muscles, normal or reduced muscle tone, • Dementia for >2 years.
loss of reflexes, and altered sensation in a radicular or • Onset of dementia before gait abnormality.
peripheral nerve distribution). • Alcohol abuse.
• Aphasia.
Blood tests to be considered • Substantial cortical atrophy and white matter involvement
• Vitamin B12. suggested by cranial CT or MRI.
• Folate.
• Vitamin E. Complications/morbidity
• Thyroid function tests. • Surgical mortality and severe residual morbidity rate: 5–15%.
• Liver function tests. • Post surgical complications are 30–40%, mostly due to intra-
• Rapid plasma reagin (RPR). cranial hemorrhage and extracranial infection, which fre-
• Fasting blood glucose. quently necessitate shunt removal and revision (see p.472).
• Serum protein electrophoresis. Less common complications include seizures and ischemic
stroke.
DIAGNOSIS
A clinical diagnosis primarily, supported by CT or MRI of the Predictors of shunt complications/morbidity
brain and LP. • Elderly.
• Initial symptoms are mental impairment of urinary
TREATMENT incontinence.
Medical • The complication rate exceeds 25% in these patients.
• No effective medical treatment is available.
• Acetazolamide, diuretics and corticosteroids are ineffective, Summary of management
despite having a role in idiopathic intracranial hypertension Management remains very difficult because of difficulties with
(pseudotumor cerebri), particularly when loss of vision has diagnostic criteria and limited evaluation of therapeutic
not occurred. Steroids are most helpful in reducing techniques by means of randomized trials. As a result,
increased ICP from vasogenic edema (that is, tumors). knowledge about prognosis, response to shunting, and
• CSF evacuation through repeated LP can dramatically predictors of a favorable and unfavorable response to shunting
reverse the NPH syndrome, but the success rate is also is minimal. In turn, practice remains haphazard and ranges
highly variable. from therapeutic nihilism to shunting nearly every patient with
suspected NPH. Most patients appear to have a real but limited
Surgical chance of improvement after a shunt based on clinical and brain
Techniques imaging features. The ratio of substantial benefit and serious
Ventriculoperitoneal or lumboperitoneal CSF diversion/shunt harm may be deceptively low. Consequently, caution and
surgery; ventriculoatrial shunting may be more difficult and reserve are required before labelling idiopathic communicating
more prone to complications such as arrhythmia and NPH as a reversible dementia.
pulmonary hypertension. Following clinical assessment, cranial CT or MRI scan, and
a CSF tap test, if there is any doubt about what to do next, we
Timing would recommend continuous external lumbar drainage for
Best done during the first 2 years after onset of symptoms and 4–5 days and shunting only those patients with considerable
signs. clinical improvement who are fit and willing to accept the
potential risks of shunt surgery.
Responsiveness A randomized controlled trial comparing the effect of
• Substantial improvement in about 30% of patients with shunting plus best medical therapy with best medical therapy
idiopathic NPH, and in 50–70% where the etiology is alone on long term outcome (death, cognitive function, gait,
known. urodynamics and handicap) is required.
• Gait disturbance is the most likely symptom to improve after
shunting. PROGNOSIS
• Surgical mortality and severe residual morbidity rate is The natural history is usually progressive deterioration. CSF
5–15%. shunting carries a success rate ranging from 25–80%, depending
• Post surgical complications are 30–40%. on patient selection and surgical skill and care.

Predictors of shunt responsiveness


• Short history.
• Known cause of hydrocephalus.
• Gait disorder is the initial and predominant feature.
Idiopathic Intracranial Hypertension (IIH) 477

IDIOPATHIC INTRACRANIAL Unconfirmed


HYPERTENSION (IIH) (BENIGN • Systemic diseases:
INTRACRANIAL HYPERTENSION, – Chronic renal failure.
PSEUDOTUMOR CEREBRI) – Systemic lupus erythematosus.
• Endocrine diseases:
– Adrenal insufficiency (Addison’s disease).
DEFINITION – Cushing’s disease.
A syndrome characterized by intracranial hypertension and – Hypoparathyroidism.
papilledema, normal sized cerebral ventricles, a normal CSF – Hypothyroidism.
composition, and no focal neurologic signs or evidence of • Thrombophilia (prothrombotic abnormalities):
a space-occupying lesion, an underlying infective process or – Polycythemia.
obstruction to CSF flow. – Thrombocytosis.
– Antiphospholipid antibodies.
EPIDEMIOLOGY – Hyperfibrinogenemia.
Incidence: – Antithrombin III deficiency.
– 0.9 per 100 000 per year. • Medication:
– 1.6 per 100 000 females per year. – Tetracycline.
– 3.3 per 100 000 women aged 15–44 years. – Minocycline.
– 7.9 per 100 000 overweight women. – Trimethoprim-sulfamethoxazole.
– 20 per 100 000 in overweight women aged 15–44 years. – Cimetidine.
• Age: adults 15–45 years of age predominantly; – Nalidixic acid.
adolescents, children, and infants with open fontanelles – Nitrofurantoin.
may also be affected. – Corticosteroids.
• Gender: F>M = 8:1 among adults; F=M in childhood – Tamoxifen.
and adolescence. – Lithium.
– Danazol.
PATHOLOGY – Levothyroxine.
Edema of the brain is present with ventricles of normal size. – Isotretinoin.
There is no obvious obstruction to CSF flow nor – Nasal fluticasone propionate (for hay fever)
abnormality of its content and no intracranial mass. Cerebral
vein thrombosis may be present, and may be secondary to CLINICAL FEATURES
compression of cerebral veins by the raised intracranial • Child or young woman of reproductive age usually.
pressure, rather than the primary cause of the raised • Obese (70%) or recent weight gain.
intracranial pressure. • Alert and generally well.
• Menstrual irregularities may be present.
ETIOLOGY AND PATHOGENESIS • A history of systemic or endocrine disease, or recent
Uncertain. Two fundamental defects of obscure origin: medication use (see above) may be present.
• Reduced CSF absorption at the level of the arachnoid
villi. Symptoms
• Increased brain intraparenchymal water: vasogenic brain • Headache (94%):
edema. – No specific site; usually generalized.
– Aching and throbbing.
Intracranial venous pressure is increased, probably due – Mild/dull.
to the raised intracranial pressure rather than venous sinus – Worse in the morning and may awaken the patient from sleep.
thrombosis being the underlying cause. Although some of – Aggravated by head movement, sitting up or standing, alco-
the risk factors for IIH (see below) are also possible risk hol, exertion, coughing, straining, sneezing and vomiting.
factors for cerebral vein thrombosis (e.g. thrombophilia), it – Relieved by lying down, and by aspirin or paracetamol
is possible that they are confounding factors, due to (in contrast to psychogenic headache).
misdiagnosis of true cases of venous sinus thrombosis as – Associated with nausea, vomiting and eventually papill-
IIH. Irrespective, IIH is probably a heterogeneous entity. edema.
In some patients there may be just one cause, such as • Transient visual obscurations or blurring of vision occurs,
venous sinus thrombosis, or severe obesity, and others may precipitated by activities, such as stooping over, straining,
have multiple risk factors which combine to precipitate IIH. and coughing, which further increase the intracranial
pressure sufficient to compromise the retinal and optic
Risk factors nerve circulation (68%).
Confirmed • Pulsatile intracranial noises (58%).
• Female gender. • Double vision (38%).
• Reproductive age group. • Visual loss (30%).
• Menstrual irregularity. • Retrobulbar pain on eye movement (22%).
• Obesity. • Minor symptoms may include: neck tenderness and
• Recent weight gain. stiffness, tinnitus, paresthesiae in the limbs distally, joint
pains, low back pain and intermittent ataxia.
• Irritability, drowsiness and apathy may be warning signals
of IIH in children.
478 Disorders of the CSF Circulation

Signs MRI/MRA (magnetic resonance angiography) or


• Papilledema (601–605) is seen, with its resultant con- digital subtraction angiography
centric constriction of the visual fields (30%), enlarged To exclude venous sinus thrombosis.
physiologic blind spots, and perhaps inferior nasal field
defects (15%), arcuate defects, cecocentral scotomas, and Other
vision loss: usually bilateral but may be unilateral or • Serial visual field perimetry: monitor.
highly asymmetric. • Serial stereoscopic ocular fundus photographs or indirect
• VIth cranial nerve paresis may occur as a false localizing ophthalmoscopy.
sign of raised intracranial pressure. • Fluorescein angiography: usually leaks diffusely with
• Other focal neurologic signs such as VIIth cranial nerve papilledema but leaves discrete spots of late focal
palsy and long tract involvement have been attributed to fluorescein with drusen.
IIH, but they are distinctly atypical.
• Patients are alert with no other focal neurologic signs (in Blood tests
contrast to patients with raised intracranial pressure, • Full blood count.
sufficient to cause papilledema, due to an expanding • Urea and electrolytes.
lesion). • Calcium.
• Thyroid function tests.
DIFFERENTIAL DIAGNOSIS • Antinuclear and antiphospholipid antibody.
• Venous sinus thrombosis: a distinctive diagnostic • Fibrinogen.
category: • Factor V Leiden gene mutation.
– Transverse, lateral or superior sagittal sinus thrombosis.
– Jugular vein obstruction. Snellen acuity, confrontational visual fields, direct
– Superior vena cava obstruction. ophthalmoscopic assessment of the degree of papilledema,
– Right heart failure. and visual evoked responses are insensitive and less useful in
• Occult mass lesion: gliomatosis cerebri (widely the follow-up of visual loss.
disseminated glioma).
• Papilledema: normal CT/MRI, abnormal CSF: DIAGNOSIS
– Guillain–Barré syndrome. Modified Dandy’s diagnostic criteria
– Cryptococcal meningitis. • Symptoms and signs of increased intracranial pressure
– Carcinomatous meningitis. (e.g. papilledema).
– Chronic brucellosis. • No localizing focal neurologic signs, in an awake and
– Cysticercosis. alert patient, other than occasional abducens nerve
– Neurosarcoidosis. paresis.
– Poliomyelitis. • Normal neuroimaging studies (enhanced CT or MRI
• Pseudopapilledema: anomalous elevation of the optic brain scan) except for small ventricles or empty sella.
nerve head: • Documented increased opening pressure (250 mm of
– Hyperopia. water or more) but a normal composition of the CSF.
– Optic nerve drusen: can also cause transient visual • Primary structural or systemic causes of elevated
obscurations and a progressive optic neuropathy rarely. intracranial venous sinus pressure excluded (e.g. venous
• Headache: sinus thrombosis, hyperviscosity syndromes and right
– Tension headache. heart failure).
– Mixed migraine/tension headache. • Benign clinical course apart from visual deterioration.

INVESTIGATIONS
CT and MRI scan of the brain
The ventricles are either normal size or small with a slit-like
third ventricle and diminished basal cisterns (606, 607).
This may be difficult to recognize in individual cases,
therefore the main reason for imaging is to (1) exclude 601
other causes of the patient’s symptoms such as tumor
masses, and (2) to exclude cerebral venous sinus thrombosis
(see p.257 for imaging of venous sinus thrombosis).
Common associated features are an ‘empty sella syndrome’
(55% of scans), a prominent cisterna magna, and thickened
optic nerve sheaths.

CSF
• Opening pressure: elevated, >25 cm (>250 mm) of
water.
• Cell count: normal.
• Protein: normal or low.
• Glucose: normal.

601 Normal optic disk.


Idiopathic Intracranial Hypertension (IIH) 479

602 603

602 Early mild papilledema. (601–605, courtesy of Mr M Wade, 603 Severe papilledema. Note the pinkish congested disc, indistinct
Department of Medical Illustrations, Royal Perth Hospital,Western disc margin, elevation of the optic nerve head, filling-in of the optic cup,
Australia.) and distension of the retinal veins.

604 605

604, 605 Right (604) and left (605) optic fundus of a patient with bilateral papilledema due to idiopathic intracranial hypertension.

606 607

606, 607 CT brain scan (606) and T2W axial MRI (607) in a patient with idiopathic intracranial hypertension.
Note the small third and lateral ventricles but otherwise normal appearing brain.
480 Disorders of the CSF Circulation

COMPLICATIONS Pharmacologic
Visual loss • Acetazolamide (Diamox), a carbonic anhydrase inhibitor
• 15–50% of patients. which inhibits CSF production: start with oral dose of
• May occur early or late. 250 mg four times a day and increase to 500 mg four
• May be sudden or gradually progressive. times daily unless adverse effects develop (paresthesias,
• May be asymptomatic. drowsiness, nausea, malaise, metabolic acidosis, altered
• Visual loss is probably due to a nerve conduction defect taste, and renal calculi).
related to the status of axoplasmic flow found in • Other diuretics: chlorthalidone 100 mg oral, daily; fruse-
papilledema. It is avoidable with appropriate monitoring mide (furosemide), glycerol (but may cause weight gain).
and treatment. • Corticosteroids: prednisone 40–60 mg/day for 10–14
days, and taper over the next 2 weeks.
PROGNOSIS • Anticoagulation if a secondary venous sinus thrombosis
• Not well known. is present.
• Often self-limiting; many people recover spontaneously
in a few weeks and most people experience spontaneous Surgical
resolution in 6–18 months, but it is a chronic process in If vision loss is severe initially or progresses in spite of
some. medical treatment:
• Optic nerve sheath decompression by fenestration is the
TREATMENT treatment of choice as it is reasonably safe with very few
Early detection and prevention of vision loss complications.
Medical • Lumboperitoneal shunting (the insertion of a shunt from
• Avoid or correct predisposing factors (see above): weight the spinal subarachnoid space to the peritoneum) has a
reduction if obese. higher risk of complications and failure.
• Serial LPs: if symptoms persist after initial LP perform
3–4 LPs within the first 2–4 weeks; further LPs are
unlikely to help.

FURTHER READING IDIOPATHIC INTRACRANIAL Radhakrishnan K, Ahlskog E, Garrity JA, Kurland


HYPERTENSION (IIH) LT (1994) Idiopathic intracranial hypertension.
Bond DW, Charlton CPJ (2001) Benign intracra- Mayo Clin. Proc., 69: 169–180.
NORMAL PRESSURE HYDROCEPHALUS nial hypertension secondary to nasal fluticasone Walker RWH (2001) Idiopathic intracranial hy-
(NPH) propionate. BMJ, 322: 897. pertension: any light on the mechanism of the
Chen IH, Huang CI, Liu HC, Chen KK (1994) Brazis PW, Lee AG (1998) Elevated intracranial raised pressure? J. Neurol. Neurosurg. Psychia-
Effectiveness of shunting in patients with nor- pressure and pseudotumor cerebri. Curr. Opin. try, 71: 1–7.
mal pressure hydrocephalus predicted by tem- Ophthalmol., 9: VI:27–32.
porary, controlled-resistance, continuous lum- Digre KB, Corbett JJ (2001) Idiopathic intracra-
bar drainage: a pilot study. J. Neurol. Neuro- nial hypertension (pseudotumor cerebri): a
surg. Psychiatry, 57: 1430–1432. reappraisal. The Neurologist, 7: 2–67.
Evidente VH, Gwinn KA (1997) 73-year-old man Go KG (2000) Pseudotumour cerebri. Incidence,
with gait disturbance and imbalance. Mayo management and prevention. CNS Drugs, 14:
Clin. Proc., 72: 165–168. 33–49.
Graff-Radford NR (1999) Normal pressure hy- Karahalios DG, Rekate HL, Khayata MH, Apos-
drocephalus. The Neurologist, 5: 194–204. tolides PJ (1996) Elevated intracranial venous
Pleasure SJ (1999) Ventricular volume and trans- pressure as a universal mechanism in pseudotu-
mural pressure gradient in normal pressure hy- mor cerebri of varying etiologies. Neurology,
drocephalus. Arch. Neurol., 56: 1199–1200. 46: 198–202.
Vanneste JAL (1994) Three decades of normal King JO, Mitchell PJ, Thomson KR, Tress BM
pressure hydrocephalus: are we wiser now? J. (1995) Cerebral venography and manometry
Neurol. Neurosurg. Psychiatry, 57: 1021–1025. in idiopathic intracranial hypertension. Neurol-
ogy, 45: 2224–2228.
Chapter Eighteen 481

Cranial
Neuropathies

OLFACTORY (IST CRANIAL NERVE) • Thus, olfactory impulses reach the cerebral cortex
NEUROPATHY without relay through the thalamus; a unique feature
among the sensory systems.
• From the prepiriform cortex, fibers project to the
DEFINITION neighboring entorhinal cortex (area 28 of Brodmann),
Disorder of the Ist cranial, or olfactory, nerve resulting in a the medial dorsal nucleus of the thalamus, and the
disturbance of smell sensation (anosmia). hypothalamus.
• Olfactory stimuli and emotional stimuli are thus strongly
ANATOMY linked, as a result of their common roots in the limbic
• Nerve fibers subserving the sense of smell have their cells system. Yet, the ability to recall an odor is nowhere near
of origin in the mucous membrane of the upper and as good as the ability to recall sounds and sights.
posterior parts of the nasal cavity.
• The olfactory mucosa contains three types of cells: PHYSIOLOGY
– Olfactory or receptor cells. • During quiet breathing, little of the air entering the
– Sustentacular or supporting cells. nostril reaches the olfactory mucosa; sniffing carries the
– Basal cells, which are stem cells and the source of both air into the olfactory crypt.
olfactory (receptor) cells and sustentacular cells during • To be perceived as an odor, an inhaled substance must
regeneration. be volatile – i.e. spread in the air as very small particles,
• The olfactory or receptor cells are bipolar neurons that and soluble in water or lipid.
each have a peripheral process (the olfactory rod), from • Intensity of olfactory sensation is determined by the
which project 10–30 fine hairs, or cilia, and several frequency of firing of afferent neurons.
central processes (or olfactory filia) which are fine • Quality of odor is probably determined by ‘cross fiber’
unmyelinated fibers that converge to form small fascicles activation, since the individual receptor cells are
enwrapped by Schwann cells and pass through openings responsive to a wide range of odors and exhibit different
in the cribriform plate of the ethmoid bone into the types of responses to stimulants.
olfactory bulb. Collectively, the central processes of the
olfactory receptor cells constitute the Ist cranial, or ETIOLOGY
olfactory, nerve. Nasal
• In the olfactory bulb, the axons of the receptor cells Odorants do not reach the olfactory receptors
synapse with mitral cells, the dendrites of which form Nasal obstruction or inflammation (rhinitis) is by far the
brush-like terminals or olfactory glomeruli. most common cause.
• The axons of the mitral cells enter the olfactory tract,
which courses along the olfactory groove of the Olfactory neuroepithelial
cribriform plate to the brain. Destruction of receptors or their axon filaments
• The olfactory tract divides into medial and lateral Congenital absence or hypoplasia of primary receptor
olfactory striae. The medial striae contains fibers from neurons:
the anterior olfactory nucleus which pass to the opposite • Kallman syndrome (congenital anosmia and hypogon-
side via the anterior commissure. Fibers in the lateral adotropic hypogonadism).
striae originate in the olfactory bulb, give off collaterals • Albino.
to the anterior perforated substance, and terminate in the
medial and cortical nuclei of the amygdaloid complex
and the prepiriform area (the lateral olfactory gyrus). The
latter represents the primary olfactory cortex, which
occupies a restricted area on the anterior end of the
hippocampal gyrus and uncus (area 34 of Brodmann).
482 Cranial Neuropathies

Disruption of the delicate filaments of the receptor cells as • Bilateral anosmia:


they pass through the cribriform plate: – Nasal obstruction or inflammation (rhinitis).
• Head injury, particularly if severe enough to cause skull – Hysteria.
fracture (608). Less common with closed head injury.
The damage may be unilateral or bilateral. Qualitative abnormalities of olfaction
• Cranial surgery. Distortions or illusions of smell (dysosmia or parosmia)
• Subarachnoid hemorrhage. • Local nasopharyngeal conditions (e.g. empyema of the
• Chronic meningitis. nasal sinuses).
• Depression.
Central
Olfactory pathway lesions Olfactory hallucinations or delusions
• Inferior frontal tumor compressing the olfactory tracts • Temporal lobe disease: complex partial seizures, anterior
(e.g. olfactory groove meningioma). temporal lobectomy.
• Large aneurysm of the anterior cerebral or anterior • Psychiatric disease: schizophrenia, depression.
communicating artery.
• Anterior meningoencephalocele (CSF rhinorrhea may be Loss of olfactory discrimination
present in certain head positions). • Alcoholics with Korsakoff ’s psychosis (degeneration of
• Meningitis. neurons in the higher order olfactory systems of the
• Refsum’s disease (hereditary ataxic neuropathy). medial temporal and thalamic regions).
• Sarcoidosis. • Temporal lobe disease: complex partial seizures, anterior
temporal lobectomy.
Parkinson’s disease
Reduced odor recognition
Multiple sclerosis • Alzheimer’s disease.
• Huntington’s disease.
Idiopathic
Usually bilateral but can be unilateral. INVESTIGATIONS
Determined by the clinical findings:
CLINICAL FEATURES • CT or MRI brain scan.
• Smell should be tested with an aromatic odor, such as an • CSF examination.
orange or cordial bottle top, and not with an acrid odor, • EEG.
such as ammonia.
• Associated signs, such as unilateral optic atrophy and TREATMENT
contralateral papilledema (Foster–Kennedy syndrome) Remove the underlying cause, if possible.
may be present in conditions such as olfactory groove
meningioma which extends posteriorly to involve the PROGNOSIS
ipsilateral optic nerve and also causes raised intracranial Depends on the cause. Anosmia due to head injury causing
pressure and papilledema. skull fracture is usually permanent. Anosmia due to closed
head injury recovers in about one-quarter of patients.
Quantitative abnormalities of olfaction
Bilateral loss or reduction of the sense of smell
(anosmia, hyposmia) 608 608 Ventral
Commonly, but not always, recognized by the patient. It surface of the
may present as impaired taste. Indeed, the patient with frontal lobes of
bilateral anosmia is usually convinced that the sense of taste the brain at
has been lost as well (ageusia), because taste depends largely autopsy showing
on the volatile particles in foods and beverages, which reach disruption of the
the olfactory receptors through the nasopharynx, and the olfactory nerves
perception of flavor is a combination of smell and taste. bilaterally
However, these patients are able to distinguish the (arrows) due to
elementary taste sensations (sweet, sour, bitter and salty). traumatic brain
injury.
Unilateral loss or reduction of the sense of smell
(anosmia, hyposmia)
Seldom, if ever, recognized by the patient.

DIFFERENTIAL DIAGNOSIS
Quantitative abnormalities of olfaction
Loss or reduction of the sense of smell (anosmia, hyposmia)
• Unilateral anosmia. Hysteria: anosmia, if unilateral, may
be on the same side as other symptoms such as anesthesia,
blindness or deafness. Hysterical anosmics don’t usually
complain of loss of taste whereas true anosmics do, but
actually show normal taste sensation on testing.
Optic (IInd Cranial Nerve) Neuropathy 483

OPTIC (IIND CRANIAL NERVE) Fibers from the lateral geniculate body sweep into the
NEUROPATHY hemisphere in two fan-shaped projections, which later come
together at the occipital cortex.
The lower fibers of the optic radiation (subserving the
DEFINITION contralateral upper visual field) sweep forward into the
Disorder of the optic (IInd cranial) nerve. anterior temporal lobe as Meyer’s loop. The upper fibers of
the optic radiation (subserving the contralateral lower visual
EPIDEMIOLOGY field) follow a more direct route back into the parietal lobe.
Quite common. All fibers then sweep back in the deep white matter of the
occipital lobe and terminate in the calcarine (visual) cortex
ANATOMY astride the calcarine fissure.
Nerve fibers from retinal nerve cells lying on the choroid at The cells subserving the upper visual fields (i.e. lower
the back of the eye come straight forward and then angle retinae) lie in the lower half of the calcarine cortex, and those
sharply to run across the surface of the retina towards the subserving the lower fields (i.e. upper retinae) are in the upper
optic nerve head (optic disc) where they enter the optic nerve. half of the cortex. The cells subserving the peripheral
The macula, situated to the temporal side of the optic nerve (temporal) visual fields are represented anteriorly (hence the
head, is the most critical part of the retina because it is finding of a temporal crescent visual field defect with an
responsible for central vision. It consists of a very densely anterior striate cortex lesion), and those subserving macular
packed mass of cells. These nerve cells are particularly sensitive vision are concentrated at the extreme tip of the occipital
to a variety of toxins and, if damaged, lead to a centrocecal lobe. The longitudinal extent of the calcarine cortex is impor-
scotoma. Fibers from the macula form the papillomacular tant in permitting striking localized visual field defects that
bundle as they shift sideways en mass and enter the temporal may occur as a result of lesions (usually vascular) in this area.
half of the optic disc. As they pass backwards along the optic
nerve, they gradually becoming more central. ETIOLOGY AND PATHOPHYSIOLOGY
At the optic chiasm the fibers in the lateral half of the Optic nerve lesion
optic nerve, coming from the lateral (temporal) half of the Optic nerve: unilateral
retina, pass straight back into the optic tract on the same Sudden monocular visual failure:
side. The fibers in the medial half of the optic nerve, coming • Ischemia: ischemic optic neuropathy (see p.487).
from the medial (nasal) half of the retina cross (decussate) • Demyelination: optic neuritis (see p.488).
in the chiasm and pass back into the optic tract on the • Cavernous sinus thrombosis.
opposite side. However, before doing so, the fibers from the • Carotico-cavernous fistula.
inferior nasal retina (receiving visual information from the
upper temporal field), having crossed the anterior inferior Progressive monocular visual failure:
optic chiasm, then loop forwards into the posterior part of • Optic neuritis.
the opposite optic nerve before turning back to pass in the • Tumor of the orbit or optic nerve: optic nerve
lateral chiasm and optic tract. Consequently, pressure in the astrocytoma, optic sheath meningioma.
posterior optic nerve can cause not only an ipsilateral blind • Dysthyroid eye disease.
eye but also a small defect in the upper temporal field of the • Paget’s disease of the skull: obstruction of foramina
opposite eye (the anterior chiasmal syndrome). The function leading to optic atrophy.
of the chiasm is to bring the information from the halves of • Anterior communicating artery aneurysm: compression
each retina that look to the right and the halves of each of optic nerve.
retina that look to the left together in the same optic tract. • Irradiation.
In the optic tract the fibers serving the identical point of • Tolosa–Hunt syndrome.
each of the two retinae have to come together. The fibers in • Meningovascular syphilis.
the anterior part of the optic tract rotate inwards through 90°, • Subacute and chronic meningitis.
which brings the fibers in the lower and upper fields together
in the medial and lateral halves of the tract respectively. In the Optic nerve (bilateral) or optic chiasm
posterior part of the optic tract the fibers fan out towards the Sudden binocular visual failure:
six layers of the geniculate body, allowing the adjacent fibers • Bilateral ischemic optic neuropathy.
from each retina to interdigitate. The macula fibers occupy a • Pituitary apoplexy.
large central wedge of the geniculate body, with the lower • Raised intracranial pressure causing bilateral severe
fields being represented medially and the upper fields laterally. papilledema.
484 Cranial Neuropathies

Progressive binocular visual failure: Visual fields


• Bilateral optic neuritis. • Tested at the bedside by confrontation testing. Routine
• Leber’s hereditary optic atrophy. screening is usually performed with a 10 mm (0.4 in)
• Adrenoleukodystrophy. white hat pin (or fingers); a 10 mm (0.4 in) red hat pin
• Metachromatic leukodystrophy. is useful when a compressive lesion of the optic nerve,
• Compression/infiltration: pituitary adenoma; cranio- optic chiasm or optic tract is suspected; and smaller pins
pharyngioma; meningioma of tuberculum sellae, olfac- (5 mm [0.2 in]) are useful for detecting small scotomata
tory groove, or sphenoid wing; epidermoid/dermoid (small field defects) in the centre of the visual fields.
tumor; carotid siphon or ophthalmic artery aneurysm, More sophisticated tests include the Amsler chart,
chordoma of the clivus; nasopharyngeal carcinoma; Tangent (Bjerrum) screen testing and Goldman
metastatic carcinoma; sarcoid granuloma. perimetry.
• Nutritional deficiency: vitamins B1 and B12 (usually • Ipsilateral scotoma: usually central, but sometimes
centrocecal scotoma). centrocecal (embracing the blind spot); or:
• Toxins: alcohols and glycerols (glycerins; e.g. methyl • Complete monocular blindness.
alcohol), heavy tobacco consumption, mercury, – An associated upper temporal field defect in the
clioquinol, iodoquinol, chronic cyanide poisoning. contralateral eye may be present with retro-orbital optic
• Drugs: amiodarone, chloramphenicol, chloroquine, nerve lesions that involve some of the nasal crossing
desferrioxamine (deferoxamine), ethambutol, quinine. fibers from the other eye in the anterior optic
• Systemic disorders: sarcoidosis, dysthyroid eye disease. chiasm/posterior optic nerve.
• Subacute and chronic meningitis (e.g. tuberculosis, • Bilateral centrocecal scotoma are suggestive of bilateral
syphilis, fungal). optic neuropathies due to toxins and drugs.
• Malignant meningitis.
• Irradiation. Pupils
• Primary hypothyroidism (causing pituitary enlargement). • Abnormal (delayed or less intense) direct response to
light (i.e. impaired afferent arc of the reflex) but intact
N.B. The chiasm is not affected by ischemia because it consensual response (preserved efferent arc). This
has a very extensive blood supply from many small arteries. commonly manifests as an afferent pupillary defect
(Marcus Gunn pupil) (see p.575).
CLINICAL FEATURES • A normal response to light occurs with blindness that is
• Visual failure and field defects, which may be monocular caused by lesions posterior to the lateral geniculate body
or binocular, sudden or gradual in onset, and transient (e.g. bilateral occipital lobe disease [cortical blindness]
or persistent. or hysteria/malingering) because the afferent arc of the
• The diagnosis and localization of the etiologic lesions pupillary reflex leaves the optic tract just before the lateral
requires a detailed examination of the visual acuity, visual geniculate body, and goes on its way to the midbrain,
fields, pupils, fundi and the first six cranial nerves, along and then to the pupil.
with the eye itself and for any general neurologic and
medical disorders. Fundi (609)
• May show features of the underlying cause (e.g. optic
Optic nerve lesion neuritis, see p.489) or the sequelae of the cause (e.g.
Visual acuity optic atrophy, papilledema).
• Reduced visual acuity in the visual field of the ipsilateral • Atrophy/degeneration of the optic nerve due to
eye, which is usually noticed early by the patient. traumatic transection, demyelination, syphilis, Leber’s
• Tested with a Snellen chart and, if necessary, with hereditary optic atrophy, or a tumor of the nerve usually
correction for refractive errors with lenses/spectacles or causes the optic disc to appear chalk white with sharp,
a pinhole. Normally the visual acuity is 6/6 in each eye, clean margins (610).
or better (i.e. 6/5 or 6/4). • Atrophy/degeneration of the optic nerve due
to/following papillitis or papilledema usually causes the
Color vision optic disc to appear a pallid, yellow-gray color with
• Important for detecting lesions of the pregeniculate optic irregular and indistinct margins, and partial obscuration
pathway, which characteristically compromise ability to and sometimes sheathing of the vessels.
detect a red object. For example, the earliest evidence of
a pituitary lesion may be a bitemporal hemianopia
specifically to a red object. Similarly, an optic tract lesion
may produce an incongruous hemianopic defect to red.
If the patient has impaired visual acuity but can still
detect a red object then a pregeniculate pathway lesion
is unlikely.
• The standard test is Ishihara color plates but those
patterns requiring accurate perception of red are of
particular value in neurology. A red hat pin can
sometimes therefore suffice.
Optic (IInd Cranial Nerve) Neuropathy 485

Optic chiasm lesion Optic radiation lesion


Visual fields • Congruous homonymous hemianopia involving central
• Initially the crossing fibers tend to be affected first, vision results if the whole radiation is involved
resulting in a bitemporal hemianopia that is lower if the (proximally by a small lesion near its origin or distally by
upper chiasm is compressed (e.g. by a craniopharyn- a large lesion such a stroke or tumor).
gioma) and upper if the lower chiasm is compressed (e.g. • Upper temporal homonymous quadrantanopia results if
by a pituitary adenoma). only the lower fibers are affected: temporal lobe lesions.
• Later, the visual field defect expands to become a • Lower temporal homonymous quadrantanopia results if
complete bitemporal hemianopia, which may remain only the upper fibers are affected: parietal lobe lesions.
unnoticed by the patient, or only cause vague symptoms
such as difficulty with close work, vertical splitting of Occipital cortex lesion
images and double vision. Unilateral
• The bitemporal hemianopia may be quite asymmetric, Homonymous hemianopia occurs with or without macular
incongruous, or spare the peripheral field (i.e. sparing. If the macular is ‘split’ the patient may complain of
scotomatous bitemporal hemianopia), but the midline seeing ‘half of things’.
vertical division between the normal nasal and abnormal
temporal fields is definite, albeit sometimes subtle and Bilateral
only detectable with a red target. • Cortical blindness occurs bilaterally and may be ignored
• Eventually, involvement of the uncrossed temporal fibers or denied by the patient.
leads to expansion of the visual field defect across the • Upper or lower altitudinal visual field defect occurs in
midline to also involve the nasal field, and the reduction both eyes if only the lower or upper calcarine cortex is
in visual acuity is then noticed by the patient, sometimes involved.
rather suddenly. • Tunnel (tubular) vision occurs due to macular sparing if
• An ipsilateral nasal field defect may very occasionally be the occipital pole is spared.
seen due to lateral compression of the chiasm (e.g. from • Bilateral central scotoma occurs if only the occipital poles
a carotid aneurysm). are involved.

Optic tract and lateral geniculate body lesion SPECIAL FORMS


• Rare. • Anterior ischemic optic neuropathy (see p.487).
• Incongruous homonymous hemianopia. • Optic neuritis (see p.489).
• Leber’s hereditary optic neuropathy (see p.491).
• Optic nerve tumor (see p.372).

609 610

609 Ophthalmoscopic view of a normal optic fundus showing a 610 Optic atrophy, showing a white optic disc with sharp, clear
healthy optic nerve head, retina, and retinal vessels. margins.
486 Cranial Neuropathies

DIFFERENTIAL DIAGNOSIS • Optic radiation:


Impaired visual acuity in one eye (monocular) – Stroke.
Reduced visual acuity (corrected) indicates a lesion of the – Tumor.
eye (cornea, lens, vitreous, retina), optic nerve, or optic • Occipital lobes:
chiasm. Visual acuity is not commonly affected by lesions of – Bilateral parieto-occipital infarction due to profound
the optic tract, optic radiation and visual cortex in the hypotension/anoxia.
occipital lobe, and, if it is, it is binocular. – Bilateral occipital infarction due to vertebrobasilar
thromboembolism.
Transient – Vertebral angiography.
• Eye: glaucoma. – Head injury.
• Retina: • Other: hysteria and malingering.
– Ischemia (amaurosis fugax).
– Migraine. INVESTIGATIONS
• Orbit: tumor. • Formal visual field testing.
• Optic nerve: demyelination: Uhthoff’s phenomenon. • Visual evoked potentials.
• Posterior optic nerve/anterior optic chiasm: hemorrhage: • MRI or CT scan of the orbits and sinuses, pituitary fossa,
arteriovenous malformation (chiasmal apoplexy). and brain.
• Other: hysteria and malingering. • Fluorescein angiography (611).
• CSF examination.
Persistent • Full blood count and ESR.
• Eye: • Fasting blood glucose.
– Cataract. • Thyroid function tests.
– Vitreous hemorrhage. • Autoantibody screen.
– Glaucoma. • Mitochondrial DNA.
• Retina: • Syphilis serology.
– Infarction: arterial or venous (central or branch artery or • Chest x-ray.
vein). • Cerebral angiography.
– Hemorrhage.
– Macular disease (e.g. senile macular degeneration): visible DIAGNOSIS
with an ophthalmoscope. Clinical, electrophysiologic and radiologic.
– Chorioretinitis: cytomegalovirus, toxoplasma infection.
• Orbit: TREATMENT
– Tumor. Determined by the cause.
– Suppuration, fungal infection (orbit or paranasal sinuses):
mucomycosis. PROGNOSIS
• Optic nerve/anterior optic chiasm (see above). Depends on the cause.
• Central: amblyopia due to a congenital squint.

Impaired visual acuity in both eyes (binocular)


Transient
• Bilateral occipital lobe ischemia: vertebrobasilar transient
ischemic attack.
• Migraine.
• Vertebral angiography.
• Systemic hypotension.
• Head injury.
• Raised intracranial or intraorbital pressure causing
bilateral papilledema; 611
Persistent
• Eye:
– Cataract.
– Glaucoma.
• Retinal:
– Infarction.
– Hemorrhage.
– Diabetic retinopathy.
– Macular disease (e.g. senile macular degeneration): visible
with an ophthalmoscope.
– Toxins: tobacco, alcohol, ethambutol, amiodarone,
isoniazid, chloramphenicol, iodoquinol.
– Retinitis pigmentosa (ultimately leads to optic atrophy).
• Optic nerve and chiasm demyelination, tumor. 611 Fluorescein angiogram of a normal optic fundus, showing no
• Optic tract: middle cranial fossa tumor. leakage of contrast material from the vessels.
Anterior Ischemic Optic Neuropathy (AION) 487

ANTERIOR ISCHEMIC OPTIC Examination


NEUROPATHY (AION) Visual acuity: varies from 6/6 to no light perception, so
normal visual acuity does not exclude AION.
Visual fields: usually nerve fiber bundle defects are
DEFINITION characteristic of an optic disc disorder but any visual field
Anterior ischemic optic neuropathy is an acute ischemia of defect can occur. An altitudinal hemi-field defect (loss of
the anterior portion of the optic nerve. either the upper or, more frequently, the lower half of the
field in one eye) is particularly common, as is inferior nasal
EPIDEMIOLOGY segmental loss and central scotoma. This altitudinal pattern
• Incidence: 10 per 100 000 per year (non-arteritic). of field loss is because of the nature of the posterior ciliary
• Age: 11–91 years, but typically over 50 years of age. arterial circulation, which is divided into upper and lower
• Gender: M=F. divisions of supply to the optic nerve.
Pupils: relative afferent pupillary defect (see p.575), as
PATHOLOGY with any unilateral optic neuropathy.
Acute ischemic infarction of the optic nerve head occurs due Ophthalmoscopy: the disc may appear normal at first,
to occlusion of the posterior ciliary arteries that are branches but within a few days it becomes pale and swollen, often
of the ophthalmic artery and supply the anterior part of the with small flame-shaped hemorrhages radiating from the
optic nerve, the choroid and outer retina. disc margin (ischemic papillopathy), distended veins and,
occasionally, cotton-wool spots due to ischemic change in
ETIOLOGY AND PATHOPHYSIOLOGY the surrounding retina (612, 613). The swelling may
Posterior ciliary artery territory ischemia involve only one segment of the disc or be more marked in
The optic disc is close to an arterial borderzone between the one segment than in another. The disc swelling is attributed
territories of supply of the two major PCAs and is therefore partly to leakage of plasma from damaged blood vessels and
liable to selective ischemia when the systemic blood pressure partly to the arrest of axoplasmic transport along damaged
falls, the intraocular pressure rises or when there is occlusive
disease of local small arteries.
• Systemic hypotension. 612
• Raised intraocular pressure.
• Thrombotic or embolic occlusion of the PCA:
– Giant cell arteritis (GCA).
– Other connective tissue diseases such as polyarteritis
nodosa.
– Atherosclerosis: patients have an increased prevalence of
hypertension and diabetes and an increased risk of
subsequent cerebrovascular and cardiovascular events.
– Embolism from the proximal carotid circulation, aortic
arch or heart.

Risk factors
• Congenitally small and crowded optic nerve heads (the
so-called ‘disc-at-risk’).
• Increasing age. 612 Fundus photograph showing a swollen optic disc, which is more
• Hypertension. marked in one segment than in another, with small flame-shaped
• Diabetes. hemorrhages radiating from the disc margin.

CLINICAL FEATURES 613


Monocular blindness
History
• Abrupt onset.
• Painless.
• Severe.
• The entire visual field is usually affected but tends to be
more severe in the lower quadrants because the upper
segment of the optic disc is more vulnerable to ischemia.
• It may be exacerbated in early stages by minor postural
change (precarious circulation).
• AION is persistent and non-progressive but can be brief
(and present as transient monocular blindness) and may
progress over several hours or days.
• Preceding amaurosis fugax may have occurred.
• Rarely, blindness is bilateral and simultaneous or develops 613 Fundus photograph showing a swollen optic disc with distended
within a few days (usually due to giant cell arteritis). veins and cotton-wool spots due to ischemic change in the surrounding
retina. (612, 613 courtesy of Mr M Wade, Department of Medical
Illustrations, Royal Perth Hospital,Western Australia.)
488 Cranial Neuropathies

nerve fibers. The disc swelling may be indistinguishable • Hypertension is present in up to half of patients, and
from that seen with raised intracranial pressure but diabetes in up to 40% of patients.
sometimes the disc has a pale appearance and, in about half
of the eyes with AION due to giant cell arteritis, the disc INVESTIGATIONS
swelling has a chalky white appearance. Later, the swelling • ESR.
subsides to be succeeded by optic atrophy with attenuation • Temporal artery biopsy: likely to remain positive in cases
of the small blood vessels on the disc surface. of GCA even if done up to 14 days after starting steroid
therapy.
DIFFERENTIAL DIAGNOSIS • Fluorescein angiography of the ocular fundus.
Sudden and persistent monocular visual failure
• Retinal infarction: TREATMENT
– Central or branch retinal artery occlusion. Arteritic AION
– Central or branch retinal vein occlusion. Immediate high dose steroids to prevent complete blindness
• Retinal or vitreous hemorrhage. (see GCA, p.234).
• Other visible retinal lesions.
• Papilledema due to raised intracranial pressure. Non-arteritic AION
• Cavernous sinus thrombosis. Acute treatment
• Carotico-cavernous fistula. • No proven effective therapy.
• Optic nerve sheath decompression surgery: making one
Sudden and persistent binocular visual failure or several openings in the dura covering the optic nerve
• Pituitary apoplexy (see p.268). just behind the eyeball, allowing CSF to escape, thus
• Bilateral occipital infarction causing cortical blindness reducing the pressure surrounding the optic nerve, has
(which is sometimes not recognized by the patient) with been shown in a randomized trial of 244 patients to be
normal pupil response to light (since visual afferents ineffective and possibly harmful.
destined for the midbrain leave the optic tract just before
the lateral geniculate body): Prevention of subsequent vascular events
– Profound hypotension (bilateral parieto-occipital • Control of vascular risk factors such as hypertension and
infarction). diabetes.
– Vertebrobasilar thromboembolism. • Long term antiplatelet therapy.
– Vertebral angiography.
• Head injury. PROGNOSIS
The prognosis for recovery of vision is variable and depends
DIAGNOSIS on factors such as the underlying cause. Many patients (up
Swelling of the optic disc is a necessary sign for the diagnosis to 56%) with non-arteritic AION recover good central
of AION. Having established the diagnosis of AION, the vision (6/18 or 20/60, or better) or are left with arcuate
crucial next step is to differentiate arteritic AION, that is and sectorial visual field defects corresponding to loss of
caused by giant cell arteritis, from non-arteritic AION. bundles of nerve fibers. In other patients, particularly those
with arteritic AION, visual loss may be complete and
Features suggestive of arteritic AION permanent.
• Older age.
• Systemic symptoms: headache, scalp tenderness, jaw
claudication, muscle aches, fatigue and weight loss.
• Premonitory visual symptoms: transient monocular visual
loss or diplopia.
• Early, massive, and progressive visual loss.
• Bilateral visual loss within days or weeks.
• Chalky-white optic disc swelling.
• Concurrent signs of retinal circulation ischemia such as
cotton wool spots or retinal infarction.
• Elevated ESR.
• Absent filling of the choroidal circulation on fluorescein
fundus angiography.

Features suggestive of non-arteritic AION


• Predominantly in 50s and 60s, but can occur at any age.
• Visual loss is typically altitudinal, especially inferiorly.
• Visual loss commonly progresses over a few days after
onset but few (<10%) worsen over more than 10 days.
• Many (up to 56%) have relatively good visual acuity.
• Recurrences in the same eye are unusual.
• Subsequent involvement of the fellow eye is common,
with an estimated 25th percentile time to bilaterality of
32 months.
Optic Neuritis 489

OPTIC NEURITIS visual failure progressing over weeks or months and


stopping short of complete blindness in the second or
third decade of life.
DEFINITION • Central scotomas are present and optic atrophy develops.
Optic neuritis is an acute, usually unilateral, inflammatory, Recovery does not occur.
demyelinating optic neuropathy. • Occasionally dementia, ataxia and spastic paraplegia are
present.
TERMINOLOGY
• Papillitis: inflammation of the optic nerve that is Isolated and idiopathic optic neuritis (probably a
sufficiently anterior to cause swelling of the optic disc. forme fruste of MS)
• Retrobulbar neuritis: inflammation of the optic nerve
that is posterior to the optic nerve head so that the direct CLINICAL FEATURES
effects of inflammation are not visible on History (symptoms)
ophthalmoscopy. • Vision becomes blurred, dimmed or is lost in one eye
over hours to days, reaching its worst in a week or so.
EPIDEMIOLOGY • Pain in and behind the eye may precede, and commonly
• Incidence: 1.40 per 100 000 person years (2.28 per accompanies, the visual blurring in about 90% of patients.
100 000 person years for women, 0.53 per 100 000 The eye is commonly painful to move or to touch.
person years for men; age-adjusted). • Bright, brief flashes of light (phosphenes) precipitated by
– Seasonal: highest in spring and lowest in winter. eye movement or unexpected sounds, particularly in the
• Prevalence (lifetime): 0.6 (95% CI: 0.3–1.0) per 1000. dark or when the eyes are closed, may occur in the acute
• Age: peak age of onset is in the 20s and 30s. stage or, more commonly, during recovery.
• Gender: F>M (2–3:1).
Examination (signs)
PATHOLOGY • Visual acuity is impaired.
Inflammatory, demyelinating optic neuropathy. • Color vision is impaired.
Simultaneous bilateral optic neuritis is very rare. • A central or paracentral scotoma, particularly by using a
red target, is present in most patients; most optic nerve
ETIOLOGY AND PATHOPHYSIOLOGY fibers transmit information from the macular which
Multiple sclerosis (MS) processes central vision and is densely innervated.
• The most common and important cause; about 50–60% Consequently, optic nerve lesions tend to affect fibers
of patients with optic neuritis eventually develop multiple that are transmitting information predominantly from
sclerosis (see p.340). the macular. However, the classic central scotoma is not
• Simultaneous bilateral optic neuritis is seldom due to always present and one of a wide variety of other visual
MS. field defects may be present. In addition, visual field
abnormalities in the fellow eye may be detected by visual
A specific infectious or inflammatory disorder field testing which may evolve over subsequent follow-
Viral infections (usually bilateral) up suggesting concurrent bilateral involvement.
• Herpes zoster virus. • A relative afferent pupillary defect (see p.575).
• Measles. • A swollen optic disc on ophthalmoscopy (papillitis) in
• Epstein–Barr virus. about one-third of cases, in whom the inflammatory
• Cytomegalovirus. demyelination is close to the optic nerve head (614).
• HIV. Continued overleaf

Other infections 614


• Syphilis.
• Tuberculosis.
• Histoplasmosis.
• Cryptococcus.

Vaccinations
• Hepatitis B.
• Influenza.

Orbital cellulitis

Meningitis and encephalitis

Neuromyelitis optica (Devic’s disease)

Leber’s hereditary optic atrophy (see p.491) 614 Ocular fundus photograph showing a swollen optic disc
• Rare, variably inherited, males are more often affected (papillitis) due to demyelination close to the optic nerve head.
than females. (Courtesy of Mr M Wade, Department of Medical Illustrations,
• Subacute bilateral (simultaneous or sequential) painless Royal Perth Hospital,Western Australia.)
490 Cranial Neuropathies

Examination (signs) (continued) • Mitochondrial DNA analysis.


• The optic disc appears normal, in the acute phase at least, • MRI of the brain, including orbits: excludes optic nerve
in about two-thirds of patients with more posterior optic compression or infiltration and shows mild to profound
nerve lesions (retrobulbar optic neuritis). changes consistent with brain demyelination in about half
• Later, when symptoms have improved, optic atrophy of patients.
may be present (with pallor of the optic disc, particularly • CSF: often adds little additional information to the MRI
on the temporal side) and sometimes subtle disturbances results. Oligoclonal banding is present in most patients
of color vision, distortion of depth perception (Pulfrich’s who go on to develop clinically definite MS but most of
phenomenon), a paracentral scotoma, and a relative these patients also have an abnormal MRI. In a minority
afferent pupillary defect. of cases, patients with optic neuritis who have oligoclonal
bands in the CSF and a normal MRI will progress to MS
DIFFERENTIAL DIAGNOSIS within the next 2 years.
Progressive monocular visual failure
• Visible ocular lesions (glaucoma, cataract, senile macular DIAGNOSIS
degeneration). Clinical.
• Optic nerve infarction.
• Anterior ischemic optic neuropathy (see p.487). TREATMENT
• Optic nerve inflammation/infection: • Oral prednisone alone has no role in the treatment of
– Perioptic neuritis. acute demyelinating optic neuritis.
– Orbital and nasal suppuration, fungal infection: • Oral high-dose methylprednisolone (500 mg daily for
– Sarcoidosis. 5 days) improves recovery from optic neuritis at 1 and
– Meningitis: subacute and chronic. 3 weeks but has no effect at 8 weeks or on subsequent
• Optic nerve tumor: attack frequency.
– Primary: astrocytoma. • Intravenous methylprednisolone (1000 mg/day) for
– Secondary: nasopharyngeal carcinoma. 3 days followed by oral prednisone (1 mg/kg/day) for
• Optic nerve compression: 11 days is used by many clinicians for acute optic neuritis
– Orbital tumor. patients with brain MRI abnormalities suggestive of
– Dysthyroid eye disease. demyelinating disease and for those with substantial visual
– Tolosa–Hunt syndrome. loss. The rationale is that it hastens recovery of vision but
– Paget’s disease of the skull. it provides no long term benefit at 1 year and it is
– Anterior communicating artery aneurysm uncertain whether it reduces the rate of developing MS.
• Optic nerve irradiation.
PROGNOSIS
Progressive binocular visual failure Eye pain
• Glaucoma. Subsides in about a week.
• Diabetic retinopathy.
• Bilateral severe papilledema. Visual acuity
• Leber’s hereditary optic neuropathy. Commonly improves slowly over a few weeks. Lack of at least
• Nutritional deficiency: vitamin B12, vitamin E. some improvement in visual acuity within the first 3 weeks
• Optic nerve toxins: after onset, or worsening of vision after termination of a
– Alcohol and glycols. course of steroids, should be considered atypical and warrant
– Heavy tobacco consumption. re-assessment of the diagnosis. At 6 months, about 94% of
– Mercury. patients have vision of 6/12 (20/40) or better and 75% have
– Clioquinol. improved to 6/6 (20/20) or better. There is frequently some
– Chronic cyanide poisoning. residual impairment of color vision. Chronic progression with
– Drugs: chloramphenicol, chloroquine, desferrioxamine no improvement, resulting in permanent blindness, is rare and
(deferoxamine), ethambutol. should provoke a search for other causes.
• Optic chiasm compression: At 5 years, about half of patients report their vision to be
– Pituitary adenoma. ‘somewhat worse’ or ‘much’ worse than it had been before
– Craniopharyngioma. the optic neuritis, but only 20% have abnormal results on
– Meningioma of the tuberculum sellae, olfactory groove all of the four tests of visual function (visual acuity, contrast
or sphenoid wing. sensitivity, color vision, and visual field mean deviation).
– Hydrocephalus and dilatation of the third ventricle. The best predictors of visual recovery are baseline visual
– Carotid siphon or ophthalmic artery aneurysm. acuity, and short length of optic nerve signal abnormality
• Optic nerve irradiation. and extracanalicular location on MRI (<17.5 mm of optic
nerve involvement), but even those with no light perception
INVESTIGATIONS can have good recovery of visual acuity.
• Full blood count and ESR.
• Antinuclear antibody. Visual fields
• Syphilis serology. Most visual field defects also return to normal, although
• Chest x-ray. new and different patterns of defects develop in some
• Visual evoked potentials: delayed conduction in the visual patients over the course of follow-up in both eyes. A
pathways. minority of patients (about 13%) develop chiasmal and
Leber’s Hereditary Optic Neuropathy (LHON) 491

retrochiasmal visual field defects at some time during the LEBER’S HEREDITARY OPTIC
first year of follow-up, particularly those with an abnormal NEUROPATHY (LHON)
brain MRI at the onset of optic neuritis.

Recurrence of optic neuritis DEFINITION


Recurrence in the same or other eye is not unusual; vision Leber’s hereditary optic neuropathy is a maternally-inherited
is less likely to recover than after the initial attack. optic neuropathy caused by a mitochondrial DNA mutation
and manifesting as an acute or subacute loss of central vision
MS in both eyes in young adults, predominantly young men.
About 15% of patient will develop clinically definite MS
within the next 2 years, 22% within 5 years, 35% in 10 years, EPIDEMIOLOGY
and about 50–60% of patients with optic neuritis will • Incidence: rare, but the commonest cause of blindness
eventually develop MS. in otherwise healthy young men.
• Age: late adolescence and early adulthood. Mean age of
MS is less likely to occur onset: 23 years; rare over the age of 50 years, but
• Following optic neuritis in childhood than in adults. reported up to 70 years of age.
• With bilateral simultaneous onset. • Gender: M>F (4:1).
• With absence of pain.
• With marked disc edema. ETIOLOGY
• If there are no oligoclonal bands in the CSF. • Maternal inheritance.
• If the brain MRI is normal (lack of pain, mild visual • Heterogeneous mitochondrial DNA (mtDNA)
acuity loss, and the presence of optic disc swelling are mutations: homoplasmic missense point mutations have
also favorable prognostic factors in patients with no brain been found in at least eight genes that encode subunits
MRI lesions). of three biochemical complexes. The most common
‘primary’ mtDNA mutations are at the following
MS is more likely to occur nucleotide positions/base pairs:
• If previous ill-defined non-specific neurologic symptoms – 11 778 (accounts for 31–90% of LHON probands, and
are present. results in substitution of histidine for arginine in subunit
• If there is an history of previous optic neuritis. 4 of complex 1 of the mitochondrial respiratory chain).
• With increased CSF IgG. – 3460 (8–15% of probands).
• If the brain MRI is abnormal with three or more lesions – 14 484 (10–15% of probands).
suggestive of demyelinating disease: – 15 257.
– At 3 years follow-up, about 10% of patients with normal • Epigenetic factors: alcohol and tobacco abuse.
baseline MRI scans develop clinically definite MS,
compared with about 30% of patients with one or two CLINICAL FEATURES
MRI signal abnormalities and 45% of patients with three • Subacute, bilateral, sequential loss of central vision which
or more MRI lesions. is typically painless but may be associated with a constant
– At 5 years follow-up, about 16% of patients with normal dull pain. The interval between involvement of the two
baseline MRI scans develop clinically definite MS, eyes is rarely more than a few months.
compared with about 51% of patients with three or more • Abnormal color vision (e.g. Ishihara plates).
MRI lesions. • Central or centrocecal visual field defects.
• Possibly, if there is a family history of MS. • Fundus:
– Acute phase: normal, or hyperemia and swelling of the
optic disc, peripapillary telangiectasia, and dilatation and
tortuosity of the retinal vasculature, with or without
hemorrhages.
615 – Chronic phase: optic nerve pallor and loss of nerve fiber
layer develop, loss predominantly in the papillomacular
bundle (615).
• Associated features include dystonia, low amplitude
postural and action tremor of the upper limbs,
myelopathy, and cardiac conduction abnormalities.
Brainstem (midbrain) involvement has also been
described.
• Family history of visual loss is present in about one-half
of patients.

615 Optic fundus, showing optic atrophy, of a man with progressive loss
of central vision in one eye followed a few months later by involvement
of the fellow eye, who was found to have pathogenic mutation of
mitochrondrial DNA at base pair 11 778. Acute stage: hyperemia of the
optic disc with peripapillary microangiopathy but without fluorescein
leakage; chronic: progression to optic atrophy is usual.
492 Cranial Neuropathies

DIFFERENTIAL DIAGNOSIS PAPILLEDEMA


• MS: a high proportion of women with a mtDNA
mutation at base pair 11778 develop an illness
indistinguishable from MS. DEFINITION
• Other hereditary optic neuropathies: Papilledema is edema of the optic papilla or disc. Not a
– Autosomal dominant optic neuropathy (Kjer’s disease): disease but a sign of an underlying condition.
symmetric insidious onset of visual loss in the first decade
of life. ETIOLOGY
– Blue-yellow dyschromatopsia: Raised intracranial pressure (bilateral papilledema)
Centrocecal scotomas. Brain parenchymal disorders
Wedges of temporal disc atrophy. • Mass lesion: abscess, infarct, hematoma, tumor.
Abnormal gene in telomeric portion of long arm of • Cerebral edema: tumor, post head injury, post brain
chromosome 3. anoxia, idiopathic intracranial hypertension (benign
• Ischemic optic neuropathy. intracranial hypertension), lead poisoning, steroid
• Neurosyphilis. withdrawal, vitamin A intoxication.

INVESTIGATIONS CSF disorders


• Goldmann perimetry: central or centrocecal scotoma. • CSF circulatory block: aqueduct stenosis, intraventricular
• Visual evoked potentials to pattern reversal: show a delay tumor, ventricular outflow block.
(>100 ms) and reduced amplitude. • Impaired CSF absorption due to raised CSF protein or
• Fluorescein angiography. altered blood products: post subarachnoid hemorrhage,
• Venous blood mtDNA analysis, using the polymerase post meningitis, Guillain–Barré syndrome, hypertrophic
chain reaction (PCR) for a point mutation: the major ones polyneuritis, spinal cord tumor.
are at nucleotide positions 11 778, 3460, and 14 484.
• Full blood count: normal. Malignant hypertension
• ESR: normal.
• VDRL/RPR, TPHA: negative. Circulatory disorders
• ECG: usually normal, but may show a conduction • Lateral venous sinus thrombosis.
abnormality. • Jugular vein thrombosis.
• CT or MRI scan of the head and orbits: normal or high • Superior vena cava obstruction.
signal within the optic nerve on MRI short time • Hyperviscosity syndrome: polycythemia rubra vera,
inversion recovery (STIR) sequence. A symmetric area multiple myeloma, macroglobulinemia.
of increased signal intensity on T2W images in the dorsal
midbrain has been reported. Metabolic disorders
• CSF: normal, or oligoclonal bands in some patients. • Hypercapnia.
• Hypocalcemia (in children in particular).
DIAGNOSIS
Typical clinical features (e.g. positive family history in half Raised intranerve pressure (unilateral papilledema)
of cases, visual loss, ophthalmoscopic findings of circum- Optic nerve disorder
papillary telangiectasic microangiography, swelling of the • Orbital or optic nerve tumor.
nerve fiber layer around the optic disc [pseudoedema]), • Optic neuritis (see p.489).
absence of staining of fluorescein angiography, and venous • Ischemic optic neuropathy (see p.487).
blood analysis by PCR showing a mtDNA point mutation • Malignant thyrotoxic exophthalmos.
at nucleotide positions 11 778, 3460, or 14 484, or possibly
4160 and 15 257. Circulatory disorders
• Central retinal vein occlusion.
TREATMENT • Cavernous sinus thrombosis.
No effective medical (e.g. acetazolamide, prednisolone, and • Carotico-cavernous fistula.
intravenous dextran) or surgical (e.g. optic nerve sheath
decompression) treatment. PATHOPHYSIOLOGY
• Disc swelling indicates abnormal elevation of the optic
PROGNOSIS disc, generally due to blocked axoplasmic flow.
• Visual acuity typically deteriorates permanently to levels • It usually requires at least 2–4 hours to develop, even
of 6/60 or worse, although spontaneous recovery of with acute increases in intracranial pressure, and so its
vision can occur even years later. absence does not exclude increased intracranial pressure
• The probability of visual recovery varies widely, of less than several hours duration. Peripapillary
depending on the mutation and age of onset. hemorrhages may be seen more acutely.
• Good visual outcome correlates strongly with younger
age at onset.
• The presence of a ‘primary’ LHON mutation does not
guarantee that a patient will lose vision; patients with the
14 484 mutation have a substantially greater chance of
spontaneous recovery of vision (up to 50%) and better
final visual acuities.
Papilledema 493

CLINICAL HISTORY • Flame retinal perivenous hemorrhages on or around the


• Visual obscurations: brief, painless, monocular or disc.
binocular blurring or complete loss of vision (‘gray-outs’), • Obscuration of vessels at the edge of the disc.
precipitated by postural changes such as bending quickly • Opacity of the peripapillary nerve fiber layer.
and standing or exercise, and resolving within seconds. • Circular retinal folds around the disc and choroidal folds
They are due to temporary optic nerve ischemia as a may be seen.
result of further obstruction to venous outflow from the • Cotton wool spots – small round white blobs known as
orbit caused by raised intracranial or intraorbital pressure cytoid bodies, indicate very severe axonal damage.
(resisting venous outflow) and a small drop in systemic • Chronic papilledema (e.g. in malignant hypertension or
blood pressure (reducing arterial inflow). idiopathic intracranial hypertension) may be complicated
• Associated symptoms of the underlying cause (e.g. by secondary changes in the macular area: the so-called
headache). macular star is edema radiating out from the macular and
seriously interfering with vision (617).
PHYSICAL EXAMINATION
Visual acuity Other
• Normal, initially at least, if the papilledema is due to Check the blood pressure, look for proptosis and determine
raised intracranial or intraorbital pressure. if the proptosis is pulsatile.
• Visual acuity becomes impaired later as raised pressure
papilledema increases and spreads towards the macular SPECIAL FORMS
or because of macular hemorrhage or optic atrophy in The Foster–Kennedy syndrome
severe cases. A rare combination of papilledema in one eye and optic
atrophy in the other eye due a frontal tumor which is large
Visual fields enough to compress one optic nerve (and cause ipsilateral
Enlarged blind spot, and sometimes constricted visual field, optic atrophy) and increase the intracranial pressure to cause
particularly infero-nasally. papilledema in the contralateral eye. The differential
diagnosis is acute anterior ischemic optic neuropathy or
Ophthalmoscopy of the ocular fundus optic neuritis in one eye and optic atrophy due to previous
• Filling in of the optic cup in the optic nerve head. optic neuritis in the other eye. The distinction can often be
• Pink, hyperemic optic disc (616). made by the presence of impaired visual acuity in both eyes
• Blurred disc margin. in the latter circumstance.
• Swelling and elevation of the optic disc.
• Engorged, non-pulsatile retinal veins; the presence of
spontaneous venous pulsations indicate the CSF pressure
is <200 mm of water at the time of the observation.

616 617

616 Ocular fundus photograph showing early papilledema in a patient 617 Ocular fundus photograph showing severe chronic papilledema in
with idiopathic intracranial hypertension. Note the hyperemic optic disc, a patient with idiopathic intracranial hypertension. Note the swollen,
blurred disc margin, and engorged retinal veins. (616, 617 courtesy of hyperemic optic disc with obliteration of the optic cup, blurred disc
Mr M Wade, Department of Medical Illustrations, Royal Perth Hospital, margin, engorged retinal veins, obscuration of vessels at the edge of the
Western Australia.) disc, retinal hemorrhages around the disc and macular star (edema
radiating out from the macular).
494 Cranial Neuropathies

DIFFERENTIAL DIAGNOSIS HORNER’S SYNDROME


Unilateral optic disc swelling with normal
vision (papilledema)
• Normal optic disc: the nasal side of the normal disc often DEFINITION
looks a little blurred, and retinal venous pulsation is Horner’s syndrome presents as unilateral ptosis, miosis and
absent in about 25% of normal people. anhidrosis due to damage to the ipsilateral oculosympathetic
• Papillophlebitis (the ‘big blind spot syndrome’): a pathway.
benign, self-limiting condition.
• Idiopathic intracranial hypertension. EPIDEMIOLOGY
• Congenital disc anomalies: Prevalence (lifetime): 0.2 (95% CI: 0.04–0.4) per 1,000.
– Deep optic cup.
– High myopia (normal corrected vision). ANATOMY AND PATHOPHYSIOLOGY
– Myelinated nerve fibers. Cervical sympathetic pathway
– Optic nerve drusen (618, 619): refractile mass in the Begins in the posterolateral part of the hypothalamus, but
optic nerve head which may produce arcuate nerve there is a considerable degree of cortical control. There are
bundle-type visual field defects and show auto- three neurons:
fluorescence with fluorescein angiography.
– Prepapillary vascular loops. First neuron
– Tilted discs. Descends from the hypothalamus, through the dorsolateral
– Megalopapilla. brainstem and cervical spinal cord, to synapse in the
intermediomedial and intermediolateral cells (the ciliospinal
Optic disc swelling with abnormal vision centre of Budge) in the lateral gray matter of the
(anterior optic neuropathy) cervicothoracic spinal cord at levels C8 and T1. These cells
• Optic neuritis (papillitis) (see p.489). give rise to preganglionic fibers.
• Anterior ischemic optic neuropathy (see p.487).
• Optic disc infiltration: Second neuron
– Granuloma: e.g. sarcoidosis. Preganglionic fibers leave the upper thoracic spinal cord,
– Tumor: e.g. leukemia, astrocytoma, lymphoma, metastases. most via the white rami of the T1–T2 ventral nerve roots
• Optic disc neovascularization. (which lie close to the apex of the lung where they cross in
• Hereditary optic neuropathy (Leber’s). the pleural covering and loop around the subclavian artery),
and proceed through the stellate ganglion to synapse in the
INVESTIGATIONS superior cervical ganglion.
Fluorescein angiography (619) shows leakage of dye from
the disc. Further investigations depend on clinical clues to Third neuron
the underlying cause (e.g. CT or MRI scan of brain and Postganglionic fibers ascend in the cervical sympathetic
orbits, screen for a procoagulant state). trunk (in close proximity to the internal carotid artery),
enter the base of the skull on the surface of the internal
PROGNOSIS carotid artery, traverse the cavernous sinus where they join
• The course of papilledema is best followed by serial visual the first (ophthalmic) division of the trigeminal nerve. They
field testing and fundus photography. Papilledema may are carried in the nasociliary branch of the ophthalmic nerve
take 6–10 weeks to resolve after intracranial pressure through the superior orbital fissure into the orbit.
returns to normal. Vasomotor fibers in the nasociliary branch of the trigeminal
• Chronic, untreated papilledema can cause loss of visual nerve traverse the ciliary ganglion without synapsing to
function and blindness. The optic disc becomes gray or supply the blood vessels of the eye. Pupillodilator fibers on
pale (secondary optic atrophy). the nasociliary branch of the trigeminal nerve continue, by

618 619 618 Ocular fundus


photograph showing optic disc
drusen, which may be
mistaken for papilledema.
Drusen is a hyaline mass that
develops during life and may
cause a central scotoma or, if it
damages nerve fiber bundles in
the optic nerve head, an
arcuate scotoma.

619 Fluorescein angiography


showing auto-fluorescence in
optic disc drusen.
Horner’s Syndrome 495

passing around the eye, as the long ciliary nerves to CLINICAL FEATURES
innervate the pupil. History
Fibers carried in the third cranial (oculomotor) nerve Asymptomatic unless the patient or others notice the ptosis
innervate the superior and inferior tarsus muscles of Müller or small pupil.
and orbitalis. These muscles assist eye opening by
attachment to the tarsal plates, opposing the action of Examination
orbicularis. When paralysed, the upper lid is ptosed and the Horner’s syndrome
lower lid pulls up – so-called ‘upside down’ ptosis, or ptosis • Ptosis of the upper lid (sympathetic fibers supply the smooth
of the lower lid – narrowing the palpebral fissure and muscle of the upper eyelids) that is partial and not complete
producing an apparent enophthalmos. (as it is in a IIIrd nerve palsy), and slight elevation (‘upward
ptosis’) of the lower lid (paresis of Müller’s muscles)
ETIOLOGY resulting in narrowing of the palpebral fissure (620).
• Damage to the cervical sympathetic pathway. Isolated ‘upside down ptosis’ of the lower lid may occur.
• A cause is found in about 60% of cases. • Pupillary constriction (miosis) occurs. The anisocoria in-
• Hypertension and diabetes mellitus are frequently creases in darkness. Occasionally pupillary involvement
present in the undiagnosed group suggesting ischemia can only be demonstrated on pharmacologic testing (see
of the oculosympathetic pathway. below).
• Anhidrosis (impaired sweating) on the ipsilateral side of
Congenital: 3% the face may occur if the lesion is proximal to the carotid
bifurcation (since sympathetic fibers to the face course
Acquired: 57% with the external carotid artery). Anhidrosis of the
Brainstem medial side of the forehead may occur if the lesion is
• Stroke: lateral medullary syndrome (5%). distal to the carotid bifurcation. Anhidrosis occurs in 5%,
• Tumor. usually with preganglionic lesions. Vascular dilatation in
the face and conjunctiva is transient.
Spinal cord • Iris heterochromia may be seen in congenital Horner’s
• Tumor. syndrome; Horner’s syndrome developing in utero or
• Cervical disc extrusion (3%). infancy interferes with pigmentation of the iris so the
• Syringomyelia. eyes are of different colors (620).
• Meningitis. • Enophthalmos is apparent and not real; it is not a feature
of oculosympathetic palsy.
Apex of lung
• Coronary artery bypass surgery (0.2–7.7%). N.B. Bilateral Horner’s syndrome from a central
• Tumor. (brainstem or cervical spinal cord) lesion can be easily
• Pneumothorax. missed because the ptosis is symmetric and the pupils are
symmetrically equal and react to light, but are small.
Neck
• Trauma.
• Carotid artery dissection, thrombosis.
• Thyroid tumors, surgery.
• Cervical rib.

Cavernous sinus and orbit


• Cluster headache (migrainous neuralgia) (12%).
• Raeder’s paratrigeminal syndrome.

620

620 Facial photograph showing a left Horner’s syndrome. Note the


narrowing of the palpebral fissure due to mild incomplete ptosis of the
upper lid and slight elevation of the lower lid.The pupil is small and
constricts to light and accommodation.
496 Cranial Neuropathies

Associated features Central preganglionic or peripheral


• Chest: open heart surgery scar. post-ganglionic lesion?
• Neck: wasting of small muscles of hand due to ipsilateral • Wait until at least 12 hours have elapsed after the cocaine
T1 radiculopathy; thyroidectomy scar. test.
• Cavernous sinus and orbit lesions: the pupil is often • Hydroxyamphetamine 1–2% or pholedrine eyedrops:
semidilated or fixed and dilated because the lesion also apply one drop to the affected eye, repeat after a few
damages parasympathetic fibers running with the IIIrd minutes, and determine the change in pupillary size after
cranial nerve to the constrictor fibers of the pupil. 30 minutes. Hydroxyamphetamine releases noradrenaline
Raeder’s syndrome also includes hemicrania with or from nerve endings and dilates pupils with an intact third
without cranial nerve III, IV, V, and VI dysfunction. order neuron. So, normal pupils dilate as do small pupils
caused by a preganglionic (central) oculosympathetic
DIFFERENTIAL DIAGNOSIS palsy. Small pupils due to a postganglionic oculosympa-
Small pupil (miosis) thetic lesion fail to dilate with hydroxyamphetamine.
• Congenital.
• Trauma to the iris (usually irregular). The site of the lesion can be deduced further by eliciting
• Pontine lesion. relevant associated clinical features.
• Argyll Robertson pupil.
• Pilocarpine and other constricting eyedrops such as INVESTIGATIONS
carbachol, metacholine, physostigmine, neostigmine, Determined by the site of the lesion in the oculosympathetic
isoflurophate, ecothiophate (echothiopate), demecarium, pathway (which is determined by the presence and absence
aceclidine. of coexistant focal neurologic symptoms and signs, and the
• Organophosphorus poisoning. results of eyedrop tests (see above).
• Opiates, barbiturates, chloral hydrate, cholinergics,
lignocaine, marijuana, phenothiazines (bilateral). Preganglionic Horner’s syndrome
• Autonomic neuropathy. • MRI brain and cervical spinal cord.
• Old age (normal). • CSF examination.
• Physiologic anisocoria: about 25% of the normal • Chest x-ray.
population have pupils of unequal size; the difference is • Chest CT scan.
<1 mm in about 20% and >1 mm in up to 5%. The
asymmetry remains constant in the light and dark. Postganglionic Horner’s syndrome
• Carotid artery imaging (ultrasound, magnetic resonance
Ptosis angiography, catheter contrast angiography).
• Congenital. • MRI neck.
• Midbrain lesion. • MRI head.
• Neurosyphilis (see p.312).
• IIIrd cranial (oculomotor) nerve palsy (see p.499). TREATMENT
• Myasthenia gravis: ptosis may be variable and asymmetric Depends on the cause.
(see p.657).
• Myopathies: myotonic dystrophy, ocular dystrophies, PROGNOSIS
mitochondrial cytopathy (chronic progressive external Depends on the cause. Generally benign in isolated, new
ophthalmoplegia), polymyositis, dysthyroidism. onset postganglionic Horner’s syndrome.
• Levator dehiscence–disinsertion syndrome due to ageing,
inflammation, trauma, surgery or ocular allergy.
• Trauma to the orbit.
• Pseudoptosis due to eyelid inflammation (allergy,
blepharitis, conjunctivitis), hemangioma, enophthalmos,
Duane’s orbital retraction syndrome, trachoma, ptosis
adiposis, plexiform neuroma, amyloid infiltration and
chronic Bell’s palsy.

DIAGNOSIS
Horner’s syndrome or not?
Horner’s syndrome is characterized by:
• Normal pupil reaction to light in both eyes.
• Anisocoria is greater in darkness than in light.
• ‘Dilatation lag’ of the smaller pupil following sudden
darkness.
• Cocaine 2–10% eyedrops: small pupil fails to dilate.
Cocaine dilates normal pupils by preventing the reuptake
of noradrenaline from sympathetic nerve endings.
Horner’s syndrome, due to disruption of the sympathetic
pathway at any level prevents the release of noradrenaline
from the terminal nerve endings and the pupil fails to
dilate in response to cocaine.
Holmes–Adie Syndrome 497

HOLMES–ADIE SYNDROME PATHOLOGY


Rare post mortem examinations reveal ciliary ganglion cell
loss.
DEFINITION
The syndrome is a combination of a Holmes–Adie (tonic) ETIOLOGY AND PATHOPHYSIOLOGY
pupil and absent or depressed deep tendon reflexes. Adie’s tonic pupil
The tonic pupil is believed to result from damage to the
EPIDEMIOLOGY ciliary ganglion or the postganglionic cholinergic para-
• Incidence: uncommon. sympathetic fibers projecting from the ganglion to the iris
• Age: adolescents or young adults. sphincter. There is denervation of the postganglionic supply
• Gender: females mainly. to the sphincter pupillae and the ciliary muscle. This may
follow infection such as herpes zoster or trauma, but is
ANATOMY AND PHYSIOLOGY OF THE usually idiopathic. It may be part of a mild polyneuropathy.
PUPIL SIZE AND REACTIONS
• Pupil size is controlled by a circle of constrictor fibers Hyporeflexia
innervated by the parasympathetic nervous system and a Degeneration in the dorsal root ganglia and dorsal columns
ring of radially arranged dilator fibers controlled by the occurs, which leads to impaired synaptic transmission in the
sympathetic nervous system (see p 575). spinal monosynaptic reflex arc, accounting for the absent
• The resting size of the pupil is governed by the light reflexes.
intensity falling on the eye and depends on the integrity
of the parasympathetic nervous system. CLINICAL FEATURES
• Changes in pupil size do not usually affect vision, and are History
usually asymptomatic. However, a fully dilated pupil on • The syndrome is usually asymptomatic, other than some
one side will not focus close up, and will cause blurring photosensitivity to bright light.
on attempted reading. • Patients may present with sudden onset of blurred vision.

Parasympathetic pathways Examination


• Light falling on the retina of one eye is conveyed in the • One pupil is larger than the other (621), but in about
optic nerve to the optic chiasm. The impulses then 20% of cases both pupils are dilated.
decussate and are conveyed in both optic tracts to both • The pupil reacts very sluggishly to light but with
lateral geniculate bodies. prolonged exposure to light it will pass through the full
• About 10% of the fibers to the geniculate bodies are para- range of constriction. When the light stimulus is
sympathetic fibers that subserve the light reflex and are removed, the pupil dilates very slowly, hence the
relayed in the periaqueductal gray matter to both Edinger– alternative name of ‘myotonic pupil’.
Westphal nuclei (see pp.500, 575) in the high midbrain. Continued overleaf
• The parasympathetic fibers join the IIIrd cranial
(oculomotor) nerve, and are carried in a superficial and
dorsal position on the nerve (which may explain the
variable abnormalities of the pupil in IIIrd cranial nerve
palsies [see p.499]), to the orbit.
• The final relay of the parasympathetic pathway is in the
ciliary ganglion, which lies in the posterior orbit on the
branch of the IIIrd nerve to the inferior oblique muscle. 621
• The postganglionic fibers then enter the eye via 8–10
short ciliary nerves which subdivide into 16–20 branches
that pass around the eye to reach the sphincter pupillae;
about 3% of the fibers innervate the sphincter pupillae
and 97% the ciliary body, which is a muscle that alters the
shape of the lens for focusing.
• The Edinger–Westphal nuclei are also stimulated by
activity in the adjacent IIIrd nerve subnuclei which
control the medial rectus muscles. Consequently,
activation of the medial recti not only leads to
convergence of the eyes but also activation of the
Edinger–Westphal nuclei which, in turn, leads to
pupillary constriction as a result of simultaneous
contraction of the sphincter pupillae and the ciliary 621 Dilated Adie pupil of a young woman who presented having woken
muscle. This is the basis of the accommodation reflex, one morning and, on looking in the mirror, noted that the left pupil was
which is a much more powerful stimulus to larger than the right. The left pupil reacted extremely sluggishly to
pupilloconstriction than the light reflex. prolonged maximal light stimulation but much better to accommodation
(light-near dissociation), after which it remained tonically constricted and
Sympathetic pathways redilated very slowly.The affected left pupil also constricted promptly to
See Horner’s syndrome (p.494). 0.1% solution of pilocarpine, indicating denervation supersensitivity.
(Courtesy of Mr Alan Hamilton, Department of Medical Illustrations, Royal
Perth Hospital,Western Australia.)
498 Cranial Neuropathies

Examination (continued) • Amyloidosis.


• Whenever a pupil reacts poorly to light, the near reflex • Myotonic pupil.
should be tested by having the patient fixate on their
own finger tip held immediately in front of their nose. INVESTIGATIONS
The tonic Adie pupil constricts with convergence (reacts Determined by suspected and likely differential diagnoses
to accommodation). Because convergence and pupil (above).
constriction depend on adequate close vision, reading
glasses may need to be worn or the patient simply asked DIAGNOSIS
to look at the tip of their nose. • Dilated pupil(s) ± depressed knee and ankle reflexes.
• Knee and ankle jerks may be depressed or absent. • Poor light reaction of the dilated pupil.
• More anisocoria in light than in darkness.
SPECIAL FORMS • Test for cholinergic denervation supersensitivity with
Ross’ syndrome pilocarpine 0.1% (i.e. low dose) or methacholine 2.5%
• The triad of Adie’s tonic pupil, hyporeflexia, and eye drops: causes Adie’s pupil (but not the normal pupil,
segmental anhidrosis constitutes Ross’ syndrome. third nerve palsy pupil, or Argyll Robertson pupil) to
• Orthostatic hypotension, reduced heart rate responses to constrict because of denervation supersensitivity of
Valsalva maneuver, and other autonomic disturbances receptors. With 1% pilocarpine, both Adie’s pupil and the
may occur in patients with Ross’ or Holmes–Adie pupil of a third nerve palsy will dilate. On slit lamp
syndrome. examination, the margin of the iris may be seen to
• Ross’ syndrome results from injury to sympathetic and undulate.
parasympathetic ganglion cells or to their postganglionic
projections. TREATMENT
No specific treatment.
DIFFERENTIAL DIAGNOSIS
Large pupil (mydriasis) PROGNOSIS
Unilateral Eventually the pupil may become small and irregular, very
• Parasympathetic nervous system lesion (i.e. IIIrd cranial much like an Argyll Robertson pupil (see p.576).
nerve lesion).
• Trauma to the iris (usually irregular).

Bilateral
• Autonomic neuropathy.
• Parinaud’s syndrome.
• Anxiety (bilateral and symmetric).
• Mydriatic and cycloplegic eyedrops that dilate the pupil
(mydriatic) and paralyze the ciliary muscle (cycloplegic)
respectively (i.e. antimuscarinic agents), such as atropine,
homatropine, cocaine, cyclopentolate, adrenaline,
phenylephrine, hydroxyamphetamine, oxyphenonium,
scopolamine, tropicamide.
• Mydriatic agents (sympathomimetic or muscarinic
agents) such as anticholinergics (atropine, belladonna,
scopolamine, propantheline, jimsonweed, nightshade
[belladonna]), tricyclic antidepressants, trihexyphenidyl,
benztropine, antihistamines (diphenhydramine, chlor-
pheniramine), phenothiazines, glutethimide, ampheta-
mines, cocaine, ephedrine, adrenaline, ethanol, botu-
linum toxin, snake venom, barracuda poisoning,
tyramine, hemicholinium, thiopental, lysergic acid
diethylamide, fenfluramine (patients on reserpine).
• Hypocalcemia.
• Hypermagnesemia.
• Brain death.
• Normal in infants (bilateral).

Light-near dissociation
• Severe anterior visual system dysfunction (e.g. severe
glaucoma, bilateral optic neuropathy).
• Neurosyphilis (Argyll Robertson pupils): associated with
miosis, irregular pupils, poor dilatation, and usually
relatively normal vision.
• Rostral dorsal midbrain lesion (Parinaud’s syndrome).
• Aberrant IIIrd cranial nerve regeneration.
• Diabetes (out of proportion to any retinopathy).
IIIrd, IVth, and VIth Cranial Nerve (Ocular Motor) Neuropathies 499

IIIRD, IVTH, AND VITH CRANIAL NERVE Two other pathways for vertical saccadic (and pursuit,
(OCULAR MOTOR) NEUROPATHIES see below) eye movements involve ascending pathways from
the pontine centres to the mesencephalic reticular
formation: the central tegmental tract, which terminates in
DEFINITION the pretectum, in the nucleus of the posterior commissure
Ocular motor neuropathies are defined as disorders of the (mediating upgaze); and a bundle that passes around the
IIIrd (oculomotor), IVth (trochlear), and VIth (abducens) nucleus of Cajal and Darkschewitz to the rostral interstitial
cranial nerves which carry lower motor neuron fibers from nucleus of the MLF (mediating downgaze).
the midbrain and pons, through the meninges, cavernous Conjugate horizontal gaze is accomplished by activation
sinus, and supraorbital fissure into the orbit to supply the of the contralateral frontal eye field, followed by activation
extraocular muscles and elevators of the upper eyelid. of the ipsilateral PPRF, and then simultaneous innervation
These disorders, along with disorders of the extraocular of the ipsilateral lateral rectus and contralateral medial
muscles that are innervated by these nerves, cause rectus, the latter through fibers that run in the medial
dysconjugate eye movements and thus binocular diplopia portion of the contralateral MLF.
(unless either eye is closed, blind or amblyopic). Conjugate vertical gaze is accomplished by activation of
the frontal eye fields, followed by activation of the dorsal
ANATOMY ‘transthalamic’ bundles and pontine centres, which project
Supranuclear pathways for voluntary vertical and to the rostral midbrain and innervate the oculomotor (IIIrd
horizontal conjugate gaze cranial nerve) nucleus, riMLF, iC, and posterior commis-
Rapid eye movements (saccades) sure. Saccades are modulated by the posterior vermis of the
Saccades are generated in area 8 of the contralateral frontal cerebellum.
cortex. Direct, and probably more importantly indirect,
projections descend through the corona radiata and anterior Smooth pursuit eye movements
limb of the internal capsule. At the level of the rostral • The object to be followed is located by a voluntary eye
diencephalon, two bundles separate: movement (saccade).
• A dorsal ‘transthalamic’ bundle, which is predominantly • The eyes ‘lock in’ on the object and then only move in
uncrossed, courses through the thalamus to terminate response to movement of the object.
diffusely in the midbrain pretectum, superior colliculus, • Focus is maintained on the macular part of the retina by
and periaqueductal gray matter. An offshoot of these pathways from the retina, optic nerve, optic tract and
fibers projects to the rostral part of the oculomotor optic radiation to the primary visual cortex (area 17),
(IIIrd cranial nerve) nucleus and to the ipsilateral rostral from where visual information is relayed to the ipsilateral
interstitial nucleus of the medial longitudinal fasciculus parieto-occipital cortex, mainly area 7 of the parietal
(riMLF) and interstitial nucleus of Cajal (iC) in the cortex and area 19 of the ipsilateral visual cortex, as well
midbrain, which are involved in vertical eye movements. as other regions of the adjacent anterior occipital and
• A more ventral ‘capsular-peduncular’ bundle descends superior temporal lobes.
through the posterior limb of the internal capsule and • From the ipsilateral parieto-occipital cortex, corticotectal
most medial part of the basis pedunculi (cerebral and corticotegmental fibers descend to the superior
peduncle), to partially decussate in the rostral pons and colliculus and rostral midbrain (vertical pursuit) and the
terminate mainly in the paramedian pontine reticular ipsilateral dorsolateral pontine nuclei (horizontal pursuit),
formation (PPRF), which in turn, projects to the VIth which project to the flocculus and dorsal vermis of the
cranial nerve (abducens) nucleus. cerebellum.
• Movements are modulated by the ipsilateral vestibulo-
The final common pathway for horizontal saccadic (and cerebellum (flocculus and nodulus).
pursuit, see below) eye movements involves the low lateral • There is also a large forward projection that gives the
pontine centres for horizontal gaze (on the right and left), frontal eye fields information as to where to direct
which comprise the PPRF, abducens and vestibular nuclei, voluntary gaze. Thus a direct relay from the eye to the
and MLF. The PPRF functions as a relay station for the visual cortex and ipsilateral parieto-occipital cortex
horizontal and probably for the vertical saccade pathways. controls ocular pursuit.
The abducens nucleus contains two groups of neurons, • Paralysis of both voluntary saccadic gaze and pursuit gaze
abducens motor neurons, which project to the ipsilateral usually indicates damage at the subthalamic level, where
lateral rectus and abducens internuclear neurons, which both pathways come together (the Roth–Bielschowsky
project via the contralateral MLF to the medial rectus syndrome).
neurons of the (contralateral) IIIrd cranial nerve
(oculomotor) nucleus. The MLFs are fasciculi (pathways)
in the medial pontine and mesencephalic tegmentum,
connecting the ocular motor nuclei of the IIIrd and VIth
cranial nerves.
500 Cranial Neuropathies

Nuclear and infranuclear pathways for voluntary VIth (abducens) cranial nerve
vertical and horizontal conjugate gaze • Nuclei. A paired group of cells in the floor of the fourth
IIIrd (oculomotor) cranial nerve ventricle, adjacent to the midline, at the level of the lower
• Nuclei: pons.
– Nuclei consist of several paired groups of nerve cells in • Pons:
the midbrain, adjacent to the midline and central to the – The VIth nerve courses ventrally and medially, traversing
aqueduct of Sylvius, at the level of the superior colliculi. the corticospinal tracts to emerge from the low pons
– The groups of nerve cells innervate the levator of the adjacent to the pontomedullary junction in the midline,
eyelid, superior and inferior recti, inferior oblique, and deep in the posterior fossa.
medial rectus. The medial and inferior recti and inferior – The intrapontine portion of the facial nerve loops around
oblique have strictly homolateral representation in the the VIth nerve nucleus before it turns anterolaterally to
oculomotor nuclei, the superior rectus receives only make its exit.
crossed fibers, and the levator palpebrae superioris has • Pre-pontine. The nerve ascends on the front of the pons
bilateral innervation in this dorsal–ventral order. and then angles sharply forwards over the tip of the
– The parasympathetic portion of the oculomotor nucleus petrous bone, and into the canal under the petroclinoid
(the so-called Edinger–Westphal nucleus) is central and ligament (Dorello’s canal) to enter the back of the
dorsal in the midbrain, and innervates the pupillary cavernous sinus.
sphincters and the ciliary bodies (muscles of accommoda- • Cavernous sinus. The nerve lies free in the sinus.
tion). • Orbit. The nerve enters the orbit through the superior
• Midbrain. The IIIrd nerve fibers course ventrally in the orbital fissure, and passes laterally to reach the lateral
midbrain, traversing the MLF, red nucleus, substantia rectus muscle.
nigra, and medial part of the cerebral peduncle.
• Interpeduncular cistern. The IIIrd nerves emerge from Extra-ocular muscles
the midbrain between the cerebral peduncles, and then • The lateral rectus muscle of one eye and the medial
splay out as they pass anteriorly, lying between the rectus muscle of the other work as a yoked pair to
posterior cerebral arteries and the superior cerebellar produce horizontal (lateral) eye movements.
arteries. They then run parallel to the posterior • The superior and inferior rectus muscles of each eye work
communicating arteries, until they enter the cavernous together (one relaxes as the other contracts) in producing
sinus. vertical eye movements. Their effect is maximal when the
• Cavernous sinus. The nerves lie between the two layers eye is abducted, that is, looking outwards, as in this
of dura that form the lateral wall of the cavernous sinus. position the line of pull of the muscles is along the
• Orbit. The nerve enters the orbit through the superior vertical axis of the eye.
orbital fissure and divides into two main branches: • The superior and inferior oblique muscles each work
– The upper branch supplies the eyelid and superior rectus together in producing vertical eye movements,
muscle. particularly when the eye is adducted, that is, looking
– The lower branch supplies the medial rectus, inferior inwards, because in this position their line of pull is along
rectus, and inferior oblique and, via the ciliary ganglion, the vertical axis of the eye.
the pupil.
ETIOLOGY AND PATHOPHYSIOLOGY
IVth (trochlear) cranial nerve Midbrain (IIIrd and IVth cranial nerves) and pons
• Nuclei. The nuclei lie in the dorsal midbrain, just caudal (VIth cranial nerve)
to those of the oculomotor nuclei and at the level of the • Stroke (see p.192).
inferior colliculi. • Migraine (ophthalmoplegic: IIIrd cranial nerve only).
• Midbrain. The fibers decussate (cross over) a short • Encephalitis (e.g. Listeria) (see p.292).
distance from their origin, around the periaqueductal • Wernicke’s encephalopathy (see p.460).
gray and in the superior medullary velum, before • MS (see p.340).
emerging on the dorsal surface of the brainstem, just • Tumor (see p.357).
caudal to the inferior colliculi. The right trochlear nerve
thus originates in the left trochlear nucleus and vice versa. Meninges
• Cavernous sinus. After encircling the brainstem, the • Posterior communicating artery aneurysm (IIIrd cranial
nerve passes forwards on the opposite side to enter the nerve only) (622).
cavernous sinus. • Basilar artery aneurysm or ectasia.
• Orbit: • Infective meningitis: acute and chronic (see pp.273, 279).
– The nerve enters the roof of the orbit through the • Malignant meningitis.
superior orbital fissure and in doing so crosses the IIIrd • Meningovascular syphilis.
nerve. • Tumor: chordoma, nasopharyngeal carcinoma.
– In the orbit it passes around a trochlea (pulley) in the • Diabetes.
anteromedial orbital roof just under the inner end of the • Sarcoidosis.
eyebrow. • Inflammatory neuropathies: Guillain–Barré syndrome
(Miller Fisher syndrome).
• Infectious neuropathies: herpes zoster, HIV infection,
borrelia (Lyme disease).
• Ischemic neuropathies (at the level of meninges and
along the course of the nerve).
IIIrd, IVth, and VIth Cranial Nerve (Ocular Motor) Neuropathies 501

Petrous bone apex (VIth cranial nerve only) CLINICAL FEATURES


• Raised intracranial pressure. Assessment of diplopia
• Infection: middle ear. The eyes are normally positioned so the image falls on
• Tumor: nasopharyngeal carcinoma, chondrosarcoma exactly the same spot on the retina of each eye. The
(623). symmetric and synchronous movement of the eyes is termed
conjugate movement or gaze (‘conjugate’ means ‘yoked’ or
Cavernous sinus ‘joined together’). The slightest displacement of either eye
• Pituitary adenoma/apoplexy (see p.268). causes diplopia. Most nerve lesions causing extra ocular
• Meningioma sphenoid wing (see p.374). muscle weakness produce obvious dysconjugate gaze, but
• Nasopharyngeal carcinoma. occasionally, mild lesions or other diseases may cause
• Metastases. diplopia in which the degree of weakness is too slight to be
• Carotid siphon aneurysm expansion or rupture. observed (i.e. the eyes appear to move conjugately but the
• Carotico-cavernous fistula. patient still complains of diplopia). This may be due to
• Cavernous sinus thrombosis. monocular diplopia (due to refractive errors, vitreous
lesions, retinal lesions in the affected eye, or hysteria) or
Superior orbital fissure binocular diplopia (due to a misalignment of the visual
• Sphenoid wing meningioma (see p.374). axes). In this situation the cover test is useful.
• Optic sheath meningioma. The fundamental principle of the cover test is that when
• Metastases. the patient looks in the direction of action of the weak
• Orbital pseudotumor. muscle, the misalignment of the visual axes increases (i.e.
• Tolosa–Hunt syndrome. the disparity in eye movements is greater), and the degree
of separation of the two images increases. The true image
Orbit remains on the macula of the ‘good eye’ but the false image
• Tumor. moves progressively further away from the macula of the
• Cellulitis. lagging eye. The false image is therefore always projected as
the outer image of the two. If the examiner asks the patient
to follow an object until double vision is maximal, and then
determines which eye must be covered to obliterate the
outer (false) image, the affected eye is identified. The weak
muscle can then be deduced from the direction of gaze of
the affected eye.

622 623

622 CT brain scan, three slices in the axial plane, of a patient with a left ocular motor
(IIIrd cranial) nerve palsy due to a ruptured aneurysm of the left posterior
communicating artery.The lower slice, on the left, shows the malalignment of the visual
axes due to the left ocular motor (IIIrd cranial) nerve palsy (the left eye is abducted due
to unopposed action of the left lateral rectus).The middle slice shows some blood in the
pre-pontine cistern and left medial temporal lobe of the brain (arrows).The higher slice,
on the right, shows subarachnoid blood in the suprasellar cistern, the left ambient cistern,
and left quadrigeminal cistern (arrows).

623 PDW axial MRI showing a large mass arising on the right side of the skull base at
the petrous apex, projecting posteriorly into the posterior fossa and anteriorly into the
temporal fossa. Note that the internal carotid artery is elevated by the mass (arrows).
This feature is typical of chondrosarcomas.The calcification (which is frequent) is better
visualized on CT scan. The patient had presented with a painful VIth nerve palsy, which is
also said to be typical, and other cranial nerve palsies had developed later.
502 Cranial Neuropathies

IIIrd cranial nerve (oculomotor nerve) lesion VIth cranial nerve (abducens nerve) lesion
Diplopia Diplopia
• Due to weakness of all extraocular muscles except the • Due to weakness of lateral rectus muscle (625).
lateral rectus and superior oblique (624). • In the primary position of gaze, there is unopposed
• In the primary position of gaze, the eye is looking ‘down action of the medial rectus, and the eye is adducted.
and out’ due to unopposed action of the lateral rectus • On attempted lateral gaze to the side of the lesion (i.e.
(which abducts the eye ‘out’) and the superior oblique abduction of the eye), the diplopia is maximal.
(which rotates the eye ‘out’ [i.e. intorsion] when the eye
is abducted and the patient attempts to look down). DIFFERENTIAL DIAGNOSIS
Ophthalmoplegia
Ptosis Latent strabismus
• Due to damage to the parasympathetic fibers, which run • Congenital strabismus (squint) causes dysconjugate gaze
with the IIIrd nerve, to the levator palpebrae superiorus. but no diplopia because of longstanding suppression of
• If complete, covers the eye and so abolishes the diplopia. vision and amblyopia in one eye.
• A latent strabismus (squint) may break down in adult life,
Dilated pupil with impaired response to light and lead to intermittent diplopia, under conditions of an
and accommodation intercurrent illness, fatigue, or drowsiness; or following
• Due to damage to the parasympathetic pupilloconstrictor the development of uncorrected impaired vision in either
nerve fibers, which run along the outside of the IIIrd eye, which makes it difficult for the patient with a
nerve. This is almost always the case when the IIIrd lifelong squint to maintain binocular vision.
nerve is compressed by a structural (‘surgical’) lesion • A history of a childhood squint or previous orthoptic
such as a posterior communicating artery aneurysm. exercises is often present.
• N.B. The pupil is usually spared (i.e. normal) if the IIIrd • The examination reveals normal monocular eye
nerve palsy is caused by ischemia to the nerve as result of movements.
occlusion of the vasa nervorum (e.g. diabetes, arteritis) • Cover test:
which supplies the inner fibers of the nerve. – Cover one of the patient’s eyes and ask the patient to fix
• The pupil may not be fully dilated, and appear to be gaze on an object held 40 cm (16 in) away.
fixed, in the case of a IIIrd nerve palsy combined with a – Uncover the eye, and the uncovered eye will be seen
Horner’s syndrome, as may occur with a cavernous sinus immediately to be deviated inwards or outwards before
lesion. quickly returning to align with the fixating eye.
– This unmasks a latent deviation requiring positive muscle
IVth cranial nerve (trochlear nerve) lesion action to keep it compensated. Fatigue allows the eye to
Diplopia drift, resulting in intermittent diplopia, such as in the
• Due to weakness of the superior oblique muscle. evenings watching TV.
• In the primary position of gaze, there is oblique
separation of the images, due to unopposed action of the Congenital oculomotor apraxia
inferior oblique which rotates the eye (i.e. extorsion). Seen in children who have difficulty with horizontal eye
The diplopia can be abolished by tilting the head to the movements and need to blink to produce them.
side contralateral to the lesion.
• The diplopia is maximal, and thus exacerbated, when Impaired conjugate vertical gaze (supranuclear
looking down and inwards (e.g. as when descending vertical gaze palsy)
stairs, or looking at one’s feet or nose), which is the • Midbrain stroke (626).
primary action of the superior oblique. • Progressive supranuclear palsy.
• Huntington’s disease, Parkinson’s disease and some other
parkinsonian syndromes.
• Hydrocephalus.
624 • Parinaud’s syndrome (e.g. pineal tumor).
• Wernicke’s encephalopathy.
• Autosomal dominant and idiopathic cerebellar ataxias.
• Vitamin E deficiency.
• Whipple’s disease.
• Hexosaminidase deficiency (GM2 gangliosidoses).
• Niemann–Pick disease.
• Miller Fisher syndrome (probably nuclear/infranuclear).
• Elderly people (particularly upgaze and convergence).

Impaired conjugate horizontal gaze (supranuclear


horizontal gaze palsy)
• Contralateral frontal lobe (eyefield 8) lesion: e.g. stroke,
tumor, abscess.
• Ipsilateral low pontine (abducens nuclear complex/pon-
624 Ptosis of the left eyelid with the left eye looking down and out tine lateral gaze centre) lesion: e.g. stroke, tumor,
due to a left oculomotor (IIIrd cranial) nerve palsy, with sparing of the demyelination (627, 628).
pupil, in a patient with diabetes mellitus.
IIIrd, IVth, and VIth Cranial Nerve (Ocular Motor) Neuropathies 503

625 626

625 The nine cardinal positions of gaze showing features of a right 626 Bilateral ptosis, supranuclear vertical gaze pareses (particularly up
abducens (VIth cranial) nerve palsy (due to trauma). Note the inability gaze), and skew deviation (left eye lower [hypotropia]) due to a rostral
to abduct the right eye on attempted lateral gaze to the right due to midbrain ischemic stroke. (624–629 courtesy of Mr M Wade,
weakness of the right lateral rectus muscle. In the primary position of Department of Medical Illustrations, Royal Perth Hospital,Western
gaze (looking straight ahead) the right eye is deviated inwards, causing Australia.)
diplopia, due to the unopposed action of the right medial rectus.

627 628

627, 628 Impaired and dysconjugate lateral gaze to the left due to a left low pontine ischemic stroke affecting the pontine lateral gaze centre.

Internuclear ophthalmoplegia (629) 629


Damage to the MLF, which connects the IIIrd nerve
nucleus above with the opposite VIth nerve nucleus below,
causes the medial rectus ipsilateral to the MLF lesion to fail
to contract synchronously with the contralateral lateral
rectus on attempted lateral gaze to the contralateral side (i.e.
away from the side of the lesion). A unilateral lesion of the
MLF therefore results in incomplete or slow adduction of
the ipsilateral eye but normal abduction of the contralateral
eye when looking away from the lesion. Usually there is
nystagmus in the abducting eye (ataxic nystagmus); the
mechanism is uncertain, but may represent a form of gaze-
paretic nystagmus. The impaired adduction of the ipsilateral
eye is not due to a medial rectus palsy because convergence
and monocular movements are reasonably normal.
Unilateral lesions may be caused by demyelination, 629 Slow and incomplete adduction of the right eye (with horizontal
infarction (paramedian pontine), and tumor; but bilateral nystagmus in the abducting left eye) on lateral gaze to the left due to a
lesions are almost pathognomonic of demyelination (MS). right internuclear ophthalmoplegia in a patient with multiple sclerosis.
504 Cranial Neuropathies

One-and-a-half syndrome Pseudo VIth nerve palsy


• Paralysis of conjugate deviation of the eyes on horizontal • Hysterical convergence spasm: the pupils are constricted,
gaze to one side (the side of the lesion) and an due to voluntary convergence. When the patient is asked
internuclear ophthalmoplegia on horizontal gaze to the to look horizontally to one side, not only do the eyes still
other side (away from the lesion); i.e. the eye ipsilateral converge, but the pupils remain constricted. Patients
to the lesion does not abduct or adduct and the with VIth nerve palsies are not trying to converge the
contralateral eye can only abduct, often with associated eyes, and therefore the pupils are not constricted.
ataxic nystagmus. • Duane’s syndrome (630, 631): a congenital anomaly
• Due to a unilateral lesion (usually infarction, demyelina- due to fibrous replacement of the lateral rectus muscle
tion, or tumor) of the MLF and pontine centre for causing impaired action of the lateral rectus muscle, and
horizontal gaze in the low pontine tegmentum. thus impaired abduction of the eye. On attempted
adduction of the eye, the eyeball retracts into the orbit,
Neuromuscular junction disorders and consequently the palpebral fissure narrows.
Myasthenia gravis: usually but not always bilateral.
Clinical points
Muscle disorders The most likely differential diagnoses can be deduced by
• Ocular myopathies: usually bilateral. accurately localizing the site of the neurologic lesion (from
• Mitochondrial cytopathies (Kearns–Sayre syndrome). the neurologic examination) and the tempo of onset (from
• Ocular muscular dystrophies. the history).
• Facioscapulohumeral dystrophy. Important physical signs to seek are:
• Myotonic dystrophy. • Anosmia.
• Orbital myositis: unilateral or bilateral. • Visual failure (uniocular or binocular).
• Orbital tumor: usually unilateral. • Visual field defects.
• Orbital trauma. • Papilledema.
• Dysthyroid eye disease: usually but not always bilateral. • Optic atrophy.
• GCA (ischemia of the orbital muscles or their nerve • Proptosis.
supply). • Pain around the eye/face/head.
• Vth cranial (trigeminal) nerve involvement.
Ptosis • Fatiguability.
• Congenital.
• Senile (usually bilateral). One of the most common scenarios in neuro-
• IIIrd cranial nerve (oculomotor nerve) lesion (see ophthalmologic practice is the acute onset of an isolated
below). complete lesion of one oculomotor nerve, with or without
• Horner’s syndrome (partial ptosis only) (see p.494). pain around the ipsilateral eye, in a middle-aged or elderly
person. If there is no history of trauma, and there are no
Cerebral hemisphere (‘cerebral ptosis’) other neurologic symptoms or signs, and particularly if the
Stroke – often non-dominant and right hemisphere. pupil is spared in a IIIrd nerve lesion, the most likely cause
is ‘ischemic’. Spontaneous recovery usually begins within
Midbrain 2–4 weeks, during which time it is important to check for
Syphilis (tabes dorsalis). and control any diabetes, arteritis, syphilis, and other
vascular risk factors. Recovery is usually complete. However,
Neuromuscular junction if recovery does not ensue, or if at onset there is pain, a
• Myasthenia gravis. history or evidence of cancer, the pupil is dilated, or there
• Lambert–Eaton myasthenic syndrome. are other neurologic signs, then it is important to exclude a
compressive lesion (by CT or MRI scan of brain ±
Muscle nasopharynx ± cerebral angiography) and meningeal
• Mitochondrial cytopathies. infection, inflammation or malignancy (CSF examination).
• Ocular muscular dystrophies.
• Myotonic dystrophy. INVESTIGATIONS, DIAGNOSIS,
• Orbital trauma. TREATMENT AND PROGNOSIS
Depend on the likely site of involvement and cause (622,
Unilateral dilated pupil 623, 632).
• Parasympathetic nervous system lesion (i.e. IIIrd cranial
nerve lesion).
• Trauma to the iris (usually irregular).
• Holmes–Adie syndrome (see p.497).
Trigeminal (Vth Cranial Nerve) Neuropathy 505

630 TRIGEMINAL (VTH CRANIAL NERVE)


NEUROPATHY

DEFINITION
Disorder of the trigeminal (Vth cranial) nerve.

EPIDEMIOLOGY
Quite common.

ANATOMY
630 Attempted lateral gaze to the left by a patient with Duane’s The largest cranial nerve, it arises from the middle of the
syndrome (congenital fibrosis of the left lateral rectus muscle) showing pons and passes forwards and laterally across the
impaired abduction of the left eye due to impaired action of the left subarachnoid space to form a large ganglion which lies over
llateral rectus muscle. the tip of the petrous bone, where the nerve divides into
three.

631 Sensory
Peripheral
First (ophthalmic) division. Innervates the skin of the
ipsilateral scalp (on top of the head), forehead, cornea and
nose via three branches: the lacrimal nerve (lateral eyelid and
brow); the frontal nerve, and its branches the supratrochlear
and suprorbital nerves (forehead to vertex); and the
nasociliary nerve (cornea, upper medial eyelid, upper side
of nose, tip of nose, alar and vestibule, and nasal mucosa).
The first division carries most of the afferent pathway of the
corneal reflex, with perhaps some contribution from the
631 Lateral gaze to the right by the same patient as in 630 with second division for the lower half of the cornea. It then
Duane’s syndrome showing retraction of the left eyeball into the orbit, enters the orbit and passes through the superior orbital
and consequent narrowing of the left palpebral fissure due to fibrosis fissure, lateral wall of the cavernous sinus (below the
(and failure to relax and stretch) of the left laterial rectus muscle. abducens nerve) to the trigeminal sensory ganglion.

Second (maxillary) division. Innervates the skin of the


632 ipsilateral lower half of the cornea and lower eyelid; cheek
and jaw, sparing the skin over the angle of the mandible
(which is innervated by the C2/C3 nerve roots); midlateral
nose and lateral part of the alar, and the mucous membranes
of the cheek and upper lip. It enters the skull through the
foramen rotundum and then passes in the extreme lower
lateral wall of the cavernous sinus to the trigeminal sensory
ganglion.

Third (mandibular) division. Innervates the skin of the


ipsilateral temple, cheek and upper lip, lower oral cavity,
anterior tongue, and gums. It then enters the skull through
the foramen ovale (with the motor fibers) and passes inferior
to the cavernous sinus to the trigeminal sensory ganglion.

Central
There are two central sensory pathways within the brainstem.

632 T1W coronal MRI showing a large mass (chondrosarcoma) arising


on the right side of the skull base at the petrous apex, projecting
posteriorly into the posterior fossa and anteriorly into the temporal
fossa.The calcification (which is frequent) is better visualized on CT scan.
The patient had presented with a painful VIth nerve palsy, which is also
said to be typical, and other cranial nerve palsies had developed later.
506 Cranial Neuropathies

Touch and afferent arc of the corneal reflex: it enters the mid- Petrous temporal bone apex (Gradenigo’s syndrome)
pons, crosses the midline and ascends through the midbrain • Inflammation: osteitis.
to the contralateral thalamus and then to the face area of the • Infection (trigeminal sensory ganglion): herpes zoster
sensory cortex in the inferior anterior parietal lobe. (usually first sensory division) (633, 634).
• Tumor.
Pain and temperature: it enters the mid-pons but does not
enter the trigeminal nucleus. It descends in the lateral part of Cavernous sinus/superior orbital fissure
the brainstem, parallel to the descending nucleus of the Vth Vascular
nerve, and adjacent to the ascending pain pathway from the • Aneurysm of carotid siphon or ophthalmic artery.
contralateral arm, leg and trunk. It then enters the descending • Carotico-cavernous fistula.
nucleus of the Vth nerve in the lower medulla and upper • Cavernous sinus thrombosis.
cervical cord (C1–2) and crosses to the opposite side.
The crossed fibers in the contralateral lower medulla and Inflammation
upper cervical spinal cord become the secondary ascending • Meningitis (acute and chronic).
tract of the trigeminal nerve (the quintothalamic tract), lying • Sarcoidosis.
adjacent to the medial lemniscus. The fibers then ascend in the • Tolosa–Hunt syndrome.
medial part of the brainstem to the midbrain, where they join
the touch pathways from the same side of the body and face, Tumor
and proceed to the contralateral thalamus and then to the face • Schwannoma (635).
area of the sensory cortex in the inferior anterior parietal lobe. • Pituitary adenoma/apoplexy.
• Meningioma sphenoid wing.
Motor • Nasopharyngeal carcinoma.
The nucleus is in the pons. Efferent fibers leave the base of • Metastases.
the skull, in the mandibular branch of the trigeminal nerve, • Malignant meningitis.
through the foramen ovale. A small branch, the nerve to the
medial pterygoid, supplies the medial pterygoid, tensor Orbit
tympani and tensor veli palatini. The main mandibular nerve • Inflammation: cellulitis.
divides into anterior and posterior trunks. The anterior • Tumor.
trunk conveys the bulk of the motor root and supplies the
muscles of mastication: temporalis, masseter and lateral Mandible
pterygoid. The posterior trunk is mainly sensory (see above) • Inflammation.
and divides into the auriculotemporal nerve, lingual nerve, • Tumor: often metastatic.
and inferior alveolar nerve.
Other
ETIOLOGY • Mixed connective tissue disease.
Pons • Systemic sclerosis.
• Stroke. • Polio (motor loss only).
• MS. • Guillain–Barré syndrome and other peripheral
• Tumor. neuropathies (motor ± sensory).
• Herpes zoster (usually first sensory division).
Medulla • Skull trauma.
• Syringobulbia. • Trichloroethylene (organic solvent) toxicity: bifacial
• Tumor. numbness.
• Organic mercury poisoning.
Cerebello-pontine angle • Isolated trigeminal sensory neuropathy.
• Acoustic neuroma.
• Meningioma. CLINICAL FEATURES
• Trigeminal neuroma. Unilateral lesion
• Glossopharyngeal neuroma. Motor loss
• Epidermoid/dermoid tumor. • Difficult to detect.
• Chordoma. • Wasting of the ipsilateral temporalis and masseter muscles
• Nasopharyngeal carcinoma. may be evident (636).
• Metastases. • The open jaw deviates to the side of the lesion, due to
• Basilar artery ectasia or aneurysm. pterygoid muscle weakness.
• Arteriovenous malformation.
Sensory loss
Base of the skull Sensory disturbance in any or all of the three sensory
• Infective meningitis. divisions, ipsilateral or contralateral to the lesion can occur,
• Malignant meningitis. depending on the location of the lesion (see below).
• Sarcoidosis.
• Meningovascular syphilis.
• Chordoma.
• Nasopharyngeal carcinoma.
• Metastases.
Trigeminal (Vth Cranial Nerve) Neuropathy 507

Lesion in the brainstem Cerebello-pontine angle and base of skull


• Sensory disturbance in all three sensory divisions can • Ipsilateral facial sensory disturbance (pain [and tem-
occur, with or without motor loss: perature] and touch [and corneal reflex] loss) in all three
– A lesion in the pons can result in ipsilateral or contra- sensory divisions and motor loss occurs.
lateral facial pain, temperature, touch and corneal reflex • Symptoms are associated other ipsilateral cranial nerve
loss, with or without motor loss. (e.g. VI, VII, VIII, IX), brainstem and cerebellar signs.
– A lesion in the medulla can cause ipsilateral or contra-
lateral facial pain and temperature loss only. Petrous temporal bone apex (Gradenigo’s syndrome)
– Usually symptoms are associated with other brainstem (e.g. Ipsilateral sensory disturbance in all three sensory divisions
lower cranial nerve and long tract) and cerebellar signs. and motor loss occurs, with associated ipsilateral abducens
(VIth cranial) nerve palsy and/or ear pain.

633 634

633, 634 Skin rash in the distribution of the mandibular branch of the
left trigeminal nerve due to herpes zoster infection.The patient also has
left facial weakness (Ramsay Hunt syndrome) due to involvement of the
left facial (VIIth cranial) nerve. (Courtesy of Dr AM Chancellor,
Neurologist,Tauranga, New Zealand.)

635 636

635 MRI examination of the brain, axial plane,T2W image, 636 Wasting of the left temporalis muscle and weakness
showing a large area of increased signal intensity, due to an of the left side of the face due to infiltration of the left
intracavernous Schwannoma of the ophthalmic division of trigeminal and facial nerves by metastatic breast carcinoma.
the right trigeminal nerve, extending from the right (Courtesy of Dr AM Chancellor, Neurologist,Tauranga,
cavernous sinus into the retro-orbital space (causing New Zealand.)
proptosis), and compressing the medial right temporal lobe.
508 Cranial Neuropathies

Cavernous sinus/superior orbital fissure lesion • Known causes need to be excluded, such as a skull base
• Sensory disturbance occurs in the first division, and tumor, mixed connective tissue disease, systemic sclerosis,
sometimes the second division if the lesion is posterior and other connective tissue diseases, but sometimes no
and inferior (i.e. not the superior orbital fissure); the cause can be found.
third division is spared. • Recovery sometimes occurs after weeks or months.
• Symptoms are associated with ocular motor (IIIrd, IVth,
and VIth cranial) nerve palsies, optic nerve or chiasm DIFFERENTIAL DIAGNOSIS
compression, and sometimes pain above and within the Pain in and around the eye
orbit. • Glaucoma.
• Proptosis, eyelid and conjunctival edema, episcleral • Iritis.
vasodilatation, and papilledema may be present if there • Optic neuritis.
is venous obstruction (e.g. cavernous sinus thrombosis) • Distal carotid or posterior communicating artery
or a carotico-cavernous fistula (also pulsating exophthal- aneurysm.
mos and orbital bruit). • Dissection of the internal carotid artery.
• The Tolosa–Hunt syndrome is the subacute onset of • Diabetic IIIrd nerve palsy.
severe unilateral orbital pain which may be accompanied • Post-herpetic neuralgia.
by sensory disturbance in first and sometimes the second • Migrainous neuralgia (cluster headache).
division of the trigeminal nerve, and ocular motor (IIIrd, • Ophthalmoplegic migraine.
IVth, and VIth cranial) nerve palsies. The ESR may be • Nasopharyngeal tumors.
raised. It is very rare and the cause is sometimes a chronic • Lesions of the petrous apex, cavernous sinus, superior
inflammation behind and/or within the orbit. It can only orbital fissure, and orbit (see above).
be diagnosed after excluding the other painful
inflammatory and neoplastic causes in the superior orbital Pain in the face
fissure listed above. • As for pain in the eye (see above).
• Trigeminal neuralgia (see p.509).
Orbital lesion • Atypical facial pain.
Sensory disturbance in the first division only. • Cervical spondylosis.
• Cerebello-pontine angle tumor.
Foramen ovale or mandibular lesion • Trigeminal neurofibroma.
Sensory disturbance in the third division only; sometimes • Isolated trigeminal sensory neuropathy.
only unilateral numbness of the chin. • Gradenigo’s syndrome.
N.B. Sometimes the sensory loss may be a very restricted • Nasopharyngeal tumor.
area within the territory of a sensory division of the • Post-herpetic neuralgia.
trigeminal nerve if only a few fibers of a division are affected, • Internal carotid artery dissection.
as in the case of a skull base tumor, for example. • Angina (usually in neck and jaw, rather than face).
• Tabetic lightning-like pains.
Bilateral lesion
Sensory Weak jaw muscles
• Bilateral facial sensory loss occurs, which may not be • Motor neuron disease.
complete (e.g. it may be in an ‘onion skin’ distribution • Myasthenia gravis.
as may occur with syringobulbia [see p.541]). • Myopathy.
• There may be a history of exposure to trichloroethylene
(an organic solvent in glue, paint stripper, and paint) or INVESTIGATIONS
organic mercury (e.g. methyl mercury discharged into These depend on the clinical syndrome and likely location
water and entering fish, or contaminated grain from and etiology:
fungicides). • Direct examination of the nasopharynx and larynx.
• There may be systemic evidence of a connective tissue • MRI or CT scan of brain, base of skull and orbits (636).
disease such as progressive systemic sclerosis, SLE and • CSF examination.
polymyositis (mixed connective tissue disease). • Full blood count and ESR.
• Fasting blood glucose.
Motor • Autoantibody screen, antiribonuclear protein.
More obvious symptoms, with weakness and wasting of the • Chest x-ray.
temporalis and masseter muscles bilaterally, are usually • Carotid angiography if a carotico-cavernous fistula is
evident. If severe, the jaw hangs open. suspected.

SPECIAL FORMS DIAGNOSIS


Isolated trigeminal sensory neuropathy Clinical.
• Uncommon.
• It may begin as a small numb patch on the face which TREATMENT
spreads to the territory of innervation of the whole Determined by the cause.
division of the nerve and then to the adjacent divisions
and even the whole face. PROGNOSIS
• Severe loss of pain sensation in the face may lead to Depends on the cause.
ulcerative lesions around the nose and of the cornea.
Trigeminal Neuralgia (Tic Douloureux, Paroxysmal Facial Pain) 509

TRIGEMINAL NEURALGIA (TIC CLINICAL FEATURES


DOULOUREUX, PAROXYSMAL History
FACIAL PAIN) Pain
• Site: face or mouth. Commonly starts in the dermatome
of the second or third division of the trigeminal nerve;
DEFINITION only 5% start in the first division.
A degenerative condition of the trigeminal nerve pathways, • Trigger factors:
characterized by severe stabbing pains in the distribution of – Talking.
one or more divisions of the trigeminal nerve. The – Chewing.
condition is known as tic douloureux (a painful spasm) – Swallowing.
because of the typical lightning-like jabs of pain. – Shaving.
– Cleaning the teeth.
EPIDEMIOLOGY – Wind blowing on the face.
• Incidence: 2–8 per 100 000 per year. • Trigger points: areas around the nose, lips or mouth
• Lifetime prevalence: 0.7% (95% CI: 0.4–1.0%) of general which, when touched, evoke a paroxysm. When sites are
population; 4% of MS patients. inside the mouth patients become hesitant about eating,
• Age: usually starts after the age of 40 years, most drinking and brushing their teeth.
commonly in the sixth and seventh decades. • Nature: stabbing/lightning or electric shock-like/pene-
• Gender: F>M (1.6:1). trating jabs of pain or clusters of stabbing pains.
• Duration: brief (seconds) and followed by long pain-free
PATHOLOGY intervals.
Focal demyelination and microneuromas are often present • Episodic pattern: pain may recur many times a day for
at the site of microvascular compression of the trigeminal weeks or months, and then may remit for months or
nerve but these features may also be found in asymptomatic years (as for cluster headache).
subjects.
Complications
ETIOLOGY Patients may become anxious and withdrawn because of the
Idiopathic pain. Oral hygiene may suffer and weight loss may occur as
The most common ‘cause’. patients attempt to avoid triggering the pain: depression,
dehydration, and even suicide can occur in severely afflicted
Vascular compression of the trigeminal nerve root patients.
Branches of the anterior or superior cerebellar arteries
impinge on the root of the trigeminal nerve where it enters PHYSICAL EXAMINATION
the mid-pons. Normal in idiopathic trigeminal neuralgia. If abnormal, such
as ipsilateral trigeminal nerve sensory loss, depressed corneal
Secondary causes reflex, or deafness; or if an aching pain persists between the
• A plaque of demyelination (MS) at the root entry zone characteristic stabs, then an underlying cause for the tic-like
in the pons accounts for 2–3% of all cases of trigeminal pains must be searched for; these include intra-axial lesions
neuralgia and up to 8% of younger patients. such as MS plaques, and extra-axial lesions that compress
• A small tumor arising from the trigeminal nerve itself the trigeminal nerve or root such as acoustic neuroma,
(e.g. neuroma) or compressing the trigeminal nerve (e.g. trigeminal neuroma and other posterior fossa tumors.
cerebello-pontine angle neurofibroma, meningioma, or
angioma). DIFFERENTIAL DIAGNOSIS
• Rarely, arteriovenous malformations, aneurysms, and • Temperomandibular joint disorders.
herpes zoster may be implicated. • Atypical dental pain.
• Phantom tooth pain.
PATHOPHYSIOLOGY • MS.
Uncertain. • Glossopharyngeal neuralgia: extremely rare. Pain is of a
similar nature (i.e. severe, unilateral, lancinating, and
Compression hypothesis episodic) but at a different site: pain arises from the
Vascular compression of the trigeminal root appears to be throat, larynx, pharynx, or pinna of the ear (i.e. in the
an important contributing factor. At posterior fossa distribution of the IXth [glossopharyngeal] nerve).
exploration, many patients are found to have a trigeminal
root that is compressed or even grooved by a blood vessel,
usually the superior cerebellar artery.
It is thought that compression results in partial demye-
lination and axonal damage, rendering the axons hyper-
excitable. Damaged axons that are near each other become
susceptible to chemical coupling. Under these conditions,
normal impulses elicited by light mechanical stimulation can
recruit nearby pain fibers, particularly if they have already
been made hyperexcitable by axonal damage. This results in
synchronous discharge and, therefore, intense pain.
510 Cranial Neuropathies

INVESTIGATIONS Surgical
CT brain scan, or ideally MRI brain scan If the pain persists in spite of best medical management, one
CT may not identify intra-axial demyelination or small extra- of several surgical procedures can be undertaken. These are
axial neurofibromas. Scans should be used particularly if the either decompressive or denervating (i.e. permanently
patient is young, bilateral pain is present (more common in damage the trigeminal nerve). However, no clinical trials
MS), or neurologic signs (e.g. deafness) (637). have established the efficacy of any surgical procedure.

Tomographic magnetic resonance angiography Decompression


MRA allows the simultaneous visualization of blood vessels Posterior fossa exploration and microvascular decompression
and neural tissue against a dark background of CSF. In a (the Jannetta procedure) consists of separating/removing
series of 50 patients with trigeminal neuralgia, neurovascular and decompressing the offending artery or vein from the
contact was demonstrated on the affected side in 51 of 55 trigeminal nerve root. The majority of patients have im-
symptomatic nerves studies and on the contralateral side in mediate relief after microvascular decompression, although
only four of 45 nerves. some later have recurrent pain. The advantage of this
procedure is that facial sensation is preserved. The operation
DIAGNOSIS has risks which include cranial hematomas, CSF leakage,
Clinical. meningitis, ipsilateral facial paresis or hearing loss (1%),
brainstem infarction (0.1%) and death (0.2%). Despite its
TREATMENT risks, microvascular decompression is currently the best first
Most patients can be managed medically. surgical option, especially late in the course of the disease.

Medical
Carbamazepine 200 mg three times daily, is the drug of
choice for idiopathic trigeminal neuralgia, being effective
initially in about three-quarters of patients. It may control
symptoms by suppressing sodium ion currents in the
trigeminal caudal nucleus or in the gasserian ganglion. A
small dose of 50–100 mg nightly is the usual starting dose
(to avoid drowsiness), escalating in weekly increments until
pain relief is achieved (or adverse effects occur). A slow 637
release preparation can also be used. The dose is continued
for 1 month or so, and can then be tapered slowly, reloading
if the pain recurs. Most responders will experience about
6–12 months of respite before recurrence. Up to one-third
of patients cannot tolerate carbamazepine in the doses
required to alleviate the pain, because of adverse effects such
as rash, nausea, drowsiness, and ataxia. Carbamazepine may
also cause hyponatremia, megaloblastic anemia (folate
interaction), aplastic anemia, agranulocytosis and
hypersensitivity reactions.
Alternatives, which may be as, or more, effective include:
• Phenytoin 200–400 mg nocte, or 100 mg three times
daily. Less effective than carbamazepine but can be given
intravenously for acute relapses of pain, in a similar
regimen to that given for status epilepticus.
• Baclofen, a gamma-aminobutyric acid-B-receptor
agonist, 10–20 mg three times daily.
• Clonazepam.
• Pimozide.
• Oxcarbazine.
• Mexiletine.
• Gabapentin.
• Lamotrigine, which is at least as potent as carbamazepine
in inactivating sodium ion currents, with fewer adverse
effects, and is effective in some cases. Lamotrigine is a
potent antiglutamatergic agent which may depress
excitatory transmission in the trigeminal caudal nucleus.
It may cause initial ataxia, diplopia, nausea, vomiting, and
blurred vision in 15–35% of patients but these are often 637 MRI brain scan,T2W image, axial section through the mid-pons of
dose related. An allergic skin rash is seen in 3–17% of a patient with trigeminal neuralgia due to multiple sclerosis.The MRI
patients. shows multiple areas of high signal, consistent with demyelination
(arrows), in the mid-pons bilaterally (and also in the deep white matter
of the temporal lobes) involving the intra-axial component of the
trigeminal nerves bilaterally.
Facial (VIIth Cranial Nerve) Neuropathy 511

Denervation FACIAL (VIITH CRANIAL NERVE)


• Percutaneous stereotactic radiofrequency thermal NEUROPATHY
rhizotomy of the trigeminal (Gasserian) ganglion. This
is a relatively simple procedure in which a probe is
inserted into the foramen ovale under local anesthesia DEFINITION
without the need for craniotomy. General anesthesia is Disorder of the facial (VIIth cranial) nerve.
required for the very painful process of coagulation. This
procedure is effective in most cases and preserves some EPIDEMIOLOGY
sensation but troublesome dysesthesia develops in about • Common.
one-sixth of cases. • Age: any age.
• Percutaneous glycerol (glycerin) rhizotomy. • Gender: M=F.
• Balloon compression of the trigeminal ganglion.
• Intracranial nerve section of the appropriate divisions of ANATOMY
the trigeminal nerve. The facial nucleus and nerve is mainly a motor nerve
supplying all the muscles concerned with facial expression
Percutaneous procedures are less invasive than on one side.
microvascular compression and are associated with low rates
of mortality and morbidity but they all create trigeminal Motor (facial muscle) innervation
nerve lesions, occasionally producing anesthesia dolorosa or Supranuclear (upper motor neuron)
keratitis. • Precentral gyrus of the cerebral motor cortex: the
innervation of the facial muscles begins in the most
PROGNOSIS lateral and inferior aspect of the precentral gyrus, near
Commonly the episodes of pain follow a relapsing and the Sylvian fissure. This region is supplied by a branch of
remitting course, with exacerbations lasting weeks to months the middle cerebral artery.
and spontaneous remissions that become shorter and less • Centrum semiovale. From the motor cortex the axons
frequent over the years. Some patients go into spontaneous pass through the centrum semiovale and converge in the
remission, and if soon after onset, may last for years. internal capsule.
Ten years after microvascular decompression surgery, • Internal capsule. The fibers to the face are located at the
70% of patients are free of pain without medication for tic. genu of the internal capsule (the fibers to the arm are
Another 4% have occasional pain that does not require long posterior to the facial fibers, and the fibers to the leg are
term medication. posterior to the arm fibers).
The frequency of recurrent pain is similar for both micro- • Midbrain cerebral peduncles. From the internal capsule
vascular decompression and percutaneous radiofrequency the motor fibers continue down through the cerebral
rhizotomy. Recurrence is more likely and occurs sooner after peduncles where the facial fibers are most medial, the
milder damage to the trigeminal nerve, produced either arm fibers are lateral to them, and the leg fibers are most
chemically or by compression with a percutaneously lateral.
positioned balloon. • Lower pons. The motor fibers continue down to the
Predictors of recurrence after microvascular decom- lower pons where they innervate the upper part of the
pression: ipsilateral facial nucleus (which controls the upper face
• Female sex. muscles) and also cross to innervate the contralateral
• Symptoms lasting >8 years before surgery. facial nucleus. Because the upper part of the facial
• Venous compression of the trigeminal root entry zone. nucleus, that controls the upper facial muscles, receives
• Lack of immediate cessation of tic. innervation from both cerebral hemispheres, a unilateral
hemisphere lesion impairs only the lower facial muscles.

Nuclear and infranuclear (lower motor neuron)


The motor nucleus of the facial nerve lies ventral and lateral
to the abducens nucleus in the lower pons. From the facial
nucleus, all the nerve fibers that innervate the ipsilateral
facial muscles ascend posteriorly and medially to hook
around the abducens nucleus before travelling forward
through the pons (just lateral to the corticospinal tract), the
subarachnoid space, the internal auditory meatus, the
petrous temporal bone, the parotid gland, and the
subcutaneous facial region to the muscles of facial expression
(except the elevators of the eyelids). Therefore, a lesion in
the facial nucleus or proximal part of the nerve, before it
gives off branches in the parotid gland, impairs all the
ipsilateral muscles of facial expression. The facial nerve also
supplies the small stapedius muscle that damps oscillations
of the tympanic membrane in response to loud sounds.
512 Cranial Neuropathies

Sensory (tongue taste) innervation Associated neurologic symptoms


• The sensory component of the facial nerve is small (the • Ipsilateral hemiparesis, hemisensory loss, hemineglect,
nervus intermedius of Wrisberg); it conveys taste hemianopia suggest a supranuclear (upper motor
sensation from the anterior two-thirds of the tongue and neuron) facial palsy.
probably cutaneous sensation from the anterior wall of • Contralateral hemiparesis, ipsilateral limb ataxia, facial
the external auditory canal. numbness and gaze palsy to the side of the facial
• The taste fibers from the anterior two-thirds of the weakness may be signs of an intra-axial low pontine
tongue first traverse the lingual nerve (a branch of the lesion and indicate a nuclear/infranuclear (lower motor
mandibular nerve) and then diverge to join the chorda neuron) facial neuropathy.
tympani (a branch of the VIIth cranial nerve); thence • Ipsilateral sensorineural deafness indicates a lesion in the
they pass through the pars intermedia and geniculate ipsilateral brainstem, cerebello-pontine angle or petrous
ganglion of the VIIth nerve to the rostral part of the temporal bone because of the close proximity of the
nucleus of the tractus solitarius in the medulla. vestibulocochlear nerve (see p.517).
• Secretomotor fibers innervate the lacrimal gland through • Ipsilateral reduced corneal reflex and ataxia are suggestive
the greater superficial petrosal nerve and the sublingual of a cerebello-pontine angle lesion. The corneal reflex
and submaxillary glands through the chorda tympani. can be assessed when ipsilateral paralysis of eyelid closure
is present by asking the patient if a stimulus is felt, and
ETIOLOGY by observing the normal consensual closure of the
Brainstem opposite eyelid and the upward roll of the eyeball when
• Stroke. the cornea is touched on the paralysed side. The last sign
• Pontine glioma. (‘Bell’s phenomenon’) is also apparent when the patient
• MS. attempts to close the eye forcefully and is a normal event,
• Abscess. albeit usually invisible.
• Ptosis is not found with isolated facial nerve lesions and,
Cerebello-pontine angle if present, may indicate a Horner’s syndrome or an
• Acoustic neuroma. oculomotor nerve lesion.
• Meningioma. • Vesicles in the external ear canal and on the palate are
suggestive of herpes zoster infection.
Geniculate ganglion • Altered taste and hyperacusis suggest a lesion of the facial
Herpes zoster (Ramsay Hunt syndrome) (638–640). nerve in the facial canal (e.g. Bell’s palsy). However,
Bell’s palsy should not be diagnosed if deafness, reduced
Facial canal corneal reflex or ptosis are present.
Bell’s palsy (see p.515). • Otitis, trauma, and steadily worsening facial weakness
suggest a lesion of the facial nerve in the petrous
Petrous temporal bone temporal bone.
• Trauma: fracture. • Parotid mass or trauma suggest a lesion of facial nerve
• Infection: Pseudomonas aeruginosa. branches.
• Tumor: metastatic, invasive meningioma. • Peripheral nervous system signs such as sensory
disturbances in the distal extremities suggest a lower
Facial nerve branches motor neuron facial palsy.
• Parotid gland tumor or infection.
• Trauma: facial lacerations. Pre-existing medical conditions which could cause or
• Leprosy. predispose to facial weakness
Risk factors for ischemic stroke and ischemic neuropathy:
Facial mononeuropathy • Increasing age.
• Ischemic: small vessel disease (e.g. vasculitis). • Hypertension.
• Sarcoidosis. • Cigarette smoking.
• Behçet’s syndrome. • Hypercholesterolemia.
• Sjögren’s syndrome. • Diabetes mellitus.
• Syphilis.
• Lyme disease. Ear and parotid gland infections

CLINICAL HISTORY Head trauma with basal skull fracture


Rate of onset of weakness
• Sudden or rapid onset within hours suggests trauma or Malignancy
a vascular etiology such as stroke or peripheral ischemic
neuropathy if the weakness is unilateral, and PHYSICAL EXAMINATION
Guillain–Barré syndrome if the weakness is bilateral. • Drooping of one side of the face with flattening of the
• Subacute or gradual onset over days or weeks is most forehead creases, widening of the palpebral fissure,
likely idiopathic (Bell’s palsy) or the result of an infection flattening of the nasolabial fold, lowering of the corner
or weakness. of the mouth, and impaired smiling and grinning are
observed at rest.
• The eye may be red and dry as a result of impaired
blinking or decreased lacrimation.
Facial (VIIth Cranial Nerve) Neuropathy 513

• The patient is unable to elevate the eyebrow, wrinkle the • Otoscopy of the external auditory canal may reveal signs of
forehead, and close the eye on the affected side. infections and tumors in the tympanic cavity which could
• Air escapes between the lips on the affected side when be relevant (e.g. vesicles of the external ear suggest herpetic
the cheeks are puffed with air. facial neuropathy [Ramsay Hunt syndrome] [639]).
• Emotional facial movements are also impaired. Examination of the oral cavity may reveal vesicles on the
• Corneal reflex is impaired because the lesion affects the palate in patients with Ramsay Hunt syndrome (640).
efferent part of this reflex arc.
• Taste may be impaired if the facial nerve is lesioned N.B. The distinction between upper and lower motor
proximal to the chorda tympani nerve branch, within the neuron facial weakness made on examination is very reliable
temporal bone, which carries taste sensation from the if made during acute weakness. However, after a partial
anterior two-thirds of the tongue. recovery has occurred, lower motor neuron facial weakness
• Speech is usually slurred (flaccid dysarthria). may occasionally resemble upper motor neuron weakness.
• Sounds may be exaggerated (hyperacusis) if the facial
nerve is lesioned proximal to the stapedius nerve branch SPECIAL FORMS
in the temporal bone, which supplies the ipsilateral Ramsay Hunt syndrome (herpes zoster
stapedius muscle to dampen loud sounds. facial paresis)
• Associated CNS, cranial nerve, or peripheral nervous • Reactivation of the varicella-zoster virus.
system signs may be present such as hemisensory loss on • Otalgia.
the contralateral side or gaze palsy on the same side as • Hearing loss.
the lower motor neuron facial weakness (suggesting a • Vesicles in the external ear canal and palate (639, 640),
low pontine lesion), multiple cranial neuropathies from which varicella-zoster virus can be easily recovered.
(suggesting basilar meningitis or vasculitis) or sensory • Seroconversion is common, with an increase in specific
disturbances in the distal extremities or muscle wasting antibody to VZV.
(which may indicate an underlying neuropathy). • Worse prognosis than Bell’s palsy.
• Treat with oral antiviral agents: aciclovir (acyclovir),
famcyclovir, valaciclovir (valacyclovir).

638 639

640

638, 639 Weakness of the left forehead (and lower facial muscles) on
attempts to raise the eyebrows (638) and herpetic skin rash involving
the external ear (639) due to herpes zoster infection (Ramsay Hunt
syndrome) of the left facial nerve.

640 Vesicles on the right side of the palate due to varicella-zoster virus
infection in a patient with a right lower motor neuron facial palsy due to
varicella-zoster virus infection (Ramsay Hunt syndrome).
514 Cranial Neuropathies

DIFFERENTIAL DIAGNOSIS INVESTIGATIONS


Upper motor neuron facial weakness MRI brain/temporal bone scan
Clinical features • Indicated when a brain or temporal bone lesion is
• Normal appearance of the forehead and width of the suspected.
palpebral fissure. • It is more sensitive than CT brain scan of the posterior
• Emotional lower facial movements, such as smiling, are fossa, which is limited by bone artifacts.
usually intact despite weakness of voluntary lower facial • Reveals gadolinium-induced enhancement of the facial
movements, because facial innervation for voluntary nerve in acute cases of Bell’s palsy.
movements comes from the pyramidal motor cortex
whereas facial innervation for emotional facial expression CT brain scan
comes from extrapyramidal cerebral regions (e.g. CT is superior to MRI in demonstrating basal skull
thalamus). fractures, if suspected.
• Preservation of eyebrow elevation, forehead wrinkling,
and eye closure. Blood serology titres
• Corneal reflex is intact because the lesion is above this Herpes simplex; herpes zoster; Lyme disease.
reflex arc.
• Associated CNS signs may be present: hemiparesis, CSF
hemisensory loss, hemineglect, hemianopia. • Indicated if possible meningitis or vasculitis (e.g. fever,
headache, nuchal rigidity, or systemic signs of vasculitis).
Causes • Inconsistently shows mildly elevated cell counts and
• Stroke: 85% ischemic, 15% hemorrhagic. protein levels in Bell’s palsy.
• Brain tumor: primary or metastatic, hemispheric or
brainstem. Electrodiagnostic studies
• Brain abscess. May be used to prognosticate recovery.

Lower motor neuron facial weakness DIAGNOSIS


Associated with multiple cranial nerve deficits Clinical.
• Infectious basal meningitis: common bacteria (pneumo-
coccus, meningococcus, Haemophilus influenzae), TREATMENT
Mycobacterium tuberculosis, Lyme borreliosis, syphilis, Determined by the cause.
fungi, HIV.
• Carcinomatous basal meningitis. PROGNOSIS
• Basilar skull tumor. Determined by the cause.

Bilateral lower motor neuron facial weakness


Rapid onset:
• Guillain-Barré syndrome (may be HIV associated).
• Sarcoidosis.
• Vasculitis.
• Botulism.
• Basal meningitis.
• Stroke.
• Lyme disease.

Slow onset or longstanding:


• Congenital hypoplasia of the facial nuclei (Möbius
syndrome).
• Myasthenia gravis.
• Muscular dystrophy and other myopathies (e.g.
facioscapulohumeral muscular dystrophy).
Acute Idiopathic Facial Paralysis (Bell’s Palsy) 515

ACUTE IDIOPATHIC FACIAL DIFFERENTIAL DIAGNOSIS


PARALYSIS (BELL’S PALSY) Lower motor neuron facial weakness
• Isolated unilateral lower motor neuron facial weakness,
without other neurologic deficits.
DEFINITION • Direct facial nerve infections: herpes viruses (e.g.
A unilateral, lower motor neuron facial paralysis that is reactivation of the varicella-zoster virus [Ramsay Hunt
probably due to acute viral inflammatory demyelination of syndrome]: vesicles in the ear from which varicella-zoster
the facial nerve causing swelling and secondary nerve virus can be easily recovered; seroconversion often, with
ischemia within the facial canal. increase in specific antibody to VZV); Lyme disease.
• Ischemic facial neuropathy due to small vessel disease
EPIDEMIOLOGY (e.g. vasculitis).
The most common cause of facial paralysis. • Sarcoidosis, Behçet’s syndrome, Sjögren’s syndrome,
• Incidence: 20–30 per 100 000 persons per year. syphilis.
• Age: any age. • Middle ear infections: common organisms, Pseudomonas
• Gender: M=F. aeruginosa.
• Middle ear tumors.
PATHOLOGY • Temporal bone fracture.
• Inflammation of the facial nerve is present within the • Temporal bone tumors: metastatic, invasive meningioma.
subarachnoid space, composed largely of lymphocytes • Parotid gland tumors or infections.
with associated demyelination or axonal destruction or • Facial lacerations.
both.
• Studies of autopsy material have shown latent herpes Upper motor neuron facial weakness
simplex virus (HSV) in the geniculate ganglia of humans. • Stroke.
• Brain tumor.
ETIOLOGY AND PATHOPHYSIOLOGY • Brain abscess.
• Uncertain.
• Probably acute viral inflammatory demyelination within INVESTIGATIONS
the facial canal, causing swelling of the nerve within the Not usually necessary, unless there is doubt about the
facial canal and secondary ischemia. diagnosis.
• Viral infection:
– HSV is the strongest association: reactivation of HSV 1 Blood serology titres
genomes. HSV might be present in the geniculate A light preponderance of elevated titres against HSV,
ganglia where it could cause a facial nerve palsy when the compared with controls but increased titres are the
virus travels down the nerve axon, perhaps infecting the exception rather than the rule.
Schwann cells.
– Mumps.
– Epstein–Barr virus. 641
– Cytomegalovirus.
– Coxsackievirus.
– Influenza.
– HIV.
• Autoimmune reaction.

Risk factors
• Diabetes: fivefold increased risk.
• Pregnancy: threefold increased risk.

CLINICAL FEATURES
• A viral prodrome is present in about 60% of patients.
• Pain behind the ear, evolving over about 48 hours before
reaching a plateau.
• Unilateral facial weakness of lower motor neuron type
(see p.511) (641) follows the pain, and may be
associated with excessive tearing due to weakness of the
orbicularis oculi (which normally holds the lacrimal
puncta against the conjunctiva).
• Ipsilateral facial numbness is a common symptom, but
objective sensory testing is normal.
• Taste may be altered.
• Sensitivity to sound (hyperacusis) may be increased, such
as when using a telephone. 641 Weakness of the left facial muscles in a patient with a left Bell’s
palsy who is attempting to close his eyes gently and grimace. He does
not demonstrate a Bell’s phenomenon (upward deviation of the
eyeballs on attempted eye closure).
516 Cranial Neuropathies

CSF Other treatments


Inconsistently shows mildly elevated cell counts and protein There is no proven place for adjunctive therapies or surgical
levels. decompression of the facial nerve in Bell’s palsy.

MRI brain scan Avoid complications


• Indicated when a brain lesion is suspected (e.g. when Exposure keratitis
more than an isolated lower motor neuron facial palsy is • Occurs if the cornea is not adequately protected.
present). • It can be avoided by using artificial tears, instilling
• MRI scans are more sensitive than CT brain scan of the lubricating paraffin ointment, and taping (rather than
posterior fossa, which is limited by bone artifacts. padding) the eye closed at night.
• Reveals gadolinium-induced enhancement of the facial • Dark glasses should be worn outdoors.
nerve in acute cases of Bell’s palsy. • Ophthalmic advice should be sought if the patient
reports eye discomfort or the eye becomes irritated
PCR techniques despite the above measures.
• These are used to amplify viral genomic sequences from • Botulinum toxin injection into the eyelid levator to
endoneurial fluid collected from the facial nerve or tissue weaken it may be considered if conservative measures fail.
of the posterior auricular muscle innervated by the facial • Tarsorrhaphy is rarely necessary in cooperative patients.
nerve in patients undergoing decompressive surgery.
• In one study, HSV type 1 genomes were amplified from PROGNOSIS
the nerve or muscle tissue of 11 of 14 patients with Bell’s 60–80% of patients recover completely. In these cases,
palsy. However, PCR does not distinguish between viral recovery usually begins within 8 weeks and is complete by
genomes that are present in a latent state and those that 6–12 months. The most favorable prognostic sign is an
are present because of reactivated virus in the course of incomplete rather than complete facial palsy. If weakness is
a productive infection. severe or complete, recovery commencing within 3 weeks
is a favorable sign. The longer the delay in return of
Electrodiagnostic studies movement the poorer the recovery.
Electroneurography may be used to prognosticate recovery, Predictors of incomplete recovery are:
but not to make the diagnosis. • Complete facial weakness.
• Pain other than in or around the ear (i.e. back of head,
DIAGNOSIS cheek, other).
A clinical diagnosis of exclusion: • Systemic hypertension, diabetes or psychiatric illness.
• Lower motor neuron facial weakness (peripheral facial • Older age.
nerve palsy). • Hyperacusis.
• No other neurologic deficits. • Decreased tearing.
• No apparent cause, other than a herpes simplex virus,
after prolonged follow-up (until the condition has Residual deficits include:
resolved or fresh clues to a specific cause appear). • Facial weakness.
• ‘Jaw winking’ and other abnormal facial movements
TREATMENT (synkinesias) caused by aberrant reinnervation of the
Corticosteroids muscles of facial expression by regenerating facial nerve
Controversial, partly because of the good prognosis of the fibers (i.e. a miswiring).
untreated condition and the failure of controlled trials to • ‘Crocodile tears’ (rare): an excessive flow of tears when
prove a beneficial effect on long term outcome. eating, caused by aberrant reinnervation of the lacrimal
Nevertheless, steroids are used empirically by some gland by regenerating facial nerve fibers.
neurologists to:
• Relieve pain that is sometimes an early feature. Recurrent facial palsy occurs in about 10% of patients. If
• Prevent progression of incomplete to complete paralysis. this occurs, alternative causes should be excluded, such as
• Prevent denervation in cases of complete paralysis. diabetes, sarcoidosis, tumors, or infection.
• Shorten the time to recovery.
• Retard development of abnormal synkinesias.

It is likely that any benefit obtained is due to steroids


used early in the course (within 1 week of onset). The usual
regime is prednisolone, 25 mg per day, orally, for 1 week,
with patient review on completion. Corticosteroids should
not be used when contraindications exist, such as diabetes,
hypertension, peptic ulcer disease, osteoporosis, glaucoma,
or pregnancy.

Acyclovir
May be effective if the underlying cause is herpes virus infec-
tion. More data are needed from a large, randomized con-
trolled and blinded trial with at least 12 months’ follow up.
Vestibular-cochlear (VIIIth Cranial Nerve) Neuropathy 517

VESTIBULAR-COCHLEAR (VIIITH Auditory apparatus


CRANIAL NERVE) NEUROPATHY Cochlear
• A fluid-filled spiral tube, which is divided into three
compartments by a roof membrane (of Reissner) and a
DEFINITION basilar membrane.
Disorder of the vestibulo-cochlear (VIIIth cranial) nerve. • Endolymph fills the closed cavity between the
membranes, and perilymph fills the cavities on either
EPIDEMIOLOGY side.
Not uncommon. • At the apex the membrane has a diameter of 0.5 mm and
at the base the membrane diameter is 0.04 mm,
ANATOMY responding to low tones and high tones respectively.
The vestibulo-cochlear nerve carries information from two
highly specialized end organs, the vestibular apparatus and Organ of Corti
the organ of Corti, both of which lie deep in the temporal • Receptor hair cells which lie on supporting tissue arising
bone where they are suspended in perilymph (which is from the basilar membrane in the cochlear spiral.
basically CSF which is in continuity with the subarachnoid • The hair processes are embedded in a thin membrane
space). Endolymph is a highly specialized fluid which has a that bridges across the organ of Corti, known as the
high protein content and fills the semicircular canals of the tectorial membrane.
labyrinth and the scala media of the cochlear. • Sound waves are transmitted by the pump-like effect of
the stapes in the oval window which creates shock waves
Vestibular apparatus that are passed to the round window where the energy
Semicircular canals dissipates. The pressure wave is transmitted through the
• Situated in the temporal bone on each side of the head. perilymph, and leads to vibration of the basilar
• Each side contains three fine tubes/canals orientated in membrane, depending on the frequency of the sound.
three different planes. When the basilar membrane oscillates, the tectorial
• When the head is in the normal upright position facing membrane has a shearing effect across the hair cells,
forwards, the lateral canal is tilted up 30° from the plane stimulating the cochlear nerve fibers.
of the floor; it is not horizontal.
• When the head is reclined backwards 60° so that it is 30° Vestibulo–cochlear nerve
from the plane of the floor, the lateral canal lies vertically The vestibulo–cochlear nerve emerges from the temporal
(i.e. at 90° from the floor). bone to enter the posterior fossa through the internal
• The end of each canal joins the utricle where there is a auditory canal along with the facial nerve. Both nerves pass
swelling called the ampulla. through the cerebello–pontine angle to enter the brainstem
• The ampulla contains the cupola and the hair cells. at the anterolateral pontomedullary junction.
• The hair cells are polarized to respond to movement in
one direction only by the position of the single Brainstem
kinocilium on each cell. Vestibular nucleus
• In the lateral canal, movement towards the utricle Occupies much of the lower lateral pontine tegmentum,
stimulates the cupola, and in the vertical canals with vertical ramifications over the entire brainstem from
movement away from the utricle stimulates the cupola. the midbrain down to the cervical spinal cord. Conse-
• The semicircular canals work as three matched pairs in quently, vestibular symptoms and signs are almost always
the three planes. present in brainstem disease but are of limited intrinsic
• Neural activity generated by the canals in transmitted in localizing value within the brainstem.
the vestibular nerve to the vestibular nucleus on the same
side of the brainstem, and then to the oculomotor nuclei. Cochlear nucleus
Receives fibers from the cochlear nerve as they enter the
Utricle pons, and transmits auditory information to the superior
• Contains an otolith organ which senses tilting of the olives bilaterally (mainly contralaterally), from where the
head. fibers ascend in the lateral lemniscus to the inferior colliculus
• The organ is plate-shaped, tilted up at the anterior end, (midbrain), medial geniculate and temporal cortex (Heschl’s
and covered with hairs polarized toward the bend. gyrus).
• Head tilt leads to variation in pressure of the calcium
carbonate crystals on the hairs because of the effect of ETIOLOGY
gravity. Brainstem (pontomedullary junction)
• Stroke.
Saccule • MS.
• Contains an otolith organ which senses angular • Tumor.
acceleration of the head.
• The organ is shaped like a shield, with a central ridge
facing forwards.
• Forward movement forces the crystals to slide down the
slope on either side in proportion to the speed and angle
of movement.
518 Cranial Neuropathies

Cerebello–pontine angle Tinnitus


• Acoustic neuroma (vestibular schwannoma) (642). • May accompany any form or cause of hearing loss.
• Meningioma. • Rarely an isolated symptom of neurologic disease unless
• Trigeminal neuroma. pulsatile and associated with a bruit that is audible to the
• Glossopharyngeal neuroma. examiner (as well as the patient).
• Epidermoid/dermoid tumor.
• Chordoma. Vertigo
• Nasopharyngeal carcinoma. • Vertigo is uncommon, particularly if there is no deafness.
• Metastases. • Associated nystagmus tends to be fairly transient and no
• Basilar artery ectasia or aneurysm. longer than any associated vertigo when caused by a
• Arteriovenous malformation. labyrinthine or a vestibular nerve lesion.

Base of skull Nystagmus


• Infective meningitis. • Horizontal or rotatory nystagmus.
• Malignant meningitis. • Fast phase beats away from the side of the lesion.
• Sarcoidosis. • Maximal on lateral gaze away from the side of the lesion.
• Meningovascular syphilis. • It tends to be fairly transient and no longer than any
• Chordoma. associated vertigo, whereas nystagmus due to a brainstem
• Nasopharyngeal carcinoma. lesion tends to persist.
• Metastases. • ‘Canal paresis’ on caloric testing: irrigating warm and cold
• Paget’s disease of the skull: obstruction of foramina. water in the external auditory canal on the side of the
vestibular nerve (or labyrinthine) lesion, leads to a shorter
Ototoxins duration of nystagmus when compared with irrigating the
• Aminoglycosides. normal ear. False negative results may occur.
• Frusemide.
Unsteadiness
CLINICAL FEATURES • Veering or falling to one side – the side of a unilateral
Deafness vestibular (or cerebellar) lesion.
• The first symptom is usually progressive, unilateral • Wide-based gait.
hearing loss (sensori-neural deafness), often with • Incoordination of finger-nose and heel-knee-shin testing
ipsilateral tinnitus. with the limbs on the side of the vestibular (or cerebellar)
• Involvement of adjacent neurologic structures in the lesion if the eyes are closed; performance is improved
brainstem, cerebello-pontine angle, or inner ear while the patient is watching.
determines the subsequent clinical course:
– Trigeminal nerve: absent corneal reflex and loss of facial Other symptoms
sensation ipsilaterally. Ipsilateral face pain and trigeminal Ototoxins tend to cause imbalance and oscillopsia rather
neuralgia are rare. than vertigo.
– Facial nerve: ipsilateral facial weakness (usually mild) with
taste seldom affected, or rarely, hemifacial spasm. Associated symptoms
– Imbalance is common but vertigo is unusual. Determined by the location of the lesion (e.g. cerebello-
pontine angle).

642 DIFFERENTIAL DIAGNOSIS


Deafness
• Cerebello-pontine angle lesion (see above).
• Acute or chronic meningitis.
• Malignant meningitis.
• Intrinsic brainstem lesion (unusual).
• Cortical lesion (most unusual).

Tinnitus
Pulsatile
• Dural arteriovenous malformation involving the
transverse sinus.
• Arteriovenous malformation in the neck, or head.
• Stenosis of the distal internal carotid artery extra- or
intracranially.
• Carotico-cavernous fistula.
• Giant intracranial aneurysm.
• Glomus jugulare tumor.
• High cardiac output (e.g. pregnancy, thyrotoxicosis,
anemia).
642 Axial section through the pons and cerebellum at autopsy
showing a vestibular schwannoma (arrows).
Vestibular-cochlear (VIIIth Cranial) Neuropathy 519

Vertigo Associated vertigo


The nature of the vertigo and nystagmus are not as reliable Nystagmus, when associated with vertigo, tends to persist
in localizing the site of the lesion as other associated when due to a brainstem lesion whereas it tends to be fairly
neurologic symptoms and signs. transient and no longer than any associated vertigo when
caused by a labyrinthine or a vestibular nerve lesion.
Peripheral vertigo
The clues to localization are not so much the features of the Brainstem lesion
vertigo but more so other features such as deafness, tinnitus • Nystagmus may be asymptomatic (i.e. present without
and pain or fullness in the ear: vertigo), ataxic (i.e. part of an internuclear ophthalmo-
• Vestibulo-cochlear nerve lesions. plegia), or rotatory with the fast phase beating away from
• Inner ear lesions. the side of the lesion.
• Nystagmus tends to persist.
Central vertigo
Brainstem lesions may cause episodic or prolonged vertigo but Cerebellar lesion
hearing is usually normal; there are usually other brainstem Horizontal gaze-evoked nystagmus on lateral gaze to the
features (e.g. diplopia, dysarthria, pyramidal and cerebellar signs), side of a cerebellar hemisphere lesion. Associated ipsilateral
and if present, nystagmus persists after the vertigo has resolved. limb ataxia, imbalance, and dysarthria are present.

Nystagmus Opsoclonus
• A to-and-fro oscillation of the eyes which may be Rapid, chaotic oscillation of the eyes in all directions of
pendular or phasic (jerk). gaze, with an interval between each saccade.
• The direction of the nystagmus may be horizontal,
vertical or rotatory. Opsoclonus–myoclonus–ataxia syndrome
• It may be present in the primary position of gaze (i.e. A subacute syndrome seen as a rare complication of
looking straight ahead) or in one or more of the other neuroblastoma in childhood, and cancer in adulthood.
eight cardinal positions of gaze.
• It is usually accentuated (or appears) on horizontal INVESTIGATIONS
and/or vertical gaze. Caloric test of vestibular function
• The nystagmus is described in terms of the direction of The most useful test of vestibular function.
gaze in which it is observed, and the direction of the fast
phase of the abnormal movements (e.g. rotatory Rationale
nystagmus on horizontal gaze to the left). The cupola can be deflected by convection currents set up
• It is seldom symptomatic (although the underlying cause in the semicircular canals if they are heated or cooled. The
may be symptomatic in the form of vertigo, for example), lateral canals can be easily stimulated by hot or cold fluid
but a few patients complain of oscillation of the visual traversing the external auditory canal.
field (oscillopsia).
• The nature of the nystagmus (and vertigo, if present) are Procedure and interpretation
not as reliable in localizing the site of the lesion as other • Position the patient’s head so that it is reclined
associated neurologic symptoms and signs. backwards 60°, and therefore lying 30° from the plane
of the floor. In this position the lateral canal lies vertically
Pendular (i.e. at 90° from the floor).
• Congenital nystagmus: rare, horizontal nystagmus equal • Examine the external auditory canal and check the
in amplitude in both eyes and in all directions of gaze eardrum is not perforated.
(horizontal and vertical). Usually vision has been poor • Ask the patient (if conscious) to look at a point straight
since childhood. ahead.
• Albinos and miners who have spent years in dim light • Inject warm water at 44°C (111°F) slowly into one ear,
may also have pendular nystagmus. at a rate of about 5 ml per second for about 45 seconds.
• The warm water heats the apex of the canal setting up a
Phasic convection current so that fluid flows away from the
• Symmetric horizontal nystagmus on lateral gaze to the left warm area towards the utricle. This stimulates the cupola.
and right may occur transiently (i.e. for a few beats) in normal Activation of the semicircular canals moves the eyes away
individuals if the direction of gaze is >30° from the midline, from the stimulated ear to the opposite side.
or if the eyes are convergent. More sustained bilateral • If the patient is conscious, they will experience vertigo
horizontal (± vertical) nystagmus is usually due to drug and will manifest nystagmus as they try to correct the
toxicity (e.g. alcohol, barbiturates, carbamazepine, phenytoin). tonic deviation of the eyes away from the stimulated ear
• Vertical nystagmus is always due to a brainstem lesion; by corrective saccades in the opposite direction.
the cause may be drug toxicity (e.g. anticonvulsants) as • The normal duration of the nystagmus is about
well as structural lesions. 2 minutes.
• Rotatory nystagmus has no localizing value. • The test can be repeated by injecting water at 30°C
• Positional nystagmus (and vertigo) which fatigues in (86°F) slowly into the same ear, and then repeating the
seconds is due to benign paroxysmal positional vertigo. warm and cold water in the other ear. The cold water
• Positional nystagmus which persists for minutes to hours cools the apex of the canal and fluid flows towards the
or longer is likely to be due to a central (brainstem or cool area away from the utricle. The cupola is inhibited.
cerebellar) lesion. The eyes move toward the stimulated ear.
520 Cranial Neuropathies

Canal paresis IXTH CRANIAL NERVE


A lesion of the semicircular canals or vestibulo-cochlear (GLOSSOPHARYNGEAL)
nerve on one side results in an incomplete or defective NEUROPATHY
response to both hot and cold water in the affected ear (e.g.
nystagmus, characterized by tonic eye movement and
corrective saccades, cannot be induced to the side of the DEFINITION
stimulation), and normal responses from the other ear. Disorder of the glossopharyngeal (IXth cranial) nerve.

Other tests EPIDEMIOLOGY


• Audiometry. Rare, particularly in isolation.
• Brainstem auditory evoked potentials (643): prolonged
interpeak latency, but false positives are common. ANATOMY
• Direct examination of the nasopharynx and larynx. Medulla
• MRI or CT brain scan with high resolution views of the Motor nucleus
posterior fossa and internal auditory canal of the • Situated in the upper part of the nucleus ambiguus in the
temporal bone, before and after i.v. contrast. medulla, and supplies stylopharyngeus (which cannot be
• CSF examination. tested clinically).
• Full blood count and ESR. • The nucleus ambiguus is the main motor nucleus of the
• Fasting blood glucose. IXth, Xth and XIth cranial nerves, and it receives bilateral
• Autoantibody screen. supranuclear innervation from the corticobulbar fibers.
• Chest x-ray.
Sensory nuclei
DIAGNOSIS • The long spoon-shaped nucleus of the tractus solitarius
Clinical. receives taste fibers from the posterior third of the
tongue, soft palate, and palatal arches (via the
TREATMENT glossopharyngeal nerve).
Determined by the cause. • The nucleus of the spinal tract of the trigeminal nerve
receives somatic sensation from the mucous membrane
PROGNOSIS of the soft palate and pharynx.
Depends on the cause.
Inferior salivatory nucleus
Gives rise to autonomic parasympathetic nerves.

Base of skull (jugular foramen)


• The glossopharyngeal nerve emerges from the medulla
in line with the vagus (cranial nerve X) and accessory
(cranial nerve XI) nerves, and enters the internal part of
the jugular foramen lying on the medial side of the
sigmoid sinus.
• The foramen angles forwards and laterally under the
petrous bone, which is excavated by the slight ballooning
643 of the sigmoid sinus as the sinus exits through the skull
2 4 6 msec to become the jugular bulb.
V • The glossopharyngeal, vagus and accessory nerves exit
I II III IV from the jugular foramen in front of the jugular bulb
(644).
VI Outside the skull
A1–Cz
VII
• The glossopharyngeal nerve descends between the
jugular vein and internal carotid artery, which ascends to
enter the carotid canal just anterior to the jugular vein as
it emerges from the jugular foramen. Here, the
glossopharyngeal nerve picks up sympathetic fibers from
the cervical sympathetic chain which has ascended into
the area on the carotid artery. Nearby is the hypoglossal
A1–Cz
(XIIth) cranial nerve, which exits the skull through the
anterior condylar canal posteromedial to the jugular
foramen and comes into close contact with the three
other nerves outside the skull.
• The glossopharyngeal nerve then loops forwards and
medially to reach the soft tissues of the oropharynx,
643 Normal brainstem auditory evoked potential (BAEP) following posterior tongue and palate.
repetitive auditory stimulation of the left ear and duplication of waves
I–VII.The sweep duration is 10 msec.
IXth Cranial Nerve (Glossopharyngeal) Neuropathy 521

• The final branches are the pharyngeal, tonsillar, and ETIOLOGY AND PATHOPHYSIOLOGY
lingual branches carrying general sensation and taste Unilateral isolated glossopharyngeal neuropathy
from the named areas. Rare:
• In its course to the pharynx, it gives off the tympanic • Neuroma of the glossopharyngeal nerve.
(Jacobson’s) nerve, which conveys the preganglionic • Meningioma.
secretomotor fibers for the parotid gland to the otic • Neurofibroma.
ganglion via the tympanic plexus and lesser petrosal • Epidermoid tumor (cholesteatoma).
nerve. The otic ganglion lies just below the foramen • Glomus or carotid body tumor (645, 646): arises in
ovale, attached to the mandibular nerve but functionally ectopic chemoectodermal tissue that is normally present
conveying information from the glossopharyngeal nerve. in the carotid body, and which may be found in the inner
Postganglionic parasympathetic fibers supply the parotid ear and jugular foramen, around the glossopharyngeal
gland, and evoke salivation, by way of the auriculo- and vagus nerves. Neoplastic changes in this tissue
temporal nerves. produce highly vascular erosive tumors which may
• An important branch, the carotid nerve, innervates the destroy the petrous temporal bone and present as a
carotid body and carotid sinus conveying, respectively, vascular nodule in the external auditory canal. These
chemoreceptor and stretch reflex information centrally tumors occur in either gender with a peak incidence in
for respiratory and circulatory reflex function. the third and fourth decades. Depending of the exact site
of the tumor, an external jugular foramen syndrome
(cranial nerves IX, X, XI) and/or a cerebello-pontine
angle syndrome (cranial nerves V, VII, VIII) may result.

Optic foramen Superior orbital fissure 644 645

Foramen
rotundum
Petrous
Foramen temporal
ovale bone
VII and VIII
enter the VI V
internal
auditory
canal

IX, X, XI
in jugular
foramen

XII in
anterior
condylar
canal Lateral sinus

644 Diagrammatic view from above of the inside of the right petrous temporal 646
bone of the skull showing the exit foraminae for the glossopharyngeal (IXth)
and other cranial nerves.

645, 646 T1W MRI post contrast (645) and carotid angiogram (646) of a
right-sided glomus jugulare tumor. Note the mass near the jugular bulb (645,
arrows) and the intense vascularity on angiography (646, arrows).
522 Cranial Neuropathies

Unilateral glossopharyngeal neuropathy patient is then asked to compare these gentle stimuli.
plus other neurologic signs Evaluation of taste sensation over the posterior third of the
• Lateral medullary infarct. tongue has no proven value in clinical diagnosis.
• Other medullary pathology (e.g. tumor, demyelination).
• Glomus or carotid body tumor. Associated clinical features
• Meningioma. These are clues to the site of involvement (e.g. intracranial
• Neurofibroma. vs. extracranial).
• Epidermoid tumor (cholesteatoma). • Long tract signs of brainstem involvement or
• Glomus or carotid body tumor. compression indicate an intracranial lesion.
• An isolated ipsilateral Horner’s syndrome (together with a
Bilateral isolated glossopharyngeal neuropathy glossopharyngeal palsy) is suggestive of a lesion outside the
Extremely rare. skull, as the cervical sympathetic ascends into the area of the
jugular foramen but does not pass through the foramen.
Bilateral glossopharyngeal neuropathy plus other • A palpable mass at the angle of the jaw may represent a
neurologic signs meningioma or neurofibroma that has extended out
• Syringobulbia. through the jugular foramen in a dumb-bell fashion, but
• Arnold–Chiari malformation. it must not be confused with the tip of the transverse
• Infection: process of the atlas which is a normal bony hard mass
– Viral: Epstein–Barr virus; infectious mononucleosis. palpable in this area.
– Mycoplasma infection.
– Chronic granulomatous diseases. Glossopharyngeal neuralgia
– Syphilis. • Attacks are rare and comprise severe, excruciating neuralgic
– Tuberculosis. pain (similar to that of trigeminal neuralgia) in the posterior
– Cryptococcal infection. pharynx, tonsillar fossa, base of the tongue and the ear
• Sarcoidosis. when fluid or food is swallowed or when coughing or
• Vasculitis: yawning, due to a ‘trigger point’ in the throat.
– Polyarteritis nodosa. • Occasionally, bradycardia and syncope occur (associated
– Wegener’s granulomatosis. with swallowing or coughing), perhaps because of cross
– SLE. stimulation, via the carotid nerve, of the carotid sinus.
– Rheumatoid arthritis. • May be associated with MS.
– Sjögren’s syndrome. • May respond to carbamazepine.
• Whipple’s disease.
• Tumor: DIFFERENTIAL DIAGNOSIS
– Chordoma. • Vagus (Xth cranial) nerve palsy.
– Epidermoid cyst. • Pharyngeal and palatal mucosal disease.
– Giant cell tumor. • Functional disorder.
– Primary leptomeningeal gliomatosis.
– Carcinomatous meningitis. INVESTIGATIONS
– Lymphoma. Suspected intracranial lesion
• CT or, preferably, MRI scan (645) of brain and base of skull.
Glossopharyngeal neuralgia • CSF examination.
• Aberrant vessels coursing across the glossopharyngeal
nerve. Suspected extracranial lesion
• Neurofibroma or cholesteatoma adjacent to the nerve • Direct examination of the nasopharynx and larynx.
causing nerve compression. • MRI scan of base of skull and sinuses.
• Intra-axial lesions such as demyelination (MS). • Full blood count and ESR.
• Paranasal sinus x-ray.
CLINICAL FEATURES • Chest x-ray.
Glossopharyngeal nerve palsy • Carotid angiography if a glomus tumor is suspected (646).
Depressed pharyngeal and palatal sensation
From a clinical point of view, the glossopharyngeal nerve is DIAGNOSIS
pure sensory because the only muscle supplied by the nerve Clinical.
(stylopharyngeus) cannot be tested clinically. Contrary to
what many students misconceive, the glossopharyngeal nerve TREATMENT
is not motor to the palate. When the gag reflex is tested the Glossopharyngeal palsy
sensory stimulus is relayed via the glossopharyngeal nerve, Determined by the cause.
but the resulting visible palatal movement is mediated by the
vagus nerve. The gag reflex therefore is too gross for accurate Glossopharyngeal neuralgia
clinical diagnosis of a glossopharyngeal lesion. Carbamazepine, baclofen or microsurgical vascular
The only way of accurately testing the integrity of the decompression if no other lesion is demonstrable.
glossopharyngeal nerve is to test pharyngeal sensation by
carefully touching each side of the palate gently with an PROGNOSIS
orange sick and then, with the patient saying ‘Aah’, Depends on the cause.
touching the posterior pharyngeal wall on each side. The
Vagus Nerve (Xth Cranial Nerve) Neuropathy 523

VAGUS NERVE (XTH CRANIAL Base of skull (jugular foramen)


NERVE) NEUROPATHY • Multiple rootlets from the extensive nuclear connections in
the brainstem leave the anterolateral medulla and form a
flat cord which enters the jugular foramen.
DEFINITION • The jugular foramen transmits the glossopharyngeal, vagus
Disorder of the vagus (Xth cranial) nerve. and accessory cranial nerves along with the sigmoid sinus.
It is adjacent to the hypoglossal canal which transmits the
EPIDEMIOLOGY hypoglossal cranial nerve. Anteriorly lies the internal carotid
Uncommon. artery with the associated sympathetic nerves which enter
the skull through the foramen lacerum.
ANATOMY • In the jugular foramen, the vagus nerve gives off a
The most widely distributed cranial nerve. meningeal branch supplying the dura of the posterior fossa.
• In the jugular foramen, and immediately below, lie the
Medulla superior and inferior ganglia, which make connections with
Dorsal nucleus of the vagus the accessory and hypoglossal nerves, and pick up fibers
• Motor: general visceral efferent to the smooth muscle of from the sympathetic plexus on the carotid artery.
the bronchi, heart, esophagus, stomach and intestine.
• Sensory: receives general visceral afferents from the Branches
esophagus and upper bowel, with cell bodies in the • The superior ganglion gives off the auricular branch, which
superior and inferior vagal ganglia. is joined by a branch from the glossopharyngeal, and is distri-
buted to the skin of the external ear with the branch of the
Nucleus ambiguus facial nerve. These fibers all ultimately enter the nucleus of
Motor: efferent to striated muscle of the pharynx and intrinsic the descending tract of the trigeminal nerve in the brainstem.
muscles of the larynx. Bilateral supranuclear innervation. • The inferior ganglion gives off the superior laryngeal nerve,
which divides into two branches. The inferior laryngeal
Nucleus of the tractus solitarius nerve supplies sensation of the mucous membrane of the
Sensory: receives fibers from the taste buds of the epiglottis larynx and conveys proprioceptive information from the
and vallecula. Shared with the glossopharyngeal nerve. neuromuscular spindles and stretch receptors of the larynx.
The external laryngeal nerve supplies the cricothyroid and
Spinal nucleus of the trigeminal nerve contributes to the pharyngeal plexus (important for speech).
Sensory: receives general somatic afferent fibers from the • Below the inferior ganglion, the cranial root of the accessory
pharynx and larynx. nerve merges into the vagus nerve, and forms the
pharyngeal branch which distributes its fibers to supply the
pharynx and larynx.
• The recurrent laryngeal nerve on the right loops under the
647 subclavian artery and on the left under the aortic arch. On
both sides it then ascends on the side of the trachea to reach
the larynx, where it supplies all the muscles of the larynx,
except the cricothyroid, and carries sensory fibers from the
mucous membranes and the stretch receptors of the larynx.

ETIOLOGY AND PATHOPHYSIOLOGY


Unilateral
Lower motor neuron
• Medullary stroke (e.g. lateral medullary infarct).
• Syringobulbia.
• Infective mononeuropathies (e.g. viral).
• Diabetes.
• Arteritis.
• Internal carotid artery dissection.
• Neuroma of the vagus nerve.
• Meningioma.
• Epidermoid/dermoid tumor.
• Glomus tumor.
• Chordoma (647).
• Extension of cerebello-pontine angle mass.
• Nasopharyngeal carcinoma.
• Metastasis.
647 T2W axial MRI through the skull base showing a large, high signal • Malignant lymph nodes in the neck.
lesion in the midline arising from the clivus and invading the post nasal • Trauma:
space (arrows). Chordomas typically affect the clivus or C1/2 causing – Accidental injury to recurrent laryngeal nerve (usually the
bone destruction and areas of calcification (in 50%).The intracranial left as it passes around the aorta) during open heart surgery.
component may compress the pons, invade cranial nerves and the sellar – Gunshot or knife wounds.
region. The main differential diagnosis radiologically is chondrosarcoma. – Skull base fracture.
524 Cranial Neuropathies

Upper motor neuron • Wasting and weakness of the ipsilateral tongue (hypoglos-
Stroke. sal nerve).
• Pain in and around the ear if bone erosion or
Bilateral inflammation occur.
Lower motor neuron • Brainstem signs if lesion is within the skull.
• Syringobulbia. • Ipsilateral Horner’s syndrome if lesion is outside the
• Motor neuron disease. skull.
• Polio.
• Multiple systems atrophy. Unilateral upper motor neuron vagus nerve palsy
• Arnold–Chiari malformation. Contralateral palatal weakness: very transient, if at all, in
• Guillain–Barré syndrome. upper motor neuron lesions because of bilateral
• Subacute/chronic infective or malignant meningitis. supranuclear control of the palate.
• Infective neuropathies.
• Diabetes. Bilateral lower motor neuron vagus nerve palsy
• Arteritis. Bilateral palatal and vocal cord weakness.

Upper motor neuron Bulbar palsy


• Motor neuron disease. • Nasal regurgitation.
• Stroke. • Nasal dysarthric speech.
• MS. • Dysphagia.
• Multiple system atrophy. • Choking and aspiration.
• Central pontine myelinolysis. • Absent gag reflex.
• Hoarse voice.
CLINICAL FEATURES • Weak cough.
Unilateral lower motor neuron vagus nerve palsy • Stridor sometimes, particularly during sleep.
Examine the patient’s palatal movement, voice and cough.
Bilateral upper motor neuron vagus nerve palsy
Motor to the ipsilateral palate and vocal cords Pseudobulbar palsy
Ipsilateral palatal weakness: • Emotional lability (common).
• Regurgitation of fluid and foods into nasopharynx and • Brisk jaw jerk (if bilateral lesions above the mid pons).
nose. • Spastic tongue: slow repetitive movements.
• Nasal speech. • Bilateral corticospinal (pyramidal) tract signs in all four
• Asymmetric palatal movement: unopposed action of the limbs.
contralateral vagus nerve allows the palate to be pulled
across to the intact side (i.e. contralateral to, or away DIFFERENTIAL DIAGNOSIS
from, the vagus lesion) when the patient says ‘Aah’ or Bilateral vagus nerve palsy
when the gag reflex is elicited. Lower motor neuron
• Myasthenia gravis.
Ipsilateral vocal cord weakness (usually a recurrent laryngeal • Myopathy (e.g. myotonic dystrophy, polymyositis).
nerve lesion if isolated vocal cord weakness): • Psychogenic vocal cord adductor palsy.
• Hoarseness.
• Loss of voice volume (dysphonia). INVESTIGATIONS
• Weak cough: unable to cough explosively (‘bovine Suspected intracranial lesion
cough’). • MRI scan of brain and base of skull (647).
• Asymmetric vocal cord movement: the paralysed vocal • CSF examination.
cord lies permanently and limply abducted to the
midline. Suspected extracranial lesion
• Direct examination of the nasopharynx and larynx.
Somatosensory from the ipsilateral ear • MRI scan of base of skull and sinuses.
Reduced sensation in the skin over the cranial part of the • Full blood count and ESR.
auricle and posterior wall and floor of the external auditory • Paranasal sinus x-ray.
meatus (auricular branch). Hence the referred pain from • Chest x-ray.
malignant disease or other irritation of the throat to the ear • Carotid angiography if a glomus tumor suspected.
and auditory canal.
DIAGNOSIS
Associated features Clinical.
Jugular foramen syndrome: lesions in or near the jugular
foramen may cause any combination of: TREATMENT
• Diminished ipsilateral palatal sensation (glossopharyngeal Determined by the cause.
nerve).
• Hoarse voice, nasal speech, dysphagia, asymmetric palatal PROGNOSIS
movement to the contralateral side (vagus nerve). Depends on the cause.
• Wasting and weakness of the ipsilateral sternomastoid
and trapezius (accessory nerve).
Spinal Accessory Nerve (XIth Cranial Nerve) Neuropathy 525

SPINAL ACCESSORY NERVE (XITH Supranuclear innervation


CRANIAL NERVE) NEUROPATHY Probably ipsilateral, because hemispheric lesions (e.g. stroke)
cause weakness of the sternocleidomastoid on the same side
as the lesion (i.e. weakness turning the head to the side of
DEFINITION the hemiparesis); and partial epileptic seizures originating
Disorder of the spinal accessory (XIth cranial) nerve. in the frontal lobe cause the head to turn away from the side
of the lesion and the epileptic focus, indicating that the
EPIDEMIOLOGY ipsilateral sternocleidomastoid is contracting.
Uncommon.
ETIOLOGY AND PATHOPHYSIOLOGY
ANATOMY Intracranial
Cranial part • Arnold–Chiari malformation.
• A detached portion of the vagus nerve. • Syringomyelia.
• It arises predominantly from the lower part of the
nucleus ambiguus and a small component from the Jugular foramen
dorsal efferent nucleus of the vagus. • Neuroma.
• The nerve rootlets emerge from the anterolateral medulla • Meningioma.
in line with the vagus. • Epidermoid/dermoid tumor.
• They are joined by the ascending spinal component, and • Glomus tumor.
then run laterally to enter the jugular foramen. • Chordoma.
• The cranial portion merges with the vagus nerve at the • Extension of cerebello-pontine angle mass.
level of the inferior vagal ganglion and is then distributed • Nasopharyngeal carcinoma.
in the pharyngeal and recurrent laryngeal branches of the • Metastasis.
vagus nerve. • Malignant lymph nodes in the neck.
• These fibers probably supply the muscles of the soft • Trauma:
palate. – Gunshot or knife wounds.
– Skull base fracture.
Spinal part • Infective mononeuropathies (e.g. viral).
• Motor to the sternocleidomastoid and upper part of the • Diabetes.
trapezius. • Arteritis.
• It arises from the anterior horn cells from C1–C5. • Subacute and chronic meningitis.
• The spinal root fibers (C1–C5) emerge from the upper
cervical cord laterally between the anterior and posterior Neck
spinal nerve roots to form a separate nerve trunk, which • Trauma.
ascends into the skull through the foramen magnum. • Malignancy.
• Once inside the skull it exits by way of the jugular
foramen in the same dural sheath as the vagus nerve. CLINICAL FEATURES
• Upon emerging from the jugular foramen, it imme- Unilateral lower motor neuron lesion
diately runs posteriorly to supply the sternocleidomastoid Weakness of the ipsilateral sternocleidomastoid and upper
and upper part of the trapezius. part of the trapezius muscles (648, 649).
• It also receives a major contribution from branches of the
ventral motor roots of C3 and C4 to form a plexus which Acute upper motor neuron lesion
passes through the posterior triangle of the neck (where Weakness of the ipsilateral sternocleidomastoid and upper
it is prone to damage by lymph node biopsy and other part of the contralateral trapezius muscles, so that the
operations) to supply the cervical musculature. patient cannot elevate the shoulder of the paralysed arm nor
turn the head towards the paralysed side.

648 649

648, 649 Wasting and weakness of the trapezius muscle due to section of the left accessory nerve in the process of lymph node on the left
biopsy for tuberculosis. (Courtesy of Dr AM Chancellor, Neurologist,Tauranga, New Zealand.)
526 Cranial Neuropathies

DIFFERENTIAL DIAGNOSIS HYPOGLOSSAL (XIITH CRANIAL


Bilateral weakness of the sternomastoids NERVE) NEUROPATHY
(and neck flexors)
• Motor neuron disease.
• Myasthenia gravis. DEFINITION
• Polymyositis. Disorder of the hypoglossal (XIIth cranial) nerve, which is
• Myotonic dystrophy. the motor nerve to the tongue.
• Acid maltase deficiency.
EPIDEMIOLOGY
INVESTIGATIONS Rare.
Electromyography to confirm denervation of the sterno-
mastoid and trapezius muscles. ANATOMY
• Arises from a nuclear column in the floor of the fourth
Suspected intracranial lesion ventricle, which was derived from the same embryologic
• MRI scan of brain and base of the skull. cell groups as the nuclei of ocular motor cranial nerves
• CSF examination. III, IV and VI.
• Fascicular fibers traverse the full sagittal diameter of the
Suspected extracranial lesion brainstem (medulla), like cranial nerves III and VI, to
• Direct examination of the nasopharynx and larynx. exit from the ventral surface between the pyramid and
• MRI scan of base of skull and sinuses. the olive.
• Full blood count and ESR. • Numerous rootlets combine to form two main fasciculi
• Paranasal sinus x-ray. with their own dural sleeves.
• Chest x-ray. • Leaves the skull through the hypoglossal canal,
• Carotid angiography if a glomus tumor suspected. immediately below the jugular foramen and descends
anteriorly to emerge between the carotid artery and
DIAGNOSIS jugular vein, crosses the inferior vagal ganglion, and
Clinical and EMG. ascends anteriorly on the hyoglossus muscle, distributing
branches to all the muscles of the tongue.
TREATMENT • Receives sympathetic fibers from the superior cervical
Determined by the cause. ganglion, some fibers from the vagus, and from the
motor roots of C1 and C2 via the ansa cervicalis.
PROGNOSIS • Fibers from the hypoglossal nucleus innervate the ipsi-
Depends on the cause. lateral styoglossus, hyoglossus, geniohyoid, and
genioglossus.
• Fibers from the C1 components supply the sternohyoid,
sternothyroid, omohyoid, thyrohyoid, and geniohyoid.
• Supranuclear innervation of the hypoglossal nucleus is
usually bilateral but may be predominantly contralateral.

ETIOLOGY
Unilateral
Lower motor neuron (nuclear/infranuclear)
Medulla:
• Glioma.
• Syringobulbia.
• Arteriovenous malformation.

Meninges:
• Neurofibroma of the hypoglossal nerve (650, 651):
usually female.
• Meningioma.
• Chordoma.

Base of skull:
• Dissecting aneurysm of the internal carotid artery.
• Glomus tumor.
• Nasopharyngeal carcinoma.
• Metastasis.
• Epidermoid/dermoid tumor.

Neck:
• Malignant disease or lymph nodes.
• Resection of malignant disease in the neck.
• Carotid endarterectomy.
Hypoglossal (XIIth Cranial Nerve) Neuropathy 527

• Central venous catheterization. 650


• Infective mononeuropathies (e.g. viral).
• Diabetes.
• Arteritis.

Upper motor neuron (supranuclear)


Stroke (transient).

Bilateral
Lower motor neuron
• Syringobulbia.
• Motor neuron disease.
• Polio.
• Arnold–Chiari malformation.
• Subacute/chronic infective or malignant meningitis.

Upper motor neuron


• Motor neuron disease.
• Stroke.
• MS.
• Multiple system atrophy.
• Central pontine myelinolysis.

CLINICAL FEATURES 651


Unilateral
Lower motor neuron hypoglossal nerve palsy
• Mild dysarthria.
• Wasting, fasciculation and weakness of one side of the
tongue (ipsilateral to the lesion), with deviation of the
tongue to the opposite side (652). During tongue
protrusion, the weak muscle cannot balance the forward
(and contralateral) push of the normal muscle on the
other side, and the normal muscle forces the tongue
forward and to the opposite side (which is towards the
side of the weak muscle and damaged nerve or nucleus).
• Laryngeal shift to one side on swallowing (contralateral
to the lesion) due to failure of the hyoid to elevate on the
paralysed side.

Upper motor neuron hypoglossal nerve palsy


• Mild dysarthria.
• Mild tongue weakness contralateral to the side of the
UMN lesion.
• Usually transient.

652

650, 651 MRI brain scans. Axial plane T1W image (650) and T2W
image (651) showing a schwannoma of the right hypoglossal nerve
(arrows), adjacent to the right ventral medulla and right vertebral
artery. (Courtesy of Dr AM Chancellor, Neurologist,Tauranga, New
Zealand.)

652 Weakness and wasting of the right side of the tongue due to a
right hypoglossal nerve palsy.The unopposed action of the normally
innervated left genioglossus muscle pushes the tongue toward the side
of the lesion.
528 Cranial Neuropathies

Bilateral fasciculations. Tongue fasciculation is best seen with the


Lower motor neuron hypoglossal nerve palsy tongue resting in the floor of the mouth.
• Difficulty manipulating food in the mouth, chewing, and
swallowing. Bulbar palsy
• Difficulty speaking (flaccid dysarthria). • Motor neuron disease.
• Other signs of a bulbar palsy may be present: • Myasthenia gravis.
– Nasal regurgitation. • Myopathy (e.g. myotonic dystrophy, polymyositis).
– Choking and aspiration.
– Absent gag reflex. INVESTIGATIONS
– Hoarse voice. Suspected intracranial lesion
– Weak cough. • MRI scan of brain and base of skull (651, 652).
– Stridor sometimes, particularly during sleep. • CSF examination.

Upper motor neuron hypoglossal nerve palsy Suspected extracranial lesion


• Severe dysarthria. • Direct examination of the nasopharynx and larynx.
• Spasticity of the tongue: slow movements. • MRI scan of base of skull.
• Other signs of a pseudobulbar palsy may be present: • Full blood count and ESR.
– Emotional lability (common). • Paranasal sinus x-ray.
– Brisk jaw jerk (if bilateral lesions above the mid pons). • Chest x-ray.
– Bilateral corticospinal (pyramidal) tract signs in all four
limbs. DIAGNOSIS
Clinical.
DIFFERENTIAL DIAGNOSIS
Bilateral hypoglossal nerve palsy TREATMENT
Normal tongue Determined by the cause.
The tongue normally flickers slightly when it is held out for
more than a few seconds, which can be mistaken for PROGNOSIS
Depends on the cause.

FURTHER READING Sellebjerg F, Nielsen HS, Frederiksen JL, Olesen FACIAL (VIIth CRANIAL NERVE)
J (1999) A randomised, controlled trial of oral NEUROPATHY
high-dose methylprednisolone in acute optic Fiedler CP, Raza SA (1998) Steroids in facial palsy
neuritis. Neurology, 52: 1479–1484. due to herpes zoster. BMJ, 316: 233–234.
OPTIC (IInd CRANIAL NERVE) Ross RT, Mathiesen R (1998) Volitional and emo-
NEUROPATHY LEBER’S HEREDITARY OPTIC tional supranuclear facial weakness. N. Engl. J.
Acheson J (2000) Optic nerve and chiasmal dis- NEUROPATHY Med., 338: 1515.
ease. J. Neurol., 247: 587–596. Riordan-Eva P, Sanders MD, Govan GG, Sweeney
Balcer LJ (1996) Optic nerve disorders. Curr. MG, Da Costa J, Harding AE (1995) The ACUTE IDIOPATHIC FACIAL PARALYSIS
Opin. Ophthalmol., 8: VI:3–8. clinical features of Leber’s hereditary optic Grogan PM, Gronseth GS (2001) Practice para-
Newman NJ (1996) Optic neuropathy. Neurolo- neuropathy defined by the presence of a path- meter steroids, acyclovir, and surgery for Bell’s
gy, 46: 315–322. ogenic mitochondrial DNA mutation. Brain, palsy (an evidence-based review). Neurology,
Shingleton BJ, O’Donoghue MW (2000) Blurred 118: 319–337. 56: 830–836.
vision. N. Engl. J. Med., 343: 556–562. Murakami S, Mizobuchi M, Nakashiro Y, et al.
HORNER’S SYNDROME (1996) Bell’s palsy and herpes simplex virus:
ANTERIOR ISCHEMIC OPTIC Bates AT, Chamberlain S, Champion M, et al. identification of viral DNA in endoneurial fluid
NEUROPATHY (1995) Pholedrine: a substitute for hydrox- and muscle. Ann. Intern. Med., 124(1 pt 1):
Hattenhauer MG, Leavitt JA, Hodge DO, Grill R, yamphetamine as a diagnostic eye drop test in 27–30.
Gray DT (1997) Incidence of non-arteritic an- Horner’s syndrome. J. Neurol. Neurosurg. Psy- Sipe J, Dunn L (2001) Aciclovir for Bell’s palsy
terior ischaemic optic neuropathy. Am. J. Oph- chiatry, 58: 215–217. (idiopathic facial paralysis) (Cochrane review).
thalmol., 123: 103–107. Keane JR (1979) Oculosympathetic paresis. Analy- The Cochrane Library, Issue 3, 2001. Oxford:
Ischaemic Optic Neuropathy Decompression Trial sis of 100 hospitalised patients. Arch. Neurol., update software.
Research Group (1995) Optic nerve decom- 36: 13–16.
pression surgery for non-arteritic anterior is- Kerrison JB, Biousse V, Newman NJ (2000) Iso- VESTIBULAR-COCHLEAR (VIIIth CRANIAL
chaemic optic neuropathy (NAION) is not ef- lated Horner’s syndrome and syringomyelia. NERVE) NEUROPATHY
fective and may be harmful: results of the Is- J. Neurol. Neurosurg. Psychiatry, 69: 131–132. Fife TD, Tusa RJ, Furman JM, et al. (2000) As-
chaemic Optic Neuropathy Decompression sessment: vestibular testing techniques in
HOLMES–ADIE SYNDROME adults and children. Neurology, 55:
Trial. JAMA, 273: 625–632.
Martinelli P (2000) Holmes-Adie syndrome. 1431–1441.
OPTIC NEURITIS Lancet, 356: 1760–1761.
Chan JW (2000) Optic neuritis in multiple scle- IXth CRANIAL NERVE
rosis: an update. The Neurologist, 6: 205–213. IIIRD, IVTH AND VITH CRANIAL NERVE (GLOSSOPHARYNGEAL) NEUROPATHY
Jin Y-P, de Pedro-Cuesta J, Söderström M, Link (OCULAR MOTOR) NEUROPATHIES Minagar A, Sheremata WA (2000) Glossopharyn-
H (1999) Incidence of optic neuritis in Stock- Kline LB, Hoyt WF (2001) The Tolosa–Hunt geal neuralgia and MS. Neurology, 54:
holm, Sweden, 1990–1995. Arch. Neurol., 56: syndrome. J. Neurol. Neurosurg. Psychiatry, 1368–1370.
975–980. 71: 577–582. HYPOGLOSSAL (XIIth CRANIAL NERVE)
Optic Neuritis Study Group (1997) The 5-year
TRIGEMINAL NEURALGIA NEUROPATHY
risk of MS after optic neuritis. Experience of
Barker FG, Jannetta PJ, Bissonette DJ, et al. Keane JR (1996) Twelfth nerve palsy: analysis of
the Optic Neuritis Treatment Trial. Neurolo-
(1996) The long term outcome of microvas- 100 cases. Arch. Neurol., 53: 561–566.
gy, 49: 1404–1413.
cular decompression for trigeminal neuralgia. Keane JR (2000) Combined VIth and XIIth cra-
Purvin V (1998) Optic neuritis. Curr. Opin. Oph-
N. Engl. J. Med., 334: 1077–1083. nial nerve palsies. A clival syndrome. Neurolo-
thalmol., 9; VI:3–9.
gy, 54: 1540–1541.
Chapter Nineteen 529

Spinal Cord Diseases


HEREDITARY SPASTIC PARAPARESIS • Neuronal cell bodies of degenerating fibers are preserved.
• Mild loss of anterior horn cells may occur.
• Dorsal root ganglia, posterior roots and peripheral nerves
DEFINITION are normal.
A group of clinically and genetically diverse disorders • There is no evidence of primary demyelination.
characterized by progressive lower limb spasticity and weakness.
ETIOLOGY AND PATHOPHYSIOLOGY
TERMINOLOGY Heterogeneous pattern of inheritance.
Hereditary spastic paraparesis (HSP) is also known as
familial spastic paraparesis and Strumpell–Lorrain syndrome. Pure uncomplicated forms
The term ‘hereditary spastic paraparesis’ is more precise • Autosomal dominant (70%): seven different loci mapped
because it indicates the genetic basis of this group of to chromosomes 2p (SPG4), 14q (SPG3), 15q (SPG6),
disorders; conditions that occur in families (familial) are not 8q24 (SPG8), 10q (SPG9), 12p (SPG10), 19q (SPG12),
necessarily inherited (genetic). 2q (SPG13). A single gene is known for autosomal
dominant HSP; the gene encodes spastin which is
EPIDEMIOLOGY responsible for SPG4 (spastic paraplegia 4).
• Uncommon. • Autosomal recessive (30%) mapped to a locus on
• Age: age at onset can be difficult to date precisely chromosome 8q12-13 (SPG5).
because incipient disease can go unnoticed unless a • X-linked (rare): mapped to a locus on Xq11.
clinical examination is performed. Most patients
experience symptom onset in the second through fourth Complex forms
decades, but there is a wide range of symptom onset • Autosomal dominant: one locus mapped to chromosome
from infancy through to age 85 years. 10q (SPG9), with associated cataract, distal amyotrophy,
motor axonal neuropathy, short stature, gastro-
CLASSIFICATION esophageal reflux, and vomiting.
Genetic • Autosomal recessive: four loci mapped to chromosomes
• Autosomal dominant (70%). 2q, 3q (SPG14), 13q (ARSACS), 15q (SPG11), and 16q
• Autosomal recessive (30%). (SPG7), all with different phenotypes. Two genes have
• X-linked (rare). been identified, paraplegin and sacsin.
• X-linked: two loci mapped to chromosome Xq28 (SPG1)
Clinical and Xq22 (SPG2). Two genes have been identified, L1
Pure (uncomplicated) HSP cell adhesion molecule and proteolipid protein (PLP).
• Most common.
Surprisingly, the five genes identified to date (spastin,
Complicated HSP paraplegin, sacsin, L1 cell adhesion molecule, and PLP) en-
Associated with: code proteins of different families, making understanding and
• Intellectual handicap. diagnosis of HSP difficult. The discovery of new genes should
• Optic neuropathy. hopefully help to clarify the pathophysiology of these disorders.
• Retinal pigmentary degeneration.
• Ophthalmoplegia. 653
• Deafness.
• Extrapyramidal signs.
• Cerebellar symptoms.
• Amyotrophy in the upper limbs.
• Skin disorders (ichthyosis).

PATHOLOGY
• Axonal degeneration occurs in the terminal/distal
portions of the longest descending and ascending tracts
of the spinal cord (653):
– Corticospinal tract fibers (crossed and uncrossed) from
pyramidal neurons in the motor cortex to the legs.
– Fasciculus gracilis (and cuneatus to a lesser extent) fibers
from the dorsal root ganglia neurons. 653 Axial section through the spinal cord showing degeneration (pallor)
– Spinocerebellar fibers to a lesser extent. in the corticospinal tracts, spinocerebellar tracts, and dorsal columns.
530 Spinal Cord Diseases

CLINICAL FEATURES patients should be evaluated thoroughly for concurrent or


History alternative neurologic disorders.
• Following normal gestation, delivery and early childhood
development, patients develop leg stiffness and gait DIFFERENTIAL DIAGNOSIS
disturbance (stumbling and tripping) due to difficulty Inherited ataxias
dorsiflexing the foot and weakness of hip flexion. • Spinocerebellar ataxias (see p.437): prominent ataxia.
• Paresthesiae below the knees are quite common. • Friedreich’s ataxia.
• Urinary urgency progressing to urinary incontinence is
a frequent, although variable, late manifestation. Inherited leukodystrophies
• A significant minority of patients are asymptomatic and • Adrenoleukodystrophy (ALD)/adrenomyeloneuropathy
unaware of signs. (AMN) (see p.166):
– MRI brain, plasma long-chain fatty acid analysis.
EXAMINATION – Childhood-onset ALD: progressive cognitive impairment
Cranial nerves and peripheral neuropathy accompany corticospinal tract
• Fundi, and extraocular and facial movements are normal signs.
(unless complex HSP: optic neuropathy, retinal pig- – Adolescent-/adult-onset AMN: cognition is normal,
mentary degeneration, ophthalmoplegia). peripheral neuropathy is variable.
• Jaw jerk may be brisk in older subjects but there is no • Metachromatic leukodystrophy (see p.169): autosomal
evidence of frank corticobulbar tract dysfunction. recessive, psychomotor retardation and peripheral neuro-
pathy are typical in children; MRI brain, arylsulfatase
Upper limbs analysis.
• Tone and muscle power are normal (unless complex • Krabbe (globoid cell) leukodystrophy (see p.171):
HSP: amyotrophy). autosomal recessive, peripheral neuropathy may be
• Hoffman’s and Tromner’s signs may be observed. absent in adolescent-/adult-onset Krabbe disease; MRI
• Deep tendon reflexes may be brisk (2–3+). brain, galactocerebrosidase analysis.
• Slight terminal dysmetria is present on finger-to-nose
testing in occasional older affected patients. Structural spinal cord abnormality
• Arnold–Chiari malformation (see p.136): ataxia, MRI of
Lower limbs posterior fossa.
• Pes cavus is generally present and usually prominent in • Cervical or lumbar spondylosis: radiculopathy,
older affected patients. radiographs and MRI spine.
• Wasting of muscles may occur but is mild and limited to • Tethered cord syndrome: MRI conus medullaris.
the anterior and posterior tibial compartment in • Tumor of the spinal cord: pain, loss of sensation,
wheelchair-dependent elderly patients. asymmetric involvement; MRI spinal cord.
• Tone is increased in the hamstrings, quadriceps and • Arteriovenous malformation of the spinal cord: saltatory
ankles. Ankle clonus is present uniformly. progression, spinal sensory level; MRI, spinal arterio-
• Weakness is most notable of the iliopsoas, tibialis anterior graphy.
and, to a lesser extent, hamstring muscles.
• Deep tendon reflexes are pathologically increased (3–4+).
Crossed adductor reflexes and extensor plantar responses
are present uniformly.
• Sensation to sharp stimuli is decreased below the knees 654 655
in occasional patients and vibratory sense is often
diminished mildly in the distal lower limbs.
• Gait: circumduction is present due to difficulty with hip
flexion and ankle dorsiflexion.

Other
Icthyosis of the skin in some patients (654, 655).

Clinical variability
The clinical phenotypic expression of ‘pure’ autosomal
dominant spastic paraparesis is highly variable. The age of
symptom onset, rate of symptom progression, and extent of
disability vary within and between HSP kindreds. Phenotype
variation is related, at least in part, to symptom duration.
However, neurologic deficits are limited to corticospinal and
dorsal column tracts subserving the lower limbs, with the
exception of a trace degree of terminal dysmetria. Additional
deficits such as visual disturbance, marked amyotrophy,
fasciculations, dementia, seizures, or peripheral neuropathy
in patients from pure HSP kindreds should not be
attributed to variant presentations of pure HSP but rather, 654, 655 Ichthyosis of the skin in a patient with complicated hereditary
spastic paraparesis.
Hereditary Spastic Paraparesis 531

• Granuloma (e.g. tuberculous) involving vertebrae and signs, ataxia, amyotrophy and peripheral neuropathy, is the
spinal cord: back pain, subacute course, spine radio- basis for classifying HSP as pure or complex and helps to
graphy and MRI. distinguish HSP from other neurologic disorders.
Otherwise, the diagnosis of HSP is a diagnosis of
Infectious disease exclusion, and should only be made after excluding treatable
• Tertiary syphilis (hypertrophic pachymeningitis): disorders, (such as vitamin B12 deficiency, dopa-responsive
VDRL/RPR, TPHA. dystonia, cervical spondylosis) and disorders with a different
• Tropical spastic paraparesis: subacute course, HTLV-I prognosis (such as familial ALS).
antibodies.
• AIDS: subacute course, HIV antibodies. Definite HSP
• Diagnosed when alternative disorders in the differential
Metabolic disorder diagnosis have been excluded.
• Subacute combined degeneration of the spinal cord: • A family history supports inheritance of an X-linked,
peripheral neuropathy, marked dorsal column involve- autosomal recessive, or autosomal dominant disorder.
ment; serum vitamin B12 concentration. • Progressive gait disturbance is present.
• Mitochondrial encephalomyopathy: short stature, • Spastic paraparesis with grade 4 hyperreflexia and extensor
retinitis pigmentosa, multiple stroke-like episodes; MRI plantar responses: occasionally, evidence of a spastic para-
brain, serum lactate and pyruvate. paresis will be found in asymptomatic subjects and, unless
• Abetalipoproteinemia (Bassen–Kornzweig disease): serial neurologic examinations have been performed, it is not
peripheral neuropathy; lipoprotein electrophoresis. possible to know whether these signs have been present from
• Vitamin E deficiency: peripheral neuropathy; serum birth (representing mild spastic cerebral palsy, for example)
vitamin E concentration. or presage a progressive gait disorder. Such subjects should
be classified as probably affected and examined serially.
Degenerative disease
• Motor neuron disease (familial/sporadic): fasciculations, TREATMENT
amyotrophy; EMG and nerve conduction studies. Specific
• There is no specific treatment to prevent, retard, or
Miscellaneous reverse the progressive disability.
• Dopa-responsive dystonia due to tyrosine hydroxylase • A trial of low dose levodopa-carbidopa is advised for all
gene mutations: diurnal fluctuation, response to low dose patients with childhood-onset progressive gait dis-
levodopa-carbidopa. turbance of uncertain etiology (particularly those with
dystonia) because hereditary progressive dystonia with
INVESTIGATIONS diurnal variation can resemble HSP and is treatable.
• MRI brain and spinal cord.
• Serum vitamin B12, E. Symptomatic
• HTLV-1 antibodies. Similar treatment approaches are used as for chronic
• VDRL/RPR, TPHA. paraplegia from other causes, such as:
• Plasma long-chain fatty acid analysis. • Regular physiotherapy and occupational therapy to
• DNA analysis. minimize contractures and abnormal postures and
maximize physical function and independence.
Electrophysiology • Oral and intrathecal baclofen (or oral dantrolene).
• Nerve conduction studies: normal. Subclinical sensory impair- • Regular local botulinum toxin injections into spastic hip
ment in peripheral nerves or spinal pathways may be detected. adductors or ankle and toe flexors/invertors (e.g. tibialis
• Somatosensory evoked potentials (SSEPs), after stimu- posterior).
lation of peripheral nerves in the lower limbs, shows • Oxybutynin for bladder dysfunction due to detrusor
conduction delay in dorsal column fibers. hyperreflexia.
• Cortical evoked potentials, used to measure neurotrans-
mission in corticospinal tracts, can produce variable results Genetic counselling
but generally show greatly reduced corticospinal tract The availability of genetic markers tightly linked with HSP
conduction velocity and amplitude of the evoked potential loci greatly improves genetic counselling but is applicable
in muscles innervated by lumbar spinal segments. However, only to counselling informative subjects from kindreds with
the cortical evoked potentials of the arms are normal or linkage to these loci who are at risk of transmitting or
show only mildly reduced conduction velocity. These find- inheriting this disorder.
ings indicate that there are decreased numbers of cor- Genetic counselling must consider that, despite complete
ticospinal tract axons reaching the lumbar spinal cord and genetic penetrance, there may be substantial variation, even
that the remaining axons have reduced conduction velocity. within the same family, in age of symptom onset, rate of symp-
• Autonomic nervous system has not been studied system- tom progression, extent of bladder involvement, and functional
atically in HSP. disability. In some HSP kindreds, the condition begins in child-
hood and is relatively non-progressive after about age 10 years.
DIAGNOSIS
The diagnosis of HSP is straightforward when the family CLINICAL COURSE AND PROGNOSIS
history indicates inheritance of progressive spastic paraparesis Gait disturbance progresses insidiously without exacerbations,
as an isolated symptom. In these cases, documenting remissions, or saltatory worsening. The evolution of the
associated deficits, such as retinitis pigmentosa, extrapyramidal disease is similar in patients with early- and late-onset.
532 Spinal Cord Diseases

SPINAL MUSCULAR ATROPHY (SMA) – The survival of motor neuron (SMN) gene.
– The neuronal apoptosis inhibitory protein (NAIP).
– P44, a subunit of the basal transcription TFIIH.
DEFINITION
SMAs are a group of common inherited disorders CLINICAL FEATURES
characterized by degeneration of lower motor neurons, • Slowly progressive symmetric muscle weakness (proximal
leading to progressive paralysis with muscular atrophy. > distal) and atrophy.
• Absent or markedly decreased deep reflexes.
EPIDEMIOLOGY • Fasciculations of the tongue.
• Incidence: childhood SMA: 1 per 6000 newborns. • Tremor of the hands.
• Age: onset varies from birth to adulthood.
• Gender: depends on inheritance (males only if X-linked). SPECIFIC DISORDERS
Type I (infantile: the acute form of WH disease)
PATHOLOGY • Severe generalized muscle weakness and hypotonia at birth
Neuronal loss occurs in the anterior horn cells and beyond in or within the next 6 months (floppy baby syndrome).
the spinal cord and brainstem, with degeneration of lower • Death from respiratory failure usually occurs within the
motor neurons, typical signs of axonal degeneration (reduction first 2 years.
of myelin and dystrophic axons with shrunken and condensed • A more slowly progressive variant is recognized.
axon structures) and neurogenic denervation of muscles (656).
Type II (intermediate)
ETIOLOGY AND PATHOPHYSIOLOGY (see Table 44) Children can sit, but cannot stand or walk unaided and
Inherited survive beyond 2 years.
Childhood SMA
Most pedigrees show an autosomal recessive pattern of Type III (juvenile-onset: Kugelberg–Welander
inheritance. disease)
• Birth prevalence: 1.2 per 100 000.
Severe SMA with arthrogryposis (a heterogeneous group of • Inheritance: usually autosomal recessive, but dominant
neuromuscular disorders) and bone fracture and X-linked forms are described.
X-linked. • Recessive types are linked to chromosome 5 (5q11.2); at
least 5% of families are not linked.
Adult SMA • Linkage in families with autosomal dominant inheritance
• Autosomal dominant and recessive forms are described. has not yet been established.
• Genetically heterogeneous. • Onset is in childhood or adolescence (3–18 years) with
• Some patients demonstrate the survival motor neuron difficult walking due to hip-girdle weakness.
gene (SMNT) deletion. • Proximal muscle weakness of upper and lower limbs is
• X-linked form (Kennedy syndrome/spinal and bulbar present, of varying severity, starting after age 18 months.
muscular atrophy). • Fasciculations are often seen.
• Bulbar musculature is spared.
All forms • Upper motor neuron signs are rarely present.
• Disease locus mapped to chromosome 5q11.2-q13.3, • The SMA is slowly progressive.
suggesting that they are allelic disorders. • Joint contractures often develop during the course of the
• Three inherited or de novo deletions: disease.
• Spinal and chest deformities are common complications.
• Otherwise, type III has a benign prognosis and is
compatible with a normal life-span.

Table 44 Etiology of SMAs


Disorder Inheritance Locus Gene 656
product

SMA I (Werdnig–Hoffman [WH]) AR 5q11-q13 SMN


SMA II (chronic infantile WH) AR
SMA III (Kugelberg–Welander) AR 5q11-q13 SMN
SMA (distal with UL predominance) AD 7p
Kennedy disease XR Xq13 SBMA
Familial ALS AD 21q22 ALS1
(SOD1)

SMA: spinal muscular atrophy; SMN: survival motor neuron protein;


SBMA: androgen receptor; ALS: amyotrophic lateral sclerosis;
SOD: Cu/Zn superoxide dismutase; AR: autosomal recessive;
656 Muscle biopsy, H&E stain, showing denervation atrophy. Several
AD: autosomal dominant; XR: X-linked recessive; UL: upper limb
fascicles consist of atrophic fibers containing dark pyknotic nuclei,
indicating denervation of muscle.
Spinal Muscular Atrophy (SMA) 533

Type IV (adult-onset) Muscle weakness


• Rare. • Muscle weakness of the trunk and limbs (proximal more
• Age of onset: after 15 years; mean age of onset 35 years. than distal; lower limbs weaker than upper).
• Proximal weakness and wasting. • Symmetric.
• Absence of weakness of extra-ocular muscles, diaphragm
X-linked bulbospinal neuropathy and the myocardium, or marked facial weakness.
(Kennedy’s syndrome) (see p.537)
• Onset is in adulthood. N.B. There are rare congenital-onset cases of SMA
• The gene is localized to Xq21-22, where a tandem repeat whose clinical picture also includes external ophthalmo-
of CAG in the androgen receptor disrupts the gene. plegia, facial diplegia, and early respiratory insufficiency.
• Features are similar to proximal SMA (symmetric muscle Wasting is often not conspicuous in SMA type I.
weakness [proximal > distal] and atrophy, and absent or
markedly decreased deep reflexes) but also perioral Other associated features
fasciculations/tremor, which usually also involve the • Fasciculations of tongue and tremor of hands. N.B.
tongue (657), and gynecomastia. Tremor of the hands is frequently observed in SMA types
• Electrophysiology tests show evidence of anterior horn cell II and III.
disease and axonal degeneration at a more peripheral level. • Absence of sensory disturbances.
• Counselling is advocated as for other X-linked • Absence of CNS dysfunction. N.B. Arthrogryposis of the
conditions. All daughters of affected males will be carriers major joints is a rare finding in a severe form of SMA
and sons will be unaffected. Carrier daughters will have type I. In SMA type I some mild limitation of abduction
a 50% risk of having an affected male. Given the CAG of the hips or extension of the knees or elbows is
repeats, this enables gene carriers to be tested and will common.
allow prenatal diagnosis. There is a reasonably good • Absence of involvement of other neurologic systems or
correlation between the number of repeats and the organs, i.e. hearing or vision.
severity of the clinical picture.
Electrophysiology
DIFFERENTIAL DIAGNOSIS • Abnormal spontaneous activity, e.g. fibrillations, positive
• Other non-inherited motor neuron diseases (see p.534). sharp waves and fasciculations by EMG.
• Limb-girdle muscular dystrophy (see p.674). • Increased mean duration and amplitude of motor unit
• Becker muscular dystrophy (X-linked dystrophinopathy): potentials by EMG.
– Family history. • In SMA types II and III, motor nerve conduction
– Clinically similar distribution of weakness but large calves velocities (MNCVs) >70% of lower limit and normal
and lordosis, and no fasciculations. sensory nerve action potentials (SNAPs).
– Serum CK: elevated 10-fold.
– EMG: myopathic. N.B. MNCVs are markedly reduced in SMA type I.
– Muscle biopsy: dystrophic muscle (not neurogenic There is a rare congenital-onset SMA with death within the
changes) and reduced intensity of dystrophin first few weeks of life in which MNCVs are very long and
immunostaining. SNAPs are absent.

INVESTIGATIONS
• Serum CK: <5 times the upper limit of normal.
• Nerve conduction studies and EMG: denervation with
neither sensory involvement nor marked decrease of
motor nerve conduction velocities.
• Muscle biopsy: evidence of skeletal muscle denervation
with groups of atrophic and hypertrophic fibers and type 657
grouping (in chronic cases) (656).
• Molecular DNA analysis: homozygous absence/mutation
of the SMNT gene.

DIAGNOSIS
Diagnosis is confirmed by demonstration of a homozygous
absence of the SMNT gene or, in rare cases, of a small
mutation.

Diagnostic criteria
Age at onset
• In SMA type I (severe form) onset ranges from prenatal
period to age of 6 months.
• In SMA type II (intermediate form) onset is before the
age of 18 months.
• In SMA type III (mild form) onset is usually after the age
of 18 months. 657 Wasting and fasciculations of the tongue in a man with X-linked
recessive bulbar and spinal neuronopathy (Kennedy’s syndrome).
534 Spinal Cord Diseases

Histopathology of muscle MOTOR NEURON DISEASES (MND)


• Groups of atrophic muscle fibers of both types.
• Hypertrophic fibers of type 1.
• Type grouping (chronic cases). DEFINITION
Motor neuron diseases are a heterogeneous group of
N.B. In early-onset cases of SMA type I these inherited and sporadic disorders of upper and lower motor
characteristic features may not be present. Instead there are neurons which lead to progressive weakness of bulbar, limb,
small fibers of both types. In SMA III there may be a thoracic, and abdominal muscles with relative sparing of
concomitant myopathic pattern. oculomotor muscles and sphincter function.
The term ‘amyotrophic lateral sclerosis’ is used to
Laboratory criteria describe this condition in North America whereas the term
Biochemistry: CK usually <5 times the upper limit of ‘motor neuron diseases’ is used on both sides of the
normal. Atlantic.

Molecular genetics EPIDEMIOLOGY


The homozygous absence/mutation of the SMNT gene in • Incidence: 1.5–2.5 per 100 000 person years, relatively
the presence of clinical symptoms is diagnostic. constant throughout the world but higher (5 per
N.B. In cases with absence/mutation of the SMNT gene 100 000 per year) in western Pacific (Guam).
further diagnostic procedures such as EMG and muscle • Prevalence: 4–6/100 000 of total population.
biopsy are no longer needed. The presence of both copies • Age: any age but the age-specific incidence rises steeply
of SMNT argues strongly against the diagnosis. with age, reaching a maximum of 1 in 10 000 per year
between 65–85 years.
TREATMENT – Sporadic MND: median age of onset: 65 years.
• No specific therapy. – Familial MND: mean age of onset: 50 years.
• A multidisciplinary team approach is required involving • Gender: M (2.5 per 100 000) >F (1.8 per 100 000):
the neurologist, GP, physiotherapist, occupational 1.4:1.
therapist, speech/swallowing therapist and social worker.
PATHOLOGY
PROGNOSIS Macroscopic
The prognosis depends on the age of onset and subtype: Brain and spinal cord often appear normal; precentral gyrus
• In SMA types I and II, there is an arrest of development and corticospinal tracts may show atrophy (658–662).
of motor milestones. Children with SMA type I are never
able to sit without support. Children with SMA type II Microscopic (663, 664)
are unable to stand or walk without aid. In SMA type III • Loss of pyramidal cells of the motor cortex:
the ability to walk is achieved. chromatolysis, spheroid formation, neuronophagia.
• In SMA type I the majority of patients have life • Degeneration of the corticospinal tracts.
expectancy <2 years. In SMA type II survival into • Degeneration of lower brainstem motor nuclei (not
adolescence or adulthood is common. In SMA type III oculomotor nuclei) in most cases.
life expectancy is most likely normal. • Cytoplasmic eosinophilic inclusions (Bunina bodies) and
ubiquitin immunoreactive inclusion bodies in
N.B. There will be certain patients who do not clearly fit degenerating cranial motor nuclei and anterior horn cells.
any one category. • Accumulation of neurofilaments (spheroids) in the
proximal axons of spinal motor neurons.
• Muscle shows features of denervation.

Subtypes
• Amyotrophic lateral sclerosis (the classical form of
MND): degeneration of corticospinal tract neurons in
the motor cortex (upper motor neurons) and brainstem
and spinal cord motor neurons (lower motor neurons)
(658–662).
• Progressive bulbar palsy: degeneration of the motor
neurons of cranial nerves IX–XII originating in the
medulla oblongata (the ‘spinal bulb’).
• Progressive muscular atrophy: loss or chromatolysis of
motor neurons of the spinal cord and brainstem.
• Primary lateral sclerosis: complete absence of Betz cells
from layer 5 of the precentral cortex, reduced numbers
of pyramidal cells in layers 3 and 5 of the precentral
cortex with accompanying laminar gliosis and shrinkage
of pyramidal neurons of all sizes.
Motor Neuron Diseases (MND) 535

658 659

660 661

658–661 Axial section through a normal cervical spinal cord (658). 662
Axial sections though an abnormal spinal cord at the cervical (659),
thoracic (660) and lumbar (661) levels showing degeneration in the
anterior and lateral corticospinal tracts, which is more prominent on the
right than the left.

662 Axial section through the spinal cord of another patient with
motor neuron disease showing degeneration in the anterior (arrows)
and lateral (arrowheads) corticospinal tracts.

663 Normal neurons in the anterior horns of the spinal cord (arrows).

664 Chromatolysis of neurons in motor neuron disease.

663 664
536 Spinal Cord Diseases

ETIOLOGY • Bulbar symptoms: 25% present with:


Unknown. – Slurred speech (dysarthria): spastic (pseudobulbar palsy),
flaccid, nasal speech (bulbar palsy), or a combination of
Inherited (5–10% of cases) the two.
Familial ALS (upper and lower motor neuron) – Difficulty swallowing (dysphagia), often with drooling
• Autosomal dominant: of saliva.
– Chromosome 21q22.1: Cu/Zn superoxide dismutase – Alteration in voice quality (dysphonia).
(SOD1) gene mutation (lowers threshold for apoptosis – Emotional lability (common in pseudobulbar palsy).
in motor neurons). • Nocturnal breathlessness due to respiratory failure is a
– Chromosome 22q: neurofilament heavy subunit. presenting symptom in 3%.
• Autosomal recessive (juvenile): chromosome 2q. • Family history: 5–10% have a family history, suggesting
autosomal dominant inheritance of MND; 20% of such
Spinal muscular atrophy (SMA) (lower motor neuron) families have mutations of the Cu, Zn SOD1 gene on
• Chromosome 5q11.2-13.3 I, II, III: homozygous chromosome 21.
deletion of exons 7 and 8.
• X chromosome: X-linked bulbospinal atrophy PHYSICAL EXAMINATION
(Kennedy’s disease): an androgen receptor defect due to Signs may be initially confined to the upper or lower motor
an abnormal trinucleotide repeat (CAG) in the first exon neurons or to one anatomic area, such as the lumbar
of the androgen receptor gene. segment of limbs, bulbar or respiratory musculature.

Sporadic (90–95% of cases) Lower motor neuron (nuclear or infranuclear) signs


• Candidate toxins: sulfur-containing. • Muscle wasting (665, 666).
• Viruses: enteroviruses (e.g. polio), retroviruses (e.g. HIV, • Fasciculations.
HTLV-I). • Muscle weakness.
• Autoimmune factors: antiganglioside antibodies. • Areflexia.

PATHOGENESIS Upper motor neuron (supranuclear) signs


What kills motor neurons in MND or, conversely, maintains • Increased (spastic) tone: may be marked, particularly in
their long life in the absence of disease is not known, but young patients.
genes controlling the cell cycle, growth factors, and • Clonus.
programmed cell death (apoptosis) may be implicated; • Hyperactive deep tendon reflexes, including brisk jaw
ageing has been considered as nothing more than the jerk in patients with bilateral corticospinal tract lesions
accumulation of disease-carrying mutations with age- above the mid-pons.
dependent expression. • Weakness in a pyramidal distribution.
• Extensor plantar responses.
Three hypotheses have been suggested
• Autoimmune attack on motor neurons by IgG Combined upper and lower motor neuron signs
antibodies against L-type calcium channels. Increased tendon reflexes in a corresponding wasted,
• Excessive formation of free radicals due to mutations in fasciculating and weak myotome.
genes such as copper-zinc SOD gene, which is an enzyme
that detoxifies the superoxide anion, a free radical formed Bulbar palsy (dysfunction of the muscles of the
as a by-product of aerobic metabolism. pharynx, larynx, and tongue innervated by cranial
• Excessive activation of inotropic glutamate receptors, nerves IX–XII originating in the medulla oblongata
such as N-methyl-D-aspartate (NMDA) and α-amino-3- [the ‘spinal bulb’])
hydroxy-5-methyl-4-isoxazole propionic acid/kainate • Flaccid dysarthria: labial sounds are usually affected first;
receptors in motor neurons, causing excitotoxicity and palatal weakness gives the speech a nasal quality and dif-
permitting the influx of sodium and calcium into motor ficulty with palatal sounds such as ‘ing’, ‘egg’, and ‘ka’.
neurons, activation of many enzymes, breakdown of • Dysphonia.
proteins and lipids and formation of free radicals. • Dysphagia.
• Weakness, wasting and fasciculation of lower facial
CLINICAL HISTORY muscles and tongue.
• Gradual onset. • Reduced voluntary and reflex movement of the palate,
• Limb weakness: 70% present with progressive asymmetric and also tongue bilaterally.
weakness of the limbs. • Reduced cough and gag reflexes.
– Often begins in the leg as a foot drop, though symptoms • Absent jaw jerk.
may start in the hands, with difficulty of fine movements,
or in the shoulder girdle. Pseudobulbar palsy (dysfunction of the muscles of
– Muscle twitches and cramps may be present, which may the pharynx, larynx, and tongue due to bilateral
be severe. corticobulbar pathway involvement rostral to lower
– Local pain may herald the onset of weakness and wasting cranial nerve nuclei)
by several weeks; pain may be severe in the late stages. • Spastic dysarthria: slow and forced speech with difficulty
– Paresthesia is an occasional symptom but the sensory enunciating labial and lingual sounds.
examination is normal. • Dysphagia: slow and difficult swallowing.
Motor Neuron Disease (MND) 537

• Reduced voluntary movement of the palate with the • Life expectancy near normal and patients remain
patient saying ‘ah’. ambulant until just before death.
• Normal reflex movement of the palate after touching the • Diagnosis: abnormal expansion of the trinucleotide
posterior pharyngeal wall with a wooden probe such as (CAG) repeat in the first exon of the androgen receptor
a tongue depressor. gene on the X chromosome.
• Brisk/exaggerated jaw jerk and facial reflexes.
• Small, contracted tongue which cannot be fully Spinal muscular atrophy (SMA) (see p.532)
protruded. • A group of genetically inherited diseases that usually
• Bilateral spastic quadriparesis. present in infancy or childhood and affect only lower
• Associated emotional lability: inappropriate and excessive motor neurons.
laughter or crying. • Inheritance is usually autosomal recessive.
• Gene locus for SMA types I–III has been mapped to
Ventilatory failure chromosome 5q. Abnormalities have been found in two
• Increased respiratory rate. genes at this locus (SMN and NAIP genes).
• Reduced vital capacity, which is worse on lying flat.

Other
• Mild cognitive impairment, particularly of frontal lobe
function, and less so memory, may be present. 665
• Signs of comorbidities such as cerebrovascular disease
and spinal degenerative disease, may be present in
20–50% of patients without overt dementia.
• Eye movements are normal, besides impaired smooth
pursuit by saccadic intrusions.
• Bladder and bowel function is usually preserved.

VARIANTS
Amyotrophic lateral sclerosis (the classical form of MND)
A unique combination of upper and lower motor neuron
signs.

Progressive bulbar palsy


About 10% of patients present in this manner and most
progress to classical MND.

Progressive muscular atrophy


About 10% of patients show only lower motor neuron signs.

Primary lateral sclerosis


Progressive symmetric spinobulbar spasticity and weakness
of the limbs, more marked in the legs (spastic quadriparesis),
and sometimes the bulbar musculature, causing a 665 A patient with severe limb weakness due to motor neuron
pseudobulbar palsy. Lower motor neuron signs are absent disease in his own environment. Note the severe wasting of the hands.
and higher cognitive function is preserved. (Courtesy of Dr AM Chancellor,Tauranga, New Zealand.)

SPECIAL FORMS
Familial MND 666
• Earlier age at onset: 6–55 years.
• Initial symptoms: arms or legs.
• Variable duration of disease: 3–38 years.

X-linked recessive bulbar and spinal muscular


atrophy (Kennedy’s syndrome) (see p.533)
• Rare.
• X-linked recessive disease affecting men aged 20–50 years.
• Patients present with dysarthria with a wasted fasciculating
tongue, prominent facial fasciculations and postural tremor.
• Proximal muscle wasting, fasciculations and weakness are
also present with hypo- or areflexia.
• Endocrine abnormalities including gynecomastia, reduced
fertility, maturity onset diabetes mellitus can occur.
• Patients have absent sensory action potentials. 666 A patient with ventilatory failure due to motor neuron disease.
• Slowly progressive. Note the severe wasting of the hands. (Courtesy of Dr AM Chancellor,
Tauranga, New Zealand.)
538 Spinal Cord Diseases

Type I SMA (Werdnig–Hoffman disease) dysarthria, pyramidal tract signs, dystonia, dyskinesias,
• Presents at birth or early infancy. supranuclear ophthalmoplegia, white cell/skin fibroblast
• Hypotonia. hexosaminidase A deficiency, electron microscopy of
• Reduced movements. rectal biopsy).
• Proximal muscle wasting and weakness.
• Hyporeflexia. Metabolic and toxic disorder
• Death before 3 years of age. • Thyrotoxicosis.
• Hyperparathyroidism.
Type II SMA • Diabetic ‘amyotrophy’.
• Presents at about 6 months of age. • Lead poisoning.
• More protracted course. • Mercury poisoning.
• Death from respiratory failure before 10 years of age. • Manganese toxicity.

Type III SMA (Kugelberg–Welander disease) Neuromuscular


• Presents at about 10 years of age. • Monomelic amyotrophy.
• Much slower progression than lower motor neuron • Multifocal motor neuropathy with conduction block
forms of MND. (defined electrophysiologically) (see p.593):
• Life expectancy normal. – A disorder of the motor axon rather than the motor
nerve cell body.
DIFFERENTIAL DIAGNOSIS – Patchy, asymmetric, progressive, mainly distal, wasting,
Initially diagnosis is incorrect in about 10% of cases; clues fasciculations and weakness, principally of the upper
are failure to progress, development of atypical features, limbs.
and the results of radiologic and neurophysiologic – Preserved reflexes.
investigations. – EMG: multifocal conduction block.
– High serum titers of anti-GM1 ganglioside antibodies
Brain often detectable.
Pseudobulbar palsy – May respond to immunomodulation with intravenous
• Parasagittal meningioma. human immunoglobulin, plasmapheresis or cyclosphos-
• Pick’s disease. phamide.
• Bilateral corticobulbar vascular lesions (e.g. brainstem • Cramp-fasciculation syndrome due to peripheral nerve
and/or capsular). hyperexcitability:
• Multiple sclerosis. – Normal muscle strength.
– Benign fasciculations.
Bulbar palsy • Myasthenia gravis.
• Myasthenia gravis. • Lambert–Eaton syndrome.
• Posterior fossa tumors or aneurysms involving medulla • Myopathy: inclusion body myositis, scapuloperoneal
or lower cranial nerves. dystrophy.
• Syringobulbia.
• Polyneuritis cranialis: low grade basal meningitis, vasculitis. INVESTIGATIONS
• Full blood count and ESR.
Creutzfeldt–Jakob disease (amyotrophic form) • Serum creatine phosphokinase (may be slightly raised),
calcium, lead levels.
Spinal cord • Thyroid function tests.
• Craniocervical junction abnormality (Chiari malformation). • Serology for syphilis (RPR, TPHA), HIV antibodies,
• Cervical radiculomyelopathy. HTLV-I antibodies.
• Cervical myelopathy and lumbosacral spondylotic • White blood cell/skin fibroblast hexosaminidase A levels.
polyradiculopathy. • Nerve conduction studies:
• Syringomyelia. – Conduction velocity of sensory fibers is normal, and of
• Arteriovenous malformation. motor fibers is normal for nerves of unaffected muscles,
• Vasculitis. and not <70% of average normal value in nerves of
• Infection: poliomyelitis, syphilis, HIV, HTLV-I. affected muscles.
• Intramedullary neoplasia: lymphoma, carcinoma. – EMG: often reveals subclinical chronic partial denerva-
• Paraneoplastic syndrome: encephalomyelitis with anterior tion in clinically unaffected muscles (e.g. increased
horn cell involvement. insertional activity, spontaneous fasciculations and
• Post-radiation myeloplexopathy. fibrillation potentials, positive sharp waves, reduction in
• Hereditary spastic paraparesis with distal amyotrophy number and increase in amplitude and duration of motor
(ethnic origin [Amish, Tunisian, Lebanese], early age of unit action potentials, reduced interference pattern) due
onset, positive family history, slow progression, pes cavus, to anterior horn cell disease.
vibration sense loss, dementia, dystonia). • MRI brain: indicated in patients with bulbar onset to
• Late-onset SMA. exclude a brainstem lesion. MRI may show atrophy of
• Kennedy’s syndrome. the precentral gyrus in primary lateral sclerosis (667), or
• Hexosaminidase A, B deficiency (adult GM2 ganglio- abnormal signal in the corticospinal tracts and cortex
sidosis) (ethnic origin [Ashkenazi Jews], intellectual (thought to be due to iron deposition) (668).
impairment, psychiatric disturbances, ataxia, stutter-
Motor Neuron Diseases (MND) 539

• MRI cervical spinal cord: particularly recommended if a 667


combination of spastic paraparesis and lower motor neuron
signs in the upper limbs are present to exclude a
compressive lesion in the neck (most commonly cervical
spondylosis). May show atrophy and increased signal in the
anterolateral parts of the spinal cord due to gliosis (669).
• Cerebrospinal fluid: normal.
• Neuropsychologic testing: may reveal frontal lobe
dysfunction.
• Ventilatory muscle strength: forced vital capacity.
• Arterial blood gases: abnormalities occur late and
indicate ventilatory failure.
• Magnetic motor cortex stimulation: prolonged central
motor conduction times: more prolonged in primary
lateral sclerosis.
• Positron emission tomography: diminished glucose [18F]
fluorodeoxyglucose uptake in the pericentral motor
cortex in primary lateral sclerosis.

DIAGNOSIS
No specific diagnostic test.

Diagnostic criteria
Presence of
• Lower motor neuron signs (including EMG features in 668
clinically normal muscles).
• Upper motor neuron signs.
• Progression of the disorder.

Diagnostic categories
• Definite MND: upper motor neuron and lower* motor
neuron signs in three regions**.
• Probable MND: upper motor neuron and lower motor
neuron signs in two regions, with upper motor neuron signs
rostral to lower motor neuron signs (thereby excluding
myelopathy, which is characterized by the reverse).
• Possible MND: upper motor neuron and lower motor
neuron signs in one region, or upper motor neuron signs
in two or three regions (e.g. monomelic MND,
progressive bulbar palsy, and primary lateral sclerosis).
• Suspected MND: lower motor neuron signs in two or
three regions (e.g. progressive muscular atrophy, and
other motor syndromes).

*Clinical or electrophysiologic evidence of chronic partial


denervation of anterior horn cell origin.
**Regions are: head, arms, thorax and trunk, legs.
669

667 MRI brain,T1WI, sagittal section near midline, showing atrophy of


the precentral gyrus (arrow) in a patient with primary lateral sclerosis.

668 PDW axial MRI of the brain at the level of the lateral ventricles
(same patient as in 669). Note the abnormal signal in the
periventricular white matter in the region of the major motor fibers
(arrow). (Courtesy of Dr R Gibson, Dept of Neuroradiology,Western
General Hospital, Edinburgh, UK.)

669 Axial T2W MRI of the cervical spinal cord in a patient with motor
neuron disease. Note the increased signal in the left anterolateral
aspect (arrow) due to gliosis of the motor tract.
540 Spinal Cord Diseases

Absence of • Spasticity: physiotherapy, baclofen, dantrolene, focal


• Sensory signs. botulinum injections.
• Sphincter disturbances. • Depression and insomnia: common: antidepressants and
• Visual disturbances. hypnotics can be used when appropriate.
• Autonomic dysfunction. • Referral to a local hospice and/or palliative care as well
• Parkinson’s disease. as terminal and home care should be given when
• Alzheimer-type dementia. appropriate.
• MND ‘mimic syndromes’ (see Differential diagnosis,
above). Specific
• Riluzole:
Supporting features – 2-amino-6-(trifluoromethoxy)benzothiazole which
• Fasciculation in one or more regions. crosses the blood–brain barrier.
• Neurogenic change in EMG studies. – It inhibits glutamate release, inactivates voltage-
• Normal motor and sensory nerve conduction (distal dependent sodium channels, and interferes with
motor latencies may be increased). intracellular events that follow transmitter binding at
• Absence of conduction block. excitatory amino acid receptors.
– May retard the course of MND (improves tracheostomy-
TREATMENT free survival by about 3 months without any effect on
Symptomatic muscle strength and neurological function) but does not
• Multidisciplinary team approach (neurologist, GP, clinical arrest the disease.
nurse, physiotherapist, occupational therapy, speech – Dose: one 50 mg tablet every 12 hours.
therapy and social worker, from which a key worker acts – Well tolerated.
as the coordinator). – Adverse effects include weakness, nausea, dizziness,
• Physiotherapy: muscle tone and strength, mobility. vertigo, sleepiness, shortness of breath and headache.
• Occupational therapy: activities of daily living, prompt – Monitoring of full blood count and liver function tests
provision of aids and appliances. is required at monthly intervals for the first 3 months of
• Speech and swallowing therapy: treatment and then every 3 months.
– Attention to swallowing techniques and function, cough – A year’s treatment costs US$7000–8000 per patient and
and texture of food. is best used early in the evolution of the disease if it is to
– If there is no bulbar weakness, a poor cough does not be used at all.
require any specific treatment unless severe enough to • Vitamin E may retard onset but not progression (cf.
cause retention of sputum (in which case, consider riluzole which may retard progression but not onset).
tracheostomy). • Nerve growth factors (e.g. brain-derived neurotrophic
• Dysphagia: growth factor, insulin-like growth factor): may have a
– Dietary and swallowing advice, and food thickeners. role in the future.
– Percutaneous endoscopic gastrostomy if oral feeding
poses a risk of aspiration. CLINICAL COURSE
– Tracheostomy with a cuffed tube, or a laryngeal diversion • Progressive.
procedure are considered, if aspiration is still a problem, • About 2% develop dementia of frontal lobe type.
but both of these deprive the patient of speech.
• Dyspepsia: common. Diaphragmatic weakness results in PROGNOSIS
esophageal reflux. Treat with antacids, H2-blockers or • Median life expectancy: 2.5–3.5 years; about 2 years for
omeprazole. patients with bulbar onset and 4 years for patients with
• Excess saliva can be treated with anticholinergic medi- onset of limb weakness.
cations (e.g. benzhexol [trihexyphenidyl], hyoscine • 25% survive 5 years from onset of symptoms.
[scopolamine]) or antidepressants ([e.g. amitriptyline, • Favorable prognostic factors:
dothiepin]). A home suction device can be used. – Early age of onset (some may survive 20 years).
Radiotherapy can be given to the parotid gland when – Progressive muscular atrophy.
these measures fail. – Primary lateral sclerosis: mean disease duration: 15 years
• Constipation: due to poor diet, reduced fluid intake and (median: 19 years).
abdominal wall weakness. Encourage fluid intake, and • Poor prognostic factors:
use laxatives (e.g. lactulose, senna [anthraquinone – Bulbar onset.
laxatives], co-danthramer). – Older age.
• Nocturnal respiratory failure (e.g. breathlessness on lying – Female sex.
flat at night, hypoxemia, hypercapnia): • Death is usually from respiratory failure due to
– Nasal intermittent positive pressure ventilation or a diaphragmatic weakness, and may be precipitated by
negative pressure system such as a cuirass can be used at aspiration pneumonia.
night, if good bulbar and limb function are present.
– If assisted ventilation is inappropriate, dyspnea should be
relieved with benzodiazepines and opiates; permanent
tracheostomy and intermittent positive pressure
ventilation are best avoided.
• Muscle cramps: quinine.
Syringomyelia 541

SYRINGOMYELIA • Paracentral parenchymal syrinx: no communication with


the central canal, fourth ventricle, or the subarachnoid
space; almost exclusively caused by spinal cord injury
DEFINITION (trauma, ischemia, viral infection).
A chronic, progressive, degenerative disorder of the spinal
cord, characterized by cavitation (syrinx [from the Greek Anatomy of the syrinx: communicating or loculated
syrinx, ‘tube’ or ‘pipe’]) of the central part of the cervical • Communicating syrinx: free communication between the
spinal cord (syringomyelia), sometimes extending caudally cavity and the subarachnoid space, and therefore free
to the thoracic and lumbar cord and rostrally to the brain exchange between the syrinx and the CSF; hence, the
stem (syringobulbia), which causes progressive neurologic fluid is the same as CSF. These syrinxes are mostly
symptoms, usually brachial amyotrophy and segmental associated with Chiari I and II hindbrain malformations.
dissociated sensory loss, as it expands. • Non-communicating syrinxes: loculated. The syrinx fluid
has a higher protein content than CSF. These are mostly
TERMINOLOGY neoplastic.
Hydromyelia refers to dilatation of the central canal of the
spinal cord. It occurs in association with obstruction at the ETIOLOGY
foramen magnum (Chiari I and II malformations, Paget’s Congenital
disease and other lesions associated with basilar invagin- Hindbrain herniation: the Chiari type I malformation (see
ation). The dilated canal is lined with ependyma and p.136) is present in many cases of syringomyelia that present
communicates with the fourth ventricle via the obex. in adulthood, and the more severe Chiari types II and III
Syringomyelia refers to cavities in the central portion of abnormalities are associated with infantile forms of syringo-
the spinal cord, unconnected with the central canal. The myelia. However, there is a school of thought that the
cavity may be central or eccentric, is lined with glial cells and Chiari I malformation of the cerebellar tonsils is acquired,
does not communicate with the fourth ventricle. It is not congenital, because of the disappearance of the
associated with arachnoid cysts, arachnoiditis, meningiomas abnormal shape and position of the tonsils after simple
and neurofibromas, cord tumors and inflammation, trauma decompressive extra-arachnoidal surgery.
and spondylotic compression. For our purposes both will be
considered together under the title ‘syringomyelia’. Acquired
• Trauma: post-traumatic spinal cord cavitation is a
EPIDEMIOLOGY progressive disorder in which initial spinal cord damage
• Prevalence: 8.4 per 100 000 population. leads to altered CSF hydrodynamics and arachnoiditis,
• Age: symptoms usually begin about 35–45 years of age, resulting in progressive expansion and extension of the
but occasionally in late childhood or adolescence. syrinx. Most cases result from motor vehicle accidents
• Gender: M=F. and affect the lower portion of the cervical segment of
the spinal cord.
ANATOMY AND PHYSIOLOGY OF CENTRAL CANAL • Surgery: intradural spinal surgery, such as resection of a
The central canal of the spinal cord is fluid-filled and lined spinal cord tumor, may lead to syringomyelia.
by ependymal cells that have open gap junctions. Normal • Inflammation: post-inflammatory syringomyelia may
products of metabolism and products of disease enter the result from infections (e.g. tubercular, fungal, parasitic)
central canal from spinal interstitial spaces (the central canal or from chemical meningitis, and is commonly associated
thus acts like a sink draining CSF) and are carried rostrally with arachnoidal scarring.
toward the fourth ventricle, with the aid of vascular
pulsations transmitted to this fluid-filled tube. The central CLASSIFICATION
canal of the spinal cord normally undergoes stenosis or • Type I. Syringomyelia with foramen magnum
occlusion. This is attributed to common viral infections obstruction and central canal dilatation:
causing inflammation of the ependyma. – With type I Chiari malformation.
– With other obstructive lesions of the foramen magnum.
PATHOLOGY • Type II. Syringomyelia without foramen magnum
An abnormal fluid-filled cyst (syrinx) is present, lined by obstruction (idiopathic).
astrocytic gliotic tissue and a few thick-walled vessels, and • Type III. Syringomyelia with other diseases of the spinal
containing clear fluid with a relatively low protein content, cord:
like CSF. – Spinal cord tumors.
It most commonly occurs in the central gray matter of – Traumatic myelopathy.
lower cervical or upper thoracic segments of the spinal cord, – Spinal arachnoiditis and pachymeningitis.
but can involve the entire length of the spinal cord and may • Type IV. Pure hydromyelia with or without hydro-
extend rostrally into the brainstem (syringobulbia) as far as cephalus.
the thalamus.
PATHOPHYSIOLOGY
Anatomy of the syrinx: axial plane of the spinal cord Unknown. There is no consensus on the pathophysiology
• Central: not continuous with the fourth ventricle or of syringomyelia, particularly in Chiari I malformations.
subarachnoid space; the most common type of syrinx. Consequently, there is a great diversity in the surgical
• Central with expansion into the paracentral parenchyma: approaches that are being used.
about 40%.
542 Spinal Cord Diseases

Hydrodynamic theory Motor weakness and wasting


The normal outflow of CSF from the fourth ventricle is Wasting and weakness of upper limb muscles is present,
prevented by a congenital failure of opening of the outlets usually the small muscles of the hand initially, when the
of the fourth ventricle. As a result, a pulse wave of CSF cavity compresses the anterior horn cells sufficient to cause
pressure, generated by systolic pulsations of the choroid them to degenerate. Fasciculations may be seen. Later,
plexuses, is transmitted through the fourth ventricle into the lower limb weakness may occur due to compression of the
central canal of the spinal cord leading to the formation of a corticospinal tracts, causing a spastic paraparesis.
central cavity that dissects along gray matter and fiber tracts.
This theory is supported by the frequent association of Reflex change
syringomyelia with congenital malformations at the Loss of reflexes occurs in the upper limbs due to
craniocervical junction that could interfere with normal flow interruption of the reflex arc in affected segments of the
of CSF, such as the Arnold–Chiari malformation, and the cord. Brisk reflexes and increased muscle tone in the legs
Klippel–Feil syndrome (fusion of one or more cervical occur (leg stiffness is a common symptom) if the lateral
vertebrae), and other congenital abnormalities such as spinal corticospinal tracts become compressed, causing a spastic
bifida and hydrocephalus. paraparesis or quadriparesis, below the level of the syrinx.
It has also been proposed that syringomyelia associated
with Chiari I malformation is produced by the action of the Horner’s syndrome
cerebellar tonsils, which partially occlude the subarachnoid Horner’s syndrome may occur in the presence of
space at the foramen magnum and act as a piston on the compression of the ipsilateral descending sympathetic fibers.
partially enclosed spinal subarachnoid space. This creates
enlarged cervical subarachnoid pressure waves that compress Associated abnormalities
the spinal cord from without, not from within, and Brainstem and cerebellum
propagate syrinx fluid caudally with each heartbeat, which Type 1 Chiari malformation: present in about 90% of
leads to syrinx progression. patients with syringomyelia (about 50% of patients with type
1 Chiari malformation have syringomyelia).
Hydrodynamic mechanism within a destructive
cervical cord lesion Skull base
A cervical cord lesion, occurring in childhood as part of a Platybasia and basilar invagination.
Chiari malformation, results in an enlarging cavity and
diverticulation of the spinal canal via a hydrodynamic Vertebral column
mechanism within a destructive cervical cord lesion. • Klippel–Feil anomaly.
• Fusion of vertebrae.
CLINICAL FEATURES • Thoracic kyphoscoliosis.
Dissociated sensory loss and its consequences
• Loss of pain and temperature sensation occurs in one or SPECIAL FORMS
two dermatomes in the upper limbs bilaterally, often in Syringobulbia
a cape-like distribution across the back and shoulders. The syrinx extends rostrally into the medulla (usually
This is due to expansion of the cavity anteriorly (and over beneath the floor of the fourth ventricle) and rarely beyond.
one or two segments) at the level of the dermatomes, • Pain and temperature sensation is reduced or lost over
thus compressing the decussating (crossing) pain and one or both sides of the face, (often in an ‘onion-skin’
temperature fibers of the lateral spinothalamic tracts. If type distribution) if the syrinx extends into the upper
the syrinx expands laterally it may cause contralateral pain cervical cord (C1,2) and compresses the spinal
and temperature loss below the level of the lesion. trigeminothalamic tract.
Consequently, patients frequently damage their skin from • Touch, proprioception and vibration sensation may be
burns and injure their joints from trauma because the reduced or lost ipsilateral to compression of the
pain goes undetected. decussating fibers of the medial lemniscus in some cases
• Infection, scars, trophic changes and joint deformities in of syringobulbia.
the upper limbs give rise to a soft fleshy appearance of • Wasting and weakness of the tongue, soft palate, pharynx
the fingers and Charcot joints. and vocal cords, causing dysphagia, dysarthria and
• Touch and position sense are retained initially dysphonia, may occur if the syrinx extends into the
(dissociated sensory loss) but later proprioceptive loss medulla and compresses the nuclei of the hypoglossal
also occurs in the limbs due to compression of the and vagus nerves (nucleus ambiguus).
posterior columns. • An internuclear ophthalmoplegia may occur with
• Pain may occur. involvement of the medial longitudinal fasciculus.
Syringomyelia 543

670–672 MRI 670 INVESTIGATIONS


cervical spine,T1W MRI spine
image, midline sagittal MRI is the imaging modality of choice. MR of the cervical
section (670), and spinal cord will usually demonstrate widening of the spinal
axial lower cervical cord, the full extent of the syrinx within the spinal cord and
(671) and upper the extent of expansion in the axial, sagittal and coronal plane,
thoracic (672) and associated lesions such as tonsillar herniation (670–674).
sections, showing The signal from the syrinx is usually of similar signal to CSF
cerebellar tonsillar on all sequences unless it is loculated and contains
ectopia and a proteinaceous material or blood breakdown products.
relatively low lying
fourth ventricle due Other imaging modalities
to a Chiari 1 • Plain cervical spine x-rays may reveal widening of the
malformation (with spinal canal but this is rather non-specific.
previous posterior • Myelography may demonstrate a widened cord; CT may
decompression), and show widening of the spinal cord and canal.
a syrinx within the • CT myelography may demonstrate the syrinx as a low
cervical and upper density area within the widened cord, but it is more
thoracic cord.The difficult to cover an entire syrinx with axial CT than it is
syrinx is at its widest with sagittal MRI.
in the upper thoracic
cord, where it has a EMG
transverse diameter Nerve conduction studies and EMG show reduced
of about 8 mm (0.3 compound muscle action potential amplitude and features of
in).The syrinx chronic partial denervation (fibrillation potentials and reduced
terminates rostrally interference pattern) in the small muscles of the hand among
at the level of the C2 patients with a moderately extensive cervical syrinx.
vertebral body.

671

673 674

672

673, 674 T2W (673) and T1W (674) midline sagittal MRI of the cervical
spine showing a congenital vertebral anomaly at C1, 2 and 3 (arrows), and
also at C5/6 (arrowhead).There is a small syrinx at the level of C4/5.The
appearance is consistent with a Klippel–Feil anomaly.
544 Spinal Cord Diseases

DIFFERENTIAL DIAGNOSIS DIAGNOSIS


• Intramedullary spinal cord tumor (primary or Diagnosis is clinical and radiologic (MRI).
secondary): rapid course, raised CSF protein.
• Extramedullary spinal tumor: tends to present with nerve TREATMENT
root pain and a spastic paraparesis due to extramedullary Conservative
cord compression. CSF protein is elevated. If a small syrinx is present associated with very slow progres-
• Hematomyelia: usually a history of trauma, sudden onset sion/deterioration. Carbamazepine, amitriptyline or trans-
and pain in the involved area. cutaneous nerve stimulation can be used if the pain does not
• Cervical spondylosis: sensory loss is usually confined to respond to simple analgesics.
the involved nerve roots.
• Motor neuron disease: wasting of the hands but no Surgical
sensory loss. If increasing neurologic deficit occurs. Spinal deformity,
• Multiple mononeuropathy: may be associated with such as kyphoscoliosis, should be first corrected. A variety
peculiar sensory deficits. Usually abrupt onset of loss of of shunts, shunt materials, and shunt locations are used but
function of one, followed by other peripheral or cranial there is no consensus, one reason being our lack of full
nerves. Sensory dissociation in the upper trunk is very understanding of the pathophysiology of syringomyelia, and
uncommon. lack of randomized trials of treatment strategies.
• Diabetic neuropathy: another (but rare) cause of Charcot
joints in the shoulders but other clinical features of Syringomyelia associated with Chiari I malformation
diabetes. The primary goal is to arrest the progression of the neuro-
• Vasculitic neuropathy (systemic: polyarteritis nodosa, logic deficits by decompressing the Chiari I malformation
rheumatoid arthritis, or non-systemic): may present with with a suboccipital craniectomy and upper cervical (C1)
distal symmetric polyneuropathy but more commonly a laminectomy in combination with a duraplasty. If scarring
mononeuropathy multiplex. occludes the outlet of the fourth ventricle, an opening is
• Leprosy: tends to affect pain and temperature sensation made in the scar to re-establish the outlet of the fourth
and may cause a syringomyelia-like syndrome. However, ventricle of treatment strategies.
leprosy preferentially affects the intracutaneous nerves
and so the pattern of sensory loss does not follow the Syringomyelia associated with Chiari II malformation
distribution of individual peripheral nerves or nerve Chiari II is a congenital malformation associated with
roots. In lepromatous leprosy, sensory loss occurs in areas myelomeningocele, hydrocephalus and often lower cranial
with low skin temperature, causing a superficial nerve abnormalities. In addition to the hindbrain herniation
resemblance to a glove and stocking distribution, but seen in Chiari I, the posterior fossa in Chiari II is too small
other cool areas such as the nose, earlobes, breasts, for the cerebellum, and there is upward herniation into the
abdomen and buttocks may also be affected. In addition middle fossa. Consequently, decompression of the posterior
there is usually a history of residence in an area where fossa in Chiari II is not effective in most patients, and may
leprosy is endemic and there are skin lesions and enlarged result in shunt malfunction and immediate herniation into
nerves. the decompression.
• Acute intermittent porphyria: patchy sensory loss
involving the trunk or arms but usually acute onset, all Post-traumatic syringomyelia
four limbs are affected before the trunk, abdominal pain, Treatment of cysts with shunts should be considered a
psychiatric symptoms, or a deficiency of erythrocyte treatment of last resort. Shunting can collapse the cyst but
porphobilinogen deaminase. often complications result in re-expansion and the need for
• Amyloidosis: loss of sensation to pain and temperature subsequent surgery and reshunting. Placing shunts in the
but autonomic dysfunction is also common. spinal cord can also add to the neurologic deficit.
• Fabry’s disease: an X-linked recessive disorder that Decompression by means of a dural graft with a bypass
frequently presents with severe burning pain in the hands for CSF may be helpful.
and feet.
• Tangier disease: may cause a syringomyelia-like syndrome PROGNOSIS
(spontaneous pain in the arms, wasting of the hand Usually slowly progressive and ultimately severely disabling
muscles, dissociated sensory loss in the upper trunk, and but some patients experience a stepwise deterioration, and
facial diplegia) due to selective loss of small myelinated others ‘plateau’ and do not progress.
and unmyelinated nerve fibers and small dorsal-root
ganglion cells, but patients have large, lobulated, yellow-
gray tonsils, and they may have hepatosplenomegaly, a
low serum cholesterol level (unless a second lipid
disorder), and nearly absent high-density lipoprotein
(HDL) and apolipoprotein A-I and A-II.
Cervical Spondylotic Myelopathy and Radiculopathy 545

CERVICAL SPONDYLOTIC ETIOLOGY AND PATHOPHYSIOLOGY


MYELOPATHY AND RADICULOPATHY Predisposing factors
• Narrowing of the spinal canal in the sagittal
(anteroposterior) plane: <11–12 mm (<0.4–0.5 in)
DEFINITION results in deformation of the cord, the degree of which
A condition in which the spinal cord (myelopathy) and/or correlates with the clinical severity of myelopathy. The
nerve roots (radiculopathy) are damaged, either directly by salient static measurement is however, the cross sectional
traumatic compression and abnormal movement, or area of the cord. Reduction in cross sectional area of the
indirectly by ischemia due to arterial compression, venous cord by 30% or more to a value of about 60 mm2 or less,
stasis, or other consequences of the proliferative bony results in symptoms and signs.
changes that characterize spondylosis. • In patients with cervical spondylotic myelopathy caused
by an ossified posterior longitudinal ligament, the
EPIDEMIOLOGY dentate ligaments fix the cord against the anterior part
Most people older than 50 years have cervical spondylosis. of the canal. Sectioning of the dentate ligaments may
Most have no symptoms apart from reduced mobility of the spread the tension in the cord over a greater segment.
cervical spine. • Hypertrophic facet and uncovertebral joints occupy space
• Incidence: 83 per 100 000 per year (cervical radiculopathy), in the root canal.
2 (95% CI: 0.5–6) per 100 000 per year (chronic spondy- • Neck motion: predisposes to spur formation and activates
lotic myelopathy). symptoms and signs of cervical spondylotic myelopathy
• Prevalence (lifetime): 0.4 (95% CI: 0.2–0.7) per 1000 (e.g. patients with athetoid cerebral palsy).
(spondylotic and compressive myelopathy). • Deflexion (i.e. extension) of the neck.
• Age: middle-aged and elderly; peak incidence in the age
group 50–54 years.
• Gender: M>F.

PATHOLOGY
Macroscopic
Spondylosis of the vertebral column
• Transverse bars occur which may extend across the
posterior aspect of the vertebrae and compress the spinal
cord. The lateral end of the transverse bars may encroach
on an intervertebral foramen and compress the nerve root. 675
• Localized bosses occur centrally or laterally, which may
compress the spinal cord.
• Intervertebral disc protrusions are commonly associated
with the bars and bosses.
• Frequently these lesions are found at more than one
vertebral level.

Spinal cord/root compression


• Indentation, flattening and distortion of the spinal cord
occurs corresponding to the spondylotic protrusions (675).
• The C6 or C7 nerve roots are affected in two-thirds of
cases of cervical radiculopathy.

Microscopic
• Demyelination of the lateral columns, ischemic change and
nerve cell damage and loss in the central gray matter, and
cavitation at the site of the compression are all present.
• Degeneration of the dorsal columns occurs rostral to the
lesions.
• Degeneration of the lateral columns occurs caudal to the
lesions.

675 Autopsy specimen of the cervical spine in the sagittal plane


showing a degenerate cervical intervertebral disc (oblique arrow),
compressing the cervical spinal cord causing a necrosis of the cervical
spinal cord at that level (horizontal arrow).
546 Spinal Cord Diseases

Causes Cervical radiculopathy (see Acute ‘slipped disc’,


• A bulging or herniated disc (676). p.550)
• Degenerated yellow ligaments. • Neck and arm pain in some (677, 678).
• A fixed subluxation due to disc degeneration. • Paresthesiae, pain and cutaneous sensory loss in a
• Microtrauma. radicular distribution.
• Wasting and weakness of proximal arm muscles or in the
Mechanism hands.
Direct • Fasciculations in a segmental distribution in the hands
Traumatic compression and abnormal movement. Acute and arms.
trauma is likely to exacerbate any pre-existing myelopathy
in a chronically distorted, narrowly confined, cord. DIFFERENTIAL DIAGNOSIS
Mechanical compression theory: the spinal cord is com- Cervical spine/spinal cord pathology
pressed by a spondylotic bar anteriorly and the ligamenta • Discogenic or degenerative cervical spine stenosis.
flava posteriorly, particularly during extension of the neck, • Vertebral body fracture: trauma, metabolic bone disease,
when the ligamenta flava bulge into the spinal canal, tumor.
decreasing its anteroposterior depth while the antero- • Tumor:
posterior dimension of the spinal cord itself increases. – Tumor of vertebral column or epidural space (or both):
People with congenitally narrow spinal canals are more Metastases.
vulnerable. Plasmacytoma or multiple myeloma.
Dentate tension theory: the anterior spondylotic bar Primary bone tumor.
displaces the spinal cord posteriorly and stretches the dentate Primary intradural tumor of spinal cord.
ligaments, which attach the lateral pia to the lateral dura. The • Infection:
attachment to the dura is fixed because the dural root sleeves – Intervertebral discitis or osteomyelitis.
are anchored in the neural foramina. Consequently, the – Epidural or subdural abscess.
spinal cord is pulled laterally by the dentate ligaments. The – Herpes zoster or other viral-based radiculopathy.
spondylotic bar may also increase dentate tension by • Vascular lesion:
interfering locally with dural stretch during neck flexion, the – Acute epidural hematoma.
resultant increase in dural stress being transmitted to the – Spinal cord infarction.
spinal cord via the dentate ligaments. – Arteriovenous malformation of the spinal cord.
– Spinal dural arteriovenous fistula.
Indirect • Amyotrophic lateral sclerosis.
The role of ischemia is uncertain. There is no correlation • Multiple sclerosis.
with atherosclerosis of major vessels or with obstruction of • Syringomyelia.
blood flow in the anterior spinal artery. Distortion and com- • Diabetic radiculoneuropathy.
pression of small vessels in the cord may have a pathogenetic • Acute inflammatory demyelinating polyradiculopathy
influence. The influence of venous stasis is uncertain. (Guillain–Barré syndrome).

CLINICAL FEATURES Brain pathology


Cervical spondylosis without foraminal or spinal canal • Parasagittal meningioma.
stenosis does not result in cervical spondylotic radiculopathy • Bilateral anterior cerebral artery territory infarction.
or myelopathy. • Bilateral corticospinal tract infarction (e.g. internal
capsule and pons).
Cervical spondylotic myelopathy • Primary lateral sclerosis.
• Neck pain in some. • Hydrocephalus.
• Numb, clumsy hands:
– Amyotrophic hand: localized wasting and weakness of INVESTIGATIONS
the hand without remarkable sensory loss or gait Plain films of the cervical spine
disturbance. Correlates with a reduced cross-sectional Useful because they demonstrate narrowing of the spinal
area of the spinal cord at the C7–T1 segments. Similar canal, osteophytes arising from the posterior surfaces of
to hands of patients with spinal muscular atrophy. degenerate discs and at the neurocentral joints, and the
– Myelopathic hand: spastic dysfunction and deficient pain alignment of the vertebral bodies. Flexion and extension
sensation due to reduction in spinal cord diameter at views may be done to look for instability with movement.
higher spinal levels. They do not, however, give any information on the degree
• Spastic paraparesis: the sine qua non for the diagnosis. of cord or root compression or the soft tissue abnormalities
• Loss of proprioception and vibratory sensation in the legs. around the spine.
• Poorly defined cutaneous sensory loss in the legs.
Plain CT (679)
CT has a limited role; it images osteophytes and calcified discs,
and accurately measures the dimensions of the bony spinal
canal, but the cervical cord and roots cannot be assessed.
Cervical Spondylotic Myelopathy and Radiculopathy 547

676 Illustration showing the effect of a cervical spondylitic 676


bar, osteophyte or degenerate intervertebral disc at the
C5/6 level, compressing the cervical spinal cord (causing a
Spinal cord
cervical myelopathy) and/or left C6 nerve root (causing a compressed by
left C6 radiculopathy). spondylitic bar

Spine of C6 vertebra

Osteophyte

C6 root damaged by
lateral osteophyte

Degenerate
intervertebral disc

Vertebral artery

677 678
C3
C4 C3
C5 T2 C4
T3 C5
T4 T2
T2 T3
T5 T4
C6 T3 T5 T2
T6
T6 C6
T1 T7 T3
T7 T1
T8 T8
C6 T9
T9 C6
T10
T10
C7 T11
T11 C7
C8 T12
T12 L1 C8
L1

677, 678 Distribution of the cervical and thoracic dermatomes on 679


the anterior aspect of the upper limb, chest and abdomen (677), and
the posterior aspect of the upper limb and back (678).

679 CT scan of the cervical spine, axial plane at C5/6 showing very
advanced spondylotic change with anterior and posterior lipping
osteophytes and a large osteophyte arising from the posterior aspect
of the C5 disc (circle) causing markedly severe central spinal canal
stenosis.
548 Spinal Cord Diseases

MRI cervical spine flexion of the neck which increases dural tension.
The imaging modality of choice (680–684). Improvement has been reported in nearly half of patients
• Sagittal T1W and T2W images show the cord and level treated with a cervical collar but the studies have been
of any compression. uncontrolled.
• Axial views at those levels further define the degree of Surgical decompression is considered in patients who are
cord and root involvement. moderately or severely disabled on initial assessment and
• Disc protrusions are seen in about 20% of neurologically who, on close follow-up, have progressive impairment of
asymptomatic people aged 45–54 years, and in about function without sustained remission. Measurement of
57% of asymptomatic people older than 64 years. cervical mobility on functional radiographs may help to
• Spinal cord impingement (a concave defect in the spinal select patients who are more likely to deteriorate and thus
cord adjacent to a site of disk bulging, without obliteration more likely to benefit from surgery, such as those with spinal
of the subarachnoid space posterior to the cord) is seen in hypermobility possibly. (N.B. Male patients with symptoms
about one-sixth of neurologically asymptomatic people and signs of prostate enlargement should first be treated for
under 65 years and one-quarter over 65 years. that problem to avoid major post operative micturition
• Cord compression (with obliteration of the posterior disturbances). Less suitable cases for surgery are those with
subarachnoid space) may also be neurologically advanced neurologic changes (e.g. existing severe
asymptomatic but usually the percentage reduction in myelopathic features and evidence of severe cord atrophy),
cord area in asymptomatic people is well below 30% associated neuropathies (i.e. due to diabetes and
(usually below 15%). Thinning of the cord opposite a disc alcoholism), and those too elderly to engage actively in a
protrusion is indicative of compression, worse if there is post operative rehabilitation programme.
increased signal in the cord at that point on T2WI. The aim of surgical therapy is to arrest the progression
Increased signal intensity on T2W images of a compressed of myelopathy by elimination of mechanical compression of
cord reflect myelomalacia, demyelination, gliosis or the dura-enclosed spinal cord, but the risks and benefits of
microcavities. An intense signal correlates with clinical surgical therapy in a large proportion of patients are
deficit and probably reflects inflammation or edema. uncertain and a randomized controlled trial is sorely needed.
• Nerve root compression can occur without significant Surgical techniques include laminectomy (which would
cord compression in which case axial views targeted to seem unhelpful if the problem is not one of compression by
the level of the patient’s symptoms (plus the level the posterior elements of the spine) with or without
immediately above and below) are required. Absence of facetectomy (which releases the tethering of the dural sac
the fat signal from within the root canal, in addition to by the dural root sleeves in the foramina, removing the fixed
obvious abnormal mass (e.g. disc or bone) and point against which the dentate ligaments can be stretched);
displacement or obliteration of the nerve is good opening the dura and sectioning the dentate ligaments (but
evidence on imaging of compression. extensive adhesion may form and involve the spinal cord),
and, perhaps preferably, removal of the spondylotic bar
Computer assisted myelography (CAM) through an anterior or posterior approach that remains
• Useful if MRI quality is suboptimal, the MRI study extradural. This removes the source of posterior pressure on
inconclusive, or the patients is unable to have MRI. the spinal cord and the source of interference with
• Useful for delineating the bone and soft tissue abnormali- stretching of the dura during cervical flexion. Fixation of
ties but it is an invasive procedure.
• It has a low complication rate, mostly due to cervical
spine hyperextension or lateral C1–C2 puncture. 680 680 MRI of the cervical
and upper dorsal spine,
DIAGNOSIS sagittal T2WI, of the same
• Diagnosis is based on clinical symptoms and signs of patient in 679 who had
spinal cord (± nerve root) compression (i.e. spastic severe Parkinson’s disease
paraparesis) with unequivocal MRI or CAM evidence of and had fallen forwards
spinal cord compression that correlates precisely with the and landed on his face. He
clinical findings. was so bradykinetic that
• Key clinical features are: he failed to move his arms
– A spastic paraparesis. forwards to cushion the fall
– Numb, clumsy hands. and consequently fell on
– Sudden quadriplegia or paraplegia after a minor fall in an his face, suffering a sudden
elderly patient. hyperextension injury to
• Diseases such as motor neuron disease and multiple the cervical spine. In the
sclerosis must be ruled out by a thorough neurologic presence of severe cervical
clinical assessment, plain cervical spine radiographs in spondylosis he
flexion and extension, in combination with MRI and if compressed the cervical
necessary CAM. spinal cord and presented
with quadriplegia of
TREATMENT sudden onset. He had a
Patients without major neurologic deficits or signs of motor level at C6/7 and a
worsening are probably best treated conservatively and sensory level at T5 on the
observed closely over time. A restraining collar may help left and T8 on the right.
limit the mobility of the cervical spine, and particularly
Cervical Spondylotic Myelopathy and Radiculopathy 549

the vertebrae bordering the spondylotic bar with a bone PROGNOSIS


graft may also help in reducing spinal mobility. This form of The natural history is not well known because most patients
surgery fails in about 25% of cases however, possibly because undergo some form of surgical treatment. Spontaneous
post operative adhesions between the anterior dura and the regression and complete remission is unusual. About one-
anterior wall of the spinal canal may cause increased tension third of patients experience a recurrence during a median
in the dura during neck flexion. time of follow-up of 5 years.

681 682

681, 682 T1W sagittal (681) and T2W axial (682) MRI of a C5/6
disc herniation. Note that the disc (arrows) protrudes posteriorly and
to the left, displacing the cord posteriorly and to the right and
obliterating the left C6 nerve root.

683 684

683, 684 T2W sagittal (683) and T2W axial (684) MRI of the
cervical spine in a patient with severe cervical spondylosis. Note the
marked narrowing of the spinal canal at multiple levels. On the axial
view the cord appears ribbon-like. Also note the increased signal within
the cord indicating gliosis.
550 Spinal Cord Diseases

ACUTE ‘SLIPPED DISC’ (seconds-minutes-hours), associated symptoms (e.g. limb


weakness or sensory disturbance, bladder or bowel dysfunc-
tion, weight loss, fever), exacerbating factors (e.g. cough,
DEFINITION sneeze, strain) and relieving factors and body positions.
Acute extrusion of the nucleus pulposus of an intervertebral • History of trauma, weight loss, malignancy, other
disc. systemic illness.
• Medication/drug history (cf. ?analgesic abuse).
EPIDEMIOLOGY • Explore secondary gain phenomenon such as litigation,
• Common. worker’s compensation issues, psychiatric problems,
• Incidence – acute cervical myelopathy related to disc: narcotic abuse.
2 (95% CI: 0.2–6) per 100 000 per year.
• Age: young adults or middle-aged. EXAMINATION
• Gait: examine a few steps while walking ‘normally’, on
PATHOLOGY toes, on heels, heel-to-toe; hopping on either leg, arising
• The prolapse is usually posterolateral rather than central. from a squat, Rhomberg’s sign.
• The lumbar discs are most often affected (L4/5 or • Spinal tenderness or spasm.
L5/S1), cervical discs occasionally (C7 root involved in • Limited range of motion of the spine.
70% of cases of cervical disc protrusion, C6 root in 20% • Aggravation of pain with movement of the spine
of cases, C5 and C8 roots in the remaining 10%), and (particularly hyperextension of the neck and downward
thoracic discs rarely. pressure on the head in the hyperextended position with
cervical disc protrusion).
ETIOLOGY AND PATHOPHYSIOLOGY • Straight-leg raising (Lasegue’s sign).
• Acute slipped disc may be spontaneous or precipitated • Limb muscle bulk, tone, power, deep tendon reflexes,
by sudden spine movement (e.g. sudden hyperextension sensation (including perianal sensation and anal tone if a
of the neck, diving, forceful chiropractic manipulations), cauda equina syndrome is suspected).
lifting and straining. • Signs of systemic disease, infection or malignancy.
• A prolapsed cervical disc compresses the nerve root and
spinal cord at that level (e.g. a laterally situated disc DIFFERENTIAL DIAGNOSIS
protrusion between the sixth and seventh cervical Painless cervical myelopathy (see p.545)
vertebrae compresses the seventh cervical root) (676), • Subacute degeneration of spinal cord.
whereas a prolapsed disc in the lumbar region (e.g. at • Motor neuron disease.
L4/5), where nerve roots are angulated, usually
compresses the root passing to the foramen below (e.g. Back pain with or without radiculomyelopathy
the L5 root) (685). • Discogenic or degenerative spinal stenosis:
• Nerve root compression usually involves only one nerve – Herniated intervertebral disc.
root on one side (perhaps two) but a central lumbar disc – Degenerative spondylosis:
prolapse may compress most of the cauda equina, Central canal stenosis.
causing multiple lumbosacral root lesions and sphincter Lateral recess stenosis.
disturbance. – Synovial cyst of facet joint.
• Spinal cord compression by central cervical or thoracic • Metabolic bone disease: osteoporotic compression fracture.
discs (at or above the level of the conus medullaris, which • Spinal trauma.
is at the level of the L1/2 intervertebral disc) causes • Spinal tumor:
upper motor neuron signs. – Primary intradural tumor of spinal cord, medullary cone
or cauda equina.
CLINICAL FEATURES – Tumor of vertebral column or epidural space (or both):
• Acute pain, tenderness and paraspinal muscle spasm at the Metastases.
level of the prolapse. N.B. Central cervical disc Plasmacytoma or multiple myeloma.
herniations, compressing the spinal cord, may be painless. Primary bone tumor.
• Nerve root (radicular) pain, which is sharp, burning or – Extraspinal retroperitoneal malignant lesion.
‘electric’, and severe; combined with sensory disturbance; • Vascular lesion:
in the dermatomal distribution of the compressed nerve – Acute epidural hematoma.
(677, 678, Table 45, 686). – Spinal cord infarction.
• Pain: exacerbated by coughing, sneezing, straining, mov- – Arteriovenous malformation of the spinal cord.
ing (bending and stooping), straight leg raising (low – Spinal dural arteriovenous fistula.
lumbar and sacral root compression), and femoral stretch • Infection:
test (upper lumbar roots); relieved by rest and particular – Intervertebral discitis or osteomyelitis.
postures. – Epidural or subdural abscess.
• Muscle wasting and weakness and depressed tendon – Urinary tract infection.
reflex in myotomal distribution (see Tables 46, 47). – Herpes zoster or other viral-based radiculopathy.
• Intra-abdominal or intrapelvic pathology:
HISTORY – Abdominal aortic aneurysm.
• Neck/back pain: location (radicular), onset (usually acute, – Posterior perforating duodenal ulcer.
?precipitating factors), timing (?nocturnal), nature (sharp), – Pancreatic disease.
intensity (severe), radiation (dermatomal), duration – Endometriosis.
Acute ‘Slipped Disc’ 551

• Degenerative hip disease. 685


• Diabetic radiculoneuropathy. Dura mater Subarachnoid Dura mater Subarachnoid
• Acute inflammatory demyelinating polyradiculopathy space space
(Guillain–Barré syndrome).
• Congenital:
– Tethered cord.
– Intraspinal lipoma.

Radiculopathy
Mononeuropathy: e.g. ulnar nerve palsy may be mistaken L5 L5
for compression of the eighth cervical root by a disc L4 L4
protrusion at C7–T1.

L5 L5

S1 S1

685 Illustration showing the effect of a lumbar disc prolapse at L4/5


which is central (illustration on the left), compressing many or all of the
nerve roots below L4, particularly the most anterior roots (L5, S1, S2); 686
and lateral (illustration on the right), compressing the L5 nerve root L1
T10
(and sometimes the S1 nerve root) and displacing the root pouch on
the side of the prolapse. T11 S4
S5 L2
S3
T12
Table 45 Nerve root (radicular) pain and sensory L1
disturbance in the dermatomal distribution of the
S3
compressed nerve (677, 678 and see p.545)
S2
C5: lateral shoulder and upper arm S4
L2
C6: lateral forearm, thumb and index finger
C7: middle finger
C8: ring and little fingers and medial forearm
L3
T1: medial forearm and upper arm
L4: pain radiates down the leg to the thigh, knee, medial lower leg
and medial malleolus and sometimes the groin L3
S2
L5: lateral lower leg; lateral malleolus and dorsum of the foot
S1: heel, lateral border and sole of the foot L4
S2: back of leg
S3,4,5: perineal and perianal region (686).
Pain: sharp,‘electric’, and severe, and exacerbated by coughing, S1
L5
sneezing, straining, moving (bending and stooping), straight leg raising
L4
(low lumbar and sacral root compression), and femoral stretch test
(upper lumbar roots). Relieved by rest and particular postures

S1

Table 46 Innervation of spinal segments and muscles S1 L5


L5
Spinal Muscle Action
segment
686 Distribution of the lumbar and sacral dermatomes in the lower
C5, C6 Deltoid Shoulder abduction abdomen, buttocks and legs.
C5, C6 Biceps Elbow flexion
C6, C7 Extensor carpi radialis Wrist extension
C7, C8 Triceps Elbow extension Table 47 MRC grading scale for evaluation of
C8,T1 Flexor digitorum profundus Hand grasp muscle strength
C8,T1 Hand intrinsics Finger abduction
L1, L2, L3 Iliopsoas Hip flexion MRC grade Muscle strength
L2, L3, L4 Quadriceps Knee extension 5 Normal strength
L4, L5, S1, S2 Hamstrings Knee flexion 4 Active power against both resistance and gravity
L4, L5 Tibialis anterior Ankle dorsiflexion 3 Active power against gravity but not resistance
L5, S1 Extensor hallucis longus Great-toe extension 2 Active movement only with gravity eliminated
S1, S2 Gastrocnemius Ankle plantar flexion 1 Flicker or trace of contraction
S2, S3, S4 Bladder and anal sphincter Voluntary rectal tone 0 No movement or contraction
552 Spinal Cord Diseases

INVESTIGATIONS • Myelography will demonstrate canal narrowing, nerve


Screen for non-discogenic causes of back pain root displacement, thickening and cut off, disc hernia-
• Plain radiography of the relevant part of the spine tions and the number of levels involved, but is invasive.
(anteroposterior and lateral projections). • Myelography with water-soluble contrast medium followed
• Full blood count. by CT (myelogram-CT) images the subarachnoid space
• ESR. and may reveal cauda equina tumors and other intradural
• Urea and electrolytes, glucose, calcium, phosphate and lesions that can mimic lumbar disc herniation.
liver function tests. • MRI is excellent for imaging the lumbar spine and shows
• Urinalysis. the features of disc herniation, canal stenosis and so on
• Prostatic specific antigen (in men older than 50 years of age). (687–691). As with CT, a pre- and post-contrast exam
is required if there has been previous surgery. T1W and
Electromyography T2W midline sagittal followed by T1W axial images are
EMG can help confirm the clinical impression of the absence routine. Absolute contraindications include the presence
or presence, site and severity of a radiculopathy and may reveal of a pacemaker or ferromagnetic implant (e.g. intracranial
an unsuspected polyradiculopathy or peripheral neuropathy. vascular clip); relative contraindications include claustro-
phobia, obesity and the need for advanced life support.
Imaging of lumbar spondylosis
• Plain films may demonstrate narrowing of individual disc DIAGNOSIS
spaces and the spinal canal, either congenital or Diagnosis is based on findings of nerve root pain, objective
secondary to degenerative joint disease, but are rather neurologic signs of spinal cord and/or nerve root compression,
non-specific and do not show the contribution from any and clear-cut evidence of a herniated disc on MRI or
soft tissue component. myelogram-CT that correlates precisely with the clinical findings.
• CT is very good and can be set up to cover the whole
lumbar spine, by imaging through each disc space and TREATMENT
the mid point of each vertebral body. Thus osteophytes, Conservative
disc herniations, ligament hypertrophy, facet joint • Reassure patients that they most likely have a benign,
hypertrophy, lateral recess stenosis and exit canal self-limited condition and that symptoms should rapidly
narrowing can be demonstrated at all the lumbar levels. resolve without active treatment. Hospitalization is
When disease is suspected at a specific level, then images usually not needed.
of the corresponding disc space plus the one above and • Simple analgesia: aspirin, non-steroidal anti-inflammatory
below are usually obtained. If there has been previous drugs, paracetamol (acetaminophen); narcotic analgesics
disc surgery the examination should be done before and are usually unnecessary.
after intravenous contrast. • Muscle relaxants (e.g. cyclobenzaprine hydrochloride or
methocarbamol) for a short term in selected patients
with pronounced paraspinal muscle spasm.
• Bed rest: short term (2–3 days), on a firm mattress or board.
687, 688 T2W 687 • Immobilization of the involved spine: avoid movements
sagittal (687) and of the involved spine and strain on the spine; for cervical
T1W axial (688) MRI disc protrusions use a neck collar that is rigid enough to
of an L5/S1 disc prevent neck movement, particularly if flexion and
(arrows) herniation. extension are painful (a soft collar may be more tolerable
at night); for lumbar disc prolapses, use a firm lumbar
corset. Use a chair with adequate back support, and
change positions frequently. Control body weight.

688
Acute ‘Slipped Disc’ 553

• Begin a physiotherapy programme: aerobic exercise as Lumbar discectomy with magnified vision is the ‘gold
tolerated but avoiding heavy lifting and repetitive bend- standard’ operation for lumbar disc disease; various closed
ing and twisting of the back. Traction, manipulation, or percutaneous procedures, such as chemonucleolysis,
ultrasound therapy, transcutaneous electric stimulation, percutaneous automated discectomy, and endoscopic
and application of heat and cold can be effective. discectomy are being trialled. Chymopapain is more
• Lumbar epidural corticosteroid injections may be of effective than placebo for lumbar disc prolapse, but
benefit in selected patients. discectomy may lead to better clinical outcomes with fewer
• Reassess the few patients who have not experienced second procedures than chymopapain.
improvement within 6 weeks. Surgical complications include great vessel and cauda
equina injury and deep spinal infection but are uncommon.
Surgical decompression
• Emergency: PROGNOSIS
– Acute cord compression. • Acute neck and low back pain, in the absence of tumor
– Acute bladder disturbance due to cauda equina and other serious underlying disease, usually resolves
compression by a central lumbar disc prolapse. rapidly within 4–6 weeks.
• Urgent: severe or progressive loss of motor function due • Lumbar discectomy is successful in 80–90% of patients,
to a monoradiculopathy; trivial root signs such as absent if properly selected.
tendon reflex are not an indication for surgery. • Patients with severe cauda equina syndrome due to
• Elective: continuing nerve root pain with a mild to massive midline disc herniation have a guarded prognosis
moderate neurologic deficit which has not responded to for neurologic recovery, even with prompt disc removal
about 6 weeks of conservative treatment. and neural decompression.

689, 690 T1W 689 691


axial pre- (689) and
post contrast (690)
MRI of the lumbar
spine in a patient
with fibrosis
following previous
lumbar disc surgery.
Note the enhancing
tissue around the
right hand side of
the spinal theca
(arrow).

691 T2W sagittal


MRI of thoracic
discs (arrows).

690
554 Spinal Cord Diseases

SPINAL EPIDURAL HEMATOMA MRI spine


MRI is the primary method of diagnosis (693). The
findings depend on the timing of the MR scan after the
DEFINITION onset of the hemorrhage. At about 36 hours after onset,
Hemorrhage in the spinal epidural space. T1W MRI of the spine usually shows an isointense
collection in the epidural space, usually anterior, with or
EPIDEMIOLOGY without cord or nerve root compression. An underlying
• Uncommon. spinal vascular malformation may be seen.
• Age: any age.
• Gender: M=F. CT spine
CT may also show an epidural mass, which does not tend
PATHOLOGY to enhance with contrast.
• Blood in the spinal epidural space.
• Direct compression of the spinal cord or roots by the Chest x-ray
mass effect of the hematoma. Hilar lymphadenopathy, pulmonary nodules or interstitial
• Spinal cord/nerve root pressure necrosis. lung disease may be seen in patients with arteritis causing
spinal epidural hemorrhage.
ETIOLOGY AND PATHOPHYSIOLOGY
• Trauma. Spinal angiography
• Spinal extradural arteriovenous malformation (692). May identify the source of the hemorrhage and even
• Vertebral body hemangioma. distinguish between the several types of vascular
• Epidural metastases. malformations and hemangioblastomas, and localize them
• Arteritis. accurately to the vertebral bodies, epidural or subdural
• Hypertension. space, or spinal cord.
• Bleeding diathesis due to a coagulopathy.
• Anticoagulant therapy.
• Cocaine injection.
• Epidural anesthesia.
• Lumbar puncture in patients with a bleeding diathesis.
• Idiopathic.

CLINICAL FEATURES 692 693


Sudden and severe neck or back pain, followed in minutes
to hours by progressive motor, sensory and sphincteric
disturbances referable to radicular spinal cord or cauda
equina origin. Other clinical features are determined by the
underlying cause (e.g. vasculitis causing associated skin rash
[693–695]).

DIFFERENTIAL DIAGNOSIS
• Hemorrhage into the spinal cord (hematomyelia) or
spinal subarachnoid space (693).
• Intervertebral disc prolapse.
• Vertebral body collapse (spinal angulation: tuberculosis,
tumor).
• Trauma.
• Spinal cord infarction.
• Epidural abscess.

INVESTIGATIONS
Blood
• Full blood count.
• ESR.
• Coagulation profile.
• Serum chemistry profile. 692 Autopsy specimen of spinal cord showing an epidural spinal
• VDRL/RPR and TPHA. arteriovenous malformation (a common cause of spinal epidural
• Antinuclear antibody titer. hematoma), containing dark clotted blood. (Courtesy of Professor BA
• Urine test for cocaine/amphetamine metabolites. Kakulas, Royal Perth Hospital,Western Australia.)

693 MRI cervical and upper thoracic spine,T1W image, sagittal plane,
showing a linear streak of high intensity due to hemorrhage in the
spinal subarachnoid space in a patient with arteritis due to
Churg–Strauss syndrome (allergic angiitis and granulomatosis).
Spinal Epidural Abscess 555

CSF SPINAL EPIDURAL ABSCESS


Blood and xanthochromia are present in the spinal fluid, if
there has been subarachnoid hemorrhage or hemorrhage
into the spinal cord (hematomyelia). DEFINITION
Pus or infected granulation tissue in the spinal epidural
DIAGNOSIS space.
MRI spine or spinal angiography in a patient with
appropriate clinical syndrome. EPIDEMIOLOGY
• Rare.
TREATMENT • Age: any age; children or adults.
• Prompt neurosurgical decompressive laminectomy, • Gender: either sex.
particularly if acute onset and neurologic function is
deteriorating. PATHOLOGY
• Microsurgical resection or embolization of the cause (e.g. • An abscess containing pus or infected granulation tissue in
if a vascular malformation is present). any part of the spinal epidural space, most commonly the
dorsal aspect of the thoracic spinal cord, where the loose
PROGNOSIS attachment of the dura permits rapid spread of infection.
Depends on the site, duration and degree of spinal cord and • Direct compression of the spinal cord or roots by the
nerve root compression, and the underlying cause of the mass effect of the abscess.
hematoma. • Spinal cord infarction secondary to pressure and
thrombophlebitis of the local venous channels.

ETIOLOGY
• Staphylococcus aureus: most common.
• Streptococci.
• Brucella (a Gram-negative coccobacillus which may infect
694 people in close contact with infected animals and
carcasses [camels, cows, goats, sheep] or after drinking
infected milk).
• Salmonella.
• Fungus.
• Tuberculosis (696) (complications include nerve root
and/or spinal cord compression, arteritis and spinal cord
infarction, spinal arachnoiditis, and spinal cord
tuberculoma).
• Anaerobic organisms.

696

695 694, 695 Hands (694) and shin


(695) of the patient with
Churg–Strauss syndrome (693)
showing the typical skin rash.

696 Spine of a patient with a


tuberculous spinal epidural
abscess due to localized spinal
meningitis (Pott’s disease of the
spine) with vertebral body
collapse and destruction of the
adjacent intervertebral discs.
556 Spinal Cord Diseases

PATHOPHYSIOLOGY INVESTIGATIONS
Seeding of the spinal epidural space or a vertebral body with Blood
micro-organisms occurs under the following circumstances: • Neutrophil leukocytosis.
• Septicemia complicating a chronic medical illness (e.g. • Raised ESR.
diabetes, infective endocarditis, immunocompromise) with • Serology may be positive for infectious organisms such
seeding of the spinal epidural space or of a vertebral body. as brucella.
• Vertebral osteomyelitis.
• Infected intervertebral disc. MRI spine
• Penetrating or non-penetrating back injury at the time The imaging method of choice (697–699). The findings
of furunculosis or other skin or wound infection. are similar to those of epidural tumors (see p.567). Bone
• Local spinal surgery: laminectomy. involvement varies as the abscess can simply sit as a cuff in
• Spinal or epidural anesthesia. the spinal canal with minimal bone involvement, though
• Lumbar puncture. more commonly bone involvement is substantial. The cord
appears normal width or narrowed, with the CSF obliterated
CLINICAL FEATURES around it at the level of the lesion, but visible above and
• Subacute onset. below, and a mass around the outside plus or minus bone
• Systemic upset: fever, headache, malaise. destruction. MRI not only demonstrates the extent of the
• Back pain and tenderness to palpation/percussion: local symptomatic lesion, but also other lesions that may be at
and severe. other levels and may be asymptomatic.
• Neck/spine rigidity.
• Nerve root pain. Spine x-ray
• Nerve root and/or spinal cord compression/infarction: Bone changes of osteomyelitis.
progressive paraparesis, sensory loss and sphincter
paralysis (urinary and fecal retention). Myelography (with or without CT)
Shows spinal cord compression. It may only show up to the
DIFFERENTIAL DIAGNOSIS lower level of the lesion, as if the block is complete, the
• Epidural hematoma (see p.554). contrast will not pass it to outline the cord above.
• Acute intervertebral disc prolapse (see p.550).
• Subdural abscess: spinal imaging often reveals a less sharp CSF
margin and a greater vertical extent of the abscess than • Cells: mild pleocytosis: <100 white cells/mm3, both
an epidural abscess. polymorphonuclear leukocytes and lymphocytes; frank
• Transverse myelitis (see p.561). pus if the needle penetrates the abscess.
• Spinal cord abscess. • Protein: quite high: 1–4 g/l (100–400 mg/dl).
• Spinal cord infarction (see p.558). • Glucose: normal.
• Culture: often sterile.

697 698 DIAGNOSIS


The definitive pre-surgical diagnosis can only be made with
an MRI scan, indicating the extent and localization of the
lesion, in combination with clinical, serologic, microbiologic
and/or pathologic information.

699

697–699 MRI lumbar spine,T1W (697) and dual echo T2W (698)
images in the sagittal plane from T11 to S3, and axial T1W (699)
images at the L3/4 disc level showing at the L3/4 disc level an anterior
epidural soft tissue mass which causes severe canal stenosis.The soft
tissue mass (arrows) is of intermediate signal intensity on T1W images
with increased signal on T2W images and minimal contrast
enhancement.The differential diagnosis includes epidural abscess,
tumor or hemorrhage.
Spinal Arachnoiditis 557

TREATMENT PATHOGENESIS
• Surgical decompression by laminectomy and drainage as • The arachnoid is an avascular membrane which lies
soon as possible if the spinal cord is compressed. between the vascular pia and the vascular dura. It has a
• Antibiotics appropriate for the infecting organism must limited capacity to participate in an inflammatory response.
be given (e.g. for brucellosis: doxycycline, 200 mg orally, • In response to injury or irritation, a reactive inflammatory
once daily, plus rifampicin (rifampin), 900 mg orally, response begins in the vascular pia and dura and progresses
once daily for at least 6 weeks, or tetracycline, 500 mg to a chronic stage with fibrous thickening of the arachnoid
orally, four times daily for 6 weeks plus streptomycin, 1g and adhesions between the pia and the dura.
daily i.m. for 3 weeks).
PATHOLOGY
PROGNOSIS • Opacification and thickening of the arachnoidal
• A fibrous or granulomatous reaction may develop at the membranes.
site of apparently successful surgical drainage and give • Adhesions between the arachnoid and dura due to
rise to a slowly progressive, then stabilizing syndrome of proliferation of connective tissue in response to an
incomplete cord compression. antecedent arachnoidal inflammation.
• If allowed to smoulder due to delayed diagnosis or • Obliteration of the subarachnoid space.
inadequate therapy, a chronic adhesive meningomyelitis • Strangulation of nerve roots and the spinal cord by
may evolve. thickened connective tissue, causing compression and
progressive vascular occlusion and ischemia of the spinal
roots and cord, and obstruction of CSF flow at the
foramen magnum, possibly causing hydrocephalus and
syringomyelia.
• Variable destruction of the peripherally placed fibers of
the spinal cord.
SPINAL ARACHNOIDITIS • Secondary degeneration of the dorsal and lateral columns
of the spinal cord.
• Usually diffuse, with a predilection for the thoracic
DEFINITION segments.
A non-infectious or post infectious aseptic inflammatory • Occasionally confined to relatively circumscribed sections
process of the leptomeninges which may or may not be of the cord or subarachnoid space, giving rise to
associated with other disease of the spine or spinal cord and loculated collections of fluid (meningitis serosa
results in a progressive overgrowth of fibrous tissue in the circumscripta).
subarachnoid space.
CLINICAL FEATURES
EPIDEMIOLOGY Asymptomatic
• Uncommon.
• Age: any age; most commonly 40–60 years of age; rarely Myeloradiculopathy
younger than 20 years. • Onset: concurrent with acute arachnoidal inflammation
• Gender: M=F. or delayed for weeks, months or even years.
• Dorsal sensory nerve root compression:
ETIOLOGY – Spine and/or radicular pain in the distribution of one or
• Myelography using oil based contrast media (pantopaque, more sensory nerve roots, initially on one side and then
Myodil [iophendylate]). These substances had to be bilaterally. Persistent and burning, stinging or aching.
removed from the CSF space by aspiration through the – Depressed deep tendon reflexes.
lumbar puncture needle once the procedure had finished. • Ventral motor nerve root compression: muscle wasting
Due to the oily nature it was impossible to remove all of and weakness (particularly with cauda equina
it. The contrast excited an inflammatory reaction leading involvement).
to arachnoiditis. One occasionally still sees plain x-rays of
the spine with little radiopaque droplets overlying the Spinal cord compression
spinal canal indicating a previous Myodil myelogram. Spastic ataxic paraparesis, slowly progressive (may follow
Modern water-soluble contrast media rarely, if ever, cause months or years after radicular symptoms).
arachnoiditis.
• Intrathecal administration of penicillin and other Signs of the underlying cause
antibiotics, spinal anesthetics (a detergent that had Previous disc surgery, Myodil (iophendylate), infection,
contaminated vials of procainamide), and repeated corti- subarachnoid hemorrhage, malignancy.
costeroid injections.
• Repeated spinal surgery (e.g. for lumbar discs).
• Subarachnoid hemorrhage.
• Meningeal infection:
– Viral: lymphocytic choriomeningitis.
– Bacterial: acute: meningococcal, gonococcal, listeria;
subacute or chronic: tuberculosis, syphilis, cryptococcus.
– Worms: cysticercosis.
• Ankylosing spondylitis.
558 Spinal Cord Diseases

DIFFERENTIAL DIAGNOSIS SPINAL CORD INFARCTION


• Spinal cord tumor (see p.567).
• Cervical and lumbar spondylotic myeloradiculopathy (see
p.545). DEFINITION
• Subacute/chronic meningitis (see p.279). Infarction of the spinal cord caused by arterial occlusion, or
less often, venous occlusion.
INVESTIGATIONS
• Can be imaged with myelography (700), CT or MRI: EPIDEMIOLOGY
• Non-ionic water-soluble myelography: shows various • Rare.
specific features including truncated or absent nerve root • Age: any age; mean 55 years.
sleeves, clumping of the nerve roots, adherence of the • Gender: M>F (2:1).
roots to the thecal membranes, and obliteration of the
thecal sac. PATHOLOGY
• CT myelography: also demonstrates these features, but • Infarction of the anterior two-thirds of the spinal cord
on axial imaging it is often easiest to appreciate that the with occlusion of the anterior spinal artery or a spinal
nerve roots are abnormally bunched together as the radicular artery.
other signs may be more subtle. The meninges may or • Infarction of one dorsal column with unilateral posterior
may not enhance. spinal artery occlusion.
• MRI: shows similar features to CT, with the most easily • Ischemic or hemorrhagic infarction with venous
appreciated sign again being bunching of the nerve occlusion.
roots. The meninges may or may not enhance.
• CSF: moderate lymphocytic pleocytosis, elevated protein PATHOPHYSIOLOGY
(sometimes extremely so); CSF may be normal or have Blood supply to the spinal cord
only a slightly raised protein in localized lumbar Anterior spinal artery
arachnoiditis. The gray and inner white matter of the anterior two-thirds
of the spinal cord (anterior and lateral columns on either
DIAGNOSIS sides) receives its blood supply via the left and right
Diagnosis is based on clinical features of a sulcocommissural arterial branches of the anterior spinal
myeloradiculopathy with characteristic CSF, and imaging
findings of obliteration of the nerve root sheaths, smooth
outline of the theca, and eventually attenuation and 700
obliteration of the thecal sac.

TREATMENT
• No effective treatment for chronic adhesive lumbar
arachnoiditis.
• Corticosteroids seem ineffective in controlling the
inflammatory reaction and preventing progression of the
disease.
• Surgery may be helpful for the rare case of localized ‘cyst’
formation and cord compression.
• Posterior rhizotomy may help relieve severe radicular
pain.

Prevention
• Avoid oil-based and ionic water-soluble contrast media
for myelography.
• Use caution in the selection and preparation of spinal
anesthetic agents.

PROGNOSIS
Variable: partial or complete recovery within a year or two,
no change, gradual progression and sometimes fatal.

700 Lumbar myelogram showing narrowing of the spinal theca, with


nerve root cut-off typical of arachnoiditis.
Spinal Cord Infarction 559

artery, which originates in its most rostral portion from the ETIOLOGY
union of paired branches of the vertebral arteries (proximal Hypoxia
to their union to form the basilar artery) at the ventral • Respiratory or cardiac arrest.
surface of the medulla oblongata and passes down the • Anemia.
anterior surface of the cord in the midline, narrowing near
the upper fourth thoracic segment. Low spinal cord perfusion
Along its discontinuous course below T4, it receives • Prolonged, severe hypotension.
segmental input from six to nine intercostal radicular arteries
arising from the aorta. The major radicular artery, the great Aorta, vertebral or radicular branch artery occlusion
ventral radicular artery or artery of Adamkiewicz, arises Embolism
usually on the left, variably from T9–T12, or less commonly • Embolism from the heart.
anywhere from T5–L2, and supplies the lumbar cord and • Atherothromboembolism from the aorta or origin of the
conus medullaris. At the lumbosacral level, radicular arteries intercostal and lumbar arteries.
are derived from larger regional vessels, the largest of which • Air embolism: nitrogen bubbles lodge in spinal veins
enters the intervertebral foramen at L2 to form the (decompression sickness).
lowermost portion of the anterior spinal artery (the terminal • Fibrocartilaginous embolism from intervertebral disc
artery) which runs along the filum terminale. rupture.
• Therapeutic renal artery embolization.
Posterior spinal arteries
The posterior one-third of the spinal cord (posterior white Aortic, vertebral and radicular artery disease
columns and part of the posterior gray columns) receives its • Atherosclerosis and atherothrombosis.
blood supply from the posterior spinal arteries which are • Dissection or rupture (701, 702).
paired and considerably smaller than the anterior spinal • Thoracic or abdominal aortic aneurysm.
artery. They receive branches from the posterolateral plexus • Aortic instrumentation (radiologic or surgical).
at various levels. • Intra-aortic balloon pump counterpulsation.
The surface of the spinal cord is richly interwoven with • Arteritis (syphilis, sarcoidosis, tuberculosis, polyarteritis
circumflex anastamoses arising from the anterior and nodosa and other connective tissue disease, drugs:
posterior spinal arteries. A relative hypovascularity exists in cocaine, amphetamine).
the mid thoracic region from about T4–T8, which is the • Thoracoplasty.
most vulnerable part of the spinal cord to ischemia. • Pneumonectomy.
Venous drainage occurs via the median posterior and the • Porto-caval shunt placement.
anterior spinal veins. The lack of venous valves may permit • Radicular artery ligation.
abdominal infectious processes to spread to the spinal cord. • Arteriovenous malformation of the spinal arteries (703).

701 702 703

701, 702 Sagittal section of the spine at autopsy showing vertebral artery thrombosis (702) secondary to 703 Autopsy specimen of a
vertebral artery dissection, which also caused brainstem and anterior upper cervical spinal cord infarction at spinal cord arteriovenous
the craniocervical junction (701). malformation.
560 Spinal Cord Diseases

Venous thrombophlebitis Spinal venous infarction syndrome


• Intra-abdominal and renal sepsis. • Subacute onset.
• Variable patterns of deficits.
Hematologic
• Sickle cell disease. Spinal TIAs
• Procoagulant states: thrombocytosis, metastatic cancer. • Painless paraparesis or quadriparesis without loss of
consciousness.
Spinal cord • Sporadic, or precipitated by postural changes in patients
• Trauma to the spine. with foramenal stenosis during cervical or lumbar
• Acute compression of the spinal cord: extension, which maximally compromises the intervertebral
– Vertebral body and disc disease. foramina through which pass spinal radicular arteries.
– Extradural masses: hematoma, abscess, tumor, fat.
– Subdural masses. DIFFERENTIAL DIAGNOSIS
• Infection within the spine. Acute cord compression
• Intrathecal lignocaine or phenol injection (spinal artery • Intervertebral disc prolapse.
thrombosis). • Vertebral body collapse/fracture (spinal angulation:
tuberculosis, tumor).
CLINICAL FEATURES • Trauma.
Anterior spinal artery syndrome • Epidural hemorrhage.
• Sudden onset.
• Flaccid paraparesis and weakness of myotomes below the Intramedullary
level of the lesion. • Hemorrhage.
• Areflexia below the level of the lesion. • Demyelination (myelitis).
• Babinski responses.
• Loss of pain and temperature sensation below the level INVESTIGATIONS
of the lesion; proprioception intact. CT/MRI spine
• Autonomic dysfunction: To be performed urgently to rule out spinal cord compres-
– Sphincter flaccidity. sion by vertebral fractures or hematomata (CT). When
– Atonic urinary bladder. repeated after several days, MRI is more likely to reveal focal
– Paralytic ileus. cord swelling, and may reveal increased signal in the cord on
• Radicular or back pain may herald these findings T2WI (704, 705), and gadolinium enhancement of the
sometimes. May mimic angina pectoris. lesion on T1WI, a similar appearance to that of transverse
• May progress over a few days. myelitis. Hemorrhage may be identified by the typical
• Later: spasticity, clonus and exaggerated deep tendon changes (see hemorrhagic brain lesions section). If per-
reflexes below the lesion: formed months or years later, MRI shows focal cord atrophy.
– Mixed or lower motor neuron deficits may occur if
infarction involves anterior horn cells (e.g. flaccidity), CSF
nerve roots or cauda equina. • Cells: normal or moderate leukocytosis (<100 mono-
– Detrusor hyperreflexia and striated sphincter dyssynergia. nuclear cells/cm3).
– Sexual dysfunction. • Protein: mild elevation (<1 g/l [100 mg/dl]).
– Orthostatic hypotension (if lesions are superior to the
origin of the greater splanchnic nerve at T4–T9). Other
– Impaired thermoregulation due to impaired vasomotor • Myelography: prone and supine, if arteriovenous
and sudomotor tone below the level of the lesion. malformation is suspected.
– Spinal dysautonomia: a paroxysmal, generalized hyper- • Spinal arteriography: if myelogram suggests AVM.
sympathetic state in which disinhibited sympathetic • Somatosensory evoked potentials: abnormal only if the
neurons of the intermediolateral cell column may mount double perfused posterior columns are infarcted.
an exaggerated response to mildly noxious stimuli such • EMG: abnormal only if infarction involves anterior horn
as a distended bladder. cells or nerve roots.

Sulcocommissural artery syndrome TREATMENT


Ipsilateral paralysis and contralateral loss of pain and • Treat the underlying cause, if possible.
temperature sensation below the level of the lesion. • Symptomatic control of pain, paresthesia, depression and
autonomic and bladder dysfunction.
Posterior spinal artery syndrome • Avoid complications of immobility: regular turning to
• Rare. avoid pressure sores, graduated compression stockings
• Impaired discrimination of direction of scratch on the and aspirin or subcutaneous heparin to prevent deep
skin of the legs. venous thrombosis, and regular bladder emptying.
• Impaired vibration and proprioception cannot be • Rehabilitation: physiotherapy, occupational therapy,
attributed to isolated destruction of the dorsal columns psychologic support, social work.
but rather indicate involvement of additional regions of
the spinal cord such as the dorsal horns.
Acute Transverse Myelitis 561

PROGNOSIS ACUTE TRANSVERSE MYELITIS


• Death: about 20%.
• No improvement: about 25%.
• Minimal improvement: about 10%. DEFINITION
• Moderate improvement: about 25%. An acute loss of sensory, motor and bladder function due
• Marked improvement: about 20%. to inflammation of a transverse (± rostral-caudal) segment
of the spinal cord.
Prognostic factors
• Recovery is impeded by autonomic dysfunction, pain, EPIDEMIOLOGY
paresthesia, depression. • Incidence: 0.1–0.5 per 100 000 population per year.
• Favorable ambulatory outcome correlates with • Age: any age (18 months–82 years), most commonly
improving neurologic examinations in the acute phase young to middle-aged adults.
and relatively preserved strength in hip abductors and • Gender: M=F.
knee extensors.
• Poor prognosis if extensive deficits are present without PATHOLOGY
initial improvement. Ranges from severe necrosis of white and gray matter over many
cord segments to predominant demyelination of white matter.
704
ETIOLOGY AND PATHOPHYSIOLOGY
Infections
• Tuberculosis.
• Syphilis.
• Borrelia (Lyme disease).
• Mycoplasma pneumoniae.
• Enteroviral infection.
• Herpes simplex types 1 and 2.
• Herpes zoster.
• Epstein–Barr virus (EBV).
• Cytomegalovirus (CMV).
• Human herpes virus-6.
• HIV.
• Measles.
• Rubella.

Immune-mediated
• Post infectious: non-specific (e.g. upper respiratory
infection) or specific (e.g. post-mycoplasma or post-
varicella).
• Post vaccination.
• Multiple sclerosis (MS).
• Connective tissue disease (e.g. systemic lupus
erythematosus [SLE]).

705 Granulomatous inflammation


Sarcoidosis.

Radiation
• Early radiation myelopathy may occur 10–16 weeks after
radiation exposure. The symptoms usually resolve
2–9 months later.
• Delayed radiation myelopathy is rare and is also
suggested only when there is a previous history of
radiation. The delay between radiation and acute
myelopathy can exceed 10 years.

N.B. In most cases, no definite cause is found although


one may subsequently declare itself with time (such as MS).

704, 705 T2W sagittal (704) and T2W axial (705) MRI of the PATHOGENESIS
cervical spine in a patient with sudden onset of right-sided neck pain • Immune-mediated.
followed by a lateral medullary syndrome, showing an infarct in the • Active infection of the spinal cord.
upper cervical cord/lower medulla.The right vertebral artery was
partially occluded, thought to be dissected. Note the increased signal in
the upper right side of the cord (arrows).
562 Spinal Cord Diseases

CLINICAL HISTORY Spinal cord infarction (see p.558)


Antecedents Arteriovenous malformation.
• Upper respiratory tract illness in 30–60% of patients.
• Vaccination. Rapidly evolving myeloradiculopathy
• Recent surgery. Lyme disease (see p.316).
• Adenocarcinoma.
Rapidly evolving polyradiculopathy
Onset Guillain–Barré syndrome (segmental form).
• Paresthesiae: usually ascending from the feet.
• Pain: typically interscapular; a Lhermitte’s symptom may INVESTIGATIONS
be present. The aim is to exclude treatable causes of a myelopathy (e.g.
• Leg weakness: usually bilateral. spinal epidural abscess, haematoma or tumor) and identify
• Sphincter disturbance: urinary retention. the underlying cause.

Evolution MRI of the spine


Smooth or stepwise progression to a nadir, usually over • The imaging modality of choice.
several days to weeks, but sometimes rapidly progresses to • The spinal cord may be focally swollen with single or
a maximum within 1 hour (in 15% of cases) and within multiple areas of increased signal within the cord on
24 hours (in 45% of cases). T2W (706, 707) and proton density images, and
hypointense signal on T1W images which may enhance
PHYSICAL EXAMINATION with gadolinium. The vertebral canal and vertebral
Bilateral, fairly symmetric cervical or thoracic bodies are normal.
myelopathy • This appearance is non-specific and occurs in MS, other
• Paraparesis: usually flaccid and of variable severity inflammatory causes and vasculitis.
(pyramidal tracts).
• Deep tendon reflexes: usually depressed or absent. MRI of the brain
• Sensory level: usually thoracic; can be dissociated sensory • Normal, unless associated encephalomyelitis, MS or
loss (e.g. loss of pain and temperature [spinothalamic] vasculitis.
but not proprioception [dorsal columns]) or loss of • Should be considered if these diagnoses (e.g. MS) are
sensation to all modalities (light touch, pain, possible.
temperature, and proprioception). The upper extent of
the lesion is typically several segments above the level of Myelography
the sensory loss. Myelography is useful if MRI cannot be tolerated because the
• Loss of sphincter tone in nearly all patients. patient is claustrophobic or obese, or has a contraindication
• Altered sexual and other autonomic function. to MRI. In the past, MRI did not always reveal angiomas,
• Postural hypotension occurs rarely. and supine myelography followed by spinal angiography was
• Ventilatory compromise (upper cervical myelopathy). the recommended investigation. However, most angiomas
are now detectable using modern MRI technology, with
Clues to the underlying cause phased array receiver coils, high resolution fast spin echo and
• Skin rash: erythema chronicum migrans: Lyme disease; dynamic scanning after a bolus of gadolinium.
vesicles and pustules: herpes virus infections.
• Palatal petechiae: EBV infection. CSF
• Lymphadenopathy and hepatosplenomegaly: EBV, CMV, • Cell count: normal (>50% of cases) or increased (usually
hepatitis, rubella. >30 lymphocytes/ml, but can be polymorphonuclear).
• Myringitis: Mycoplasma pneumoniae infection. • Protein: normal or, more often, raised; if very high and
• Oligoarticular arthritis: Lyme disease. xanthochromia is present, suspect severe cord swelling
with block.
DIFFERENTIAL DIAGNOSIS • Glucose: normal.
Enlarging parasagittal cerebral lesion • Oligoclonal bands: sometimes present.
• Encephalomyelitis. • Acid fast bacilli: not seen.
• Bilateral anterior cerebral artery territory infarction. • Viral and syphilis serology.
• Parasagittal meningioma. • Polymerase chain reaction to amplify DNA from human
• Hydrocephalus. herpes virus 6 and other infectious agents.
• Focal cerebral edema.
N.B. There is no correlation between CSF analysis and
Spinal cord compression the extent of neurologic impairment.
• Extradural abscess (see p.555).
• Extradural hematoma (see p.554). Blood
• Extradural tumor (see p.567). • Full blood count.
• ESR.
Acute ascending necrotizing myelitis • Urea, creatinine, electrolytes, glucose, and liver function
• Lymphoma. tests.
• Carcinoma. • Antinuclear antibody screen (antinuclear antibody, anti-
• Thrombophlebitis. dsDNA, anticardiolipin antibody, rheumatoid factor).
Acute Transverse Myelitis 563

• VDRL, TPHA, FTA. • Acute lupus myelopathy is generally treated with high
• Hepatitis B surface antigen tests. dose methylprednisolone and pulse dose cyclophos-
• Mantoux tests. phamide.
• Serology: herpes simplex, herpes zoster, HIV, • Zoster myelitis: intravenous aciclovir (acyclovir).
mycoplasma, Lyme disease, EBV, CMV, rubella. • Neurosyphilis: penicillin.
• Tibial somatosensory evoked potentials: usually • Tuberculous myelopathy: antituberculous therapy.
unrecordable bilaterally. • HTLV-1 or radiation myelopathy: no satisfactory therapy
• Cortical and spinal motor stimulation: normal motor at present.
response in upper limbs, absent motor response in the
lower limbs. PROGNOSIS
• Visual evoked potentials. Recovery of neurologic function
• Extremely variable: about two-thirds recover to variable
DIAGNOSIS degrees and one-third remain paraplegic.
• Acute or subacute painful spinal cord syndrome (usually • May also be protracted.
thoracic), with disturbance of motor, sensory and
sphincter function. Predictors of a poor outcome
• Spinal segmental level of sensory disturbance with a well • Pain.
defined upper limit. • Catastrophic onset of symptoms.
• No evidence of spinal cord compression. • High ‘deficit score’ at onset.
• Abnormal spinal cord imaging.
TREATMENT • Abnormal somatosensory evoked potentials.
General nursing and comprehensive rehabilitation
programme N.B. CSF findings are not predictors of outcome.
• Bladder care: intermittent catheterization.
• Bowel care: bulk laxatives. Recurrence of transverse myelitis (relapsing
• Elastic stockings. isolated transverse myelitis)
• Physiotherapy. • Rare.
• Analgesia: tricyclic antidepressants, carbamazepine • Distinct from MS and infrequently a harbinger of MS.
(usually ineffective), transcutaneous electric nerve • Causes include: SLE, antiphospholipid antibody syn-
stimulation (TENS), and guanethidine blocks. drome, isolated angiitis of the CNS, HIV, herpes simplex
• Intensive care, including mechanical ventilation, may be infections, and spinal arteriovenous malformations.
required with lesions involving the upper cervical cord.
MS
Specific • Develops in about 1–10% of cases.
• Short course of high-dose steroids: intravenous • Predictors of MS:
methylprednisolone, usually 1 g/day for 3 days or – More subacute onset.
500 mg/day for 5 days (anecdotal reports of benefit, no – Partial or asymmetric lesion.
controlled trials). – Oligoclonal bands in CSF.
• Recurring spinal cord dysfunction in MS manifests most
often as a partial cord syndrome, generally of subacute
706, 707 T2W 706 or chronic onset and with associated evidence of
sagittal (706) and dissemination at onset or in the early follow-up period.
T2W axial (707)
MRI of the cord in
a patient with
transverse myelitis.
Note the increased
signal in the cord at
C3–C6 levels with
swelling. 707
564 Spinal Cord Diseases

HUMAN IMMUNODEFICIENCY VIRUS CLINICAL FEATURES


(HIV) MYELOPATHY • Painless, often slowly progressive, spastic ataxic
paraparesis without a definite sensory level.
• Variable sensory disturbance in the feet and legs
DEFINITION (predominantly of proprioception and vibration sense).
Involvement of the spinal cord by HIV-1, a lentivirus, which A marked sensory ‘level’ is unusual and should suggest
is a family of viruses known to cause chronic neurologic other diagnoses such as a spinal mass.
disease in their animal hosts, as a result of primary infection • Dysesthesiae may occur, but are often due to a
and opportunistic infections in immunosuppressed patients. concomitant polyneuropathy.
• Urinary hesitancy is common but urinary retention and
EPIDEMIOLOGY loss of sphincter control does not occur usually until late
Incidence and prevalence in the illness.
• Unknown. • Often there are features of coexistent HIV
• About one-third of AIDS patients show varying degrees encephalopathy (see p.301).
of vacuolar myelopathy and about 10% of AIDS patients
will show clinical features of a myelopathy. For unknown DIFFERENTIAL DIAGNOSIS
reasons, HIV myelopathy is very uncommon in HIV- • Vitamin B12 deficiency: presents over weeks or months
infected children. as a subacute spinal cord syndrome with intense
• The most common cause of myelopathy in AIDS is paresthesia and a combination of pyramidal and dorsal
vacuolar myelopathy, which is detected in as many as 40% column signs.
of autopsies. • Infection of the spinal cord:
– HTLV-1 associated myelopathy (‘tropical spastic
PATHOLOGY paraparesis’) in adults from endemic regions or with
There are two separate pathologic entities (see below), but other risk factors (e.g. Afro-Caribbeans). Lifetime
in some cases multinucleated cell myelitis with evidence of prevalence: 0.04 (95% CI: 0–0.2) per 1000. A
productive local HIV infection may coexist with the progressive spastic paraplegia develops over a number of
vacuolar changes. years; however, sphincter disturbance is the rule and
considerable neuropathic lower limb pain is common.
Vacuolar myelopathy – Toxoplasmosis.
• Multiple vacuoles are present in the myelin sheaths of the – Varicella-zoster granulomatous myelitis.
white matter of the posterior and lateral columns of the – Herpetic necrotizing myelitis.
spinal cord, particularly the cervical and high thoracic – CMV.
cord. Vacuolization of myelin sheaths is due to the – Tuberculosis.
accumulation of foamy macrophages and microglia, with – Schistosomiasis.
relative preservation of axons. – Syphilis.
• Infrequent lipid-laden macrophages. • MS and other demyelinating disease of the spinal cord.
• Separation of the myelin lamellae seen on electron micro- • Sarcoidosis.
scopy; the changes are not confined to a particular tract. • Spondylotic myelopathy.
• Lymphoma or other neoplastic mass lesions of the spine
Multinucleated giant cell myelitis (e.g. astrocytoma, ependymoma, lipoma, hemangioma,
Multinucleated giant cell infiltrates that do not seem to have metastases).
a predilection for any particular part of the cord. • Dural arteriovenous malformation: the most common
type of spinal angioma. Usually affects the thoracolumbar
PATHOGENESIS segments and tends to present in middle-aged men as a
Vacuolar myelopathy chronic progressive myelopathy with symptoms that may
There is no evidence that the pathogenesis of vacuolar fluctuate or be aggravated by exercise. A combination of
myelopathy is related to a direct effect of HIV infection as upper and lower motor neuron signs may be present.
the disorder has been described in immunocompromised • Hereditary spastic paraparesis: usually a family history of
non-AIDS patients. This suggests that it is a consequence autosomal dominant inheritance, but may be autosomal
of an as yet undefined opportunistic infection. The myelin recessive.
damage may be the result of indirect mechanisms such as • Adrenomyeloneuropathy: X-linked inherited disorder
the release of toxic cytokines, particularly tumor necrosis that affects males and some heterozygous females with a
factor, or another metabolic or toxic insult. progressive myelopathy. A peripheral neuropathy and
adrenal insufficiency may be present.
Multinucleated giant cell myelitis • Radiation myelopathy: steadily progressive spastic para-
Related at least in part to the direct effects of the infiltrating plegia months to years after radiotherapy. Pathologically
cells as immunohistochemical studies have demonstrated there is necrosis of the irradiated cord segments with
various HIV antigens. The precise mechanisms of spinal obliterative changes in blood vessels in the same region.
cord damage are likely to be similar to those that operate in • Lathyrism: endemic in parts of India and presents as a
HIV encephalopathy (see p.301). subacute or chronic spastic paraparesis in people who
regularly ingest chickling pea vetch over several months.
It is thought to be caused by a toxin in the chickling pea.
Radiation-induced Encephalo-myelo-radiculopathy 565

INVESTIGATIONS RADIATION-INDUCED ENCEPHALO-


• MRI of the spine or myelogram appears normal or shows MYELO-RADICULOPATHY
mild cord atrophy only but helps to exclude other
possible causes.
• Full blood count and film (?vitamin B12 deficiency). DEFINITION
• Serum vitamin B12. An iatrogenic disease of the nervous system arising as a
• Serology for HIV, herpes virus. complication of radiation therapy.
• Blood CD4 lymphocyte count.
• CSF: non-specific changes of elevated WBC, elevated EPIDEMIOLOGY
protein and IgG. There is no correlation between levels • Incidence: decreasing.
of HIV-1 RNA viral load in the CSF and the presence or • Age: any age.
severity of AIDS myelopathy. HIV can be cultured from • Gender: either sex.
CSF.
PATHOLOGY
DIAGNOSIS Mild-moderate disease
• Negative MRI examination of the spine and brain. Spongy appearance, demyelination, depletion of
• Positive HIV antibody test. oligodendrocytes and partial preservation of axons.
• Blood CD4 lymphocyte (T helper cell) count <200 ×
106/l (normal 800–1200 × 106/l). Severe disease
• Culture of HIV in CSF may be positive. • Coagulation necrosis, typical of infarction, of focal or
• HIV may be demonstrable by immunochemistry in multifocal regions of the white matter and, less so, gray
spinal cord tissue at autopsy. matter of the brain or spinal cord.
• Softening and liquefaction, with cyst formation, may
TREATMENT occur in some areas.
• No clinical trials of therapy have been performed in this • Arterioles: hyaline thickening and fibrinoid necrosis of
disorder. the vessel wall, with thrombosis.
• Anecdotal data suggest that vacuolar myelopathy does • Secondary degeneration of the ascending and descending
not respond to antiretroviral therapy (see p.301) but tracts.
multinucleated cell myelitis may.
• Other treatment is symptomatic and supportive, and ETIOLOGY
includes antispasticity agents (e.g. baclofen), manage- Radiation
ment of sphincter dysfunction, physiotherapy, occupa- • Tissue-dependent: the brain and spinal cord appear more
tional therapy and provision and education in the use of sensitive than the peripheral nerves to irradiation
walking aids. damage.
• Counselling is frequently required for patient and carers • Dose- (total and fractional) dependent: a daily dose
early in the illness. >2 Gy (>200 rads), weekly dose >9 Gy (>900 rads), total
• Medical power-of-attorney should be recommended to dose >60 Gy (>6000 rads) (40 Gy in 25 fractions for
patients early in the illness. spinal cord) is associated with an increased risk of
• Regular respite care is beneficial to patient and carers radiation injury.
during the illness and palliative care facility is usually • Time-dependent: a total given over a period of
required to manage late complications of dementia. <30–70 days is associated with an increased risk of
radiation injury.
CLINICAL COURSE/PROGNOSIS • Risk factors: hyperthermia treatment for cancer
HIV myelopathy has a variable course: predisposes to radiation myelopathy.
• Patients may remain ambulatory with only modest leg
weakness. PATHOPHYSIOLOGY
• Occasionally a more fulminant course occurs, with • Irreversible radiation damage to small intracranial blood
progression to paraplegia over a few months. vessels.
• High plasma HIV RNA values and rapidly declining • The relative contribution of irradiation and concurrent
CD4 lymphocyte number and percentage predict chemotherapy can be difficult to determine in some
progression to AIDS and death. patients.

CLINICAL SYNDROMES AND FEATURES


Multifocal and diffuse encephalopathy
• Subacute onset.
• Focal or multifocal neurologic dysfunction.
• Partial or generalized seizures.
• Impaired mental function: progressive dementia (which
may be accompanied by gait ataxia and sphincter and
motor dysfunction).
• Pituitary dysfunction.
• Raised intracranial pressure eventually.
• History of whole-brain irradiation (e.g. for tumor or
acute lymphoblastic leukemia).
566 Spinal Cord Diseases

Multiple cranial neuropathies MRI


Usually follows radiation therapy of nasopharyngeal tumors. • Previous radiotherapy involving a section of the spine can
be recognized on T1WI as diffuse increased signal in the
Mild ‘early’ transient sensory myelopathy bone marrow of several adjacent vertebral bodies
• Occurs several (3–6) months after irradiation. (decreased on conventional T2WI).
• Paresthesiae in the limbs, evoked or exacerbated by neck • The spinal cord may also be atrophic at that level.
flexion (Lhermitte symptom/sign). • Increased signal in the cord on T2WI may be due to
• Resolves spontaneously in most cases after a few months. radiation myelitis (708) or residual tumor (if that was the
original reason for the therapy).
Acute myelopathy
Developing over hours to days, due to spinal cord CSF
infarction. Normal except for slightly elevated protein.

Delayed chronic progressive myelopathy DIAGNOSIS


• Incidence: about 2–3% of treated patients (but difficult Histologic.
to estimate because many patients die of their malignant
disease before the cord lesion matures). TREATMENT
• Latent period: 14 months post-irradiation on average; Symptomatic
usual range 4 months–19 years. • Corticosteroids (dexamethasone) may reduce the edema
• Onset: insidious, but exceptionally it may be subacute, surrounding the lesion in some patients, with concurrent
within a few hours. regression of symptoms, but the effects are often
• Sensory symptoms: paresthesiae and dysesthesiae of one temporary.
or both feet (and hands, if cervical cord involved) or a • Surgical debulking may also be temporarily effective but
Lhermitte symptom /sign. results are indifferent.
• Motor symptoms, such as weakness of one or both legs, • Antithrombotic therapy (e.g. aspirin, warfarin) remains
usually follow the sensory loss, and may complete a to be evaluated.
Brown–Sequard syndrome or transverse myelopathy.
• Autonomic dysfunction may occur. Prevention
• Local pain is notably absent (in contrast to spinal Radiation injury to the nervous system is preventable if the
metastases). total and fractional dose of radiation is delivered according
to established guidelines.
Motor lumbosacral radiculopathy (lower motor
neuron syndrome) PROGNOSIS
Radiation-induced encephalo-myelo-radiculopathy usually
Sensorimotor lumbosacral plexopathy has a progressive clinical course, but periods of apparent
Usually following irradiation for pelvic neoplasia. arrest for several years may occur.

DIFFERENTIAL DIAGNOSIS
Diffuse brain injury (radiation encephalopathy) 708 708 T2W sagittal
• Normal pressure hydrocephalus. MRI of the whole
• Leukodystrophy. cord showing an
• Alzheimer’s disease. area of increased
signal in the upper
Focal brain injury (radiation encephalopathy) thoracic cord
• Brain tumor. (arrow).The patient
• Stroke. had received
radiation for
Mild ‘early’ transient sensory myelopathy lymphoma and then
• Cervical spondylotic myelopathy. developed long tract
• MS. signs.The increased
signal is the result of
Delayed chronic progressive myelopathy radiation myelitis.
• Spinal tumor recurrence or metastases: usually local pain The bone marrow
and evidence of a mass lesion. changes of radiation
• MS. are not visible on
this T2W image.
INVESTIGATIONS
CT brain scan
Low density, contrast-enhancing lesion(s).
Spinal Cord Tumors 567

SPINAL CORD TUMORS Primary source:


• Breast (often multiple extradural deposits).
• Bronchus (usually single spinal lesion).
DEFINITION • Prostate.
Tumors of the spinal cord. • Melanoma.
• Multiple myeloma.
EPIDEMIOLOGY • Lymphoma.
• Incidence: 10 (95% CI: 5–18) per 100 000 per year. • Renal cell carcinoma.
Primary spinal cord tumors represent about 15% of • Gastrointestinal carcinoma.
primary CNS tumors. Metastases to the spine are far
more common. Intradural but extramedullary (rare)
• Age: Leptomeningeal metastases of carcinoma or lymphoma
– Young and middle-aged adults predominantly. causing malignant meningitis.
– Intramedullary tumors are more common in children.
– Extramedullary tumors are more common in adults. Intramedullary (very rare)
• Gender: M=F. Carcinoma of the bronchus is the main source.
Metastases tend to arise in the thoracic region and
PATHOLOGY usually spread from the vertebral body into the anterior and
Primary tumors of the spine anterolateral parts of the dural space. They are most unusual
Extradural: common (45–55%) in the cervical spine except when due to local spread from
• Neuroblastoma. adjacent regions such as nasopharyngeal carcinoma.
• Lymphoma and leukemia.
• Myeloma/plasmacytoma. Associations
• Primary bone tumors. Syringomyelia is frequently associated with intramedullary
• Hemangioma of bone. tumors.
• Sarcoma.
CLINICAL FEATURES
Intradural but extramedullary (outside the spinal cord The spinal cord and its nerve roots are contained within the
but beneath the dura, i.e. in the leptomeninges or roots): rigid vertebral canal and are therefore vulnerable to
common (40–50%) compression, particularly in those individuals with
• Meningioma (anywhere along the cord). constitutionally narrow vertebral canals. The clinical features
• Neurofibroma (commonly thoracic). depend on the site and extent to the spinal cord lesion as
• Sarcoma (F>M). well as the rate of growth.
• Vascular tumors. Compression of the spinal cord and nerve roots at a
• Chordoma (commonly sacral region). given level may give rise to:
• Epidermoid. • Segmental pain or sensory disturbance at the level of the
• Dermoid (often sacral region). lesion due to dorsal root irritation. The pain is
• Malignant meningitis. exacerbated by sneezing, coughing, and straining.
Cervical cord compression may cause Lhermitte’s
Intramedullary (inside the spinal cord): uncommon (5%) symptom/sign of a sudden, unpleasant, electric shock-
• Ependymoma (many arise from filum terminale): 60%. like sensation down the back and into the legs, and
• Astrocytoma (mostly cervical): 25%. occasionally the arms, after flexing the neck. It is due to
• Oligodendroglioma. dysfunction of the cervical spinal cord from a number of
• Hemangioblastoma: may cause hematomyelia. causes (e.g. compression, MS, subacute combined
• Lipoma. degeneration of the cord, acute radiation myelopathy)
• Epidermoid. and may also occur in some sensory neuropathies
• Dermoid. (cisplatin, pyridoxine abuse).
• Teratoma. • A segmental lower motor neuron lesion at the level of
compression.
Secondary tumors (metastases) of the spine • Upper motor neuron signs, including bladder
Most spinal tumors are metastases, rather than primary dysfunction, below the level of the lesion. The upper
benign or malignant tumors. motor neuron deficit is hardly ever unilateral but may be
asymmetric.
Extradural (most common site of metastases) • Sensory loss to some or all modalities (usually joint
• 85% are bony metastases in the vertebral column which position sense) below the level of the lesion with a
involve the spinal canal directly or via the intervertebral distinct upper level; the sensory symptoms and signs tend
foramina. to begin in the feet and ascend to just below the
• The minority arise from hematogenous dissemination to anatomic level of the lesion. Sensory loss may be mild or
the extradural space. even absent depending on whether and which ascending
spinal cord pathways are involved.
• Autonomic changes below the level of the lesion, and
even an ipsilateral Horner’s syndrome with severe cervical
cord compression.
568 Spinal Cord Diseases

Three typical clinical syndromes Subacute onset


Myelopathy (sensori-motor spinal tract syndrome) • Epidural abscess (e.g. pyogenic, tuberculous and fungal
• Gradual onset. granulomatous lesions).
• Progressive course. • Paraneoplastic necrotizing myelopathy.
• Symmetric or, more commonly asymmetric, motor
and/or sensory deficits at and below the level of the Slow onset
lesion, together with bladder and bowel dysfunction. • Spondylosis.
• Syringomyelia.
Myelo-radiculopathy • Tethered cord syndrome (see Spinal dysraphism, p.139).
• Segmental pain and sensory change in the distribution of • Radiation myelopathy.
one or more sensory nerves. • Meningeal carcinomatosis.
• Segmental cramps, fasciculations, wasting, weakness,
reflex loss in the distribution of one or more motor INVESTIGATIONS
nerves. • Full blood count and ESR.
• Spinal pain and tenderness. • Liver function tests: secondary deposits.
• Associated myelopathy: asymmetric spastic para- or • Syphilis serology.
quadriparesis, sensory level below which pain and • Urine: hematuria with renal cell carcinoma.
temperature perception and joint position sense is
reduced, and detrusor hyperreflexia. Chest x-ray
• Infection with tuberculosis.
Central cord (syringomyelic) syndrome • Tumor: primary or secondary.
• Segmental dissociated sensory loss and amyotrophy. • Paravertebral shadow (neurofibromatosis, Pott’s disease).
• Early incontinence.
• Later corticospinal weakness. Spine x-ray
• Vertebral body collapse (osteoporosis, infection, tumor).
Extramedullary tumors • Erosion of vertebral bodies or pedicles by tumor.
• Tumors usually involve a few segments with root com- • Scalloping of posterior margin of vertebral body (chronic
pression (usually dorsal) and progressive compression of tumor).
the cord to cause an incomplete or complete transection • Vertebral body alignment: subluxation, spondylolisthesis.
syndrome. • Intervertebral disc space narrowing (spondylosis) or
• Pain and local tenderness is the initial symptom of destruction.
extradural metastases in about 95% of adults. It is a good • Intervertebral exit foramina (oblique views of neck)
localizing sign of the level of the lesion. Spinal pain at enlarged in neurofibroma, reduced in spondylosis.
night is highly suggestive of a tumor. • Spinal canal width, osteophytes.
• Muscular weakness, wasting and sensory loss is present
in the distribution of the affected roots. Imaging
• There is a variable pattern of spastic paraparesis and If the patient has established primary malignant disease,
sensory loss below the lesion, as well as bladder (less unequivocal clinical features of progressive spinal cord
commonly bowel) dysfunction, if the cord is compressed. and/or root compression, and plain x-ray changes of a
• Vascular spinal cord syndromes may occur, and are metastatic deposit in the appropriate site to cause the
suggestive of metastases (and abscesses) because they neurologic lesion, there may be nothing to gain from
tend to occlude spinal vessels. further investigation (with MRI or myelography),
particularly in the presence of multiple metastases and a
Intramedullary tumors poor prognosis. The aim of management is usually palliative.
Tumors may extend over multiple levels and give rise to a If there is no known primary malignancy, then the spine
clinical picture similar to syringomyelia (see p.541). Asym- must be imaged with MRI or myelography and, if a
metric location within the cord results in asymmetric clinical compressive mass is found that has the features of a tumor
signs. (i.e. not those of a disc or spondylotic bar), then a tissue
diagnosis may be possible by CT-guided percutaneous
Other features suggestive of the cause biopsy or surgery.
• Skin lesions, e.g. café-au-lait spots of neurofibromatosis;
pigmented melanoma. CT scan
• Breast mass or other evidence of primary cancer. CT scan of the vertebral canal with contrast enhancement
• Weight loss, malaise: metastases. may demonstrate tumors and delineate the extraspinal
extent of tumors.
DIFFERENTIAL DIAGNOSIS
Acute onset Myelography followed by CT scan (709–711)
• Intervertebral disc prolapse. Useful if MRI (see below) is not available. Widening of the
• Vertebral body collapse (spinal angulation: tuberculosis, spinal cord in the antero-posterior (AP) view may be due to
tumor). anterior or posterior cord compression but widening in the AP
• Trauma. and lateral view suggests cord tumor, myelitis, or a syrinx.
• Epidural hemorrhage.
• Intramedullary hemorrhage: arteriovenous malformation.
• Spinal cord infarction.
Spinal Cord Tumors 569

CSF MRI spine


Lumbar puncture may precipitate acute deterioration if The imaging method of choice as it displays bones and soft
there is cord compression and may damage the spinal cord tissues impinging on spinal cord and roots better than
if it is tethered to the low lumbar or sacral vertebral bodies. myelography and CT, and it images inside the spinal cord
CSF cytology may reveal malignant cells. Froin syndrome: and can detect intramedullary tumors (712, 713) and
xanthochromia with clotting of CSF, due to isolation or exclude a syrinx, which myelography cannot do. However,
loculation of the CSF below the site of the tumor which has it may need to be backed up by plain x-rays or CT to
blocked the circulation of CSF from above. examine bone detail and if a percutaneous imaging-guided
biopsy is to be planned.

709, 710 Lateral 709


(709) and antero-
posterior (710)
views from a
myelogram in a
patient with
symptoms of cord
compression. Note 710
the dura is
compressed inwards
towards the cord in a
pattern typical of
extradural lesions.
This was a secondary
deposit. Note the
absent pedicle of the
vertebral body
(arrow) indicating
bony involvement.

711 712 713

711 Myelogram of the lumbar region from a patient with breast carcinoma and symptoms of multiple nerve root compression, showing cut-off of
the lumbar nerve roots, and an irregular outline of the dura due to multiple meningeal secondary breast cancer nodules.

712, 713 MRI cervical spine, sagittal (712) and axial (713) sections showing a hyperdense mass (arrows) in the left hemicord (causing a
Brown–Séquard syndrome) due to metastatic malignant melanoma.
570 Spinal Cord Diseases

Extradural lesions If a secondary deposit is found without a known primary


Lesions arise outside the dura and compress the cord from the source, then further investigation is indicated because it
outside, either circumferentially or mainly anteriorly or helps general and specific management strategies:
posteriorly depending on the site (714). They are most • Breast and pelvic examination.
commonly secondary deposits from bronchus, breast, prostate, • Serum liver function tests.
renal, and testicular carcinomas, other adenocarcinomas or • Acid phosphatase and protein electrophoresis.
lymphomas. Bone involvement varies as the tumor can simply • Mammography.
sit as a cuff in the spinal canal with minimal bone involvement, • Bone scan (skeletal survey is more sensitive in myeloma).
though more commonly bone involvement is substantial. On • Fecal occult bloods and, if positive, colonoscopy or
imaging, the cord appears normal width or narrowed, with the barium enema.
CSF obliterated around it at the level of the lesion, but visible • Urine occult blood and, if positive, ultrasound imaging
above and below, and a mass around the outside plus or minus of the renal tract.
bone destruction. MRI is best as it demonstrates not only the • Ultrasound of the liver.
extent of the symptomatic lesion, but also the presence of
other asymptomatic deposits at other levels. Myelography (± DIAGNOSIS
CT) may only show up to the lower level of the lesion, as if MRI spine and tissue biopsy in a patient with appropriate
the block is complete, the contrast will not pass it to outline clinical features.
the cord above.
TREATMENT
Intradural extramedullary lesions (meningiomas, • Rapidly progressive spinal cord compression by
schwannomas, neurofibromas, lipomas, arachnoid cysts) extradural tumor calls for emergency MRI, immediate
Also best seen on MRI (715, 716). They appear as a mass high dose corticosteroids, and a tissue diagnosis either
within the CSF space displacing (and compressing) the cord by CT-guided percutaneous biopsy or open surgical
to one side. Meningiomas are isointense on T1WI, enhance biopsy.
and are commonest in the thoracic region in females. Nerve • If the tumor is benign it should be removed surgically if
sheath tumors may contain mixed signal elements and lie technically possible.
within the nerve root exit canal as well as the spinal canal. • If the tumor is malignant, the spinal cord and roots
There are often other stigmata of neurofibromatosis. They should be decompressed by surgery, radiotherapy,
usually enhance. Lipomas have characteristic signal and chemotherapy or hormonal therapy, depending on
usually lie in the lumbar region. Arachnoid cysts can be very tumor type and the clinical circumstances; surgical
difficult to demonstrate and may be better seen on decompression is probably no more effective and should
myelography including delayed films (with CT if possible) be avoided except perhaps in acute cases of malignant
to watch for delayed filling of the cyst. The precise site of tumor compression.
the cyst may be difficult to demonstrate on imaging in terms
of its relationship to the dural sac.
714 714 T1W midline sagittal
Intramedullary tumors MRI showing metastatic
Tumors cause expansion of the spinal cord which may fill extradural lesions in the
and obliterate the CSF space if pronounced. Primary upper thoracic and upper
(astrocytoma, ependymoma, hemangioblastoma) or lumbar regions (arrows).
secondary tumors (metastases from breast carcinoma) are Note the involvement of the
the main neoplastic causes of cord expansion (717, 718, vertebral bodies in the
719). Non-neoplastic causes include MS, ADEM, AIDS, malignant process as shown
sarcoid, SLE, syrinx, hemorrhage, infarct, AVM. MRI is the by the altered marrow signal.
imaging modality of choice as it demonstrates the full extent
of the lesion, and the signal characteristics give some clues
as to the composition and likely type of lesion. Astrocytomas
mainly affect the thoracic and cervical cord, usually are large,
diffuse and involve the whole thickness of the cord, are
often cystic and often enhance. Ependymomas are usually
more focal, well circumscribed, enhancing, with evidence of
cyst formation and hemorrhage. They tend to occur around
the conus and filum terminale. Hemangioblastomas have
cystic and solid components (the latter enhancing
markedly), may appear eccentric, have hemorrhagic
components, and sometimes flow voids can be seen due to
the intense capillary network and sinus channels.
Angiography shows a capillary blush and draining vein.
Metastases appear as lumps which enhance within the cord.

Isotope bone scanning


Also widely used to look for bone involvement by malignant
secondary (and some primary tumors) but is rather limited
and will not be discussed further here.
Spinal Cord Tumors 571

• If a secondary deposit is found without a known primary PROGNOSIS


source, then further investigation is indicated (see above). • Prognosis depends on the underlying cause and duration
• Operative fixation of the vertebral column may be and degree of spinal cord and nerve root compres-
worthwhile in patients with unremitting pain due to sion/infiltration.
spinal instability. • In contrast to brain tumors, many spinal tumors are
benign and produce their effects mainly by compression
of the spinal cord rather than by invasion.

715 716

715, 716 T1W sagittal (715) and T2W axial (716) MRI of the
cervical spine showing a meningioma at C2 level (arrows). Note how
the lesion is surrounded by CSF and pushes the dura away from the
cord in a pattern typical of an intradural extramedullary lesion.

717 718 719

717 T2W sagittal MRI of the lumbar spine showing a lobulated mass (arrow) in the lower lumbar canal within the dura.This was an ependymoma.

718, 719 T2W (718) and T1W (719) sagittal MRI of a cord astrocytoma. Note how the whole cord appears expanded and the signal within it is
diffusely abnormal.There appear to be cystic and solid elements.
572 Spinal Cord Diseases

Treatment ACUTE TRANSVERSE MYELITIS


FURTHER READING Miller RG, Rosenberg JA, Gelinas DF, et al. Al Deeb SM, Yaqub BA, Bruyn GW, Biary NM
(1999) Practice parameter: The care of the pa- (1997) Acute transverse myelitis. Brain, 120:
tient with amyotrophic lateral sclerosis (and 1115–1122.
HEREDITARY SPASTIC PARAPARESIS evidence-based review). Report of the Quali- De Seze J, Stojkovic T, Breteau G, et al. (2001)
Fink JK, Heiman-Patterson T, for the Hereditary ty Standards Subcommittee of the American Acute myelopathies. Clinical, laboratory and
Spastic Paraplegia Working Group (1996) Academy of Neurology. Neurology, 52: outcome profiles in 79 cases. Brain, 124:
Hereditary spastic paraplegia: advances in ge- 1311–1323. 1509–1521.
netic research. Neurology, 46: 1507–1514. Quality Standards Subcommitte of the American
Academy of Neurology (1997) Practice advi- HUMAN IMMUNODEFICIENCY VIRUS
Furukawa Y, Graf WD, Wong H, et al. (2001)
sory on the treatment of amyotrophic lateral MYELOPATHY
Dopa-responsive dystonia simulating spastic
sclerosis with riluzole: Report of the Quality Geraci A, Di Rocco A, Liu M, et al. (2000) AIDS
paraplegia due to tyrosine hydroxylase (TH)
Standards Subcommittee of the American myelopathy is not associated with elevated
gene mutations. Neurology, 56: 260–263.
Academy of Neurology. Neurology, 49: HIV viral load in cerebrospinal fluid. Neurol-
McDermott CJ, White K, Bushby K, et al. (2000)
657–659. ogy, 55: 440–442.
Hereditary spastic paraparesis: a review of new
Swash M (2000) Pharmacoeconomics and motor Lehky TJ, Flerlage N, Katz D, et al. (1996)
developments. J. Neurol. Neurosurg. Psychia-
neuron disease. J. Neurol. Neurosurg. Psychia- Human T-cell lymphotropic virus type II-as-
try, 69: 150–160.
try, 68: 410–414. sociated myelopathy: clinical and immunolog-
McDermott CJ, Dayaratne RK, Tomkins J, et al.
ic profiles. Ann. Neurol., 40: 714–723.
(2001) Paraplegin gene analysis in hereditary
SYRINGOMYELIA Machigashira N, Yoshida Y, Wang S, Osame M
spastic paraparesis (HSP) pedigrees in north-
Heiss JD, Patronas N, DeVroom HL, et al. (2001) HTLV-1-associated myelopathy/trop-
east England. Neurology, 56: 467–471.
(1999) Elucidating the pathophysiology of sy- ical SPASDC paraparesis with pseudohypo-
Reid E, Dearlove AM, Whiteford ML, Rhodes M,
ringomyelia. J. Neurosurg., 91: 553–562. parathyroidism. Neurology, 56: 104–106.
Rubinsztein DC (1999) Autosomal dominant
Small JA, Sheridan PH (1996) Research priorities Manns A, Hisada M, La Grenade L (1999)
spastic paraplegia. Refined SPG8 locus and ad-
for syringomyelia: A National Institute of Human T-lymphotropic virus type I infection.
ditional genetic heterogeneity. Neurology, 53:
Neurological Disorders and Stroke workshop Lancet, 353: 1951–1958.
1844–1849.
summary. Neurology, 46: 577–582.
Tallaksen CME, Dürr A, Brice A (2001) Recent SPINAL CORD TUMORS
advances in hereditary spastic paraplegia. Curr. CERVICAL SPONDYLOTIC MYELOPATHY Byrne TE (1992) Spinal cord compression from
Opin. Neurol., 14: 457–463. AND RADICULOPATHY epidural metastases. N. Engl. J. Med., 327:
White KD, Ince PG, Lusher M, et al. (2000) Casey ATH, Crockard HA, Bland JM, et al. 614–619.
Clinical and pathologic findings in hereditary (1996) Surgery of the rheumatoid cervical Tattersall R, Turner B (2000) Brown–Séquard and
spastic paraparesis with spastin mutation. Neu- spine for the non-ambulant myelopathic pa- his syndrome. Lancet, 356: 61–63.
rology, 55: 89–94. tient – too much, too late? Lancet, 347:
1004–1007.
SPINAL MUSCULAR ATROPHY
Fairbank J (1998) Trials and tribulations in cervi-
59th ENMC International Workshop: Spinal Mus-
cal spondylosis. Lancet, 352: 1165–1166.
cular Atrophies: recent progress and revised di-
Fowler CJ (1999) Neurological disorders of mic-
agnostic criteria 17–19 April 1998, Soest-
turition and their treatment. Brain, 122:
duinen, The Netherlands (1999) Neuromus-
1213–1231.
cular Disorders, 9: 272–278.
Levine DN (1997) Pathogenesis of cervical
MOTOR NEURON DISEASE spondylotic myelopathy. J. Neurol. Neurosurg.
General Psychiatry, 62: 334–340.
Rowland LP, Schneider NA (2001) Amyotrophic Rowland LP (1992) Surgical treatment of cervical
lateral sclerosis. N. Engl. J. Med., 344: spondylotic myelopathy: Time for a controlled
1688–1700. trial. Neurology, 42: 5–13.

Epidemiology ACUTE SLIPPED DISC


Piemonte and Valle D’Aosta Register for Amy- Deen HG Jr (1996) Diagnosis and management
otrophic Lateral Sclerosis (PARALS) (2001) of lumbar disc disease. Mayo Clin. Proc., 71:
Incidence of ALS in Italy. Evidence for a uni- 283–287.
form frequency in Western countries. Neurol- Gibson JN, Grant IC, Waddell G (1999) The
ogy, 56: 239–244. Cochrane review of surgery for lumbar disc
Traynor BJ, Codd MB, Corr B, et al. (1999) In- prolapse and degenerative lumbar spondylosis.
cidence and prevalence of ALS in Ireland, Spine, 24: 1820–1832.
1995–1997. A population-based study. Neu- Vinas FC, Wilner H, Rengachary S (2001) The
rology, 52: 504–509. spontaneous resorption of herniated cervical
discs. J. Clin. Neuroscience, 8: 542–546.
Etiology
Shaw PJ (1999) Motor neurone disease. BMJ, SPINAL EPIDURAL HEMATOMA
318: 1118–1121. Samkoff LM, Daras M, Kleiman AR, Koppel BS
(1996) Spontaneous spinal epidural
Clinical haematoma. Another neurological complica-
Katz JS, Wolfe GI, Andersson PB, et al. (1999) tion of cocaine? Arch. Neurol., 53: 819–821.
Brachial amyotrophic diplegia. A slowly pro-
gressive motor neuron disorder. Neurology, SPINAL CORD INFARCTION
53: 1071–1076. Cheshire WP (2000) Spinal cord infarction mim-
Kuipers-Upmeijer J, De Jager AEJ, Hew JM, et al. icking angina pectoris. Mayo Clin. Proc., 75:
(2001) Primary lateral sclerosis: clinical, neu- 1197–1199.
rophysiological, and magnetic resonance find- Cheshire WP, Santos CC, Massey EW, Howard JF
ings. J. Neurol. Neurosurg. Psychiatry, 71: Jr (1996) Spinal cord infarction: aetiology and
615–620. outcome. Neurology, 47: 321–330.
Polkey MI, Lyall RA, Moxham J, Leigh PN Iseli E, Cavigelli A, Dietz V, et al. (1999) Prog-
(1999) Respiratory aspects of neurological dis- nosis and recovery in ischaemic and traumatic
ease. J. Neurol. Neurosurg. Psychiatry, 66: spinal cord injury; clinical and electrophysio-
5–15. logical evaluation. J. Neurol. Neurosurg. Psy-
chiatry, 67: 567–571.
Diagnosis Nacimiento W, Noth J (1999) What, if anything,
Belsh JM (1999) Diagnostic challenges in ALS. is spinal shock? Arch. Neurol., 56:
Neurology, 53 (Suppl 5): S26–S30. 1033–1035.
Satran R (1988) Spinal cord infarction. Stroke, 19:
529–532.
Chapter Twenty 573

Autonomic Nervous
System Disorders
AUTONOMIC NEUROPATHY thoracic root (T1) and course through the ventral roots and
white rami to synapse in the superior cervical ganglion.
Postganglionic fibers travel from the superior cervical
DEFINITION ganglion along the common carotid artery to its bifurcation.
Autonomic neuropathy is a disorder of the autonomic The pupillodilator fibers and fibers responsible for sweating
nervous system. on the medial aspect of the forehead then follow the course
of the internal carotid artery to the carotid siphon. At the
ANATOMY petrous temporal bone and cavernous sinus, they join the
The autonomic nervous system consists of two major ophthalmic (first) division of the trigeminal nerve and travel
divisions: sympathetic (thoracolumbar division) and to the pupil. Interruption of these fibers causes ipsilateral
parasympathetic (craniosacral outflow). ptosis and miosis.
Preganglionic fibers responsible for sweating and
Sympathetic nervous system (720) vasomotor control in the face accompany the second and
Efferent third thoracic nerve roots (T2,T3) and synapse in the
Sympathetic nerve fibers descend from the hypothalamus superior cervical ganglion. Postganglionic fibers travel along
and other regions of the brain, through the lateral the external carotid artery in the periarterial plexus before
tegmentum of the midbrain, pons and medulla, to the passing along the peripheral branches of the trigeminal
intermediolateral cell columns of the spinal cord between nerve to the skin.
T1 and L2, where they synapse with neurons of the Preganglionic sympathetic fibers to the bladder travel
preganglionic sympathetic efferents. The nerve fibers exit from the intermediolateral columns in the T11-L2 segments
from the spinal cord through the anterior roots, join the of the spinal cord to the ventral roots, white rami and
proximal part of the nerve root, and then leave the root via hypogastric plexus. Postganglionic fibers join the subserosal
the white rami communicantes to join the sympathetic chain plexus on the bladder wall and innervate mainly the trigone
which consists of a series of ganglia and nerve fibers and neck of the bladder.
extending from the base of the skull to
the coccyx. 720 Intermediolateral column of spinal cord
Preganglionic sympathetic fibers,
which are myelinated and cholinergic,
Posterior root
enter the sympathetic chain through
the white rami, may synapse in the
nearest ganglion, pass up or down the
sympathetic chain before making their
synapse, or pass through the chain to
White ramus
synapse at more peripheral ganglia
such as the celiac or other mesenteric Anterior root Gray ramus
ganglia.
Blood vessels
Postganglionic sympathetic fibers, Sympathetic
which are unmyelinated and arise from chain
Adrenal medulla
the ganglia of the sympathetic chain,
join the main nerve trunks by way of Sweat gland
the gray rami communicantes and are Some muscle vessels
distributed to the sweat glands
(cholinergic) and blood vessels of the Gut
Celiac
skin (adrenergic vasoconstrictor) and ganglion
muscles (cholinergic vasodilator). The
peptides released may play a role in the 720 Schematic diagram of the sympathetic nervous system showing the thoracic spinal cord
control of vasomotor tone and the and nerve roots at one level in cross-section, preganglionic myelinated cholinergic fibers (solid
regulation of regional blood flow. blue line), postganglionic unmyelinated noradrenergic fibers (dashed blue line) and postganglionic
Preganglionic sympathetic fibers to unmyelinated cholinergic fibers (dashed red line). (Modified from McLeod JG, Lance JW, Davies L
the eye leave the spinal cord at the first (1995). Introductory Neurology, 3rd edn, Blackwell Science, Carlton, Austrialia.)
574 Autonomic Nervous System Disorders

Afferent PHYSIOLOGY
Mainly conveys painful sensation. Blood pressure and heart rate
• These are controlled by baroreflex pathways.
Parasympathetic nervous system (721) • Baroreceptors in the carotid sinus, aortic arch and other
Efferent thoracic regions respond to alterations in blood pressure,
• Parasympathetic preganglionic nerve fibers descend from and send information, from the carotid sinus (via afferent
the hypothalamus and other regions of the brain (e.g. fibers in the glossopharyngeal nerve) and from the aortic
Edinger–Westphal nucleus) to the brainstem and the arch and thoracic low pressure receptors (via fibers in the
intermediolateral columns of the sacral spinal cord. vagus nerve), to the nucleus of the tractus solitarius in
• The nerve fibers exit from the brain stem in cranial the brainstem.
nerves III, VII, XI, X, and from the sacral spinal cord in • Efferent fibers are carried in the vagus nerve to the heart
the S2, S3, S4 nerve roots. and in the sympathetic nerves to the heart, mesenteric
• The fibers in the S2–4 nerve roots travel in pelvic nerves vascular bed and blood vessels in the skin and muscles.
to form a diffuse subserosal network in the bladder wall. • A fall in blood pressure results in a compensatory
• Preganglionic fibers are long and post ganglionic fibers increase in heart rate and peripheral vasoconstriction
are short because the ganglia lie close to the innervated (particularly in the splanchnic vascular bed) to maintain
structures. blood pressure.

Parasympathetic fibers innervate the following: Sweating


• Pupillary and ciliary muscles (via cranial nerve III, the The sweat glands are innervated mainly by cholinergic
ciliary ganglion, and ciliary nerve). postganglionic sympathetic fibers and are under
• Lacrimal, submandibular and sublingual glands (cranial hypothalamic control.
nerve VII, and branches of the greater superficial petrosal
and chorda tympani nerves). Bladder and bowel
• Parotid gland (cranial nerve IX). Bladder
• Thoracic and abdominal viscera (cranial nerve X). Afferent:
• Bladder smooth muscle fibers, large bowel, and genital • Parasympathetic afferent fibers convey sensations of
system (sacral outflow). bladder fullness and pain of overdistension and
overcontraction. They also mediate the reflex for bladder
Pre- and postganglionic fibers in the parasympathetic contraction through S2–4 segments.
nervous system are cholinergic. • Sympathetic afferent fibers transmit painful sensation
from the trigone area, and a vague sensation of bladder
fullness. They play no part in the micturition reflex.
721 • Somatic fibers in the pudendal nerve convey sensations
from the urethra and sphincter of urethral pain and
Iris temperature, urine passing and bladder emptying.
Ciliary body III

Efferent:
Lacriminal gland VII
• Parasympathetic efferent impulses cause the detrusor
Submandibular
and sublingual IX muscles of the bladder to contract and the bladder neck
glands to shorten, thus allowing the passage of urine.
Parotid gland X • Sympathetic efferents may inhibit the detrusor but
appear to play little part in the act of micturition. During
ejaculation, sympathetic activity causes the bladder neck
Heart to contract, preventing retrograde ejaculation.
Lungs • Somatic fibers in the pudendal nerve (S2–4) innervate the
GI tract external sphincter of the bladder and the anal sphincter,
causing relaxation of the sphincter during micturition (as
the detrusor is contracting). It can also be voluntarily
contracted to terminate the act of micturition.

Pupillary reflexes
Light reflex
Rectum
• The normal pupil contracts briskly in the eye to which
S2
the light is directed (direct response) and also in the
Bladder S3 opposite eye (consensual response).
Genitalia S4 • The afferent pathway is from the retina, along the optic
nerve, to the optic tract, where fibers separate from those
destined for the lateral geniculate body and go to the
pretectal region and then to part of the cranial nerve III
nucleus (Edinger–Westphal nucleus) of both sides.
721 Schematic diagram of the parasympathetic nervous system. • The efferent pathway is from both Edinger–Westphal
(Modified from McLeod JG, Lance JW, Davies L (1995). Introductory nuclei via the IIIrd cranial nerves to the pupils, account-
Neurology, 3rd edn, Blackwell Science, Carlton, Austrialia.) ing for both the direct and consensual light reflexes.
Autonomic Neuropathy 575

Accommodation reflex Disorders associated with peripheral neuropathy


• Normal pupils constrict as the eyes converge (‘near Autonomic dysfunction which is clinically important:
response’). • Diabetes.
• Pathways descend from the parieto-occipital cortex to • Acute inflammatory radiculoneuropathy (Guillain–Barré
the Edinger–Westphal nucleus in the midbrain, and then syndrome).
as pupilloconstrictor fibers in both IIIrd nerves to the • Acute intermittent porphyria.
pupil. • Primary and familial amyloidosis.
• Familial dysautonomia (Riley–Day syndrome; dopamine
Relative afferent pupillary defect β-hydroxylase deficiency, hereditary sensory and
Also known as the Marcus Gunn pupil or swinging torch autonomic neuropathy-3 [HSAN3]).
sign. If the IIIrd cranial nerve is intact bilaterally, then the • Chronic sensory and autonomic neuropathy.
contribution of the optic nerve to the light reflex can be
assessed by swinging a torch from one eye to the other Autonomic dysfunction which is not usually clinically
every 2 seconds or so. If there is impaired conduction in one important:
optic nerve relative to the other, the direct response of light • Alcoholism and nutritional diseases.
in the affected eye and consensual response in the • Toxic neuropathies (vincristine sulfate, acrylamide, heavy
unaffected eye will be suboptimal. Swinging the torch from metals).
the affected eye to shine light in the unaffected eye evokes • Metabolic disorders (vitamin B12 deficiency, chronic renal
a brisker and more profound direct and consensual response failure, chronic liver disease).
so that the pupils in both eyes will constrict further (and • Charcot–Marie–Tooth disease.
realize the paradox that shining a light in the unaffected eye • Malignancy.
actually leads to further constriction of the pupil). Swinging • Rheumatoid arthritis.
the torch back to shine in the affected eye leads to dilatation • Systemic lupus erythematosus.
of both pupils. The effect may not be obvious initially, but • Chronic inflammatory neuropathy.
after one or two swings of the torch, the pupillary response • HIV infections.
of the affected eye rapidly fatigues (see p.484). • Prion disorders: fatal familial insomnia.
• Chagas’ disease (South American trypanosomiasis): may
ETIOLOGY AND PATHOPHYSIOLOGY target the autonomic plexus intrinsic to the heart and
Autonomic disorders may be localized (e.g. Holmes–Adie gut.
pupil) or generalized (e.g. primary chronic autonomic
failure). Drugs
• Antidepressants.
Disorders affecting CNS • Anti-hypertensive drugs.
Primary autonomic failure • Barbiturates.
• Pure autonomic failure (PAF) or idiopathic orthostatic • Phenothiazines.
hypotension (IOH). • Atropine.
• Autonomic failure with Parkinson’s disease (AF-PD). • Epidural anesthesia.
• Autonomic failure with multiple system atrophy (AF-
MSA). CLINICAL FEATURES
Autonomic dysfunction
Spinal cord lesions above T6 The most common clinical manifestations of autonomic
• Trauma. dysfunction are:
• Transverse myelitis. • Postural (orthostatic) hypotension; impotence.
• Syringomyelia. • Disorders of bladder function.
• Tabes dorsalis. • Abnormalities of sweating.
• Vasomotor disturbances.
Wernicke’s encephalopathy
Sympathetic adrenergic failure
Miscellaneous disorders • Postural hypotension.
• Cerebrovascular disease. • Ejaculatory failure.
• Brainstem tumors.
• Multiple sclerosis. Sympathetic cholinergic failure
• Adie’s syndrome. Anhidrosis.
• Tabes dorsalis.
Parasympathetic failure
Disorders affecting the peripheral nervous system • Fixed heart rate.
Disorders with no associated peripheral neuropathy • Sluggish urinary bladder and bowel.
• Acute and subacute autonomic neuropathy. • Erectile failure.
• Pandysautonomia.
• Cholinergic dysautonomia.
• Botulism.
• Bilateral lumbar sympathectomy.
576 Autonomic Nervous System Disorders

Pupillary reflexes Respiratory system


Light reflex • Stridor.
• Lesions of cranial nerve III cause ipsilateral pupil • Inspiratory gasps.
dilatation. Shining a light into either eye fails to evoke a • Apneic episodes.
direct or consensual response in the affected eye because
of the defect in the efferent arc of the reflex. However, Gastrointestinal system
shining a light in the eye ipsilateral to the lesion (the • Constipation.
affected eye) evokes a consensual reaction in the eye • Diarrhea (occasionally).
contralateral to the lesion (the unaffected eye). • Dysphagia.
• Bilateral occipital lobe lesions (beyond the optic nerves
and tracts) which cause bilateral blindness are associated Renal and urinary bladder
with preserved direct and consensual reflexes. • Nocturia.
• Frequency.
Accommodation reflex • Urgency.
Any lesion involving the pupilloconstrictor fibers will impair • Incontinence.
the reaction of accommodation. Selective impairment of • Retention of urine.
accommodation may occur in some midbrain lesions.
Reproductive system
Relative afferent pupillary defect Erectile and ejaculatory failure.
Also known as the Marcus Gunn pupil or swinging torch
sign (see p.575). ASSOCIATED FEATURES
Involvement of other neurologic systems (see
Adie’s pupil (tonic pupil of Adie; Holmes–Adie syndrome) Multiple systems atrophy, p.435)
(see p.497) • Extrapyramidal dysfunction (Parkinsonism).
• Cerebellar dysfunction.
Horner’s syndrome (see p.494) • Pyramidal dysfunction.
• Peripheral neuropathy: peripheral autonomic fibers may
Argyll Robertson (AR) pupil be damaged in peripheral neuropathies, particularly those
• Described by Douglas Argyll Robertson, an Edinburgh affecting small myelinated and unmyelinated fibers (e.g.
physician. diabetes, primary amyloidosis), or those which cause acute
• Small, irregular pupils that are non-reactive to light, but demyelination of the preganglionic sympathetic fibers and
reactive to accommodation (AR pupils are ‘accommo- the vagal afferent and efferent fibers (e.g. Guillain–Barré
dation retaining’.) They dilate poorly with mydriatics. syndrome). The usual clinical manifestations are postural
• The site of the lesion is uncertain but is probably in the hypotension, impairment of heart rate control, loss of
pretectal region. sweating, and impotence.
• Causes include syphilis and diabetes.
SPECIAL CONDITIONS
Clinical features of autonomic failure PAF or IOH
Neurologic system (see Multiple systems atrophy, p.435). • Degeneration of postganglionic (i.e. peripheral)
• Extrapyramidal dysfunction (Parkinsonism). sympathetic fibers.
• Cerebellar dysfunction. • Age of onset: 40–60 years.
• Pyramidal dysfunction. • Gender: M=F.
• Postural hypotension (dizziness and weakness on
Eye standing or walking).
• Anisocoria. • Impotence.
• Horner’s syndrome (see p.494). • Bladder disturbances.
• Dry eyes, rarely excessive lacrimation. • Loss of sweating.
• Normal neurologic examination.
Sudomotor system • Slowly progressive course.
• Sweating abnormalities (anhidrosis usually, but
occasionally gustatory facial sweating in a diabetic). Autonomic failure associated with multiple system
• Heat intolerance. atrophy (see p.435)

Cardiovascular system DIFFERENTIAL DIAGNOSIS


Postural (orthostatic hypotension): symptoms include Postural (orthostatic) hypotension
lightheadedness, dizziness, faintness, visual disturbances or • Dehydration.
loss of consciousness (syncope) on standing upright. It is • Blood loss.
usually defined as a fall in systolic blood pressure of at least • Drugs.
4 kPa (30 mmHg) or to <10.7 kPa (80 mmHg) within a
minute or two of standing, but such falls are neither Argyll Robertson pupils
sensitive nor specific (e.g. they can occur in the normal Pseudo-Argyll Robertson pupils: pupils that do not react to
asymptomatic elderly, cardiac failure, Addison’s disease). light but are not small or irregular. May occur in midbrain
lesions.
Autonomic Neuropathy 577

DIAGNOSIS The patient assumes the semi-recumbent posture and is


The clinical approach to the patient with clinical features of asked to expire forcefully, maintaining a column of mercury
dysautonomia aims to determine: at 5.3 kPa (40 mmHg) pressure for 10–15 seconds, while
• Whether autonomic function is affected. the EKG and BP are continuously recorded (intra-arterial
• Which organ systems are affected. catheter for BP).The ratio of the longest pulse interval to
• The site of the lesion (central, preganglionic and the shortest pulse interval recorded on the EKG during the
postganglionic). maneuver (the Valsalva ratio) normally exceeds 1.45 in
• The cause of the lesion. young adults. Increased age and parasympathetic and sym-
pathetic neuropathies are associated with a reduced ratio.
INVESTIGATIONS
Is autonomic function normal or abnormal? Isometric contraction
• Normally, sustained isometric contraction (e.g. a firm
Change of blood pressure with posture (postural hypotension) handgrip) for up to 5 minutes increases the heart rate
• Normally, changing from the supine to the standing (and thus cardiac output) and total peripheral resistance
position alters the blood pressure very little. (by peripheral vasoconstriction), leading to an increase
• A fall exceeding 4/2 kPa (30/15 mmHg) is abnormal, in systolic and diastolic BP. Diastolic BP rises by 2 kPa
indicating significant postural hypotension. (15 mmHg) or more.
• Autonomic neuropathies affecting sympathetic efferent
Change of heart rate with posture (reflex tachycardia; the fibers are associated with an impaired response.
30:15 ratio)
• Normally, changing from the supine to the standing Response to emotional and other stimuli
position increases the heart rate by about 11–29 beats • Normally, mental arithmetic, a loud noise, or cold or
per minute, reaching a maximum after 15 beats, after painful stimuli usually evoke peripheral vasoconstriction,
which a reflex bradycardia ensues, reaching a maximum increase the total peripheral resistance and arterial blood
at about the 30th beat. pressure. In some normal people, this response may not
• The ratio of the R–R interval (heart period) on an EKG be present.
corresponding to the 30th and 15th beats, is the 30:15 • Autonomic neuropathies affecting sympathetic efferent
ratio. The ratio is normally considerably greater than one, fibers are associated with an impaired response.
but varies with age.
• Autonomic neuropathies affecting the afferent or efferent Sweating
fibers of the reflex arc reduce the rise in heart rate associated • Autonomic neuropathies affecting sympathetic efferent
with a change of posture, and reduce the 30:15 ratio. fibers are associated with impaired sweating.
• The thermoregulatory sweat test (TST) determines the
Change of heart rate with deep breathing (sinus arrhythmia) distribution of sweat loss (anhidrosis). The skin is
• Normally, inspiration is associated with an increase in covered with a powder such as alazarine red or
heat rate and expiration with a decrease in heart rate quinazarine that changes color when sweating occurs.
(sinus arrhythmia). This difference is less pronounced in The patient is warmed by radiant heat sufficient to raise
older individuals. the body temperature by 1°C (1.8°F).
• Autonomic neuropathies involving the vagus nerve are • Local sweating may also be tested by injection or ion-
associated with impaired variation of the heart rate with tophoresis of acetylcholine which stimulates an axon reflex,
respiration (loss of sinus arrhythmia). or pilocarpine which stimulates the sweat glands directly.

Valsalva maneuver Pupillary responses


Normally, forced expiration against a closed glottis or • Metacholine (methacholine) 2.5%, installed into the
mouthpiece (Valsalva maneuver) alters the heart rate (HR) conjunctival sac, does not affect the size of the normal
and blood pressure (BP): pupil but it causes pupillary constriction if para-
• Phase I: BP rises due to the raised intrathoracic pressure sympathetic innervation is impaired. This is because of
associated with the Valsalva causing mechanical the denervation supersensitivity of the constrictor muscle
compression of the aorta and increased peripheral of the pupil.
resistance. A compensatory fall in HR, mediated by the • Epinephrine 0.1% has no effect on the normal pupil but
parasympathetics in the vagus nerve, follows. causes pupillary dilatation if postganglionic sympathetic
• Phase II: BP falls due to the fall in venous return (associated innervation is impaired, also because of denervation
with continuously high intrathoracic pressure) and thus fall supersensitivity.
in stroke volume. A compensatory rise in HR (due to • Cocaine 4% eye drops cause dilatation of the normal
parasympathetic vagal withdrawal) and total peripheral pupil because cocaine blocks the re-uptake of noradrena-
resistance (due to peripheral vasoconstriction) follows. line (norepinephrine), but if peripheral sympathetic in-
• Phase III: forced expiration ceases and venous return nervation is impaired, pupillary dilatation does not occur.
increases rapidly, increasing the stroke volume and blood • Tyramine 2% eye drops produce dilatation of the normal
pressure and decreasing the heart rate. pupil because tyramine releases noradrenaline (norepine-
• Phase IV: BP rises further and overshoots because the phrine) from peripheral nerve terminals, but if sympathetic
peripheral circulation is still vasoconstricted for up to 30 innervation is impaired, noradrenaline (norepinephrine) is
seconds because of the increase in peripheral sympathetic depleted and pupillary dilatation does not occur.
vasoconstrictor tone induced by stages II–III. Therefore,
stage IV is a measure of sympathetic vascular tone.
578 Autonomic Nervous System Disorders

Where is the lesion (i.e. site) and what is the • Elevate the head of the bed by 10–15 cm (4–6 in)
functional deficit? (reduces renal arterial pressure and thus increases the
In addition to the above tests, a number of other tests of secretion of renin, resulting in retention of sodium and
baroreflex sensitivity, vasomotor control, and bladder water, and increased blood volume).
control exist but are more difficult to perform. • Elastic stockings with lower abdominal constriction
(reduce the volume of the venous capacitance bed).
Sudomotor function • Drugs:
Quantitative sudomotor axon reflex test (Q-SART): a test – 9-α-fluorohydrocortisone (fludrocortisone), a mineralo-
of postganglionic sympathetic sudomotor function. If the corticoid which increases circulating effective blood
TST is abnormal (i.e. anhidrosis) and the Q-SART is volume and probably sensitizes peripheral blood vessels
normal, then the site of the sympathetic lesion is to catecholamines, is the most useful drug. The dose is
preganglionic. 0.1 mg/day and can be increased. The main adverse
effects are supine hypertension, fluid retention (and heart
Vasomotor function failure), worsening of diabetes mellitus, and potassium
• Skin vasomotor reflexes: measured by laser doppler flow depletion.
meters (e.g. on the pulp of index finger) in response to – Indomethacin 25–50 mg three times per day, but may
stimuli such as standing, Valsalva, cold water. Limited if cause headache.
the patient is nervous and resting skin blood flow is – Alpha antagonists: ephedrine 15–50 mg three times
minimal. daily; methylphenidate hydrochloride (Ritalin) 5–10 mg
• Axonal flare response: measure at same time as Q-SART. three times daily, and midodrine, a recently introduced
• Veno-arteriolar reflex: a test of post ganglionic adrenergic α-adrenergic agonist which acts by improving arterial and
function. venous constriction in response to standing. May be
limited by supine hypertension which itself may respond
Heart period (R–R interval) recordings (see above) to beta blockade.
Responses to deep breathing, Valsalva maneuver, and – Ambulatory norepinephrine infusion via intravenous
standing. indwelling catheter and an infusion pump.
• Atrial tachypacing may help in patients who do not have
What is the cause? a tachycardia on standing up.
Underlying and associated disorders, such as diabetes
mellitus and amyloidosis (see above), need to be excluded. Bladder dysfunction
Seropositivity for antibodies that bind to or block ganglionic Frequency of micturition
acetylcholine receptors identifies patients with various forms Anticholinergic drugs:
of autoimmune autonomic neuropathy and distinguishes • Propantheline bromide 15 mg two to four times daily.
these disorders from other types of dysautonomia. • Penthienate bromide 5 mg three to four times daily.
• Tricyclic antidepressant drugs (e.g. amitriptyline
TREATMENT 25–100 mg/day).
Aims to remove the underlying cause if possible and relieve
symptoms. Botulinum toxin injections into the detrusor muscle
(2.5 MU of Botox injected per single site, up to 30
Postural hypotension injections).
• General advice:
– Maintain hydration and salt intake (150 mmol/l Distended bladder with incomplete emptying
[150 mEq] salt). Cholinergic drugs: bethanechol chloride 10 mg three to
– Avoid dehydration, diuretics and other hypotensive drugs four times daily.
and deconditioning.
– Rise slowly from the lying and sitting positions, particularly Gastroparesis
in the morning, after hot baths and heavy meals. • Metoclopramide 10 mg before meals and at night.
– Avoid straining and extremes of temperatures. • Cisapride 15–40 mg daily in two to four divided doses.
– Wear light clothes.

FURTHER READING Naumann M, Jost WH, Toyka KV (1999) Vernino S, Low PA, Fealey RD, et al. (2000)
Botulinum toxin in the treatment of Autoantibodies to ganglionic acetylcholine
neurological disorders of the autonomic receptors in autoimmune autonomic
nervous system. Arch. Neurol., 56: 914–916. neuropathies. N. Engl. J. Med., 343: 847–855.
Donofrio PD, Caress JB (2001) Autonomic dis- Oldenburg O, Mitchell A, Nurnberger J, et al..
orders. The Neurologist, 7: 220–233. (2001) Ambulatory norepinephrine treatment
Kauffmann H (2000) Primary autonomic failure: of severe autonomic orthostatic hypotension.
three clinical presentations of one disease? J. Am. Coll. Cardiol., 37: 219–223.
Ann. Intern. Med., 133: 382–384. Schatz IJ (2001) Treatment of severe autonomic
Mathias CJ (1997) Autonomic disorders and their orthostatic hypotension. Lancet, 357:
recognition. N. Engl. J. Med. 336: 721–724. 1060–1061.
Chapter Twenty-one 579

Diseases of the
Peripheral Nerve
PERIPHERAL NEUROPATHY Epineurium Fascicles Perineurium 722

DEFINITION
A disorder of any or all of the peripheral nerves or nerve
roots.
Regional
EPIDEMIOLOGY artery
• Incidence: 69 per 100 000 per year.
• Lifetime prevalence: 3 per 1000.
Nutrient
• Age: any age, but usually the middle-aged and elderly. artery
• Gender: M=F.

ANATOMY (722–725) 722 Diagrammatic representation of peripheral nerve trunk and its
A peripheral nerve consists of a cell body and about six blood supply. Regional arteries arranged longitudinally outside the
fascicles, each of which contains many myelinated and nerve trunk give rise to nutrient arteries that penetrate the epineurium
unmyelinated axons. Each fascicle has small nutrient blood and form an anastomosis. Small arterioles penetrate the perineurium
vessels which are integral to its function. of the fascicles and form an endocapillary network.

723 Diagrammatic 723


representation of Dorsal root
components of the
peripheral nerve trunks.
Peripheral nerve Afferent fibers
Efferent fibers
Post-ganglionic
sympathetic
efferents
Preganglionic Ventral
sympathetic efferents root

724 725

724 Transverse section of a peripheral nerve stained with toluidine 725 Electron micrograph of a transverse section through a normal
blue.The circular shaped myelin is dark blue. myelinated nerve fiber, showing the axon in the center surrounded by
several layers of myelin.The major dense lines of the myelin arise by
the fusion of the inner surfaces of the Schwann cell surface membrane.
580 Diseases of the Peripheral Nerve

Anatomic classification selectively affect small unmyelinated nerve (e.g. alcohol and
• Focal neuropathy or mononeuropathy. amyloid) leading to the characteristic autonomic disturbance
• Multifocal neuropathy. seen in these neuropathies. Inflammatory and vasculitis
• Generalized polyneuropathy. disorders often lead to multiple, random pathologic lesions
within the nerve.
PATHOLOGY (726–728)
Peripheral neuropathy results from a disturbance of Pathologic classification
structure and function of the peripheral sensory, motor and • Demyelinating.
autonomic neurons. The disturbance of structure and • Axonal.
function may affect the:
• Cell body: neuronopathy. ETIOLOGY
• Axon: axonopathy. Worldwide, the most common cause of peripheral nerve
• Myelin sheath: myelinopathy. disease is leprosy (see Differential diagnosis below).

Neuronopathy CLINICAL HISTORY


• Anatomy: anterior horn cells (AHC) or dorsal root As the peripheral nerves supply motor power, sensation, and
ganglia (DRG). autonomic function, patients with peripheral nerve disease
• Pathology: degeneration of central and peripheral may experience symptoms of any of these functions.
processes.
• Examples: spinal muscular atrophy (AHC), toxic Symptoms
neuronopathy, paraneoplastic neuropathy (DRG). Sensory
• Numbness and tingling, usually beginning in the feet
Axonopathy (supplied by the longest nerves) are common symptoms
• Anatomy and pathology: metabolic derangement within of patients with sensory axonal neuropathies, particularly
axon (e.g. uremia); begins distally and progresses those involving damage to the cell body (e.g. vitamin B12
proximally: ‘dying-back’ neuropathy; secondary deficiency).
breakdown of myelin sheath. • Pain beneath the sole, and a feeling as if the socks are
• Clinical features: distal (legs>arms), symmetric, mixed ruffled are other common symptoms.
sensorimotor neuropathy. • Upper limb symptoms, if present, generally include
• Recovery: slow, often incomplete. difficulty picking up small objects such as pins, numbness
• Causes: alcohol, diabetes, uremia, Fabry’s disease. or a sandpaper feeling on the fingers.

Myelinopathy Motor
• Anatomy and pathology: damage to myelin sheath or • Limb weakness.
Schwann cell (with sparing of axon); segmental • Falls.
demyelination ± conduction block. • Double vision.
• Clinical features: profound weakness, mild sensory loss • Difficulty swallowing.
(relative sparing of small myelinated and unmyelinated • Slurred speech.
fibers). • Shortness of breath.
• Recovery: rapid, within weeks; repeated demyelination
and remyelination; Schwann cell proliferation (‘onion Autonomic disturbances
bulb’ layering) around the axon. • Postural hypotension.
• Causes: Guillain–Barré syndrome. • Impotence.
• Bladder dysfunction.
Wallerian degeneration • Diarrhea.
• Anatomy: interruption of a healthy axon. • Constipation.
• Pathology: distal degeneration of axon and myelin • Loss of sweating.
sheath.
• Clinical features: immediate weakness and sensory loss. Sometimes these symptoms are preceded by severe pain,
• Recovery: slow and incomplete. and are usually due to pathologies that affect small fibers
• Causes: trauma, ischemia. within nerves, such as amyloid, alcohol, Guillain–Barré
syndrome) and diabetes (20–40% of patients).
Disorders of the peripheral nerve can primarily damage
myelin (e.g. Guillain–Barré syndrome) or the axon (e.g. Bulbar dysfunction
vincristine and many other drugs), or can interfere with the Due to cranial neuropathies which may occur in some
blood supply of the peripheral nerve leading to areas of peripheral neuropathies:
necrosis in the distribution of the small nutrient vessels (e.g. • Double vision.
polyarteritis nodosa). Many toxic processes that produce • Slurred speech.
slowly progressive damage to nerve cell bodies lead to a • Difficulty swallowing.
‘dying-back’ type of neuropathy, in which the longest axons • Respiratory problems.
show damage initially and the neuropathic symptoms ascend
from the feet. Alternatively, pressure on one portion of a History targeted to the underlying etiology
nerve at a site of entrapment can lead to myelin loss and, if A careful history is essential, and should be targeted to
severe, axonal damage. Some pathologic processes include the following details:
Peripheral Neuropathy 581

• Family history: at least 25% of neuropathies without an Multifocal neuropathy


obvious cause are genetic in origin. A thoroughly • Inflammation:
researched family tree often reveals details of forgotten – Leprosy.
relatives with foot deformity or death at an early age with – Lyme disease.
an unusual inherited disorder. – Sarcoidosis.
• Drug exposure. • Infiltration:
• Alcohol intake. – Amyloidosis.
• Systemic disease (e.g. diabetes). – Malignancy.
• Environmental toxins. • Immune reaction:
• HIV. – Immunization.
• Travel (infections). – Foreign serum and proteins.
• Malignancy. • Trauma: multiple nerve injuries.
• Vascular disease:
PHYSICAL EXAMINATION – Diabetes.
Lower motor neuron lesion – Polyarteritis nodosa.
Any or all of the following in any combination: – Rheumatoid arthritis.
• Muscle wasting and fasciculations, commonly in distal – Sjögren’s syndrome.
muscles of hands and feet. – Systemic lupus erythematosus.
• Muscle weakness (e.g. limbs, face, eyelids, diaphragm). – Wegener’s granulomatosis.
• Depressed or absent reflexes. – Vasculitis (non-systemic).
• Sensory loss: pain and heat sensation (small fiber); fine
touch, proprioception, two-point discrimination (large
fiber). 726
Myelin
• Autonomic disturbance (postural hypotension, loss of
sinus arrhythmia) may be present. Axon

Chronic denervation
Trophic changes:
• Skin: thin, red, shiny, transparent, indolent ulcers
(repeated trauma).
• Nails: transversely striped, thickened, brittle.
• Hair: disappears from denervated areas.
• Joints, bones: immobility: disuse atrophy.

Underlying cause
• Pes cavus (inherited). Skin or
• Diabetic retinopathy. muscle
• Hepatomegaly (alcohol). A B C D

DIFFERENTIAL DIAGNOSIS
Anatomic 727 728
Focal neuropathy or mononeuropathy
• Local entrapment.
• Mononeuritis.
• Trauma.

726 Diagrammatic representation of pathologic changes in peripheral


nerve fibers. A: normal myelinated nerve fiber; B:Wallerian
degeneration of the axon and myelin following section or crush of
nerve fiber; C: axonal degeneration (with secondary myelin
degeneration) of the distal portion of nerve fiber in ‘dying-back’
neuropathy; D: segmental demyelination, with degeneration of the
myelin in one internode but preservation of the axon.

727 Teased nerve fiber (osmium tetroxide) showing segmental


demyelination (yellow axons [myelin normally appears black]) and
active Wallerian degeneration due to focal interruption of axons
(numerous myelin ovoids which appear as black blobs).

728 Teased nerve fiber showing segmental demyelination (of the


bottom half of the strip).
582 Diseases of the Peripheral Nerve

Generalized polyneuropathy • Hereditary sensory and autonomic neuropathy (HSAN type


• Alcohol. 1) – autosomal dominant, small fibre loss, acromutilation).
• Diabetes. • Hypothyroidism.
• Uremia. • Lyme disease.
• Vitamin B12 deficiency. • Paraneoplastic sensory neuropathy.
• Guillain–Barré syndrome. • Vasculitic neuropathies.
• Chronic inflammatory demyelinating polyneuropathy
(CIDP). Autonomic neuropathies
• Hereditary. • Amyloidosis (primary or familial).
• Diabetes.
Clinical • Guillain–Barré syndrome.
Acute onset • HSAN 3 (autosomal recessive; Riley–Day syndrome).
• Critical illness polyneuropathy. • Porphyria (acute intermittent porphyria).
• Diabetes, uremia (rarely).
• Diphtheria. Pathologic
• Guillain–Barré syndrome. Demyelinating neuropathies
• Malignancy. Acute onset:
• Porphyria. • Diphtheria.
• Serum sickness (post immunization). • Guillain–Barré syndrome (AIDP).
• Toxic (e.g. arsenic, nitrofurantoin).
Subacute/chronic onset:
Predominantly motor • Hereditary neuropathies:
• Botulism. – Hereditary motor and sensory neuropathy (HMSN)
• Charcot–Marie–Tooth disease. types 1 and 3.
• Diabetes (diabetic amyotrophy). – Refsum’s syndrome.
• Diphtheria. – Metachromatic leukodystrophy.
• Guillain–Barré syndrome. – Krabbe’s disease.
• Lead. • Inflammatory neuropathies:
• Porphyria. – Chronic inflammatory demyelinating polyneuropathy
(CIDP).
Predominantly sensory – Motor neuropathy with multifocal conduction block.
• Amyloidosis (primary or familial). – Chronic inflammatory polyneuropathy associated with
• Diabetes (distal sensory polyneuropathy). paraproteinemia.
• Fabry’s disease. – Inflammatory neuropathy associated with HIV infection.
• Guillain–Barré syndrome. • Malignancy: some acute and subacute neuropathies
• Hereditary sensory and autonomic neuropathies. associated with carcinoma and lymphoma.
• Hypothyroidism. • Metabolic neuropathies:
• Infections: – Diabetes (sometimes).
– HIV infection. – Uremia (sometimes).
– Leprosy. • Toxic neuropathies:
– Lyme disease. – Amiodarone.
– Syphilis (tabes dorsalis). – Hexacarbons.
• Mitochondrial cytopathy. – Perhexilene (perhexiline) maleate.
• Neoplastic (carcinomatous) and paraneoplastic
neuropathy. Axonal degeneration
• Sarcoidosis. Acute:
• Sjögren’s syndrome. • Guillain–Barré syndrome (AMAN).
• Toxicity: • Toxins.
– Arsenic. • Porphyria.
– Thallium. • Vasculitis (polyarteritis nodosa, systemic lupus erythe-
• Uremia. matosus).
• Vasculitis.
• Vitamin B12 or thiamine deficiency. Chronic:
• Hereditary:
Radicular – HMSN type 2.
• Diabetic truncal neuropathy. – Giant axonal neuropathy.
• Lyme disease. – Hereditary ataxias.
• Sjögren’s syndrome. • Inflammatory: systemic lupus erythematosus, sarcoid.
• Infectious: leprosy.
Painful neuropathies • Paraneoplastic:
• Alcohol, nutritional deficiencies. – Lymphoma.
• Arsenic. – Carcinoma of the lung.
• Cryoglobulinemia. • Metabolic:
• Diabetes (acute painful neuropathy). – Diabetes.
Peripheral Neuropathy 583

– Uremia. • Hexacarbon neuropathy.


• Toxic: • IgM kappa paraproteinemia neuropathy.
– Alcohol. • Metachromatic leukodystrophy.
– Drugs (dapsone, gold, isoniazid, metronidazole, • Krabbe’s disease.
platinum, vincristine, vitamin B6). • Fabry’s disease.
• Deficiencies:
– Vitamin B12. Diagnosis of a chronic axonal neuropathy, the most
– Thiamine. common neuropathy, is not usually helped by nerve biopsy.
– Vitamin E. About one-third of patients experience persistent increased
– Nicotinamide. pain at the biopsy site.
• Miscellaneous:
– Primary amyloid. Other investigations
Core screen
INVESTIGATIONS • Full blood count with differential.
Nerve conduction studies and electromyography (EMG) • ESR.
• The most useful investigations in peripheral nerve • Fasting blood glucose and hemoglobin A1C.
disease. • Serum vitamin B12 level.
• Determines whether a neuropathy is present, its • Serum protein electrophoresis.
distribution (e.g. focal or generalized) and in some cases • Chest x-ray.
the precise location (e.g. carpal tunnel), whether the
process is axonal or demyelinating, and also whether Targeted
there is any evidence of conduction block. • Thyroid function tests.
• Conduction block implies a focal area of demyelination • Antinuclear antibodies, rheumatoid factor, antineutrophil
which is characteristic of diseases such as CIDP or cytoplasmic antibody, cryoglobulins.
multifocal motor neuropathy. • HIV, Lyme disease, hepatitis B and C, syphilis serology.
• Serum vitamin E.
Nerve conduction velocity (NCV) • Peripheral autonomic skin response: to determine if a
Depends on the: small fiber neuropathy.
• Diameter of the fiber. • Anti-Hu-antibody (encephalomyelitis with sensory
• Distance between the nodes of Ranvier. neuropathy).
• Integrity of the myelin sheath. • CSF: cells, protein, glucose, cytology, serology.

Segmental demyelination: DIAGNOSIS


• NCV reduced by >40% of mean normal values. • Does the patient have a peripheral neuropathy?
• Conduction block. • What type of neuropathy is it (anatomy, pathology)?
• Slowing of F-wave responses. • What is the course of the neuropathy (acute [1–4 weeks],
subacute [1–3 months], chronic [>3 months], or
Axonal neuropathies: relapsing)?
• NCV normal or slightly reduced. • What is the cause of the neuropathy?
• NCVs are those of the fastest conducting fibers (large
myelinated axons). TREATMENT
• Specific treatment of the underlying cause if possible (e.g.
Action potential amplitude vitamin B12 replacement).
The amplitude is an indication of the number of motor units • Ameliorate the decline in function and optimize
present. adaptation to any disability:
• Segmental demyelination: CMAP amplitude: normal or – Physiotherapy: to maintain muscle strength.
slightly reduced (dispersed). – Occupational therapy: orthotic aids, wheelchair.
• Axonal neuropathies: CMAP amplitude: declines – Podiatry (cf. foot deformity [pes cavus] and sensory loss).
progressively with fallout of axons.

Nerve biopsy
Sural nerve biopsy may reveal characteristic histopatho-
logical appearances, and be helpful in patients suspected to
have the following conditions:
• Chronic demyelinating neuropathy where immunologi-
cally-mediated disorders (e.g. CIDP) requiring treatment
with steroids or other immunosuppressants are better
managed with supportive histology.
• Charcot–Marie–Tooth disease type 1.
• Vasculitic neuropathies (e.g. multiple mononeuropathies).
• Sarcoidosis.
• Leprosy.
• Amyloid.
• Giant axonal neuropathy.
584 Diseases of the Peripheral Nerve

HEREDITARY NEUROPATHIES galactosidase, and resulting in accumulation of tri-


hexosylceramide in various tissues.
• Clinical features: painful neuropathy affecting hands and
DEFINITION feet, with pain crises in the palms and soles. Other
• A genetically heterogeneous group of several familial and manifestations affect the eye (angiokeratomata with lipid
slowly progressive disorders of peripheral nerves and inclusions on microscopy), eye (corneal opacities in 80%),
anterior horn cells of the spinal cord which are inherited heart (arrhythmias) and kidney (renal failure and
as an autosomal dominant, autosomal recessive, or X- birefringent lipid on microscopy).
linked trait, and which have overlapping clinical and • Treatment: symptomatic; the pain usually responds to
electrophysiologic characteristics. carbamazepine.
• They were formerly known as Charcot–Marie–Tooth
(CMT) disease, or peroneal muscular atrophy but, Porphyrias
following recent major advances in the understanding of • Inheritance: autosomal dominant.
the genetics of hereditary motor and sensory neuropathy • Etiology: impaired porphyrin metabolism.
(HMSN), they are now classified according to their • Clinical features: predominantly motor peripheral neuro-
genetic and etiologic basis. pathy which may mimic Guillain–Barré syndrome or multi-
ple mononeuropathy. The course is relapsing and exacer-
EPIDEMIOLOGY bations are usually triggered by medications, such as female
• Prevalence: 8–41 per 100 000 population. sex hormones, benzodiazepines, and anti-epileptic drugs.
• Age: onset in second or third decade of life. Neuropsychiatric manifestations are seen in acute intermit-
• Gender: M=F. tent and variegate porphyria. The relapsing course and
psychiatric symptoms may lead to a misdiagnosis of hysteria.
CLASSIFICATION • Investigations: the urine turns red-brown on standing,
HMSN CMT] with known genetic basis due to urinary porphyrins.
HMSN (CMT) type 1 (see p.585) • Treatment: supportive and symptomatic with avoidance
• HMSN (CMT) 1A. of precipitating medications. Other family members
• HMSN (CMT) 1B. should be considered for testing.
• HMSN (CMT) X.
Familial amyloid polyneuropathies
HMSN (CMT) type 3 (see p.585) • Inheritance: autosomal dominant.
• Pathology and etiology: small fiber neuropathy due to
HMSN with unknown genetic basis deposition of amyloid in the extracellular space. The most
Demyelinating neuropathies common fibrillar proteins deposited as amyloid are
• Autosomal dominant. transthyretin apolipoprotein A-1 (transthyretin is
• Autosomal recessive. produced in the liver) and gelsolin. For transthyretin
alone there are 40 amyloidogenic point mutations
Axonal neuropathies (chromosome 18q). The commonest mutation causes the
• HMSN (Charcot–Marie–Tooth) type 2 (see p.585) substitution of methionine for valine at position 30 which
• Complex HMSN. results in a late onset, progressive, painful, predominantly
sensory neuropathy, formerly called the Portuguese type
Hereditary neuropathy with liability to pressure or familial amyloid neuropathy type 1.
palsies (see p.587) • Clinical features: pain, loss of pain and temperature sen-
Etiology: autosomal dominant; deletion, or sometimes a sation, autonomic dysfunction, cardiomyopathy, and occa-
point mutation, at 17p 11.2. sional vitreous, renal and other organ involvement (729).
• Diagnosis: by nerve biopsy.
Syndromic hereditary peripheral neuropathies • Treatment: symptomatic, but liver transplantation in
Refsum’s disease (heredopathia atactica polyneuritiformis) young adults can halt the progression of this subtype.
• Inheritance: autosomal recessive.
• Age: onset in first or second decade.
• Pathology and etiology: demyelination due to abnormal 729 729 Macroglossia
storage of phytanic acid as a result of a defect in alpha- due to amyloid
oxidation of beta-methylated fatty acids. infiltration of the
• Clinical features: night blindness (retinitis pigmentosa); tongue in a patient
dry, scaling, ichthyosis-like skin; polyneuropathy. with primary
Cerebellar ataxia and cardiac abnormalities are late amyloidosis.
features. Cochlear hearing loss occurs in 80% of cases
• Treatment: a low phytanate diet is rarely very successful.

Fabry’s disease
• Prevalence: 1 per 40 000 (rare).
• Age and gender: 10–30 year old males.
• Inheritance: X-linked (Xq22).
• Pathology and etiology: a disorder of glycolipid
catabolism associated with defective lysosomal alpha-
Hereditary Motor and Sensory Neuropathy (Charcot–Marie–Tooth Disease) 585

HEREDITARY MOTOR AND Axonal neuropathies


SENSORY NEUROPATHY • HMSN (CMT) type 2: caused by mutations to a number
(CHARCOT–MARIE–TOOTH DISEASE) of as yet unknown genes. Distinguished from HMSN 1 by:
– Autosomal recessive inheritance; the responsible genes
have not been identified.
DEFINITION – Axonopathy rather than demyelination (i.e. sparse
A general name for a group of genetic disorders affecting demyelination and no onion bulbs).
peripheral nerves. First described by Jean Martin Charcot • Complex HMSN: other categories of HMSN with
and Pierre Marie in France in 1886. associated features have yet to be fully characterized.
Some have pyramidal features (retained reflexes and
EPIDEMIOLOGY extensor plantar responses), optic atrophy, deafness, or
• The most common inherited peripheral neuropathy. pigmentary retinal degeneration.
• Prevalence: 5/100 000.
• Age: onset in second or third decade of life. PATHOLOGY
• Gender: M=F. HMSN (Charcot–Marie–Tooth) type 1
• Nerve fiber loss with segmental demyelination and
HMSN (Charcot–Marie–Tooth disease [CMT]) remyelination in surviving fibers.
• HMSN type 1: up to 80% of cases: • ‘Onion bulbs’, comprising concentrically proliferated
– HMSN (CMT) 1A: 70% of patients with HMSN 1. Schwann cells surrounding surviving myelinated fibers,
– HMSN (CMT) X: 25% of families with HMSN 1. are characteristic beyond adolescence.
• HMSN type 2: up to 20% of cases.
HMSN (Charcot–Marie–Tooth) type 2
ETIOLOGY A predominantly axonal neuropathy (neuronal form).
Genetically heterogeneous: autosomal dominant, autosomal
recessive, X-linked and spontaneous cases occur. PATHOPHYSIOLOGY
HMSN (Charcot–Marie–Tooth) type 1
HMSN (CMT) with known genetic basis The mutations disrupt myelin and Schwann cell function in
HMSN (CMT) type 1 a number of complex ways, leading to segmental
• Autosomal dominant inheritance. demyelination and secondary axonal damage of the
• Caused by mutations in one of several genes expressed peripheral nervous system. It is the secondary axonal damage
in Schwann cells, the myelin producing cells of the which is the major cause of weakness in HMSN (CMT) 1.
peripheral nervous system.
– HMSN (CMT) 1A: duplication of the gene coding for a CLINICAL FEATURES
22 kDa peripheral nerve myelin protein 22 (PMP 22), a HMSN (CMT) type 1
Schwann cell adhesion molecule, on the short arm of • Widely variable, even within families.
chromosome 17, at position 17p 11.2. • Symptoms develop in childhood (particularly HMSN
– HMSN (CMT) 1B: point mutations in the protein zero 1A) and include: ‘weak ankles and legs’, ‘tripping’, ‘can’t
(P0) gene, which encodes the major PNS myelin run’, ‘can’t walk on heels’, ‘poor balance’.
structural protein. • Progressive, predominantly distal muscle wasting and
– HMSN (CMT) X: point mutations in the connexin-32 weakness involves mainly the legs. Advances cases show
gene on the X chromosome, which encodes the gap ‘inverted champagne bottle’-shaped legs due to atrophy
junction channel protein connexin-32 which is expressed of the peroneal and calf muscles and relative sparing of
in myelinating Schwann cells but not incorporated in the the quadriceps muscles.
myelin sheath. • Reduced or absent deep tendon reflexes.
– Point mutations in the early growth response gene 2 • Thickened greater auricular nerves may be visible in the
(EGR-2), or Krox 20, encoding a zinc finger neck, and enlarged ulnar and peroneal nerves may be
transcription factor expressed in myelinating Schwann palpable in some patients.
cells. • Severe childhood cases with marked nerve thickening are
a feature of Déjérine–Sottas disease (or HMSN 3).
HMSN (CMT) type 3: • Upper limb tremor may be present, and when prominent
• Déjérine–Sottas syndrome (a severe and early-onset is known as Roussy–Levy syndrome.
form). • Pes cavus is common as the weakness progresses.
• Caused by some point mutations in the PMP22 gene. • Scoliosis is sometimes present.
• High stepping gait due to foot drop tends to develop late.
HMSN with unknown genetic basis • Weakness in the hands is present in some patients, and
Demyelinating neuropathies may cause difficulty writing and manipulating small
• Autosomal dominant: HMSN 1 non-A non-B is the objects, but is seldom severe.
term applied to the disease in families where linkage to • Claw hands in some patients, late in the disease.
chromosomes 17 and 1 have been excluded; their • Sensory involvement is slight, and positive sensory
genotypes await identification. symptoms such as tingling or pain are not expected.
• Autosomal recessive: a rare, severe, early-onset disorder • Autonomic function is preserved.
mapping to chromosome 8q. Some forms have focally • Positive family history is common: many people have
folded myelin sheaths on nerve biopsy. mild symptoms and may not seek medical help until
another family member has more severe problems.
586 Diseases of the Peripheral Nerve

HMSN type 2 TREATMENT


• Dominant or recessive inheritance. • Treatment is a team approach incorporating neurologists,
• Similar clinical presentation to type I but may not GPs, genetic counsellors, physiotherapists, podiatrists, and
develop symptoms until the fourth or fifth decade and if necessary orthotists and orthopedic surgeons.
may produce more extensive wasting. • There is still no specific pharmacologic treatment.
• The emphasis is on symptomatic management, aimed
Recessive disorders are usually of earlier onset, more primarily at maintaining lower limb function and
clinically severe, and less variable in their clinical expression preventing forefoot drop creating pes cavus and other
because there are two mutant genes. Dominant disorders deformities by regular physiotherapy (e.g. stretching,
are less severe, of later onset, and more clinically variable exercises) and providing appropriate splints, ankle-foot
because there is one mutant gene and one normal gene. and in-shoe orthotics and footwear. In children the aim
is to prevent bone deformity caused by muscle imbalance.
DIFFERENTIAL DIAGNOSIS • As with any genetic disorder, genetic advice to the patient
Other hereditary motor and sensory neuropathies and family is of paramount importance. Coming to terms
(see p.584) with having a genetic disorder can be a difficult process
• Hereditary neuropathy with liability to pressure palsies for the patient and other existing family members.
(see p.587). Furthermore, the risk of passing on the condition to their
• Refsum’s disease. children needs to be evaluated and recognized.
• Fabry’s disease.
• Porphyria. PROGNOSIS
• Familial amyloid polyneuropathy. • Depending on the type, HMSN has the potential to
cause significant disability if ignored and prevention
Distal myopathy measures are not initiated early and maintained.
Miyoshi myopathy (dysferline positive staining): • The level of disability therefore depends on how early the
• Autosomal recessive inheritance. patient is diagnosed and whether there has been
• Onset: 15–30 years. consistent use of splints and compliance with exercise.
• Wasting and weakness of muscles of posterior • If patients are well monitored and aware of their
compartment of leg (e.g. calves), which may be limitations, many can lead active lives with little need of
asymmetric. reliance on medical care. Nevertheless, many do have to
• Ankle reflexes may be absent. modify their activities, for example, by avoiding jobs that
• Marked elevation of serum creatine kinase. involve fine hand movements or constant standing.
• Normal nerve conduction studies.
• Myopathic EMG findings.
• Hypertrophic muscle fibers with patchy necrosis,
occasional regenerating fibers, and positive dysferlin
staining.
• Variable progression.

INVESTIGATIONS
Electrophysiologic studies
HMSN type 1
Uniform and severe slowing of peripheral motor conduction
velocity (usually <38 m/sec in the median nerve) due to
peripheral demyelination.

HMSN type 2
A predominantly axonal neuropathy (neuronal form), in
which nerve conduction velocities are normal or near 730
normal but the compound muscle action potentials are of
reduced amplitude.

Molecular DNA analysis

Sural nerve biopsy (730)

DIAGNOSIS
Establish from clinical and neurophysiologic assessment of
the type of HMSN (CMT), and the pattern of inheritance
in the family:
• HMSN (CMT) type 1: DNA analysis for the three types
of HMSN (CMT) 1.
• HMSN (CMT) type 2: clinical and neurophysiologic; 730 Teased fibers from a sural nerve showing segmental
there is no direct DNA test. demyelination in several strips in a patient with HMSN (CMT) type 1.
Hereditary Neuropathy with Liability to Pressure Palsies (HNPP) 587

HEREDITARY NEUROPATHY • Patients may have mild talipes cavus or other features sug-
WITH LIABILITY TO PRESSURE gestive of a generalized inherited disorder of peripheral nerve.
PALSIES (HNPP) • Tendon reflexes are often depressed or abolished.
• Pes cavus or skeletal deformities are rare.
• There is no nerve hypertrophy or CNS involvement.
DEFINITION
An autosomal dominant inherited disease characterized by DIFFERENTIAL DIAGNOSIS
recurrent episodes of acute weakness or sensory disturbance, Clinically, HNPP can be difficult to distinguish from other
due to single or multiple peripheral nerve palsies, often inherited neuropathies, notably hereditary neuralgic
precipitated by minor compression or traction. amyotrophy (HNA) and CMT. Patients with HNPP may
manifest only recurrent brachial plexus palsy or a familial
EPIDEMIOLOGY tendency to brachial plexus palsy. However, these episodes,
• Incidence: unknown, but most neurologists see at least associated with diffuse nerve conduction abnormalities, are
one family with this condition. usually painless, without amyotrophy, and usually associated
• Age: onset is usually by the second decade of life. with other mononeuropathies.
• Gender: in some families the disorder predominates in The confusion with CMT may relate to the possible
males, but this may reflect a difference in everyday presence of pes cavus and abolished ankle jerks or peroneal
activities. amyotrophy in HNPP or to tomaculous changes in CMT.

PATHOLOGY Hereditary neuralgic amyotrophy


• Nerve fiber loss with demyelination and remyelination • Recurrent episodes of acute painful arm weakness.
(731). • Amyotrophy.
• Focal, large, sausage-shaped enlargement/thickening • A focal axonal disorder with focal electrophysiologic
(‘tomacula’) of the myelin sheath after teasing and abnormalities.
staining of individual nerve fibers in biopsy specimens of
the sural or another cutaneous nerve. These Charcot–Marie–Tooth disease
abnormalities are also found in carriers in whom pressure • Recurrent acute nerve palsies are absent.
palsies have yet to occur. • The median MNCV is markedly reduced in CMT1A,
• Electron microscopy reveals the tomacula which mostly below 30 m/sec.
correspond to regions of uncompacted myelin lamellae, • Conduction blocks are absent.
folded back longitudinally or circumferentially.
Other causes of multiple mononeuropathy
ETIOLOGY AND PATHOPHYSIOLOGY • Vasculitis.
Autosomal dominant inheritance. The gene defect is a large • Diabetes.
1.5 Mb interstitional deletion of DNA at chromosome 17p
11.2-12. The deleted region appears uniform in unrelated INVESTIGATIONS
pedigrees. It includes the gene for peripheral myelin protein Nerve conduction studies
22 (PMP-22), and involves the same chromosomal region Electrophysiologic evidence of a mild generalized
that is duplicated in Charcot–Marie–Tooth type 1A disease neuropathy with slowing of motor and sensory nerve
(also known as hereditary motor and sensory neuropathy type conduction velocities or conduction block at common nerve
1a). This region is now called the ‘CMT1A/HNPP entrapment sites (pressure points), such as the ulnar groove
monomer unit’. This finding suggests that the two disorders or fibula head, in clinically affected and unaffected nerves.
may be reciprocal products of unequal crossover during It is the coexistence of mild-to-moderate slowing of nerve
meiosis: HNPP with a single allele (monosomy, through conduction in clinically unaffected nerves that is the crucial
deletion) and HMSN 1A with three alleles (trisomy, through
reduplication). The genetic relationship between the two
diseases may explain why some patients show the phenotype 731
of a symmetric demyelinating sensorimotor polyneuropathy
after a series of pressure palsies, or sometimes as the
presenting feature.

CLINICAL FEATURES
History
• Recurrent episodes of painless compressive neuropathy
(e.g. peroneal neuropathy) or plexopathy at common
pressure points, often induced by relatively minor
trauma, such as repeated occupational squatting and
kneeling or carrying heavy objects on the shoulders.
• Onset is usually acute, but progressive weakness may
occur.

EXAMINATION 731 Nerve biopsy, transverse section, showing demyelinating


• Generally yields little information besides a focal neuropathy. A few enlarged myelin rings remain but most axons are
compressive neuropathy or plexopathy. devoid of myelin and there is early ‘onion bulb’ formation.
588 Diseases of the Peripheral Nerve

finding. HNPP is the only hereditary neuropathy in which GUILLAIN–BARRÉ SYNDROME (GBS)
conduction blocks occur. (ACUTE INFLAMMATORY
Bilaterally delayed median distal motor latency and DEMYELINATING POLYRADICULO-
reduced sensory velocity in the palm-wrist segment and a NEUROPATHY [AIDP])
delayed distal motor latency or reduced motor nerve
conduction velocity in the peroneal nerve are highly
suggestive of HNPP when there is a family history of DEFINITION
HNPP. Bilaterally normal median nerve DML and sensory An acute, reactive, self-limited, autoimmune disease,
velocity at the wrist would exclude this possibility. triggered by a preceding viral or bacterial infection, and
characterized by progressive muscle weakness and
Electromyography respiratory paralysis associated with absent deep tendon
Secondary signs of denervation are found in severe cases. reflexes, which develops over a period of 3–4 weeks. The
name honors the French neurologists who described an
Molecular analysis acute idiopathic polyneuritis in two soldiers in 1916.
This is a rapid and reliable diagnostic tool. The 17p 11.2
deletion is found in most families (22/24 to date). Since EPIDEMIOLOGY
the possibility of genetic heterogeneity exists, combined • Incidence: about 2 per 100 000 per year.
molecular and electrophysiologic examinations can be • Age: any age, but rare in infancy. The incidence increases
performed, obviating the need for nerve biopsies to with age after 40 years of age, and is maximal at 50–74 years.
diagnose HNPP. • Gender: M>F.

Nerve biopsy PATHOLOGY


Specimens show myelin redundancy or duplication in • Early lymphocyte infiltration in spinal roots and peri-
discrete areas of the myelin internode. These sausage-shaped pheral nerves, and deposition of activated complement
thickenings of myelin sheaths, most easily detected on components along the outer Schwann-cell surface mem-
teased fiber preparations, were designated by Madrid and brane of myelinated nerve fibers. Vesicular disruption of
Bradley as ‘tomacula’, which led to coining of the term myelin sheaths progresses from outward to inward.
‘tomaculous neuropathy’. The tomaculous changes are not • Subsequent recruitment of macrophages and invasion of
specific to HNPP and occur, although with a lesser nerve fibers, followed by macrophage-mediated seg-
frequency and intensity, in other inherited neuropathies such mental (multifocal) demyelination of peripheral nerves.
as HNA, CMT, and Déjérine–Sottas disease. • Variable disruption and loss of nerve axons accompanies
inflammatory demyelination in severe cases, and is
DIAGNOSIS thought to be a secondary ‘bystander’ effect, possibly
• Painless. caused by intense inflammation, edema, and swelling of
• No amyotrophy. nerves. The degree of axonal loss is an important
• Diffuse nerve conduction abnormalities. determinant of recovery and prognosis.
• Median MNCV is often reduced but rarely below • Repair and remyelination begin when the immune
40 m/sec. reactions cease.
• Slowing of MNCV in other nerves is heterogeneous and • Denervation atrophy of groups of muscle fibers if there
frequently marked in entrapment sites. is axonal degeneration (732).
• Conduction blocks in one or more nerves are suggestive
of HNPP.
• Molecular DNA analysis – 17p11.2 deletion.
• Nerve biopsy – tomaculae.

TREATMENT 732
• Inform patients and their siblings about the practical
implications of the diagnosis.
• Practices to avoid include:
– Sitting with legs crossed.
– Wearing rucksacks.
– Choosing a profession where frequent kneeling is necessary.
– Careless positioning of limbs during general anesthesia.
• Genetic counselling can help identify children who may
be affected.
• There is no effective medical or surgical treatment.

PROGNOSIS
The individual palsies are often slow to recover (conduction
blocks may last for weeks, months or even several years) but
they do tend to recover completely. However, repeated 732 Muscle biopsy showing denervation atrophy of groups of muscle
episodes do occur and may result in some degree of residual fibers due to axonal degeneration.There is a reduction in size of
deficit (e.g. foot drop) and absent tendon reflexes. muscle fibers within the denervated motor units (and enlargement of
intact motor units due to collateral innervation).
Guillain–Barré Syndrome (GBS) 589

ETIOLOGY AND PATHOPHYSIOLOGY CLINICAL FEATURES


An autoimmune disorder triggered by exposure to an Acute inflammatory demyelinating
antigen that is usually infectious. polyradiculoneuropathy (AIDP) (85–90% of cases)
• Progressive symmetric weakness of the limbs which
Antecedent exposure to an antigen develops acutely (within days) or subacutely (up to
Infection 4 weeks), and progresses over a period of 1–8 weeks in
50–60% of cases give a history of a recent viral or other an ascending fashion (caudal to rostral), reaching a
infection. plateau, and then spontaneously resolving.
• Paresthesiae in the hands and feet: not as prominent as
Bacteria: motor signs.
• Campylobacter jejuni: the most frequent antecedent • Back pain (30% of cases).
pathogen (26–41% of cases). A gastrointestinal pathogen • Depressed or absent deep tendon reflexes.
which causes gastroenteritis and diarrhea. Associated with • Cranial neuropathies, particularly facial neuropathies:
the Miller Fisher variant and an acute motor-axonal (50% of cases).
neuropathy variant of GBS (see below). • Progression of disability for up to 4 weeks.
• Mycoplasma pneumoniae.
The common initial symptoms are numbness and tingling
Viruses: in the lower limbs, which are soon followed by weakness in
• Cytomegalovirus (CMV): 10–22% of cases of GBS, the legs and arms, which may be proximal as well as distal
particularly young women presenting with prominent and sometimes more pronounced in the proximal muscles.
involvement of the sensory and cranial nerves. Many About a half of patients develop cranial neuropathies, usually
have high serum titers of antibodies reacting with GMs bilateral facial paresis (of lower motor neuron type). Ptosis
gangliosides and with sulfated glycolipids. and double vision occur in about 10% of cases. Back pain is
• Epstein–Barr virus (EBV). a major symptom in about one-third of patients. Sensation
• Varicella-zoster virus. is usually impaired in a glove and stocking distribution but
• HIV: a GBS-like syndrome most commonly occurs profound sensory loss is uncommon. Sphincter disturbances
around the time of seroconversion, but the CSF has a are present in only a small proportion of cases and suggest
lymphocytic pleocytosis. another diagnosis such as spinal cord compression.

Vaccination: DIFFERENTIAL DIAGNOSIS


• Rabies. Anxiety or hysteria
• Swine influenza. In the early stages of the illness there may be few objective
neurologic signs and patients may be misdiagnosed as being
Autoimmune response anxious or hysterical, and sent home, only to soon become
The infecting organism induces humoral immune responses very weak with respiratory paralysis.
(e.g. production of antibodies to GM1, GM2 and GQ1b)
and cellular immune responses that, because of the sharing Spinal cord lesions
of homologous epitopes (molecular mimicry) between the Transverse myelitis.
lipopolysaccharides of the infecting organism and the surface
components of peripheral nerves (e.g. ganglioside), cross- Anterior horn cell lesions (part of spinal cord)
react with surface components of peripheral nerves. Poliomyelitis: caused by an enterovirus (like GBS is
Subsequent pathogenic mechanisms are heterogeneous. predominantly caused by an enteral agent).
The humoral immune response results in complement
activation on the outer Schwann cell plasmalemma, which Peripheral nerve disorders
leads to entry of calcium into the cell with subsequent • Vasculitis.
activation of PLA and protease, resulting in demyelination. • HIV infection: may cause a syndrome like GBS but the
The cellular immune response involves macrophages and T CSF white cell count is usually elevated (>30 × 106/l).
cells which attack healthy myelin in peripheral and cranial • Diphtheria: more likely to have a bulbar onset, lead to
nerves, resulting in block in conduction of nerve impulses. respiratory failure, evolve more slowly, take a biphasic
However, the immune responses are more complex, and course and cause death or long term disability.
host factors contribute substantially to disease susceptibility • Lyme disease.
and to the clinical pattern of GBS. • Lymphoma.
Immune reactions against target epitopes in Schwann cell • Carcinoma of the lung.
surface membrane or myelin result in acute inflammatory • Diabetes.
demyelinating polyradiculoneuropathy (85% of cases), and • Acute intermittent porphyria.
reactions against epitopes contained in the axonal • Heavy metals and other toxins.
membrane cause acute axonal forms of GBS (15% of cases). • Alcohol-related acute axonal neuropathy (in combination
with malnutrition).
• Vitamin B12 deficiency.
590 Diseases of the Peripheral Nerve

Neuromuscular junction disorders Acute motor axonal neuropathy (AMAN)


• Myasthenia gravis. (10–20% of cases of GBS)
• Botulism. • Originally described during summer epidemics in
children of farm families in northern China.
Muscle disorders • Antecedent enteric Campylobacter jejuni infection
• Hypokalemia. (heterostrain 019) in 80% of cases.
• Periodic paralysis. • IgG and activated complement components bind to the
• Polymyositis. axolemma at nodes of Ranvier in large motor fibers.
Macrophages are attracted to these nodes and track
INVESTIGATIONS underneath the detached myelin lamellae along the
• Vital capacity: should be measured every 2–4 hours in periaxonal space, dissecting the axon from the overlying
the initial stages. Schwann cell and compact myelin. The axolemma, in
• Nerve conduction studies and EMG: contact with invading macrophages, is focally destroyed,
– Demyelination: slowing of nerve conduction velocities, resulting in selective loss of motor axons distally or along
prolongation of distal and F-wave latencies, and the entire length due to axonal degeneration. Distal
conduction block. sensory fibers are intact.
– Axonal degeneration: reduction in amplitude of sensory • Little or no demyelination.
nerve action potentials (SNAP) and compound muscle • Little or no lymphocyte infiltration.
action potentials (CMAP), EMG evidence of • Normal SNAP and CMAP.
denervation. • Normal conduction velocities and latencies.
• Lumbar puncture: CSF white cell count is normal and • Early denervation.
protein elevated. • High titers of antibodies to GM1, GD1a and GD1b which
• Full blood count. parallel the clinical course.
• ESR. • The variable severity of the axonal destruction is reflected
• Serum electrolytes, urea and creatinine. by the variable time span of recovery, and overall good
• Blood glucose. prognosis in many children.
• Serology for Campylobacter jejuni (serotypes HS-2, 4 • Reasonable prognosis, similar to AIDP, in contrast to
and 19), Mycoplasma pneumoniae, CMV, EBV, varicella- AMSAN: mortality 4%.
zoster, HIV, Lyme disease.
• Stool culture (Campylobacter jejuni): may be positive for Miller Fisher syndrome (MFS)
several weeks after the end of the diarrheal illness. • Ophthalmoparesis.
• Ataxia.
VARIANTS • Areflexia.
AIDP is the most common, and traditional, condition • No significant weakness or sensory disturbance.
causing GBS. But recently there have been described other • Antibodies to GQ1b ganglioside in 96% of cases of MFS
conditions which also cause, or fall within, GBS (e.g. (the only form of GBS where a serum test has sensitivity
AMSAN, AMAN, MFS). and specificity) which parallel the disease course.
• The antibodies recognize epitopes that are expressed
Acute motor-sensory axonal neuropathy (AMSAN) specifically in the nodal regions of oculomotor nerves,
• Abrupt onset of severe generalized paralysis and muscle but also in dorsal-root ganglion cells and cerebellar
atrophy. neurons.
• Association with antecedent diarrheal or flu-like illness. • Antecedent infection sometimes: e.g. Campylobacter spp.
• Acute axonal degeneration of motor and sensory nerve
fibers, extending to proximal nerve roots, due to a Acute sensory loss with absent reflexes
primary attack on axons, rather than Schwann cells; scant • A predominantly sensory AIDP.
lymphocytes and little evidence of demyelination.
• Low amplitude or absent SNAP and CMAP on distal Acute autonomic neuropathy (acute
supramaximal stimulation. pandysautonomia)
• Antiganglioside antibodies (i.e. anti-GM1) are common. • Postural hypotension.
• Rapid progression. • Impaired sweating, lacrimation, bladder and bowel
• Delayed and very poor recovery. function.

Chronic inflammatory demyelinating


polyneuropathy (see p.593)
• Similar pathogenesis to AIDP.
• Slower onset, over weeks or months.
• Course may be relapsing and remitting, or progressive.
• Abnormal nerve conduction studies.
• Responds to treatment with corticosteroids, immuno-
suppressive agents, plasmapheresis and intravenous
immunoglobulin.
Guillain–Barré Syndrome (GBS) 591

DIAGNOSIS • Exchange 200–250 ml plasma/kg total in 4–5 exchanges


Diagnostic criteria for typical GBS over a 7–14-day period.
Features required for diagnosis • Restore intravascular volume with albumin or artificial
• Progressive weakness in both arms and both legs. plasma solution.
• Areflexia. • Wait for a response, which may take 3–4 weeks.

Features strongly supporting diagnosis Intravenous human immunoglobulin:


• Progression of symptoms over days to 4 weeks. • 0.4 g/kg daily for 5 days is equally effective as plasma-
• Relative symmetry of symptoms. pheresis, and is safer and more readily administered, but
• Mild sensory symptoms or signs. is more expensive and difficult to access. Again, wait for
• Cranial nerve involvement, especially bilateral weakness a response.
of facial muscles.
• Recovery beginning 2–4 weeks after progression ceases. Corticosteroids and immunosuppressive drugs:
• Autonomic dysfunction. • No evidence of benefit.
• Absence of fever at onset.
• High concentration of protein in CSF, with fewer than If intravenous immunoglobulin (IVIG) or plasma exchange
10 leucocytes/mm3. are not successful, consider:
• Typical electrodiagnostic features. • Repeating the IVIG (but not often: ‘don’t throw good
money after bad’).
Features excluding diagnosis • Steroids after IVIG (but not steroids alone).
• Diagnosis of botulism, myasthenia, poliomyelitis, or toxic • ?Tryptophan staphylococcal exchange column.
neuropathy. • Do not use sequential plasma exchange and IVIG; the
• Abnormal porphyrin metabolism. combination is as effective as either alone but is not
• Recent diphtheria. superior to either alone.
• Purely sensory syndrome, without weakness.
About 10% of responders have a limited relapse after
TREATMENT either treatment with IVIG or plasma exchange. At the time
The key to management is to anticipate complications it is not known which of these patients will go on to develop
before they occur, and implement appropriate prevention CIDP (see p.593) or not. Firstly, re-treat them with the
strategies. previously effective IVIG or plasma exchange. If this is not
effective, consider steroids (?CIDP).
General
• Careful nursing, particularly attention to the patient’s Intensive care
vital signs, fluid and nutritional status, comfort (e.g. Problems
pain), emotional status, and care of the trachea, pharynx, • Respiratory failure: 25% (reduced from 30% by IVIG and
mouth, eyes, skin, bladder and bowels. plasma exchange).
• Tracheostomy or intubation and artificial ventilation may • Cardiovascular dysautonomias
be necessary. • Medical complications:
• Feeding by nasogastric tube, percutaneous gastrostomy – Pneumonia: 25% in ICU patients.
tube, or intravenous routes may be required. – Urinary tract infection: 18%.
• Bladder and bowel infections require prompt treatment. – Hyponatremia: 14%.
• Deep vein thrombosis prophylaxis: subcutaneous heparin – Pulmonary embolism.
5000 U bd. – Gastrointestinal bleeding: 5%.
• Physiotherapy should begin immediately. – Gut perforation: 3%.
• Splints to prevent foot- and wrist-drop may be required. – Hypercalcemia (after third week): 3%.
• Occupational therapy. – Sepsis/shock: 1%.
• Speech and swallowing therapy. • Iatrogenic: tube and line insertion: pneumothorax: 3%.
• Psychologic support and counselling may be necessary, • Patient comfort and communication.
particularly for patients on ventilatory support.
• Social worker. Respiratory failure
• GBS support groups. • The best measure of diaphragmatic power is mid-
inspiratory flow rate.
Specific • Vital capacity is a poor measure of diaphragmatic power;
Plasma exchange (PE) and high-dose IV immunoglobulin it is a volumetric measure and thus an epiphenomenon.
(IVIG) improve the rate of motor recovery in at least 60% • Vital capacity (VC):
of patients. They are of equivalent efficacy, and there is no – 30 ml/kg: poor cough, poor airway clearance.
additional benefit from combining the treatments consecu- – 20 ml/kg (about 1.5 l): compromise of the sigh
tively. CSF filtration is proposed as a new treatment for mechanism (yawn) and reduced tidal volume; alveoli at
GBS, which may also be effective when combined with PE peripheral and base of lungs collapse, resulting in pul-
or IVIG; future studies are needed. monary arteriovenous shunting, ventilation/perfusion
mismatch, mild hypoxia, and stimulus to increase the
Plasmapheresis (plasma exchange): respiratory rate.
• Start as soon as possible, certainly within the first – 12–15 ml/kg: loss of sigh; intubate and ventilate.
2 weeks. – 10 ml/kg: hypoventilation and carbon dioxide retention.
592 Diseases of the Peripheral Nerve

Dysautonomia CLINICAL COURSE AND PROGNOSIS


• Urinary retention in 15–20% of cases (do not confuse Course
with a spinal cord lesion). • The interval from onset to peak disability may vary from
• Cardiovascular: hours to weeks.
– Pulse: no beat-to-beat variation; vagotomized. • About 30% reach their maximum deficit within 7 days;
– Arrhythmias: others progress for up to 4 weeks.
Tachycardia (5–30%; differential diagnosis of a fixed • About 60% of cases are unable to walk at the height of
tachycardia is a pericardial effusion; the onset of tachy- their illness.
cardia in third week: suspect pulmonary embolism). • Respiratory function is impaired in about half of patients,
Bradycardia (2–10%; often with suctioning and eye ball and about 20–30% require assisted ventilation.
pressure).
Asystole (1%). Recovery
Ventricular arrhythmias. • One-third of patients begin to show signs of recovery
– Blood pressure may swing wildly (e.g. from 8 to within 2 weeks.
26.7 kPa [60 to 200 mmHg] systolic), and leads to • One-third begin to recover in the second to fourth
cardiac arrest. This is due to a centrally mediated vaso- weeks.
depressor response leading to sympathetic vasodilatation • One-third may have to wait up to 3 months before
and hypotension. definite improvement is evident.

Hyponatremia Outcome
Due to a natriuresis as a result of pulse secretion of atrial • Good recovery (includes paresthesiae, mild weakness):
natriuretic factor. The patient loses water as well as salt, so 80%.
do not restrict fluids. • Unsteady gait with or without orthosis: 5%.
• Walk with callipers: 5%.
Pulmonary embolism • Wheelchair-bound: 3%.
• Tends to occur after 2–3 weeks. • Chronic or relapsing course: 3%.
• Completely avoidable. • Mortality: 5%.
• Intermittent pneumatic compression air boots are most
effective. Mortality
• Subcutaneous heparin may lose its effectiveness after Most common causes of death: complications of respiratory
about 8–10 days unless the dose is increased (e.g. failure, pulmonary embolism, cardiac arrhythmias,
doubled). Check the APTT about 40 minutes after the autonomic failure, infection.
heparin dose, and if not twice normal, then increase the
dose of heparin. Prognostic factors
• DVTs most commonly arise in pelvic veins. • Advanced age.
• Can be diagnosed with plethysmography. • Axonal injury (electrophysiologic evidence).
• Mechanical ventilation for 3 months: the patient will be
Ileus wheelchair bound.
• Suspect if abdominal pain.
• May lead to perforation and peritonitis.
• Ranitidine and cissapride (cisapride) promote gut
motility.

Iatrogenic
Beware sensory deafferentation and motor weakness when
inserting a nasogastric tube: patients may not cough if the
tube goes into bronchi, alveoli, or even through the lung
into pleural cavity and mediastinum. Always perform an x-
ray after inserting a nasogastric tube and before feeding;
deaths have occurred by feeding into the mediastinum!

General treatment
• Early intubation: don’t wait until the patient is breathless;
intubate when VC is about 12 ml/kg.
• Treat severe pain with epidural analgesics (or try
narcotics or high dose i.v. steroids).
• ICU techniques for infection surveillance and DVT
prophylaxis.
• Nutrition, skin and eye care.
• Communication and psychologic support: to
communicate with mute patients, use a transparent
plexiglass with grid and letters, and phrases such as
‘please turn me’, ‘suction me’, ‘I’m in pain’; and watch
the patient’s eyes as they look at these phrases.
Chronic Inflammatory Demyelinating Polyneuropathy (CIDP) 593

CHRONIC INFLAMMATORY occurrence of fasciculations (see pp.534, 538).


DEMYELINATING POLYNEUROPATHY • Serum IgM anti-GM1 ganglioside antibodies are present
(CIDP) in about 40% of patients. The role of anti-GM1
antibodies remains obscure because they are neither
sensitive nor specific to the disease. Alone they do not
DEFINITION mediate conduction block or block sodium channels.
A subacute or chronic demyelinating polyneuropathy with • The diagnostic hallmark is the electrophysiologic
a chronic relapsing-remitting, monophasic or progressive demonstration of multiple, persistent localized sites of
course, lasting months to years. motor conduction block (reduction in amplitude and
area of compound muscle action potential by >50%,
EPIDEMIOLOGY across the block), but sensory conduction remains
• Incidence: 0.15 per 100 000 per year (crude). normal in the same nerve segments.
• Prevalence: 2 per 100 000. • There have been no controlled trials of therapy but many
• Age: any age, mean and median age of onset: about 50 open studies have reported benefit from intravenous
years. immunoglobulin (IVIG) and from oral or intravenous
• Gender: M≥F. cyclophosphamide therapy.
• IVIG, 2 g/kg (1.0 g/kg on 2 consecutive days, or
PATHOLOGY 2.0 g/kg in 24 hours), is the preferred initial treatment.
Endoneurial inflammation and primary demyelination of spinal It appears to induce short term improvement in up to
roots and peripheral nerves without infection or vasculitis. 80% of patients, but treatment usually needs to be
repeated monthly (which is expensive). Long term IVIG
ETIOLOGY AND PATHOPHYSIOLOGY is effective in maintaining improvement in about 60% of
• Probably an autoimmune response caused by both cases, but it does not eradicate the disease. Prognostic
cellular and humoral immune processes: factors for a response to immunoglobulin treatment
– Associated with an increased frequency of HLA DR3. include lower age at onset, lower number of affected
– Target antigens remain elusive. limb regions, creative kinase level <180 U/l, elevated
• Macrophages penetrate the Schwann cell basement anti-GM1 antibodies, and definite conduction block.
membrane, ingest the myelin sheath and denude the • Oral cyclophosphamide (1–3 mg/kg/day) may help to
axon. If the inflammation is severe, the axons may induce a sustained remission but evidence of efficacy is
undergo ‘bystander’ degeneration. lacking and its adverse effects are significant. It should,
therefore, be reserved for patients who require frequent
CLINICAL FEATURES IVIG infusions to maintain improvement. However,
• Typically subacute onset and progressive (over >8 weeks) because of the very high cost of IVIG, in practice, several
asymmetric weakness and/or numbness of the distal and patients are treated with cyclophosphamide.
proximal limbs, with pain, sensory ataxia, and areflexia. • The course is slowly progressive.
• Occasionally patients present with nerve root pain, a pure
motor syndrome, and ataxic sensory variant, with Sensory ataxic CIDP
mononeuritis multiplex, involvement of lower limbs only
or one-limb only, or with repeated episodes of acute Pure motor CIDP
Guillain–Barré syndrome.
• Limb or back pain is not uncommon at presentation or DIFFERENTIAL DIAGNOSIS
relapse. Chronic autoimmune inflammatory neuropathies
• A postural tremor occasionally occurs. • Chronic inflammatory demyelinating polyneuropathy.
• It is rarely severe enough to require ventilatory support. • Multifocal motor neuropathy with conduction block.
• Paraproteinemic demyelinating neuropathy.
CLINICAL VARIANTS • Chronic relapsing axonal neuropathy.
Multifocal motor neuropathy with conduction block
• An uncommon demyelinating motor neuropathy. Chronic inflammatory neuropathies are distinguished
• Originally described as a subgroup of CIDP and some from other chronic neuropathies by reduced or absent
authorities still regard it as such, whereas others argue reflexes, raised CSF protein, usually demyelinating
that it is a distinct entity. neurophysiology and lack of a family history.
• Predilection for young adults.
• Immune-mediated. Other chronic inflammatory neuropathies
• Clinically it is characterized by slowly progressive, patchy, • Vasculitis/connective tissue disease (axonal) (see p.597).
asymmetric limb weakness, usually beginning and • Infection (axonal, demyelinating): leprosy, herpes zoster,
becoming most prominent in the forearms but the lower HIV.
limbs may be affected (usually later).
• Weakness may be associated with cramps, muscle Toxins
wasting, fasciculations, decreased tendon reflexes and • Alcohol (axonal).
sometimes minor sensory symptoms. • Chemicals (axonal): acrylamide, carbon disulfide, heavy
• The clinical features are often confined to the territory metals.
of individual nerves. • Drugs: amiodarone (demyelinating); dapsone, disulfiram,
• Clinical misdiagnosis as motor neuron disease is possible isoniazid, metronidazole, nitrofurantoin, phenytoin,
because of the predominant motor phenotype and platinum, vincristine (axonal).
594 Diseases of the Peripheral Nerve

Metabolic (axonal) DIAGNOSIS


• Diabetes mellitus. Diagnostic criteria
• Acromegaly. • Symptomatic motor or sensory demyelinating polyradicu-
• Myxedema. loneuropathy of presumed autoimmune origin, causing
• Uremia. progressive or relapsing weakness of more than one limb.
• Onset extending over 2 months or more (progression
Nutritional deficiencies (axonal) over 2 months distinguishes CIDP from the acute
• Vitamin B12. demyelinating neuropathy of Guillain–Barré syndrome).
• Pyridoxine. • Electrophysiologic (slowing or block of nerve
• Thiamine. conduction) or morphologic features of demyelinating
• Niacin. or demyelinated nerve fibers on sural nerve biopsy.
• Folic acid. • No detectable alternative cause of neuropathy (e.g.
exclusion of monoclonal gammopathy; HIV infection;
Paraneoplastic (axonal, demyelinating) hereditary, metabolic and paraneoplastic disorders; and
• Carcinoma. ischemic neuropathy).
• Polycythemia rubra vera.
TREATMENT
Miscellaneous (axonal) First line
• Primary amyloidosis. • Prednisolone or IVIG is the recommended initial
• Celiac disease. treatment. A recently published trial comparing oral
• Primary biliary cirrhosis. prednisolone (tapering from 60 mg to 10 mg daily over
6 weeks) with IVIG (2.0 g/kg given over 1–2 days)
Hereditary neuropathy (HMSN type 1) showed slightly, but not significantly, more improvement
• HMSN type 1 positive genetic test. after IVIG than prednisolone; the mean difference
between the groups in change in disability grade, being
CIDP is distinguished from HMSN type 1 by: 0.16 (95% CI: –0.35–0.66) (Hughes et al., 2001). For
• Ability to date the onset of symptoms. patients who respond to their first course of treatment
• Positive sensory symptoms (e.g. tingling). and do not require prolonged medication, IVIG is
• No pes cavus. associated with slightly more short term efficacy, but
• Palpable nerve thickening (e.g. greater auricular nerve). slightly more short term adverse effects than oral steroids
• Proximal and distal weakness. (i.e. the choice of treatment is evenly balanced). For
• Raised CSF protein. patients who require long term treatment, IVIG may be
• Asymmetric clinical and electrophysiologic findings. preferable because of the known serious adverse effects
of prolonged steroid treatment.
INVESTIGATIONS • Steroids (e.g. prednisolone 120 mg alternate days,
CSF reducing after 2 weeks and over 3 months; or
• White cell count: normal. prednisolone 60 mg each morning [mane] for 2 weeks,
• Protein: elevated in 50%. 40 mg mane for 1 week, 30 mg mane for 1 week, 20 mg
mane for 1 week, and then 10 mg mane for 1 week) are
Nerve conduction studies usually effective. Osteoporosis prophylaxis should be
Slowed conduction, particularly in motor nerves, and there may considered.
be variable degrees of multifocal conduction block. In chronic • IVIG 0.4 g/kg/day for 5 days was effective in about
and severe cases, the superimposition of axonal degeneration two-thirds of patients in a randomized, double-blind,
may make the distinction from axonal neuropathy difficult. placebo-controlled study, but is expensive, invasive and
there are theoretical concerns with its long term safety
Sural nerve biopsy (733) (i.e. transmission of infectious agents, as it is a human
• Indicated only if the diagnosis is uncertain, because blood product). Other regimes (e.g. IVIG on days 1, 2
unpleasant dysesthesiae may result as an adverse effect. and 21 in a dose of 1g/kg) have also been shown in trials
• Changes are non-specific: chronic demyelination with to be more effective than placebo. Some patients with
relatively little inflammatory infiltrate, or sometimes chronic progressive disease have a long lasting remission
axonal change from Wallerian degeneration. after a single course of treatment and patients with
• Increased numbers of macrophages express the inflam- relapsing disease can be maintained and stabilized by a
mation-associated antigens MRP14, 25F9 and MRP8. single treatment before the expected relapse. Other
patients need to be treated every 2–12 weeks. The
MRI of cervical and lumbosacral regions precise mechanisms of IVIG action in CIDP are not
• May show nerve hypertrophy, sometimes of a massive known. Anti-idiotypic neutralization of antibodies,
degree. binding of complement, and blockade of macrophages
• Increased signal intensity on T2W images and nerve may prevent the ongoing inflammatory demyelination.
enlargement correlate with demyelinative foci, and gado- • Plasma exchange (10 exchanges over 4 weeks), is
linium enhancement with disease activity (progression or effective in about 80% of newly diagnosed patients, but
relapse), emphasizing the influence of the blood–nerve most require long term immunosuppression (usually
barrier. Where the barrier is deficient, significant nerve azathioprine with or without steroids) for stabilization.
hypertrophy is not uncommon suggesting a continuing • Plasma exchange and IVIG probably confer equal benefit
process of demyelination and remyelination. in the short term.
Neuralgic Amyotrophy 595

Second line NEURALGIC AMYOTROPHY


• Azathioprine, cyclophosphamide, cyclosporine A, and
interferon alpha-2a have not been tested in randomized
controlled trials, but have been reported to be effective DEFINITION
in some patients. A vague and incompletely understood condition which
• Azathioprine or cyclosporine may sometimes be effective affects the brachial plexus on one side, and usually on only
in resistant cases. Cyclosporine A may be effective in one occasion in life. Many variants exist, such as
small maintenance doses of <5 mg/kg, which may avoid involvement of separate nerves, cranial nerves, and even the
the most serious complication of cyclosporine A, that of lumbosacral plexus; and hereditary and recurrent forms. It
dose-dependent reversible nephrotoxicity. Some patients is sometimes called the ‘Parsonage Turner syndrome’,
have entered long lasting remissions following its use for following its original description in 1948 by Parsonage and
a limited period. Turner, and ‘brachial plexus neuropathy’.
• Interferon alpha-2a and interferon-β1a may provide an
alternative therapy for patients who do not respond to EPIDEMIOLOGY
first line therapies but remains to be evaluated in • Incidence: 1.64 per 100 000 per year.
controlled trials. • Age: any age.
• Combinations of steroids and immunoglobulin or • Gender: M=F.
another immunosuppressive agent are commonly used
empirically. PATHOLOGY
Uncertain. Abundant, multifocal mononuclear inflam-
N.B. If treatment fails, the diagnosis should be re- matory cell infiltrates were seen in biopsies of the brachial
evaluated and a paraprotein re-sought. plexus in four patients with typical clinical features, but in
whom there was also focal enlargement or enhancement of
PROGNOSIS lesions on CT or MRI scans of the plexus, or a severe or
• About 80% of patients respond to treatment, which may progressive course suggesting an alternative possible
need to be continued for many years. malignant disorder.
• About 13% of patients deteriorate sufficiently to become
permanently dependent, bed-bound or chair-bound. ETIOLOGY AND PATHOPHYSIOLOGY
About 87% of prevalent patients are able to walk without • Unknown; possibly an autoimmune reaction to
walking aids or other assistance, and 54% are still being preceding immunization or infection (high levels of
treated. complement-fixing antibodies against peripheral nerve
• About half of patients have a relapsing remitting course. myelin have been detected in the acute phase of the
• The mean duration of the disease amongst prevalent illness in three patients, which fell during subsequent
cases is about 7 years (median 5 years). recovery, compared with 25 controls).
• Most often arises in healthy individuals with no
antecedent events.
• Rarely, inherited as an autosomal dominant disorder
which is heterogeneous genetically (i.e. not all map to
chromosome 17q25).
• Several potential causal factors have been implicated but
remain unproven.

Injection with foreign material


• Preceding vaccination, particularly against tetanus.
• Botulinum toxin A injections.

Infection
733 • Parvovirus B19 ± CMV.
• Salmonella.
• Leptospirosis.
• Yersiniosis.

Other
• Surgery.
• Parturition.
• Strenuous exercise.

PATHOGENESIS
Focal conduction block plays a significant part in the
pathogenesis of neuralgic amyotrophy, which is generally
regarded as an axon loss process.

733 Sural nerve biopsy showing multiple regions of segmental


demyelination.
596 Diseases of the Peripheral Nerve

CLINICAL FEATURES • Episodes may be precipitated by pregnancy and non-


History specific illness, and tend to recur.
• Usually healthy prior to onset. • Congenital anomalies, such as syndactyly and hypotelor-
• Pain in the shoulder and neck: ism, are common in affected members of these families.
– Onset quite sudden, and often in the evening or night.
– Typically severe, deep and sometimes excruciating. Recurrent episodes of neuralgic amyotrophy
Rarely mild. Rare.
– May radiate to the medial part of the scapula or down
the arm to involve the whole arm or part of the upper DIFFERENTIAL DIAGNOSIS
arm or forearm. • Guillain–Barré syndrome: usually involves the legs first,
– Lasts from a few hours to a few weeks; usually several days. and pain is not commonly severe and prominent.
The pain may linger as a dull ache for some time, however. • Poliomyelitis: the CSF is usually abnormal (unlike
• Mild numbness may be present over the deltoid muscle neuralgic amyotrophy).
in the rough distribution of the axillary nerve; • Rucksack paralysis: direct mechanical injury to the
paresthesiae occur rarely. brachial plexus from wearing a heavy back pack.
• Severe, and often unilateral, weakness of the muscles of • Compression of the brachial plexus during sleep: unlikely
the shoulder, arm or both develops over a few hours, to cause severe pain.
frequently overnight, during the pain or more commonly • Isolated lesions of the long thoracic nerve to serratus
just after it has mainly subsided. The weakness is maximal anterior: need to exclude a local cause.
soon after onset, but occasionally it progresses. • Radiation therapy: may cause painful lesions of the
brachial plexus but usually a relevant history and scarring,
Examination and slower onset.
• Early wasting and flaccid weakness of one or any • Multifocal motor neuropathy: bilateral, but asymmetric,
combination of muscles of the shoulder and arm: serratus painless, and slower in onset.
anterior, rhomboids, supra- and infraspinatus, deltoid, • Lead poisoning: may cause bilateral brachial neuropathy
latissimus dorsi, or one or more arm muscles innervated but usually painless.
by major nerves or their branches (734, 735).
• Normal, depressed or absent deep tendon reflexes of the arm. INVESTIGATIONS
• Sensory loss is less apparent and, if present, is usually in Nerve conduction studies
the area of innervation of the axillary nerve. • Motor conduction velocity to proximal muscles may be slow.
• Sensory nerve action potentials may be reduced.
CLINICAL VARIANTS
Involvement of single nerves or their branches
rather than a large part of the brachial plexus 734
These nerves usually have their origin in the brachial plexus,
e.g.:
• Long thoracic nerve to serratus anterior.
• Suprascapular nerve.
• Axillary nerve.
• Anterior interosseous nerve.
• Lateral antebrachial cutaneous nerve.
• Median nerve trunk.
• Median palmar cutaneous branch.

In these cases, it has been proposed that the pathology


is probably within the corresponding fascicle of the brachial
plexus rather than a separate trunk of the respective nerve.
However, this would not explain occasional cases with 735
involvement of nerves remote from the brachial plexus such
as the lower cranial nerves (facial nerve, vagus nerve,
recurrent laryngeal nerve, accessory nerve, and cranial
nerves IX, X, XI and XII combined) and the phrenic nerve.

Bilateral involvement
Occurs in about 5–10%, but up to one-third, of cases.

Lumbosacral plexus involvement


Rare.

Familial neuralgic amyotrophy


• Inherited as an autosomal dominant trait. 734, 735 Winging of the right scapula due to neuralgic amyotrophy of
• Associated with a mutation in distal chromosome 17q. the brachial plexus causing weakness of the serratus anterior (supplied
• Genetically distinct from hereditary neuropathy with by the long thoracic nerve). (Courtesy of Dr AM Chancellor,
liability to pressure palsies. Neurologist,Tauranga, New Zealand.)
Vasculitic Neuropathy 597

Needle EMG • Polyarteritis nodosa: affects medium-sized arteries (e.g.


Usually reveals normal paraspinal muscles and multifocal cerebral, gastrointestinal, renal), and causes peripheral
axonal lesions within the brachial plexus or its branches, but neuropathy in about half of patients, usually as a
can be consistent with an isolated lesion of a peripheral nerve. mononeuritis multiplex or symmetric polyneuropathy.
Presents at any age and is twice as common in men than
MRI brachial plexus women. It is characterized commonly by malaise, fever,
If a local compressive or infiltrative cause is suspected (e.g. weight loss, and tachycardia; and may be associated with
neuroma). an acute abdomen, vascular event of the brain or heart,
or severe or treated hypertension. Anti-neutrophic
DIAGNOSIS cytoplasmic antibodies are rare, and when present, may
Typical clinical picture of rather sudden onset of neuropathic indicate coincident vasculitis of small vessels, and may be
shoulder pain and weakness, supported by EMG studies, and associated with a lupus anticoagulant and alpha1-
followed by spontaneous resolution of pain and gradual, but antitrypsin deficiency.
often incomplete, resolution of weakness. • Wegener’s granulomatosis: a systemic necrotizing granu-
lomatous vasculitis of the upper and lower respiratory
TREATMENT tract, with or without glomerulonephritis or arthralgia.
• Physiotherapy, particularly passive (and later active) exercises Multiple mononeuropathy or polyneuropathy may occur,
of the shoulder joint for patients with impaired shoulder as may cranial neuropathies. Anti-neutrophil cytoplasmic
elevation, to prevent a ‘frozen shoulder’ syndrome. antibody is positive in 90% of cases.
• No effective medical treatment; no convincing evidence • Giant cell arteritis (see p.234): may be complicated by
for corticosteroids. multiple mononeuropathy, as well as mononeuropathy
• Therapeutic interventions should perhaps be directed to and polyneuropathy.
patients with persistent conduction block with the aim
of eradicating the block and possibly minimizing Vasculitis associated with systemic
subsequent axon loss. Intravenous immunoglobulin is autoimmune disease
worthy of future study. • Rheumatoid arthritis: vasculitis may affect small arteries,
arterioles, capillaries, and venules, and often causes few
PROGNOSIS symptoms. When present, it is associated with long-standing
Pain usually resolves within 1–2 weeks but weakness may not disease, seropositivity, and florid subcutaneous nodule
recover for months or even years. In one series of 99 patients, formation. Polyneuropathy, and particularly multiple
one-third had made a functional recovery after one year, mononeuropathy, are rare. More common causes are joint
three-quarters after 2 years, and 89% at 3 years. Weakness of deformities and synovial swellings causing entrapment
the shoulder muscles had recovered by the end of 1 year in neuropathies, and medication-induced neuropathy.
60% of cases whereas no patient with predominantly lower • Sjögren’s syndrome.
plexus involvement recovered normal function. • Systemic lupus erythematosus: occasionally presents with
a multiple mononeuropathy or chronic inflammatory
demyelinating polyneuropathy.

VASCULITIC NEUROPATHY Vasculitis associated with other conditions


• Cryoglobulinemia, particularly mixed (often hepatitis C
positive).
DEFINITION • Behçet’s disease: a small vessel vasculitis with mono-
Ischemia and infarction of one or more peripheral nerves nuclear cell infiltrate in the skin, gastrointestinal ulcers
due to vasculitis of the vasa nervorum. and the CNS (occasionally), characterized by relapsing
ocular lesions and recurrent oral and genital ulcers,
EPIDEMIOLOGY meningo-encephalitis, and rarely peripheral neuropathy,
• Rare. usually in the form of multiple mononeuropathy.
• Age: any age. • Human immunodeficiency virus (HIV) infection.
• Gender: M=F. • CREST syndrome (calcinosis, Raynaud’s phenomenon,
esophageal dysmotility, sclerodactyly and telangiectasis).
PATHOLOGY • Non-systemic vasculitis of peripheral nerves.
• Vasculitis of the vasa nervorum (50–300 μm diameter). • Angiitis associated with amphetamine use.
• Ischemia and infarction of one or more peripheral nerves. • Paraneoplastic vasculitis: lymphoma, small cell lung
carcinoma.
ETIOLOGY
Primary vasculitis
• Allergic granulomatosis and angiitis (Churg–Strauss
syndrome): a granulomatous necrotizing vasculitis which
predominantly affects the lungs. A prodromal phase of
asthma and eosinophilia commonly precedes the onset
of systemic vasculitis, which may involve one or more
peripheral nerves. Other features include vasculitic rash,
glomerulonephritis and positive anti-neutrophil cyto-
plasmic antibody in 60–70%.
598 Diseases of the Peripheral Nerve

CLINICAL FEATURES Mass lesions


• Depend on the size and extent of the nerves involved: Suspect if the affected nerves lies close together.
– Large nerve infarction: multiple mononeuropathy.
– Smaller nerve infarction: asymmetric sensorimotor INVESTIGATIONS
neuropathy. Nerve conduction studies
– Nerve ischemia: distal symmetric sensorimotor neuropathy. Normal or mildly decreased nerve conduction velocities,
• Onset is typically rapid over several weeks. compound muscle action potentials, and sensory nerve
• Pain is prominent: deep, aching and neuropathic. action potentials in the involved nerves. Conduction block
• Symptoms and signs of the underlying disease may or in a few cases.
may not accompany the neurologic deficits.
• Multiple mononeuropathy: EMG
– Several peripheral or cranial nerves become involved, one Typical changes of an axonal neuropathy (e.g. denervation
after another, within days or weeks. and reinnervation) in most cases.
– Pain, paresthesia, loss of sensation, and muscle weakness
in the dermatomes and myotomes supplied by the Blood tests
diseased nerve(s). • ESR
– As more nerves become involved, the clinical picture • Antinuclear antibodies.
resembles a polyneuropathy. • Antineutrophic cytoplasmic antibodies.
• Symmetric polyneuropathy: in polyarteritis nodosa and • Rheumatoid factor.
Wegener’s granulomatosis this pattern is as common as
multiple mononeuropathy. Tissue biopsy (muscle or sural nerve)
A nerve biopsy (736–738) is essential as a basis for long
DIFFERENTIAL DIAGNOSIS term immunosuppression. It is positive in 90% of cases of
Multiple mononeuropathy vasculitis if the nerve is abnormal neurophysiologically. An
Hematologic disorders additional muscle biopsy increases the diagnostic yield:
• Acute myeloid leukemia. • Eosinophilic infiltrates: suggestive of Churg–Strauss
• Myelodysplastic syndrome. syndrome.
• Plasmacytoma. • Epineurial and endoneurial granulomas, with secondary
• Paraproteinemia. demyelination, are diagnostic of sarcoidosis.
• Waldenström’s macroglobulinemia.
• Lymphoma. DIAGNOSIS
A histologic diagnosis requiring confirmation by tissue biopsy.
Inflammation/infection
• Meningococcal septicemia. TREATMENT
• Infective endocarditis. Corticosteroids are the mainstay of treatment despite the
• Cutaneous polyarthritis, in children. lack of evidence from controlled clinical trials. Steroids
• Non-vasculitis, steroid-responsive form. combined with cyclophosphamide are generally required for
• Sarcoidosis (see p.350): a chronic multisystem disease of severe forms, or those with systemic features of vasculitis.
unknown cause. There is infiltration of affected tissues The drawbacks of cyclophosphamide include opportunistic
(most commonly lung) by T lymphocytes and mono- infection, gonadal failure and bladder carcinoma (15%). For
nuclear phagocytes, with formation of non-caseating patients with giant cell arteritis, steroids should be
epithelioid granuloma. The central and peripheral nervous continued for many months, or even years; the clinical
system is involved in about 5% of cases, and the peripheral features and ESR are useful measures for titration of dosage.
nervous system alone in about 3%. The most common
neurologic presentation is cranial neuropathy, frequently PROGNOSIS
involving the facial nerve. The course may be fluctuating, Depends on the cause:
as with Wegener’s granulomatosis. Peripheral nervous • Polyarteritis nodosa: poor in untreated patients.
system involvement is usually as a mononeuropathy Corticosteroid therapy is associated with partial or
multiplex, which may be accompanied by lesions of the complete recovery of neurologic function in about half
CNS and cranial nerves, but can also be as a symmetric of patients over a few months to years.
polyneuropathy, compressive median neuropathy due to • Rheumatoid arthritis: outcome varies, and may not be
synovitis, or multifocal sensory neuropathy with large that bad.
areas of sensory loss over the trunk. • Systemic lupus erythematosus: tends to recover
spontaneously.
Other conditions • Wegener’s granulomatosis: peripheral (and cranial) neuro-
• Diabetes mellitus. pathies may resolve spontaneously over several hours.
• Neurofibromatosis. • Sarcoidosis: corticosteroids are the treatment of choice
• Jellyfish stings. but the response is extremely variable.
• Non-systemic form confined to nerve or nerve and
Symmetric sensorimotor neuropathy (see p.579) muscle: much better prognosis and an excellent response
to therapy.
Guillain–Barré syndrome (see p.588)
Fulminant forms of vasculitic neuropathy (e.g.
Churg–Strauss syndrome) may present like this.
Herpes Zoster Infection 599

736 HERPES ZOSTER INFECTION

DEFINITION
A predominantly dermatologic condition caused by
reactivation of the varicella-zoster virus which is latent in
nerve cells.

EPIDEMIOLOGY
Varicella-zoster virus is a ubiquitous infectious agent. More
than 90% of the adult population in the western world, and
50% in tropical countries, are infected with the virus.
• Incidence of shingles: 100–225 per 100 000 general
population per year.
• Cumulative incidence: at least 20% of all adults suffer
from zoster at some time.
• Age: any age, but the chances rise with age. 5% of cases
737 occur in children younger than 15 years of age.
• Gender: M=F.

PATHOLOGY
• Dorsal root ganglia: inflammation (mononuclear cell
infiltration) with intranuclear varicella-zoster virus
inclusions in neurons and satellite cells; neuronal loss and
necrosis. Destruction of ganglion cells is prominent in
areas of hemorrhage. Sclerosis occurs months to years
after the acute attack.
• Posterior nerve roots: inflammation, destruction of
myelin and axonal swelling within 1–2 weeks after the
onset of rash, followed by macrophage and fibroblastic
proliferation.
• Posterior columns: myelin breakdown in severe
infections.

738 ETIOLOGY AND PATHOPHYSIOLOGY


• Transmission is largely through inhalation of infectious
aerosols. Direct contact with active varicella or zoster
lesions less often leads to infection.
• Incubation period: 10–21 days.
• Primary infection in the non-immune host is chickenpox.
• Following the acute, often trivial, primary infection, the
virus becomes latent in nerve cells.
• In the immune host, reactivation of the virus results in
zoster, which is predominantly an infection of the skin
(shingles).

Risk factors
• Increasing age.
• Immunosuppression.
• HIV infection.
736–738 Sural nerve biopsy from a patient with ischemic neuropathy
due to polyarteritis nodosa. H&E stain, x100 magnification (736), and
H&E, x400 (737), show infiltration of the arteriolar wall by neutrophils,
and fibrinoid necrosis of the vessel wall as a homogenous pink. Martius
scarlet blue stain for fibrin, x400 (738), shows the fibrin (of the
fibrinoid necrosis) as red.
600 Diseases of the Peripheral Nerve

CLINICAL FEATURES COMPLICATIONS


• Site of involvement of nerve root dermatomes: • Aseptic meningitis: some degree is often, although not
– Trigeminal nerve: 12% of patients. always present, particularly in the early stages.
– Cervical: 17%. • Encephalitis (see p.292).
– Thoracic: 50%. • Cerebral infarction via infiltration and arteritis of major
– Lumbar: 10%. intracranial arteries.
– Sacral: 5%. • Brachial plexus neuropathy.
• Pain, and sometimes paresthesia, in the region of the • Myelitis.
involved dermatome may precede the skin eruption by up • Acute ascending polyradiculopathy.
to 3 weeks. This may lead to misdiagnosis (e.g. of chole- • Mononeuropathy (median, ulnar, long thoracic,
cystitis) and inappropriate treatment (e.g. cholecystectomy). recurrent laryngeal, or phrenic nerves).
• Skin lesions (vesicular eruption) and sensory loss in the • Post-herpetic neuralgia: point-prevalence in community
affected dermatomes (739–742). The vesicles are initially studies is about 19% at 1 month, 7% at 3 months, 3% at
clear, become turbid, begin to crust within 5–10 days, 1 year, and 1% at 3 years. Usually mild to moderate
and occasionally leave residual scars. severity. Risk factors include increased age (>60 years),
• Low grade fever, general malaise, headache, neck stiffness, and prodrome of dermatomal pain before appearance of rash,
regional lymphadenopathy may accompanying symptoms. and severity of pain and rash.
• In 3–5% of patients with cutaneous zoster, concomitant
infection of the ventral roots at the same level of the INVESTIGATIONS
spinal cord lead to weakness of the affected myotome • Swab and culture infected vesicles.
(‘segmental zoster paresis’). • Viral serology.
• Pain usually abates after the skin has healed, but at least • Electromyography: may show evidence of motor
10–20% of patients develop post-herpetic neuralgia. denervation in paraspinal muscles adjacent to involved
sensory roots, indicating concomitant involvement of
ventral roots (clinically or subclinically).

739 740

741 742

739–741 Vesicular skin rash in the distribution of the T5 dermatome, 742 Vesicular skin rash over the left side of the sacrum in a patient
around the trunk in the mid thoracic level, due to herpes zoster with a left L5 radiculopathy due to herpes zoster infection.
infection of the right T5 dorsal root ganglia.
Human Immunodeficiency Virus (HIV) Neuropathy 601

TREATMENT HUMAN IMMUNODEFICIENCY VIRUS


Herpes zoster (HIV) NEUROPATHY
Acyclovir
• 800 mg five times a day, orally for 7 days (intravenously if
immunocompromised). DEFINITION
• Speeds healing and reduces the period of pain and skin Peripheral neuropathy (‘neuritis’ in its true sense) caused by
lesions. infection with the human immunodeficiency virus.
• Reduces the incidence of post-herpetic neuralgia.
• Combining acyclovir with oral prednisolone does not EPIDEMIOLOGY
prevent the occurrence of post-herpetic neuralgia. • Prevalence: uncommon; about 1–2% of HIV positive
• A related drug to acyclovir, such as valacyclovir or fam- individuals. Much more common in the late stages of the
ciclovir, is an alternative. As they have better bioavail- disease.
ability, they can be given three times daily. Famciclovir • Age: young adults.
oral 500 mg or 750 mg three times daily, given within • Gender: M≥F.
72 hours of the onset of rash for 7 days, but once- or
twice-daily dosing interval in patients with renal PATHOLOGY
impairment, decreases the duration and symptoms of Acute and chronic inflammatory demyelinating
acute herpes zoster and post-herpetic neuralgia. Adverse polyneuropathy
effects are few. Segmental demyelination.

Post-herpetic neuralgia Distal, symmetric, painful, predominantly sensory


Analgesia axonal neuropathy
• Topical: • The most common peripheral nerve syndrome associated
– Lignocaine gel 5%. with HIV infection, occurring clinically in about one-
– Capsaicin. third of patients late in the disease course, and demon-
– Acetylsalicylic acid (aspirin). strable pathologically in up to 100% of small series (743).
• Oral: tricyclic antidepressants. • A progressive axonal degeneration with endoneurial and
• Parenteral: epineurial inflammation may be found on nerve biopsy.
– Ketamine.
– Narcotics. Sensory ataxic neuropathy
– Intravenous lidocaine. Ganglioneuronitis.
• Nerve blockade/solvents:
– Selective sympathetic or somatic nerve blockade.
– Alcohol.
– Chloroform.
– Low-energy laser.
• Systemic and intralesional corticosteroids: ineffective.

Antiviral therapy
• Famciclovir (500 mg or 750 mg three times a day for
7 days) halves the duration of post-herpetic neuralgia (by
about 2 months on average from 163 days [4 months]
to 63 days [2 months]) in immunocompetent patients,
particularly those over 50 years of age.
• Valaciclovir (valacyclovir).
743
PROGNOSIS
• Herpes zoster infection usually resolves spontaneously
over a week or so, particularly with supplementation in
the form of antiviral agent.
• Recovery from post-herpetic neuralgia occurs in most
cases, but may take up to 2 years.

743 Sural nerve biopsy, teased osmicated preparation, three nerve


fibers. In the upper and middle fibers paranodal demyelination is seen
and in the lowermost fiber there are myelin ovoids indicating Wallerian
degeneration.
602 Diseases of the Peripheral Nerve

Mononeuritis multiplex and progressive • Several subtypes of peripheral neuropathy: the most
polyradiculopathy common patterns involve several rather than single nerves.
Necrotizing arteritis of the vasa nervorum (744, 745).
Acute inflammatory demyelinating polyneuropathy
Isolated mononeuropathy without apparent local • Subacute onset over days.
compression (e.g. facial nerve, median nerve, lateral • Progressive motor weakness, beginning in the lower
cutaneous nerve of thigh, common peroneal nerve) extremities and ascending.
• Areflexia.
Iatrogenic dose-related toxic neuropathy • Variable sensory and autonomic symptoms.
• Some patients progress to respiratory insufficiency
ETIOLOGY AND PATHOPHYSIOLOGY requiring mechanical ventilatory support.
Acute inflammatory demyelinating polyneuropathy
• A rare event which occurs early in the course of HIV Chronic inflammatory demyelinating
infection and may represent a reaction to seroconversion. polyneuropathy
• Presumed to result from an immune reaction that • Progressive motor weakness, beginning in the lower
transiently causes peripheral demyelination. extremities and ascending.
• Secondary to perivascular replication of HIV in • Areflexia.
infiltrating mononuclear cells. • More prominent sensory symptoms than AIDP.
• Usually recovers completely. • May follow a progressive or relapsing course over months

Chronic inflammatory demyelinating Distal, symmetric, painful, predominantly


polyneuropathy sensory axonal neuropathy
May occur at any disease stage, presumably as a result of • Distal symmetric sensory involvement beginning in the
immune mediated demyelination. feet and gradually ascending in the legs.
• Severe neuralgic pain; burning or shooting in character.
Distal, symmetric, painful, predominantly sensory • Feet are numb, and paresthesiae in upper and lower
axonal neuropathy limbs are usually present.
• Etiology unknown. • Examination reveals impairment of vibration and pinprick
• Rare in the early stages of infection. sensation distally, loss of or diminished angle reflexes with
• Clear temporal relation between onset of symptoms or variable depression of other reflexes and sensory ataxia.
abrupt change in severity of symptoms and • Motor weakness in distal muscles may be present but this
cytomegalovirus (CMV) infection. is overshadowed by the sensory symptoms.

Mononeuritis multiplex and progressive Sensory ataxic neuropathy


polyradiculopathy
Usually arise in AIDS patients with CD4 counts <50 and Progressive polyradiculopathy
CMV infection. Cauda equina syndrome.

Iatrogenic dose-related toxic neuropathy Mononeuritis multiplex


Due to treatment for HIV with dideoxynucleotides, Bilateral radial nerve palsy.
particularly ddC.
Isolated mononeuropathy without apparent
CLINICAL FEATURES local compression
• Onset especially in the late stages of the disease, when • Facial neuropathy.
the CNS is also commonly involved. • Median neuropathy.

744 745

744, 745 Nerve biopsy showing mild inflammatory cell infiltrate around the veins of the vasa nervorum (744) and marked neutrophil infiltration
(dark, nucleated cells) and fibrinoid necrosis (homogeneous pink substance) of the arteriolar wall of the vasa nervorum (745).
Human Immunodeficiency Virus (HIV) Neuropathy 603

• Lateral cutaneous nerve of thigh neuropathy. Sural nerve biopsy


• Common peroneal neuropathy. Performed if the differential diagnosis includes other
treatable conditions that can be diagnosed by sural nerve
DIFFERENTIAL DIAGNOSIS biopsy (see Peripheral neuropathy, pp.579, 583).
Acute and chronic inflammatory demyelinating
polyneuropathy (see pp. 589, 593) TREATMENT
General principles
Distal, symmetric, painful, predominantly sensory • Correct any metabolic deficiency and discontinue
axonal neuropathy potential neurotoxic agents, particularly dideoxynucleo-
• Toxic effects of dideoxynucleoside antiretroviral agents, sides, if possible.
which may mimic or exacerbate this neuropathy. • Physiotherapy to prevent contracture formation and
• Other potentially neurotoxic agents commonly used in improve strength and function.
HIV patients such as dapsone, isoniazid, and vincristine. • Occupational therapy.
• Vitamin B12 deficiency is common in these patients. • Subcutaneous heparin 5000 U bd and compression
• Lymphoma may produce neuralgia by direct invasion or stockings to prevent deep vein thrombophlebitis in non-
by paraneoplastic effects. ambulatory patients.
• Diabetes mellitus. • Disabling sensory symptoms (neuralgic pain) may
• Alcoholic neuropathy. respond to any of the following:
– Amitriptyline.
Sensory ataxic neuropathy due to ganglioneuronitis – Carbamazepine (starting with 100 mg bd and increasing
• Paraneoplastic syndrome. by 200 mg weekly to 800–1200 mg/day if tolerated and
• Sjögren’s syndrome. needed).
– Diphenylhydantoin (phenytoin) (100 mg tds).
Mononeuritis multiplex – Gabapentin (300–600 mg tds).
• Vasculitis. – Mexiletene (mexiletine) (150 mg bd, increasing to 300
• Herpes zoster infection. mg bd as required), if the above measures fail.
• Narcotic analgesia, if the above measures fail.
INVESTIGATIONS
CD4 lymphocyte count Acute inflammatory demyelinating polyneuropathy
Most opportunistic complications appear after significant • May recover spontaneously.
immunosuppression has occurred, as reflected by CD4 • If significant motor impairment:
lymphocyte counts of 200/mm3 or less. – Plasma exchange totalling 200–250 ml/kg divided in five
exchanges over a 2-week period with 5% albumin
Plasma HIV viral RNA load replacement. Or:
– Pooled human immunoglobulin 0.4 g/kg daily for
Serology 5 days given as an i.v. infusion at 0.05–4.0 ml/kg/hour
• HIV. as tolerated.
• CMV. • Electively intubate patients with impending respiratory
failure (vital capacity <1000 cm3) until adequate
CSF ventilatory function returns.
• Acute and chronic inflammatory demyelinating
polyneuropathy. Chronic inflammatory demyelinating
• Prominent elevation of CSF protein. polyneuropathy
• Moderate lymphocytic pleocytosis, in contrast with the • Maintenance therapy with one treatment every
non-HIV-infected population with acute IDP. 2–4 weeks of pooled human immunoglobulin (or plasma
• A polymorphonuclear pleocytosis suggests CMV infection. exchange). Intervals can be gradually extended as
response occurs.
Electrophysiologic studies • Corticosteroid therapy probably should not be used
Acute and chronic inflammatory demyelinating because of the potential for facilitating opportunistic
polyneuropathy: infections.
• Prominent slowing and motor nerve conduction blocks.
• Prolonged or absent F-wave responses. Distal, symmetric, painful, predominantly sensory
• Variable degrees of axonal damage and denervation. axonal neuropathy
• Direct HIV therapy has variable benefit.
Distal, symmetric, painful, predominantly • Treat disabling sensory symptoms (neuralgic pain) as
sensory axonal neuropathy above.
• Diminished amplitudes and conduction in sural nerves
and variable amplitude decrements in other nerves with Iatrogenic dose-related toxic neuropathy
relatively preserved conduction. Discontinue potential neurotoxic agents, particularly
• EMG may detect symmetric denervation and dideoxynucleosides, if possible.
reinnervation.
604 Diseases of the Peripheral Nerve

PROGNOSIS Multibacillary leprosy


Mononeuritis multiplex and progressive • ≥6 skin lesions and may be skin-smear positive.
polyradiculopathy • Equivalent to BB, BL, and LL diseases of Ridley–Jopling
Usually progressive unless antiviral treatment with classification.
ganciclovir is given.
There is a continuous spectrum from paucibacillary or
Acute inflammatory demyelinating polyneuropathy tuberculoid leprosy (TT) with high cell-mediated immunity
Natural history is similar to that of Guillain–Barré syndrome and low acid-fast bacilli counts, to multibacillary or
in the non-HIV-infected population and usually recovers lepromatous leprosy (LL) with low cell-mediated immunity
completely. and high acid-fast bacilli counts.

Chronic inflammatory demyelinating ETIOLOGY AND PATHOPHYSIOLOGY


polyneuropathy Mycobacterium leprae is the causative bacterium of leprosy.
• Neurologic symptoms may be relapsing or progressive. It is an obligate intracellular parasite which grows best at
• Residual neurologic impairment is common. 27–30°C (81–86°F), hence its predilection for cooler areas
of the body. It is most probably spread by the respiratory
Iatrogenic dose-related toxic neuropathy route. Most people are not susceptible to leprosy and after
Toxic symptoms may persist for 6–8 weeks after typical exposure will not develop it. Those who do, after an
discontinuation. incubation period of several years, may have only a single
lesion (indeterminate leprosy), which often self-heals. When
self-healing does not occur and when the single lesion is not
treated, the disease may progress to the paucibacillary or
multibacillary stages.
LEPROSY Those who develop the disease, particularly the
multibacillary type, demonstrate an impaired cell-mediated
immune response to M. leprae. The source of the impaired
DEFINITION immune response is uncertain but probably genetic. M.
An ancient disease caused by the obligate intracellular leprae selectively enters the small sensory intracutaneous
parasite Mycobacterium leprae. nerve endings, and leads to granulomatous inflammation
with thickening of the affected nerve or nerves.
EPIDEMIOLOGY
Leprosy occurs mostly in tropical areas of the world, but Tuberculoid (localized) leprosy
also in cooler regions of China, Japan and Korea. M. leprae often involves only a single nerve, most commonly
• Incidence: unchanged: 685 000 new patients registered the posterior tibial nerve (sensory), the ulnar and median
throughout the world in 1997, many at an early stage of nerves or the common peroneal nerve. The superficial
the disease, and 42% were multibacillary. branch of the radial nerve is affected only later in the
• Prevalence: varies among countries, but most cases arise disease. The bacteria slowly proliferate, spreading from the
in developing countries, led by India and Brazil, where intracutaneous nerve endings in a centripetal direction to
the prevalence is as high as 1% or more. Less than one thicker parts of the nerve. Eventually motor branches are
million cases registered worldwide in 1998, compared also involved. If nerves are infected at well-recognized
with 5.4 million in 1985 (probably due to the entrapment sites (e.g. the carpal tunnel, ulnar groove, and
introduction of multi-drug therapy [rifampicin tarsal tunnel) the nerve may swell leading to secondary
(rifampin), dapsone, and clofazimine] by the World compression and dysfunction.
Health Organization).
• Age: any age. Lepromatous leprosy
• Gender: M>F (1.5:1). Many bacilli with high infectivity lead to an extensive and
diffuse form of the disease.
CLASSIFICATION
Ridley–Jopling immunologic classification (based on CLINICAL FEATURES
sensorimotor, skin smear and skin biopsy findings): Incubation period is at least 3 years and often longer.
• Indeterminate (I).
• Tuberculoid (TT). History
• Borderline tuberculoid (BT). • The first and major symptoms are altered sensation: pain,
• Mid-borderline (BB). paresthesia and pruritus.
• Borderline lepromatous (BL). • Patchy hypopigmentation of the skin with analgesia is
• Lepromatous (LL). commonly, but not always present at an early stage.

WHO classification
Paucibacillary leprosy
• ≤5 skin lesions with no bacilli on skin smears.
• Equivalent to I, TT, and BT diseases of Ridley–Jopling
classification.
Leprosy 605

Examination Lepromin skin test


• Fusiform and tender thickening/enlargement of one or Utilizes a suspension of killed M. leprae, and is used as a
more peripheral nerves over part of their course in measure of the cell-mediated immune response, but its
superficial locations (e.g. the ulnar nerve at the elbow, usefulness is limited. It is generally positive in paucibacillary
the median and superficial radial cutaneous nerve at the cases and negative in multibacillary leprosy. A positive
wrist, the common peroneal nerve at the head of the response in normal individuals is associated with a reduced
fibular, and the greater auricular nerve in the neck). likelihood of developing the disease.
Detected by brushing the skin carefully with a fingertip.
• Hypopigmented areas of skin may be present, but can be DIAGNOSIS
difficult to detect in whites (746, 747). • Suspect in any patient with skin lesions, sensory loss
• Loss of pain sensation (and often anhidrosis) in the and/or enlarged nerve(s), particularly if an immigrant or
affected skin areas despite ability to distinguish blunt a local returning home after having lived in an endemic
from sharp sensations, and preserved position sense and area.
vibration sense. • The diagnosis rests mainly on the skin and peripheral
• Painless ulcerations of the fingers and toes may be nerve, but other areas can be affected such as the eyes,
present if affected. nose and testicles.
• Acid-fast bacilli must be demonstrated for a definitive
DIFFERENTIAL DIAGNOSIS diagnosis.
Thickening of nerves
• Hypertrophic forms of hereditary neuropathies: often TREATMENT
other family members are affected because of an Multi-drug therapy (rifampicin [rifampin], dapsone, and
autosomal dominant inheritance. clofazimine), administered for a minimum period of 2 years
• Von Recklinghausen’s disease (neurofibromatosis): often or until after a skin smear is negative for acid-fast bacilli,
other family members are affected because of an whichever is later, is recommended by the World Health
autosomal dominant inheritance. Organization.
• Refsum’s disease (phytanic acid deficiency): autosomal
recessive.
• Amyloidosis.

INVESTIGATIONS
Nerve conduction studies 746
May reveal electrophysiologic involvement of other nerves
in cases where only a single nerve is affected clinically.

DNA amplification
Detection of M. leprae in tissue by the polymerase chain
reaction.

Phenolic glycolipid-1 (PGL-1) antibodies

Stained smears of nasal swabs


May detect bacilli (often transmitted by nasal secretions) if
there is diffuse involvement.

Skin smears
• Take from skin lesions and the ears, elbows and/or
knees. 747
• Small slits are made in pinched skin (to avoid bleeding),
the edges of the cut skin are scraped, and the tissue fluid
obtained is smeared on a slide and stained for acid-fast
bacilli. The bacterial index can range from 0 (none found
in 100 oil-immersion fields) to 6+ (over 1000 bacilli per
field).

Skin biopsy
Taken from entirely within a skin lesion, focusing on the
extent and type of infiltrate and involvement of dermal
nerves.

Sensory nerve biopsy


Performed if skin biopsy is uninformative and leprosy is still
suspected. 746, 747 Patchy hypopigmentation of the skin in an Australian
Aboriginal patient with normally pigmented skin and hypopigmented
areas on the skin of the hands (746) and knee (747) due to leprosy.
606 Diseases of the Peripheral Nerve

Single-lesion paucibacillary leprosy • Some paraproteins (mostly polyclonal) react with GM1
Single dose of rifampicin (rifampin) 600 mg, ofloxacin 400 gangliosides or disialosyl groups on gangliosides GD1b,
mg, and minocycline 100 mg (ROM). GT1b, and GQ1b.

Paucibacillary leprosy Systemic disorders


Rifampicin (rifampin) 600 mg monthly, supervised, and • Multiple myeloma.
dapsone 100 mg daily, unsupervised, for 6 months. • Osteosclerotic myeloma (solitary or multiple plasma-
cytomas), or the POEMS (polyneuropathy, organo-
Multibacillary leprosy megaly, endocrinopathy, monoclonal paraproteinemia,
Rifampicin (rifampin) 600 mg and clofazimine 300 mg and skin hyperpigmentation) syndrome.
monthly, supervised; and dapsone 100 mg and clofazimine • Waldenström’s macroglobulinemia.
50 mg daily, unsupervised, for at least 12 months, if not 24 • Amyloidosis.
months. • Cryoglobulinemia.
• Other lymphoproliferative disorders:
Adverse effects – Non-Hodgkin’s lymphoma.
Uncommon – Leukemia.
• Dapsone: mild hemolytic anemia (can be severe with – Castleman’s disease.
glucose-6-phosphate dehydrogenase deficiency), agranu- – Hypersensitivity lymphadenopathy.
locytosis and skin eruptions. Safely taken in pregnancy.
• Clofazimine: gastrointestinal upset and skin pigmentation PATHOLOGY (748)
(which clears when the drug is discontinued). Safely MGUS
taken in pregnancy. • The IgM polyneuropathy is usually of demyelinating type
• Rifampicin (rifampin): rarely causes side effects given as and predominantly causes large fiber sensory dysfunction.
a once monthly dose of 600 mg, but experience in • Immunocytochemistry may show binding of IgM to
pregnancy is limited. myelin with widening of the interperiod line within
myelin.
PROGNOSIS • Electron microscopy may show characteristic widening
If appropriately treated, the average relapse rate is slightly of external myelin lamellae.
>1% for paucibacillary cases and <1% for multibacillary cases. • The IgG polyneuropathy may be an axonal neuropathy,
If relapse does occur, the patient should be re-treated with and there may be antibodies directed against sulfatide or
the same regimen since drug resistance is very unlikely if the chondroitin sulfate C, both of which are epitopes on the
patient’s M. leprae was originally fully sensitive to the drugs axon.
used.
Systemic disorders
Multiple myeloma
• Usually a dying-back peripheral neuropathy with
destruction of both axons and myelin; presumably
PARAPROTEINEMIC NEUROPATHIES primary axonal degeneration with secondary
demyelination.
• Amyloid deposition is present in about one-third of
DEFINITION cases.
A group of neuropathies associated with the presence of • Neoplastic root infiltration occurs rarely.
excessive amounts of abnormal immunoglobulins, which are
usually monoclonal serum proteins (termed M protein or
M spike) and are the product of a single clone of plasma
cells.

EPIDEMIOLOGY 748
• Prevalence: account for 10% of demyelinating
neuropathies and 10% of neuropathies of otherwise
unknown etiology.
• Age: more common in the elderly (i.e. >50 years of age).
• Gender: M>F.

ETIOLOGY
Monoclonal gammopathy of uncertain significance (MGUS)
(most common):
• Comprises two-thirds of patients with paraproteinemic
neuropathy.
• Among patients with MGUS and neuropathy, the
paraprotein is of any immunoglobulin (Ig) class, but is
usually IgM (60%) and sometimes with κ light chains,
and less commonly IgG (30%) and IgA (10%). 748 Transverse section though a nerve fascicle showing loss of
myelinated fibers and axonal degeneration.
Paraproteinemic Neuropathies 607

Osteosclerotic myeloma • Myelin associated glycoprotein (MAG) is the most


• A rare plasma-cell dyscrasia characterized by single or common target epitope. The carbohydrate epitope on
multiple plasmacytomas that manifest as sclerotic bone myelin-associated glycoprotein also reacts with the HNK-
lesions. 1 epitope and there is shared reactivity with two other
• Accounts for 3–5% of myelomas. myelin proteins, P0 and PMP-22, and a sulfated
• 85% of patients present with a demyelinating glycosphingolipid sulfate-3-glucuronyl paragloboside.
polyneuropathy, with secondary axonal degeneration, • A few patients with a sensory form of axonal neuropathy
and with or without inflammation. have autoantibodies (usually IgG) directed against
• The M protein, which is usually IgG or IgA in low sulfatide or chondroitin sulfate C, both of which are
concentration, is present in 90% of cases, and virtually epitopes on the axon.
always with a λ subtype of light chain. • In the CANOMAD syndrome (chronic ataxic neuropathy,
• Essentially the same disease as the POEMS syndrome. ophthalmoplegia, M protein, agglutination, anti-disialosyl
antibodies), the antibody binds to human dorsal roots and
Waldenström’s macroglobulinemia dorsal root ganglia and to femoral and oculomotor nerves.
• Demyelination, but distal axonopathy and sensory • In osteosclerotic myeloma, the deposition of light chains
neuropathy occur rarely. in the endoneurium suggests that the paraprotein has a
• The IgM paraprotein is derived from lymphocytoid cells proximate role in nerve damage. Greatly elevated levels
that proliferate in the marrow and lymph nodes, much of proinflammatory cytokines, such as tumor necrosis
the same as in IgM MGUS, from which Waldenström’s factor, have also been implicated.
macroglobulinemia may arise. • In amyloidosis, the pathogenesis of the generalized
sensory neuropathy is uncertain; both a direct toxic effect
Amyloidosis of amyloid and vascular insufficiency have been
Amyloid polyneuropathies are of two types: proposed. Amyloid concentration in the flexor
• Inherited amyloidosis-associated neuropathy. retinaculum causes carpal tunnel syndrome (see p.630).
• Primary (non-familial) systemic amyloidosis-associated
neuropathy: CLINICAL FEATURES
– More common than inherited amyloidosis-associated Heterogeneous clinical picture.
neuropathy.
– An M spike is present in serum or urine in 90% of Peripheral neuropathy associated with MGUS
patients, usually consisting of IgG with a λ light chain or IgM (usually κ) paraproteinemic neuropathy syndrome with
the light chain alone. antibodies to myelin-associated glycoprotein
– A symmetric sensorimotor small-diameter sensory fiber • A relatively homogeneous subgroup clinically.
axonal polyneuropathy with amyloid deposition. • Men, over 50 years of age are principally, but not
exclusively, affected.
Cryoglobulinemia • Slowly progressive, predominantly sensory, sensorimotor
• Cryoglobulins are proteins (usually IgG or IgM) which demyelinating neuropathy characterized by foot
precipitate when cooled, re-dissolve when warmed, and numbness, paresthesiae, imbalance and gait ataxia
are deposited as immune complexes in blood vessels. progressing over a few months. Touch, joint position and
• The immunoglobulin may be monoclonal, both vibration sensation in the legs (referable to conduction
monoclonal and polyclonal (mixed essential in large fibers) are most affected.
cryoglobulinemia), or polyclonal. • Aching, discomfort, dysesthesiae or lancinating pains
• Neuropathy is quite common in patients who have mixed occur in half the patients.
essential cryoglobulinemia, the type unassociated with • An upper limb postural tremor is often present.
lymphoproliferative diseases, chronic infections, or • Weakness of the distal leg muscles with variable wasting
autoimmune disorders. occurs as the illness advances.
• The neuropathy is a confluence of complete and • A few patients have a pure motor disorder.
incomplete multiple axonal mononeuropathies caused by
vasculitis in numerous nerve fascicles. IgG and IgA paraproteinemic neuropathy syndromes
• More heterogeneous clinical picture.
PATHOPHYSIOLOGY • Clinical features and response to treatment commonly
• Some of the abnormal immunoglobulins have properties resemble CIDP, although a few have a sensory form of
of antibodies which are directed against components of axonal neuropathy.
the myelin sheath or axolemma.
• Others have an uncertain pathophysiologic role Neuropathic syndrome due to paraproteins (mostly polyclonal)
intermediate between that of proteins associated with which react with GM1 gangliosides
neuropathies and proteins associated with Purely motor syndrome.
lymphoproliferative disorders.
• The nerves may also be damaged by deposition of the Neuropathic syndrome due to paraproteins specific for disialosyl
amyloid byproduct of the circulating paraprotein. groups on gangliosides GD1b, GT1b and GQ1b
• Immunoglobulin M antibodies are more likely to be • Progressive sensory ataxic neuropathy due to altered
pathogenic than IgG or IgA. position sense.
• CANOMAD syndrome develops in a subgroup of these
patients.
608 Diseases of the Peripheral Nerve

Systemic malignant disorders DIFFERENTIAL DIAGNOSIS


(precede or accompany neuropathy) Chronic inflammatory demyelinating
Multiple myeloma polyneuropathy (see p.593)
• Neuropathy is present neurophysiologically in about one- • About one-quarter of patients with CIDP also have a
third of patients, and clinically in about 5–10% of paraproteinemia (which is of uncertain relevance).
patients. • The cardinal EMG feature of CIDP (a focal block of
• Usually mixed sensorimotor peripheral neuropathy but electric conduction in motor nerves) sometimes also
purely sensory or relapsing and remitting forms occur. occurs in MGUS.
• Autonomic dysfunction may be present in some patients, • The CSF protein level is usually elevated in both CIDP
and suggests the presence of systemic amyloidosis. and MGUS.
• Weakness and numbness of the distal limbs appear • Both CIDP and MGUS respond to immunomodulating
subacutely over several weeks, beginning occasionally in treatment.
the upper limbs.
Multifocal motor neuropathy (see CIDP, p.594):
Osteosclerotic myeloma, or the POEMS syndrome • A purely motor disorder of middle-aged men,
Commonly presents with a slowly progressive neuropathy characterized by slowly progressive, painless weakness
which is mainly motor and demyelinating, and may be part that is asymmetric and confined to one limb.
of the POEMS syndrome (polyneuropathy associated with • Linked to high titers of IgM antibodies directed at GM1
organomegaly, endocrinopathy, M protein, skin thickening ganglioside on myelin membranes. The circulating
and hyperpigmentation, and clubbing). paraprotein is often polyclonal, but monoclonal in 20%
of cases.
Waldenström’s macroglobulinemia • Electric conduction block in the proximal or middle
• Sensorimotor neuropathy occurs frequently, very similar segments of motor nerves, with normal sensory
to that which occurs in IgM paraprotein associated conduction in the same nerves.
neuropathies of benign type. • 90% of patients respond to immune globulin, but require
• Fatigue, weight loss, and bleeding dominate the clinical repeated infusions.
picture.
• Paresthesiae and numbness in the feet are followed by Motor neuron disease (see p.534)
weakness and wasting of the lower legs, causing foot
drop and a steppage gait, and months later, by arm Multiple myeloma
weakness. • Absence of systemic features of myeloma (bone pain,
fatigue, anemia, hypercalcemia, renal insufficiency).
Amyloid neuropathy • Smaller amount of paraprotein (<3 g/dl serum, and
• A symmetric sensorimotor small fiber polyneuropathy is usually 0.75–1.5 g/dl).
the presenting feature in 15% of patients. • Amyloidosis (cf. sensory form of axonal neuropathy that
• Numbness in the feet is the most common presenting symp- may occur with IgG MGUS).
tom, but the signature symptoms are burning and aching
pains with lancinating electric sensations and loss of pain and INVESTIGATIONS
temperature sensation in the distal parts of the limbs. Nerve conduction studies
• Autonomic symptoms can be extreme, particularly IgM paraproteinemic demyelinating neuropathy with anti-
postural hypotension, diarrhea (also from infiltration of myelin-associated glycoprotein antibodies
the gut wall), impotence and bladder dysfunction. • Slowing of motor nerve conduction velocity which is
• Carpal tunnel syndrome is common. more distal than proximal.
• Systemic symptoms include weight loss, and those • Distal motor latencies prolonged representing a marked
referable to amyloid deposition in other organs, such as distal accentuation of conduction slowing.
the heart and kidneys. • Absence of conduction block.

Cryoglobulinemia Multiple myeloma


• The most common clinical picture is that of a Neurophysiologic evidence of axonal damage, which may
progressive, symmetric, distal sensorimotor neuropathy be severe.
(due to a confluence of incomplete mononeuropathies)
combined with one or two clearly recognizable Waldenström’s macroglobulinemia
mononeuropathies (e.g. wrist or foot drop). Neurophysiologic evidence of demyelination, but
• The other typical presentation is that of a multiple occasionally a distal axonopathy or sensory neuropathy.
mononeuropathy.
• Onset is acute in about one-third of patients who have Amyloid neuropathy
multiple mononeuritis. Features of a symmetric sensorimotor small fiber axonal
• Pain is almost always present at onset. neuropathy.
• Paresthesiae and Raynaud’s phenomenon are precipitated
by cold in some patients. Cryoglobulinemia
• Weakness may be multifocal or generalized. Neurophysiologic evidence of axonal damage in multiple
nerves.
Paraproteinemic Neuropathies 609

Immunoglobulins and serum protein electrophoresis IgM MGUS


• Immunoglobulins can be detected by immunoelectro- • Generally more refractory to treatment than IgG or IgA
phoresis or the more sensitive immunofixation tests. MGUS.
• The detection of cryoglobulins requires that the blood • May respond to same regimens as IgG or IgA MGUS,
specimen is transported to the laboratory in a warm- particularly if chlorambucil or cyclophosphamide is added
water bath. in a dose sufficient to reduce the amount of M protein.
• Patients with anti-MAG antibodies generally require
CSF prolonged therapy with monthly plasma exchange and
CSF protein is often raised, and in osteosclerotic myeloma continuous oral or pulsed intravenous cyclophosphamide.
the CSF protein is almost invariably elevated to >1 g/l • Interferon-alpha was beneficial in one trial.
(>100 mg/dl).
Systemic disorders
Bone marrow examination or radiologic Multiple myeloma
skeletal survey Removing the paraprotein by plasma exchange has no
Performed if suspected plasmacytoma or myeloma (e.g. consistent effect on the neuropathy.
patients with IgG and IgA paraproteins, particularly if
resistant to treatment). Osteosclerotic myeloma
• Treatment of solitary bone lesions (e.g. resection,
Nerve biopsy focused radiotherapy, or chemotherapy) may stabilize or
MGUS: most common improve the neuropathy in about half of patients but the
• Immunocytochemistry may show binding of IgM to response may not be forthcoming for several months.
myelin with widening of the interperiod line within • Plasma exchange is generally ineffective, as in neuropathy
myelin. associated with myeloma.
• To rule out amyloid deposition in patients with sensory
axonal neuropathy due to suspected IgG MGUS. Waldenström’s macroglobulinemia
• Plasma exchange may slow the progression of the
Amyloid neuropathy neuropathy.
Confirms the diagnosis in 90% of cases. • Prednisone, melphalan, and chlorambucil may be helpful.

Biopsy of other tissues Amyloid neuropathy


Amyloid neuropathy • Prednisone and melphalan prolongs survival in a small
Amyloid is detected in biopsies of bone marrow and rectal proportion of patients for several years but has little effect
mucosa in 70% and 80% of cases, respectively. on the neuropathy.
• Autologous stem-cell transplantation may stabilize or
TREATMENT improve the condition in a few patients in the short term
Treatment of the underlying cause may improve the at least.
neuropathy.
Cryoglobulinemia
MGUS • Corticosteroids, cyclophosphamide, and plasma exchange
IgG or IgA MGUS are variably successful in stabilizing the neuropathy.
• Plasma exchange (a total of 220 ml/kg, given in four or • Interferon alpha is promising in cases associated with
five treatments) has been shown in a controlled trial to hepatitis C.
sometimes afford at least short-term benefit within days
or weeks of administration in about one-third of patients. PROGNOSIS
• Intravenous immunoglobulin in high dose (0.4 g/kg MGUS
body weight daily for 5 days) appears to benefit some • Usually benign, mild and stable, but about 20% of
patients. patients will in time acquire a malignant plasma-cell
• Corticosteroids, sometimes in combination with disorder, usually myeloma.
immunosuppressants, may be effective but is more often • The syndrome of predominantly sensory neuropathy due
ineffective. to IgM κ paraprotein with antibodies to myelin-
• Cyclophosphamide, melphalan, azathioprine, chlorambucil, associated glycoprotein is associated with a benign clinical
fludarabine and interferon-alpha have been used. course.
• Immunoadsorption (in which IgG and immune • Neuropathies associated with benign paraproteins of the
complexes are removed by passing the patient’s blood IgG and IgA class respond better to treatment than those
through a plastic column containing covalently bound associated with an IgM paraprotein. However, patients
staphylococcal protein) generally produces only transient with IgM paraprotein neuropathies may improve after
amelioration, and needs to be repeated every few plasma exchange or intravenous immunoglobulin,
months. particularly when used in association with
cyclophosphamide or chlorambucil.
• In patients with polyneuropathy associated with IgM
monoclonal gammopathy, antibody tests to MAG (myelin-
associated glycoprotein) SGPG (sulfoglucuronyl para-
globoside), and sulfatide do not have prognostic value.
610 Diseases of the Peripheral Nerve

DIABETIC NEUROPATHY • Nerve conduction velocity is reduced.


• Rapidly corrected by establishing diabetic control.

DEFINITION Generalized polyneuropathies


Neuropathic complications of diabetes mellitus. Distal symmetric predominantly sensory
(and motor) axonal polyneuropathy
EPIDEMIOLOGY • The most common type of diabetic neuropathy.
• Incidence: 54 (95% CI: 33–83) per 100 000 per year. • Very common in diabetes, particularly with poorly
• Lifetime prevalence: 2 (95% CI: 1–3) per 1000 controlled disease of long duration.
population. One of the most common causes of
peripheral neuropathy: Pathology: a distal axonal degeneration of dying back type
– At the time of diagnosis, 8% of non-insulin dependent with relative preservation of dorsal root ganglion cells (749).
diabetics have definite or probable neuropathy, compared This may well be a central-peripheral distal axonopathy in
with 2% of an age-and sex matched control population. which there is also a rostral degeneration of nerve fibers in
– After 10 years of follow-up, the prevalence of neuropathy the dorsal columns of the spinal cord. An important aspect
increases to 42% among diabetic patients and to 6% in is a failure of axonal regeneration, which probably
controls. contributes to the lack of reversibility of the neuropathy once
– At any one time, about 34% of insulin dependent it is established, even with good glycemic control.
diabetics and 26% of non-insulin dependent diabetics Pathophysiology: it remains to be established whether the
have distal symmetric polyneuropathy; and 58% of insulin mechanism is a direct metabolic effect or whether it is
dependent diabetic patients aged 30 years or more have secondary to hypoxia from microvascular disease. A major
distal symmetric polyneuropathy metabolic abnormality in nerve is the accumulation of
– At any one time, about 17% of diabetics have at least one sorbitol because of increased flux in the polyol pathway
abnormal test of autonomic function but, besides erectile secondary to hyperglycemia. The sorbitol in diabetic nerve
dysfunction, only 2.4% report symptoms attributable to is not sufficient in quantity to produce osmotic damage but
autonomic dysfunction. it is possible that it may have deleterious effects on neural
• Age: adults, increases with duration of diabetes. metabolism. However, trials with aldose reductase inhibitors
• Gender: M=F. that reduce the production of sorbitol have failed so far to
show any substantial effects on diabetic polyneuropathy.
PATHOLOGY Other possible metabolic disturbances of relevance include
Axonal degeneration of nerve fibers of all sizes, both alterations in the metabolism of essential fatty acid and non-
myelinated and unmyelinated. enzymatic glycation of proteins.
Clinical: distal symmetric predominantly sensory (and
ETIOLOGY AND PATHOPHYSIOLOGY motor) loss characterized by sensory impairment in a glove
• Diabetes renders nerves vulnerable to injury and may also and stocking distribution and distal motor weakness. The
involve the vasa nervorum. sequelae of longstanding severe distal sensory loss, such as
• Hyperglycemia play a central role in the pathogenesis of neuropathic joints, may be present (750).
diabetic peripheral neuropathy. Treatment: strict control of blood glucose concentrations
by an insulin pump or multiple daily insulin injections can
Risk factors for neuropathy prevent or greatly diminish the risk of developing
• Poor glycemic control. neuropathy. This treatment however, is only applicable to
• Duration of diabetes. patients with type I insulin-dependent diabetes and only a
• Age. small proportion of them. Good glycemic control can only
• Height. be achieved in practice in about 25% of patients. Once
• Male gender. DSSP is established, it fails to improve significantly even
• Alcohol consumption. with satisfactory glycemic control. Treatment is therefore
required that will prevent the occurrence of neuropathy or
Risk factors for neuropathy in insulin- halt its deterioration if present.
(but not non-insulin) dependent diabetics Prognosis: after 25 years of diabetes, about half will have
• Systemic hypertension. developed neuropathy. Further research is required to help
• Cigarette smoking. identify, perhaps by genetic markers, those patients who are
• Hyperlipidemia. more susceptible to developing neuropathy.

CLINICAL SYNDROMES Autonomic neuropathy


Onset • Symptoms include nocturnal diarrhea, postural
May be acute, but more commonly insidious. hypotension and syncope.
• Mild degrees are common in both type I and type II
Rapidly reversible phenomena diabetes, but severe forms are virtually only encountered
Hyperglycemic neuropathy in type I diabetic patients.
• Presumably related directly to hyperglycemia or to a
metabolic abnormality correlated with it (e.g. hypoxia, a
switch to anerobic glycolysis in the diabetic nerve).
• Patients with severe uncontrolled hyperglycemia.
• Uncomfortable sensory symptoms, mainly in the legs.
Diabetic Neuropathy 611

Acute painful sensory neuropathy Other focal peripheral nerve lesions are likely to result
• Uncommon. from an abnormal susceptibility of diabetic nerve to
• Mechanism: uncertain. compression. The reasons for this is uncertain, but in non-
• Severe burning or aching pain, mainly in the legs but diabetic individuals entrapment neuropathies are related to
sometimes more widespread. longitudinal axoplasmic displacement away from the site of
• Precipitants include treatment with insulin. compression and the consequent distortion and breakdown
• Examination reveals intense cutaneous contact of the myelin sheath of larger myelinated fibers. The basal
hyperesthesia but only mild sensory loss. lamina surrounding nerve fibers is known to be abnormally
• May be associated with uncontrolled hyperglycemia and rigid in patients with diabetic neuropathy, possibly due to
precipitous weight loss. increased cross linking of collagen because of abnormal
• Nerve biopsy shows acute axonal degeneration. glycation related to advanced glycation end product (AGE)
• Prognosis: resolves over several months with adequate formation.
glycemic control. In some patients with proximal lower limb diabetic
neuropathy, inflammatory lesions, including vasculitis,
Focal and multifocal neuropathies affecting small epineurial vessels, are present in peripheral
More common in diabetics than in the general population. nerves, raising the possibility of a superimposed auto-
immune process.
Pathogenesis
The abrupt onset of a diabetic IIIrd cranial nerve palsy is Cranial neuropathies
consistent with an ischemic basis and this has been The IIIrd and VIIth cranial nerves are affected particularly.
supported by sound pathologic studies. The pathology is a
focal demyelination, accounting for the usually satisfactory Thoracoabdominal radiculoneuropathy
recovery that occurs, presumably by remyelination.
Although nerve ischemia usually gives risk to axonal loss
rather than segmental demyelination, it is possible that
demyelination in focal diabetic lesions is the result of
reperfusion injury (which is known to produce
demyelination).

749 750

749 Teased nerve fiber (osmium tetroxide) showing segmental 750 Deformed (Charcot) ankle joints in a patient with distal
demyelination(yellow axons [myelin normally appears black]) and active symmetric sensory neuropathy due to diabetes.
Wallerian degeneration due to focal interruption of axons (numerous
myelin ovoids which appear as black blobs).
612 Diseases of the Peripheral Nerve

Focal limb mononeuropathies (including entrapment • May occur against a background of a chronic, symmetric
and compression neuropathies) polyneuropathy.
• Located at the well-known sites of entrapment or • Severe pain in the anterior thigh may be present at onset.
external compression, and commonly involve the median • Weakness of the quadriceps and iliopsoas is most marked,
nerve at the wrist (751) and ulnar nerve at the elbow. but the adductor muscles (obturator nerve) may also be
Other common sites are the radial and peroneal nerves weak.
(as in patients without diabetes), as well as the superficial • Absent knee jerk.
branch of the radial nerve (cheiralgia paresthetica). • Sensory deficits are rare.
• Symptomatic carpal tunnel syndrome is found in about • Weight loss is usual.
11% of patients with diabetes mellitus (751). • Differential diagnosis includes autoimmune vasculitis:
• Not uncommonly superimposed on a polyneuropathy, MRI scan showing enhancement of the lumbar nerve
which may be symptomatic or asymptomatic and only roots suggests autoimmune vasculitis rather than diabetes
evidenced from nerve conduction studies. A coexistent as the cause.
polyneuropathy is found in about 80% of diabetics with • Nerve conduction studies and EMG usually show
an ulnar nerve palsy, and about 20% of diabetics with features consistent with axonal degeneration of the
carpal tunnel syndrome. lumbar spinal nerve roots.
• Frequently it is difficult to determine whether the cause • Pathologic studies show inflammatory change or small
is external pressure or intrinsic focal nerve ischemia or infarcts in the lumbosacral plexus and trunks of the
infarction secondary to occlusion of small blood vessels femoral and other (e.g. obturator) nerves.
supplying the nerve. • Treatment is symptomatic although steroids have been
• Femoral neuropathy is not caused by entrapment. used in severe cases despite the diabetes.
• Acute painless peroneal neuropathy: usually caused by • At least some degree of recovery begins within weeks of
compression to at least some degree in diabetics. onset and continues over 12–18 months; if no
• Brachial plexus neuropathy: improvement at all has occurred after several months the
– Unilateral or bilateral. diagnosis is in doubt.
– May be associated with a typical radiculo-plexopathy of • Recovery is incomplete in nearly half of patients.
the legs (symmetric or asymmetric) and a background
generalized sensorimotor polyneuropathy. Superimposed chronic inflammatory demyelinating
– Onset: subacute or gradual. polyneuropathy
• CIDP is more frequent in diabetics.
Proximal diabetic neuropathy (diabetic amyotrophy, • A secondary autoimmune process may be responsible.
‘diabetic radiculo-plexopathy’, ‘lower limb asymmetric
motor neuropathy’) DIFFERENTIAL DIAGNOSIS
• Acute, painful, unilateral or asymmetric proximal leg Predominantly sensory neuropathies
weakness with particular involvement of the sensorimotor • Diabetes.
territory of the L2–L4 nerve roots (weakness of hip • Thiamine deficiency.
flexion and knee extension, absent knee jerk, and • Malignancy.
numbness of the anterior thigh and leg). • Leprosy.
• Onset in middle-aged or elderly diabetics. • Hereditary sensory neuropathies.
• May be the first manifestation of diabetes. • Amyloid.
• May arise in patients with diabetes that is mild and well • Uremia.
controlled.
Predominantly motor neuropathies
• Guillain–Barré syndrome.
751 • Porphyria.
• Diphtheria.
• Botulism.
• Lead.
• Charcot–Marie–Tooth disease.
• Disorders of the neuromuscular junction or muscle.

INVESTIGATIONS
• Nerve conduction studies: a more pronounced decrease
in sensory and motor compound action potential
amplitudes than in nerve conduction velocities,
consistent with axonal degeneration.
• Blood glucose: fasting.
• Hemoglobin A1C.
• Nerve biopsy if the diagnosis remains uncertain.

DIAGNOSIS
A typical clinical and neurophysiologic profile in a diabetic
751 Wasting of the left abductor pollicis brevis muscle (arrow) in a patient, after excluding differential diagnoses.
patient with a median neuropathy at the wrist due to diabetes.
Further Reading 613

TREATMENT PROGNOSIS
• Good diabetic control probably prevents the develop- • Depends on the intensity of glycemic control.
ment of neuropathy. • Poor glycemic control and low plasma concentrations of
• Pancreatic transplantation may relieve the progression. insulin independent of concentrations of glucose are
• Aldose reductase inhibitors do not appear to be effective. associated with increased risk of development and
• Recombinant human nerve growth factor 0.1µg/kg was progression of neuropathy.
not effective in a recently published randomized • Autonomic neuropathy in diabetes probably carries a
controlled trial involving 1019 patients with diabetic poor prognosis (i.e. increased risk of death).
polyneuropathy.
• Intravenous immunoglobulin may be helpful for diabetic
amyotrophy, but controlled trials are needed.
• Painful neuropathy may respond to tricyclic
antidepressants; gabapentin 900 mg/day is probably
ineffective or only minimally effective.

HEREDITARY NEUROPATHIES CHRONIC INFLAMMATORY


FURTHER READING De Jonghe P, Timmerman V, Nelis E, et al. DEMYELINATING POLYNEUROPATHY
(1999) A novel type of hereditary motor and Duarte J, Martinez AC, Rodriguez F, et al. (1999)
sensory neuropathy characterised by a mild Hypertrophy of multiple cranial nerves and
PERIPHERAL NEUROPATHY phenotype. Arch. Neurol., 56: 1283–1288. spinal roots in chronic inflammatory demyeli-
Epidemiology Lupski JR (2000) Recessive Charcot-Marie-Tooth nating neuropathy. J. Neurol. Neurosurg. Psy-
MacDonald BK, Cockerell OC, Sander JWAS, disease. Ann. Neurol., 47: 6–8. chiatry, 67: 685–687.
Shorvon SD (2000) The incidence and life- Dyck PJ, Dyck PJB (2000) Atypical varieties of
time prevalence of neurological disorders in a HEREDITARY MOTOR AND SENSORY chronic inflammatory demyelinating neu-
prospective community-based study in the NEUROPATHY ropathies. Lancet, 355: 1293–1294.
UK. Brain, 123: 665–676. Kamholz J, Menichella D, Jani A, et al. (2000) Haq RU, Fries TJ, Pendlebury WW (2000)
Martyn CN, Hughes RAC (1997) Epidemiology Charcot-Marie-Tooth disease type 1. Molecu- Chronic inflammatory demyelinating
of peripheral neuropathy. J. Neurol. Neurosurg. lar pathogenesis to gene therapy. Brain, 123: polyradiculoneuropathy. A study of proposed
Psychiatry, 62: 310–318. 222–233. electrodiagnostic and histologic criteria. Arch.
HEREDITARY NEUROPATHY WITH Neurol., 57: 1745–1750.
Etiology Hughes R, Bensa S, Willison H, et al. (2001) Ran-
Bird SJ, Rich MM (2000) Neuromuscular com- LIABILITY TO PRESSURE PALSIES
Duborg O, Mouton P, Brice A, et al. (2000) domized controlled trial of intravenous im-
plications of critical illness. The Neurologist, 6: munoglobulin versus oral prednisolone in
2–11. Guidelines for diagnosis of hereditary neu-
ropathy with liability to pressure palsies. Neu- chronic inflammatory demyelinating
romuscular Disorders, 10: 206–208. polyradiculoneuropathy. Ann. Neurol., 50:
Subtypes
195–201.
Bryer MA, Chad DA (1999) Sensory neu-
GUILLAIN–BARRÉ SYNDROME McLeod JG, Pollard JD, Macaskill P, et al. (1999)
ronopathies. The Neurologist, 5: 90–100.
Feasby TE, Hartung H-P (2001) Drain the roots. Prevalence of chronic inflammatory demyeli-
Holland NR, Crawford TO, Hauer P, et al.
A new treatment for Guillain–Barré syndrome. nating polyneuropathy in New South Wales,
(1998) Small-fibre sensory neuropathies: clin-
Neurology, 57: 753–754. Australia. Ann. Neurol., 46: 910–913.
ical course and neuropathology of idiopathic
Hahn AF (1998) Guillain-Barré syndrome. Mendell JR, Barohn RJ, Freimer ML, et al. (2001)
cases. Ann. Neurol., 44: 47–59.
Lancet, 352: 635–641. Randomised controlled trial of IVIg in un-
Windebank AJ, Blexrud MD, Dyck PJ, Daube JR,
Hartung H-P (1999) Infections and the Guillain- treated chronic inflammatory demyelinating
Karnes JL (1990) The syndrome of acute sen-
Barré syndrome. J. Neurol. Neurosurg. Psychi- polyradiculoneuropathy. Neurology, 56:
sory neuropathy: clinical features and electro-
atry, 66: 277. 445–449.
physiologic and pathologic changes. Neur-
Hughes RAC, Gregson NA, Hadden RDM, Pestronk A (1998) Multifocal motor neuropathy:
ology, 40: 584–591.
Smith KJ (1999) Pathogenesis of Diagnosis and treatment. Neurology, 51
Investigation Guillain–Barré syndrome. J. Neuroimmunol., (Suppl 5): S22–S24.
Gabriel CM, Howard R, Kinsella N, et al. (2000) 100: 74–97. Van den Berg-Vos RM, Franssen H, Wokke JHJ,
Prospective study of the usefulness of sural Hughes RAC (2001) Sensory form of Guil- et al. (2000) Multifocal motor neuropathy:
nerve biopsy. J. Neurol. Neurosurg. Psychia- lain–Barré syndrome. Lancet, 357: 1465. Diagnostic criteria that predict the response to
try, 69: 442–446. Logina I, Donaghy M (1999) Diphtheritic immunoglobulin treatment. Ann. Neurol., 48:
McLeod JG (1995) Investigation of peripheral polyneuropathy: a clinical study and compari- 919–926.
neuropathy. J. Neurol. Neurosurg. Psychiatry, son with Guillain-Barré syndrome. J. Neurol.
58: 274–283. Neurosurg. Psychiatry, 67: 433–438. VASCULITIC NEUROPATHY
Said G (2001) Value of nerve biopsy? Lancet, 357: Oh SJ, La Ganke C, Claussen GC (2001) Sen- Moore PM (2000) Vasculitic neuropathies. J.
1220–1221. sory Guillain–Barré syndrome. Neurology, 56: Neurol. Neurosurg. Psychiatry, 68: 271–276.
Wolfe GI, Nations SP (2001) Guide to autoanti- 82–86.
body testing in peripheral neuropathies. The Seneviratne U (2000) Guillain-Barré syndrome. HERPES ZOSTER INFECTION
Neurologist, 7: 195–207. Postgrad. Med. J., 76: 774–782. Choo PW, Galil K, Donahue JG, et al. (1997)
Wollinsky KH, Hülser P-J, Brinkmeier H, et al. Risk factors for post herpetic neuralgia. Arch.
Management (2001) CSF filtration is an effective treatment Intern. Med., 157: 1217–1224.
Hughes RAC (2000) Management of chronic pe- of Guillain–Barré syndrome. A randomized Cunningham AL, Dworkin RH (2000) The man-
ripheral neuropathy. Proc. R. Coll. Physicians clinical trial. Neurology, 57: 774–780. agement of post-herpetic neuralgia. BMJ, 321:
Edin., 30: 321–327. 778–779.
614 Diseases of the Peripheral Nerve

Helgason S, Petursson G, Gudmundsson S, Sig- Jacobsen RR, Krahenbuhl JL (1999) Leprosy. DIABETIC NEUROPATHY
urdsson JA (2000) Prevalence of postherpetic Lancet, 353: 655–660. Apfel SC, Schwartz S, Adornato BT (2000) Effi-
neuralgia after a first episode of herpes zoster: Young D (2001) Leprosy and the genome – not cacy and safety of recombinant human nerve
prospective study with long term follow up. yet a burnt-out case. Lancet, 357: 1639–1640. growth factor in patients with diabetic
BMJ, 321: 794–796. polyneuropathy. A randomized controlled
Tyring S, Barbarash RA, Nahlik JE, et al., and the NEURALGIC AMYOTROPHY trial. JAMA, 284: 2215–2221.
Collaborative Famciclovir Herpes Zoster Study Lo Y-L, Mills KR (1999) Motor root conduction Courtney AE, McDonnell GV, Patterson VH
Group (1995) Famciclovir for the treatment in neuralgic amyotrophy: evidence of proximal (2001) Human immunoglobulin for diabetic
of acute herpes zoster: effects on acute disease conduction block. J. Neurol. Neurosurg. Psy- amyotrophy – a promising prospect? Postgrad.
and postherpetic neuralgia. A randomized, chiatry, 66: 586–590. Med. J., 77: 326–328.
double-blind, placebo-controlled trial. Ann. Watts GDJ, O’Briant KC, Borreson TE, et al. Gorson KC, Schott C, Herman R, et al. (1999)
Intern. Med., 123: 89–96. (2001) Evidence for genetic heterogeneity in Gabapentin in the treatment of painful dia-
Wood MJ, Johnson RW, McKendrick MW (1994) hereditary neuralgic amyotrophy. Neurology, betic neuropathy: a placebo controlled, dou-
A randomized trial of acyclovir for 7 days or 56: 675–678. ble blind, cross over trial. J. Neurol. Neurosurg.
21 days with or without prednisolone for PARAPROTEINEMIC NEUROPATHIES Psychiatry, 66: 251–252.
treatment of acute herpes zoster. N. Engl. J. Eurelings M, Moons KGM, Notermans NC MacDonald BK, Cockerell OC, Sander JWAS,
Med., 330: 896–900. (2001) Neuropathy and IgM M-proteins. Shorvon SD (2000) The incidence and life-
Wood MJ, Kay R, Dworkin RH, Soong SJ, Whit- Prognostic value of antibodies to MAG, time prevalence of neurological disorders in a
ley RJ (1996) Oral acyclovir therapy acceler- SGPG, and sulfatide. Neurology, 56: 228–233. prospective community-based study in the
ates pain resolution in patients with herpes Natov N (1995) Pathogenesis and therapy of neu- UK. Brain, 123: 665–676.
zoster: a meta-analysis of placebo-controlled ropathies associated with monoclonal gam- Thomas PK (1999) Diabetic neuropathy: mecha-
trials. Clin. Infect. Dis., 22: 341–347. mopathies. Ann. Neurol., 37 (Suppl 1): nisms and future treatment options. J. Neurol.
S32–S42. Neurosurg. Psychiatry, 67: 277–281.
LEPROSY
Croft RP, Nicholls PG, Steyerberg EW, et al. Pollard JD, Young GAR (1997) Neurology and
(2000) A clinical prediction rule for nerve- the bone marrow. J. Neurol. Neurosurg. Psy-
function impairment in leprosy patients. chiatry, 63: 706–718.
Lancet, 355: 1603–1606. Ropper AH, Gorson KC (1998) Neuropathies as-
Hietaharju A, Croft R, Alam R, et al. (2000) sociated with paraproteinaemia. N. Engl. J.
Chronic neuropathic pain in treated leprosy. Med., 338: 1601–1606.
Lancet, 356: 1080–1081.
Chapter Twenty-two 615

Mononeuropathies
DORSAL SCAPULAR NERVE (TO ANATOMY
RHOMBOIDS) NEUROPATHY Course of the dorsal scapular nerve (to rhomboids):
• Arises from the upper trunk of the brachial plexus (752),
carrying fibers from the C4 and C5 nerve roots.
DEFINITION • Pierces the medial scalenus muscle.
Dysfunction of the dorsal scapular nerve to rhomboids. • Innervates the levator scapulae, which elevates the
scapula.
EPIDEMIOLOGY • Courses along the medial border of the scapula to
Uncommon. innervate the rhomboids, which adduct the medial
border of the shoulder blade.

752

Dorsal scapular nerve to rhomboids


Nerve to subclavius
C4
Long thoracic nerve to serratus anterior
Suprascapular nerve to supraspinatus and infraspinatus C5
Lateral cord C6

C7
Pectoralis minor
Posterior cord T1
Musculocutaneous nerve
Axillary nerve T2
Short head of biceps
Coracobrachialis Scalenus anterior
Medial pectoral nerve
Lateral pectoral nerve
Radial nerve
Median nerve Medial cord
Ulnar nerve
Subscapular nerves
Medial cutaneous nerve of forearm to subscapularis and
Medial cutaneous nerve of arm teres major

Thoracodorsal nerve
to latissimus dorsi

752 Diagram of the brachial plexus. (Adapted from Aids to examination of the peripheral nervous system (1986). Baillière Tindall, London.)
616 Mononeuropathies

ETIOLOGY LONG THORACIC NEUROPATHY


• Neuroma (753, 754).
• Neuralgic amyotrophy may involve the dorsal scapular
nerve. DEFINITION
• No descriptions of isolated entrapment or other Dysfunction of the long thoracic nerve to serratus anterior.
compressive lesions.
EPIDEMIOLOGY
EXAMINATION Uncommon.
Stand behind the patient and ask the patient to put their
hand behind their back, face the palm of the hand ANATOMY
backwards, and to push backwards against the resistance of Course of the long thoracic nerve to
your hand. The muscle bellies of the rhomboids can be felt serratus anterior
medial to the medial border of the scapula and sometimes • Arises from the motor roots of C5, C6 and commonly
seen (755). also from C7.
• Courses downward through and in front of the medial
DIFFERENTIAL DIAGNOSIS, INVESTIGATIONS, scalenus muscle, and descends further dorsal to the
TREATMENT AND PROGNOSIS brachial plexus, along the medial wall of the axilla and
As for neuralgic amyotrophy (see p.595). innervates the serratus anterior muscle.

ETIOLOGY
• Inherited brachial plexus neuropathy: rare.
• Trauma:
– Carrying heavy loads on the back (‘rucksack paralysis’).
– Pushing loads above the head.
– Fall on outstretched arms.
– Trauma to shoulder or lateral part of the chest.
• Athletic activities:
753 – Archery.
– Ballet.
– Volleyball.
• Surgery to the chest wall:
– Transaxillary breast augmentation.
– Axillary node dissection (malignant melanoma, breast
carcinoma).
– Thoracostomy for pneumothorax.
– Scalenotomy and cervical/first rib resection for thoracic
outlet syndrome.
• Chiropractic manipulation.
• Neuralgic amyotrophy (see p.595): often involves
additional nerves.
• Borrelia infection (Lyme disease): rare.
• Radiation therapy: for breast cancer.

HISTORY
754 • Difficult elevating the upper arm (e.g. shaving or
combing hair).
• Dull shoulder ache, mainly because of strain on the
shoulder muscles and ligaments in the absence of the
serratus anterior muscle tightening the scapula against
the rib cage.

EXAMINATION
• Stand behind the patient and inspect for winging of the
scapula, which may be present in the resting position.
Then, ask the patient to push against a wall with both
arms slightly flexed at the elbow, or elevate the arms to
a forward position.
• Weakness is indicated by winging of the scapula on the
affected side(s) (and sometimes also abduction of the
arm) (735, 756).
753, 754 T1W coronal pre- (753) and T1W coronal post contrast
(754) MRI of the brachial plexus showing a neuroma. Note the
discrete mass closely related to the nerves in the left supraclavicular
fossa which enhances following contrast (arrows).
Long Thoracic Neuropathy 617

• If weakness is so severe that the patient cannot flex the • If there has been axonal damage more than 2–3 weeks
extended arm at the shoulder, do this for the patient and previously, spontaneous muscle fiber potentials in the
then ask the patient to push the fist forward against your form of fibrillation potentials, positive sharp waves, or
other hand. Look for winging of the scapula during this both may be seen and heard (‘denervation activity’).
maneuver. During voluntary contraction, there will be poor
• Elevation of the arm may become possible if you press recruitment of motor units, and a reduction in the EMG
the patient’s scapula against the chest wall, and thereby pattern. If partial denervation is followed by collateral re-
take over the function of the paralysed serratus anterior. innervation, enlarged and polyphasic motor unit
potentials (MUPs) will be seen. Recent re-innervation is
DIFFERENTIAL DIAGNOSIS characterized by polyphasic MUPs with unstable
• C6 or C7 radiculopathy: but usually additional weakness configuration.
of extensors of the arms, wrist or fingers. • The EMG features of degeneration are only present in
• Myopathy: weakness is usually bilateral and involving the muscles innervated by the nerve.
additional muscles of the shoulder and upper arm.
• Disruption of the serratus anterior muscle: TREATMENT
– Rheumatoid arthritis. • Conservative.
– Fracture of the scapula. • Surgical stabilization of the scapula:
• Trapezius muscle weakness: may also cause winging of – Wait until at least 2 years have elapsed before even
the scapula (upper part), particularly on abduction of the considering surgery (i.e. await spontaneous recovery).
arm. – Shoulder function usually remains impaired despite the
operation.
INVESTIGATIONS – The fixation may give way after several years.
Nerve conduction studies
Motor latencies from stimulation at Erb’s point to the PROGNOSIS
serratus anterior may be prolonged. • Depends on the cause and severity of the neuropathy.
• Usually recovers spontaneously in most patients with
EMG neuralgic amyotrophy or partial traumatic injury, even
• Place your (the examiner’s) index finger and ring finger following complete loss of function.
in the intercostal spaces on either side of the fifth (or
sixth) rib between the anterior and middle axillary lines
(to avoid puncture of the intercostal muscles and pleura)
and introduce the needle electrode between these fingers
at an acute angle until it touches the rib, and then
withdraw it slightly.

755 756

Patient

Examiner

755 The patient is pressing the palm of the hand backwards against 756 The patient is pushing against a wall and there is winging of the
the examiner’s hand.The muscle bellies of the rhomboids (arrow) can right scapula due to weakness of the right serratus anterior muscle.
be felt and sometimes seen.
618 Mononeuropathies

SUPRASCAPULAR NEUROPATHY • Idiopathic:


– Acute onset: presumably autoimmune, inflammatory
neuropathy.
DEFINITION – Subacute onset: possible entrapment by transverse
Dysfunction of the suprascapular nerve. superior or inferior scapular ligament.
• Local mass:
EPIDEMIOLOGY – Ganglion cyst.
Uncommon. – Hematoma associated with fracture.
– Sarcoma.
ANATOMY – Chondrosarcoma.
Course of the suprascapular nerve (757) – Metastases.
• Arises from upper trunk of brachial plexus, carrying fibers
from the C5 and C6 nerve roots. HISTORY
• Passes under the trapezius muscle and courses from the • Precipitating factors may include vigorous exercise, such
upper border of the scapula beneath the transverse as baseball, tennis, or volleyball; and prolonged cradling
superior scapular ligament. of a mobile telephone between an ear and a shoulder.
• Innervates supraspinatus and infraspinatus muscles. • Pain at the superior margin of the scapula, radiating
• Fibers to infraspinatus pass separately through the towards the shoulder, is common but not invariable; it
spinoglenoid notch, which is covered by the transverse may be painless.
inferior scapular ligament. • Difficulty with movements of the shoulder may be
present.
Entrapment sites (757)
• Suprascapular notch. EXAMINATION
• Spinoglenoid notch. • Stand behind the patient and inspect the supra- and
infraspinatus muscles from behind and above, for
ETIOLOGY wasting/atrophy.
• Trauma: • Test the supraspinatus muscle by asking the patient to
– Heavy lifting. abduct the upper arm, from the adducted position,
– Blow on shoulder. against resistance (758). This is because the first 30° of
• Athletic activities: forceful and extreme movement of the abduction of the upper arm are effected by the
shoulder (ballet [professional], baseball, boxing, fencing, supraspinatus muscle (not the deltoid muscle). The
pitching, tennis, volleyball). muscle belly can be felt and sometimes seen.

757
Suprascapular nerve

Compression
C5 sites
C6

Supraspinatus
muscle

Infraspinatus
muscle

757 Course of the suprascapular nerve and sites of compression.


Suprascapular Neuropathy 619

• Test the infraspinatus muscle by asking the patient to EMG


externally rotate the upper arm at the shoulder (from the • Needle examination of the supra- and infraspinati may
adducted position, with the elbow flexed) against help identify the presence and severity of denervation
resistance (759). The examiner supports the patient’s activity in these muscles.
elbow and prevents abduction of the arm by holding the • Needle examination of other muscles may distinguish
elbow with one hand and resisting the movement of lesions of the suprascapular nerve from lesions of the
external rotation of the shoulder with the other hand cervical nerve roots or brachial plexus.
placed around the patient’s wrist.
• There should be no restriction of passive shoulder MRI scan
movements and the deltoid muscle is of normal size and If no improvement with rest, scan to exclude local
strength (cf. soft tissue injury of the shoulder), except for compression by a mass lesion.
a small minority of cases in which the suprascapular nerve
lesion may cause a frozen shoulder. DIAGNOSIS
• Increasing pain upon moving the arm across the chest is Requires the presence of focal neurologic deficits, and not
non-specific and also occurs with other painful disorders just regional pain (as with entrapment syndromes of other
of the shoulder. peripheral nerves).

DIFFERENTIAL DIAGNOSIS TREATMENT


Non-neurogenic disorders of the shoulder Depends on the cause:
• Soft tissue injuries of the shoulder (‘frozen shoulder’) • Conservative:
which lead to pain, inhibited mobility of the shoulder – Await natural history.
joint, wasting and weakness of muscles around the – Avoid provoking activities if possible.
shoulder (especially the supraspinatus but also the – Hydrocortisone injections may be effective but
deltoid). unproven.
• Rotator cuff syndrome. • Surgical decompression: resection of the transverse
• Tendinitis of the supraspinatus muscle. suprascapular ligament ± widening of the spinoglenoid
notch.
Neurogenic
• C5 or C6 radiculopathy: but usually pain radiates into PROGNOSIS
the arm, and the biceps reflex is diminished. • Depends on the cause and severity of the neuropathy.
• Neuralgic amyotrophy: may be confined to one or both • Following surgery, about half of patients experience
of the spinatus muscles and, if so, is probably auto- immediate pain relief, and most of the remainder recover in
immune rather than due to entrapment. the next few months.

INVESTIGATIONS
Nerve conduction studies
Motor latencies from stimulation of the brachial plexus at
Erb’s point to the supraspinatus may be prolonged.

758 759

Patient Patient

Examiner
Examiner

758 Testing the supraspinatus muscle.The arm is abducted against 759 Testing the infraspinatus muscle.The upper arm is externally
resistance. rotated against resistance.
620 Mononeuropathies

AXILLARY NEUROPATHY HISTORY


• Weakness of abduction of the shoulder >30° above the
horizontal.
DEFINITION • Precipitating injury (see above).
Dysfunction of the axillary nerve.
EXAMINATION
EPIDEMIOLOGY • Wasting (atrophy) of the deltoid muscle, evident
Uncommon. particularly when the patient is seated and inspected from
above and behind.
ANATOMY • Prominence of the acromion and head of the humerus
Course of the axillary nerve (due to deltoid wasting).
• Arises from the posterior cord of brachial plexus, carrying • Weakness of the anterior and middle parts of the deltoid
fibers from the C5 and C6 nerve roots. muscle, elicited by asking the patient to keep the arm
• Passes just below the shoulder joint, encircles the abducted in the horizontal plane against resistance. (N.B.
humerus from behind until it is under the deltoid muscle The first 30° of abduction of the upper arm are effected
at the posterior aspect of the shoulder and then passes by the supraspinatus muscle.)
through the quadrilateral space, defined by teres minor • Weakness of the posterior part of the deltoid, elicited by
above, teres major below, the long head of the triceps retracting the abducted arm against resistance.
medially, and the neck of the humerus laterally. • The teres minor muscle cannot be examined in isolation
• Innervates the deltoid and teres minor muscles. because it acts together with the infraspinatus muscle
• The sensory branch, the upper lateral cutaneous nerve of (suprascapular nerve) in external rotation of the upper
the upper arm, follows a short and separate route, and arm.
innervates a small area of skin overlying the deltoid • Loss of sensation to light touch and pin prick in a small
muscle. area of skin overlying the deltoid muscle (760).

ETIOLOGY DIFFERENTIAL DIAGNOSIS


• Trauma: Non-neurogenic disorders of the shoulder
– Subcapital fracture of the humerus. • Soft tissue injuries of the shoulder (‘frozen shoulder’)
– Anterior-inferior dislocation of the shoulder joint (e.g. which lead to pain, inhibited mobility of the shoulder
blow at the tip of the shoulder) or attempted joint, wasting and weakness of muscles around the
repositioning of this type of dislocation. shoulder (especially the supraspinatus but also the
– Blunt trauma of the shoulder. deltoid).
• Iatrogenic: • Rotator cuff syndrome.
– Intramuscular injections into deltoid.
– Faulty positioning on operating table with upper arm Neurogenic
elevated to 90° for 4.5 hours. • C5 radiculopathy: but usually pain radiating into the
– Birth injury. arm, and the biceps reflex is diminished.
• Athletic activities: • Neuralgic amyotrophy: may be confined to one or both
– Volleyball. of the spinatus muscles and, if so, is probably auto-
immune rather than due to entrapment.

760 INVESTIGATIONS
Nerve conduction studies
Motor latencies from stimulation of the brachial plexus at
Erb’s point to the deltoid muscle may be prolonged.

EMG
• Needle examination of the deltoid muscle, and perhaps
also the teres minor muscle (which is difficult to localize)
may help identify the presence and severity of
denervation activity in these muscles.
• Needle examination of other muscles may distinguish
lesions of the axillary nerve from lesions of the cervical
nerve roots or brachial plexus.

X-ray of shoulder and upper humerus


Performed if an history of trauma, or joint deformity.

760 Area of skin innervated by the axillary nerve (upper lateral


cutaneous nerve of the upper arm).
Musculocutaneous Neuropathy 621

DIAGNOSIS MUSCULOCUTANEOUS
Requires the presence of isolated weakness of the deltoid NEUROPATHY
(and teres minor muscles) and sensory loss over the deltoid.
Can be confirmed by EMG.
DEFINITION
TREATMENT Dysfunction of the musculocutaneous nerve.
Depends on the cause:
• Conservative: mobilize the shoulder joint by active and, EPIDEMIOLOGY
if necessary, passive exercises if the deltoid is weak, to Very rare in isolation.
avoid a frozen shoulder syndrome (particularly in older
patients). ANATOMY
• Surgical: nerve grafting should be considered in patients Course of the musculocutaneous nerve (761)
with axillary neuropathy due to trauma if there are no • Arises from the lateral cord of brachial plexus, carrying
signs of recovery of paralysis of the deltoid muscle after fibers from the C5, C6 and C7 nerve roots.
>4 months. • Passes through the axilla, pierces the coracobrachialis
muscle (giving off branches to it), descends between the
PROGNOSIS biceps and brachialis muscles (giving off branches to both
Depends on the cause and severity of the neuropathy: parts of the biceps muscle and the brachial muscle) and
• Partial lesions tend to recover spontaneously. continues as the lateral cutaneous nerve of the forearm,
• Neuralgic amyotrophy (an autoimmune inflammatory which pierces the fascia lateral to the tendon of the biceps
response) may recover very slowly over many months. muscle and just above the elbow, and innervates the skin
of the radial part of the volar side of the forearm as far as
the wrist.
• Innervates the coracobrachialis, biceps and brachialis
muscles.
• The sensory branch, the lateral cutaneous nerve of the
forearm, innervates the skin of the radial part of the volar
side of the forearm as far as the wrist.

ETIOLOGY
• Trauma:
– Dislocation of the shoulder joint.
761 – Fracture (closed) of the clavicle.
– Shoulder operations for habitual luxation (dislocation)
or instability of the clavicle.
– Axillary node dissection for malignant melanoma or
breast carcinoma.
Coracobrachialis – Penetrating injury of the upper arm (gunshot or knife).
• Athletic activities: strenuous exercise of the arms (e.g.
weight-lifting, repetitive push-ups [e.g. 500 times]).
Musculocutaneous • Neuralgic amyotrophy.
nerve • Opportunistic infection by Capnocytophaga canimorsus,
probably of the vasa nervorum.
Biceps • Compression:
– Sleep.
– Excessive exercise with elbow extension and forearm
pronation.
Brachialis
HISTORY
• Numbness or paresthesia of the lateral/radial part of the
forearm.
• Pain in the elbow and forearm may be present.
• Weakness of flexion of the elbow (with the forearm
supinated).
• Precipitating injury (see above).

761 Diagram of the musculocutaneous nerve and the muscles which


it supplies.
622 Mononeuropathies

EXAMINATION DIAGNOSIS
• Weakness of the biceps and brachialis muscles, elicited by Requires the presence of isolated weakness of the biceps,
flexion of the elbow, with the forearm in full supination, brachialis, and coracobrachialis and sensory loss over the
against resistance. Also there is some weakness of lateral (radial) border of the forearm. Can be confirmed by
supination of the forearm with the elbow in flexion EMG.
(biceps muscle).
• Weakness of the coracobrachial muscle may be present TREATMENT
exceptionally, evident by some weakness on elevation of Depends on the cause:
the arm. • Conservative.
• Loss of superficial sensation on the skin of the radial part • Surgical: only if direct penetrating trauma with severe
of the volar side of the forearm as far as the wrist (762). axonal injury or complete interruption of nerve
continuity.
DIFFERENTIAL DIAGNOSIS
Non-neurogenic PROGNOSIS
Ruptured biceps tendon: no sensory loss, and on Depends on the cause and severity of the neuropathy; partial
contraction of the biceps muscle a hardening mass evolves lesions tend to recover spontaneously.
under the insertion of the pectoralis major muscle.

Neurogenic
C6 radiculopathy: but this is usually accompanied by
sensory loss in the hand; weakness of supination with the
forearm extended (supinator muscle [radial nerve]), and
wrist extension (at least toward the radial side [extensor
carpi radialis muscle]), and absence of an obviously visible
muscle belly of the brachioradialis muscle on flexion of the
elbow (radial nerve).

INVESTIGATIONS
Nerve conduction studies
• Decreased or absent sensory nerve action potential
(SNAP) and delayed sensory conduction in the lateral
cutaneous nerve of the forearm as measured by means of
an antidromic technique. A decreased or absent SNAP
reflects axonal degeneration distal to the spinal ganglion;
sensory root lesions proximal to the spinal ganglion cause
no degeneration of the peripheral sensory axon, and 762
thereby do not influence the SNAP.
• Motor latencies from stimulation of the brachial plexus
at Erb’s point to the biceps muscle may be prolonged.

EMG
• Needle examination of the biceps, brachialis, and
coracobrachialis muscles may help identify the presence
and severity of denervation activity in these muscles.
• Needle examination of other muscles may distinguish
lesions of the musculocutaneous nerve from lesions of
the C6 cervical nerve root or brachial plexus.

X-ray of clavicle, shoulder and upper arm


Performed if an history of trauma, or joint deformity.

762 Area of skin innervated by the musculocutaneous nerve (lateral


cutaneous nerve of the forearm).
Radial Neuropathy 623

RADIAL NEUROPATHY Wrist


The superficial terminal branch of the radial nerve passes
superficially on the lateral/radial side of the forearm, over
DEFINITION the styloid process just proximal to the wrist (where it lies
Dysfunction of the radial nerve. exposed and is easily compressed), and towards the dorsum
of the thumb. It supplies the lateral/radial part of the
EPIDEMIOLOGY dorsum of the hand and ends in five dorsal digital nerves,
Reasonably common. of which two supply the dorsum of the thumb (except the
nail area), two supply the dorsum of the index finger
ANATOMY (proximal to the middle phalanx), and one supplies the first
Course of the radial nerve (763) phalanx of the middle finger.
Arises (together with the axillary nerve) from the posterior
cord of the brachial plexus, carrying fibers from the C5, C6, ETIOLOGY
C7, C8 and T1 nerve roots. Axillary lesions
• Rare.
Axilla • Compression: long crutches, but usually also involves the
Courses through the axilla, giving off branches to the triceps median and ulnar nerves.
muscles, then down between the medial and lateral heads • Local trauma.
of the triceps muscle, before winding around the back of the
mid-humerus, in the spiral groove (where it is most often
damaged by compression, particularly the motor fibers).

Upper arm
In the spiral groove, two sensory nerves, the posterior
cutaneous nerve of the upper arm and the posterior
cutaneous nerve of the forearm, leave the radial nerve to
supply a small area of skin on the dorsal aspect of the upper
Axillary nerve
763
arm and a larger area of the skin on the dorsal aspect of the
forearm respectively.
At the distal third of the upper arm, the nerve gives off
Deltoid
the lower lateral cutaneous nerve of the upper arm which
innervates the skin of the lateral and posterior surface of the
distal third of the upper arm and a small portion of the back Teres minor
of the proximal forearm. The radial nerve then pierces the
intermuscular septum between the brachialis muscle and Triceps, long head
lateral head of triceps, and gives off branches to the
brachioradialis muscle and extensor carpi radialis longus and Triceps, lateral head
brevis. Triceps,
medial head
Within 3 cm (1.2 in) of the humero-radial joint (above
or below it) the nerve divides into a deep motor branch
(which continues as the posterior interosseous nerve) and a
superficial sensory branch (the superficial branch of the Radial nerve
radial nerve).
Brachioradialis
Forearm
The posterior interosseous nerve passes through and
Extensor carpi radialis longus
supplies the supinator muscle and then runs dorsal to the
interosseous membrane of the forearm to innervate the Extensor carpi radialis brevis
extensor carpi ulnaris, all extensor muscles of the fingers and Spinator Posterior
thumb, and abductor pollicis longus muscle. Extensor carpi ulnaris interosseous
Extensor digitorum nerve
Extensor digiti minimi
Abductor pollicis longus
Extensor pollicis longus
Extensor pollicis brevis
Extensor indicis

763 Diagram of the axillary and radial nerves and the muscles which
they supply.
624 Mononeuropathies

Upper arm lesions Finger lesions (dorsal digital nerves)


• Trauma: supracondylar fracture of the humerus. • Professional and daily use of scissors (thumb).
• External compression against the spiral groove: • Palmar ganglion.
– Intoxicated sleep (e.g. with the arm folded over the back
of a chair or resting on a hard ridge [‘Saturday night HISTORY
palsy’, ‘Parkbanklähmung’, paralysie des ivrognes’]). Axillary lesions
– Improper positioning during general anesthesia. • Short history of weakness stretching the elbow, wrist, all
– Prolonged (e.g. 3 hours) shooting practice in a kneeling fingers and thumb.
position with the upper arm resting on the ipsilateral knee. • Pain is not prominent.
– Akinetic rigid syndromes (e.g. Parkinson’s disease)
causing severe immobility. Upper arm lesions
– Hereditary liability to pressure palsies (see p.587): palsy • Often sudden onset of inability to extend wrist, fingers
after normal sleep. and thumb.
– Lipoma adjacent to the nerve. • Numbness or paresthesia of the lateral/radial part of the
– Traumatic aneurysm of the radial artery. forearm.
– Callus bone formation following fracture of the shaft of • Pain in the elbow and forearm may be present.
the humerus. • Precipitating injury or predisposing factors (e.g. alcohol
– Myositis ossificans of the shaft of the humerus. or drug intoxication; see above).
– Prolonged labor or forceps extraction in the neonate, or
repeated blood pressure measurement in the premature Forearm (posterior interosseous nerve) lesions
infant. (supinator syndrome)
• Athletic activities (compression by lateral head of triceps • Slowly progressive onset of symptoms.
or a fibrous arch at the lower part of the humeral • Initially, difficulty stretching the little finger (it gets
groove): curled up during tasks such as retrieving something from
– Involving extension at the elbow against strong a trouser pocket).
resistance. • Later, inability to extend metacarpophalangeal joint of
– ‘Windmill’ pitching motion of competitive softball. the little finger and then similar weakness begins in other
• Medical mononeuropathy: fingers, one after the other.
– Diabetes mellitus. • Consequently, difficulty playing the piano but writing
– Arteritis. remains normal and grip powerful if the fingers are
• Nerve tumor. passively placed around an object.
• Pain is uncommon.
Forearm (posterior interosseous nerve) lesions • Bilateral symptoms may occur.
• Congenital hemihypertrophy of the supinator muscle.
• Entrapment of the motor branch of the radial nerve, the Wrist lesions
deep radial nerve or the posterior interosseous nerve, • Shooting pain in the radial side of the wrist.
between ‘normal’ anatomic structures, at the level of the • Painful paresthesia in the thumb and index finger evoked
supinator muscle. by touching or knocking the radial side of the wrist.
• Accessory brachioradialis muscle. • Reduced sensation on the radial side of the hand (764).
• Dislocation of the elbow, fracture of the ulna with
dislocation of the radial head, or Monteggias’s fracture.
• Arthroscopy of the elbow joint. 764
• Rheumatoid arthritis of the elbow joint.
• Traumatic aneurysm of the posterior interosseous artery.
• Arteriovenous fistula for hemodialysis.
• Lipoma.
• Intramuscular myxoma.
• Cysts.
• Ganglia.

Wrist lesions (superficial terminal branch


of radial nerve)
• Handcuffs and other tight wrist bands (e.g. watch band).
• Direct injury:
– Accidental.
– Post-surgery:
Stenosing tenosynovitis (de Quervain’s disease).
Open reduction of fractures of radius and ulna.
Shunt operations for hemodialysis.
• Transposition of a flexor tendon towards the thumb.
• Repeated movements of pronation and supination of
forearm, or abduction and adduction of the wrist with
the forearm in pronation and the wrist in flexion. 764 Areas of skin innervated by the radial nerve (superficial terminal
• Stenosing tenosynovitis (de Quervain’s disease). branch of radial nerve).
Radial Neuropathy 625

EXAMINATION Wrist lesions


Axillary lesions Reduced sensation over the lateral/radial part of the dorsum
• Weakness of the triceps and all muscles extending wrist, of the hand, and the dorsum of the thumb (except the nail
fingers and thumb. area), index finger (proximal to the middle phalanx) and
• Mild decreased sensation on the back of the upper arm first phalanx of the middle finger.
and forearm, in the web between index finger and
thumb, and the radial side of dorsum of the hand.

Upper arm lesions


• Dropped hand and fingers due to weakness of extensors
of wrist and metacarpophalangeal joints (765, 766).
• Spares triceps muscle and sensation in the upper arm, and
often posterior cutaneous nerve of forearm leading to
preserved sensation in the web between the index finger
and thumb.
• Weakness of brachioradialis (elbow flexion with forearm
pronated [not supinated cf. biceps]), supinator
(supination of forearm with elbow extended [not flexed,
cf. biceps]).
765
Forearm (posterior interosseous nerve) lesions
(supinator syndrome)
• Dropped fingers without dropped hand: inability to
extend the fingers and thumb at the metacarpophalangeal
joints. Despite severe weakness of extensor carpi ulnaris,
wrist extension remains possible because extensor carpi
radialis muscles function normally (because the branch to
extensor carpi radialis leaves the main stem of the radial
nerve above the elbow and proximal to entry of the
supinator muscle). If extensor carpi ulnaris is weak, a
distinct radial deviation of the extended hand occurs when
the patient is asked to make a fist.
• There may be some extension of the index finger at the
interphalangeal joints (by contraction of the lumbrical
muscles innervated by the median nerve) and some
weakness of the supinator muscle, but brachioradialis
remains powerful (unless the lesion is above the elbow).
• The interossei may appear to be weak (although they are
not weak) with any radial nerve lesion above the wrist
because of weakness of finger extension; in order for the
fingers to be abducted they need to be extended first. If
the fingers are supported on a flat surface, the action of
the interossei can be assessed and, in the event of a radial
nerve lesion, the fingers can spread apart (to some extent
at least), whereas in an upper motor neuron lesion, a T1
root lesion, or a combined lesion of the radial and ulnar
nerves, there is weakness of finger abduction.

766

765, 766 Upper arm scar indicating the


site of injury to the radial nerve (765), and
dropped hand and fingers due to weakness
of extensors of wrist and
metacarpophalangeal joints following upper
arm radial nerve injury (766).
626 Mononeuropathies

DIFFERENTIAL DIAGNOSIS EMG


Neurogenic • Needle examination of muscles innervated by the radial
• Central weakness (upper motor neuron lesions): dropped nerve may help identify the presence and severity of
hand and fingers but also weakness of the radial wrist denervation activity in these muscles, and help localize
extensors and interossei, an involuntary extension of the the site and severity of a radial nerve lesion; fibrillation
wrist when making a fist or gripping an object, and other potentials and other signs of axonal degeneration may be
upper motor neuron signs such as increased limb tone found in all or some of the muscles supplied by the radial
and deep tendon reflexes and an extensor plantar nerve that are distal to the nerve lesion. For example,
response. EMG evidence of denervation in the extensor capri
• Spinal muscular atrophy: not infrequently begins with ulnaris (and more distal muscles innervated by the radial
partial deficits, such as weakness of extensors of the wrist, nerve) but not in the extensor carpi radialis longus and
but weakness is often present in muscles of the hand and brevis is consistent with the supinator syndrome.
forearm beyond the innervation of the radial nerve. • Needle examination of other muscles may distinguish
• C7 radiculopathy: weakness in the triceps and extensors lesions of the radial nerve from lesions of the C7 cervical
of the fingers but the wrist extensors (mainly C6 root) nerve root, brachial plexus or upper motor neuron.
are mostly spared, the triceps reflex is depressed, and the
history almost invariably begins with pain (mainly in the X-ray of shoulder, humerus, elbow joint
neck and extensor aspect of upper arm) and pins and Performed if a history of trauma, or joint deformity.
needles in the middle (and index) finger.
• Neuralgic amyotrophy (see p.595). DIAGNOSIS
Requires the presence of isolated weakness of muscles and
Non-neurogenic loss of sensation of skin innervated by the radial nerve.
• Diseases of extensor tendons.
• Compartment syndrome of the deep extensor muscles of TREATMENT
the forearm. Upper arm lesions
Depends on the cause, but generally conservative with
INVESTIGATIONS functional aids (767).
Nerve conduction studies
• Decreased or absent sensory nerve action potential Surgical exploration
(SNAP) and delayed sensory conduction in the lateral • If slowly progressive radial nerve palsy (e.g. if nerve
cutaneous nerve of the forearm occurs as measured by compression by a structural local mass lesion such as
means of an antidromic technique. A decreased or absent callus bone formation, myositis ossificans, aneurysm of
SNAP reflects axonal degeneration distal to the spinal the radial artery, or lipoma).
ganglion; sensory root lesions proximal to the spinal • If direct penetrating trauma with severe axonal injury or
ganglion cause no degeneration of the peripheral sensory complete interruption of nerve continuity (e.g.
axon, and thereby do not influence the SNAP. immediate and complete radial nerve palsy after a
• Motor latencies from stimulation of the brachial plexus complex fracture of the humerus).
at Erb’s point to the biceps muscle may be prolonged. • Possibly for immediate and complete radial nerve palsy
The main trunk of the radial nerve and its final motor after a closed fracture of the humerus, but remembering
(and sensory) branches can be stimulated at several sites, that MRI and even CT provide a lot of information and
so that conduction can be studied in all segments of that recovery is the rule even in radial nerve palsies
interest. immediately after a fracture of the humerus, particularly
if nerve conduction studies show that the deficit is mainly
caused by conduction block.

Conservative
Partial radial nerve lesion (e.g. following humeral fracture).

Forearm (posterior interosseous nerve) lesions


767 Surgical exploration
If the nature of the nerve lesion has been shown to be
consistent with compression, and the site established,
decompression surgery is likely to be effective.

PROGNOSIS
Depends on the cause and severity of the neuropathy: partial
lesions tend to recover spontaneously and over about 6–8
weeks. Neurophysiologic studies can help determine
prognosis.

767 Forearm splint to facilitate hand function.


Median Neuropathy 627

MEDIAN NEUROPATHY Anomalies


• Fibers from the median nerve in the forearm may cross
to the ulnar nerve (the Martin–Gruber anastomosis) in
DEFINITION 15–30% of normal individuals and almost all, if not all,
Dysfunction of the median nerve. with Down’s syndrome. The most common variety, in
60% of cases with this variant, is that fibers of the anterior
EPIDEMIOLOGY interosseous nerve travel with the ulnar nerve to
Common. innervate muscles normally innervated by the median
nerve. In 35% of cases, the anastomosing fibers from the
ANATOMY median nerve supply muscles normally innervated by the
Course of the median nerve (768) ulnar nerve (e.g. adductor pollicis and the first dorsal
Arises from the lateral and medial cords of the brachial interosseous).
plexus, carrying fibers from the C5–C8 and T1 nerve roots. • The median nerve may innervate the hypothenar muscles
via an anomalous branch, arising from its course in the
Axilla carpal tunnel.
Emerges from the axilla with the radial and ulnar nerves and • An ‘all median hand’ may occur rarely.
the axillary artery and vein, through the inelastic axillary • The deep motor branch of the ulnar nerve may
sheath. communicate with the median nerve in the hand
(Riche–Cannieu anastomosis).
Upper arm • The flexor pollicis brevis may be innervated entirely by
Descends through the upper arm and bicipital sulcus, giving the ulnar nerve or have double innervation by the
off no branches, to the elbow, where it lies medial to the median and ulnar nerves.
brachial artery. • An anastomotic branch of the radial nerve may innervate
the abductor pollicis brevis muscle very rarely.
Elbow • The musculocutaneous nerve may innervate muscles
Courses through the antecubital fossa, medial to the biceps normally innervated by the median nerve.
tendon and beneath the bicipital aponeurosis, where it gives
off branches to the pronator teres, flexor carpi radialis,
palmaris longus and flexor digitorum superficialis muscles. 768
Forearm
Proximally, the nerve lies deep, between the two heads of
the pronator teres muscle. Distal to the pronator teres the
anterior interosseous nerve arises. It is a motor nerve which
descends anterior to the interosseous membrane (joining
the radius and ulna) and between the flexor digitorum
profundus (I and II) and flexor pollicis longus, which it Median nerve
innervates, together with the pronator quadratus muscle.

Wrist and hand


• A few centimeters proximal to the wrist, the palmar
cutaneous branch leaves the main trunk of the median
nerve and travels over the transverse carpal ligament to
the thenar eminence. It innervates the skin on the
proximal half of the radial side of the palm.
• Immediately proximal to the wrist the median nerve Pronator teres
Anterior
becomes more superficial and enters the carpal tunnel. Flexor carpi radialis interosseous
Palmaris longus nerve
• Within, or distal to, the carpal tunnel the recurrent
Flexor digitorum
motor branch to the thenar eminence arises and supeficialis
Flexor digitorum
innervates the abductor pollicis brevis, opponens pollicis, profundus I, II
and flexor pollicis brevis muscles. More distal branches Flexor pollicis
supply the superficial head of the flexor pollicis brevis and longus
the first and second lumbrical muscles.
• The digital nerves innervate the skin on the volar aspect Pronator
of the thumb, index finger, middle finger, radial half of quadratus
the ring finger, and the adjoining portion of the palm; Abductor pollicis brevis
and the dorsal aspect of the entire middle finger, and Flexor pollicis brevis
middle and terminal phalanges of the index and ring Opponens pollicis
finger (radial half). First lumbrical Second lumbrical

768 Diagram of the median nerves and the muscles which it supplies.
Note: the white rectangle signifies that the muscle indicated receives a
part of its nerve supply from another peripheral nerve.
628 Mononeuropathies

ETIOLOGY • Intrinsic lesions of the skin, tendons or bony structures


Axillary lesions of the fingers.
• Trauma.
• Iatrogenic: HISTORY
– Repositioning of shoulder luxation. Pronator teres syndrome
– Axillary angiography causing a false aneurysm or • Pain along the ventral surface of the proximal forearm.
inflammatory fibrosis. • Paresthesia in the median nerve distribution in the hand
• Lipoma. (thumb, index, middle and ring fingers), but not as
intense as that experienced in carpal tunnel syndrome
Upper arm lesions (see p.630).
• Tourniquet paralysis.
• Nerve tumor. Anterior interosseous syndrome
• Soft tissue tumor: non-Hodgkin lymphoma. • Subacute onset of weakness of the index finger and
• Closed reduction after supracondylar fracture of the thumb.
humerus. • Pain may be present.
• Sleep paralysis from pressure of a partner’s head on the
upper arm (‘honeymoon palsy’ or ‘paralysie des Carpal tunnel syndrome (see p.630)
amoureux’).
Palmar cutaneous branch of median nerve
Elbow lesions Pain, paresthesia and numbness in the palm, particularly the
Entrapment thenar region.
• Between the two heads of the pronator teres muscle
(pronator teres syndrome). Digital branches of the median nerve
• By a fibrous band between the deep head of the pronator Reduced sensation in adjoining halves of two fingers and in
muscle and the flexor digitorum superficialis (anterior the web space between them (common digital nerve lesion),
interosseous syndrome): rare, <1% of median neuropathies. or in one-half of a single finger (proper digital nerve lesion).

Compression EXAMINATION
• Externally, by heavy objects being carried. Pronator teres syndrome
• Externally during general anesthesia. • Weakness in one or more muscles in the hand innervated
• Tight band for the treatment of lateral epicondylitis by the median nerve (e.g. abductor pollicis brevis, flexor
(‘tennis elbow’). pollicis longus, opponens pollicis, flexor digitorum
• Anterior interosseous artery. profundus I and II).
• Persistent median artery in the forearm. • Sensory loss in the entire median nerve distribution in
• Hematoma or false aneurysm after a puncture of the the hand (thumb, index, middle and ring fingers) or,
brachial artery. more commonly, restricted to the radial aspect of the
• Venepuncture or intravenous catheterization or injection index finger and ulnar side of the thumb (769).
in the forearm. • Pressure on the pronator teres muscle may produce pain
• Vascular shunts for hemodialysis. along the ventral surface of the proximal forearm or,
• Fibrous brand from the supracondylar process to the more convincingly, paresthesia in the median nerve
medial epicondyle of the humerus, or Struthers’ ligament. distribution in the hand.
• Fibrous band at the humeral head of the pronator
muscle. Anterior interosseous syndrome
• Synovial bursa from partial rupture of distal biceps • Weakness of flexion of the interphalangeal joint of the
brachii tendon. thumb (flexor pollicis longus) and terminal phalanx of
• Closed reduction of a dislocated elbow. the index finger (flexor digitorum profundus of the index
• Intra-articular fracture of the humerus. finger), and sometimes less severe weakness of pronator
quadratus (which should be tested with the elbow
‘Overuse’ of pronator muscle, probably in combination flexed).
with one of the anatomic factors listed above. • The patient is unable to form a small circle by pinching
the terminal phalanx of the thumb and index finger
Carpal tunnel lesions (see p.630) together: the so-called ‘pinch sign’ (770).
• The ‘straight thumb sign’ is elicited by asking the patient
Palmar cutaneous branch of the median nerve to attempt to grasp something.
• Ganglion of the flexor carpi radialis tendon, immediately • No sensory loss.
proximal to the wrist flexion crease.
• Entrapment within the antebrachial fascia. Carpal tunnel syndrome (see p.630)
• An atypical palmaris longus muscle.
• Iatrogenic injury following carpal tunnel release. Palmar cutaneous branch of the median nerve
• Loss of sensation in the skin of the proximal half of the
Digital branches of the median nerve radial side of the palm, particularly the thenar region.
• Frequent use of scissors (e.g. by orthopedic surgeons). • The Hoffmann–Tinel sign (distal tingling on percussion
• The rim of the hole in bowling balls (‘bowler’s thumb’). of the nerve) may be present.
Median Neuropathy 629

Digital branches of the median nerve Anterior interrosseous syndrome


• Sensory impairment in the adjoining halves of two • EMG abnormalities exclusively in the flexor digitorum
fingers and in the web space between them if injury to a profundus (I and II), flexor pollicis longus, and pronator
common digital nerve in the palm. quadratus muscles, without involvement of muscles of
• Sensory impairment in one-half of a single finger if injury the arm and shoulder (cf. neuralgic amyotrophy).
to a proper digital nerve distal to the division of a • Usually no slowing in conduction of the anterior
common digital nerve to the two fingers. interosseous nerve.
• Sensation at the tip of the finger may be spared, • Normal motor and sensory conduction in the main trunk
depending on the overlap with the proper digital nerve of the median nerve.
of the other half-finger.
X-ray of elbow joint, forearm, wrist
DIFFERENTIAL DIAGNOSIS Performed if an history of trauma, or joint deformity.
Neuralgic amyotrophy: weakness is preceded or
accompanied by acute or severe pain in the shoulder or DIAGNOSIS
upper arm. Requires the presence of isolated weakness of muscles and
loss of sensation of skin innervated by the median nerve,
INVESTIGATIONS confirmed by EMG.
Nerve conduction studies and EMG
Pronator teres syndrome TREATMENT
• Fibrillation potentials and other signs of axonal Depends on the cause.
degeneration may be found in the forearm and hand
muscles innervated by the median nerve, including the PROGNOSIS
pronator teres muscle itself. Therefore, EMG does not Depends on the cause and severity of the neuropathy: partial
allow differentiation from more proximal lesions of the lesions tend to recover spontaneously and over about 6–8
median nerve. weeks. Neurophysiologic studies can help determine the
• Nerve conduction studies are usually normal. prognosis.

769 770

769 Areas of skin innervated by the median nerve.

770 Weakness of flexion of the interphalangeal joint of the thumb


(flexor pollicis longus) due to an anterior interosseous nerve palsy
resulting in an inability to form a small circle by pinching the terminal
phalanx of the thumb and index finger together: the so-called ‘pinch
sign’ (i.e. the thumb is straight, not bent, and so this is only a semi-circle).
630 Mononeuropathies

CARPAL TUNNEL SYNDROME (CTS) CLINICAL FEATURES


Symptoms
• Paresthesia (e.g. tingling), with or without numbness and
DEFINITION pain, involving the palmar surface of the hand
A compression neuropathy of the median nerve at the wrist. (particularly the thumb, index, middle and ring fingers
innervated by the median nerve) and often extending
EPIDEMIOLOGY proximally into the forearm, arm and even neck.
• Incidence: 0.1% per year. • The symptoms are frequently worse at night (waking the
• Prevalence: at least 2.1% of the population: patient ‘brachialgia paresthetica nocturna’) and after use
– Symptoms (hand pain/numbness/tingling): 14.4% (95% of the hand, and characteristically are relieved by rapid
CI: 13–16%) of the general population. flexion and extension movements of the wrist (unlike the
– Clinically certain CTS: 3.8% (95% CI: 3.1–4.6%). discomfort associated with soft tissue injury and
– Neurophysiologic CTS: 4.9% (95% CI: 4.1–5.8%). arthritis).
– Clinical and neurophysiologic CTS: 2.7% (95% CI: • Often bilateral, but symptoms are usually markedly worse
2.1–3.4%). on one (mostly the dominant) side.
• Age: most common in middle age.
• Gender: M<F (1:1.4). Signs
Median nerve provocative tests
PATHOLOGY • Tinel’s sign (paresthesia in the median nerve territory
Focal demyelination (and, if severe enough, axonal elicited by gentle tapping/percussion over the flexor
degeneration) of the median nerve in the carpal tunnel retinaculum of the carpal tunnel).
beneath the flexor retinaculum at the wrist with deformation • Phalen’s sign (paresthesia in the median nerve territory
of the myelin lamellae by the mechanical stress. elicited by asking the patient to flex the wrists and keep
the wrists in forced flexion for 60 seconds).
PATHOPHYSIOLOGY • In most cases, these maneuvers are negative.
Chronic increased tissue pressure within the carpal tunnel,
causing chronic focal compression of the median nerve, with Severe nerve compression
deformation of the myelin lamellae, and probably also nerve • Sensory loss over some or all of the digits innervated by
ischemia (which could account for the intermittent the median nerve (thumb, index, middle and
paresthesia that occurs at night or with wrist flexion). radial/lateral half of ring fingers).
• Wasting and weakness of abductor pollicis brevis (thumb
ETIOLOGY abduction), flexor pollicis brevis (thumb flexion at the
• Idiopathic: 45% of cases of CTS. interphalangeal joint), and opponens pollicis (opposition
• Obesity (body mass index ≥25 kg/m2): present in of thumb to little finger) (771, 772).
45–70% of cases of CTS.
• Pregnancy, hormonal agents (e.g. oral contraceptive pill), DIFFERENTIAL DIAGNOSIS
and oophorectomy: present in 10% of cases of CTS. • Cervical (C6, C7, C8) radiculopathy.
• Active blue collar workers (3.5%; vs. active white collar • Brachial plexopathy.
workers: 1.7%; p=0.03); hard work with hands and • Proximal median neuropathy.
repetitive wrist movements and vibrations. • Diffuse peripheral neuropathy.
• Diabetes: present in 3–6% of cases of CTS. • Motor neuron disease.
• Rheumatoid arthritis and other inflammatory joint • Spondylotic myelopathy.
diseases of the wrist: 2–5%. • Syringomyelia.
• Thyroid disorders: 3%. • Multiple sclerosis.
• Wrist (Colles’) fracture or sprain: 10%.
• Carpal tunnel/hand surgery: 1%. INVESTIGATIONS
• Miscellaneous others: Nerve conduction studies
– Carpal bone hypertrophic osteoarthropathy. Ensure the temperature of the hands is at least 30°C (86°F).
– Acromegaly. • Median nerve distal sensory latency (index or middle
– Flexor tenosynovitis. finger-wrist).
– Anomalous flexor tendons. • Wrist-palm sensory conduction velocity.
– Proximal origin of lumbrical muscles. • Ulnar nerve distal sensory latency (fifth finger-wrist).
– Aberrant muscles. • Sensitivity only about 70% in diagnosing CTS based on
– Bifid median nerve. history and examination, and yet also a high rate of false-
– Persistence of median artery and other vascular variants. positives (about 76%).
– Cysts and tumors in the carpal tunnel.
– Amyloidosis. MRI of the median nerve in the carpal tunnel
– Hemodialysis. • May image the compressed median nerve.
Carpal Tunnel Syndrome (CTS) 631

DIAGNOSIS Ultrasound
The key to diagnosis is a careful history and examination, 20 sessions of ultrasound (1 MHz, 1.0 W/cm2, pulsed
including a search for the underlying cause. There is no mode 1:4, 15 minutes per session) applied to the area over
consensus as to whether CTS is a clinical or electrophysio- the carpal tunnel, on a once daily basis (5 sessions per week)
logical diagnosis, and no standard diagnostic criteria have for the first 2 weeks, and then twice weekly for the next 5
been established. However, normal electrophysiologic weeks, may result in satisfying short and medium term (6
studies do not rule out CTS. Indeed, carpal tunnel months) benefits for patients with mild to moderate
syndrome should be suspected on the history because it is idiopathic CTS. If effective, the mechanism may be an anti-
not until the condition is moderate to severe that relevant inflammatory effect of such treatment, or stimulation of
sensory and motor deficits are detectable on physical nerve regeneration or nerve conduction.
examination (and nerve conduction studies).
Yoga-based intervention
Clinically certain CTS A yoga-based intervention consisting of 11 yoga-postures
• Recurring nocturnal and/or activity-related pain, designed for strengthening, stretching, and balancing each
numbness or tingling involving palmar aspects of at least joint in the upper body, along with relaxation, given twice
two of the first four fingers at least twice weekly during weekly for 8 weeks may be associated with pain reduction
the preceding 4 weeks. and improvement in grip strength and the Phalen sign.
• Positive nerve percussion and/or wrist flexion tests.
• Median nerve sensory and/or motor deficit is supportive
but not necessary for the diagnosis. 771
Electrophysiologic CTS
Median-ulnar peak sensory latency difference ≥0.8 ms.

TREATMENT
Avoid or treat any precipitating/causal factors
• Limit repetitive wrist and hand movements.
• Achieve ideal body weight.
• Treat underlying disease (e.g. arthritis) appropriately (e.g.
with splinting and non-steroidal anti-inflammatory
drugs).
• Diuretics may be helpful, particularly for patients who
develop CTS late in pregnancy.

Immobilization with splints


Palmar wrist splints, maintaining the wrist in a neutral or 772
slightly extended position, and worn at night may be
appropriate and helpful, particularly when symptoms are
mainly nocturnal.

Local injection of corticosteroid and


anesthetic into the carpal tunnel
A single injection of 1 ml of fluid containing a long-acting
corticosteroid (methylprednisolone 40 mg) and local
anesthetic (lignocaine 10 mg) by a 3 cm long 0.7 mm
needle, inserted 4 cm proximal to the carpal tunnel (wrist
crease), between the tendons of the radial flexor muscle and
the long palmar muscle, and angled acutely (about 10–20°
depending on the thickness of the wrist) toward the carpal
tunnel improves the symptoms of CTS in about three-
quarters of patients at 1 month after treatment (compared
with 20% of patients who are not treated), and is still
effective at 1 year in half of patients. It is also safe; there are
no adverse effects. The purpose of the lignocaine is to afford
a painless injection and to ensure that the injection was
carried out properly by demonstrating diminished sensation
in the median nerve territory after the injection.
N.B. Corticosteroid injections into the carpal tunnel may
damage the median nerve and tendons, and any treatment
benefits may be of short duration. Success is less likely in
patients with structural abnormalities or with denervation 771, 772 Wasting of the thenar eminence of the left hand due to
on electromyography. weakness and wasting of the left abductor pollicis brevis as a result
of a median neuropathy at the left wrist due to compression in the
carpal tunnel.
632 Mononeuropathies

Surgical decompression (endoscopic, open) Elbow


Rationale Enters the ulnar groove behind the medial epicondyle of the
Aims to create the appropriate circumstances under which humerus, where the nerve is exposed and most easily injured.
the nerve can recover, and thus the symptoms resolve. It
does not aim to improve nerve function itself. The capacity Forearm
of the nerve to recover also depends on the age of the • Enters through the so-called ‘cubital tunnel’ which is
patient, coexisting disease, and the severity and duration of confined by ligaments and the medial epicondyle, and
the deficit. reduces in size when the elbow is flexed.
• The ulnar nerve courses between the two heads of the
Possible indications flexor carpi ulnaris muscles, giving off branches to this
• No improvement with conservative (non-surgical) muscle and to the flexor digitorum profundus III and IV
management. (ring and little finger).
• Deterioration clinically or on serial electrodiagnostic • The palmar cutaneous branch originates in the middle of
studies. the forearm and supplies the skin of the proximal part of
• Nerve conduction block localized on nerve conduction the hypothenar eminence.
studies. • The dorsal cutaneous branch arises about 5 cm (2 in)
proximal to the wrist crease and innervates the skin on
Possible outcomes the dorsal side of the hand on the ulnar side, extending
• Immediate benefit should be realized if ‘positive’ from the wrist to the little finger, proximal phalanx of the
symptoms (i.e. pain and paresthesiae) dominate in a ring finger, and part of the base of the middle finger.
patient with neurophysiologically confirmed CTS.
• Improvement will be slow, or not at all, if the main Wrist and palm
clinical features are ‘negative’ (i.e. loss of sensory and/or The ulnar nerve, together with the ulnar artery and vein,
motor function); axons regrow at only 1–2 mm enters Guyon’s canal, passing over the transverse carpal
(0.04–0.08 in)/day. The main goal of surgery in these ligament (the median nerve passes under it), and beneath the
patients is prevention of further loss of function. palmar fascia bridging the pisiform bone and the hamulus of
the hamate bone. Within Guyon’s canal, at the inner side of
Adverse effects the pisiform bone, it divides into its two terminal branches:
Some patients complain of a vague lack of strength in the the superficial terminal branch, which is almost purely sensory,
wrist following section of the flexor retinaculum. and the deep terminal branch, which is purely motor.
The superficial terminal branch innervates the skin on
PROGNOSIS the distal part of the hypothenar eminence and then
• Depends on the cause and outcome of treatment. terminates as digital nerves which supply the skin on the
• An increasingly recognized cause of work disability. palmar side of the little finger and ring finger, and the dorsal
side of the tips of these two fingers, distal to the parts
supplied by the dorsal cutaneous branch. The superficial
terminal branch also innervates the palmaris brevis muscle.
ULNAR NEUROPATHY In the palm, the deep terminal motor branch innervates
consecutively the abductor, opponens and flexor of the little
finger, the third and fourth lumbricals, the interossei and,
DEFINITION finally, the adductor pollicis. Consequently, ulnar nerve
Dysfunction of the ulnar nerve. entrapment proximal to the wrist generally causes sensory
and motor signs, whereas entrapment distal to the wrist
EPIDEMIOLOGY causes motor deficits only.
Reasonably common.
ETIOLOGY
ANATOMY Axillary and upper arm lesions
Course of the ulnar nerve (773) • Rare.
Arises from the lower trunk and medial cord of the brachial • Compression:
plexus, carrying fibers from the C8 and T1 (and sometimes – Long crutches or inappropriately tied tourniquets: but
also from C7) nerve roots. usually also involves the median and radial nerves.
– Chondroepitrochlaris muscle, arising from pectoralis
Axilla major and crossing over the neurovascular bundle in the
Travels through the axilla in close proximity to the median axilla, before inserting into the brachial fascia and medial
nerve and axillary artery. epicondyle of the humerus.
– Aneurysm of the axillary or brachial artery.
Upper arm • Median sternotomy (?traction on brachial plexus).
Halfway down the upper arm, the nerve pierces the
intermuscular septum and descends on the dorsomedial side Elbow lesions
of the upper arm between the intermuscular septum and the Common: 87% of all ulnar neuropathies, and often bilateral.
medial head of the triceps muscle. It gives off no branches There are two potential sites of compression.
in the upper arm.
Ulnar Neuropathy 633

The shallow condylar groove (where the nerve lies directly – Rheumatoid synovial cyst.
beneath the skin) – Ruptured medial head of triceps muscle.
• External pressure: – Trauma.
– ‘Drivers’ elbow’: resting the elbow against the lower • Hypertrophic ulnar neuropathy.
edge of the car window. • Leprosy.
– Prolonged bed rest. • Nerve tumors.
– Prolonged leaning with the elbow on a desk (e.g. shoe
workers, telephone operators). The cubital tunnel (the aponeurosis of the flexor carpi
– Malpositioning during general anesthesia. ulnaris forms the roof and the medial ligament of the
– Hemodialysis shunt, with proliferating granulation tissue. elbow joint forms the floor)
– Steroid injection for medial epicondylitis, with Flexion of the elbow increases pressure in the cubital tunnel
undetected dislocation of the nerve. Repeated flexion/extension of the elbow (e.g. diamond
• Deformities or abnormalities at the elbow joint: sorters, or operating the handle of a mounted drill or a
– Aneurysm of the ulnar collateral artery. chain-saw).
– Arthrosis.
– Arthritis, chondromatosis. Forearm lesions
– Diffuse idiopathic skeletal hyperostosis (Forestier’s • Uncommon; 1% of all ulnar neuropathies.
disease). • Fibrous or fibrovascular bands at the distal part of flexor
– Epineural cyst. carpi ulnaris.
– Heterotopic calcifications. • Greenstick fracture of the ulna.
– Hypertrophic ulnar neuropathy. • Compartment syndrome of the forearm (e.g. forearm
– Ganglia. hemorrhage associated with bleeding diathesis).
– Gout. • Local hypertrophic neuropathy.

Wrist and palm lesions


773 Uncommon: 12% of all ulnar neuropathies.

External pressure
The most common cause:
• Cyclist’s palsy: pressure from handlebars.
• Occupational: handling screwdrivers, pliers, knives,
shears, pneumatic drills.
• Crutches and cane use.
• Handcuffs.

Ulnar nerve
Acute injury
• Penetrating wounds.
• Fractures of the radio-ulnar joint, hamulus of the hamate
bone, or fifth metacarpal bone.
• Carpo-metacarpal luxation.

Acquired lesions
• Benign giant cell tumor.
Flexor carpi • Benign chondroblastoma of the hamate.
ulnaris • Ganglion.
• Lipoma, with hemorrhage.
• Nodular synovitis.
Flexor digitorum • Pathologic calcifications, idiopathic or associated with
profundus III, IV scleroderma.
• Pyrophosphate arthropathy.
• Thrombo-angiitis of the ulnar artery.
Digiti minimi:
Adductor pollicis Abductor Congenital anomalies
Flexor pollicis brevis Opponens • Accessory muscles: palmaris brevis, palmaris longus, or
1st dorsal interosseous Flexor abductor digiti minimi.
1st palmar interosseous • Anomalous origin of flexor digiti minimi.
Fourth lumbrical • Anomalous course of ulnar nerve, beneath flexor retinaculum.
Third lumbrical • Duplication of the tendon of the flexor carpi ulnaris
muscles, with splitting of the ulnar nerve.
• Fusion between pisiform and hamate bones.
• Reversed palmarus longus muscle.
773 Diagram of the ulnar nerve and the muscles which it supplies. • Thickened piso-hamate ligament.
N.B.The white rectangle signifies that the muscle indicated receives a • Tortuosity of the ulnar artery.
part of its nerve supply from another peripheral nerve.
634 Mononeuropathies

Infections • If the patient has a radial neuropathy (in isolation or


• Leprosy. combination), it is not possible to test precisely finger
• Tuberculosis. abduction because the fingers need to be extended first. In
• Phlegmon of the palm. this situation, finger abduction should be tested with the
palm faced downwards on a hard surface, and the fingers
Hereditary liability to pressure palsies (see p.587) resting in an extended position but, even then, the
movements will not be quite normal even if the ulnar nerve
HISTORY is preserved.
Axillary and upper arm lesions • The flexor digitorum profundus III and IV are tested by
Same as elbow lesions unless evidence of concomitant median testing flexion of the distal interphalangeal joint of the little
or radial neuropathy. and ring finger against resistance, while the middle proximal
interphalangeal joint is fixed in extension.
Elbow and forearm lesions • The flexor carpi ulnaris often escapes compression at the
Sensory symptoms elbow but if not, there may only be a slight radial deviation
• Numbness, pins and needles, and occasionally burning pain of the hand on wrist flexion; wrist flexion remains generally
in the ring and little finger, and sometimes the hypothenar unaffected because of the action of the (median-innervated)
region (but not outside the sensory area of the ulnar nerve, flexor carpi radialis.
nor causing the patient to wake at night, in contrast to the • Severe ulnar neuropathy gives rise to the so-called ‘ulnar
carpal tunnel syndrome). claw hand’ with guttering of the dorsum from atrophy of
• Leaning on the elbow (e.g. against the lower edge of a car the interosseous muscles and third and fourth lumbricals,
window) or flexing the elbow (e.g. reading in bed) may hyperextension of the fourth and fifth metacarpophalangeal
precipitate or exacerbate the symptoms. joints, mild flexion of the interphalangeal joints and
• The elbow or the nerve in the ulnar groove may be painful abduction of the little finger (779, 780).
or tender to touch.
Wrist and palm lesions
Motor symptoms Deep and superficial branch: just proximal to or within
Weakness and clumsiness of fine finger movements (e.g. Guyon’s canal
zipping trousers, releasing a belt, buttoning and unbuttoning • Pure weakness, and possibly wasting, of all hand muscles
clothing, sewing, guitar playing) may be the presenting innervated by the ulnar nerve (adductor pollicis,
symptoms, depending on the fascicles involved, but usually interosseous muscles, hypothenar muscles [abduction of
follow the sensory symptoms. little finger], and palmaris brevis [no skin dimple on
extreme abduction of the little finger]).
EXAMINATION • Sparing of flexor carpi ulnaris and flexor digitorum
Axillary and upper arm lesions profundus (as for more proximal lesions at the elbow).
Same as elbow lesions unless evidence of concomitant median • Loss of sensation on palmar aspect of the little finger and
or radial neuropathy. ulnar half of the ring finger, corresponding to the
cutaneous innervation of the superficial terminal branch.
Elbow and forearm lesions • Normal sensation over the proximal part of the hypo-
Sensory signs thenar and the dorsal part of the ring and little fingers.
Altered sensation on the little finger (palmar and dorsal sides), • Little pain.
ring finger (ulnar half), and often adjoining parts of the hand,
but may be restricted to the tips of the little and ring fingers, Deep branch, proximal part: at the pisiform bone
or even absent (774). • Pure weakness, and possibly wasting, of all hand muscles
innervated by the ulnar nerve, excluding palmaris brevis
Autonomic signs (which contracts during strong abduction of the little
• Uncommon. finger and causes indentation of the skin overlying the
• Furrowed or atrophic nails of the little and ring fingers. hypothenar).
• Trophic skin lesions may also be present in the cutaneous • Normal sensation (the lesion is beyond the branching of
distribution of the ulnar nerve. the superficial terminal branch within Guyon’s canal).

Motor signs Deep branch, distal part: at (or distal to) the hamate bone
• Weakness (and wasting) of first dorsal interosseous and • Weakness and wasting of the radial hand muscles
hypothenar eminence (adduction of the thumb and little innervated by the ulnar nerve (adductor pollicis,
finger respectively) may be the only initial signs (775–778); interossei), with sparing of function of the hypothenar
the flexor carpi ulnaris and flexor digitorum profundus muscles, and palmaris brevis muscle.
muscles are rarely wasted (medial border of forearm) or • Normal sensation.
weak in the early stages.
• A useful test of thumb adduction is to ask the patient to Superficial branch: at or distal to Guyon’s canal
squeeze a sheet of paper between the base of the thumb • Weakness of palmaris brevis muscle (no skin dimple on
and the index finger; weakness of the adductor pollicis is extreme abduction of the little finger).
present if the interphalangeal joint of the thumb flexes, due • Loss of sensation of the little and ring finger (ulnar half)
to use of the median-innervated flexor pollicis longus on the palmar side.
(Froment’s sign).
Ulnar Neuropathy 635

774 Approximate area in which light touch and pain sensation are 774
reduced in an ulnar nerve lesion. N.B. Nerve root, plexus and cord
lesions do not ‘split’ the ring finger.

775 776

775, 776 Severe ulnar neuropathy causing wasting of the dorsal interossei (775) and wasting of the adductor pollicis and an ‘ulnar claw hand’ (776).

777 778

777, 778 Compressive mononeuropathy of the deep palmar branch of the ulnar nerve in a cyclist, causing wasting (and weakness) of the interossei.

779 780

779, 780 Severe ulnar neuropathy due to forearm injury causing the so-called ‘ulnar claw hand’ with flexion of the interphalangeal joints and
abduction of the little finger (777) and hyperextension of the fourth and fifth metacarpophalangeal joints (780).
636 Mononeuropathies

DIFFERENTIAL DIAGNOSIS EMG


• Central weakness (upper motor neuron lesions): • May identify ulnar nerve lesions that have led to axonal
diminished adduction of the little finger as an isolated degeneration even when nerve conduction velocity is
sign in the upper limb may also occur in patients with normal.
supranuclear lesions. • May distinguish lesions of the T1 cervical nerve root,
• Motor neuron disease (amyotrophic lateral sclerosis): brachial plexus or upper motor neuron.
brisk deep tendon reflexes and extensor plantar
responses. MRI scan of site of compression
• Spinal muscular atrophy: no sensory signs, but differs May identify compressive soft tissue masses (ganglia, cysts,
from a pure motor ulnar neuropathy in that weakness is tumors).
beyond the distribution of the ulnar nerve.
• Syringomyelia: may mimic an ulnar neuropathy with Cervical spine x-ray
muscle wasting and weakness and sensory loss in the Not required if clinical and neurophysiologic findings point
hands, but the sensory loss is dissociated (impaired pain to a lesion at the elbow. Otherwise, very common findings
and temperature with intact position and vibration sense) in the middle aged and elderly population, such as
and often outside the ulnar nerve territory (on the ‘foraminal narrowing’, tend to be over-interpreted.
forearms, shoulders and trunk), the upper limb reflexes
depressed or absent and sometimes there are long tract MRI scan of cervical spine
signs. May exclude suspected syringomyelia or intramedullary
• Spondylotic myelopathy: may cause weakness and cervical spine tumor, but cavitation of the spinal cord may
wasting of intrinsic hand muscles, but is usually also be present without causing symptoms.
associated with urinary incontinence and long tract signs
(e.g. corticospinal tracts and posterior columns). DIAGNOSIS
• T1 radiculopathy (e.g. cervical rib and band, Pancoast Requires the presence of isolated weakness of muscles
tumor): weakness and wasting of the small muscles of the exclusively in the distribution of the ulnar nerve (i.e. with
hand, but also involves the thenar muscles and a largely intact muscle contour of the thenar eminence)
Horner’s syndrome may be present. and/or loss of sensation of skin innervated by the ulnar
• Cervical radiculopathy (e.g. C8 nerve root compression nerve.
by a rare lateral disc prolapse between the vertebrae of
C7 and T1): the finger flexors rather than the intrinsic TREATMENT
hand muscles are weak. Prevention
• Lower brachial plexus lesion: weakness and sensory loss Adequate support of the arms in bedridden patients,
outside the distribution of the ulnar nerve. particularly paralysed arms.

INVESTIGATIONS Conservative treatment


Nerve conduction studies • Avoid elbow flexion of >30°:
Motor nerve conduction studies – Keep elbow extended as much as possible.
• Stimulation of the ulnar nerve at the wrist, and above – Avoid leaning on the elbows or crossing the arms while
and below the elbow (with the elbow flexed at 90° or sitting.
135°), while recording the compound muscle action – Hold the telephone receiver with the other hand.
potential (CMAP) from an ulnar-innervated muscle in – Wear a towel or thermoplastic splint at night.
the hand (e.g. abductor digiti minimi, first dorsal • Avoid direct nerve compression:
interosseous), may reveal slowing of conduction across – Place a pillow under the arm when sitting at a desk.
the elbow segment (of >10 or 11 m/s compared with – Use a book-stand if reading for a prolonged period.
the conduction velocity in the elbow) and/or a reduction
in amplitude of the CMAP with proximal stimulation, if Surgical treatment
there is a lesion of the ulnar nerve at the elbow. • If chronic compression excluded and if no improvement
• Conduction velocity in the deep motor branch can be in symptoms or if persistent disabling paresthesiae, pain,
compared with ulnar and median distal motor latencies or motor deficits.
to the second interosseous space. • If habitual subluxation or luxation of the ulnar nerve
from the ulnar groove.
Sensory conduction studies • If nerve compression by an arthrotic elbow joint.
• An ulnar neuropathy at the elbow may cause a reduced
or absent SNAP at the little finger, while the SNAP of PROGNOSIS
the dorsal cutaneous branch is spared. This combination Depends on the cause and severity of the neuropathy; partial
alone therefore cannot reliably localize the site of an lesions tend to recover spontaneously and over about 6–8
ulnar nerve lesion at the wrist. weeks. Severe ulnar neuropathy caused by compression at
• For a suspected lesion of the superficial branch, sensory the elbow may take 6–12 months after operation to recover,
conduction in the segment between the wrist and ring and may not recover at all. Neurophysiologic studies can
finger (superficial branch) can be compared with that in help determine the prognosis.
the median nerve fibers from the same finger.
Lateral Cutaneous Nerve of the Thigh Neuropathy 637

LATERAL CUTANEOUS NERVE OF THE Trauma


THIGH NEUROPATHY • Avulsion fracture of the anterior superior iliac spine.
• Anterolateral thigh injury.

DEFINITION Iatrogenic
Dysfunction of the lateral cutaneous nerve of the thigh. • Misguided intramuscular injections.
• Abdominoplasty.
EPIDEMIOLOGY • Transfemoral angiography.
Not uncommon. • Gastroplasty for morbid obesity.
• Groin flap (plastic surgery).
ANATOMY • Laparoscopic repair of inguinal hernia.
Course of the lateral cutaneous nerve of the thigh • Laparoscopic cholecystectomy.
• Arises from the L2 and L3 nerve roots (781). • Lithotomy position for gynecologic procedures.
• Descends from the lateral border of the psoas muscle, • Removal of bone graft from ileum.
across the iliacus muscle, and just medial to the anterior • Renal transplantation.
superior iliac spine it is enclosed between two folds of the • Rotational osteotomy of acetabulum for congenital
lateral attachment of the inguinal ligament. dysplasia of the hip.
• Leaves the pelvis in various ways:
– Through a notch between the anterior superior iliac Tumors
spine and the inferior area of the iliac spine. • Psoas muscle tumor.
– Through the inguinal ligament, sometimes with two or • Osteoid osteoma of the hip.
three separate branches.
– Medial to the inguinal ligament, near the femoral nerve. The most common causes, after idiopathic syndrome, are
• Enters the upper anterior thigh in a fibrous canal within pregnancy, tight clothing or belts, scars, iliac bone grafts and
the fascia lata, and abruptly changes course from roughly intrapelvic masses.
horizontal to vertical; the degree of angulation is
influenced by flexion and extension of the hip.
• A few centimeters distal to the inguinal ligament,
the nerve divides into several branches which 781
innervate the skin on the lateral part of the thigh, Iliohypogastric nerve
as far down as the knee, but not beyond the upper
Ilioinguinal nerve
ridge of the patella.

ETIOLOGY Psoas muscle


Idiopathic entrapment
Nerve to
iliacus
Familial
Autosomal dominant meralgia paresthetica. Genitofemoral nerve

External compression Femoral nerve


• Pocket watch.
• Pregnancy.
Pudendal nerve
• Seat-belt trauma. Nerve to levator
• Tight trousers. ani and external
sphincter
Superior and
inferior gluteal nerves
Exercise or postural factors Perineal nerve

• Pregnancy. Sciatic nerve Dorsal nerve of


penis or clitoris
• Body building.
Nerve to Inguinal canal
• Gymnastics. sartorius muscle Ilioinguinal nerve
• Falling asleep in the siddha yoga position. Genitofemoral nerve:
Genital branch
Femoral branch
Cutaneous
nerves of thigh: Obturator nerve
Lateral Branches to:
Intermediate Obturator externus
Medial Adductor longus
Adductor brevis
Nerves to quadriceps: Adductor magnus
Rectus femoris Gracilis
Vastus lateralis Cutaneous
Vastus intermedius
Vastus medialis Posterior cutaneous
nerve of thigh
Saphenous nerve Sciatic nerve
781 Diagram of the lumbosacral plexus. (Reproduced with Common peroneal
permission from Aids to examination of the peripheral nervous Tibial
system 1986. Baillière Tindall, London.)
638 Mononeuropathies

HISTORY TREATMENT
• Variable pain, tingling, burning, and numbness in the Depends on the cause.
skin of the lateral thigh, commonly underlying where
one’s hands would rest in one’s trouser pockets, and Conservative
rarely as far down as the knee (‘meralgia paresthetica’) • Remove constrictive items around the waist.
(782). • Cool with ice packs three times daily.
• Pressure on the presumed point of entrapment, medial • Non-steroidal anti-inflammatory drugs.
to the anterior superior iliac spine, and hyperextension • Injection of steroid and an analgesic agent (e.g.
of the hip; standing or walking may aggravate the lignocaine) at the suspected trigger point at the inguinal
unpleasant sensations, and hip flexion may alleviate them. ligament.
• Occasionally, the syndrome is bilateral. • Transcutaneous nerve stimulation.

EXAMINATION Surgical
• Altered sensation of the skin in the center of the territory • Often a last resort.
innervated by the nerve; sensation to touch may be • Usually involves removal of a 4 cm (1.6 in) segment of
reduced, unpleasant, or even painful. the nerve (hopefully containing a neurinoma), at its
• Local hypertrichosis or alopecia may result from frequent passage through the inguinal ligament. The resulting
rubbing of the skin or dysfunction of autonomic sensory deficit does not usually lead to anesthesia
innervation. dolorosa.
• Neurolysis is an alternative to nerve transection. Re-
DIFFERENTIAL DIAGNOSIS exploration after an initially unsuccessful neurolysis is
L2 radiculopathy: usually unsuccessful.
• L2 vertebral body metastases.
• Retroperitoneal tumor. PROGNOSIS
• Thoracic vertebral body collapse. About one-quarter of patients show spontaneous recovery
within months or years.
INVESTIGATIONS
Nerve conduction studies
• Limited because it is not possible to stimulate or record
at a short distance proximal to the suspected site of
compression.
• The site for stimulation or recording is not standardized
because of the variable course of the nerve. It is therefore
determined by eliciting maximal sensations in the lateral
aspect of the thigh.
• The more distal nerve segment is therefore studied.
Results should always be compared with those of the
unaffected side (e.g. bilateral absence of SNAPs may
occur in normal subjects). Any asymmetric slowing of
conduction implies loss of the thickest fibers, which is 782
often accompanied by reduced amplitude of the SNAPs.
• Somatosensory evoked potentials (SSEPs) may also be
used to diagnose lesions of the lateral cutaneous nerve of
the thigh. To distinguish a lesion of the nerve from a
radiculopathy or plexopathy, the dermatomal SSEP of
the ilioinguinal nerve can be used for comparison.

EMG
Needle examination should be normal and, if abnormal,
may distinguish lesions of the lumbar nerve roots, lumbar
plexus and femoral nerve.

Lumbar spine x-ray


If there are associated symptoms (e.g. back pain, weight
loss) or if the area of sensory disturbance extends beyond
that innervated by the lateral cutaneous nerve of the thigh.

Other
• MRI upper lumbar spine.
• Abdominal and pelvic ultrasound.

DIAGNOSIS
Requires the presence of isolated sensory loss confined to
the distribution of the lateral cutaneous nerve of the thigh. 782 Area of skin innervated by the lateral cutaneous nerve of the thigh.
Posterior Cutaneous Nerve of the Thigh Neuropathy 639

POSTERIOR CUTANEOUS NERVE Trauma


OF THE THIGH NEUROPATHY Wounds at the dorsal aspect of the thigh.

Iatrogenic
DEFINITION • Misguided intramuscular injections in the buttock.
Dysfunction of the posterior cutaneous nerve of the thigh. • Gluteal thigh flaps for reconstruction of the vagina
following resection of infiltrating carcinoma.
EPIDEMIOLOGY
Uncommon. Tumors
• Colorectal tumor.
ANATOMY • Hemangiopericytoma.
Course of the posterior cutaneous nerve of the thigh: • Venous malformation.
• Arises from the lower part of the lumbosacral plexus,
carrying fibers from the S1, S2, and S3 nerve roots. HISTORY
• Descends together with the inferior gluteal nerve Paresthesia and numbness in the skin over the lower part of
through the greater sciatic notch, below the piriform the buttock and the posterior aspect of the thigh.
muscle.
• Enters the thigh at the lower border of the gluteus EXAMINATION
maximus and close to the sciatic nerve, giving off Altered sensation of the skin over the lower part of the
branches to the skin of the perineum and scrotum (or buttock and the posterior aspect of the thigh may or may
labia majora). not be present (783).
• Descends superficially over the hamstring muscles to the
popliteal fossa, supplying fibers to the skin over the lower DIFFERENTIAL DIAGNOSIS
part of the buttock, the dorsal aspect of the thigh, and S2 radiculopathy.
the proximal third of the calf.
INVESTIGATIONS
ETIOLOGY EMG
External compression Needle examination may identify lesions of the S2 nerve
Prolonged sitting on the buttocks: roots, or sacral plexus.
• Sedentary occupation.
• Extensive cycling. DIAGNOSIS
• Prolonged gymnastic exercises performed on the Requires the presence of isolated sensory loss confined to
buttocks. the distribution of the posterior cutaneous nerve of the
thigh.

TREATMENT
Depends on the cause.
• Conservative: avoid pressure to the lower buttock and
dorsal thigh.
783 Area of skin 783 • Surgical: removal of the responsible mass lesion.
innervated by the
posterior cutaneous
nerve of the thigh.
640 Mononeuropathies

FEMORAL NEUROPATHY Penetrating injuries


Inflammation
• Inguinal lymphadenitis.
DEFINITION • Rheumatoid bursitis of the iliopsoas muscle.
Dysfunction of the femoral nerve. • Heterotopic calcification.

EPIDEMIOLOGY Irradiation
Uncommon. Vincristine toxicity

ANATOMY Saphenous nerve


Course of the femoral nerve Iatrogenic
• Arises from the L2, L3 and L4 nerve roots within the • Arthroscopy or menisectomy.
psoas muscle. • Saphenous vein graft.
• Descends along the lateral border of the psoas muscle, • Stripping of varicose veins, endoscopic dissection of
within the fascia between the psoas and the iliacus muscles. perforating veins, cryosurgery.
• Proximal to the inguinal ligament it gives off branches • Arterial reconstruction in the thigh.
to the iliopsoas muscle.
• Descends further, beneath the inguinal ligament, lateral Compression
to the femoral artery, and gives off branches to the • Entrapment:
sartorius and the four parts of the quadriceps muscle. – At the medial side of the knee.
• Distal to the inguinal ligament, sensory branches arise: – In the subsartorial canal.
the intermediate cutaneous nerve of the thigh, and the – By a branch of the femoral artery.
medial cutaneous nerve of the thigh, which innervate the • Neurilemmoma
skin on the anterior and medial aspects of the thigh. • Bursitis of the pes anserinus, distal to the adductor canal.
• The terminal branch of the femoral nerve, the purely
sensory saphenous nerve, passes within the quadriceps Irradiation
muscle through Hunter’s canal, and about 10 cm (4 in)
above the knee it leaves this canal and gives of the infra- HISTORY
patellar branch, which innervates the skin on the medial Femoral nerve
aspect of the knee down to the tuberosity of the tibia. • Sudden falls caused by buckling of the knee, particularly
• More distally, the saphenous nerve descends along the if walking on an uneven road surface, climbing up an
medial side of the tibia and the anterior surface of the incline, or descending a staircase (i.e. when the body
medial malleolus, to supply the skin at the anterior and weight has to be supported with some knee flexion).
medial surface of the knee, and the medial aspect of the • Deep, severe, nerve trunk pain, with or without numb-
lower leg, including the ankle and arch of the foot. ness and paresthesia on the anterior aspect of the thigh
or inner aspect of the lower leg, may be present,
ETIOLOGY depending on the cause. Pain is common in diabetic
Femoral nerve amyotrophy and psoas hematoma, which may mimic
Compression (in the iliac fossa most commonly, and in the femoral neuropathy, and is minimized by full flexion of
inguinal region) the hip. Acute pain in the flank of the abdomen is
• Spontaneous hematoma in the psoas or iliacus muscle: reported particularly by patients with hematomas in the
– Bleeding diathesis: anticoagulation, hemophilia (may be iliacus muscle that compress the femoral nerve.
bilateral).
– Ruptured aortic aneurysm. Saphenous nerve
– Trauma. Pain, numbness and paresthesia on the inner aspect of the
• Abdominal or pelvic surgery: knee (if the infrapatellar branch is involved) and lower leg.
– Hematoma after nerve block.
– Catheterization via the femoral artery causing retro- EXAMINATION
peritoneal hematoma or pseudoaneurysm in the groin. Femoral nerve
– Laparoscopy. • Weakness of iliopsoas and the quadriceps, with iliopsoas
– Stapling in laparoscopic hernia repair. tested in the supine position, and quadriceps tested at a
– Inadvertent suturing. mechanical disadvantage (because it is such a massive and
– Retractor blades (self-retaining) used in abdominal surgery. strong muscle) with the knee in flexion and the patient
– Vaginal delivery. sitting and trying to bring the lower leg forward against
– Vaginal hysterectomy in lithotomy position (extreme resistance. This method also avoids testing the knee in
abduction and exorotation of the thighs may stretch the extension where a locked joint would have to be wrenched
femoral nerve). open (which is painful and mild weakness easily missed).
– Abdominal hysterectomy. • The most reliable proof of normal strength of the
– Hip abscess after parturition. quadriceps is the ability to rise from the floor, whilst
– Hip arthroplasty. squatting on one knee, transferring the weight to the
– Renal transplantation, associated with pressure from other leg, flexed at the knee, and extending the other leg.
retractor blades, hematoma or ischemia from ‘stealing’. Of course, with advanced age this is determined more
• Synovial cyst of the hip. and more by non-neurologic factors such as joint
• Prolonged pressure on the abdomen after intoxication. stability, and the test becomes less reliable.
Femoral Neuropathy 641

• A decreased or absent knee jerk is consistent, but not specific. EMG


• Wasting of the anterior aspect of the thigh invariably Needle examination helps to distinguish femoral neuropathy
occurs after some time. from diabetic amyotrophy which is an axonal neuropathy
• Loss of sensation over the anterior and medial aspect of (rather than demyelination) with features of denervation in
the thigh and the medial aspect of the lower leg is the quadriceps as well as the hip adductors and paravertebral
present (784, 785). muscles.
• With progressive weakness, the knee is locked in a hyper-
extended position (genu recurvatum) during walking. CT scan of the pelvis
Reliably detects mass lesions in the pelvis that may cause
Saphenous nerve femoral neuropathy.
Reduced sensation on the inner aspect of the knee (if the
infrapatellar branch is involved) and lower leg. DIAGNOSIS
Requires the presence of isolated weakness of the iliacus,
DIFFERENTIAL DIAGNOSIS iliopsoas, sartorius and/or quadriceps femoris and sensory
• Lumbosacral plexopathy or lumbar radiculopathy (L2, loss over the anterior thigh and medial lower leg. Can be
L3, L4): it is crucial to carefully assess the strength of the confirmed by EMG.
adductor muscles of the hip because weakness of the hip
adductors is not consistent with a femoral neuropathy TREATMENT
and more suggestive of a lesion in the lumbosacral Depends on the cause.
plexus, lumbar nerve roots L2, L3, L4; or obturator
nerve. Conservative
• Diabetic lumbar radiculopathy/plexopathy: an axonal Painful paresthesia may be alleviated with carbamazepine or
plexopathy (rather than demyelination) with EMG phenytoin.
features of denervation in the quadriceps as well as the
hip adductors and paravertebral muscles, indicating a Surgical
proximal root lesion. • Only if direct penetrating trauma with severe axonal
injury or complete interruption of nerve continuity.
INVESTIGATIONS • Retroperitoneal hematomas are usually decompressed
Nerve conduction studies surgically or aspirated but this is to relieve pain; there is
The saphenous nerve can be examined using peripheral no evidence that it improves recovery.
nerve conduction studies and with SSEPs.
PROGNOSIS
Depends on the cause and severity of the neuropathy.
Femoral neuropathy after operation, from stretch or
compression tends to be followed by spontaneous recovery,
although this may take weeks or even months.

784 785

Area supplied by the


lateral cutaneous
nerve of the thigh
Area supplied by the
lateral cutaneous Area supplied by the
nerve of the thigh obturator nerve
Area supplied by the
obturator nerve
Area supplied by the
Posterior cutaneous femoral nerve
nerve of the thigh

Cutaneous branch of Cutaneous branch of


the peroneal nerve the peroneal nerve

Area supplied by the cutaneous


branches of the tibial nerve

784, 785 Area of skin innervated by the femoral nerve.


642 Mononeuropathies

OBTURATOR NEUROPATHY DIFFERENTIAL DIAGNOSIS


• Lumbosacral plexopathy or lumbar radiculopathy (L2–
L4).
DEFINITION • Femoral neuropathy: weakness of quadriceps muscle and
Dysfunction of the obturator nerve. depressed knee jerk.
• Osteitis or other disorders of the symphysis: pain in the
EPIDEMIOLOGY groin and medial part of the thigh similar to the
Rare, in isolation. neuralgic pain of an obturator neuropathy, but usually
local tenderness.
ANATOMY
Course of the obturator nerve (786)
• Arises from the union of the ventral branches of the L2,
L3 and L4 nerve roots within the belly of the psoas
muscle.
• Emerges on the medial border of the psoas muscle.
• Descends over the sacroiliac joint, along the wall of the
pelvis.
• Enters the obturator canal and gives off anterior branches
and posterior branches, before entering the thigh.
• The posterior branches innervate the adductor muscles
of the thigh (adductor brevis, adductor longus, and
adductor magnus) which stabilize the hip.
• The anterior branch ends as a sensory nerve innervating 786
an area of skin on the inner side of the thigh.
Iliacus
ETIOLOGY Femoral nerve
Psoas
• Obturator hernia. Obturator nerve
• Normal labor.
• Scar formation in the thigh.
• Pelvic masses (frequently also cause a femoral
neuropathy): Adductor brevis
– Psoas muscle hematoma.
– Retroperitoneal schwannoma. Quadriceps femoris:
Adductor longus
– Endometriosis Rectus femoris
• Iatrogenic: Vastus lateralis Gracilis
– Hip surgery, damaging the nerve by overstretching, Vastus intermedius
retractor blades, fixation screws, and cement.
Vastus medialis Adductor magnus
– Fixation of acetabular fracture.
– Urologic surgery with prolonged hip flexion.
– Intrapelvic surgery.
– Laparoscopic dissection of pelvic nodes.
– Gracilis flap operations. Common peroneal
nerve
HISTORY Superficial peroneal Deep peroneal nerve
• Weakness in the leg. nerve
Peroneus longus Tibialis anterior
• Pain, numbness and paresthesia on the inner aspect of
the knee (if the infrapatellar branch is involved) and Extensor digitorum
Peroneus brevis longus
lower leg. Extensor hallucis
longus
EXAMINATION
• Weakness of hip adduction.
• Depressed adductor muscle tendon reflex. Peroneus tertius
• Normal power of quadriceps muscle and normal knee
jerk.
• Reduced sensation on the inner aspect of the thigh Extensor digitorum
(787). brevis
• Broad based gait may be present.

786 Diagram of the obturator, femoral and common peroneal nerves


and the muscles that they supply.
Gluteal Neuropathy 643

INVESTIGATIONS GLUTEAL NEUROPATHY


Nerve conduction studies
No nerve conduction technique is described.
DEFINITION
EMG Dysfunction of the superior and/or inferior gluteal nerves.
Needle examination may indirectly confirm the diagnosis by
demonstrating denervation activity confined to the hip EPIDEMIOLOGY
adductors. Uncommon.

CT scan of the pelvis ANATOMY


May detect a mass lesion in the pelvis causing obturator Course of the superior gluteal nerve (788)
neuropathy. • Arises from the L4, L5 and S1 nerve roots.
• Descends over the piriformis muscle, through the
DIAGNOSIS suprapiriform foramen, and then between the gluteus
Requires the presence of isolated weakness of the hip medius and minimus muscles.
adductors, with or without sensory loss over the inner thigh. • Innervates the gluteus medius and minimus and the
Can be confirmed by EMG. tensor fasciae latae muscles.

TREATMENT Course of the inferior gluteal nerve (788)


Depends on the cause. • Arises from the L5, S1 and S2 nerve roots.
• Conservative: painful paresthesia may be alleviated with • Descends through the infrapiriform foramen, dorsolateral
carbamazepine or phenytoin. to the sciatic nerve.
• Surgical: retroperitoneal hematomas are usually • Innervates the gluteus maximus muscle.
decompressed surgically or aspirated but this is to relieve
pain; there is no evidence that it improves recovery. ETIOLOGY
Superior gluteal nerve
PROGNOSIS Trauma
Depends on the cause and severity of the neuropathy. • Fall on the buttocks (acute transient entrapment of the
superior gluteal nerve between the piriformis muscle and
the major sciatic incisure, or more permanent damage
due to secondary muscle fibrosis).
• Intramuscular injections in the buttocks:
– Incorrectly placed injections may damage the sciatic
nerve and superior gluteal nerve.
– Correctly placed injections, in the upper outer quadrant
of the buttock, may cause a partial lesion, with weakness
of only the tensor fasciae latae muscle.
• Hip surgery via a posterior approach (the so-called
‘Hardinge approach’).
787

788

L4
L5
Gluteus medius
S1 Gluteus minimis
S2
Superior Tensor fasciae latae
gluteal nerve
Inferior gluteal Gluteus maximus
nerve

787 Area of skin innervated by the obturator nerve. 788 Diagram of the gluteal nerves and the muscles that they supply.
644 Mononeuropathies

Compression SCIATIC NEUROPATHY


Coma or anesthesia injections: may damage the superior
gluteal nerve and the sciatic nerve.
DEFINITION
Inferior gluteal nerve Dysfunction of the sciatic nerve.
Compression
Colorectal tumor (often there is concurrent involvement of EPIDEMIOLOGY
the posterior cutaneous nerve of the thigh, with altered Not uncommon.
sensation of the inferior lateral buttock).
ANATOMY
Bilateral gluteal neuropathy Course of the sciatic nerve (789)
• Prolonged labor. • Arises from the L4, L5, S1, S2 and S3 nerve roots.
• Spondylolisthesis of L4 on L5 (perhaps by entrapment • In the pelvis (or gluteal region below) the fibers that will
within the piriform muscle). eventually form the tibial and common peroneal nerves
are arranged in two separate divisions: the medial and
CLINICAL FEATURES lateral trunk respectively. The peroneal nerve fibers
Superior gluteal nerve within the lateral division of the sciatic nerve are more
• Pain in the buttock. prone to compression than the tibial nerve fibers in the
• Difficulty walking (weakness of gluteus medius and medial division, so that a partial lesion of the sciatic nerve
minimus leads to weakness of hip abduction, which at this level may be indistinguishable from that of a
causes defective tilting of the pelvis and difficulty peroneal nerve palsy in the leg, including preservation of
swinging the contralateral leg forward). the ankle jerk.
• Leaves the pelvis through the sciatic foramen.
Inferior gluteal nerve • Courses below the piriformis muscle, although the
• Pain in the buttock. division that is destined to become the peroneal nerve
• Difficulty walking, and particularly descending stairs and (and sometimes the entire nerve) may pierce the
arising from a chair, due to weakness of hip extension piriformis muscle, or pass over it.
(gluteus maximus). • In the gluteal region, the nerves passes laterally and then
vertically downwards. Beneath the gluteus maximus it
DIFFERENTIAL DIAGNOSIS lies between the greater trochanter and the ischial
Proximal myopathy (all symmetric and slowly tuberosity, just posterior to the hip joint.
progressive) • At the inferior part of the buttock, the nerve is situated
• Dystrophinopathy. superficially in the subgluteal space amongst collagen and
• Limb girdle dystrophy. fat tissue. This is a site where the nerve may be damaged
• Polymyositis. by an inflammatory process or wrongly applied injection
• Acid maltase deficiency. fluids that can easily spread.
• The nerve descends dorsal to the femur, between the
Hip joint disorder knee flexor muscles.
• The medial division consecutively supplies the semi-
INVESTIGATIONS tendinosus, biceps femoris (long head) and semimem-
EMG branosus muscles, and contributes to the innervation of
Needle examination of the gluteii identifies denervation the adductor magnus muscle.
activity. • The lateral division innervates the short head of the
biceps femoris.
MRI scan pelvis • The sciatic nerve terminates at the proximal part of the
popliteal fossa, where it divides into the tibial and
DIAGNOSIS common peroneal nerve.
Requires the presence of isolated weakness of the gluteal • The main sciatic trunk does not have sensory branches
muscles. Can be confirmed by EMG. but the nerve is accompanied by the posterior cutaneous
nerve of the thigh (S1–S3) which innervates the skin of
TREATMENT the dorsal aspect of the thigh and the proximal part of
Depends on the cause. the calf.
• Conservative.
• Surgical: if more or less total nerve damage and no ETIOLOGY
evidence of recovery. Pelvis
Tumors
PROGNOSIS • Metastatic carcinoma.
Depends on the cause and severity of the neuropathy. • Neurofibroma, schwannoma.
• Lipoma.
Sciatic Neuropathy 645

Vascular abnormalities Gluteal region


• Arteriovenous malformation. Compression
• False aneurysm of the abdominal aorta. • Prolonged sitting:
• Unruptured aneurysm of the common iliac artery or – Alcoholic- or drug-induced stupor on a toilet, or hard
internal iliac artery. rail.
• Ruptured aneurysm of the hypogastric artery. – Anesthesia in a sitting position.
– Meditation/sleeping in a cross legged position.
Endometriosis • Coma (usually drug induced with rhabdomyolysis).
• ‘Backpocket sciatica’ from credit cards or coins.
Childbirth
Cesarian section with epidural anesthesia. Vascular lesions
• Hematoma (bleeding diathesis).
Infection • Persistent sciatic artery.
Clostridium septicum, with immuno-incompetence. • False aneurysm, loop or abnormal collateral of the
inferior gluteal artery or superior gluteal artery.

Tumors
• Neurofibroma, schwannoma.
• Lipoma.
• Lymphoma.
• Hemangiopericytoma.
789
Gluteus medius Gluteus miniums Iatrogenic
Superior gluteal nerve • Hip surgery (total hip arthroplasty):
Piriformis Tensor fasciae latae – Ipsilateral: trochanteric wiring or extruding cement.
– Contralateral: rhabdomyolysis.
Inferior gluteal nerve
• Intramuscular injections that are not placed in the upper
and outer quadrant of the buttock or, even if
Gluteus maximus appropriately placed, cause muscle necrosis or fibrosis.
Sciatic nerve • Scoliosis surgery: Harrington’s operation.
• Femoral fracture surgery: closed nailing.
Semitendinosus • Heart surgery: mechanical compression or femoral artery
Biceps, long head occlusion.
• Femoral artery catheterization.
Semimembranosus Biceps, short head
Trauma
Adductor magnus • Hip fracture–dislocation.
• Femur fracture.
Tibial nerve Common peroneal
nerve
‘Pyriformis syndrome’
An anatomic variation in which the piriformis muscle
Gastrocnemius, compresses the sciatic nerve as it emerges from the pelvis
medial head through the greater sciatic notch. There must be objective
Gastrocnemius,
lateral head neurologic and neurophysiologic signs of sciatic nerve
Soleus
dysfunction (not just pain in the buttock); reproduction of
pain with deep palpation via the gluteal or rectal route;
Tibialis posterior negative EMG findings in the paraspinal muscles;
Flexor digitorum
Flexor hallucis longus
appropriate radiologic studies excluding lumbosacral nerve
longus root compression, and masses in the paravertebral area,
lower pelvis and sciatic notch; negative CSF examination
Tibial nerve (i.e. no signs of inflammation that could reflect nerve root
inflammation or infection); and ultimately confirmation by
operation and subsequent relief with nerve decompression.
Medial plantar nerve Lateral plantar nerve
to: to: Thigh
Abductor hallucis Abductor digiti minimi Tumors
Flexor digitorum brevis Flexor digiti minimi
Flexor hallucis brevis • Neurofibroma, schwannoma.
Adductor hallucis • Lipoma.
Interossei
Vascular lesions
Aneurysm of persistent sciatic artery or popliteal artery.

789 Diagram of the gluteal, sciatic, tibial and common peroneal nerves
and the muscles that they supply.
646 Mononeuropathies

CLINICAL FEATURES INVESTIGATIONS


• Wasting and weakness of the muscles innervated by the Nerve conduction studies
sciatic nerve (see above): knee flexion (‘hamstring • Motor nerve conduction studies are difficult to
muscles’– semitendinosus, semimembranosus, and biceps undertake, unless needle electrodes are used for
femoris), and all movements of the foot and toes. stimulation, because the nerve lies so deeply.
• Decreased or absent biceps femoris jerk. • Abnormal late responses (soleus H-reflex, foot muscle F-
• Decreased or absent ankle jerk. N.B. The peroneal nerve waves) are non-specific and may reflect a lesion of the
fibers within the lateral division of the sciatic nerve are lumbosacral nerve roots or plexus or sciatic nerve, but
more prone to compression than the tibial nerve fibers diminished sensory nerve action potentials of the sural
in the medial division, so that a partial lesion of the sciatic and/or superficial peroneal nerve are unlikely with
nerve may be indistinguishable from those of a peroneal radiculopathy.
nerve palsy, including preservation of the ankle jerk.
However, if weakness of muscles innervated by the EMG
peroneal nerve is accompanied by a depressed ankle • Needle EMG can be extremely helpful in distinguishing
reflex, a lesion of the sciatic nerve or spinal roots is likely. sciatic nerve lesions from other lesions.
Isolated lesions of the medial division of the sciatic nerve • Denervation activity outside the territory of supply of the
are far less common, usually non-compressive (e.g. sciatic nerve indicates a lumbosacral radiculopathy or
neurilemmoma), and manifest clinical features similar to plexopathy.
a tarsal tunnel syndrome (see tibial nerve, p.647). • Denervation in the paraspinal muscles at the lumbosacral
• Diminished sensation of the posterior thigh and calf if level is typical of a lumbosacral radiculopathy, but even
there is associated damage to the posterior cutaneous in the absence of paraspinal muscle denervation EMG
nerve of the thigh, otherwise there may be sensory loss can still identify denervation in the distribution typical of
in the territory of supply of the common peroneal and a root lesion.
tibial nerve branches of the sciatic nerve (790). • Sciatic nerve lesions are more commonly characterized
by EMG signs of axonal loss than demyelination.
DIFFERENTIAL DIAGNOSIS • Denervation in the short head of biceps femoris
• L5 or S1 radiculopathy (deficits of motor, reflex and distinguishes a lesion of the lateral division of the sciatic
sensory function in the typical distribution of the nerve from the common peroneal nerve.
myotome (tibialis anterior and posterior [L5] and • EMG abnormalities may be very limited in particular
gastrocnemius/soleus and ankle jerk [S1]) and cases, such as sciatic nerve tumors which may damage
dermatome (the dorsum of the foot and big toes [L5] only a single, or very few fascicles. This can make it
and the lateral border of the foot [S1]): difficult to localize the lesion accurately.
– Disc herniation.
– Radiculitis due to infection (e.g. borrelia). MRI of the lumbosacral spine, pelvis
• Lumbosacral plexopathy. or gluteal region
Reliably detects compressive lesions such as an intervertebral
disc or paraspinal mass.

DIAGNOSIS
Requires the presence of isolated weakness of the hip flexors
and muscles below the knee, and sensory loss over the
lateral lower leg and foot. Can be confirmed by EMG.
790
TREATMENT
Depends on the cause.
• Conservative: painful paresthesia may be alleviated with
carbamazepine or phenytoin. A systematic review of 19
randomized controlled trials of conservative treatments
for sciatica indicates that neither traction, non-steroidal
antinflammatory drug therapy (piroxicam, indomethacin,
phenylbutazone) nor exercise therapy was unequivocally
effective. Epidural steroids may be effective for sub-
groups of nerve root compression. More trials are
needed.
• Surgical: fasciotomy may be indicated if local pressure has
caused rhabdomyolysis of the gluteal compartment.

PROGNOSIS
Depends on the cause and severity of the neuropathy.

790 Area of skin innervated by the sciatic nerve (i.e. common


peroneal nerve and tibial nerve).
Tibial Neuropathy 647

TIBIAL NEUROPATHY Synovial cyst


• Knee joint.
• Superior tibio-fibular joint.
DEFINITION
Dysfunction of the tibial nerve. Ankle strain (stretch injury)

EPIDEMIOLOGY Tumors of the nerve sheath


Not uncommon.
Tibial neuropathy at the ankle (in the tarsal tunnel)
ANATOMY Entrapment
Course of the tibial nerve • Idiopathic.
Proximal to the ankle • Poorly fitting footwear.
• Arises from a separate trunk within the ventral part of
sciatic nerve at a variable level above the knee. Trauma to the foot
• Carries fibers from the L4, L5, S1, S2 and S3 nerve roots. • Fractures or soft tissue injuries.
• In the popliteal fossa and calf, the nerve lies deep and • Strained flexor digitorum accessorius muscle.
well protected. • Post-traumatic fibrosis.
• In the popliteal fossa, the tibial nerve gives off the medial
cutaneous nerve (and the peroneal nerve gives off the Hypermobile ankle joint
lateral sural cutaneous nerve). In the calf, these two
branches unite to form the sural nerve. Hypertrophy of the abductor hallucis muscle
• In the calf, the tibial nerve innervates the two heads of
gastrocnemius, soleus, tibialis posterior, flexor digitorum Inflammation
longus and flexor hallucis longus muscles. • Rheumatoid arthritis.
• Leprosy (suspect residents of areas where leprosy in
At the ankle endemic, particularly when nerves are palpable).
• The tibial nerve descends behind and below the medial • Tenosynovitis.
malleolus through the tarsal tunnel which is superficial
and covered by the laciniate ligament, or flexor Metabolic disorders
retinaculum, that extends between the malleolus and the • Hypothyroidism.
medial aspect of calcaneus. The tarsal tunnel also contains • Hyperlipidemia (deposition of fat).
the posterior tibial artery and the tendons of tibialis
posterior, flexor digitorum longus and flexor hallucis Tumors
longus. • Ganglion arising from flexor hallucis longus tendon
• Within or immediately beyond the tarsal tunnel, the tibial sheath.
nerve branches into the medial and lateral plantar nerves • Intraneural ganglion.
and the calcaneal nerve. • Lipoma.
• The calcaneal nerve is purely sensory and carries • Nerve sheath cyst.
sensation from the medial side of the heel. • Neurilemmoma.
• The medial plantar nerve innervates the abductor hallucis
and short flexor digitorum muscles. Varicose veins
• The lateral plantar nerve supplies the flexor and abductor
digiti minimi, abductor hallucis and the interossei. Calcaneal neuropathy (sensory branch of the tibial
• The plantar nerves also carry sensory fibers from the sole nerve at the heel)
of the foot, and from their six terminal branches to the Compression below the deep fascia of the abductor hallucis
toes: the medial plantar proper digital nerve (from the muscle (often in athletes)
great toe), the lateral plantar proper digital nerve (from
the little toe) and four interdigital nerves (from the Medial plantar neuropathy
middle three toes). Entrapment
• The interdigital nerves course between the distal ends of the • Idiopathic:
metatarsal bones and each divide into two digital nerves. – Immediately distal to the tarsal tunnel.
– At the entrance of the fibromuscular tunnel behind the
ETIOLOGY navicular tuberosity.
Tibial neuropathy proximal to the ankle • Tight shoes.
Hematoma in the popliteal fossa • Prolonged standing on the rungs of a ladder with soft
• Blunt injury. shoes.
• Rupture of the popliteus muscle. • Immobilization in a bed with prolonged pressured
against the footboard (usually in hospital).
Entrapment • Local callus, fasciitis or arthritis.
• Idiopathic.
• Beneath a transverse fibrous band between the Physical exercise (‘jogger’s foot’)
gastrocnemius muscles.
• Beneath a tendinous arch at the origin of the soleus Inflammation
muscle. Leprosy.
648 Mononeuropathies

Tumors CLINICAL FEATURES


• Synovial cyst. Tibial neuropathy proximal to the ankle
• Tibialis posterior tendon cyst. • Wasting and weakness of plantar flexion (gastrocnemius,
• Malignant schwannoma. soleus) and inversion (tibialis posterior), and toe flexion
• Plantar fibromatosis. (flexor digitorum longus):
– The calf muscles (gastrocnemius and soleus) are often
Lateral plantar neuropathy tested together (plantar flexion) but because they are so
Trauma to the foot strong, it is difficult to detect even marked weakness if
Fractures or soft tissue injuries. plantar flexion of the foot is tested against the examiner’s
hand with the patient supine. If the patient can walk, it is
Compression of its first branch between the intrinsic muscles preferable to examine the patient walking on their toes (i.e.
of the sole without the heel touching the floor) and then standing on
one leg, supporting the body weight on the toes, and then
Entrapment hopping on the forefoot, without the heel touching the
• Tight shoes. floor. Of course, some people, such as the elderly, may not
• Prolonged standing on the rungs of a ladder with soft be able to perform this maneuver due to other
shoes. impairments, despite normal power of the calf muscles.
• Immobilization in a bed with prolonged pressured – The soleus muscle can be tested reasonably selectively by
against the footboard (usually in hospital). examining plantar flexion against resistance with hip and
• Local callus, fasciitis or arthritis. knee flexed and the patient supine.
• Decreased or absent ankle jerk.
Medial plantar proper digital neuropathy • Diminished sensation of the heel (calcaneal nerve), sole
• Ill-fitting shoes. of the foot (medial and lateral plantar nerves) and dorsal
• Scar tissue after bunion surgery. aspect of the toes (digital nerves) (792).
• Arthritis of the first metatarsophalangeal joint.
Tibial neuropathy at the ankle (in the tarsal tunnel)
Lateral plantar proper digital neuropathy (Posterior) tarsal tunnel syndrome
Traumatic neuroma secondary to bunion. • Unilateral usually, but may be bilateral.
• Pain (often burning) in the sole of the foot.
Plantar interdigital neuropathy (Morton’s • Paresthesia in the toes.
metatarsalgia) • Symptoms may only be present at night or while
Compression of one of the interdigital nerves of the foot, standing or exercising, such as walking, jogging, or
which arise from the medial and lateral plantar nerves at the playing tennis.
point where they course between the metatarsal heads, just • Wasting and weakness of the intrinsic muscles at the sole
before they divide into two digital nerves (791). The most of the foot (abductor hallucis and short flexor digitorum
common site of compression is between the third and muscles, and flexor and abductor digiti minimi, abductor
fourth metatarsal bones, particularly in women. The heads hallucis and the interossei) is usually asymptomatic and the
of the metatarsal bones and the deep transverse ligament muscles are difficult to see or test separately. Indeed, many
contribute to the compression. The swelling around the normal individuals are unable to fan their toes. However,
nerve consists mainly of fibrous tissue; the term ‘neuroma’ there may be a difference compared with the normal,
is a pathologic misnomer. particularly on palpating the muscle bulk of both feet.
• Sensory loss in the sole of the foot and toes in the
distribution of the medial (most commonly) or lateral
791 plantar nerve or both.
• Hoffmann–Tinel sign: local pain, paresthesia and an
electric sensation radiating across the sole of the foot
Tibial nerve
towards the toes, particularly the medial toes, elicited by
pressure or percussion over the tarsal tunnel, beneath the
medial malleolus, with the foot everted.
• An underlying cause may be apparent such as abnormal
postures of the foot, hypermobility of the ankle joint,
local masses and a perforating ulcer of the foot (cf.
leprosy).
Lateral plantar nerve
Medial plantar nerve
Calcaneal neuropathy (sensory branch of the tibial
nerve at the heel)
Pain in one or both heels.
Inter digital nerves Compression site Plantar neuropathy
• Isolated paresthesia and sensory loss, usually in the
Digital nerves
medial part of the sole (the medial plantar nerve is more
commonly affected).
• Weakness of the intrinsic foot muscles is usually
791 Compression site in Morton’s metatarsalgia. asymptomatic (see above).
Tibial Neuropathy 649

Medial plantar proper digital neuropathy Tibial neuropathy at the ankle (tarsal
• Pain, paresthesia and sensory loss on the medial side of tunnel syndrome)
the great toe. • Musculoskeletal pain (no paresthesia, and normal
• Tenderness and thickening of the nerve. conduction in the tibial nerve):
– Plantar fasciitis.
Lateral plantar proper digital neuropathy – Stress fractures.
Pain, paresthesia and sensory loss on the lateral side of the – Bursitis.
little toe. • Lumbar spinal stenosis (if pain and paresthesia in the foot
and toes during exercise).
Plantar interdigital neuropathy (Morton’s • S1 radiculopathy: may cause paresthesia in the sole of the
metatarsalgia) foot, but usually other signs, such as absent ankle jerk
• Severe burning pain in the sole of the foot, between the and weakness of the calf muscles.
heads of the relevant metatarsal bones (usually the third • Morton’s neuralgia: shooting pain evoked by pressure
and fourth), and radiating to the relevant toes (usually over one of the metatarsal bones (i.e. painful trigger
the third and fourth), with associated local paresthesia points over the sole of the foot).
and numbness. The pain may also radiate proximally. • Medial plantar neuropathy: pain is precipitated by
• The symptoms are initially precipitated or exacerbated by pressure distal to the medial malleolus (as opposed to
weight bearing on the feet (standing or walking), immediately below it) and the clinical symptoms and
external pressure between the heads of the metatarsal signs do not involve the lateral sole.
bones, or passive dorsiflexion of the toes, but later • Distal polyneuropathy, especially diabetic neuropathy,
become continuous, unless footwear is removed. causing burning feet may be quite similar to bilateral
• A sensory deficit may be present on the adjoining sides tarsal tunnel syndrome, but there are usually additional
of the toes. signs such as depressed ankle jerks and more extensive
sensory loss on both the ventral and dorsal aspects of the
DIFFERENTIAL DIAGNOSIS feet.
Tibial neuropathy proximal to the ankle • Sciatic nerve tumor may even initially mimic a tarsal
• S1 radiculopathy (deficits of motor, reflex and sensory tunnel syndrome.
function in the typical distribution of the myotome
(gastrocnemius/soleus and ankle jerk [S1]) and Plantar neuropathy
dermatome (the lateral border of the foot [S1]): S1 radiculopathy: may cause paresthesia in the sole of the
– Disc herniation. foot, but usually other signs, such as absent ankle jerk and
– Radiculitis due to infection (e.g. borrelia). weakness of the calf muscles.
• Lumbosacral plexopathy.
Plantar interdigital neuropathy (Morton’s
metatarsalgia)
• Tarsal tunnel syndrome.
• Interdigital neuroma.
• Stress fracture or avascular necrosis of a metatarsal bone.
• Soft tissue injury to the foot.

INVESTIGATIONS
Nerve conduction studies
• A tibial neuropathy proximal to the ankle may be
detected by stimulating the tibial nerve in the popliteal
792 fossa and the ankle, and measuring the conduction
velocity between these points as well as the amplitude of
the motor evoked potential.
• Compression of the tibial nerve at the ankle (in the tarsal
tunnel) may be suspected if there is delayed motor
conduction (distal latency) in the medial and lateral
plantar nerves as assessed by stimulating at the ankle and
recording from the abductor hallucis and abductor digiti
minimi respectively. However, in order to distinguish
between a lesion in the tarsal tunnel and a more distal
lesion, both of which may affect one or more branches
of the tibial nerve, it is necessary to stimulate the tibial
nerve proximal to the tunnel and the plantar nerves distal
to the tunnel. Nevertheless, motor nerve conduction
velocity is not infrequently normal in tarsal tunnel
syndrome and the diagnosis may depend on sensory
conduction studies which are more sensitive.

792 Area of skin innervated by the tibial nerve.


650 Mononeuropathies

EMG PERONEAL NEUROPATHY


Needle EMG may be helpful if signs of denervation are
present in the intrinsic foot muscles, particularly abnormal
motor unit potentials, because fibrillation and fasciculation DEFINITION
potentials may occur in healthy people, particularly in the Dysfunction of the peroneal nerve.
abductor hallicus (and also the gastrocnemius).
EPIDEMIOLOGY
Ultrasound, x-ray or MRI of the popliteal fossa, One of the most common mononeuropathies.
lower leg, tarsal tunnel or foot
Frequently detects any mass lesion compressing the tibial ANATOMY
nerve (e.g. hematoma) or affecting the nerve (e.g. tumor). Course of the peroneal nerve
Proximal to the ankle
DIAGNOSIS • The common peroneal nerve arises from a separate
Requires the presence of isolated weakness of plantar division within the sciatic nerve at a variable level above
flexion, ankle inversion and toe flexion, a depressed or the knee, sometimes as far proximal as the upper thigh.
absent ankle jerk, and sensory loss over the sole of the foot. • It carries fibers from the L4, L5, and S1 nerve roots.
Can be confirmed by EMG. • In the popliteal fossa, the lateral cutaneous nerve of the
calf arises from the common peroneal nerve to innervate
TREATMENT the skin of the upper third of the lateral lower leg (793).
Depends on the cause. • Below the knee the common peroneal nerve winds
around the head of the fibula, at which point it rests
Tibial neuropathy proximal to the ankle directly on the periosteum and is particularly prone to
Treat the cause. compression, stretch or other trauma.
• It then courses through the fibular tunnel (a fibro-
Tibial neuropathy at the ankle (tarsal tunnel osseous canal between the insertion of the peroneus
syndrome) longus muscle and the fibula) and enters the
• Change footwear if it is poorly fitting. compartment of the peroneal muscles, where it divides
• Antilepromatous drugs, if appropriate. into a deep and a superficial branch.
• Surgical decompression of the retinaculum: generally • The deep peroneal nerve innervates the tibialis anterior,
successful, as in the carpal tunnel syndrome. extensor digitorum longus and extensor hallucis longus
muscles.
Calcaneal neuropathy (sensory branch of the tibial • The deep peroneal nerve continues to descend as the
nerve at the heel) terminal branch of the deep peroneal nerve and lies
Surgical decompression. superficially at the dorsum of the ankle and foot where
it innervates the extensor digitorum brevis muscle and a
Medial plantar proper digital neuropathy small area of skin in the web between the first and second
• Change footwear or insert protective padding. toes (794).
• Surgical excision of the nerve with concurrent • The superficial peroneal nerve gives off motor branches
bunionectomy, if present, often provides dramatic relief to the peroneus longus and brevis muscles about 10 cm
of persistent symptoms. (4 in) proximal to the lateral malleolus and, in about
one-quarter of individuals, gives rise to a terminal motor
Plantar interdigital neuropathy (Morton’s branch (the accessory deep peroneal nerve).
metatarsalgia) • The sensory branch of the superficial peroneal nerve
• Review footwear and ensure appropriate fit without tight pierces the deep fascia of the lower leg and supplies the
pressure on the sole. Supplement with padding if skin of the lower half of lateral lower leg and dorsum of
necessary. the foot.
• Injection of local anesthetic together with corticosteroid • A sural communicating branch joins the medial
affords recovery among one-third, and is associated with cutaneous nerve of the calf, originating from the tibial
later recovery after 2 years in another third. nerve, to form the sural nerve, which supplies the skin
• Carbamazepine taken orally may be helpful. on the lateral side of the heel, the sole and the little toe
• Surgical excision of the abnormal tissue is radical but (see pp.647, 654).
frequently successful. A dorsal approach may facilitate
mobilization and prevent infection. A new ‘neuroma’ ETIOLOGY (CAUSES IN ISOLATION
may form later. OR COMBINATION)
• Neurolysis, by incision of the intermetacarpal ligament Common peroneal neuropathy (or sometimes
above resection, is an alternative. only deep or superficial branch) at the level of
the fibular head
PROGNOSIS
Depends on the cause and severity of the neuropathy. Postural change
• Prolonged squatting or kneeling (e.g. gardening,
strawberry picking, farming, delivering a baby).
• Sitting with legs crossed habitually.
• Bed rest, particularly with a bed rail.
Peroneal Neuropathy 651

Running Compression of terminal branches of the


superficial peroneal nerve
Weight loss • Entrapment during sleep or sitting.
• Starvation (e.g. voluntary, prisoners of war). • Epidermoid cysts.
• Gastrointestinal or malignant disease. • Fascial bands.
• Anorexia nervosa. • Cannulation of foot veins.
• Arthroscopic knee surgery.
Iatrogenic • Intermittent pneumatic compression as prophylaxis for
• Plaster casts. deep vein thrombosis.
• Tight bandages.
• Arthroscopy of the knee. Lateral cutaneous nerve of calf lesions
• Arthrodesis (intramedullary) of the knee. Isolated compression.
• High tibial osteotomy.
• Malpositioning during anesthesia.

Congenital abnormalities
• Constriction band.
• Hereditary liability to pressure palsies.

Trauma
• Fracture of the femur or fibula. 793
• Inversion trauma of the foot.

Vascular
Hematoma in the popliteal fossa.

Tumors
• Exostosis of the fibular head or intraosseous cyst.
• Osteochondroma.
• Cysts of the lateral meniscus or tibio-fibular joint.
• Neurofibroma.
• Intraneural cysts.
• Hemangioma.
• Lipomatosis of the popliteal fossa due to steroids.

Pretibial myxoedema

Herniation of the gastrocnemius muscle

Peroneal neuropathy between the fibular


head and the ankle
Anterior compartment syndrome (deep peroneal nerve
compression) 793 Area of skin innervated by the common peroneal nerve.
• Swelling of necrotic muscle.
• Aneurysm of the tibial artery.
794
Lateral compartment syndrome (superficial
peroneal nerve compression)
• Compression as the superficial peroneal nerve as it
courses through the deep fascia of the lower leg after
giving off motor branches to the peroneal muscles.
Causes include athletic exercise (most commonly) and
callus formation after midshaft fracture of the fibula.
• Swelling of necrotic muscle.
• Idiopathic (may be bilateral).

Peroneal neuropathy at the ankle


Compression of the terminal branch of the deep peroneal
nerve at the anterior aspect of the ankle (‘anterior tarsal
tunnel syndrome’)
• Tight footwear (shoelaces, ski boots).
• Local contusion.
• Ganglion.
• Talotibial exostoses. 794 Area of skin innervated by the deep peroneal nerve.
652 Mononeuropathies

CLINICAL FEATURES Lateral compartment syndrome (superficial


Common peroneal neuropathy at the level peroneal nerve compression)
of the fibular head • Weakness of eversion of the ankle (short and long
• The extent of the deficits depends on whether the lesion peroneal muscles), if possible to test and if the cause
involves the fascicles of the deep or the superficial involves the superficial nerve before it gives off motor
peroneal nerve or both, as well as the duration and branches to the peroneal muscles about 10 cm (4 in)
severity of the compression. Isolated deficits in the proximal to the lateral malleolus. Otherwise the only
distribution of the deep or superficial peroneal nerve are neurologic deficits may be pain and sensory loss on the
most commonly due to a lesion of a separate fascicle at dorsum of the foot.
the level of the head of the fibula than a more distal • Sensory loss over the lower half of the lateral calf and
lesion involving a single one of the two branches. dorsum of the foot.
• Foot drop due to weakness of dorsiflexion of the ankle
(tibialis anterior; deep peroneal nerve), leading to a Peroneal neuropathy at the ankle
typical stepping gait, in which there is compensatory Compression of the terminal branch of the deep peroneal
overaction of the hip and knee flexors to clear the nerve (‘anterior tarsal tunnel syndrome’)
sagging foot during the swing phase. With mild • Painful paresthesia in the web space between the first and
weakness, the gait may be normal but the foot swings second toes.
with less clearance from the floor than the normal side • Wasting of the extensor digitorum brevis on inspection.
and the patient may not be able to walk on the heel of
the affected side. With moderate weakness of tibialis Compression of terminal branches of the
anterior, the foot may still land on the heel, but superficial peroneal nerve
immediately afterwards the ankle dorsiflexors give way Pain and numbness in part of the dorsum of the foot.
giving rise to a characteristic, flat-footed sound.
• Weakness of ankle eversion (peroneal muscles; superficial Lateral cutaneous nerve of calf lesions
peroneal nerve) and extension of the toes (extensor Pain and/or sensory loss in the popliteal fossa and lateral
digitorum longus and brevis and extensor hallucis part of the calf.
longus; deep peroneal nerve).
• Normal ankle jerk. DIFFERENTIAL DIAGNOSIS
• Altered sensation (numbness or paresthesia) of the lateral Common peroneal neuropathy at the level
part of the lower leg and dorsum of the foot (superficial of the fibular head
peroneal nerve) and/or web between the first two toes • Pyramidal tract lesions (e.g. stroke): cause weakness of
(deep peroneal nerve). The sensory deficit is often less the flexor muscles of the leg, particularly dorsiflexion of
extensive, however, than might be expected from the the ankle, and so may be confused with a peroneal
anatomic distribution of the innervation by the nerve(s). neuropathy, but because other flexor muscles of the leg
Pain is uncommon, particularly when the neuropathy is are affected, the gait pattern is different: there is
due to external compression. circumduction of the hip and the foot tends to be
dragged, rather than causing a stepping gait. Further-
N.B. Confusingly, dorsiflexors of the ankle and toes are more, the muscle tone and deep tendon reflexes are
flexor muscles in the physiologic sense because they shorten increased and the plantar response extensor.
the leg, but have been called extensor muscles by • Anterior horn cell lesions (e.g. motor neuron disease,
anatomists. spinal muscular atrophy): may present with wasting and
weakness of the muscles innervated by the common
Peroneal neuropathy between the fibular peroneal nerve, but usually there is clinical or
head and the ankle electrophysiologic evidence of denervation in at least
Compartment syndromes (see below) three of the four regions formed by the head, arms,
• Severe pain and red discoloration of the skin in the trunk and legs, and no sensory loss.
affected part of the lower limb. The pain may arise • L5 radiculopathy (e.g. L4/5 intervertebral disc herni-
rapidly over hours and may also remit. ation or, less commonly, an extremely lateral prolapse of
• Precipitated or exacerbated by strenuous physical exercise the L5/S1 disc): also causes some weakness of the
(e.g. running). extensors of the ankle and toes, nerve and sensory loss in
• Dorsalis pedis arterial pulse may be absent. the lateral lower leg, but weakness of the extensor
hallucis longus is most pronounced and there is also
Anterior compartment syndrome (deep weakness of ankle inversion due to denervation of the
peroneal nerve compression) tibialis posterior muscle.
• Weakness of dorsiflexion of the ankle (tibialis anterior) • Sciatic neuropathy: may present with a foot drop because
and toes (extensor hallucis longus and extensor the lateral division of the sciatic nerve largely corresponds
digitorum longus), if it is possible to test in the presence with the peroneal nerve, and is more prone to compres-
of severe pain. sion than the portion corresponding to the tibial nerve.
• Sensory loss in the web between the first and second • Compartment syndrome (anterior or lateral): often
toes. involve the deep or superficial peroneal nerves.
• Distal myopathy: usually bilateral and involves muscles
outside those innervated by the peroneal nerves.
Peroneal Neuropathy 653

INVESTIGATIONS Surgical decompression


Common peroneal neuropathy at the level • Clinical features compatible with entrapment of the distal
of the fibular head part of the superficial peroneal nerve where the nerve
Nerve conduction studies pierces the fascia, 10 cm (4 in)above the lateral malleolus.
• May be detected by stimulating the peroneal nerve at the • Penetrating trauma, including immediate or delayed
ankle, and below and above the fibular head while peroneal palsy after surgery in the region of the knee,
recording from the extensor digitorum brevis muscle, which may have disrupted the continuity of the nerve
and identifying local slowing and/or conduction block. and which necessitates immediate exploration.
Recording from tibialis anterior increases the chance of • Local mass lesion causing slowly progressive deficit such
detecting conduction block. as a nerve tumor, lipoma, cyst, or ganglion.
• Nerve conduction studies may also give a clue to the • Compartment syndrome, in which case immediate
prognosis. A reduced or absent SNAP of the superficial fasciotomy is indicated.
peroneal nerve examined in a distal segment is consistent
with axonal loss in the peroneal nerve and indicates a PREVENTION
worse prognosis than conduction block. High risk patients
• Coma.
EMG • Bedridden with leg paralysis: Guillain–Barré syndrome.
Needle EMG may be particularly helpful if no conduction • Leg in plaster that reaches as high as the head of the
abnormalities are found in the nerve segment across the fibula.
fibular head to differentiate a suspected common peroneal
neuropathy from an L4/5 radiculopathy, plexopathy or Strategies of prevention
sciatic neuropathy. Important muscles to study are the short • Protect the common peroneal nerves at the head of the
head of biceps femoris (innervated by the lateral division of fibular by soft padding around the head of the fibula.
the sciatic nerve), and the tibialis posterior (supplied by L5 • Make an appropriate window in plaster casts near the
but not the peroneal nerve). head of the fibula.
• Instruct patients with a leg plaster to check the strength
CT or MRI scan of the brain, spinal cord, lumbosacral of the toe extensors each day and if weak, to open the
spine or leg plaster.
Frequently detects any lesion of the pyramidal tract or disc • Use care with removing a lower leg plaster by sawing or
protrusion. cutting.

DIAGNOSIS
Requires the presence of foot drop due to weakness of
dorsiflexion of the ankle (tibialis anterior), eversion of the
ankle (peroneal muscles) and extension of the toes (extensor
digitorum longus and brevis and extensor hallucis longus);
a normal ankle jerk; and/or altered sensation of the lateral
part of the lower leg and dorsum of the foot (superficial
peroneal nerve) and/or web between the first two toes
(deep peroneal nerve). Can be confirmed by EMG.

TREATMENT
Depends on the cause.

Conservative
• Most cases are due to external compression or stretch
and recover spontaneously through remyelination within
weeks or months with a conservative approach of
observation and avoidance of further compression. A
lightweight plastic orthosis which does not further
compress the peroneal nerve at the head of the fibular
should be used to facilitate a safer and more comfortable
gait.
• Prolonged and severe compression during a long
anesthetic or deep coma may result in a persistent deficit
due to severe axonal damage (identified by EMG
examination). Surgical decompression is ineffective.
654 Mononeuropathies

SURAL NEUROPATHY • The sural nerve descends the calf more laterally, between
the Achilles tendon and the lateral malleolus.
• It curves around the lateral malleolus and ends at the
DEFINITION lateral border of the foot.
Dysfunction of the sural nerve. • It innervates the skin of the lateral side of the ankle, and
lateral border of the sole, up to the base of the fifth toe
EPIDEMIOLOGY (796).
Uncommon.
ETIOLOGY
ANATOMY Sural neuropathy in the popliteal fossa
Course of the sural nerve (795) • Baker’s cyst.
• Arises from the confluence of the medial cutaneous nerve • Arthroscopy, or other operations in the popliteal fossa
of the calf (from the tibial nerve) and the lateral (e.g. for varicose veins).
cutaneous nerve of the calf (from the common peroneal
nerve). Sural neuropathy in the calf
• The medial cutaneous nerve of the calf arises from the • Tight chains, lacing or elastic socks around the calf.
tibial nerve in the popliteal fossa and descends in the • High-topped footwear.
middle of the calf between the two heads of • Pressure against a hard ridge.
gastrocnemius. After it has pierced the fascia it is joined • Calf muscle biopsy.
beneath the skin by the branch from the lateral
cutaneous nerve of the calf, which originates from the Sural neuropathy at the ankle:
common peroneal nerve. • Sitting with, or on, crossed ankles.
• Adhesions after soft tissue injury.
• Avulsion fracture of the base of the fifth metatarsal bone.
795 • Fractured sesamoid bone in the peroneus longus tendon.
• Osteochondroma.
• Neuroma.
• Ganglion.
L4 • Sural nerve biopsy (complicated by persistent pain in
L5 about 5% of cases and infections or delayed wound
S1 healing at the site of biopsy in about 20% of patients).
S2
S3
S4 CLINICAL FEATURES
S5 • Pain, paresthesia or numbness of the lateral ankle or sole.
Sciatic nerve • Local pressure may aggravate the sensory symptoms
(Hoffmann–Tinel sign) and indicate the site of the
lesion.

DIFFERENTIAL DIAGNOSIS
S1 radiculopathy.

796
Common
peroneal nerve
Tibial nerve

Lateral cutaneous
Medial cutaneous nerve of the calf
nerve of the calf

Sural nerve

795 Origin and course of the sural nerve. 796 Area of skin innervated by the sural nerve.
Pudendal Neuropathy 655

INVESTIGATIONS PUDENDAL NEUROPATHY


Nerve conduction studies
The amplitude of the SNAP is normally large and easy to
elicit with lesions proximal to the sensory ganglion (e.g. S1 DEFINITION
radiculopathy) but are decreased or absent with lesions Dysfunction of the pudendal nerve.
causing axonal degeneration distal to the sensory ganglion
(e.g. of the sural nerve). EPIDEMIOLOGY
Not uncommon.
DIAGNOSIS
Requires the presence of isolated sensory loss the skin of the ANATOMY
lateral side of the ankle, and lateral border of the sole, up to Course of the pudendal nerve (797)
the base of the fifth toe. Can be confirmed by EMG. • Arises from the S2, S3 and S4 nerve roots and innervates
most of the perineum.
TREATMENT • Descends from the pelvis below the piriformis muscle,
Depends on the cause: crosses the sacrospinous ligament, and enters the
• Advice how to avoid external compression, if causal (e.g. perineum through the lesser sciatic notch.
special postures with crossed ankles, calves against hard • Courses anteriorly, with its associated blood vessels, along
ridges, high-topped boots or tight chains). the intrapelvic wall within a tunnel in the dense obturator
• Surgery (neurolysis or nerve section) for compression by fascia (the obturator canal or Alcock’s canal) and
post-traumatic fibrosis or tumors. terminates by dividing into three branches:
– The inferior rectal (hemorrhoidal) nerve, which may also
arise from S3 and S4, supplies the external anal sphincter,
the perianal skin and the mucosa of the lower anal canal.
– The perineal nerve innervates the muscles of the
perineum (e.g. bulbocavernosus), the erectile tissue of
the penis, the external urethral sphincter, the distal part
of the mucous membrane of the urethra and the skin of
the perineum and labia/scrotum.
– The dorsal nerve of the clitoris/penis courses forward in
Alcock’s canal, pierces the urogenital diaphragm, and
sends a branch to the corpus cavernosum before running
forward on the dorsum of the clitoris/penis to innervate
the skin, prepuce and glans.

ETIOLOGY
External compression
• Prolonged bicycle ride.
• Colposcopy with suture through the sacrospinal ligament
and subsequent nerve entrapment.
• Operation on hip fracture using perineal post.

797 The pudendal nerve and its branches. 797

S2

S3

S4
Pubic bone

Pudendal nerve Striated urethral


sphincter
Inferior rectal nerve
Dorsal nerve of
External anal sphincter the penis
Perineal nerve
Bulbocavernosus muscle
656 Mononeuropathies

Stretch injury INVESTIGATIONS


• Childbirth: may cause anal sphincter muscle division, and Anorectal manometry
additional damage to the pudendal nerve. Vaginal May assess the function of the pudendal nerve to some
delivery also causes compression of the pudendal nerves. degree.
• Straining during defecation.
• Pelvic fracture or surgery. Urodynamics
May assess the function of the pudendal nerve to some
Polyneuropathy degree.
• Diabetic.
• Alcoholic. Nerve conduction studies
Infrarectal stimulation of the terminal parts of the pudendal
CLINICAL FEATURES nerves by mounting disposable surface electrodes on the
History index finger of a disposable glove and recording the evoked
• Incontinence of urine or feces (pudendal neuropathy is muscle potential from the external anal sphincter (inferior
a common cause of fecal incontinence, mainly lesions in rectal nerve) and urethral sphincter (perineal nerve), the
the distal segment of the pudendal nerve or in the latter via electrodes mounted on a Foley catheter, is possible.
inferior rectal nerve).
• Impotence. EMG
• Altered sensation in one-half or all of the labia Needle EMG of the external anal sphincter, external urethral
majora/penis and perineum. sphincter (difficult and unpleasant for the patient),
bulbocavernosus and puborectalis muscles may be helpful.
Examination
• Absent anal reflex (S3 and S4 nerve roots and pudendal Reflex responses
nerve): normally, pricking the perianal skin elicits a reflex Stimulation of the clitoris/penis and recording from the
contraction of the external anal sphincter which can be anal sphincter and bulbocavernosus.
seen as a dimpling of the perianal skin.
• Absent bulbocavernosus reflex (S3 and S4 nerve roots Motor and somatosensory evoked potentials
and pudendal nerve): normally, compression of the glans Can be used to study central as well as peripheral nerve
penis evokes a contraction of the bulbocavernosus conduction to and from the perineal region respectively.
muscle, which is best assessed clinically by palpation.
CT or MRI scan of the sacral spine or pelvis
DIFFERENTIAL DIAGNOSIS
• Conus medullaris or cauda equina lesion. DIAGNOSIS
• Polyneuropathy. Requires the presence of incontinence of urine or feces,
• Disturbance of vascular supply or autonomic innervation impotence, and altered sensation in half or all of the labia
(e.g. diabetes). majora/penis and perineum.
• Structural abnormalities of the pelvic floor and relevant
viscera. TREATMENT
Depends on the cause.

FURTHER READING Etiology Treatments


Stevens JC, Beard CM, O’Fallon WM, Kurland Dammers JWHH, Veering MM, Vermeulen M
LT (1992) Conditions associated with carpal (1999) Injection with methylprednisolone
tunnel syndrome. Mayo Clin. Proc., 67: proximal to the carpal tunnel: randomized
SUPRASCAPULAR NEUROPATHY 541–548. double blind trial. BMJ, 319: 884–886.
Antoniadis G, Richter HP, Rath S, et al (1996). Ebenbichler GR, Resch KL, Wiesinger GF, Uhl F,
Suprascapular nerve entrapment: experience Pathology Ghanem A-H, Fialka V (1998) Ultrasound
with 28 cases. J. Neurosurg., 85: 1020–1025. Jenkins PJ, Sohaib A, Akker A, et al. (2000) The treatment for treating the carpal tunnel syn-
Van Zandijcke M, Casselman J (1999) Supras- pathology of median neuropathy in drome: randomized ‘sham’ controlled trial.
capular nerve entrapment at the spinoglenoid acromegaly. Ann. Intern. Med., 133: BMJ, 316: 731–735.
notch due to a ganglion cyst. J. Neurol. Neu- 197–201. Garfinkel MS, Singhal A, Katz WA, et al. (1998)
rosurg. Psychiatry, 66: 245. Yoga-based intervention for carpal tunnel syn-
Diagnosis
D’Arcy CA, McGee S (2000) Does this patient drome. A randomized trial. JAMA, 280:
CARPAL TUNNEL SYNDROME 1601–1603.
Epidemiology have carpal tunnel syndrome? JAMA, 283:
Atroshi I, Gummesson C, Ornstein E, Ranstam J, 3110–3117. SCIATIC NEUROPATHY
Rosen I (1999) Prevalence of carpal tunnel Franzblau A, Werner RA (1999) What is carpal Vroomen PCAJ, de Krom MCTFM, Slofstra PD,
syndrome in a general population. JAMA, tunnel syndrome? JAMA, 282: 186–187. Knottnerus JA (2000) Conservative treatment
282: 153–158. Witt JC, Stevens JC (2000) Neurological disor- of sciatica: A systematic review. Journal of
MacFarlane GJ (2001) Identification and preven- ders masquerading as carpal tunnel syndrome: Spinal Disorders, 13: 463–469.
tion of work-related carpal-tunnel syndrome. 12 cases of failed carpal tunnel release. Mayo
Lancet, 357: 1146–1147. Clin. Proc., 75: 409–413.
Chapter Twenty-three 657

Neuromuscular
Junction
Disorders
MYASTHENIA GRAVIS (MG) Thymus
• Thymoma occurs in 10% of patients, which may be
locally invasive.
DEFINITION • Medullary hyperplasia, characterized by lymphoid
Myasthenia gravis is an acquired autoimmune follicles with germinal centers, occurs in about 60% of
neuromuscular disorder in which serum IgG antibodies patients, usually those presenting before 40 years of age.
(anti-AChRAb) to acetylcholine receptors (AChR) in the
post-synaptic membrane of the neuromuscular junction lead ETIOLOGY
to receptor loss, and skeletal muscle fatigue and weakness. Autoimmune
Myasthenic crisis is when weakness of the muscles of • Anti-AChRAb reduce the number of AChRs by
respiration is severe enough for the patient to require accelerated endocytosis and degradation of the receptors,
mechanical ventilation. functional blockade of acetylcholine (ACh) binding sites
and complement-mediated damage to the AChRs.
EPIDEMIOLOGY • B lymphocytes produce the anti-AChRAb but T
• Prevalence: 5–18 cases per 100 000 population. lymphocytes have a key role in the autoimmune response.
• Lifetime prevalence: 0.4 (95% CI: 0.2–0.7) per 1000. • The origin of the autoimmune response is unsolved. The
• Incidence: 3(95% CI: 0.8–7) per 100 000 per year. thymus has been implicated because about 75% of
Incidence is age- and sex-related, with one peak in the patients have thymic abnormalities (85% thymic
second and third decades affecting mostly women and a hyperplasia [germinal-center formation] and 10–15%
peak in the sixth and seventh decades affecting mostly thymoma) and thymectomy improves most patients.
men. • Genetic factors and abnormalities of immune regulation
• Age: any age, from early childhood to old age. may increase the likelihood of MG: there is a moderate
– Thymoma-associated myasthenia gravis has a peak association with the HLA antigens B8 and DRw3; a
incidence at 40–50 years of age. stronger association with HLADQw2 is still contro-
– Non-thymoma-associated MG has a peak incidence at versial.
10–30 and 60–70 years of age. • A wide variety of other autoimmune diseases are
• Gender: M=F. associated with MG (see below).

PATHOLOGY Disorders or factors that may exacerbate MG


End-plate abnormalities • Thyroid disease: hyperthyroidism, hypothyroidism.
A decrease in number (by about two-thirds) of nicotinic • Infection.
acetylcholine receptors in the post-synaptic membrane at • Drugs: aminoglycoside antibiotics (streptomycin,
neuromuscular junctions of skeletal muscle, with gentamicin, kanamycin, neomycin), polymyxin, colistin,
simplification of postsynaptic folds and widening of the curare, quinidine, quinine and antiarrhythmic agents
synaptic cleft. The degree of reduction of AChRs generally such as procainamide.
correlates with the severity of MG.
658 Neuromuscular Junction Disorders

PATHOPHYSIOLOGY transmission. Impairment of transmission induces


• Muscle contraction depends on effective neuromuscular increased transcription of AChR genes. Consequently,
transmission. virtual complete recovery can occur in patients with MG
• Effective neuromuscular transmission depends on the if the autoimmune attack can be controlled.
number of interactions between ACh molecules released
from the nerve terminal, and AChRs on the post-synaptic CLINICAL FEATURES
membrane of the neuromuscular junction (798). • The clinical hallmarks are muscular weakness and
• The nicotinic ACh receptor is a glycoprotein that fatiguability.
projects through the muscle membrane and is composed • The weakness tends to increase with repeated activity and
of five subunits (two a, one b, one d, and one g or e improves with rest.
subunit), arranged like barrel staves around a central • Weakness usually occurs in a characteristic distribution.
channel. Each of the two a subunits has an ACh-binding
site that is located extracellularly. Ocular muscles
• In the resting state the ion channel of the AChR is closed. • The eyelid and extraocular muscles are the first muscles
• When ACh binds to the binding sites of both a subunits to be involved in about 65% of patients, and are affected
of the AChR, the receptor’s cation channel opens at some stage of the disorder in >90% of patients, causing
transiently, allowing the rapid passage of cations and ptosis (799) and diplopia, which are typically asymmetric.
producing a localized electric end-plate potential. • Weakness remains confined to the eyelid and extraocular
• If the amplitude of this potential is sufficient, it generates muscles in about 15% of patients (ocular myasthenia).
an action potential that spreads along the length of the • At the bedside, fatiguable ptosis can be demonstrated by
muscle fiber, triggering the release of calcium from asking the patient to maintain upward gaze without
internal stores and leading to muscle contraction. blinking for 30–60 seconds.
• At the normal neuromuscular junction, the end-plate
potentials are more than sufficient to generate muscle Facial and bulbar muscle weakness
action potentials consistently, without failures. • Leads to loss of facial expression (a characteristic
• At the myasthenic neuromuscular junction, the decreased ‘flattened’ or ‘snarling’ smile), inability to whistle, and
number of AChRs results in end-plate potentials of difficulty with speech (‘mushy’ or nasal speech), chewing
diminished amplitude, which fail to trigger action and swallowing.
potentials in some fibers. When transmission fails at many • Weakness of the neck extensors and muscles of
junctions, the power of the whole muscle is reduced, mastication may necessitate the patient to use one of
causing muscle weakness. When contractions are their hands to prop up their jaw.
repeated, due to repeated nerve stimulation, the amount
of ACh released per nerve impulse normally declines Limb weakness
(runs down) after the first few impulses because the • Generalized weakness develops in about 85% of patients
nerve terminal is not able to sustain its initial rate of and may affect the proximal and distal limb muscles, neck
release. If there is a reduced number of AChRs, as is the extensors (and diaphragm).
case in MG, this ACh ‘rundown’ results in progressive • Initially, it may be episodic (e.g. sudden weakness of the
failure of transmission at more and more junctions and legs) and precipitated by emotional stress or infection.
muscle power progressively declines, causing fatiguability. • Characteristically, the weakness is increased by exercise,
• AChRs normally undergo continuous turnover at the tone is normal and deep tendon reflexes are generally
neuromuscular junction, depending on regulating brisk.
influences of the motor nerves and neuromuscular • At the bedside, fatiguable limb weakness can be demon-
strated by asking the patient to elevate the outstretched
arm against resistance for 30–60 seconds, and comparing
798 the strength with the opposite rested limb.
Axon
Vesicles
Mitochondria 799
Release site

Nerve terminal ↓ ACh

ACh receptors AChE

Muscle
Endplate

798 Diagrammatic representation of the neuromuscular junction, 799 Bilateral ptosis in a patient with myasthenia gravis.
showing the site of acetylcholine receptors of the post synaptic
membrane. (ACh = acetylcholine; AChE = acetylcholinesterase.)
Myasthenia Gravis (MG) 659

Respiratory muscle weakness Botulism


May lead to shortness of breath and, in severe cases, • Generalized weakness.
ventilatory failure. • Dilated pupils.
• Ophthalmoplegia.
Associated other immune conditions • Nerve conduction studies: incremental response in
Susceptibility is determined through immune response repetitive nerve stimulation.
genes (e.g. HLA):
• Neuromyotonia (continuous muscle fiber activity caused Inflammatory demyelinating polyradiculoneuropathies
by peripheral nerve hyperexcitability). • Acute (Guillain–Barré) motor type.
• Thyroid disease: hyperthyroidism occurs in about 5% • Miller–Fisher syndrome.
(3–8%) of patients with MG, and hyper- and • Chronic.
hypothyroidism may aggravate myasthenic weakness.
• Polymyositis. Hyperthyroidism
• Rheumatoid arthritis. • Exacerbates MG.
• Systemic lupus erythematosus. • Generalized weakness.
• Thyroid function tests are abnormal.
SPECIAL FORMS
Neonatal myasthenia ‘Functional’ disorder
• A transient illness, lasting <1 month. MG is sometimes misdiagnosed as a ‘functional’ disorder,
• It occurs in 1/8 babies of myasthenic mothers. perhaps because of the fatiguing characteristics of the muscle
• It is due to placental transfer of maternal anti-AchRAb weakness, its worsening with stress, and the lack of other
to the baby. neurologic signs.
• Arthrogryposis (severe joint contractures occurring in
utero) is occasionally present. Ocular myasthenia
Progressive external ophthalmoplegia (mitochondrial
Juvenile myasthenia cytopathy)
Myasthenia in patients younger than 18 years of age is • Ptosis, diplopia.
termed juvenile myasthenia. The illness is similar to MG in • Generalized weakness in some cases.
young adults. • Mitochondrial DNA: abnormal.

Penicillamine-induced myasthenia Oculopharyngeal muscular dystrophy


• This is similar to adult MG, including the presence of
anti-AchRAb. Grave’s disease
• It usually resolves over several months after withdrawal • Diplopia.
of the drug. • Exophthalmos.
• Thyroid-stimulating immunoglobulin is present.
DIFFERENTIAL DIAGNOSIS
Generalized myasthenia Intracranial mass compressing cranial nerves
Congenital myasthenic syndromes • Ophthalmoplegia.
• Rare. • Cranial nerve weakness.
• Inherited, usually as recessive disorders. • CT or MRI brain abnormality.
• Early-onset.
• Not autoimmune disorders; anti-AchRAb is undetectable. Bulbar myasthenia
• Sophisticated electrophysiologic and immunocyto- Brainstem stroke
chemical tests required for diagnosis.
• Most forms are non-progressive. Motor neuron disease (pseudobulbar palsy)

Drug-induced myasthenia INVESTIGATIONS


• Penicillamine: Anti-AChRAb assay
– Triggers autoimmune myasthenia. • 85–90% of patients with generalized MG and 60% of
– Recovery within weeks after drug withdrawal. patients with ocular disease have elevated titers of serum
• Curare, procainamide, quinines, aminoglycosides: antibodies to AChR that are detectable by an immuno-
– Weakness in normal people. precipitation assay.
– Exacerbation of MG. • Anti-AchRAb is specific to MG and is therefore useful in
– Recovery after drug withdrawal. diagnosis.
• In an individual patient, the antibody titer generally
Lambert–Eaton syndrome correlates with the severity of the disease, as shown by
• Weakness, fatigue, areflexia. the response to plasma exchange and, in the longer term,
• 60% have associate small cell lung cancer. to immunosuppressive drugs. Correlation of titer and
• Nerve conduction studies: incremental response on disease severity across patients, however, is poor (i.e. a
repetitive nerve stimulation. patient with mild disease can have a high titer and a
• Antibodies to calcium channels are present. patient with severe disease can have a low titer) probably
because of antibody heterogeneity.
Venoms (snakes, scorpions, spiders) Continued overleaf
660 Neuromuscular Junction Disorders

Anti-AChRAb assay (continued) • The nerve to a clinically weak or proximal muscle is stimu-
• The antibody remains detectable in many patients in lated supramaximally at a rate of 3 per second and the
clinical remission. muscle action potentials are recorded from surface elec-
• Among the 10–20% of patients with MG who do not trodes over the muscle. A progressive decrement in the
have detectable anti-AChRAb, antibodies to another amplitude of the evoked action potential of 15% or more is
neuromuscular junction protein muscle specific kinase considered abnormal (802). The test is generally unpleasant
(MuSK) are present. for the patient, and false positives and negatives may occur.

Antibodies to muscle specific kinase (MuSK) Single fiber EMG


• Inhibit the function of MuSK • This test detects delayed or failed neuromuscular
• MuSK is a receptor tyrosine kinase that is an essential transmission in pairs of muscle fibers supplied by
component of the developing neuromuscular function. branches of a single nerve fiber (803, 804).
• A sensitive but not highly specific test. Positive in about
Anti-striated muscle antibody assay 90% (88–92%) of cases, its specificity is limited because
Detectable in >90% of patients with thymoma and in about of positive findings in other disorders of nerves,
30% of other patients with MG. neuromuscular junction, and muscle.

Anticholinesterase (edrophonium chloride) Chest x-ray and CT or MRI of the mediastinum


test (800, 801) (thymoma, thymic hyperplasia)
Edrophonium (Tensilon) is an anticholinesterase with a rapid • CT and MRI reliably reveal enlargement of the thymus
onset (30 seconds) and short duration (5 minutes) of action. gland.
• The thymus is normally detectable until mid-adulthood but
Indications persistence of the thymus in a patient with MG who is over
• The diagnosis has to be made immediately because of 40 years of age, or an increase in its size in any patient on
disease severity. repeated scanning, raises the possibility of a thymoma.
• The patient is sero-negative for anti-AchRAb. • If negative initially, it should be repeated after 2–3 years
in those who are positive for anti-AchR and antistriated
Procedure muscle antibody.
• The test is undertaken in hospital where full resuscitative
equipment must be readily available. Screen for other disorders
• Pre-treat with atropine, 0.6 mg i.v. to counteract para- • Antinuclear antibody.
sympathetic overstimulation and avoid occasional adverse • Antithyroid antibodies.
effects. • Rheumatoid factor.
• Assess the strength of an objectively weak muscle or group • Fasting blood glucose.
of muscles (e.g. degree of ptosis or diplopia; vital capacity). • Thyroid function tests.
• Inject a test dose of edrophonium (Tensilon) 1–2 mg i.v. • Pulmonary function tests.
• If no adverse effects develop, inject a further 5–6 mg. • Tuberculin test.
• A positive response is indicated by an obvious • Bone densitometry in older patients.
improvement in strength within 1 minute (e.g. abolition • Mitochondrial DNA analysis (if ocular myasthenia).
of ptosis, an increase in the length of time that the arm
can be kept outstretched, and increase in vital capacity) Screen for disorders that may interfere with
which disappears within 5 minutes. immunosuppressive therapy
• This test can be combined with EMG measurements of • Hypertension.
neuromuscular transmission. • Unsuspected infections such as tuberculosis.
• Peptic ulcer.
An alternative, less commonly used, procedure • Occult gastrointestinal bleeding.
• Administer atropine intramuscularly into one leg and • Renal disease.
wait for a placebo response. • Diabetes.
• If negative, administer neostigmine 1.5 mg i.m. into the • Osteoporosis.
other leg.
Tests
The atropine counteracts parasympathetic overstimu- • Blood pressure, fundoscopsy, EKG, serum creatinine.
lation and also serves as a placebo control for motor effects. • Occult blood.
Neostigmine i.m. provides more time for tests and is less • Fasting blood glucose.
risky than i.v. edrophonium (i.e. from anaphylaxis). • Chest x-ray.
• Pulmonary function tests.
EMG • Tuberculin test.
Results can be misleading in patients who are already receiv- • Bone densitometry in older patients.
ing anticholinesterase medication; in such patients the drug
should, if possible, be withdrawn for 1 week before the study. DIAGNOSIS
• As a diagnosis of MG almost commits the patient to long
Repetitive nerve stimulation term medical or surgical treatment that carries substantial
• A useful supplement to the clinical diagnosis when risks, every effort should be made to establish the
antibody testing is negative. diagnosis before initiating treatment.
Myasthenia Gravis (MG) 661

• Anti-AchRAb testing is reliable, and a positive result is Group I


virtually definitive in the majority of patients, although a • Onset: early (<40 years).
negative assay result does not exclude MG. The other • Gender: F>M.
tests listed above may be useful in patients with doubtful • Weakness: generalized usually.
anti-AchRAb results. • Thymus: hyperplasia.
• Other conditions that mimic MG should be excluded, • Anti-AChRAb titers: high.
and associated conditions that may influence the choice
of treatment should be searched for. Group II
• Gender: M=F.
CLASSIFICATION • Weakness: generalized weakness usually.
Four main groups of acquired myasthenia gravis can be • Thymus: thymoma (which may be benign or locally
distinguished which have implications for treatment: invasive).
• Anti-AChRAb titers: intermediate.

800, 801 Myasthenic ‘snarl’ 800 801


(800) as the patient tries to
open his eyes, due to facial
weakness, before an
edrophonium test. Restored
muscle power (801) within
minutes of injection of
edrophonium 5 mg i.v.
(Courtesy of Dr AM
Chancellor, Neurologist,
Tauranga, New Zealand.)

802 803

804
802 Supramaximal repetitive ulnar nerve stimulation at 5 Hz showing
ten successive muscle action potentials recorded over abductor digiti
minimi and a decrement in amplitude of more than 15% between the
first and fourth response.

803, 804 Single fiber EMG. 803 Normal ‘jitter’ of the second,
compared with the first muscle fiber, supplied by a single nerve fiber.
The responses of the second muscle fiber, relative to the first, are
virtually superimposed upon one another due to the absence of any
delay in neuromuscular transmission. 804 Myasthenia gravis patient in
whom ‘jitter’ is substantially increased due to variable delay in
neuromuscular transmission.
662 Neuromuscular Junction Disorders

Group III perioperative infection. If the vital capacity is below 2 l,


• Onset: late (>40 years). plasmapheresis should be carried out preoperatively.
• Gender: M>F.
• Weakness: generalized or ocular. Technique
• Thymus: atrophy/involution. Thymectomy should remove as much of the thymus as
• Anti-AChRAb titers: low. possible. Although a cervical incision with mediastinoscopy
is associated with a smaller scar and less postoperative pain,
Group IV it has not been confirmed that this method consistently
• Weakness: ocular only or generalized. enables the thymus to be removed completely. A sternal-
• Thymus: atrophy/involution. splitting approach with exploration extending into the neck
• Anti-AChRAb titers: absent, but autoantibodies to other optimizes removal of all thymic tissue and related fat.
muscle cell targets are implicated. Epidural morphine minimizes postoperative pain and
thereby enhances respiratory effort. For a few days after
MODES OF THERAPY thymectomy, the requirement for anticholinesterase
In general, four methods of treatment are used: medication may be decreased and so only about three-
• Anticholinesterase agents. quarters of the preoperative dose needs to be given
• Thymectomy. intravenously postoperatively.
• Immunosuppression.
• Short term immunotherapies. Outcome
The benefits of thymectomy are usually delayed until
Anticholinesterase medication months to years after surgery. Clinical remission occurs in
• Most useful in mild cases. about one-third of patients and remission in another half,
• Pyridostigmine (Mestinon) 30–120 mg p.o., or which is substantially better than if not treated surgically.
neostigmine bromide 15–30 mg p.o.; both given every Mechanism of effect
3–4 hours except at night. The mechanism by which thymectomy produces benefit
• Neostigmine can also be given subcutaneously or in MG is still uncertain. Possible reasons include removal of
intramuscularly (15 mg p.o. is equivalent to 1 mg a source of continued antigenic stimulation, removal of a
subcutaneously or intramuscularly), preceded by atropine reservoir of B cells secreting AChR antibodies, and
0.5–1 mg. correcting a disturbance of immune regulation. Whatever,
• Pyridostigmine usually produces a smoother response. generally, anti-AChRAb levels fall after thymectomy.
• Adverse effects are caused by parasympathetic stimulation:
pupillary constriction; colic; diarrhea; increased salivation, Immunosuppressive treatment
sweating, lacrimation, and bronchial secretions. Indications
• Gastrointestinal adverse effects can be controlled with Immunosuppressive treatment should be considered when
propantheline bromide, atropine or loperamide. weakness is not adequately controlled by anticholinesterase
drugs and is sufficiently distressing to outweigh the risks of
Thymectomy possible adverse effects of immunosuppressive drugs.
Thymectomy has two purposes: to prevent the spread of
thymoma and to induce remission, or at least improvement, Duration
permitting a reduction in immunosuppressive medication. Treatment must be continued for a prolonged period, often
permanently.
Indications
Thymic tumors must be removed surgically since they may Corticosteroids
spread locally and become invasive, though they rarely The most consistently effective immunosuppressive drugs but
metastasize. The tumor and remaining thymus gland should they have the largest array of adverse effects. When initiating
be removed as completely as possible. If not, or if it is invasive, steroid therapy in patients with moderate to severe generalized
marker clips should be placed at the tumor site during surgery weakness, the patient should be hospitalized because of the
and focused radiation treatment carried out postoperatively. risk of transient steroid-induced exacerbation in the first few
After removal of a thymoma, some patients become weaker, weeks in up to one-half of patients. This risk is minimized by
presumably as a loss of a suppressive effect of the thymoma, beginning with a daily dose of 10–20 mg of prednisone and
and require further immunosuppressive treatment. increasing it gradually by about 5 mg every 2–3 days,
Thymectomy is indicated for generalized MG in patients according to the patient’s clinical condition and response, until
who are between adolescence and about 60 years of age. a satisfactory response is achieved or the dose reaches
The only reason for delaying thymectomy in pre-pubertal 50–60 mg a day. However, the favorable effect of steroids may
children, if possible, is because of the role of the thymus in take 2–4 weeks to become apparent and maximal benefit may
development of the immune system. It is uncertain whether not be realized until after 6–12 months or more.
thymic tissue persists in patients with MG who are older After about 3 months of daily high-dose treatment, a
than 60 years. switch to an alternate day regime may help to minimize
Thymectomy has also been undertaken in purely ocular adverse effects but a small dose of prednisone may be
MG with good results. Thymectomy should never be required on the ‘off ’ day if muscle strength fluctuates
performed as an emergency procedure. The patient’s strength significantly. Over the next few months to years, the total
and respiratory function in particular should be optimized dose should be tapered slowly (e.g. 5 mg/month) to the
preoperatively, but not with immunosuppressive agents if they minimum effective dose. Few patients are able to be weaned
can possibly be avoided because they increase the risk of off prednisone completely.
Myasthenia Gravis (MG) 663

The mechanisms by which corticosteroids work in MG TREATMENT


may include a reduction in anti-AChRAb levels, diminished Treat or avoid disorders and drugs that interfere
anti-AChRAb reactivity of peripheral blood lymphocytes, with neuromuscular transmission
and increased synthesis of AChRs. • Thyroid disease: hyperthyroidism, hypothyroidism.
• Infection.
Azathioprine • Drugs: aminoglycoside antibiotics (streptomycin,
Azathioprine is metabolized to the cytotoxic derivative 6- gentamicin, kanamycin, neomycin), polymyxin, colistin,
mercaptopurine and acts predominantly on T cells. It is curare, quinidine, quinine and antiarrhythmic agents
most useful for patients in whom steroids are contraindi- such as procainamide.
cated, in whom steroids evoked an insufficient response, or
as an adjunct to permit a reduction in steroid dose. The Anticholinesterase medication to enhance
drug is well tolerated but up to 10% of patients have an neuromuscular transmission
idiosyncratic influenza-like reaction (fever, myalgia and • First line treatment for all patients.
malaise) and it takes many months to 1 year to show its • Pyridostigmine (Mestinon) 30–60 mg every 4 hours has
therapeutic effect. an effect within 30 minutes that peaks at 2 hours and
Treatment begins with a test dose of 50 mg daily for gradually declines thereafter. The timing of administra-
1 week and, if tolerated, the dose is gradually increased to tion should be tailored to the patient’s needs, i.e. to
the target dose of 2–3 mg/kg total body weight (not lean avoid peaks and valleys of strength, and before meals or
body mass) per day. If patients are also taking allopurinol, physical activity. The maximum daily dose rarely exceeds
the dose of azathioprine must be reduced by as much as 75% 120 mg every 3 hours. Higher doses may increase
and the white blood cell count and mean corpuscular volume weakness (receptor desensitization or rarely cholinergic
monitored closely. Complete blood count and liver function block). A sustained-release preparation of Mestinon
tests should be undertaken weekly for the first 8 weeks and (Timespan) should only be used at bedtime, and only if
every 3 months thereafter. Treatment is usually lifelong. the patient experiences significant weakness during the
night or in the early morning.
Combined prednisolone and azathioprine • Provides symptomatic relief in most patients, particularly
Among patients with moderate or severe symptoms, a when weakness is mild, but the improvement is often
randomized controlled trial showed that the combination incomplete and wanes after a few weeks or months.
results in lower doses of prednisolone, fewer relapses, longer • If weakness interfering with normal activities persists,
periods of remission, and fewer adverse effects than additional measures should be considered as described
prednisolone alone. below.

Methotrexate Ocular cases


Weekly methotrexate, 5–10 mg, is an alternative to the few • Anticholinesterase medication suffices for a few patients.
patients who are unable to tolerate, and need, azathioprine. • Prednisolone, in relatively low dose (e.g. 30 mg on
alternate days), is commonly effective for these patients
Mycophenolate mofetil who have persistent symptoms despite anticholinesterase
A novel and potent immunosuppressive agent, which blocks medication.
purine synthesis in activated T and B lymphocytes. In an
open-label study, the addition of mycophenolate mofetil 1 g Thymoma patients
twice daily for 6 months to corticosteroids improved Group II patients
function within 2 weeks to 2 months in 8 out of 12 patients. • Thymectomy is indicated because the tumor can infiltrate
No major adverse effects were observed. Randomized local mediastinal structures.
controlled trials are required. • The myasthenic symptoms do not usually respond to
removal to the thymus and tumor in these patients.
Short term immunotherapies • Immunosuppressive drug treatment (prednisolone and/or
Plasma exchange azathioprine) is often effective in controlling myasthenic
Three to five daily exchanges of 50 ml/kg body weight may symptoms before thymectomy and afterwards.
produce striking short term clinical improvement. The
therapy is useful when combined with immunosuppressive Patients without thymoma
drug treatment, if patients have severe disease and can Group I patients
enable patients who might normally require hospital • Thymectomy is indicated for patients in group I (see
admission to remain at home. above) with moderate or severe weakness; about 75% of
patients will show improvement or remission.
Intravenous immunoglobulin (IVIG) • Immunosuppressive drug treatment should be
IVIG 0.4 g/kg body weight for 5 days may be as effective considered in those who fail to respond to thymectomy
as a 5-day course of plasma exchange. or who are too ill to undergo surgery.

Group III and IV patients


Immunosuppressive drug treatment should be considered
in these patients who have persistent symptoms despite
anticholinesterase medication; about 80% of patients treated
with prednisolone and/or azathioprine at optimal dose
regimens show marked improvement or remission.
664 Neuromuscular Junction Disorders

Pregnant patients with myasthenia LAMBERT–EATON MYASTHENIC


Weakness does not usually increase during pregnancy, but SYNDROME (LEMS)
may do so in the postpartum period for a few months:
• Maintain existing (pre-pregnancy) treatments, including
immunosuppressive drugs, during the pregnancy; there DEFINITION
is no evidence that prednisolone or azathioprine, as used Lambert–Eaton myasthenic syndrome is an antibody-
in MG, are teratogenic. mediated autoimmune disorder of neuromuscular
• Plasma exchange can also be used if needed. transmission, characterized by muscle weakness, hyporeflexia
or areflexia and autonomic dysfunction.
Neonatal myasthenia
General care of the infant (intubation and assisted HISTORY
ventilation, nasogastric feeding, and removal of pharyngeal In 1953 Anderson and colleagues reported a case of a
secretions) should be provided and anticholinesterase 47 year old man with a bronchial neoplasm, progressive
medications given. muscle weakness and hyporeflexia who developed
progressive apnea following administration of succinyl-
Myasthenic crisis choline. They concluded that there was ‘strong clinical
Crisis may be precipitated by infection, initiation of evidence for believing that the severe muscle weakness was
corticosteroid treatment at high dosage, or inadequate of the myasthenic type’.
treatment: In 1956, at a meeting of the American Physiological
• Maintain the airway and provide assisted ventilation and Society, Lambert, Eaton and Rooke presented a report of
anticholinesterase medication. six patients with defective neuromuscular transmission
• Intravenous immunoglobulin (IVIG) therapy or plasma associated with malignant neoplasms. They identified that
exchange can be used if needed. some of the clinical and electrophysiologic features were
different from what was expected in myasthenia gravis.
Cholinergic crisis Subsequently in 1957, Eaton and Lambert summarized the
Crisis occurs due to excess anticholinesterase medication: clinical and electrophysiologic characteristics of the
• Maintain the airway and provide assisted ventilation and myasthenic syndrome.
atropine, if not already being given.
• Withdraw anticholinesterase medication temporarily, and EPIDEMIOLOGY
reintroduce later at a reduced dose. • Incidence: rare.
• Immunosuppressive drug therapy and/or plasma • Age: onset >40 years in >80% of patients.
exchange can be used if needed. – Carcinoma-associated LEMS: mean age at presentation
58 years (rarely before 30 years of age).
PROGNOSIS – LEMS without carcinoma: mean age at presentation
The prognosis for patients with MG has improved 48 years.
dramatically in recent years. Now, with optimal care, the • Gender: M≥F (?dependent on smoking patterns).
mortality rate is zero. Remission or substantial improvement
can be expected in 80% of patients and most patients lead ETIOLOGY AND PATHOPHYSIOLOGY
normal lives but take immunosuppressive medication indefi- The release of neurotransmitters at presynaptic motor nerve
nitely. Without treatment, 20–30% will die within 10 years. terminals of the neuromuscular junction and autonomic
neurons depends on the influx of calcium through the
presynaptic voltage-gated calcium channels (VGCC).
LEMS is caused by IgG autoantibodies directed against
the presynaptic VGCC, particularly the P/Q type of VGCC,
leading to inhibition of calcium flux, a reduction in release
of acetylcholine from the motor nerve terminal into the
synaptic cleft of the neuromuscular junction, and muscular
weakness.
There are two broad groups of patients with LEMS:
those with an underlying malignancy and those without, but
both share the same clinical and electrophysiologic
characteristics.

Malignancies associated with LEMS


• Small cell carcinoma of the lung (SCLC).
• Lymphoproliferative disorders.
• Carcinoma of the breast, colon, stomach, gall bladder,
kidney, ovary, and bladder.
• Adenocarcinoma of the lung, pancreas and prostate.
• Intrathoracic carcinoid.

Many patients with LEMS (60%) have an associated


SCLC. The SCLC tumor cells express VGCCs of L, N, and
P/Q subtypes, and appear to provide the antigenic stimulus
Lambert–Eaton Myasthenic Syndrome (LEMS) 665

for antibody production which cross react with VGCCs in CLINICAL FEATURES
the nervous system. The prevalence of LEMS in patients Onset of symptoms
with SCLC is 3%. In patients presenting with LEMS the Usually gradual and insidious, occasionally subacute.
chance of having an underlying SCLC falls sharply after 2
years and becomes very small after 4–5 years. There is a Cardinal clinical features
significant association of LEMS with HLA-B8, which is Limbs and trunk
stronger in the group without carcinoma. • Weakness and fatiguability of upper and lower limb
Both LEMS groups (with and without malignancy) also muscles (proximal>distal) occurs, with muscle pain and
show an association with immunologic disorders, as stiffness. A temporary increase in strength can return
suggested by the presence of autoimmune diseases in about after voluntary exercise (unlike myasthenia gravis [MG]),
25% of patients, organ-specific antibodies in about 40% of but the weakness can be exacerbated by prolonged
patients, and also non-organ-specific antibodies. The exercise, hot bath or hot weather.
prevalence of autoantibodies is higher in the group with no • Respiratory muscle weakness (spontaneous or induced
underlying carcinoma. by anesthesia).
• Depressed or absent deep tendon reflexes.
Immunologic disorders associated with LEMS • Post-tetanic potentiation of tendon reflexes (i.e. after
• Addison’s disease. sustained contraction of the appropriate muscle for
• Celiac disease. 10–15 seconds).
• Diabetes mellitus of juvenile-onset. • Peripheral paresthesiae may occur.
• Pernicious anemia. • Poor response of weakness to edrophonium and
• Psoriasis. neostigmine (unlike myasthenia gravis).
• Rheumatoid arthritis. • Marked sensitivity to curare (as in MG).
• Scleroderma.
• Sjögren’s syndrome. Cranial nerves (70% of patients)
• SLE. • Symptoms: often mild and transient, such as eyelid
• Thyroiditis. drooping, diplopia, slurred speech, dysphagia, difficulty
• Vitiligo. chewing, weaker voice, head lolling.
• Signs: rare except for ptosis (54% of patients [805, 806])
and neck flexion weakness (34% of patients); other rare signs
include jaw weakness, facial weakness, and palatal weakness.

806

805

805, 806 Bilateral ptosis, proximal limb wasting and weakness, and
gynecomastia in a patient with Lambert–Eaton myasthenic syndrome
associated with small cell carcinoma of the lung.
666 Neuromuscular Junction Disorders

Autonomic features (sympathetic and parasympathetic, Edrophonium test


80% of patients) May be positive, but the response is usually weaker than in
• Dry mouth. MG.
• Impotence.
• Constipation. Screen for an underlying malignancy and
• Poor bladder and bowel control. immunologic disorder
• Impaired sweating. • Full blood count.
• Tonic pupils. • ESR.
• Orthostatic hypotension/lightheadedness. • Urea and electrolytes.
• Impaired esophageal and intestinal motility. • Plasma glucose.
• Liver function tests.
N.B. Severe autonomic dysfunction may be found on • Thyroid function tests.
testing even when symptoms are minimal. • Autoantibody screen.
• Vitamin B12.
Features of underlying malignancy • Chest x-ray.
SCLC, lymphoma, and other cancers listed above. • Sputum cytology.
• Urinalysis.
DIFFERENTIAL DIAGNOSIS • Stool occult blood.
• MG.
• Myopathy. DIAGNOSIS
• Chronic fatigue syndrome. Diagnosis is based on the clinical findings and results of
investigations.
INVESTIGATIONS
Antibodies to VGCCs TREATMENT
• Antibodies to P/Q type VGCCs are present in >90% of The treatment strategy depends on the severity of the
patients, particularly those with carcinoma-associated symptoms, the degree of response to symptomatic treatment,
LEMS. and the presence or absence of an associated malignancy.
• Antibodies to N type VGCCs are present in <50% of
patients, but are more common in LEMS associated with Treatment of muscle weakness and
primary lung cancer. autonomic symptoms
3,4-diaminopyridine
Electrodiagnosis • Blocks voltage-gated potassium channels which leads to
Electrophysiologic tests are useful for diagnosis as well as prolongation of the action potential at motor nerve
for monitoring the course of the illness: terminals and the open time of the VGCCs, resulting in
• Amplitude of compound muscle action potential increased influx of calcium enhancing quantal
(CMAP) to a single supramaximal stimulus: decreased neurotransmitter release.
(normal or near normal in MG). • It improves muscle strength and autonomic disturbances
• Post activation potentiation: marked increase in CMAP without serious adverse effects.
amplitude by >100% immediately after maximal • The optimal dose varies from 5 mg tds to 25 mg qid.
voluntary contraction (an increase in CMAP may be seen • Onset of beneficial effect occurs within 20 minutes of
in MG, and when present is less marked than in LEMS). oral dose.
• Post activation exhaustion: a decrease in the CMAP • The beneficial effect lasts about 4 hours.
amplitude 2–4 min after maximal voluntary muscle • The maximum response occurs 3–4 days of treatment,
contraction (as also occurs in MG). due to cumulative effect of the drug.
• Repetitive nerve stimulation at slow rates (2–5 Hz): • Adverse effects include peri-oral paresthesia and seizures
decremental pattern (MG also has a decremental pattern (rarely).
in which >10% decrement is considered to be abnormal).
• Repetitive nerve stimulation at fast rates (30–50 Hz): 3,4-diaminopyridine plus anticholinesterase
incremental pattern of over 2–20 times (MG may show drugs (e.g. neostigmine)
an incremental pattern, but it is usually less marked than Anticholinesterases, such as neostigmine, produce mild or
in LEMS). no improvement alone, but seem to potentiate the effects
• Single fiber electromyography (SFEMG): increased jitter of 3,4-diaminopyridine.
and intermittent impulse blocking, which improve with
higher firing rates (MG shows increased jitter and Guanidine and pyridostigmine
impulse blocking which get worse with higher firing Guanidine effectively reduces symptoms but it may have
rates). serious adverse effects which include bone marrow toxicity,
• In vitro microelectrode studies: nephrotoxicity, hepatotoxicity, dermatitis and atrial fibrillation.
– Miniature end-plate potential (MEPP) amplitudes:
normal (small or undetectable in MG).
– End-plate potential (EPP) quantal content: low (normal
in MG).
– Distribution of end-plate potential amplitudes: Poisson’s
distribution (normal distribution in MG).
Further Reading 667

Treatment of severe weakness Treatment of any underlying malignancy


Plasma exchange • The specific treatment of the underlying tumor usually
Repeated plasma exchanges may improve symptoms in both results in improvement or remission of symptoms, and
groups of LEMS, having a peak effect at 2 weeks and the only further treatment required may be continuation
subsiding by 6 weeks. Protein A immunoadsorption of 3,4-diaminopyridine.
removes IgG from plasma selectively. • If specific treatment for the tumor fails to resolve
symptoms, further treatment with prednisolone should
Intravenous immunoglobulin (IVIG) be considered, or, if severely weak, plasma exchange or
Given at 1 g/kg body weight/day for 2 days, IVIG IVIG.
significantly increases muscle strength compared with
placebo, peaking at 2–4 weeks and lasting up to 8 weeks, in No response to treatment, and screening for
patients with LEMS and no carcinoma. The clinical malignancy
response is associated with a significant fall in antibodies to Steroids or steroids and immunosuppressants
VGCCs. There are no data on IVIG therapy in LEMS • Long term prednisolone may be beneficial, and if
associated with carcinoma. remission is achieved, the dose can be tapered to the
minimum maintenance dose.
• Prednisolone combined with azathioprine may be more
effective than prednisolone alone in LEMS not associated
with carcinoma (but no randomized trial evidence has
been obtained).

FURTHER READING Gronseth GS, Barohn RJ (2000) Practice parame- LAMBERT–EATON MYASTHENIC
ter: thymectomy for autoimmune myasthenia SYNDROME
gravis (an evidence-base review). Report of the General
Quality Standards Subcommittee of the Amer- Seneviratne U, de Silva R (1999) Lambert-Eaton
GENERAL ican Academy of Neurology. Neurology, 55: myasthenic syndrome. Postgrad. Med. J., 75:
Vincent A, Palace J, Hilton-Jones D (2001) Myas- 7–15.Kissel JT, Franklin GM, and the Quality 516–520.
thenia gravis. Lancet, 357: 2122–2128. Standards Subcommittee of the American
Academy of Neurology (2000) Treatment of Original descriptions
myasthenia gravis. A call to arms. Neurology, Anderson HJ, Churchill-Davidson HC, Rochard-
MYASTHENIA GRAVIS
55: 3–4. son AT (1953) Bronchial neoplasm with myas-
Epidemiology
Pallace J, Newsom-Davis J, Lecky B, and the thenia: prolonged apnoea after administration
Robertson NP, Deans J, Compston DAS (1998)
Myasthenia Study Group (1998) A ran- of succinlycholine. Lancet, 2: 1291–1293.
Myasthenia gravis: a population based epi-
domised, double-blind trial of prednisolone Eaton LM, Lambert EH (1957) Electromyogra-
demiological study in Cambridgeshire, Eng-
alone or with azathioprine in myasthenia phy and electrical stimulation of nerves in dis-
land. J. Neurol. Neurosurg. Psychiatry, 65:
gravis. Neurology, 50: 1778–1783. eases of the motor unit: observations on a
492–496.
Task Force of the Medical Scientific Advisory myasthenic syndrome associated with malig-
Clinical subtypes Board of the Myasthenia Gravis Foundation of nant tumours. JAMA, 163: 117–124.
Aarli JA (1999) Late-onset myasthenia gravis. America (2000) Myasthenia gravis: recom- Lambert EH, Eaton LM, Rooke ED (1956) De-
Arch. Neurol., 56: 25–27. mendations for clinical research standards. fect of neuromuscular conduction associated
Neurology, 55: 16–23. with malignant neoplasms. Am. J. Physiol.,
Investigations Tindall RSA, Rollins JA, Phillips TJ, et al. (1987) 187: 612–613.
Hoch W, McConville J, Melms A, et al. (2001) Preliminary results of a double-blind, ran-
Autoantibodies to the receptor tyrosine kinase Treatment
domised, placebo-controlled tiral of cy-
MuSK in patients with myasthenia gravis with- Sanders DB, Massey JM, Sanders LL, Edwards LJ
closporine in myasthenia gravis. N. Engl. J.
out acetylcholine receptor antibodies. Nat. (2000) A randomised trial of 3,4-diaminopy-
Med., 316: 719–724.
Med., 7: 365–368. ridine in Lambert–Eaton myasthenic syn-
Clinical research drome. Neurology, 54: 603–607.
Treatment Task Force of the Medical Scientific Advisory
Ciafaloni E, Massey JM, Tucker-Lipscomb B Board of the Myasthenia Gravis Foundation of
(2001) Mycophenolate mofetil for myasthenia America (2000) Myasthenia gravis: recom-
gravis: An open-label pilot study. Neurology, mendations for clinical research standards.
56: 97–99. Neurology, 55: 16–23.
Evoli A, Batocchi AP, Minisci C, et al. (2001)
Therapeutic options in ocular myasthenia
gravis. Neuromuscular Disorders, 11:
208–216.
668 Chapter Twenty-four

Muscle Disorders
X-LINKED DYSTROPHINOPATHIES Becker’s muscular dystrophy (BMD)
A milder allelic form in which some muscle fibers express
dystrophin. First recognized by Becker in 1955 as a distinct
DEFINITION benign form of X-linked myopathy.
Recessive disorders of muscle (Duchenne’s and Becker’s
muscular dystrophy) caused by a mutation in the short arm, EPIDEMIOLOGY (see Table 48)
locus 21, of the X chromosome (Xp21), in the enormous • Incidence:
gene that codes for the protein dystrophin. – DMD: 1 per 3500 male births.
Dystrophin is a filamentous protein present in striated – BMD: one-fifth to one-tenth that of DMD (i.e. about 1
and cardiac muscle and other tissues which is expressed at per 35 000 births).
the periphery of the muscle fiber in the sarcolemmal • Prevalence:
membrane. Its function is to support the muscle membrane – DMD: 2.5 per 100 000 population (relatively low, com-
during muscle contraction and prevent destruction of pared with incidence, because the disease is usually fatal
muscle fibers in response to shearing forces. before the third decade).
– BMD: higher than that of DMD because of its relatively
CLASSIFICATION benign natural history.
Duchenne’s muscular dystrophy (DMD) • Age:
The most severe dystrophinopathy, in which practically no – DMD: onset in early childhood (muscle necrosis and
dystrophin is detected in skeletal muscle by immuno- serum enzyme elevation can be found in neonates).
cytochemistry. – BMD: onset in teenage years or early 20s.
• Gender:
– Almost all patients are male because the inheritance is as
an X-linked recessive trait.
807 – Girls are affected very rarely due to autosomal
translocation or if there is only one X chromosome (e.g.
Turner’s syndrome).

807–809 Normal muscle. H&E stain, x30 magnification (807). Note


the arrangement of the muscle fibers in fascicles, the close
interdigitation of the muscle fibers, and the muscle fiber nuclei are
nearly all subsarcolemmal. NADH-tr. x100 (808): a mosaic
arrangement of fibers of various staining intensities.Type 1 fibers are
darkly stained and Type 2 fibers are paler, but this distinction is clearer
in myosin ATPase preparation.Type 2A fibers tend to be darker than
type 2B fibers.The intermyofibrillar substance has a finely granular
appearance. Electron microscopy (809, bar = 1 ␮m).

808 809
X-linked Dystrophinopathies 669

810
Table 48 Epidemiology of X-linked dystrophinopathies
DMD BMD
Incidence 1/3500 1/35 000
Onset 2–7 years 3 years–adult
Creatine kinase 50x normal 10x normal
Wheelchair-bound By 12 years of age After 12 years of age
Respiratory failure By 20 years of age After 20 years of age
Dystrophin Absent Reduced (patchy
expression)

PATHOLOGY
Light microscopy (807–811)
• Abnormal variations in fiber size.
• Fiber splitting. 811
• Central nuclei.
• Replacement by fat and fibrous tissues.

Dystrophin immunostaining (812–814)


• DMD: <3% of fibers have dystrophin (813).
• BMD: severe (i.e. in a wheelchair by 15–20 years of age):
3–15% dystrophin; mild (i.e. ambulatory >20 years of
age): >20% dystrophin; partial staining of circumference
of muscle fibers (814).

810 Duchenne muscular dystrophy. Muscle biopsy showing variability of


fiber size, rounded fibers, and distortion of the muscle structure by
extensive deposition of fibrous and fatty tissue between the muscle fibers.

811 Duchenne muscular dystrophy. An area of regenerative activity in 812


muscle: regenerating fibers are usually smaller than surrounding fibers
and contain central, enlarged, vesicular nuclei, with prominent nucleoli.

812–814 Cryostatic sections of human muscle biopsies stained with


fluorescent labelled monoclonal antibodies to dystrophin, a cytoskeletal
protein located at the periphery of muscle fibers beneath the
sarcolemma from: normal muscle (812), showing characteristic even
staining of the sarcolemma; muscle from an individual with Duchenne’s
muscular dystrophy (813) showing virtually no staining of dystrophin
(<3%); muscle from an individual with Becker’s muscular dystrophy
(814), showing only dull green background staining which is reduced in
intensity and varies both between and within fibers.

813 814
670 Muscle Disorders

ETIOLOGY AND PATHOPHYSIOLOGY Year 10


• The dystrophinopathies involve X-linked inheritance. Unable to walk or stand and are dependent on a wheelchair.
• A mutation in the short arm, locus 21, of the X chromo-
some (Xp21), in the gene that codes for dystrophin. Mid teens
• Null mutations result in no protein production and a Loss of upper limb function occurs.
severe progressive phenotype (DMD), whereas missense
mutations result in phenotypes of intermediate severity Late teens–early twenties
(BMD). The dystrophin gene is the largest gene • Usually fatal.
associated with a disease that has been identified (2.4 • Death is usually related to pulmonary infections,
million base pairs). It consists of at least 85 exons with respiratory failure, gastrointestinal complications or
introns making up 98% of the gene. cardiomyopathy.
• In about one-third of patients, DMD is the result of a
new mutation; one-third of all patients have a family Carriers
history consistent with X-linked recessive inheritance, and • About 8% of female carriers have limb-girdle myopathies.
one-third are born to unwitting carriers. • A small number of carriers may also suffer from isolated
• The mutation rate in the gene is unusually high cardiomyopathy.
(7–10 × 10-5/gene/generation), probably because of its
large size. BMD
• Dystrophin links the cytoskeleton to the sarcolemma. • Onset is usually after 12 years of age (3–20 years of age).
Absence of dystrophin results in loss of dystrophin- • Patients show similar clinical features to DMD but
associated glycoprotein and disruption of the linkage milder, and myocardial and intellectual involvement is
between the cytoskeleton and the extracellular matrix, less common.
leading to sarcolemmal instability and muscle cell necrosis. • Calf pain during exercise and myoglobinuria are common.
• Patients may live up to many decades with mild to
CLINICAL FEATURES moderate symptoms that can be indistinguishable from
DMD those of limb-girdle dystrophies.
• Progressive symmetric proximal muscle weakness of the
upper and lower limb girdles with hypertrophy of calf DIFFERENTIAL DIAGNOSIS
muscles. • Congenital myopathies of the nemaline and central core
• Cardiomyopathy is always present. types.
• Non-progressive intellectual impairment occurs in at least • Limb-girdle muscular dystrophies (see p.674).
10% and up to one-third of patients and is present from • Spinal muscular atrophy (see p.534):
childhood; special schooling is required for some patients. • X-linked and autosomal dominant forms of muscular
• Smooth muscle can be affected, causing paralytic ileus, dystrophy, e.g. Emery–Dreifuss muscular dystrophy:
gastric dilatation and atony of the bladder wall. – The defective gene for the commoner X-linked
Emery–Dreifuss muscular dystrophy (EDMD), STA at
Year 2 Xq28, when normal, codes for a 34 kD nuclear
• Motor developmental delay is noticeable after the first membrane protein named ‘emerin’, which is expressed
year. in skeletal muscle and myocardium but is still of
• Onset of walking may be delayed beyond 18 months of unknown function. The gene, LMNA at 1q21, for the
age. autosomal dominant form of EDMD encodes other
nuclear membrane proteins, laminins A and C.
Year 5 – Onset in early childhood of progressive humeroperoneal
• Difficulty running and climbing stairs, have frequent falls. muscle weakness and wasting, and contractures of elbow
• Show tip-toe walking with a waddling gait. flexors, Achilles tendons and paraspinal muscles. Cardiac
• Gower’s sign: in order to stand up from the floor, involvement is common with conduction defects, brady-
children employ their hands to ‘climb up their legs’ cardias, and heart blocks. Appropriately timed insertion
(815, 816). of a pacemaker may prevent a fatal conduction defect.
• They have weakness of lower limbs, pelvic muscles and – The diagnosis of EDMD depends on mutation analysis
lower trunk. rather than protein immunohistochemistry (at present).
• Hyperlordosis with a prominent abdomen.
• Enlarged but weak calf muscles (pseudohypertrophy), INVESTIGATIONS
due to excess fat and connective tissue plus large but Blood
ineffective muscle fibers. • Serum creatine kinase: markedly elevated up to the
thousands or tens of thousands of units in DMD, and
Years 6–9 also elevated in BMD and female carriers.
• Joint contractures of the iliotibial bands, hip flexors, and • Molecular genetics: DNA analysis in blood leukocytes
heel cords are present. using the polymerase chain reaction (PCR) is abnormal
• Thoracic deformity. in more than two-thirds of DMD patients.
• Electrophoresis (western blotting) biochemically
demonstrates the deficiency of a particular protein, such
as dystrophin.
X-linked Dystrophinopathies 671

Urine DIAGNOSIS
Myoglobinuria after anesthetic exposure (DMD). DMD
The diagnosis can be confirmed by muscle biopsy showing
EKG absence or near absence of dystrophin on immunostaining
Features of a cardiomyopathy may be present (also in female (813). DNA analysis in blood leukocytes using PCR is
carriers). abnormal in more than two-thirds of DMD patients.
Prenatal diagnosis from chorionic villi is also feasible.
EMG
Myopathic features occur early in the course. Later, the PREVENTION
number of motor units that are activated decreases and Prenatal diagnosis and carrier detection
tissue may even become unexcitable. Conventional strategies of detecting female carriers by pedigree
analysis, clinical assessment (some mild muscle weakness),
Muscle biopsy serum creatine kinase (CK) determination (elevated in about
Obtain from mildly involved muscles rectus abdominus 70% of cases), EMG, and histologic study of muscle biopsy
rather than from gastrocnemius or deltoid muscles because specimens, are now complemented by dystrophin analysis in
of worsening caused by immobilization. muscle biopsy, PCR analysis of heterozygotes, and restriction
fragment length polymorphism analysis.
Light microscopy (810, 811) Fetal abnormalities can be detected using molecular
• Abnormal variations in fiber size. techniques in samples of chorionic villi during early pregnancy
• Fiber splitting. (after 8 weeks) or by amniocentesis. Muscle biopsy from fetus
• Central nuclei. or dystrophin immunostaining can be obtained after 19 weeks
• Replacement by fat and fibrous tissues. of gestation with ultrasound guidance.
DNA analysis by molecular genetic studies is much more
Dystrophin immunohistochemistry staining accurate and thus now essential for detecting female carriers
• Staining uses labelled monoclonal antibodies to of DMD and BMD, as well as for prenatal diagnostic
dystrophin, a cytoskeletal protein located at the periphery analysis of chorion villus biopsy material.
of muscle fibers and beneath the sarcolemma.
• In DMD, the majority of fibers fail to show any staining Genetic counselling
(813). Genetic counselling is important for all families with an
• In BMD the intensity of staining is reduced and varies affected male. A known carrier has one chance in two of
both between and within fibers, as it does in the rare giving birth to a DMD boy or to a carrier female. About
female carriers of DMD who manifest symptoms of the one-third of patients, however, do not have family histories.
disease (814). It is often necessary to know the specific gene mutation
• Staining differentiates benign from malignant myopathies. occurring in a family, which is usually accomplished by
initially studying an affected male in the family. However, a
Role major problem in genetic counselling in DMD is that some
With the advent of molecular diagnosis and in cases with mothers transmitted a mutation to more than one offspring
documented family histories, muscle biopsy might not be
required as much as in the past to establish the diagnosis.
In less-documented cases, muscle biopsy can be very useful
in distinguishing DMD, which is a severe disease, from 816
other diseases that present with similar clinical features but
which have a more favorable prognosis (e.g. congenital
myopathies of the nemaline and central core types).

815

815, 816 A boy with Duchenne muscular dystrophy rising from the
floor, and climbing up his legs (Gowers’ sign). (Courtesy of Professor
BA Kakulas, Neuropathology Department, Royal Perth Hospital,
Australia.)
672 Muscle Disorders

which they did not have in their somatic cells (in their Other strategies for treatment (currently
peripheral blood leukocytes). This has been attributed to being researched)
germline mosaicism. This means that prenatal testing may • Elucidate the function of the dystrophin–glycoprotein
have to be considered in subsequent pregnancies if a mother complex.
has had an affected son, because it cannot be guaranteed • Restore the link between subsarcolemmal cytoskeleton
that his dystrophy is the result of a new mutation and is and the extracellular matrix by:
therefore unlikely to recur. – Replacing the missing protein (dystrophin):
Myoblast transfer: not successful to date.
TREATMENT Gene therapy: difficult.
A multidisciplinary team approach to ongoing management – Up-regulating compensating proteins (utrophin).
(medical, genetic, physiotherapy, occupational therapy, – Up-regulating proteins with missense mutations
nursing, social work) is required. (sarcoglycans).
• Gene therapy is in the experimental preclinical stage.
Family and patient education
Physical, emotional, educational and financial implications PROGNOSIS
should be discussed. DMD
Most boys start using a wheelchairs by age 12 years and die
Physiotherapy in their 20s.
• Physiotherapy aims to preserve mobility and prevent
early contractures, which are common in flexor muscles BMD
of hips, knees and ankles. The natural history is much less predictably than that of
• Passive range of motion exercises and appropriate DMD but inability to walk occurs later than with DMD.
orthotics may prolong ambulation. Patients become confined to a wheelchair at 12–40 years or
• Exercises to maintain strength in functional muscles and later, and many individuals marry, have children, and survive
prevent obesity (which further impairs ambulation and well into middle age and beyond. Death usually occurs at
respiratory function) are also helpful. 30–60 years of age.
• Patients who can no longer walk may benefit from splints
(at night, kept in a neutral position), braces, crutches,
and surgery (e.g. scoliosis surgery), and should try to
stand at least 3 hours/day in divided periods. However,
excessive exercise can be detrimental. FACIO-SCAPULO-HUMERAL
MUSCULAR DYSTROPHY (FSHD)
Respiratory therapy
• Breathing exercises or playing wind instruments may be
beneficial. DEFINITION
• In the late stages, intermittent positive pressure ventilation An autosomal dominant inherited disorder characterized by
is useful, particularly when CO2 is being retained. weakness of the facial, scapulohumeral, anterior tibial and pelvic
girdle muscles; sometimes associated with retinal vascular
Medication disease, sensory hearing loss and, in severe cases, even abnor-
Prednisolone 0.75 mg/kg/day by oral administration may malities of the CNS due to a genetic defect on chromosome 4.
be appropriate in special circumstances such as acute
deterioration. Neuromuscular strength may improve after EPIDEMIOLOGY
1 month of treatment and the maximum effect is reached • Prevalence: 1:20 000; the third most common hereditary
by 3 months. In controlled studies, the benefits lasted for disease of muscle (after Duchenne and myotonic
3 years. Adverse effects include insomnia, difficult behavior dystrophy).
and gastrointestinal symptoms. The mechanism by which • Age of onset: any age, but usually 10–50 years of age, and
steroids may stabilize the clinical course of DMD may be mostly in the second and third decades. A large proportion
that prednisone slows muscle destruction. of gene carriers remain asymptomatic for decades.
Oxandrolone, an anabolic steroid, may be useful before • Gender: M=F.
initiating corticosteroid therapy because it is safe, accelerates
linear growth, and may slow the progress of weakness. PATHOLOGY
Avoid anesthetics with halothane and succinylcholine Dystrophic muscle
because patients may develop episodes that resemble • Disruption of muscle architecture.
malignant hyperthermia. Adverse effects may be reduced by • Abnormal variation in fiber size.
using non-depolarizing muscle relaxants. • Degeneration and regeneration of muscle fibers.
• Replacement of muscle by fat and fibrous tissue.
Gene therapy
• Gene therapy is experimental. ETIOLOGY AND PATHOPHYSIOLOGY
• Myoblast transplant therapy has succeeded in the mdx Autosomal dominant inheritance with high penetrance: the
mouse model but has failed in patients to date. gene is located to the distal chromosome 4q35 where there
• Gene therapy administered through viral vectors or are normally between 10 and 100 almost identical tandemly
liposomes is being tested in animal models. arranged repeating units each of 3.3 kb, making up an area
that is greater than 35 kb in length. In patients with FSHD,
a number of these repeats are deleted, due to large deletions
Facio-scapulo-humeral Muscular Dystrophy (FSHD) 673

of variable size, so that the area is <35 kb in length (as • FSHD also involves the trapezii, rhomboids, and serratus
measured on an electrophoretic gel). No gene has been anterior scapular fixators. Scapular winging is maximized
identified within the area of this deletion, suggesting that with forward movement because of the serratus anterior
these deletions execute a position effect on a more weakness (see p.616). The pectoralis is very atrophic.
centromeric located gene. In other words, the deletions do Deltoid and rotator cuff musculature are better preserved.
not appear to disrupt a transcribed gene but are thought to • Later in the disease, weakness involves the lower limbs,
interfere with the expression of a gene or genes located particularly the anterolateral leg compartment (foot
proximal to the deletions. dorsiflexors) and finally the hip girdles. Truncal weakness
may also occur.
CLINICAL FEATURES • Side-to-side asymmetry of muscle wasting and weakness
• Weakness of the facial, scapulohumeral, anterior tibial, is frequent and often striking.
and pelvic girdle muscles occurs (817, 818). • Heart muscle is spared usually but a predilection for atrial
• The spectrum of clinical severity is wide, even within tachyarrhythmias exists.
families: some have only minimal facial involvement and • An exudative retinopathy is present in one-third of cases.
normal life expectancy, and others are wheelchair bound • Retinal vascular tortuosities are usually subclinical in
in childhood with marked kyphoscoliosis and ventilatory most cases.
failure. • High frequency sensory hearing loss can be present but
• Although weakness is present in over 95% of affected is usually asymptomatic.
individuals by age 20 years, up to one-third of patients • Abnormalities of the CNS are present in severe cases.
have no symptoms (i.e. do not recognize the weakness),
and most present to medical attention because of DIFFERENTIAL DIAGNOSIS
involvement of the shoulder rather than the facial muscles. • Autosomal dominant scapuloperoneal syndromes:
• However, the first sign is weakness of certain facial different pattern of involvement and progression.
muscles, including the orbicularis oculi, orbicularis oris, • Neurogenic disorders.
and zygomaticus, but the actual onset of the weakness can • Nemaline myopathy.
be difficult to establish because abnormal features may be • Desmin storage.
interpreted as nothing more than family traits. Bell’s • Mitochondrial myopathy.
phenomenon and drooping of the lower lip are present, • Polymyositis.
and patients may be unable to whistle. The masseter, • Centronuclear myopathy.
temporalis, and extraocular muscles are not involved, and
the bulbar and respiratory muscles are also spared.

817 818

817, 818 Wasting and weakness of the muscles of the face, upper arms and shoulders with prominent weakness of the pectoral muscles and
winging of the scapulae in a patient with facioscapulohumeral muscular dystrophy.
674 Muscle Disorders

INVESTIGATIONS LIMB-GIRDLE MUSCULAR


• Serum CK: may be raised 2–7 times normal, but this DYSTROPHIES (LGMD)
varies by age and gender (elevated in three-quarters of
affected males and two-fifths of affected females).
• EMG: myopathic. DEFINITION
• Muscle biopsy: may reveal minimal myopathic changes A clinically and genetically heterogeneous group of
or severe dystrophic muscle, depending in part on the autosomal-dominant and autosomal-recessive inherited
choice of muscle biopsied. A significant inflammatory muscle disorders.
component may be seen.
• Blood: leukocyte DNA analysis: affected individuals have EPIDEMIOLOGY
small restriction fragments of 10–35 kb containing the • Uncommon.
deletion, whereas unaffected individuals have restriction • Age: onset at any age but predominantly between first
fragments of 50–300 kb. and fourth decades.
• Gender: M=F.
DIAGNOSIS
• Patients with characteristic facial and shoulder weakness, ETIOLOGY
and an autosomal dominant family history almost always • A small minority (<10%) of cases are inherited as an
have the same molecular defect. autosomal dominant trait (type 1) and are relatively mild.
• The 4q35 deletion that causes FSHD can be identified • The majority are inherited as autosomal recessive traits
in 86–95% of cases with the disease (sensitivity), and is (type 2) , which is often more severe and resembles DMD.
>98% specific for the disease. • At least three dominant subtypes and eight recessive
• The 5–14% of affected individuals in whom a deletion cannot subtypes have been identified.
be identified is attributed to: (1) technical difficulties with • One recessive subtype (LGMD 2A) is caused by a
degraded or partially digested DNA; (2) translocation deficiency of a muscle-specific protease (calpain 3).
between 4q35 and homologous regions on 10q complicating • Four other recessive subtypes (LGMD 2C, D, E and F)
the interpretation of the Southern blot data and, rarely, (3) are caused by deficiencies of particular sarcoglycans
FSHD not linked to 4q (locus heterogeneity). In such cases, (dystrophin associated glycoproteins) which form part of
if the clinical suspicion of FSHD remains high, further testing the dystrophin-associated protein complex of muscle
with other 4q35 and 10q probes as well as more extensive membrane.
study of the kindred with linkage analysis may be required.
The dystrophin-associated complex
PRESYMPTOMATIC AND PRENATAL DIAGNOSIS • A large oligomeric complex in the sarcolemmal
Restriction enzyme DNA fragments associated with the membrane of skeletal muscle, which provides an
gene have been found to be greater than 35 kb in length in important structural link between the actin-cytoskeleton
individuals who do not have muscular dystrophy; in affected and the extracellular matrix in skeletal muscle (819).
individuals fragments are always less than this (as measured • The complex consists of several subcomplexes beneath
by electrophoretic gel). the extracellular matrix (laminin-2, α-dystroglycan,
Furthermore, there is a general tendency for shorter glycoprotein complex [sarcoglycans and β-dystroglycan],
fragments (i.e. larger deletions) to be associated with earlier dystrophin and F-actin cytoskeleton).
onset and more severe cases of the disease; in a sporadic case,
for example, this can give an idea of the likely prognosis. Laminin-2 (merosin)
• Laminin-2 is located in the extracellular matrix at the
TREATMENT periphery of muscle fibers.
Myopathy • Absent in children with congenital muscular dystrophy
No specific treatment for the weakness. (onset in newborn) who have a nonsense mutation (no
functioning protein produced).
Physiotherapy • Deficient in children and even adults who have a
Posterior plastic ankle orthotics can correct foot drop. A trunk missense mutation in the merosin gene and produce
brace may help patients with prominent lumbar lordosis and merosin but in reduced quantities.
abdomen. A motorized wheelchair with adjustable height • It is also expressed around blood vessels in the brain.
control is invaluable for non-ambulatory patients. Hence MRI T2W images show a leukodystrophic
pattern, but the intellect is usually normal.
Exudative retinopathy
Photocoagulation may be necessary to prevent retinal
detachment.

PROGNOSIS
• Many patients are only mildly affected; most affected
individuals remain able to work, often adapting remark-
ably well to profound weakness in distinct muscle groups.
• The rate of disease progression is variable but usually very
slow.
• It is severely disabling for >15% who eventually become
dependent on wheelchairs.
Limb-girdle Muscular Dystrophies (LGMD) 675

α-Dystroglycan
• Is a 97 kDa precursor which is encoded by a single gene, Table 49 Classification of limb-girdle muscular
mapped to chromosome 3p21, and has a novel laminin dystrophies
(merosin) receptor, which connects the sarcolemma to Disorder Gene location Gene product
the extracellular matrix.
• It is located outside the cell, and binds the extracellular Autosomal dominant
matrix component laminin-2 with high affinity. LGMD 1A 5q22-q34 (Genetic anticipation, no cardiac
• It is found in muscle and also the neuromuscular involvement)
junction, peripheral nerve, brain, kidney and embryo. 1B 1q11-21 Caveolin-3 (cardiac arrhythmias
may occur early)
Sarcoglycans: α (adhalin), β, γ 1C 3p25 Caveolin-3 (a scaffolding protein)
• These are abnormal in some autosomal recessive forms 1D 6q23
of limb girdle muscular dystrophy (sarcoglycanopathies). 1E 5q31
• The spectrum of disease (sarcoglycanopathies) is similar Autosomal recessive
to DMD and BMD, and determined by the nature of the LGMD 2A 15q15.1-q21.1 Calpain-3
mutations (nonsense vs. missense). 2B 2p13 Dysferlin (cf. Miyoshi myopathy)
• Girls present like DMD or BMD, with gradual onset of 2C 13q12 γ-sarcoglycan
progressive proximal limb weakness (e.g. difficulty 2D 17q12-q21.33 α-sarcoglycan (adhalin)
ascending stairs and running), lumbar lordosis, 2E 4q12 β-sarcoglycan
prominent calf muscles, and tight tendoachilles. 2F 5q33-q34 δ-sarcoglycan
• Cardiomyopathy may occur. 2G 17q11-q12 ?
• CK is high. 2H 9q31-q34.1 ?
• Muscle biopsy dystrophic, but a normal amount of
dystrophin is present. Staining for antibodies to
sarcoglycans reveals a deficiency of one or more of the
sarcoglycans.

Dysferlin
Basal lamina/extracellular matrix
819
• Is localized in muscle membrane but its role is unknown
at present.
• Dysferlinopathies have their onset in late teens or
adulthood.
• Weakness begins in the lower limbs only: distal Laminin-2 Sarcolemma/plasma
(merosin) membrane
(gastrocnemius) weakness (plantar flexion: difficulty Sarcoglycans
standing on toes) and painful calves. α-dystroglycan
• Weakness progresses slowly and may involve the biceps α
(only) in the upper limbs. ε γ
• CK is elevated (1000s). δ β
• Muscle biopsy is dystrophic (necrotizing myopathy).
β-dystroglycan
• Loss of ambulation occurs at 30–40 years of age.
Syntrophins
β-Dystroglycan (inside the cell) Dystrophin
This binds to the cysteine-rich domain of dystrophin. Dystrobrevin

Dystrophin
-COOH
Dystrophin links the cytoskeleton to the sarcolemma.
F-actin
CLASSIFICATION (see Table 49) Intracellular
NH2-

819 Diagrammatic representation of the dystrophin-associated


complex in the muscle fiber membrane. Specific muscular dystrophies
are caused by deficiencies of dystrophin (Duchenne and Becker
muscular dystrophy), a particular sarcoglycan (limb-girdle muscular
dystrophy), or laminin-2 (merosin) (congenital muscular dystrophy).
676 Muscle Disorders

PATHOLOGY Autosomal recessive LGMD


No characteristic histologic features are present (820, 821). Sarcoglycanopathy
• Sarcoglycanopathy generally occurs as childhood-onset,
Dystrophic muscle but may arise in adults.
• Dystrophic muscle shows disruption of muscle architecture. • Calf hypertrophy is common (824, 825).
• Variation in fiber size, with very hypertrophied fibers and • Early involvement of the scapular muscle, deltoid and
small fibers. pelvic girdle.
• Prominent internal nuclei especially in hypertrophied fibers. • Serum CK is normal or mildly elevated.
• Fiber splitting in some fibers, especially in severely
involved fibers and late in disease. Calpainopathy
• Degeneration and regeneration of muscle fibers. • Onset occurs in patients aged 8–15 years generally, but
• Increased fibrous tissue. adult-onset is fairly common.
• Atrophic muscles, scapular winging, sparing of hip
Immunohistochemistry adductors, and Achilles tend contractures are present.
Deficiency of a muscle specific protease (calpain-3) or a • Serum CK is 10 times normal values.
particular sarcoglycan can be found.
Dysferlinopathy
PATHOPHYSIOLOGY • Presentation occurs in late teens or early twenties.
Disruption of the dystrophin–glycoprotein complex in • There is posterior distal lower limb involvement early on,
skeletal muscle leads to several forms of limb-girdle (and with inability to stand on the toes.
other forms of) muscular dystrophy. • Serum CK is massively elevated (may be >100 times
normal values).
CLINICAL FEATURES
General DIFFERENTIAL DIAGNOSIS
• Proximal muscle weakness occurs in pelvic and upper • BMD.
girdle muscles, sparing the face (822, 823). • Emery–Dreifuss muscular dystrophy: contractures are a
• Cardiac involvement may occur. very prominent feature.
• Chronic polymyositis.
Autosomal dominant LGMD • Mitochondrial cytopathy.
Dominant family history. • Metabolic myopathy: adult-onset acid maltase deficiency.
• Endocrine and drug-induced myopathies.
LGMD 1A • MG.
• Onset occurs in patients aged 18–35 years. • Spinal muscular atrophy.
• Affected individuals may have dysarthria and Achilles
tendon contractures. INVESTIGATIONS
• Serum CK is normal or mildly elevated. Blood
• Serum creatine kinase: usually elevated. If normal or mild
LGMD 1B elevation in active disease, suspect autosomal dominant
• Onset occurs in patients aged 4–38 years. LGMD. If elevated >100 times normal in the early
• Cardiac conduction defects are a prominent complication. stages, suspect dysferlinopathy.
• Serum CK is normal or mildly elevated. • Molecular genetics: DNA analysis in blood leukocytes
using PCR.
LGMD 1C (caveolin deficiency) • Electrophoresis (western blotting): biochemically
• Onset occurs in patients aged 5 years. demonstrates the deficiency of a particular protein.
• Serum CK is 4–25 times normal values.

820 821

820, 821 Muscle biopsy specimens in a patient with limb-girdle muscular dystrophy showing variation in fiber size, prominent internal nuclei and
degenerative and regenerative changes.
Limb-girdle Muscular Dystrophies (LGMD) 677

• Linkage analysis to dominant LGMD loci if dominant DIAGNOSIS


family history and family structure is suitable. Direct detection of the mutation is the final proof of
diagnosis and allows carrier detection/prenatal diagnosis.
EKG However, given the complexity of the genes and the
Features of a cardiomyopathy may be present. heterogeneity of the mutations, this is best directed by the
results of protein analysis. Therefore, these dystrophies
EMG should be diagnosed using a combination of clinical studies,
Myopathic, no specific features. protein studies and genetic studies, which may only be
carried out in specialized centers.
Muscle biopsy
Light microscopy: dystrophic muscle (820, 821) TREATMENT
• Disruption of muscle architecture. A multidisciplinary team approach (medical, genetic
• Variation in fiber size. counselling, physiotherapy, occupational therapy, speech
• Degeneration and regeneration of muscle fibers. therapy, psychology, social work) is required.
• Increased fibrous tissue.
PROGNOSIS
Immunocytochemistry Variably progressive.
Use at least two sarcoglycan antibodies as well as dystrophin
antibodies:
• Dystrophin: generally normal, but may be abnormal in
sarcoglycanopathy.
• Sarcoglycans: a specific sarcoglycan
may be absent or reduced. Sarco-
glycans are normal in calpainopathy 822 823
and dysferlinopathy.
• Dysferlin.

If abnormalities are detected on


immunocytochemistry with dystrophin
or sarcoglycan antibodies, then the full
range of sarcoglycan antibodies should
be used to attempt to pinpoint the
primary deficiency.

Immunoblotting
Immunoblotting is necessary to examine
calpain-3. Multiplex blotting may also
show which sarcoglycan is primarily
involved.

822, 823 The limb-girdle muscular


dystrophy phenotype: note wasting of the
proximal two-thirds of the deltoids (with
pseudohypertrophy of the distal one-third),
forearm flexors, thigh adductors, and medial
heads of the gastrocnemius muscles.

824 825

824, 825 Calf hypertrophy in a 30 year old


man with a sarcoglycanopathy.
678 Muscle Disorders

MYOTONIC DYSTROPHY Sternocleidomastoid muscles


Wasting and weakness out of proportion with shoulder and
posterior neck muscles.
DEFINITION
A autosomal dominant disorder due to a mutation causing Limb muscles
an expanded CTG trinucleotide repeat in the DMPK gene Predominantly distal muscle wasting (831) and weakness
on chromosome 19. The disease has varied and diverse (particularly the hands), which occurs later. Proximal limb
expression in many body systems, including skeletal muscle, muscles are usually preserved until the late stages.
brain, eye, heart, thyroid, pancreas and gonads, but the Consequently, patients usually remain ambulatory, unlike
cardinal clinical features of myotonia, muscle wasting, typical those with other dystrophies.
facies and cataract are almost diagnostic.
Diaphragm
EPIDEMIOLOGY • Hypoventilation.
The most common form of muscular dystrophy in adults. • Opiates and barbiturates may induce respiratory failure.
• Incidence: 14/100 000 live births. • Depolarizing relaxants may increase myotonia.
• Prevalence: 1 in 20 000.
• Age: any age. Skeletal muscle myotonia
• Gender: M=F. • Delayed muscle relaxation occurs after contraction.
• It is often a sign rather than a symptom: patients seldom
ETIOLOGY AND PATHOPHYSIOLOGY complain of stiffness, but do complain of dysarthria
• Myotonic dystrophy shows an autosomal dominant (myotonia of the tongue as well as facial weakness).
inheritance of an expanded triplet cytosine-thymine- • It may be elicited by percussion of the tongue or thenar
guanine (CTG) repeat in the DMPK (myotonic eminence, or by failing to release the grip after a
dystrophy protein kinase) gene on chromosome 19 handshake.
(19q13.2-13.3). Normal individuals have up to about 38 • True myotonia is diminished by repetitive muscle
repeats; a disease allele may consist of 50 to several contractions.
thousand repeats.
• The length of the trinucleotide repeat correlates directly Malignant hyperpyrexia
with the severity of the disease and inversely with the age
of onset. Eyes
• DMPK is a protein-kinase, a ubiquitous enzyme related to Subcapsular cataracts.
protein phosphorylation. The enzyme is widely distributed
which may explain the generalized multisystem nature of Heart
the disorder. However, the mutation is located in a part of Conduction defects
the DMPK gene that does not code for protein. It remains • Symptomatic primary heart muscle disease is very rare
unclear how a mutation that does not interrupt the and hence routine echocardiography is not indicated
DMPK protein coding sequence has a dominant effect (unlike in BMD/DMD) but cardiac conduction
with such severe consequences. problems (heart block and arrhythmias) are common and
a major cause of morbidity and mortality.
PATHOLOGY • The perioperative period is a particularly dangerous time
Muscle with respect to development of tachyarrhythmias and
• Large numbers of internal nuclei, often occurring in long heart block; the anesthetist must always be made aware
chains, together with sarcoplasmic masses and ring-fibers of the diagnosis of myotonic dystrophy.
(826). • There is a broad correlation between the likelihood of
• Increased variability in fiber size. cardiac disease and both the severity of skeletal muscle
• Increased endomysial and interfascicular fibrous tissue. involvement and the length of the trinucleotide repeat.
• Moth-eaten fibers.
• Individual necrotic and regenerating fibers are rare. Brain
• Type 1 fibers appear smaller than Type 2 fibers. Excessive daytime sleepiness
• The most common non-muscular symptom, being
CLINICAL FEATURES present in 75% of patients.
Many patients have predominant systemic symptoms rather • Patients may sleep most of the day.
than neuromuscular symptoms, and some are never • Sleepiness is broadly correlated with overall disease
diagnosed with myotonic dystrophy. severity.
• It is most frequently due to a disturbance of central
Skeletal muscle wasting and weakness mechanisms, but occasionally is due to nocturnal
Facial muscles hypoventilation, with or without obstructive sleep apnea,
• Hatched and thin face. as a result of respiratory muscle weakness.
• Frontal balding (early).
• Temporalis and masseter atrophy (827–830). Mental apathy, low intellect, and perhaps progressive dementia
• Ptosis (not as severe as in MG or Kearns–Sayre
syndrome) and occasionally diplopia due to extraocular Skull
muscle involvement. • Hyperostosis.
• Dysphagia (oropharyngeal involvement). • Small pituitary fossa.
Myotonic Dystrophy 679

Gastrointestinal tract 826


Smooth muscle involvement
• Abdominal pain (55% of patients).
• Dysphagia (45%): skeletal muscle dysfunction in the
upper esophagus and pharynx.
• Vomiting (35%).
• Diarrhea (33%).
• Constipation (33%).
• Coughing when eating (33%).
• Anal incontinence (30%).
• Cholelithiasis.

Thyroid
Hypothyroidism or hyperthyroidism.

827 828 826 Longitudinal section of muscle


from a patient with myotonic
dystrophy showing long chains of large
numbers of internal nuclei.

827–830 Frontal alopecia, myopathic


facies (e.g. ptosis), and wasting of the
temporalis, masseters, sternomastoids
in a man (827, 828) and a woman
(829, 830) with myotonic dystrophy.
Note that the woman’s head is tilted
backwards to compensate for the
ptosis. She also has an ‘inverted smile’.

831 Wasting of the distal limb


muscles such as the forearm, anterior
tibial and calf muscles in myotonic
dystrophy.

831

829 830
680 Muscle Disorders

Pancreas • Generalized myotonia, worse in the cold and relieved by


Diabetes mellitus. warmth or exercise.
• Patients show diffuse muscle hypertrophy and little if any
Gonads weakness.
Tubular testicular atrophy, impotence and poor libido in men.
N.B. These patients tend to tolerate general anesthetics Sodium channelopathies
poorly. Paramyotonia congenita (autosomal dominant):
• Attacks of myotonia and flaccid weakness, usually precipita-
SPECIAL FORMS ted by cold, but also by hyperkalemia and beta-blockers.
Congenital myotonic dystrophy • Myotonia is relieved by hypokalemia or acetazolamide.
• Present since birth. • There is overlap with the periodic paralyses, since in some
• The abnormal gene is transmitted exclusively through patients the weakness is associated with hyper- or
maternal inheritance. hypokalemia, and hyperkalemic periodic paralysis results
• The fetus shows hydramnios and reduced fetal movements. from mutations in the same sodium channel gene as
• Neonates show respiratory distress, bilateral facial weakness paramyotonia congenita. Hypokalemic periodic paralysis is
and hypotonia. a calcium channelopathy (see p.693).
• In children mental retardation is a major feature; 75% of
patients need special education, and 75% of the remainder Distal myopathy
at mainstream schools require further assistance to achieve Neurogenic
minimum standards in reading and counting. • HMSN 2 (Charcot–Marie–Tooth–2).
• There is a high prevalence of cardiac and gastroinstestinal • Distal spinal muscular atrophy.
involvement: 20% die of cardiac dysfunction and 15% are • Scapuloperoneal syndrome.
still incontinent of feces at age 5 years.
• 25% die before 18 months of age, and 50% survive into the Myopathic
mid-30s. • Myotonic dystrophy.
• Fascioscapulohumeral dystrophy.
Myotonic dystrophy type 2 • Desminopathy.
• Myotonic dystrophy type 2 is linked to the long arm of • Mitochondrial myopathy.
chromosome 3 in a single large family. • Congenital myopathies.
• Clinical features are the same as those with myotonic • Glycogen storage disease.
dystrophy type 1, although the degree of facial and limb • Inclusion body myositis.
weakness and the ptosis are more like that of a mild, rather
than severe, myotonic dystrophy type 1. INVESTIGATIONS
Blood
Proximal myotonic myopathy (PROMM) • Fasting glucose.
• Proximal muscle weakness and myotonia predominates but • Thyroid function tests.
distal weakness may also develop in some individuals with • Serum IgG reduced.
PROMM. • Serum CK: normal or mildly elevated.
• The disease has a similar frequency as classic myotonic
dystrophy. Slit lamp examination
• It is linked to the same region of chromosome 3 as Cataracts.
myotonic dystrophy type 2.
• Patients show a less severe impairment than classic myotonic EKG
dystrophy . • Abnormalities of cardiac conduction are common, even in
• Congenital disease is absent. patients without cardiac symptoms, and progress over time.
• An annual EKG is recommended because EKG changes are
DIFFERENTIAL DIAGNOSIS often a predictor of development of cardiac symptoms.
Myotonia clinically • Ventricular late-potentials may be predictive of ventricular
• Myotonic dystrophy (adults). arrhythmias. However, potentially fatal heart block or
• Myotonia congenita (dominant and recessive forms). rhythm disturbances can occur despite a normal EKG. As
• Paramyotonia congenita ± hyperkalemic periodic paralysis. the perioperative period is a particularly dangerous time with
• Hypothyroid myopathy. respect to development of tachyarrhythmias and heart block,
• Drugs: 20,25-diazacholesterol (lipid lowering agent), the anesthetist must always be made aware of the diagnosis
triparanol (lipid lowering agent), beta-blockers, and of myotonic dystrophy.
depolarizing muscle relaxants (e.g. succinylcholine) may
cause or exacerbate myotonia. 24-hour EKG recording
This is indicated if cardiac symptoms are present or a significant
Non-dystrophic myotonias change occurs on the EKG.
Chloride channelopathies
Myotonia congenita: EMG
• Thompsen’s disease (autosomal dominant): presents in Multiple myotonic discharges.
infancy.
• Becker’s disease (autosomal recessive): present in childhood. Molecular DNA analysis
PCR technique: triplet repeat expansion.
Polymyositis 681

DIAGNOSIS POLYMYOSITIS
Clinical and EMG, but confirmed by molecular DNA analysis.

TREATMENT DEFINITION
The main goals are prevention and treatment of systemic An acquired proximal inflammatory myopathy characterized
disease. by progressive proximal muscle weakness, elevated serum
creatine kinase, and the presence of inflammatory infiltrates
Myotonia in muscle.
In general, patients do not complain much about myotonia:
• Phenytoin 100–200 mg bd orally can alleviate disabling EPIDEMIOLOGY
and bothersome myotonia with reasonable efficacy and • Prevalence: 1 in 100 000.
safety. • Age at onset: >18 years of age.
• Nifedipine 10–20 mg three times daily may help. • Gender: F≥M.
• Antimyotonic drugs such as quinine sulfate,
disopyramide (100–200 mg three times daily) and PATHOLOGY
procainamide (250–500 mg four times daily) prolong the Acute
PR interval and can impair cardiac conduction. • Predominantly endomysial (in the fascicles) inflammatory
Disopyramide has anticholinergic adverse effects. cell infiltrates (T cells), surrounding or partially invading
Procainamide may cause nausea, diarrhea, skin rash, individual muscle fibers (832).
confusion, and SLE-like syndrome and agranulocytosis. • B cells are infrequent.
• A short course of prednisolone may help particularly • T cells are of the cytotoxic type.
severe cases sometimes. • Single fiber necrosis with phagocytosis.
• ‘Moth-eaten’ fibers.
N.B. Any improvement in myotonia may not necessarily • Atrophy of both fiber types.
realize functional benefit for patients whose symptoms are more • Angular atrophic fibers.
as a result of weakness than myotonia. • Increased central nucleation.
• No perifascicular atrophy or microangiopathy.
Excessive daytime sleepiness due to
central mechanisms Chronic
• Lifestyle advice: take short naps at convenient times, such as • Marked variation in fiber size (833).
after meals, to minimize disruption of daily activities. • Hypertrophied fibers.
• Methylphenidate may be helpful, but there have been no • Central nucleation.
long term studies of risks and benefits. • Fiber splitting.
• Regenerating and necrotic fibers.
Excessive daytime sleepiness due to hypercapnia • Endomysial and perifascicular fibrosis.
Normal assisted ventilation. • Inflammatory cell exudates.
• Focally distributed architectural changes in individual fibers.
Fecal incontinence
Procainamide 300 mg bd. ETIOLOGY AND PATHOPHYSIOLOGY
Idiopathic polymyositis
PROGNOSIS T cell mediated cytotoxicity of muscle fibers.
Sudden death is well recognized, and may be due to heart
block or arrhythmia.

832 833

832 Polymyositis. Inflammatory infiltrate between muscle fibers within 833 Chronic polymyositis, transverse section of muscle, x140, H&E.
the fascicle (endomysial) and adjacent to necrotic fibers and small There is marked variability in muscle fiber size, central nucleation, a
intrafascicular blood vessels. marked increase in fibrous tissue, and a diffuse inflammatory cell
response. Some fibers are conspicuously rounded.
682 Muscle Disorders

Systemic autoimmune or connective tissue diseases Evolution of symptoms


• Mixed connective tissue disease. Weakness usually evolves slowly over months or, frequently,
• Systemic lupus erythematosus. several years, such that gross muscle wasting may be evident
• Rheumatoid arthritis. at the time of diagnosis.
• Scleroderma.
• Polyarteritis nodosa (834). Underlying carcinoma
• Sjögren’s syndrome. No link.

Infection DIFFERENTIAL DIAGNOSIS


Viral myositis • Myositis associated with autoimmune disorders (MCTD,
• HIV. SLE, RA, PSS, PAN).
• Human T-cell lymphotropic virus type 1. • Polymyalgia rheumatica: pain with limitation of
• Benign acute myositis: movement (which may be misinterpreted as muscle
– Influenza A and B. weakness), but the CK is normal and muscle biopsy
– Parainfluenza. shows only minimal abnormalities.
– Adenovirus 2. • Endocrine myopathy: thyroid disease.
• Acute rhabdomyolysis: • Metabolic myopathy: osteomalacic myopathy.
– Influenza A and B. • Toxic myopathy: alcohol, opiates, chloroquine, D-peni-
– Echo 9. cillamine, zidovudine.
– Adenovirus 21. • Muscular dystrophy.
– Herpes simplex. • Myotonic dystrophy.
– Epstein–Barr virus. • MG.
• Epidemic pleurodynia: Coxsackie B5 (also B1, 3 and 4). • Spinal muscular atrophy.
• Neuropathy.
Bacterial myositis
Acute suppurative myositis: INVESTIGATIONS
• Staphylococcus aureus. Blood
• Streptococcus. • Serum CK level: helpful in diagnosis and monitoring
• Yersinia. response to therapy. Levels may be normal or only
• Anerobic organisms. moderately raised (up to five times normal).
• ESR: usually, though not invariably, elevated.
Fungal myositis • Anti Jo-1 antibody, found in 30% of cases, is associated
with lung infiltrates and greater risk of cardiomyopathy.
Parasitic myositis
• Protozoa: EKG
– Toxoplasmosis. Occasionally shows heart block.
– Sarcosporidiosis.
– Trypanosomiasis. EMG
– Amebiasis. • Useful for confirming active or inactive myopathy and
• Cestodes: for excluding neurogenic disorders.
– Cysticercosis.
– Coenurosis.
– Hydatidosis.
– Sparganosis.
• Nematodes
– Trichinosis.
– Toxocariasis. 834
Drugs
• Penicillamine, zidovudine.

CLINICAL FEATURES (see Table 50)


Presentation of symptoms
• Slow onset (weeks to months).
• Usually symmetric weakness of proximal limb muscles,
typically involving the pelvic more than the shoulder
girdle, occasionally with pain and muscle tenderness.
• Weight loss, neck weakness, dysphagia and voice change
are common.
• No skin rash, eye and facial muscle involvement, family
history of neuromuscular disease, history of exposure to
myotoxic drugs or toxins, endocrinopathy, neurogenic 834 Polyarteritis nodosa. In this muscle biopsy there is intimal
disease, or dystrophy is present. thickening and infiltration of the walls of two small arteries by
macrophages and lymphocytes.
Polymyositis 683

• Shows increased spontaneous activity with fibrillations, • Encouragement of mobility (particularly in the elderly)
positive sharp waves, and complex repetitive discharges; and physiotherapy to prevent contractures are essential.
and myopathic potentials characterized by short- • A high protein diet is advisable.
duration, low-amplitude polyphasic motor units on • Attention to swallowing, adequacy of ventilation, and
voluntary activation. These findings are non-specific and precautions against deep venous thrombosis must be
occur in a variety of acute, toxic and active myopathic considered.
processes.
• Mixed myopathic and neurogenic potentials (polyphasic Corticosteroids
units of short and long duration) may also be present as • Oral prednisolone 1 mg/kg/day on alternate days.
a consequence of the regeneration of muscle fibers and • Combination therapy with methotrexate or azathioprine
chronicity of the disease. can be considered.
• Intravenous methylprednisolone: 0.5 g/day for 3 days
Muscle biopsy (pulse therapy) if symptoms are not controlled with oral
Endomysial infiltration by mononuclear inflammatory therapy.
infiltrates (predominantly T8 lymphocytes), surrounding
and invading muscle fibers (832–834). Avoiding complications of corticosteroid treatment
• Keep initial dose of prednisolone <1 mg/kg/day, if
DIAGNOSIS possible, particularly if mild disease, concern about
A diagnosis of exclusion, based on clinical features, sup- adverse effects (e.g. postmenopausal) or remission
ported by laboratory investigations, of which the most im- induced.
portant is muscle biopsy. Patients do not have (1) family his- • Begin to reduce high dose prednisolone (1 mg/kg/day)
tory, (2) exposure to myotoxic drugs or toxins, (3) anoth- within 4–6 weeks.
er acquired muscle disease caused by endocrine, metabolic, • Begin combination therapy at 4–6 weeks as a steroid
or neurogenic disease, (4) certain sporadically occurring dy- sparing agent if the disease is slow to come under control
strophies (dystrophinopathies, sarcoglycanopathies, dysfer- (i.e. introduce a second line agent earlier rather than
linopathies), and (5) inclusion body myositis. later, as opposed to continuing prednisolone for
2–3 months: too long really).
TREATMENT • Keep maintenance dose <10 mg/day (or 20 mg alternate
• Empirical. days).
• Treatment has non-selective effects on the immune • Monitor muscle performance regularly (manual,
system. myometer or isokinetic dynamometry, and functional
• Efficacy has not been proven in randomized trials. assessment).
• No treatment is uniformly effective. • Use prophylactic calcium supplements and biphos-
• Adverse effects can be serious. phonates, particularly in postmenopausal women.

Table 50 Major clinical features of dermatomyositis, polymyositis and inclusion body myositis (IBM)
Clinical features Dermatomyositis Polymyositis IBM
Male: female 1:2 1:1 3:1
Age at onset Any age >18 years >50 years
Site of weakness Limb girdle Limb girdle Asymmetric
Respiratory muscle involvement In severe cases No No
Dysphagia Frequent Rare 40% of patients
Muscle tenderness Frequent Infrequent Infrequent
Skin involvement Frequent No Absent
Involvement of other systems Lung, heart No No
Associations
Connective tissue diseases PSS, MCTD Yes <15% of cases
Autoimmune diseases Uncommon Common Uncommon
Viruses Not proven HIV, HTLV-1 Not proven
Parasites and bacteria No Yes No
Drug-induced myotoxicity Yes Yes No
Carcinoma 20% No No
Familial No No Yes, in some cases
Course Acute/subacute Subacute/chronic Chronic
684 Muscle Disorders

Steroid resistance DERMATOMYOSITIS


Second line agents
• Methotrexate: can be given as a single weekly oral dose.
• Azathioprine 2.5 mg/kg/day. DEFINITION
An idiopathic inflammatory myopathy with characteristic
Third line agents cutaneous manifestations.
• Cyclosporine.
• Cyclophosphamide. EPIDEMIOLOGY
• Plasma exchange. • Prevalence: 1 in 100 000.
• Age at onset: any age, affects children and adults.
Problems in management Comprises 95% of childhood myositis.
• Late diagnosis. • Gender: F>M (2:1).
• Steroid complications: cushingoid features.
• Ocular cataracts or glaucoma. PATHOLOGY
• Diabetes. Inflammatory infiltration of muscle occurs by macrophages
• Infection. and T and B lymphocytes. B cells are frequent, and T cells
• Osteoporosis. are of the helper type. The inflammatory infiltrates are
• Myopathy: predominantly perivascular, or in and around the
– If prednisone >10 mg/day for a long period. interfascicular septa, rather than within the fascicles.
– Detect by declining strength on myometry. Therefore, they do not penetrate the muscle fiber
– Normal CK. membrane. Damage appears to occur through compression,
– Myopathic EMG with spontaneous activity. rather than infiltration, of the fiber.
– Biopsy if uncertain. Microangiopathy: intramuscular blood vessels show
• Steroid resistance. endothelial hyperplasia with tubuloreticular profiles, fibrin
• Relapses: it is not common to be able to withdraw thrombi (particularly in children) and obliteration of
treatment and avoid relapse, but using a small capillaries. Capillary numbers are reduced, there are
maintenance dose also does not seem to prevent relapses. immunoglobulin deposits on vessel walls and blood vessel
• Immunodeficiency. endothelial cells show ultrastructural changes. The capillary
loss and consequent ischemia causes microinfarcts and may
Treatment of relapses be responsible for the sometimes striking perivascular
• Increase dose of prednisolone to 30–50 mg/day. atrophy seen.
• Add methotrexate or azathioprine. The necrotic, degenerating and regenerating muscle
• Add intravenous immunoglobulin if patients do not fibers are mostly in groups involving a portion of a muscle
show prompt improvement over 3 weeks. fasciculus and are often the result of microinfarcts within the
muscle.
Other ‘last resort’ forms of treatment Perifascicular atrophy, characterized by two to 10 layers
• Plasmapheresis. of atrophic fibers at the periphery of the fascicle is found in
• X-ray irradiation. about 90% of children and at least 50% of adults with
• Thoracic outlet drainage. dermatomyositis and is diagnostic of dermatomyositis, even
• Thymectomy. in the absence of inflammation.
• Stem cell transplant.
ETIOLOGY
PROGNOSIS • Unknown in most cases.
The response is less favorable than in dermatomyositis, • Caused or exacerbated by drugs in a few patients:
particularly in those with a long history at presentation. – Hydroxyurea.
Immunosuppressive therapy usually causes improvement – Quinidine.
and prevents further progression but significant improve- – Non-steroidal anti-inflammatory drugs.
ment may not occur in some. Antibody to signal recogni- – Penicillamine.
tion peptide, found in 5% of cases, is associated with a – 3-hydroxy-3-methylglutaryl coenzyme A-reductase
fulminant course and resistance to treatment. The 5-year inhibitors (‘statins’).
survival rate for treated patients is nearly 80%. Up to 30%
of patients may be left with residual muscle weakness. PATHOPHYSIOLOGY
A humorally-mediated microangiopathy (e.g. antibodies
against capillary endothelial cells with complement
activation) with muscle fiber inflammation occurring
secondarily to focal ischemia.

CLINICAL FEATURES
Muscle disease
• Initial symptoms include subacute onset of myalgias,
fatigue and weakness, manifested as difficulty climbing
stairs, raising the arms for actions such as shaving or
brushing hair, rising from a squatting or sitting position,
or a combination of these features.
Dermatomyositis 685

• Weakness typically involves the proximal muscles sym- Systemic features


metrically, and the pelvic more than the shoulder girdle. • Raynaud’s phenomenon: generalized arthralgias accom-
• Pain and tenderness on palpation of the muscles is panied by morning stiffness; a symmetric non-deforming
variable. arthritis involving the small joints of the hands, wrists and
• The course is slowly progressive during a period of weeks ankles; or both, may be present in up to 25% of patients.
to months. • Esophageal disease, manifested by dysphagia, occurs in
• Difficulty swallowing (dysphagia) or symptoms of about 15–50% of patients. There are two main forms:
aspiration may reflect involvement of striated muscle in – Proximal dysphagia is caused by involvement of striated
the pharynx or upper esophagus. muscle of the pharynx or proximal esophagus, correlates
• Dysphonia. with severity of the muscle disease, and responds to
steroid treatment.
Cutaneous manifestations – Distal dysphagia is due to involvement of non-striated
• Heliotrope rash (835): a violaceous to dusky erythe- muscle and is more common in patients who have an
matous rash, with or without edema, in a symmetric overlap with scleroderma or another collagen-vascular
distribution involving the perorbital skin. The rash may disorder.
be mild and only comprise a slight discoloration along • Pulmonary disease: occurs in about 15–30% of patients,
the eyelid margin. particularly those with esophageal disease, and is usually
• Gottron’s papules: slightly raised violaceous papules and due to an interstitial pneumonitis. Less common causes
plaques, with or without a slight scale and rarely a thick include the muscle disease itself (causing hypoventilation
psoriasiform scale, over bony prominences, particularly or aspiration), and treatments for the muscle disease (caus-
the metacarpophalangeal joints, proximal interphalangeal ing opportunistic infections or drug-induced hypersen-
joints, and distal interphalangeal joints. Papules may also sitivity pneumonitis). Associated with a poor prognosis.
be found overlying the elbows, knees, feet, or a combin- • Cardiac disease: present in up to 50% of patients but
ation of these. Within the lesions there is commonly uncommonly symptomatic. Disorders include conduc-
telangiectasia. tion defects and primary end-rhythm disturbances, and
• An erythematous to violaceous psoriasiform dermatitis even less commonly congestive heart failure, pericarditis,
involving the scalp. and valvular disease. Associated with a poor prognosis.
• Malar erythema. • Calcinosis of the skin (firm, yellow, or flesh-colored
• Poikiloderma (which is the combination of atrophy, nodules, usually over bony prominences, and occasionally
dyspigmentation, and telangiectasia) on sun exposed skin extruding through the surface of the skin) or muscle
such as the extensor surfaces of the arms, the ‘V’ of the (generally asymptomatic) is unusual in adults but may
neck, and the upper back (shawl sign). occur in up to 40% of children and adolescents with
• Violaceous erythema on the extensor surfaces. dermatomyositis.
• Nailfold changes: periungual telangiectases and/or
hypertrophy of the cuticle, and small hemorrhagic Malignancy
infarcts in the hypertrophic area. • About 20–25% have associated malignancy, before the
onset of myositis, concurrently with myositis, or after the
onset of dermatomyositis.
• Malignancy is more common in older patients
(>50 years) but may even occur in children.
• The site of malignancy can be predicted by the patient’s
835 age (e.g. testicular cancer in young men, colon and
prostate cancer in elderly men).
• Gynecologic malignancy, particularly ovarian carcinoma,
is common. Nasopharyngeal carcinoma is common
among Asians with dermatomyositis.

Evolution of symptoms
• Symptoms are usually subacute over several weeks.
• Evolution may be rapid with severe weakness, dysphagia
and respiratory muscle involvement, sometimes requiring
ventilatory support. Muscle pain, tenderness, and
swelling, if present, tend to equate with a rapid onset.

SPECIAL FORMS
Childhood dermatomyositis
• Childhood dermatomyositis is more common than
childhood and adolescent polymyositis.
• Onset is usually insidious and mistaken for a viral-type
illness or dermatitis, but a fulminant onset and course
may occur.
• It is commonly characterized as a vasculitis and has
835 A blue–purple discoloration of the eyelids, cheeks and nose by a greater potential for calcinosis than adult disease.
typically heliotrope and slightly edematous rash of dermatomyositis.
686 Muscle Disorders

DIFFERENTIAL DIAGNOSIS Muscle biopsy


Muscle weakness • Biopsies are obtained under local anesthetic from a
• Muscular dystrophy. moderately involved muscle, typically quadriceps or
• Myotonic dystrophy. deltoid (see Pathology, above).
• Steroid myopathy. • Light microscopy shows microvascular injury with
• Neuropathy. perifascicular fiber atrophy, a perimysial inflammatory
infiltrate dominated by B and T4 lymphocytes; deposits
Heliotrope rash and photosensitive of membrane attack complex confirm the role of
poikilodermatous eruption complement.
• Systemic lupus erythematosus: a heliotrope rash is rarely seen.
• Scleroderma: a heliotrope rash is rarely seen. Skin biopsy

Gottron’s papules PROGNOSTIC TESTS


• Systemic lupus erythematosus. Tests and examinations are determined by the patient’s age
• Psoriasis: distinct histopathology. and gender, and commonly include rectal, vaginal and
• Lichen planus: distinct histopathology. breast examination for assessment of malignant disease.
Tests include mammography, sigmoidoscopy, EKG, chest x-
Scalp erythematous to violaceous ray and pulmonary function tests, barium swallow and
psoriasiform dermatitis esophageal motility studies, abdominal scanning (ultrasound
• Psoriasis. or CT), testing for fecal occult blood and basic hematologic
• Seborrheic dermatitis. and biochemical studies. Repeat each year for the first
3 years after diagnosis or whenever new symptoms arise.
Facial erythema
• Systemic lupus erythematosus. DIAGNOSIS
• Rosacea. Based on clinical features, supported by laboratory investi-
• Seborrheic dermatitis. gations, of which the most important is muscle biopsy.
• Atopic dermatitis.
TREATMENT
INVESTIGATIONS General
Diagnosis • Bedrest combined with a range-of-motion exercise
ESR is usually elevated programme if patients have progressive weakness.
• Raise the head of the bed and avoid meals before
Serum muscle enzymes bedtime in patients with dysphagia.
CK, aldolase, lactate dehydrogenase, alanine aminotrans-
ferase levels: Muscle disease
• CK is the most specific and widely used. Treatment for muscle disease is controversial because of the
• It is helpful in the diagnosis and monitoring the response absence of controlled clinical trials.
to therapy.
• Levels may be raised up to 20 times normal, particularly First line therapy
in acute cases, whereas levels may be normal or only Oral prednisolone 0.5–1 mg/kg bodyweight/day for at
moderately raised (up to five times normal) in polymyo- least 1 month after myositis has become clinically and
sitis, and are often normal in inclusion body myositis. enzymatically inactive. The dose is then gradually reduced,
generally over a period lasting 1.5 to two times as long as
Serologic tests the period of active intense treatment, but depending on
• Antinuclear antibody: commonly positive. clinical progress and, to some extent, serum CK levels.
• Antibodies to Mi-2, and antinuclear antibody, are specific but Convert to an alternate day regime after a few months.
not sensitive, being found in only about 25% of patients. N.B. It is easier to manage patients who have presented
• Antibodies to Jo-1 are predictive of pulmonary acutely or subacutely, in whom the response to treatment is
involvement but are rare. usually more obvious and a high CK levels falls, than those
• Antibodies to Ro (SS-A) are found in rare cases. with a slowly progressive disorder: if a response occurs at all,
it is slow, and the CK level may have been normal or little
EMG elevated at presentation.
• EMG is useful for confirming active or inactive myopathy
and for excluding neurogenic disorders. Adjunct or second line therapy
• Shows a myopathic pattern of increased spontaneous activity Immunosuppressive agents: about 25% of patients will not
with fibrillations, positive sharp waves, and complex repetitive respond to systemic corticosteroids and 25–50% will develop
discharges; and myopathic potentials characterized by short- substantial steroid-related adverse effects. Therefore, early
duration, low-amplitude polyphasic motor units on voluntary intervention with steroid-sparing agents, such as methotrexate,
activation. These findings are non-specific and occur in a azathioprine (2.5 mg/kg bodyweight), cyclophosphamide,
variety of acute, toxic and active myopathic processes. mycophenolate mofetil, chlorambucil, or cyclosporine may be
• Mixed myopathic and neurogenic potentials (polyphasic effective in inducing or maintaining a remission.
units of short and long duration) may also be present as About 50–75% of patients respond to an immuno-
a consequence of the regeneration of muscle fibers and suppressive agent with an increase in strength, a decrease in
chronicity of the disease. enzyme concentrations, or a decrease in corticosteroid dosage.
Inclusion Body Myositis (IBM) 687

Other therapy INCLUSION BODY MYOSITIS (IBM)


• High-dose intravenous immunoglobulin has been shown
to be effective for recalcitrant dermatomyositis in a
double-blind, placebo-controlled trial (N. Engl. J. Med., DEFINITION
1993; 329: 1993–2000). The indications are usually Inclusion body myositis is an idiopathic inflammatory
resistance to steroids and second line agents, adverse myopathy.
reactions to immunosuppressants and severe relapses.
The regimen may be 0.4 g/kg/day for 5 days followed EPIDEMIOLOGY
by monthly three day courses for up to 6 months (unless • Prevalence: uncommon but the most common cause of
no response within 3 months). acquired myositis in patients over 50 years of age.
• Largely anecdotal reports have described success with • Age at onset: after 50 years of age.
pulsed methylprednisolone, combination immunosup- • Gender: M>F (3:1).
pressive therapy and whole body irradiation. • Race: more common in whites than blacks.
• Plasmapheresis was ineffective in a placebo-controlled
trial (N. Engl. J. Med., 1992; 326: 1380–1384). PATHOLOGY
• Inflammatory infiltrates (predominantly T cells)
Skin disease surrounding or invading individual non-necrotic muscle
• Patients should avoid sunlight or use a broad-spectrum fibers (endomysial inflammation) (like that seen in
sunscreen with a high sun protective factor, if they are polymyositis) are often present but rarely extensive.
photosensitive. • Basophilic granular inclusions are distributed around the
• Hydroxychloroquine hydrochloric acid 200–400 mg/day edge of slit-like vacuoles (rimmed vacuoles) (836).
is effective in about 80% of patients when used as a • Eosinophilic cytoplasmic inclusions.
steroid-sparing agent. • Angulated fibers, often in small groups.
• Chloroquine phosphate 250–500 mg/day can be used • Intranuclear or intracytoplasmic 15–18 nm tubulo-
if patients are not responsive to hydroxychloroquine. filaments in muscle fibers on electron microscopy.
• Periodic ophthalmologic examinations and blood counts
are required for patients on continuous antimalarial ETIOLOGY AND PATHOPHYSIOLOGY
therapy. Sporadic
• Methotrexate 15–35 mg/week can also be used. • Uncertain; autoimmune, viral and degenerative theories
prevail.
PROGNOSIS • Partly mediated by cytotoxic T cells, with a secondary
Adverse prognostic factors inflammatory response to degenerating muscle (which is
• Increasing age. why the amount of inflammation is variable and the
• Severe myositis (frequently correlates with the degree of response to immunosuppression is poor).
weakness and the serum CK concentrations). • Multiple mitochondrial DNA deletions are present in
• Dysphagia or dysphonia. 75% of patients.
• Cardiopulmonary involvement. • Amyloidogenic protein deposition has been reported in
• Malignant disease. affected muscle and increased cellular prion protein has
• Poor response to corticosteroid therapy. also been described.

Prognosis Hereditary
• If treated early, most patients will respond well, with many Autosomal recessive rimmed vacuolar myopathies
showing full recovery of muscle function. In most cases the Gene locus 9p1-q1.
disease will burn itself out, although this may take many
years during which time treatment has to be continued. Autosomal dominant rimmed vacuolar myopathies
• Interstitial lung disease and other pulmonary disorders are • Gene locus 14q.
an important but under-recognized cause of late • Oculopharyngeal muscular dystrophy.
morbidity. Similarly, cardiac involvement is common with
conduction defects, rarely leading to complete heart block,
arrhythmias and congestive heart failure. 836
Malignancy
The myositis may follow the course of malignant disease (a
paraneoplastic course) or may follow its own course
independent of treatment of the malignant disease.

836 Eosinophilic (pink) cytoplasmic inclusions and basophilic (blue)


granular inclusions located at the periphery of narrow vacuoles (rimmed
vacuoles) in a muscle fiber of a patient with inclusion body myositis.
688 Muscle Disorders

CLINICAL FEATURES METABOLIC AND ENDOCRINE


• Gradual onset. MYOPATHIES
• Painless weakness of distal and proximal muscles
(particularly long finger flexors [flexor digitorum
profundus], wrist flexors and quadriceps femoris) which DEFINITION
may be asymmetric. Metabolic and endocrine myopathies are a large,
• Dysphagia in up to 30% of cases. heterogeneous group of inherited and acquired disorders of
• Muscle wasting can be marked. muscle due to a disturbance of metabolism.
• Early loss of deep tendon reflexes.
• Numbness and sensory symptoms may suggest an EPIDEMIOLOGY
associated peripheral sensory neuropathy. • Uncommon.
• Age: usually infants and children, but may present in
DIFFERENTIAL DIAGNOSIS adulthood (particularly endocrine myopathies).
• Polymyositis. • Gender: usually M=F.
• Neuropathy: e.g. diabetic amyotrophy.
ETIOLOGY AND PATHOPHYSIOLOGY
INVESTIGATIONS Disorders of glycogen metabolism
Blood Acid maltase deficiency
• Serum CK level: often normal or mildly raised. • An autosomal-recessive glycogen storage disorder.
• ESR: normal in 80% of cases. • Caused by a deficiency of lysosomal alpha-glucosidase
which results in impaired lysosomal conversion of
EMG glycogen to glucose so that glycogen accumulates in the
• A mixed neuromyopathic pattern may be seen. liver, heart, CNS and muscle.
• 30% of patients have EMG signs of axonal neuropathy. • The genetic abnormality maps to chromosome 17.

Muscle biopsy McArdle’s disease


Biopsy may show inflammation, predominantly an endo- • Autosomal recessive.
mysially-located mononuclear infiltrate with cytotoxic T cells • Caused by a myophosphorylase (a-1,4-glucan ortho-
invading non-necrotic muscle fibers; rimmed vacuoles and phosphate glycosyltransferase) deficiency, which results
filamentous inclusions in muscle are characteristic. The in impaired conversion of glycogen to glucose-1-
inclusions contain ubiquitin and tau protein, and are phosphate in muscle.
pathologically similar to Alzheimer neurofibrillary tangles. • The gene for myophosphorylase maps to chromosome
11q13. At least 16 different mutations have been
DIAGNOSIS identified, the most frequent of which is a nonsense
Highly likely if a clinical phenotype of asymmetric muscle mutation in exon 1 (R49X), which causes the substi-
weakness with prominent wrist flexor, finger flexor and knee tution of an arginine (CGA) with a stop codon (TGA),
extensory muscle involvement is present. A definitive and a missense mutation (W797R) in exon 20.
diagnosis requires electron microscopy of muscle biopsy • Glycogen breakdown is impossible during a sudden burst
specimens showing muscle fibers with rimmed vacuoles of muscle activity; ATP generation is compromised;
containing 15–18 nm tubulofilaments and small amyloid muscle cell pH is shifted to alkaline.
deposits, in addition to light microscopy evidence of
endomysial T cell infiltration of non-necrotic muscle fibers. Phosphofructokinase deficiency
An accurate diagnosis is important because inappropriate • Autosomal recessive.
treatment can cause adverse effects. • Caused by impaired conversion of fructose-6-phosphate
to fructose-1,6-diphosphate in muscle.
TREATMENT
• Numerous forms of immunosuppressive treatment have Debranching enzyme system deficiency
been tried without benefit: most patients are resistant to • Autosomal recessive.
immunosuppressive treatment. High-dose steroids are • Caused by impaired hydrolysis of glycogen to glucose-1-
rarely beneficial clinically despite often reducing a raised phosphate.
CK.
• In some who are not old and frail, the condition may Disorders of lipid metabolism
stabilize with a 3–6 month trial of prednisolone • Long chain fatty acids are a major source of muscle
combined with methotrexate (or azathioprine). energy and are consumed during muscular activity.
• Anabolic steroids (e.g. clenbuterol 20 µg/day–20 µg bd) • Carnitine is involved in the transport of free fatty acids
are being trialled. into mitochondria.
• A physiotherapy/strength training program is also
important. Muscle carnitine deficiency
Lipids accumulate in muscle which is deficient in carnitine.
PROGNOSIS
Gradual deterioration is usual, with increasing weakness of Carnitine-O-palmitoyltransferase deficiency
the neck, trunk, and distal arm muscles, and extensive • Autosomal recessive.
weakness and wasting in the legs. The disease can progress • Caused by impaired transport of free fatty acids into
to severe generalized weakness and disability. mitochondria.
Metabolic and Endocrine Myopathies 689

• The abnormal gene maps to chromosome 1. • Fluorinated corticosteroids, such as dexamethasone and
triamcinolone, have more myopathic potential. The dose
Mitochondrial myopathies (see p.162) required to cause myopathy varies among individuals.
Reduced ATP generation by oxidative phosphorylation.
Addison’s disease and other forms of hypoadrenalism
Endocrine disorders May occur as one component of adrenoleukodystrophy (see
Thyrotoxic myopathy p.166).
Muscle stiffness and weakness.
Acromegaly
Thyrotoxic periodic paralysis (see p.694)
Electrolyte disorders
Dysthyroid eye disease (exophthalmic ophthalmoplegia) (837) Calcium
• Hyperparathyroidism:
Hypothyroid myopathy – Primary hyperparathyroidism: adenoma.
– Secondary hyperparathyroidism: typically secondary to
Muscle weakness renal disease.
Thyroid hormone has a regulatory role on the transcription • Osteomalacia.
of numerous muscle genes encoding both myofibrillar and
calcium-regulatory proteins. Potassium
• Iatrogenic usually.
Steroid myopathy (838) • Secondary periodic paralyses (see p.693).
• About 70% of patients with Cushing’s syndrome have • Primary hyperaldosteronism.
muscle weakness.
Malignant hyperthermia
• An autosomal dominant susceptibility to a number of
drugs, particularly anesthestics such as halothane and suc-
837 cinylcholine. Other drugs include tricyclic antidepressants,
monoamine oxidase inhibitors, methoxyflurane, ketamine,
enflurane, diethyl ether, and cyclopropane.
• Due to a malfunction of the calcium channel of the sarco-
plasmic reticulum (the ryanodine receptor). The abnormal
ryanodine receptor may accentuate calcium release.
• The gene for the ryanodine receptor maps to chromo-
some 19 (13-1).
• Fast, uncontrolled increase in skeletal muscle metabolism
associated with rhabdomyolysis occurs and may occur in
association with dystrophinopathies and central core
congenital myopathy.

CLINICAL FEATURES
837 Dysthyroid eye disease (exophthalmic ophthalmoplegia) showing Disorders of glycogen metabolism
left eyelid retraction, and exophthalmos. Dysconjugate vertical eye Acid maltase deficiency
movements were present. • Infantile form: Pompe’s disease: a generalized glyco-
genosis with severe cardiomyopathy, hypotonia, macro-
838 glossia, cardiomegaly and hepatomegaly. Death occurs in
infancy.
• Adult form: a slowly progressive myopathy affecting
predominantly the diaphragm (hence respiratory failure),
biceps, shoulder, and thigh adductor muscles. There is
little or no heart disease.

McArdle’s disease
• Males are affected more than females (4:1).
• Onset in childhood or adolescence usually.
• Attacks of exercise intolerance with muscle pain (myalgia)
and stiffness, often precipitated by brief, strong exercises.
• Fatigue.
• Cramps and dark urine (myoglobinuria) usually develop
during adulthood.
838 Muscle biopsy of a patient with a steroid myopathy showing Type • Seizures.
II muscle fiber atrophy, which may be seen in disuse, cachexia, • Renal failure may occur secondary to myoglobinuria.
myasthenia gravis as well as a steroid effect. Myosin ATPase stain, pH • Not progressive usually.
9.4.Type 1 fibers are pale and Type 2 fibers are darkly stained. Fibers
showing an intermediate reaction are usually Type 1.
690 Muscle Disorders

Phosphofructokinase deficiency: Hypothyroid myopathy


• Clinically similar to McArdle’s disease. • More common in women.
• A mild hemolytic tendency is sometimes present. • Aching and painful muscles.
• Cramps.
Debranching enzyme system deficiency • Enlarged muscles.
• Infancy: growth retardation, hepatomegaly, mild • Muscle weakness (rare).
myopathy, and seizures which tend to improve after • Myedema (ridging of muscle on percussion).
puberty. • Respiratory muscle weakness may be present.
• Adults: mild weakness of hands and legs. Cardiomyopathy • Slow-recovery reflexes.
is a late complication.
Steroid myopathy
Disorders of lipid metabolism • Proximal muscle weakness, earlier and worse in the lower
Muscle carnitine deficiency limbs than upper limbs, and sometimes painful. Wasting
Myopathy develops in infants or children. is late.
• Difficulty climbing stairs.
Carnitine-O-palmitoyltransferase deficiency
• Most patients are male and present in adolescence. Addison’s disease and other forms of hypoadrenalism:
• Intermittent attacks occur without warning; precipitated • Myalgia and muscle cramps.
by prolonged exertion, fasting or a high fat diet. Muscle • Proximal muscle weakness.
cramps, muscle pain, and dark urine (myoglobinuria) are • Painful flexion contractures in the legs sometimes.
present, with normal muscle strength during attacks. • Fatigue and lassitude.
• Exposure to cold, viral infections, and general anesthesia • Postural hypotension.
can also precipitate rhabdomyolysis. • Confusional state, stupor and coma.
• Renal failure (due to myoglobinuria) and respiratory
failure may ensue. Acromegaly
• Patients have a normal capacity to perform short, • Increased muscle bulk.
demanding exercise. • Improved strength initially but later muscle wasting and
weakness occur.
Endocrine disorders • Non-specific headache.
Typically mild to moderate proximal muscle weakness. • Associated entrapment neuropathy (e.g. carpal tunnel
syndrome).
Thyrotoxic myopathy • Sensorimotor peripheral neuropathy (sometimes with
• Common. enlarged nerves).
• Proximal muscle weakness and some wasting occurs; • Visual field defects.
occasionally the bulbar and respiratory muscles only are • Hypopituitarism.
affected (cf. myasthenia, p.657). • Obstructive sleep apnea.
• Fatigue. • Complications of diabetes and hypertension.
• Heat intolerance.
• Normal or augmented reflexes. Electrolyte disorders
• Fasciculations sometimes (cf. motor neuron disease, Hyperparathyroidism and osteomalacia
p.534). Proximal and often painful muscle weakness, mainly affecting
• Associated with hypokalemic periodic paralysis and the legs and associated with mild wasting.
myasthenia gravis.
Hypokalemia and hyperkalemia
Thyrotoxic periodic paralysis (see p.694) Cause of generalized weakness.
• Very rare.
• Orientals particularly are affected. Malignant hyperthermia
• During general anesthesia or after exposure to a causal
Dysthyroid eye disease (exophthalmic ophthalmoplegia) (837) drug.
• Can be quite asymmetric. • Rapid elevation of temperature which may rise to 43°C
• Difficulty of upgaze initially (inferior rectus infiltrated (109°F).
early). • Tachycardia.
• Diplopia. • Muscle rigidity (e.g. begins with trismus).
• Lid retraction. • Areflexia.
• Exophthalmos. • Coma.
• Conjunctival and lid edema.
• Exposure keratopathy. DIFFERENTIAL DIAGNOSIS
• Ptosis often. Muscle cramps or pain on exercise
• Little pain (grittiness or fullness). • Disorders of glycogenolysis or glycolysis (e.g. McArdle’s
• Eventually raised intraocular pressure and blindness may disease).
occur. • Carnitine-O-palmitoyltransferase deficiency.
• The patient is usually but not necessarily thyrotoxic. • Mitochondrial myopathies, including zidovudine toxicity.
• Toxic myopathy caused by clofibrate and related drugs.
• Hypothyroid myopathy.
Metabolic and Endocrine Myopathies 691

Myoglobinuria • Muscle biopsy: vacuolar myopathy with high glycogen


Myoglobin is found in muscle and if there is severe and content, and acid maltase deficiency.
acute muscle injury it is released into the blood and appears • Chorionic villi biopsy: for prenatal diagnosis.
in the urine (myoglobinuria). The urine is colored brown
in severe cases and reacts to benzidine; if there is neither McArdle’s disease
hematuria nor hemoglobinemia a positive test strongly • Serum CK: elevated after exercise.
suggests myoglobinuria. The muscle enzymes are always • Urine: myoglobinuria occasionally.
raised and the muscles are tender, weak and sometimes • EMG: myopathic.
swollen. In severe cases acute renal failure occurs. • Forearm ischemic exercise produces no increase in
• Disorders of glycogenolysis or glycolysis (e.g. McArdle’s venous lactate levels.
disease). • Muscle biopsy: subsarcolemmal deposits of glycogen at
• Carnitine-O-palmitoyltransferase deficiency. the periphery, undetectable histochemical reaction of
• Duchenne muscular dystrophy after anesthetic exposure. phosphorylase.
• Acute alcoholic myopathy.
• Acute viral myositis and other acute fulminating Phosphofructokinase deficiency
inflammatory myopathies. Forearm ischemic exercise produces no increase in venous
• Malignant hyperthermia crisis. lactate levels.
• Neuroleptic malignant syndrome (see p.133).
• Status epilepticus. Debranching enzyme system deficiency
• Excessive exercise. • Infancy: fasting hypoglycemia and ketonuria.
• Heat stroke. • Adults: serum CK: elevated.
• Crush injury of muscle. • Forearm ischemic exercise produces no increase in
• Snake bite. venous lactate levels.
• Muscle biopsy: excess glycogen.
Myotonia clinically
• Myotonic dystrophy (adults). Disorders of lipid metabolism
• Myotonia congenita (dominant and recessive forms). Muscle carnitine deficiency
• Paramyotonia congenita ± hyperkalemic periodic • Normal blood level of carnitine.
paralysis. • Muscle biopsy: accumulation of lipid which is deficient
• Hypothyroid myopathy. in carnitine.

Respiratory insufficiency (possible and prominent) Carnitine-O-palmitoyltransferase deficiency


• Acid maltase deficiency (adult-onset). • Urine: myoglobinuria.
• Acute myopathy after administration of high-dose • Serum CK: normal but often raised following the attacks.
corticosteroids and muscle paralysing agent (e.g. status • Muscle biopsy at the time of an attack: may show lipid
asthmaticus). accumulation and little CPT activity.
• Myotonic dystrophy.
• Nemaline myopathy. Endocrine disorders
• Centronuclear myopathy. Typically a myopathic EMG and the non-specific finding of
• Myopathy with cytoplasmic bodies. Type II fiber atrophy on muscle biopsy.

Thyrotoxic myopathy Thyrotoxic myopathy


• Hypokalemic periodic paralysis. • Serum CK: normal.
• Myasthenia gravis. • EMG: myopathic.
• Motor neuron disease.
Thyrotoxic periodic paralysis (see p.694)
Malignant hyperthermia Plasma potassium is usually low but can be normal.
Neuroleptic malignant syndrome (see p.133): also present
with high fever, rigidity, tachycardia, and rhabdomyolysis, Dysthyroid eye disease (exophthalmic ophthalmoplegia)
but it is of slower onset over days to weeks, not familial, and • Thyroid function tests: the patient is usually but not
usually triggered by drugs that block central dopaminergic necessarily thyrotoxic.
pathways, such as phenothiazines, lithium and haloperidol, • Thyroid antibodies: often positive.
or can occur after discontinuation of L-dopa (levodopa) for • Response to thyrotrophin releasing hormone: abnormal.
Parkinson’s disease. • CT scan of the orbits: enlarged extraocular muscles.

INVESTIGATIONS Hypothyroid myopathy


Disorders of glycogen metabolism • Serum CK: may be grossly elevated.
Acid maltase deficiency • Thyroid function tests: low thyroxine. TSH may be
• Serum CK: slightly elevated. elevated if primary hypothyroidism.
• EMG: non-specifically myopathic but myotonic • Urine myoglobin: rhabdomyolysis may be present.
discharges may occur (although patients do not have
myotonia).
• Blood leukocyte or urine: acid maltase deficiency.
692 Muscle Disorders

Steroid myopathy Dysthyroid eye disease (exophthalmic ophthalmoplegia)


• Serum CK: normal. • Restore the euthyroid state.
• EMG: normal insertional activity and no spontaneous • Tarsorrhaphy to protect the cornea.
activity. • Surgical correction of diplopia if necessary.
• Muscle biopsy (838). • Severe cases: high doses of corticosteroids, cyclosporine,
and even orbital decompression have been used to save
Addison’s disease and other forms of hypoadrenalism sight.
Serum electrolytes: hyponatremia and hyperkalemia.
Hypothyroid myopathy
Acromegaly Restore the euthyroid state.
Serum CK: sometimes elevated.
Steroid myopathy
Hyperparathyroidism and osteomalacia • Change to a non-fluorinated steroid.
Serum CK: usually normal. • Reduce steroid dose to the lowest possible therapeutic
level.
Malignant hyperthermia • Administer the steroid on an alternate daily basis if
• Arterial blood gases: metabolic acidosis. needed.
• Serum CK: precipitous rise, sometimes to 10 000 times • Adequate diet and exercise should assist recovery.
the normal values.
• Blood coagulation profile: disseminated intravascular Hyperparathyroidism
coagulation. • Primary hyperparathyroidism: remove the adenoma.
• Urine: myoglobinuria. • Secondary hyperparathyroidism (typically to renal
disease):
DIAGNOSIS – Partial parathyroidectomy.
The diagnosis usually depends on histochemistry or – 1,25 dihydroxycholecalciferol.
biochemical assay of muscle biopsy material. – 1-alpha tocopheral.
• Osteomalacia: vitamin D therapy.
PREVENTION
Malignant hyperthermia Malignant hyperthermia
• Screen the relatives of affected patients by muscle biopsy; Treatment depends on the severity, which is often related to
abnormal muscle contracture in vitro is induced by the dosage and duration of anesthesia.
caffeine or halothane.
• Barbiturate, nitrous oxide, and opiate non-depolarizing Mild cases
relaxant anesthesias should not induce malignant Discontinue the anesthetic.
hyperthermia.
More severe cases
TREATMENT • Dantrolene 2 mg/kg i.v. every 5 min, up to 10 mg/kg:
Correct the metabolic defect if possible. inhibits calcium release from the sarcoplasmic reticulum.
• Correct associated hyperkalemia (not with calcium).
Disorders of glycogen metabolism • Increase ventilation.
Acid maltase deficiency • Correct the acid–base disturbance: give i.v. sodium
• No specific therapy at present but promising results of bicarbonate 2–4 mg/kg.
myophosphorylase gene transfer in myoblasts in vitro • Cool the patient: cooling blankets and cold i.v. fluids
have been reported in McArdle’s disease. until temperature reaches 38°C (100°F).
• Inspiratory exercises are useful. • Volume load with diuretics if myoglobinuria is present.
• Give steroids for the acute stress reaction.
McArdle’s disease
Oral creatine supplementation may improve skeletal muscle PROGNOSIS
function. In most cases, the weakness reverses when the metabolic
defect is corrected but improvement may take weeks to
Disorders of lipid metabolism months.
Carnitine-O-palmitoyltransferase deficiency The prognosis for malignant hyperthermia is more
No specific therapy. guarded but mortality can be greatly diminished by
recognizing the syndrome and treating it appropriately.
Endocrine disorders
Thyrotoxic myopathy
• Correct the hyperthyroidism
• Symptomatic therapy with beta-blockers.
• Glucocorticoids can be used in thyroid storm to block
the peripheral conversion of T4 to T3.

Thyrotoxic periodic paralysis (see p.694)


Correct the thyrotoxicosis and plasma potassium if low.
Hypokalemic Paralysis 693

HYPOKALEMIC PARALYSIS • Uterosigmoidostomy.


• Laxative abuse.

DEFINITION CLINICAL FEATURES


A rare but treatable clinical syndrome, representing a • Muscular weakness, particularly of the lower extremities is
heterogeneous group of disorders, characterized by acute present, which can be severe and generalized with marked
systemic weakness and low serum potassium. potassium depletion (e.g. virtually total paralysis including
respiratory, bulbar and cranial musculature).
EPIDEMIOLOGY • Deep tendon reflexes may be decreased or absent.
• Rare. • Consciousness and sensation are preserved.
• Age: youth and young adults predominantly. • Underlying etiology may be determined from the age of
• Gender: M>F. onset, race, family history, medications and underlying
disease states.
ETIOLOGY AND PATHOPHYSIOLOGY
• Symptomatology results from the increased ratio between SPECIAL FORMS
intra- and extracellular potassium concentrations, which Familial (primary) periodic paralysis (FPP)
modifies membrane polarization and thereby alters the Epidemiology
function of excitable tissues such as nerve and muscle. • Age of onset: early in life (e.g. puberty), rarely after 25 years
• Most cases are due to alteration in the transcellular of age.
distribution of potassium and the others to actual potassium • Gender: M>F.
depletion from renal or extrarenal losses. • Race: Caucasians typically.

Causes of hypokalemia Etiology and pathophysiology


Transcellular shift of K (no depletion) • Autosomal dominant inheritance (2/3 of all cases of
• Familial or primary hypokalemic periodic paralysis (most periodic paralysis).
cases). • Mutations in the gene (on chromosome 1q32) encoding
• Thyrotoxic periodic paralysis (839, 840). the skeletal muscle voltage-gated calcium channel α-1
• Barium poisoning. subunit (CACNL1A3) account for most cases.
• Insulin excess. • Mutations in the gene coding for the skeletal muscle
• Alkalosis. voltage-gated sodium channel α subunit (SCN4A)
account for a minority of cases.
Actual K depletion • Weakness occurs in association with hypokalemia (but can
Renal losses: also occur with normokalemia, or hyperkalemia).
• Excessive mineralocorticoids (primary and secondary However, alterations in potassium regulation are well
aldosteronism, liquorice ingestion, glucocorticoid excess). documented. Total body potassium stores remain adequate,
• Renal tubular diseases (renal tubular acidoses, leukemia, but serum potassium decreases due to potassium migration
Liddle’s syndrome, antibiotics, carbonic anhydrase inhibitors). into muscle cells which causes the muscles to become
• Diuretics. electrically inexcitable.
• Magnesium depletion. • The exact method of potassium translocation is not known
but is secondary to an abnormality in muscle membrane.
Extra-renal losses: (? An increase in muscle membrane sodium or calcium
• Dietary deficiency. permeability associated with an inherited defect within
• Diarrhea. skeletal muscle voltage-gated sodium or calcium
• Rectal villous adenoma. channels.)
• Fistulas. Continued overleaf

839 840

839, 840 Thyrotoxic patient presenting with periodic paralysis. Exophthalmos due to thyrotoxicosis (839). Lateral view of the neck showing
thyroid goiter (840).
694 Muscle Disorders

Etiology and pathophysiology (continued) Etiology


• Also associated with paramyotonia congenita, myotonia Any cause of thyrotoxicosis but usually Graves disease.
congenita, and generalized myotonia (i.e. both hyper- Attacks only occur during hyperthyroidism.
kalemia and hypokalemia can cause paralysis).
Pathogenesis
Clinical features Uncertain; perhaps a decrease in activity of the calcium pump.
• Episodic attacks of muscle weakness with no stiffness.
• Precipitants: decrease in blood potassium levels, rest, sleep, Clinical features
carbohydrate intake or insulin administration (attacks Similar to FPP, but also signs of hyperthyroidism, which are
almost never occur during vigorous physical activity). frequently, but not always subtle (839, 840). Paralytic
• Frequency: varies from daily to yearly. attacks can be induced by the thyrotoxic state, and
• Duration: from 3–4 hours to a day or more. carbohydrate and insulin administration but only if the
patient is hyperthyroid, and not euthyroid.
Differential diagnosis
Hyperkalemic familial periodic paralysis: Treatment
• Usually starts in infancy or childhood. • Acute attack: potassium administration. Concurrently,
• Attacks can be precipitated or induced by fasting, cold, begin to correct the hyperthyroid state and avoid preci-
pregnancy and potassium loading. pitating factors (e.g. extreme exertion, heavy carbo-
• Muscles can be stiff and in some patients there is myotonia hydrate intake, and alcohol ingestion).
of the eyelids, tongue, thumb and forearm muscles. • Prophylaxis: beta-adrenergic blocking agents reduce the
• During attacks the serum potassium is often raised frequency and severity of attacks while measures to control
(above 5.0 mmol/l [0.5 mEq/l]), as is the urine thyrotoxicosis are being instituted. As serum potassium
excretion of potassium and the CK level. levels are normal between paralytic attacks, prophylactic
• Associated with a genetic abnormality at chromosome 17q. potassium administration is unlikely to be helpful.
Furthermore, acetazolamide is not helpful (unlike in FFP),
Diagnosis and may even exacerbate attacks of paralysis in TPP.
Low serum potassium during a paralytic attack, and
exclusion of secondary causes of hypokalemia. Barium poisoning
• A rare cause of hypokalemic paralysis.
Treatment • Most cases are due to suicidal or accidental ingestion of
• Oral potassium 0.2–0.4 mmol/kg (0.2–0.4 mEq/l), barium.
repeated at 15–30 minute intervals depending on the • The mechanism of hypokalemia is transcellular shift of K+.
response of the patient (muscle strength), EKG, and • The paralysis is treated with potassium replacement.
serum potassium level.
• If the patient is unable to swallow, or is vomiting, DIFFERENTIAL DIAGNOSIS
intravenous potassium chloride 20 mmol per 100 ml CNS
normal saline hourly, while monitoring clinical status and • Cataplexy.
serum potassium, may be necessary. • Sleep paralysis associated with narcolepsy.
• Glucose in the diluent should be avoided as it can cause • Multiple sclerosis.
a further intracellular shift of potassium and reduction in • Transient ischemic attack of the brain.
serum potassium levels. • Hyperventilation syndrome.
• Poliomyelitis.
Prophylaxis against recurrent periodic attacks
• Acetazolamide 250–750 mg/day (agent of choice), or Peripheral nerve
spironolactone 100–200 mg/day or triamterine. Guillian–Barré syndrome (see p.588).
• Acetazolamide abolishes attacks in most patients, perhaps
because of the metabolic acidosis it produces. As Neuro-muscular junction
acetazolamide lowers serum potassium, it may be • Myasthenia gravis (see p.657).
necessary in some patients to supplement potassium and • Lambert–Eaton myasthenic syndrome (see p.664).
to avoid high carbohydrate meals. • Botulism.
• Chronic acetazolamide therapy may be associated with • Diphtheria.
renal calculus (for which patients should be monitored).
Muscle
Thyrotoxic periodic paralysis (TPP) • Polymyositis.
Epidemiology • Dermatomyositis.
• The most common acquired form of periodic paralysis. • Acute inflammatory myopathy (viral/parasitic).
• Age of onset: second to fourth decades (similar to
thyrotoxicosis). Metabolic/toxic
• Gender: M>F (20:1). • Electrolyte abnormalities.
• Race: Orientals (90% of cases), Caucasians, native • Porphyria.
American Indians, Blacks, and Aborigines. • Medications (e.g. opiates).
• Family history of TPP: rare. • Alcoholism.
• Hypoglycemic disorders.
• Endocrine disorders.
Further Reading 695

INVESTIGATIONS TREATMENT
• Serum potassium. • Identify and treat the cause (e.g. correction of the
• EKG. hyperthyroid state).
• Thyroid function tests. • Replace potassium: oral potassium 0.2–0.4 mmol/kg
(0.2–0.4 mEq/l), repeated at 15–30 minute intervals
DIAGNOSIS depending on the response of the patient (muscle
• Consider the diagnosis of hypokalemic paralysis in any strength), EKG, and serum potassium level.
patient presenting with a sudden onset, areflexic, pure • Avoid precipitating factors (e.g. extreme exertion, heavy
motor weakness involving one or more limbs, without carbohydrate intake and alcohol ingestion).
alteration in level of consciousness or sphincter function.
• Serum potassium is <3.5 mmol/l (3.5 mEq/l) during an PROGNOSIS
attack (usually much lower). • Generally favorable, if appropriately diagnosed and
• EKG shows U waves, ST segment sagging, and flattening managed.
and inversion of T waves, but these changes do not • Deaths from respiratory failure and arrhythmia have been
correlate well with the severity of the hypokalemia. reported.
• Response of muscle power to provocative testing with glu-
cose, insulin, potassium, and cold: for patients whose at-
tacks are infrequent, but potentially hazardous and patients
must be carefully monitored during their performance.

Gamez J, Navarro C, Andreu AL, et al. (2001) Au- Webster G, Beynon H (1999) Weak at the knees.
FURTHER READING tosomal dominant limb-girdle muscular dystro- Lancet, 354: 1696.
phy. A large kindred with evidence of anticipa-
tion. Neurology 56: 450–454. METABOLIC AND ENDOCRINE
Pollitt C, Anderson LVB, Pogue R, et al. (2001) MYOPATHIES
X-LINKED DYSTROPHINOPATHIES
The phenotype of calpainopathy: diagnosis based Duyff RF, Van den Bosck J, Laman DM, et al.
Dubowitz V (2000) What is muscular dystrophy? J.
on a multidisciplinary approach. Neuromuscular (2000) Neuromuscular findings in thyroid dys-
R. Coll. Physicians Lond., 34: 464–468.
Disorders, 11: 287–296. function: a prospective clinical and electrodiag-
Emery AEH (1998) The muscular dystrophies.
66th/67th ENMC Sponsored International Work- nostic study. J. Neurol. Neurosurg. Psychiatry, 68:
BMJ, 317: 991–995.
shop (1999) The limb-girdle muscular dystro- 750–755.
Emery AEH (2000) Emery-Dreifuss muscular dys-
phies. 26–28 March 1999, Naarden, The Nether- Fernández R, Navarro C, Andreu AL, et al. (2000)
trophy – a 40 year retrospective. Neuromuscular
lands. Neuromuscular Disorders, 9: 436–445. A novel missense mutation (W797R) in the
Disorders, 10: 228–232.
myophosphorylase gene in Spanish patients with
Fenichel GM, Griggs RC, Kissel J, et al. (2001) A
MYOTONIC DYSTROPHY McArdle disease. Arch. Neurol., 57: 217–219.
randomized efficacy and safety trial of oxan-
Moxley RT III, Udd B, Ricker K (1998) Proximal Haller RG (2000) Treatment of McArdle disease.
drolone in the treatment of Duchenne dystrophy.
myotonic myopathy (PROMM) and other prox- Arch. Neurol., 57: 923–924.
Neurology, 56: 1075–1079.
imal myotonic syndromes. Neuromuscular Dis- Klein I, Ojamaa K (2000) Thyroid (neuro)myopa-
Flanigan KM (1999) The muscular dystrophies: up-
orders, 8: 519–520. thy. Lancet, 356: 614.
date on genetics and appropriate testing. Neurol-
Thornton CA, Ashizawa T (1999) Getting a grip on Pari G, Crerar MM, Nalbantoglu J, et al. (1999)
ogist, 5: 113–121.
the myotonic dystrophies. Neurology, 52: 12–13. Myophosphorylase gene transfer in McArdle’s
Griggs RC (2000) Techniques for ventilatory sup-
disease myoblasts in vitro. Neurology, 53:
port. Clinical equipoise. Neurology, 55: 615. POLYMYOSITIS 1352–1354.
Jay V, Vajsar J (2001) The dystrophy of Duchenne. Amato A, Barohn RJ (2000) Evaluation and treat- Preedy VR, Adachi J, Veno Y, et al. (2001) Alco-
Lancet, 357: 550–552. ment of the idiopathic inflammatory myopathies. holic skeletal muscle myopathy: definitions, fea-
Mendell JR, Buzin CH, Feng J, et al. (2001) Diag- The Neurologist, 6: 267–287. tures, contribution of neuropathy impact and di-
nosis of Duchenne dystrophy by enhanced de- Callen JP (2000) Dermatomyositis. Lancet, 355: agnosis. Eur. J. Neurol., 8: 677–687.
tection of small mutations. Neurology, 57: 53–57. Selim MH (2001) Neurologic aspects of thyroid dis-
645–650. Dalakas MC (1991) Polymyositis, dermatomyositis, ease. The Neurologist, 7: 135–146.
Nomori H, Ishihara T (2000) Pressure-controlled and inclusion-body myositis. N. Engl. J. Med., Vorgerd M, Kubisch C, Burwinkel B, et al. (1998)
ventilation via a mini-tracheostomy tube for pa- 325: 1487–1498. Mutation analysis in myophosphorylase deficiency
tients with neuromuscular disease. Neurology, 55: Dalakis MC, Illa I, Dambrosia J.M. (1993) A con- (McArdle’s disease). Ann. Neurol., 43: 326–331.
698–702. trolled trial of high dose intravenous immune Vorgerd M, Grehl T, Jäger M, et al. (2000) Crea-
globulin as treament for dermatomyositis. N.. tine therapy in myophosphorylase deficiency
FACIO-SCAPULO-HUMERAL Engl. J. Med., 329: 1993–2000. (McArdle disease). A placebo-controlled
MUSCULAR DYSTROPHY Dalakis MC (2001) Progress in inflammatory my- crossover trial. Arch. Neurol., 57: 956–963.
Ricci E, Galluzzi G, Deidda G, et al. (1999) opathies: good but not good enough. J. Neurol.
Progress in the molecular diagnosis of fa- Neurosurg. Psychiatry, 70: 569–573.
Miller FW, Leitman SF, Cronin ME (1992) Con- HYPOKALEMIC PARALYSIS
cioscapulohumeral muscular dystrophy and cor-
trolled trial of plasma exchange and leukaphere- Ahlawat SK, Sachdev A (1999) Hypokalaemic paral-
relation between the number of KpnI repeats at
sis in polymyositis and dermatomyositis. N. Engl. ysis. Postgrad. Med. J., 75: 193–197.
the 4q35 locus and clinical phenotype. Ann.
J. Med., 326: 1380–1384. Benatar (2000) Neurologic potassium channelopa-
Neurol., 45: 751–757.
thy. Q. J. Med., 93: 787–797.
Tawil R, Figlewicz DA, Griggs RC, Weiffenbach B,
INCLUSION BODY MYOSITIS Griggs RC, Ptácek LJ (1999) Mutations of sodium
and the FSH Consortium (1998) Facioscapulo-
Askanas V, Engel WK (2001) Inclusion-body myosi- channels in periodic paralysis. Can they explain
humeral dystrophy: a distinct regional myopathy
tis: newest concepts of pathogenesis and relation the disease and predict treatment? Neurology, 52:
with a novel molecular pathogenesis. Ann. Neu-
to aging and Alzheimer disease. J. Neuropathol. 1309–1310.
rol., 43: 279–282.
Exp. Neurol., 60: 1–14. Lin S-H, Lin Y, Halperin ML (2001) Hypokalaemia
LIMB-GIRDLE MUSCULAR DYSTROPHIES Dalakis MC, Koffman B, Fujii M, et al. (2001) A and paralysis. Q. J. Med., 94: 133–139.
Bushby KMD (1999) Making sense of the limb-gir- controlled study of intravenous immunoglobulin Ptácek LJ (1998) The familial periodic paralyses and
dle muscular dystrophies. Brain, 122: 1403–1420. combined with prednisone in the treatment of nondystrophic myotonias. Am. J. Med., 104:
Chou F-L, Angelini C, Daentl D, et al. (1999) Cal- IBM. Neurology, 56: 323–327. 58–70.
pain III mutation analysis of a heterogeneous Nakano S, Shinde A, Kawashima S, et al. (2001) In- Sternberg D, Maisonobe T, Jurkat-Rott ET (2001)
limb-girdle muscular dystrophy population. Neu- clusion body myositis. Expression of extracellular Hypokalaemic periodic paralysis type X caused by
rology, 52: 1015–1020. signal-regulated kinase and its substrate. Neurol- mutations at codon 672 in the muscle sodium
ogy 56: 87–93. channel gene SCN4A. Brain, 124: 1091–1099.
696

Index
Page numbers in bold type indicate major anemia 222 atrophy
discussions of topics anencephaly 139 lobar 414–16
aneurysms 251 multiple systems (MSA) 263, 435–7,
atrial septal 211 474, 576
A cavernous sinus syndrome 354, 355 olivopontocerebellar (OPCA) 435
abducens nerve 18, 500 saccular 250 see also muscular atrophy
neuropathy 499–504 angiitis attention, assessment of 14
abetalipoproteinemia 531 allergic 597 attention deficit disorder (ADD) 131,
abscess granulomatous 228, 352 133
intracranial 284–7 hypersensitivity 227, 228 auditory nerve 19
spinal epidural 555–7 necrotizing 227, 228 autonomic neuropathy 573–8, 582, 590,
absences (petit mal epilepsy) 68 see also giant cell arteritis; vasculitis 610
accessory nerve see spinal accessory nerve anosmia 482 axillary nerve 620
accommodation reflex 575, 576 anterior compartment syndrome 651, 652 axillary neuropathy 620–1
acid maltase deficiency 688, 689, 691, anterior interosseous syndrome 628, 629 axonal degeneration 36, 582–3
692 anterior ischemic optic neuropathy axonopathy 580
acoustic neuroma 108, 383–4 (AION) 487–8
acquired immunodeficiency syndrome anterior spinal artery syndrome 560
(AIDS) 323, 324 anterior tarsal tunnel syndrome 651, 652 B
dementia complex 301 antiphospholipid antibodies (APAs) 218 back pain 550–1, 552
myelopathy 564 antiphospholipid syndrome 218–24 see also slipped disc
see also human immunodeficiency virus antithrombin III 223–3 bacterial infections
(HIV) anxiety 589 botulism 123
acromegaly 690, 692 aortic valve sclerosis/calcification 211 intracranial abscess 284–7
acute disseminated encephalomyelitis arachnoid cysts 570 leprosy 544, 604–6
(ADEM) 337–9 arachnoiditis, spinal 557–8 Lyme disease 316–17
acute idiopathic facial paralysis (Bell’s Argyll Robertson (AR) pupils 314, 576 meningitis 273–8
palsy) 515–16 Arnold–Chiari malformation 136–9, 530 myositis 682
acute inflammatory demyelinating see also Chiari malformations neurosyphilis 312–15
polyradiculoneuropathy arteriosclerosis 202 tetanus 287–8
(AIDP) 588–92, 602, 603 arteriovenous fistula 246 tuberculous meningo-encephalitis
acute motor axonal neuropathy (AMAN) arteriovenous malformation (AVM) 153, 281–3
590 154, 156, 245–9, 251 Whipple’s disease 289–90
acute motor-sensory axonal neuropathy dural 564 ballism 123
(AMSAN) 590 spinal 246, 443, 530 Baltic myoclonus 88, 89
acute suppurative myositis 682 arteritis 299 Bassen–Kornzweig disease 531
acute transverse myelitis 561–3 giant cell (GCA) 105, 234–7, 597 Becker’s muscular dystrophy (BMD) 668,
Addison’s disease 689, 690, 692 temporal (TA) 237 670–2
adenomas, pituitary 379 arylsulfatase A (ASA) deficiency 169 Behçet’s disease 597
growth hormone-producing 380 asterixis 127, 454 Bell’s palsy 515–16
macroadenomas 382 astrocytoma 364, 368, 370, 371 benign paroxysmal positional vertigo
microadenomas 382 anaplastic 364, 370, 371 110
adrenoleukodystrophy (ALD) 166–8, spinal cord 570 berry aneurysms 250
530 ataxia 127, 165, 436, 530 bilateral vestibular failure 108
adrenomyeloneuropathy (AMN) 166–8, autosomal dominant cerebellar 437–41 Binswanger’s disease 262
530, 564 early onset, of unknown etiology bladder dysfunction 578
adult-onset globoid cell leukodystrophy 442–3 multiple sclerosis 342, 348
(Krabbe disease) 171–2, 530 Friedreich's 441–4 normal pressure hydrocephalus 473–4
AIDS see acquired immunodeficiency multiple sclerosis 349 bladder function 574
syndrome progressive myoclonic 88 blindness see visual disturbances
akinetic mutism 54 with isolated vitamin E deficiency 442 blood pressure 574
akinetic seizures 68–9 ataxia telangiectasia 358, 444–6 see also hypertension
alcohol withdrawal 120 atherogenesis 204 Borrelia burgdorferi 316
allergic granulomatosis 597 atheroma 204 botulism 659
Alzheimer’s disease (AD) 263, 407–14, atherosclerosis 202, 204 bradyarrhythmia
416, 431, 474 atherosclerotic ischemic stroke 202–8 Parkinson’s disease 422
ammonia, liver failure and 454 plaque rupture 205 syncope 59
amnesia, transient global 187 atherothrombosis 205, 206 brain abscess 284–7
amyloid angiopathy 238 see also thromboembolism; thrombosis brain death 51–2, 54, 451
amyloidosis 544, 607, 608, 609 athetosis 123 brain imaging see imaging
amyotrophic lateral sclerosis (ALS) 534, atonic seizures 68–9 brain tumors 357–63
536, 537 atrial fibrillation 210–11 metastases 358, 396, 397, 398
anal reflex 22 atrial septal aneurysm 211 see also specific tumors
Index 697

brainstem auditory evoked potentials cholestanolosis 442 cryoglobulinemia 607, 608, 609
(BAEPs) 40–1 cholesterol reduction, stroke and 200 Cryptococcus neoformans meningitis
brainstem myoclonus 127 cholinergic crisis 664 318–20
breath-holding attacks 71 chorea 122–4, 127, 418, 420 cysticercosis 326–9
bulbar palsy 524, 528, 536, 538 choriocarcinoma 389 intraparenchymal 327
progressive 534, 537 choroid plexus papilloma 366 racemose 327
bulbocavernosus reflex 21 chronic inflammatory demyelinating cysts
polyneuropathy (CIDP) 590, 593–5, arachnoid 570
602, 603, 608 colloid 366
C chronic paroxysmal hemicrania (CPH) cytomegalovirus (CMV) 589
calcaneal neuropathy 647, 648, 650 104, 105
calpainopathy 676 chronic progressive external
Campylobacter jejuni 589 ophthalmoplegia (CPEO) 164, 165 D
CANOMAD syndrome 607 Churg–Strauss syndrome 597 Dandy–Walker syndrome 138, 140
carbon monoxide poisoning 450, 452 cigarette smoking, stroke and 200 deafness 518
carcinoma cingulate herniation 44 debranching enzyme system deficiency
choriocarcinoma 389 cisternography 26 688, 690, 691
embryonal 388 clinical history see neurologic history degenerative diseases
naevoid basal cell 358 clonus 127 Alzheimer’s disease (AD) 407–14
cardioembolic stroke 209–15 Clostridium tetani 287 ataxia telangiectasia 444–6
carnitine-O-palmitoyltransferase deficiency cluster headache 103–5 autosomal dominant cerebellar ataxias
688–9, 690, 691, 692 cluster-tic syndrome 104 437–41
carotid artery dissection 224–6 coagulation disorders 222–3 axonal degeneration 36, 582–3
carpal tunnel syndrome (CTS) 630–2 colloid cyst 366 diffuse Lewy body disease (DLBD)
cataplexy 57, 58, 175 color vision 15 430–1
cauda equina syndrome 141, 474 optic neuropathy and 484 Friedreich’s ataxia 441–4
cavernous malformation 246 see also visual disturbances frontotemporal dementia (FTD)
cavernous sinus syndrome 354–5 coma 44–50, 54 414–16
bilateral 355 communication, assessment of 15 hepatolenticular degeneration 157–61
cavernous sinus thrombosis 258 compartment syndrome 652 Huntington’s disease (HD) 417–20
cavernous sinus/superior orbital fissure anterior 651, 652 multiple systems atrophy (MSA) 435–7
506, 508 lateral 651, 652 Parkinson’s disease (PD) 420–30
central cord syndrome 568 compound muscle action potential progressive supranuclear palsy (PSP)
central motor conduction 37 (CMAP) 35, 37 432–4
central pontine myelinolysis 457–9 compression neuropathies 612 subacute combined degeneration of the
cerebellar herniation 136 carpal tunnel syndrome 630–2 spinal cord 443, 531
cerebral circulation imaging see imaging femoral neuropathy 640 Déjérine–Sottas syndrome 585
cerebral infarction 192, 202, 299 gluteal neuropathy 644 delirium 44, 48
cerebral ischemia 256 lateral cutaneous nerve of the thigh delusions, olfactory 482
cerebral venous thrombosis 257–60 neuropathy 637 dementia 175, 264, 411–14, 418–19
cerebrospinal fluid (CSF) median neuropathy 628 Alzheimer’s disease 263, 407–14
circulation disorders 468–80 peroneal neuropathy 651, 652 familial 412
hydrocephalus 468–72 plantar interdigital neuropathy 648, frontotemporal dementia (FTD)
idiopathic intracranial hypertension 649 414–16
(IIH) 477–80 posterior cutaneous nerve of the thigh HIV-associated 301–3, 308
normal pressure hydrocephalus neuropathy 639 Huntington’s disease 418
(NPH) 473–6 pudendal neuropathy 655 Lewy body type 263
circulation physiology 468 radial neuropathy 624 normal pressure hydrocephalus 473
examination 31–3 saphenous nerve 640 Parkinson’s disease 263, 423
complications 32 sciatic neuropathy 645 pseudo-dementia 412
indications/contraindications 32 ulnar neuropathy 633 vascular 261–5, 416, 474
normal values 32–3 computerized tomography (CT) 23–4 demyelination
technique 31 CT angiography 28 acute inflammatory demyelinating
ceruloplasmin deficiency 159 CT cisternography 26 polyradiculoneuropathy (AIDP)
cervical dystonia 115 CT myelography 29 588–92, 602, 603
cervical spondylosis 544, 545–9 see also specific conditions chronic inflammatory demyelinating
cervical spondylotic myelopathy/radicu- confusion 44, 48 polyneuropathy (CIDP) 590, 593–5,
lopathy 545–9 consciousness 44 602, 603, 608
Charcot joints 314 contrast angiography 26–8 delayed post-anoxic 450
Charcot–Marie–Tooth (CMT) disease coordination assessment 20, 22 multifocal demyelinating
584, 585–6, 587 corneal response 18 leukoencephalomyelitis 299
type 1 585 coronary heart disease 211 segmental 36–7
type 2 585, 586 cortical myoclonus 126 depression 416, 420
Chiari malformations cranial dystonia 115 Alzheimer’s disease 414
type I 136, 138, 541, 542, 544 cranial nerves 15–19 multiple sclerosis 349
type II 136, 138–9, 541, 544 palsy 354 Parkinson’s disease 429
type III 136, 541 see also specific nerves dermatomyositis 402, 403, 684–7
type IV 137 cranial neuropathies see neuropathy; childhood 685
chickenpox 299 specific nerves dermoid tumors 390
childhood craniopharyngioma 377–9 diabetic amyotrophy 612
dermatomyositis 685 Creutzfeldt–Jakob disease (CJD) 330–5, diabetic neuropathy 544, 610–13
juvenile myasthenia 659 416, 440 diabetic radiculo-plexopathy 612, 641
juvenile myoclonic epilepsy (JME) amyotrophic form 332 diastematomyelia, rachischisis 141, 142
86–7 Brownell–Oppenheimer variant 331 diencephalon, herniation 45
migraine 98 Heidenhain variant 331 diffuse Lewy body disease (DLBD)
chloride channelopathies 680 new variant CJD 332, 334 430–1
698 Index

diplopia 501–2 encephalopathy (continued) fasciculations 127


disseminated intravascular coagulation progressive myoclonic 88, 126 febrile convulsions 70
224, 403 radiation-induced 565–6 femoral nerve 640
dizziness see vertigo static myoclonic 125–6 femoral neuropathy 640–1, 642
dorsal scapular nerve 615 subcortical arteriosclerotic (SAE) 474 fibrillation potentials 38
neuropathy 615–16 toxic 126, 455 Foster–Kennedy syndrome 493
double vision, multiple sclerosis 342 encephalotrigeminal vascular syndrome Friedreich’s ataxia 441–4
see also visual disturbances 151–2 frontotemporal dementia (FTD) 414–16
Down’s syndrome 410 endocarditis fundoscopy 16
driving, epilepsy and 76 infective 215–18 fungal infections
drop attacks 62, 71 marantic 403 meningitis 276, 318–20
Duchenne’s muscular dystrophy (DMD) enhanced physiologic tremor (EPT) 120 mucormycosis of the nervous system
668, 670–2 Enterobacter 284 321–3
dysferlinopathy 676 entrapment neuropathies 612
dyskinesias 129–30 lateral plantar neuropathy 648
Parkinson’s disease 423, 428–9 medial plantar neuropathy 647 G
paroxysmal 129, 130 median neuropathy 628 gait
tardive 129, 130 radial neuropathy 624 assessment 22
dysosmia 482 saphenous nerve 640 hereditary spastic paraparesis and 530
dysraphism 139–42 tibial neuropathy 647 multiple sclerosis and 342
dysthyroid eye disease 690, 691, 692 ependymoma 364, 365–6, 368–9, 370, Parkinson’s disease and 422
dystonia 113–18, 121, 123, 127, 175 371 slipped disc and 550
cervical 115 spinal cord 570 galactosylceramide ß-galactosidase
cranial 115 epidermal nevus syndrome 152 deficiency 171
dopa-responsive 531 epidermoid tumors 389 gastroparesis 578
laryngeal 115 epidural hemorrhage germ cell tumors of the CNS 388–92
myoclonic 116 acute 178 germinoma 388, 392
paroxysmal 116 spinal epidural hematoma 554–5 Gerstmann–Straussler–Scheinker syndrome
epilepsy 57, 65–82, 70–1, 83, 86, 88 416
classification 65–6 giant cell arteritis (GCA) 105, 234–7,
E juvenile myoclonic epilepsy (JME) (Janz 597
eclampsia 76 syndrome) 86–7 Glasgow coma scale 47
edema, cerebral 358 myoclonic epilepsy with ragged-red glaucoma 105
cytotoxic 358 fibers (MERRF) syndrome 164, 165 glioblastoma 364, 368, 370, 371
vasogenic 358, 362 progressive myoclonic epilepsies (PME) glioblastoma multiforme 364
electroencephalography (EEG) 33–5 86, 87–90, 126 glioma 358, 364–71
definition 33 special forms of 70 optic nerve 148, 372–3
interpretation 35 status epilepticus (SE) 82–5 gliomatosis cerebri 366
recordings 34 stroke and 201 glossopharyngeal nerve 19
role 35 see also seizures palsy 522
technique 33–4 epileptic myoclonus 124–5 glossopharyngeal neuralgia 522
see also specific conditions episodic paroxysmal hemicrania (EPH) glossopharyngeal neuropathy 520–2
electromyography (EEG) 38–40 104 gluteal nerve
abnormal muscle activity 38–40 erythrocyte abnormalities 222 inferior 643, 644
normal muscle activity 38 essential thrombocythemia 222 superior 643–4
see also specific conditions essential tremor (ET) 119–22, 423 gluteal neuropathy 643–4
embolism 186, 209 evoked potentials 40–2 glycogen metabolism disorders 688,
cardiogenic 192, 209–15 brainstem auditory evoked potentials 689–90, 691, 692
pulmonary, Guillain–Barré syndrome (BAEPs) 40–1 GM2 gangliosidoses 172
592 somatosensory evoked potentials Gottron’s papules 685, 686
spinal cord infarction 559 (SSEPs) 42 Gradenigo’s syndrome 506, 507
systemic 216 visual evoked potentials (VEPs) 40 grand mal epilepsy 69
thromboembolism 190, 206, 207 see also specific conditions granuloma
Emery–Dreifuss muscular dystrophy examination see neurologic examination non-caseating 352
(EDMD) 670 exophthalmic ophthalmoplegia 690, 691, spinal 531
empty sella syndrome 380–1 692 granulomatosis
encephalitis 324 allergic 597
subacute and chronic 279–80 Wegener’s 597
toxoplasmic 323–6 F granulomatous angiitis 228, 352
tuberculous meningo-encephalitis F-waves 37 Grave’s disease 659
281–3 Fabry’s disease 544, 584 growth hormone excess 380
viral 292–5, 338 facial nerve 19 Guillain–Barré syndrome (GBS) 588–92,
encephalomyelitis 300 facial neuropathy 511–14 596, 598
acute disseminated (ADEM) 337–9 facial sensation 18
limbic 402 facio-scapulo-humeral muscular dystrophy
postinfectious 293, 337–9 (FSHD) 672–4 H
varicella-zoster virus 299–300 fainting see syncope H-reflex 37
encephalomyelopathy, mitochondrial 531 familial adult myoclonic epilepsy (FAME) Haemophilus influenzae 273–4, 278
encephalopathy 88, 126, 134, 258 86 Hallervorden–Spatz disease 162
delayed post-anoxic 450, 451 familial amyloid polyneuropathy 584 hallucinations
hepatic (HE) 452–7 familial hemiplegic migraine (FHM) 96, hypnagogic 57
hypertensive 265–7 97 olfactory 482
hyponatremic 458–9 familial periodic paralysis (FPP) 693–4 headache 91–105, 236
hypoxic 449–52 familial spastic paraparesis 529 classification 91
metabolic 126, 454 family history 12–13 cluster headache 103–5
porto-systemic 452, 453 fasciculation potentials 38–9 history/examination 91–2, 94
Index 699

headache (continued) Horner’s syndrome 16, 225, 494–6 imaging, brain (continued)
idiopathic intracranial hypertension cavernous sinus syndrome 354 single photon emission computed
477 syringomyelia 542 tomography (SPECT) 26
muscle contraction/tension-type human immunodeficiency virus (HIV) cerebral circulation 26–8
101–3 589 contract angiography 26–8
subarachnoid hemorrhage 250, 252, concurrent infection with neurosyphilis CT angiography 28
254 315 magnetic resonance angiography
thunderclap headache 99, 258 HIV myelopathy 564–5 (MRA) 28
see also migraine HIV neuropathy 601–4 ultrasound 28
hearing loss 518 HIV-associated cognitive/motor spine 29–30
heart rate 574, 577 complex (HIV-CMC) (HIV dementia) CT myelography 29
heliotrope rash 685, 686 301–3, 308 magnetic resonance imaging 30
hemangioblastoma 152 Huntington’s disease (HD) 416, 417–20 myelography 29
capillary 393 juvenile Westphal variant 418 see also specific conditions
spinal cord 570 hydrocephalus 468–72 inclusion body myositis (IBM) 687–8
hemangioma 152 acoustic neuroma and 383 infantile paralysis 309–11
hematoma communicating 468 infantile spasms 69
spinal epidural 554–5 ex-vacuo 469 infarction
subdural 177–80 normal pressure hydrocephalus (NPH) cerebral 192, 202
hematomyelia 544 469, 473–6 diffuse white matter 262
hemifacial spasm 127 obstructive 468, 474 migrainous 98
hemiplegic migraine 97 rachischisis and 142 pituitary apoplexy 268
familial (FHM) 96 subarachnoid hemorrhage and 255–6 spinal cord 558–61
hemorrhage hydromyelia 541 vascular dementia and 262
epidural 178 hygroma, subdural 178 venous 260
intracerebral 192, 198, 299 hyperglycemic neuropathy 610 see also stroke
primary (PICH) 238–44 hyperkalemia 690 infective endocarditis 215–18
subarachnoid 178, 192, 198 hyperparathyroidism 690, 692 inflammatory disorders
subdural hematoma 177–80 hyperprolactinemia 380, 381 acute disseminated encephalomyelitis
vascular dementia and 262 hypersensitivity angiitis 227, 228 (ADEM) 337–9
see also stroke hypertension acute inflammatory demyelinating
hemorrhagic stroke idiopathic intracranial 477–80 polyradiculoneuropathy (AIDP)
intracerebral hemorrhage 192, 198, primary intracerebral hemorrhage and 588–92, 602, 603
299 238 cavernous sinus syndrome 354–5
primary intracerebral hemorrhage stroke and 200 chronic inflammatory demyelinating
(PICH) 238–44 hypertensive encephalopathy 265–7 polyneuropathy (CIDP) 590, 593–5,
subarachnoid hemorrhage 178, 192, hyperthyroidism 659 602, 603, 608
198, 249–56 hyperviscosity 222 multiple sclerosis (MS) 340–9
heparin-induced thrombocytopenia 224 hypnic headache 104 neurosarcoidosis 350–3
hepatic encephalopathy (HE) 452–7 hypoadrenalism 380, 689, 690, 692 see also specific disorders
hepatolenticular degeneration 157–61 hypoglossal nerve 19 intracerebral hemorrhage 192, 198
hereditary hemorrhagic telangiectasia palsy 527–8 intracranial abscess 284–7
(HHT) 153–6 hypoglossal neuropathy 526–8 intracranial pressure
hereditary neuralgic amyotrophy 587, hypoglycemia 62 monitoring 470, 475–6
596 hypogonadism 380 raised 359, 362, 384
hereditary neuropathies 584, 605 hypokalemia 690, 693 hydrocephalus 469
Guillain–Barré syndrome (GBS) hypokalemic paralysis 693–5 papilledema 492
588–92, 596, 598 familial periodic paralysis (FPP) 693–4 intravenous digital subtraction
hereditary motor and sensory thyrotoxic periodic paralysis (TPP) 694 angiography (IV DSA) 26–7
neuropathy (HMSN) 584, 585–6 hyponatremia 256, 592 ischemic stroke
type 1 585, 586 hypopituitarism 380 atherosclerotic 202–8, 221–2
type 2 585, 586 hypoprolactinemia 380 lipohyalinosis/microatheroma 204, 238
hereditary neuropathy with liability to hyposmia 482 cardioembolic 209–15
pressure palsies (HNPP) 587–8 hypothyroid myopathy 690, 691, 692 thrombophilic 218-24
hereditary spastic paraparesis (HSP) hypothyroidism 380 isotope cisternography 26
529–31, 538, 564 hypoxia 449, 450–1
heredopathia atactica polyneuritiformis spinal cord infarction 559
584 hypoxic encephalopathy 449–52 J
herniation hysteria 589 Jansky–Bielschowsky disease 89
cerebellar 136 Janz syndrome 86–7
cingulate 44 jaundice, neonatal 175
diencephalon 45 I jaw jerk 19
uncal 46 idiopathic intracranial hypertension (IIH) JC virus (JCV) 307
herpes simplex virus encephalitis (HSE) 477–80 jugular foramen syndrome 524
295–8 IgA paraproteinemic neuropathy syn- juvenile myasthenia 659
herpes zoster infection 599–601 dromes 607, 609 juvenile myoclonic epilepsy (JME) 86–7
facial paresis 513 IgM paraproteinemic neuropathy syn-
varicella-zoster virus encephalomyelitis dromes 607, 608, 609
299–300 imaging K
hexosaminidase A deficiency 172, 442, brain 23–6 Kayser–Fleischer (K–F) ring 158
538 computerized tomography (CT) Kearns–Sayre syndrome (KSS) 164
hexosaminidase B deficiency 538 23–4 Kennedy’s syndrome 532, 533, 537
history see neurologic history magnetic resonance imaging (MRI) Korsakoff’s psychosis 460, 461, 462
HIV see human immunodeficiency virus 24–6 Krabbe’s disease 171–2, 530
Hoffmann–Tinel sign 648 positron emission tomography (PET) Kuf’s disease 89
Holmes–Aidie syndrome 497–8 26 Kugelberg–Welander disease 532, 538
700 Index

L magnetic resonance imaging (MRI) mitral valve (continued)


Lafora body disease 88, 89 (continued) sclerosis/calcification 211
Lambert–Eaton myasthenic syndrome see also specific conditions Miyoshi myopathy 586
(LEMS) 401, 402, 403, 659, 664–7 malignant angioendotheliosis 222 Mollaret’s meningitis 291
laryngeal dystonia 115 malignant hyperthermia 689, 690, 691, monoclonal gammopathy of uncertain
late waves 37 692 significance (MGUS) 606, 607, 609
late-onset GM2 gangliosidosis 172–3 Marcus Gunn pupil 575 mononeuritis multiplex 602, 603, 604
lateral compartment syndrome 651, 652 mass lesions see tumors mononeuropathies
lateral cutaneous nerve of the thigh 637 masticatory muscles 19 axillary neuropathy 620–1
neuropathy 637–8 measles virus 304 carpal tunnel syndrome (CTS) 630–2
lateral plantar neuropathy 648 medial plantar neuropathy 647–8, 649 dorsal scapular nerve neuropathy
lathyrism 564 median nerve 627 615–16
Leber’s hereditary optic neuropathy median neuropathy 627–9 femoral neuropathy 640–1, 642
(LHON) 489, 491–2 carpal tunnel syndrome 630–2 focal limb mononeuropathies 612
Leigh’s disease 165 medulloblastoma 366–7 gluteal neuropathy 643–4
leprosy 544, 604–6 Meige syndrome 115 lateral cutaneous nerve of the thigh
lepromatous 604 MELAS (mitochondrial neuropathy 637–8
multibacillary 604, 606 encephalomyopathy, lactic acidosis, and long thoracic neuropathy 616–17
paucibacillary 604, 606 stroke-like episodes) syndrome 163, median neuropathy 627–9
tuberculoid 604 164, 165 multiple 544, 587, 598
leukemia 222 memory, assessment of 15 musculocutaneous neuropathy 621–2
leukocyte abnormalities 222 Ménière’s disease 109, 111–12 obturator neuropathy 642–3
leukodystrophy 167, 442, 530 meningioma 374–6 peroneal neuropathy 650–3
adrenoleukodystrophy (ALD) 166–8 olfactory groove 375 posterior cutaneous nerve of the thigh
adult-onset globoid cell (Krabbe’s parasagittal fronto-parietal 375 neuropathy 639
disease) 171–2, 530 sphenoidal wing 375, 376 pudendal neuropathy 655–6
metachromatic (MLD) 169–70, 530 spinal cord 570 radial neuropathy 623–6
leukoencephalitis, acute hemorrhagic 338 tuberculum sellae 375, 376 sciatic neuropathy 644–6, 652
leukoencephalomyelitis, multifocal meningitis suprascapular neuropathy 618–19
demyelinating 299 acute aseptic (viral) 276, 291–2 sural neuropathy 654–5
leukoencephalopathy, progressive Mollaret’s meningitis 291 tibial neuropathy 647–50
multifocal (PML) 307–9 acute pyogenic (bacterial) 273–8 ulnar neuropathy 632–6
Lewy body type dementia 263, 436 acute syphilitic 313 Morton’s metatarsalgia 648, 649, 650
diffuse Lewy body disease (DLBD) fungal 276, 318–20 Morton’s neuralgia 649
430–1 malignant 279 motor nerve conduction studies 35
light reflex 574, 576 protozoal 276 motor neuron disease (MND) 138, 531,
limb-girdle muscular dystrophies (LGMD) subacute and chronic 279–80 534–40, 544, 636, 652
674–7 tuberculous 276 familial 537
limbic encephalomyelitis 402 serous 281 movement disorders 71, 113–35
lipid metabolism disorders 688–9, 690, meningocele 140, 141, 142 chorea 122–4
691, 692 meningoencephalitis 338, 352 dyskinesias 129–30
lipohyalinosis 204, 238 tuberculous 281–3 dystonia 113–18
lipoma, spinal cord 570 meningoencephalocele 140, 141 essential tremor (ET) 119–22
Lisch nodules 148 meningomyelocele Huntington’s disease 418
Listeria monocytogenes meningitis 274 Arnold–Chiari malformation 136–9, myoclonus 124–8
liver disease 159, 453, 454 530 neuroleptic malignant syndrome (NMS)
lobar atrophy 414–16 rachischisis 141, 142 133–5
locked-in syndrome 54, 55–6 menstrual migraine 98, 100–1 Tourette syndrome (TS) 130–3
long thoracic nerve 616 mental state assessment 14–15 mucormycosis of the nervous system
long thoracic neuropathy 616–17 mesial temporal sclerosis (MTS) 68 321–3
Louis–Bar syndrome 444–6 metachromatic leukodystrophy (MLD) multi-sensory dizziness/disequilibrium
lower limb examination 21–2 169–70, 530 108, 109
lumbar puncture 31–3 metastatic lesions 396–400 multifocal demyelinating
complications 32 brain 358, 396, 397, 398, 400 leukoencephalomyelitis 299
indications/contraindications 32 meningeal 396, 397, 398, 399, 400 multifocal motor neuropathy 608
normal values 32–3 skull/dural 396, 397 with conduction block 538, 593
technique 31 spinal cord 567, 570 multifocal neurologic syndrome 343
lumbar radiculopathy 641, 642 microatheroma 204 multinucleated giant cell myelitis 564
lumbar spondylosis 552 migraine 71, 95–101, 104–5, 108, 109 multiple mononeuropathy 544, 587, 598
lumbosacral plexopathy 641 aura 71, 96, 97, 98, 187 multiple myeloma 606, 608, 609
lumbosacral polyradiculopathy 141 childhood 98 multiple sclerosis (MS) 108, 340–9, 443,
lupus anticoagulant (LA) 220, 221 familial hemiplegic (FHM) 96, 97 458, 492
Lyme disease 316–17 hemiplegic 97 acute transverse myelitis and 563
lymphoma 385–8, 564 menstrual migraine 98, 100–1 optic neuritis and 489, 491
ophthalmoplegic 97 multiple systems atrophy (MSA) 263,
vertebrobasilar 97 435–7, 474, 576
M migrainous neuralgia 103–5 muscle carnitine deficiency 688, 690, 691
McArdle’s disease 688, 689, 691, 692 Miller Fisher syndrome (MFS) 590 muscle contraction-type headache 101–3
Machado–Joseph disease (MJD) 438–9 mini-mental state examination 14, 15, muscle disorders
macrocytosis 465 411 dermatomyositis 684–7
magnesium depletion 460 miosis 496 facio-scapulo-humeral muscular
magnetic resonance angiography (MRA) mitochondrial DNA (mtDNA) 162–3 dystrophy (FSHD) 672–4
28 mitochondrial (oxidative phosphorylation) hypokalemic paralysis 693–5
magnetic resonance imaging (MRI) diseases 89, 162–6, 531 inclusion body myositis (IBM) 687–8
brain 24–6 mitral valve limb-girdle muscular dystrophies
spine 30 prolapse 211 (LGMD) 674–7
Index 701

muscle disorders (continued) myotonia congenita 680 neuropathy (continued)


myotonic dystrophy 678–81 myotonic dystrophy 678–81 facial 511–14
polymyositis 681–4 congenital 680 femoral 640–1, 642
X-linked dystrophinopathies 668–72 proximal myotonic myopathy glossopharyngeal 520–2
see also muscular atrophy; myopathy (PROMM) 680 gluteal 643–4
muscle power 20, 21 type 2 680 hereditary see hereditary neuropathies
muscle tone 20, 21 HIV neuropathy 601–4
muscular atrophy Holmes–Aidie syndrome 497–8
peroneal 584 N Horner’s syndrome 494–6
progressive 534, 537 nevoid basal cell carcinoma 358 hyperglycemic 610
post-polio 311 narcolepsy 56–8 hypoglossal 526–8
spinal (SMA) 532–4, 536, 537–8, 626, necrotizing angiitis 227, 228 lateral cutaneous nerve of the thigh
636, 652 Neisseria meningitidis 273–4 637–8
musculocutaneous nerve 621 neonatal myasthenia 659, 664 long thoracic 616–17
musculocutaneous neuropathy 621–2 nephrotic syndrome 224 median 627–9
myasthenia gravis (MG) 657–64 nerve conduction studies 35–7 multifocal motor neuropathy 608
juvenile myasthenia 659 central motor conduction 37 with conduction block 538, 593
neonatal myasthenia 659, 664 late waves 37 multiple mononeuropathy 544, 587,
ocular myasthenia 659, 663 motor nerves 35 598
penicillamine-induced myasthenia 659 pathologies 36–7 musculocutaneous 621–2
myasthenic crisis 664 repetitive stimulation studies 37 obturator 642–3
Mycobacterium bovis 281 sensory nerves 35–6 ocular motor neuropathies 499–504
Mycobacterium leprae 604 see also specific conditions olfactory 481–2
Mycobacterium tuberculosis 281 neuralgia optic 344, 483–6, 494
mydriasis 498 glossopharyngeal 522 anterior ischemic (AION) 487–8
myelinopathy 580 migrainous 103–5 Leber’s hereditary (LHON) 489,
myelitis 324 Morton’s 649 491–2
acute ascending necrotizing 562 post-herpetic 601 papilledema 492–4
acute transverse 561–3 trigeminal 104, 509–11 paraneoplastic syndromes 402–3
multinucleated giant cell 564 neuralgic amyotrophy 595–7, 629 paraproteinemic 606–9
myelography 29 hereditary 587, 596 peripheral 579–83
myeloma neuroborreliosis 316–17 peroneal 650–3
multiple 606, 608, 609 neurofibrillary tangles 408 plantar 648, 649
osteosclerotic 607, 608, 609 neurofibromas 148 lateral 648
myeloneuropathy 465, 466 spinal cord 570 medial 647–8, 649
myelopathy 562 neurofibromatosis (NF) 147–50, 358, plantar interdigital 648, 649, 650
cervical 474 605 posterior cutaneous nerve of the thigh
HIV myelopathy 564–5 type 1 (NF-1) 147–8, 150 639
paraneoplastic syndromes 402 type 2 (NF-2) 147, 148, 150 pudendal 655–6
radiation 561, 564, 565–6 neuroleptic malignant syndrome (NMS) radial 623–6
spinal cord tumors and 568 133–5 sciatic 644–6, 652
spondylotic 636 neurologic examination 13–23 spinal accessory nerve 525–6
see also myelitis; spinal cord cranial nerves 15–19 suprascapular 618–19
myeloradiculitis 282 general assessment 13–14 sural 654–5
myocardial contusion 212 lower limbs 21–2 syncope and 59
myoclonic dystonia 116 mental state (higher mental function) tibial 647–50
myoclonic epilepsy with ragged-red fibers 14–15 trigeminal 505–8
(MERRF) syndrome 164, 165 spine 15 ulnar 632–6
myoclonic seizures 68, 124–5 upper limbs 20–1 vagus nerve 523–4
myoclonus 123, 124–8, 290 neurologic history 11–13 vasculitic 544, 597–8
Baltic 88, 89 family history 12–13 vestibular-cochlear 517–20
brainstem 127 medications 12 see also polyneuropathy
cortical 126 systems review 12 neurosarcoidosis 350–3
epileptic 124–5 neuroma neurosyphilis 312–15
nocturnal 127 acoustic 108, 383–4 asymptomatic 312
myoglobinuria 691 plexiform 148 concurrent HIV infection 315
myokymia 39 neuromuscular junction disorders 40, 590 neurovascular syndromes 181–269
myopathy 40, 403 Lambert–Eaton myasthenic syndrome antiphospholipid syndrome 218–24
distal 652, 680 (LEMS) 401, 402, 403, 659, 664–7 arteriovenous malformation (AVM)
metabolic and endocrine myopathies myasthenia gravis (MG) 657–64 153, 154, 156, 245–9, 251
688–92 neuromyotonia 659 central nervous system vasculitis
hypothyroid 690, 691, 692 neuronal ceroid lipofuscinosis 89 227–33
steroid 689, 690, 692 neuronopathy 37, 580 cerebral venous thrombosis 257–60
thyrotoxic 689, 690, 691, 692 neuropathy 165, 581 classification 181–3
Miyoshi 586 acute idiopathic facial paralysis (Bell’s dissection of the carotid and vertebral
proximal 644 palsy) 515–16 arteries 224–7
see also muscle disorders; muscular acute motor axonal neuropathy giant cell arteritis (GCA) 105, 234–7
atrophy (AMAN) 590 hypertensive encephalopathy 265–7
myositis acute motor-sensory axonal neuropathy infective endocarditis 215–18
bacterial 682 (AMSAN) 590 pituitary apoplexy 268–9
dermatomyositis 402, 403, 684–7 amyloid 608, 609 primary intracerebral hemorrhage
inclusion body (IBM) 687–8 autonomic 573–8, 582, 590, 610 (PICH) 238–44
parasitic 682 axillary 620–1 stroke 192–201
polymyositis 402, 403, 681–4 calcaneal 647, 648, 650 atherosclerotic ischemic stroke
viral 682 diabetic 544, 610–13 202–8, 221–2
myotonia 39, 678, 680, 681, 691 dorsal scapular nerve 615–16 cardioembolic stroke 209–15
702 Index

neurovascular syndromes (continued) palsy (continued) poliomyelitis (infantile paralysis)


subarachnoid hemorrhage (SAH) 178, cranial nerves 354 (continued)
192, 198, 249–56 glossopharyngeal nerve 522 post-polio syndrome 311
transient ischemic attacks (TIA) 70, hypoglossal nerve 527–8 polyarteritis nodosa 597
178, 186–92 progressive supranuclear (PSP) 263, polycythemia rubra vera 222
vascular dementia 261–5 432–4 polymyalgia rheumatica (PMR) 236, 237,
new variant CJD (nvCJD) 332, 334 pseudobulbar 524, 536–7, 538 682
Niemann–Pick disease type C (NP-C) supranuclear horizontal gaze palsy 502 polymyositis 402, 403, 681–4
174–5 supranuclear vertical gaze palsy 175, polyneuropathy 582, 610
NINDS–AIREN criteria for vascular 433, 502 acute inflammatory demyelinating 602,
dementia 264 see also paralysis 603
nocturnal myoclonus 127 papilledema 478, 492–4 chronic inflammatory demyelinating
normal pressure hydrocephalus (NPH) papillitis 489 (CIDP) 590, 593–5, 602, 603, 608
469, 473–6 papilloma, choroid plexus 366 distal 649
nutritional deficiency paralysis familial amyloid 584
vitamin B12 deficiency 463–7, 474, acute idiopathic facial (Bell’s palsy) locked-in syndrome 56
564 515–16 polyradiculopathy
vitamin E deficiency 442, 531 hypokalemic 693–5 lumbosacral 141
Wernicke–Korsakoff syndrome 460–2 familial periodic paralysis (FPP) progressive 602, 604
nystagmus 518, 519 693–4 porphyria 584
thyrotoxic periodic paralysis (TPP) acute intermittent 544
690, 691, 692, 694 positron emission tomography (PET) 26
O infantile 309–11 post-herpetic neuralgia 601
obsessive–compulsive disorder 131, 133, respiratory 450 post-polio progressive muscular atrophy
416 rucksack paralysis 596 311
obtundation 44 sleep paralysis 58 post-polio syndrome 311
obturator nerve 642 see also palsy posterior cutaneous nerve of the thigh
obturator neuropathy 642–3 paramyotonia congenita 680 639
ocular motor neuropathies 499–504 paraneoplastic syndromes 400, 401–5 neuropathy 639
ocular myasthenia 659, 663 paraproteinemias 222 posterior spinal artery syndrome 560
oculomotor nerve 18, 500 paraproteinemic neuropathies 606–9 postinfectious encephalomyelitis 293,
neuropathy 499–504 parasympathetic nervous system 574 337–9
olfactory nerve 15 Parkinsonian tremor 120 postural hypotension 64, 428, 576, 577,
olfactory neuropathy 481–2 parkinsonism 424 578
oligodendroglioma 364–5, 368, 370, 371 Parkinson’s disease (PD) 420–30, 431, pregnancy
olivopontocerebellar atrophy (OPCA) 433, 436, 474 epilepsy and 76, 87
435 dementia 263 myasthenia and 664
one-and-a-half syndrome 504 parosmia 482 prothrombotic states 222, 224
ophthalmoplegia 355, 502 paroxysmal dyskinesias 129, 130 primary CNS lymphoma 385–8
exophthalmic 690, 691, 692 paroxysmal dystonias 116 primary intracerebral hemorrhage (PICH)
internuclear 503 paroxysmal facial pain 509–11 238–44
progressive external 659 paroxysmal nocturnal hemoglobinuria primary lateral sclerosis 534, 537
ophthalmoplegic migraine 97 222 primary nerve cell degeneration 37
opsoclonus 519 Parsonage Turner syndrome 595 primary orthostatic tremor 120
optic nerve 15–16 patent foramen ovale 212 primary progressive aphasia 416
glioma 148, 372–3 penicillamine-induced myasthenia 659 prion disease 330–5
optic neuritis 489–91 perception, assessment of 15 progressive bulbar palsy 534, 537
optic neuropathy 344, 483–6, 494 perilymph fistula 108 progressive multifocal
anterior ischemic (AION) 487–8 peripheral neuropathy 579–83 leukoencephalopathy (PML) 307–9
Leber’s hereditary (LHON) 489, permanent vegetative state (PVS) 52–5 progressive muscular atrophy 534, 537
491–2 peroneal muscular atrophy 584 progressive myoclonic ataxia 88
organ of Corti 517 peroneal nerve 650 progressive myoclonic encephalopathies
Osler–Rendu–Weber syndrome 153–6 peroneal neuropathy 650–3 88, 126
osteomalacia 690, 692 pheochromocytoma 62 progressive myoclonic epilepsies (PME)
osteosclerotic myeloma 607, 608, 609 phosphofructokinase deficiency 688, 690, 86, 87–90, 126
oxidative phosphorylation diseases 691 progressive polyradiculopathy 602, 604
162–6 Pick’s disease 414–16 progressive supranuclear palsy (PSP) 263,
pinealomas 368, 390, 392 432–4
pituitary apoplexy 268–9, 381 prolactinoma 380
P pituitary tumors 379–83 pronator teres syndrome 628, 629
pachymeningitis, syphilitic 443 plantar interdigital neuropathy 648, 649, prosthetic heart valves 211
pain 650 protein C 223
history 12 plantar neuropathy 648, 649 protein S 223
multiple sclerosis 342, 349 lateral 648 prothrombotic states 218–24
neuropathy 582 medial 647–8, 649 protozoal infections
on exercise 690 plantar reflex 21 meningitis 276
Parkinson’s disease 423 platelet abnormalities 222 myositis 682
paroxysmal facial pain 509–11 POEMS syndrome 606, 608 toxoplasmic encephalitis 323–6
slipped disc 550 poikiloderma 685, 686 proximal diabetic neuropathy 612
trigeminal neuralgia 509 poliomyelitis (infantile paralysis) 309–11, proximal myotonic myopathy (PROMM)
trigeminal neuropathy 506 596 680
see also headache acute anterior horn cell poliomyelitis pseudobulbar palsy 524, 536–7, 538
palsy 310 pseudopapilledema 478
Bell’s 515–16 non-paralytic poliomyelitis 310 pseudoseizures 71
bulbar 524, 528, 536, 538 post-polio progressive muscular atrophy pseudotumor cerebri 477
progressive 534, 537 311 ptosis 495, 496, 502, 504
Index 703

pudendal nerve 655 sclerosis (continued) spinal cord (continued)


pudendal neuropathy 655–6 primary lateral 534, 537 epidural hematoma 554–5
pulmonary arterial venous malformation tuberous 143–6, 358 hereditary spastic paraparesis (HSP)
212 see also atherosclerosis; multiple sclerosis 529–31, 538, 564
pupillary responses 16, 574–5, 576, 577 Segawa’s disease 115 infarction 558–61
accommodation reflex 575, 576 segmental demyelination 36–7 infection 344, 546, 550, 561, 564
light reflex 574, 576 seizures 68–70, 187 inflammation 344
relative afferent pupillary defect 575, akinetic 68–9 see also myelitis
576 atonic 68–9 subacute combined degeneration 443,
pyriformis syndrome 645 brain tumors and 359, 362 531
myoclonic 68, 124–5 syringomyelia 541–4, 636
see also epilepsy tethered cord syndrome 142
R semicircular canals 517 tumors 344, 443, 530, 544, 546, 550,
rachischisis 139–42 senile plaques 408 567–71
radial nerve 623 sensation metastatic lesions 567, 570
radial neuropathy 623–6 assessment 21 see also myelopathy
radiation-induced encephalo-myelo- hereditary spastic paraparesis and 530 spinal cord syndrome
radiculopathy 561, 564, 565–6 multiple sclerosis and 342 acute non-compressive 344
radiculopathy 636, 646, 649, 652 sensory nerve action potential (SNAP) 36 chronic non-compressive 344
cervical spondylotic 545–9 sensory nerve conduction studies 35–6 spinal muscular atrophy (SMA) 532–4,
lumbar 641, 642 serotonin syndrome 134 536, 537–8, 626, 636, 652
lumbosacral polyradiculopathy 141 sexual dysfunction, multiple sclerosis 342, spinal venous infarction syndrome 560
radiation-induced 565–6 348 spine, examination of 15
spinal cord tumors and 568 shingles 299–300 see also imaging
raised intracranial pressure 359, 362, 384 shock 450 splenomegaly 175
hydrocephalus 469 short-lasting unilateral neuralgiform spondylosis 545
papilledema 492 headache with conjunctival injection cervical 544, 545–9
Ramsay Hunt syndrome 513 and tearing (SUNCT) syndrome 104 lumbar 552
Raynaud’s phenomenon 685 shoulder disorders 619, 620 spondylotic myelopathy 636
reflex tachycardia 577 Shy–Drager syndrome (SDS) 435 stance assessment 22
reflexes 20, 21–2 sialidosis 89 Staphylococcus aureus 284, 555
anal reflex 22 sick sinus syndrome 211 startle reflexes 127
bulbocavernosus reflex 21 sickle cell (SC) disease 222 static myoclonic encephalopathies 125–6
H-reflex 37 single photon emission computed status epilepticus (SE) 82–5
plantar reflex 21 tomography (SPECT) 26 Steele–Richardson–Olszewski syndrome
pupillary 16, 574–5, 576, 577 sinoatrial disease 211 432
accommodation reflex 575, 576 sinus arrhythmia 577 steroid myopathy 689, 690, 692
light reflex 574, 576 sinusitis 105 strabismus 502
trigeminovascular reflex 96 idiopathic cavernous 354, 355 Streptococcus milleri 284
Refsum’s disease 584, 605 sleep disorders 58, 62, 131 Streptococcus pneumoniae
repetitive stimulation studies 37 narcolepsy 56–8 intracranial abscess 284
respiratory failure, Guillain–Barré sleep apnea 57 meningitis 273–4, 278
syndrome 591 sleep paralysis 58 striatonigral degeneration (SND) 435,
respiratory insufficiency 691 sleepiness, excessive 678, 681 436
respiratory paralysis 450 see also narcolepsy stroke 192–201, 266, 458, 652
resuscitation, coma 50 slipped disc, acute 550–3 ischemic stroke
retinitis pigmentosa 165 smoking, stroke and 200 atherosclerotic ischemic 202–8,
rheumatic vascular disease 211 sodium channelopathies 680 221–2
rheumatoid arthritis 597 somatosensory evoked potentials (SSEPs) lipohyalinosis/microatheroma 204,
Ross’ syndrome 498 42 238
rubral tremor 120 spasmodic torticollis 115 cardioembolic 209–15
rucksack paralysis 596 spasticity thrombophilic 218-24
hereditary spastic paraparesis (HSP) hemorrhagic stroke
529–31, 538, 564 intracerebral hemorrhage 192, 198,
S multiple sclerosis 348 299
saccades 499 speech disturbance primary intracerebral hemorrhage
saccular aneurysms 250 transient ischemic attack 186 (PICH) 238–44
saccule 517 vagus nerve neuropathy 524 subarachnoid hemorrhage 178, 192,
sagittal sinus thrombosis 258 Spielmeyer–Vogt disease 89 198, 249–56
Sandhoff disease 172 spina bifida aperta 141 stroke-like syndrome 258
saphenous nerve 640, 641 spina bifida occulta 140, 141 Strumpell–Lorrain syndrome 529
sarcoglycanopathy 676 spinal accessory nerve 19 stupor 44, 48
sarcoidosis 350–3, 598 neuropathy 525–6 Sturge–Weber disease 151–2
Schilder disease 167 spinal artery syndrome subacute sclerosing panencephalitis (SSPE)
schizophrenia 57, 416 anterior 560 304–6
schwannoma, spinal cord 570 posterior 560 subarachnoid hemorrhage (SAH) 178,
sciatic nerve 644 spinal cord 192, 198, 249–56
sciatic neuropathy 644–6, 652 acute slipped disc 550–3 subcortical arteriosclerotic encephalopathy
sclerosis acute transverse myelitis 561–3 (SAE) 474
amyotrophic lateral (ALS) 534, 536, arachnoiditis 557–8 subdural empyema 284–7
537 arteriovenous malformation 246, 443, subdural hematoma 177–80
aortic valve 211 530 subdural hygroma 178
diffuse cerebral sclerosis of Schilder cervical spondylotic myelopathy/ suffocation 450
167 radiculopathy 545–9 sulcocommissural artery syndrome 560
mesial temporal (MTS) 68 compression 545, 550, 557, 560, 567 supinator syndrome 624, 625
mitral valve 211 epidural abscess 555–7 supranuclear horizontal gaze palsy 502
704 Index

supranuclear vertical gaze paresis 175, tremor 123, 127 venous sinus thrombosis 260, 478
433, 502 enhanced physiologic tremor (EPT) vertebrobasilar dissection 224–7
suprascapular nerve 618 120 vertebrobasilar ischemia 61, 108
suprascapular neuropathy 618–19 essential tremor (ET) 119–22, 423 vertebrobasilar migraine 97
sural nerve 654 intention/terminal tremor 119 vertigo 107–10, 519
sural neuropathy 654–5 multiple sclerosis 349 benign paroxysmal positional 110
sweating 574, 577 Parkinsonian tremor 120 Ménière’s disease 109, 111–12
sympathetic nervous system 573–4 position-specific/task-specific tremors multiple sclerosis 342
syncope 59–64, 70 120 vestibular neuronitis 111
syphilis 312–15 primary orthostatic tremor 120 vestibular-cochlear neuropathy 518
concurrent HIV infection 315 rubral tremor 120 vestibular neuronitis 111
meningovascular 312, 313–14 Treponema pallidum 312, 315 vestibular-cochlear nerve 19, 517
pachymeningitis 443 trigeminal nerve 18–19 vestibular-cochlear neuropathy 517–20
syringobulbia 541, 542 trigeminal neuralgia 104, 509–11 viral infections
syringomyelia 541–4, 636 trigeminal neuropathy 505–8 acute aseptic meningitis 291–2
syrinx 541 isolated trigeminal sensory neuropathy cytomegalovirus (CMV) 589
systemic lupus erythematosus 597 508 encephalitis 292–5, 338
trigeminovascular reflex 96 herpes simplex virus encephalitis (HSE)
trochlear nerve 18, 500 295–8
T neuropathy 499–504 herpes zoster infection 599–601
tabes dorsalis 312, 314 Tropheryma whippelii 289 facial paresis 513
tachyarrhythmia tuberculoma 281 varicella-zoster virus
reflex tachycardia 577 tuberculosis 555 encephalomyelitis 299–300
syncope 59 tuberculous meningitis 281 HIV see human immunodeficiency virus
Taenia solium 326 tuberculous meningo-encephalitis 281–3 myositis 682
Tangier disease 544 tuberous sclerosis 143–6, 358 poliomyelitis (infantile paralysis) 309–11
tapeworm infection 326–9 tumors 352, 357–405, 521 progressive multifocal leukoen-
myositis 682 acoustic neuroma 383–4 cephalopathy (PML) 307–9
tardive dyskinesia 129, 130 brain tumors 357–63 subacute sclerosing panencephalitis
tarsal tunnel syndrome 647, 648, 649, cavernous sinus syndrome 354, 355 (SSPE) 304–6
650 craniopharyngioma 377–9 visual acuity 15, 486
anterior 651, 652 germ cell tumors of the CNS 388–92 multiple sclerosis and 341–2
Tay–Sachs disease 172 gliomas 364–71 optic neuritis and 490
telangiectases 153, 154, 246 optic nerve 372–3 optic neuropathy and 484
ataxia telangiectasia 358, 444–6 intracardiac 212 papilledema 493
hereditary hemorrhagic telangiectasia meningioma 374–6 visual disturbances 236, 486, 488
(HHT) 153–6 metastases to the CNS 396–400 idiopathic intracranial hypertension
temporal arteritis (TA) 237 spine 567, 570 477, 480
tension-type headache 101–3 pituitary tumors 379–83 monocular blindness 487–8, 490
teratomas 389, 390 primary CNS lymphoma 385–8 multiple sclerosis 349
tetanus 287–8 spinal cord 344, 443, 530, 544, 546, ocular motor neuropathies 501–2
tethered cord syndrome 142 550, 567–71 optic neuritis 489
thiamine deficiency 460 von Hippel–Lindau disease (VHL) optic neuropathy 483–4
thromboembolism 190, 206, 207 392–4 anterior ischemic (AION) 487
see also atherothrombosis; embolism see also specific tumors Leber’s hereditary (LHON) 489, 491-2
thrombophilia 218–24 Turcot’s syndrome 358 transient ischemic attack 196
thrombosis 205, 206, 219 visual evoked potentials (VEPs) 40
cavernous sinus 258 visual fields 16
cerebral venous 257–60 U optic neuritis and 490–1
deep cerebral vein 259 ulnar nerve 632–3 optic neuropathy and 484, 485
cortical venous 258 ulnar neuropathy 632–6 papilledema 493
prothrombotic states 218–24 ultrasound 28 vitamin B12 deficiency 463–7, 474, 564
sagittal/transverse sinus 258 uncal herniation 46 vitamin E deficiency 442, 531
venous sinus 260, 478 Unverricht–Lundborg disease 88, 89 von Hippel–Lindau disease (VHL) 358,
thrombotic thrombocytopenic purpura upper limb examination 20–1 392–4
(TTP) 223 utricle 517 von Recklinghausen’s neurofibromatosis
thunderclap headache 99, 258 147, 605
thymoma 660, 663
thyrotoxic myopathy 689, 690, 691, 692 V
thyrotoxic periodic paralysis (TPP) 690, vagus nerve 19 W
691, 692, 694 vagus nerve neuropathy 523–4 Waldenström’s macroglobulinemia 222,
thyrotoxicosis 120 Valsalva maneuver 577 607, 608, 609
tibial nerve 647 varicella-zoster virus encephalomyelitis Wallerian degeneration 36, 580
tibial neuropathy 647–50 299–300 Wegener’s granulomatosis 597
tic douloureux 509–11 see also herpes zoster infection Werdnig–Hoffman (WH) disease 532,
tics 123, 127, 130–3 vascular dementia 261–5, 416, 474 538
tinnitus 518 NINDS–AIREN criteria 264 Wernicke–Korsakoff syndrome 460–2
Tolosa–Hunt syndrome 354 vascular diseases see neurovascular Wernicke’s disease 460, 461
Tourette syndrome (TS) 130–3 syndromes West’s syndrome 69
toxoplasmic encephalitis 323–6 vascular malformations 238, 246 Whipple’s disease 289–90
transient ischemic attacks (TIA) 70, 178, see also arteriovenous malformation Wilson’s disease (WD) 157–61
186–92 vasculitic neuropathy 544, 597–8 writer’s cramp 115
of the eye 187–8 vasculitis 597
spinal 560 central nervous system 227–33
see also stroke vegetative state 52, 451 X
transverse sinus thrombosis 258 venous infarction 260 X-linked dystrophinopathies 668–72

You might also like