Neurologic Aspects of Systemic Disease Part II [1st Edition] 9780702044335, 9780702040870

Systemic disease involves several parts of the body or the complete system. Neurologic Aspects of Systemic Disease, a tw

1,241 74 44MB

English Pages 736 [575] Year 2014

Report DMCA / Copyright

DOWNLOAD FILE

Polecaj historie

Neurologic Aspects of Systemic Disease Part II [1st Edition]
 9780702044335, 9780702040870

Table of contents :
Content:
Series PagePage ii
CopyrightPage iv
Handbook of Clinical Neurology 3rd SeriesPage v
ForewordPage viiMichael J. Aminoff, François Boller, Dick F. Swaab
PrefacePage ixJosé Biller, José M. Ferro
ContributorsPages xi-xiv
Chapter 39 - The neurologic complications of bariatric surgeryPages 587-594Joseph R. Berger, Divya Singhal
Chapter 40 - Neurologic manifestations of inflammatory bowel diseasesPages 595-605José M. Ferro, Sofia N. Oliveira, Luis Correia
Chapter 41 - Gluten-related neurologic dysfunctionPages 607-619Marios Hadjivassiliou, Andrew P. Duker, David S. Sanders
Chapter 42 - Neurologic manifestations of malabsorption syndromesPages 621-632Ronald F. Pfeiffer
Chapter 43 - Commonly used gastrointestinal drugsPages 633-643Annu Aggarwal, Mohit Bhatt
Chapter 44 - Neurologic manifestations of acute liver failurePages 645-659Sudhir Datar, Eelco F.M. Wijdicks
Chapter 45 - Portosystemic encephalopathyPages 661-674Karin Weissenborn
Chapter 46 - Neurotoxicity of commonly used hepatic drugsPages 675-682Christine L. Ahrens, Edward M. Manno
Chapter 47 - Neurology of the pituitaryPages 685-701Shanika Samarasinghe, Mary Ann Emanuele, Alaleh Mazhari
Chapter 48 - Thyroid disease and the nervous systemPages 703-735Clare A. Wood-allum, Pamela J. Shaw
Chapter 49 - Neurologic disorders of mineral metabolism and parathyroid diseasePages 737-748Lily Agrawal, Zeina Habib, Nicholas V. Emanuele
Chapter 50 - Neurologic complications of disorders of the adrenal glandsPages 749-771Tulio E. Bertorini, Angel Perez
Chapter 51 - Diabetic neuropathyPages 773-785Gerald Charnogursky, Hong Lee, Norma Lopez
Chapter 52 - Neurologic disorders associated with disease of the ovaries and testisPages 787-798Jorge C. Kattah, William C. kattah
Chapter 53 - Neurologic complications of multiple endocrine syndromesPages 799-808Jorge C. Kattah, William C. Kattah
Chapter 54 - Commonly used endocrine drugsPages 809-824Mário Miguel Rosa, Teresa Dias
Chapter 55 - Disorders of purines and pyrimidinesPages 827-838Roger E. Kelley, Hans C. Andersson
Chapter 56 - Porphyria and its neurologic manifestationsPages 839-849Jennifer A. Tracy, P. James B. Dyck
Chapter 57 - Disorders of heavy metalsPages 851-864France Woimant, Jean-Marc Trocello
Chapter 58 - Disorders of bone and bone mineral metabolismPages 865-887Monica Komoroski, Nasrin Azad, Pauline Camacho
Chapter 59 - Hydrosoluble vitaminsPages 891-914Jasvinder Chawla, David Kvarnberg
Chapter 60 - Neurologic aspects of cobalamin (B12) deficiencyPages 915-926Neeraj Kumar
Chapter 61 - The neurology of folic acid deficiencyPages 927-943E.H. Reynolds
Chapter 62 - Disorders of body temperaturePages 947-957Camilo R. Gomez
Chapter 63 - Neurology and divingPages 959-969E. Wayne Massey, Richard E. Moon
Chapter 64 - Neurologic complications of carbon monoxide intoxicationPages 971-979Kerstin Betterman, Surju Patel
Chapter 65 - Lightning and thermal injuriesPages 981-986Arthur Sanford, Richard L. Gamelli
Chapter 66 - Venomous snake bites, scorpions, and spidersPages 987-1001S.A.M. Kularatne, Nimal Senanayake
Chapter 67 - Anemias excluding cobalamin and folate deficienciesPages 1005-1014Stephanie Dublis, Shefali Shah, Sucha Nand, Elise Anderes
Chapter 68 - Neurologic complications of sickle cell diseasePages 1015-1025Akila Venkataraman, Robert J. Adams
Chapter 69 - Neurologic aspects of lymphoma and leukemiasPages 1027-1043Matthew Mccoyd, Gregory Gruener, Patrick Foy
Chapter 70 - Bleeding diathesis and hemophiliasPages 1045-1059Chirag Amin, Anjali Sharathkumar, Anne Griest
Chapter 71 - Thrombophilic statesPages 1061-1071Ida Martinelli, Serena Maria Passamonti, Paolo Bucciarelli
Chapter 72 - Chronic myeloproliferative diseasesPages 1073-1081João Forjaz de Lacerda, Sofia N. Oliveira, José M. Ferro
Chapter 73 - Neurologic aspects of plasma cell disordersPages 1083-1099Urszula Sobol, Patrick Stiff
Chapter 74 - Neurologic manifestations of Henoch–Schönlein purpuraPages 1101-1111Maxime D. Bérubé, Normand Blais, Sylvain Lanthier
Chapter 75 - Hemolytic uremic syndromePages 1113-1123Kathleen Webster, Eugene Schnitzler
Chapter 76 - Commonly used drugs in hematologic disordersPages 1125-1139Elise Anderes, Sucha Nand
IndexPages I1-I46

Citation preview

HANDBOOK OF CLINICAL NEUROLOGY Series Editors

MICHAEL J. AMINOFF, FRANC¸OIS BOLLER, AND DICK F. SWAAB VOLUME 120

EDINBURGH LONDON NEW YORK OXFORD PHILADELPHIA ST LOUIS SYDNEY TORONTO 2014

ELSEVIER B.V. Radarweg 29, 1043 NX, Amsterdam, The Netherlands © 2014, Elsevier B.V. All rights reserved. No part of this publication may be reproduced or transmitted in any form or by any means, electronic or mechanical, including photocopying, recording, or any information storage and retrieval system, without permission in writing from the publisher. Permissions may be sought directly from Elsevier’s Rights Department: phone: (þ1) 215 239 3804 (US) or (þ44) 1865 843830 (UK); fax: (þ44) 1865 853333; e-mail: [email protected]. You may also complete your request on-line via the Elsevier website at http://www.elsevier.com/permissions. This book and the individual contributions contained in it are protected under copyright by the Publisher (other than as may be noted herein). ISBN: 9780702040870 British Library Cataloguing in Publication Data A catalogue record for this book is available from the British Library Library of Congress Cataloging in Publication Data A catalog record for this book is available from the Library of Congress Notice Knowledge and best practice in this field are constantly changing. As new research and experience broaden our understanding, changes in research methods, professional practices, or medical treatment may become necessary. Practitioners and researchers must always rely on their own experience and knowledge in evaluating and using any information, methods, compounds, or experiments described herein. In using such information or methods they should be mindful of their own safety and the safety of others, including parties for whom they have a professional responsibility. With respect to any drug or pharmaceutical products identified, readers are advised to check the most current information provided (i) on procedures featured or (ii) by the manufacturer of each product to be administered, to verify the recommended dose or formula, the method and duration of administration, and contraindications. It is the responsibility of practitioners, relying on their own experience and knowledge of their patients, to make diagnoses, to determine dosages and the best treatment for each individual patient, and to take all appropriate safety precautions. To the fullest extent of the law, neither the Publisher nor the contributors or editors, assume any liability for any injury and/or damage to persons or property as a matter of products liability, negligence or otherwise, or from any use or operation of any methods, products, instructions, or ideas contained in the material herein. The Publisher

Printed in China

Commissioning Editor: Thomas E. Stone Development Editor: Michael Parkinson Project Manager: Anitha Kittusamy Ramasamy Designer/Design Direction: Alan Studholme

The Publisher's policy is to use paper manufactured from sustainable forests

Handbook of Clinical Neurology 3rd Series Available titles Vol. 79, The human hypothalamus: basic and clinical aspects, Part I, D.F. Swaab ISBN 9780444513571 Vol. 80, The human hypothalamus: basic and clinical aspects, Part II, D.F. Swaab ISBN 9780444514905 Vol. 81, Pain, F. Cervero and T.S. Jensen, eds. ISBN 9780444519016 Vol. 82, Motor neurone disorders and related diseases, A.A. Eisen and P.J. Shaw, eds. ISBN 9780444518941 Vol. 83, Parkinson’s disease and related disorders, Part I, W.C. Koller and E. Melamed, eds. ISBN 9780444519009 Vol. 84, Parkinson’s disease and related disorders, Part II, W.C. Koller and E. Melamed, eds. ISBN 9780444528933 Vol. 85, HIV/AIDS and the nervous system, P. Portegies and J. Berger, eds. ISBN 9780444520104 Vol. 86, Myopathies, F.L. Mastaglia and D. Hilton Jones, eds. ISBN 9780444518996 Vol. 87, Malformations of the nervous system, H.B. Sarnat and P. Curatolo, eds. ISBN 9780444518965 Vol. 88, Neuropsychology and behavioural neurology, G. Goldenberg and B.C. Miller, eds. ISBN 9780444518972 Vol. 89, Dementias, C. Duyckaerts and I. Litvan, eds. ISBN 9780444518989 Vol. 90, Disorders of consciousness, G.B. Young and E.F.M. Wijdicks, eds. ISBN 9780444518958 Vol. 91, Neuromuscular junction disorders, A.G. Engel, ed. ISBN 9780444520081 Vol. 92, Stroke – Part I: Basic and epidemiological aspects, M. Fisher, ed. ISBN 9780444520036 Vol. 93, Stroke – Part II: clinical manifestations and pathogenesis, M. Fisher, ed. ISBN 9780444520043 Vol. 94, Stroke – Part III: Investigations and management, M. Fisher, ed. ISBN 9780444520050 Vol. 95, History of neurology, S. Finger, F. Boller and K.L. Tyler, eds. ISBN 9780444520081 Vol. 96, Bacterial infections of the central nervous system, K.L. Roos and A.R. Tunkel, eds. ISBN 9780444520159 Vol. 97, Headache, G. Nappi and M.A. Moskowitz, eds. ISBN 9780444521392 Vol. 98, Sleep disorders Part I, P. Montagna and S. Chokroverty, eds. ISBN 9780444520067 Vol. 99, Sleep disorders Part II, P. Montagna and S. Chokroverty, eds. ISBN 9780444520074 Vol. 100, Hyperkinetic movement disorders, W.J. Weiner and E. Tolosa, eds. ISBN 9780444520142 Vol. 101, Muscular dystrophies, A. Amato and R.C. Griggs, eds. ISBN 9780080450315 Vol. 102, Neuro-ophthalmology, C. Kennard and R.J. Leigh, eds. ISBN 9780444529039 Vol. 103, Ataxic disorders, S.H. Subramony and A. Durr, eds. ISBN 9780444518927 Vol. 104, Neuro-oncology Part I, W. Grisold and R. Sofietti, eds. ISBN 9780444521385 Vol. 105, Neuro-oncology Part II, W. Grisold and R. Sofietti, eds. ISBN 9780444535023 Vol. 106, Neurobiology of psychiatric disorders, T. Schlaepfer and C.B. Nemeroff, eds. ISBN 9780444520029 Vol. 107, Epilepsy Part I, H. Stefan and W.H. Theodore, eds. ISBN 9780444528988 Vol. 108, Epilepsy Part II, H. Stefan and W.H. Theodore, eds. ISBN 9780444528995 Vol. 109, Spinal cord injury, J. Verhaagen and J.W. McDonald III, eds. ISBN 9780444521378 Vol. 110, Neurological rehabilitation, M. Barnes and D.C. Good, eds. ISBN 9780444529015 Vol. 111, Pediatric neurology Part I, O. Dulac, M. Lassonde and H.B. Sarnat, eds. ISBN 9780444528919 Vol. 112, Pediatric neurology Part II, O. Dulac, M. Lassonde and H.B. Sarnat, eds. ISBN 9780444529107 Vol. 113, Pediatric neurology Part III, O. Dulac, M. Lassonde and H.B. Sarnat, eds. ISBN 9780444595652 Vol. 114, Neuroparasitology and tropical neurology, H.H. Garcia, H.B. Tanowitz and O.H. Del Brutto, eds. ISBN 9780444534903 Vol. 115, Peripheral nerve disorders, G. Said and C. Krarup, eds. ISBN 9780444529022 Vol. 116, Brain stimulation, A.M. Lozano and M. Hallett, eds. ISBN 9780444534972 Vol. 117, Autonomic nervous system, R.M. Buijs and D.F. Swaab, eds. ISBN 9780444534910 Vol. 118, Ethical and legal issues in neurology, J.L. Bernat and H.R. Beresford, eds. ISBN 9780444535016 Vol. 119, Neurologic Aspects of Systemic Disease, J. Biller and J.M. Ferro, eds. ISBN 9780702040863

Foreword

Although neurology and psychiatry are closely linked specialties, many neurologists see their specialty as part of internal medicine. Indeed, neurology departments in the United States often began as divisions within departments of internal medicine, attesting to their special relationship. With the evolution of neurology as an independent discipline, it has become particularly important for its practitioners to remain familiar with the neurologic aspects of systemic diseases as well as with the systemic aspects of neurologic disorders. This has been recognized since the Handbook of Clinical Neurology was founded by Pierre Vinken and George Bruyn, with volume 1 appearing in December 1968. That first series concluded in 1982 and was followed by a second series, edited by them, that concluded—in turn—in 2002. We then took over as editors of the current third series, with volume 79 appearing in late 2003 and several volumes appearing annually since then. Two volumes (38 and 39) were published in the first series of the Handbook, focusing on the neurologic manifestations of systemic diseases. The second series included a further three volumes (63, 70, and 71) on the same topic, published in 1993 and 1998, with one of us (MJA) serving as an editor of those volumes. Advances in the field, but especially in immunology, genetics, imaging, pharmacotherapeutics, and intensive care, since that time have necessitated a reappraisal of the field and the publication of three new volumes on the topic. We are therefore particularly delighted at the publication of this scholarly contribution to the medical and neurologic literature and welcome it as part of the Handbook. We believe that it will appeal not only to neurologists but to physicians in all specialties, helping in their interactions with each other and with their patients. Professors Jose´ Biller and Jose´ M. Ferro have together produced an authoritative, comprehensive, and up to date account of the topic and have assembled a truly international group of authors with acknowledged expertise to contribute to these important multifaceted volumes. We are grateful to them and to all the contributors for their efforts in creating such an invaluable resource. We are confident that clinicians in many different disciplines will find much in these volumes to appeal to them. It is a pleasure, also, to thank Elsevier, our publishers – and in particular Tom Stone, Michael Parkinson, and Kristi Anderson – for unfailing and expert assistance in the development and production of these volumes. Michael J. Aminoff Franc¸ois Boller Dick F. Swaab

Preface

Medicine has always been in a state of evolution and today, more than ever, with the accelerated growth of scientific knowledge, patients are evaluated and treated by teams of physicians. The extensive body of knowledge and the major scientific and clinical advances in neurology and internal medicine are again drawing both specialties closer together. Whatever the subspecialty area of interest, the nature of modern clinical medicine calls for multidisciplinary collaborative efforts to better meet the needs of individual patients. The aim of these three volumes of the third series of the Handbook of Clinical Neurology is to integrate and provide a thorough framework of the core neurologic manifestations of a wide array of systemic disorders. Each chapter provides a critical appraisal and extensive background information regarding the variety of presentations of each disorder, the characteristic clinical course, the typical neurologic manifestations of each disease, and current therapeutic strategies. Comprehensive and updated references also bring forth valuable resources for further topical reading and research. Our intended audience includes experienced practitioners and residents in neurology, neurosurgery, and internal medicine, as well as other health care professionals in different subspecialties caring for these challenging patients. We have purposely divided these three volumes into chapters uniformly organized by organ system, which are further divided by specific conditions and disease categories. Volume I is dedicated to the neurologic aspects of cardiopulmonary diseases, renal disorders, and selective rheumatologic and musculoskeletal disorders. Volume II encompasses core neurologic aspects of gastrointestinal and hepatobiliary disorders, endocrinologic diseases, and a gamut of metabolic, nutritional and environmental conditions. Volume III concentrates on the neurologic aspects of hematologic and oncologic disorders, organ transplantation, infectious diseases, and tropical neurology. It also includes a miscellaneous group of disorders including neurodermatology, neurological complications of pregnancy, iatrogenic neurology, neuromuscular disorders in the intensive care setting, posterior reversible leukoencephalopathy and reversible cerebral vasoconstriction syndromes, neuro-Behcet’s, complications of neuroimaging, neurotraumatology, and observations pertaining to neurology in the developing world. These volumes go beyond the scope of classic neurology and examine the neurologic manifestations of a wide range of medical conditions, spanning most areas of medicine, that neurologists, neurosurgeons, internists, and other specialists must diagnose and treat in everyday practice. We are hopeful that these three volumes will contribute to the best possible care of patients with these disorders, and that the readership will find the material informative, authoritative, reliable, and stimulating We are extremely grateful to all the contributors from across the globe, who by sharing their knowledge and expertise made these volumes possible. To bring to fruition a work of this magnitude requires a highly professional editorial effort, and for this we thank Linda Turner for her wonderful organizational skills and administrative expertise, and Mike Parkinson and the editorial staff at Elsevier for their unfailing dedication, professionalism, and expert assistance in the development and production of these three volumes. Jose´ Biller, MD Jose´ M. Ferro, MD

Contributors

R.J. Adams South Carolina Stroke Center of Economic Excellence and Medical University of South Carolina Stroke Center, Charleston, SC, USA C.L. Ahrens Neurological Intensive Care Unit, Cleveland Clinic Cleveland, OH, USA A. Aggarwal Center for Brain and Nervous System, Kokilaben Dhirubhai Ambani Hospital and Medical Research Institute, Mumbai, India L. Agrawal Division of Endocrinology and Metabolism, Loyola University Medical Center, Maywood, IL, USA C. Amin Indiana Hemophilia and Thrombosis Centre, Indianapolis, IN, USA E. Anderes Division of Hematology and Oncology, Department of Medicine, Loyola University Chicago, Stritch School of Medicine, Maywood, IL, USA H.C. Andersson Hayward Genetics Center, Tulane University School of Medicine, New Orleans, LA, USA N. Azad Edward Hines Jr. VA Hospital, Hines, IL, USA

T.E. Bertorini Department of of Neurology, Methodist University Hospital and Department of Neurology, University of Tennessee Health Science Center, Memphis, TN, USA K. Betterman Department of Neurology, Penn State College of Medicine, Hershey, PA, USA M. Bhatt Center for Brain and Nervous System, Kokilaben Dhirubhai Ambani Hospital and Medical Research Institute, Mumbai, India N. Blais Department of Haematology, Centre Hospitalier de l’Universite´ de Montre´al, and Faculty of Medicine, Universite´ de Montre´al, Montreal, QC, Canada P. Bucciarelli A. Bianchi Bonomi Hemophilia and Thrombosis Center, Department of Internal Medicine and Medical Specialties, Fondazione IRCCS Ca’ Granda, Ospedale Maggiore Policlinico, University of Milan, Milan, Italy P. Camacho Loyola University Osteoporosis and Metabolic Bone Disease Center, Loyola University Medical Center, Maywood, IL, USA

J.R. Berger Department of Neurology, University of Kentucky College of Medicine, Lexington, KY, USA

G. Charnogursky Division of Endocrinology and Metabolism, Loyola University Chicago, Stritch School of Medicine, Maywood, IL, USA

M.D. Be´rube´ Department of Neurology, Centre Hospitalier de l’Universite´ de Montre´al, and Faculty of Medicine, Universite´ de Montre´al, Montreal, QC, Canada

J. Chawla Department of Neurology, Hines VA Hospital, Hines and Department of Neurology, Loyola University Medical Center, Maywood, IL, USA

xii CONTRIBUTORS L. Correia C.R. Gomez Department of Gastroenterology and Hepatology, Neurological Institute of Alabama, Birmingham, Hospital de Santa Maria, University of Lisbon, AL, USA Lisbon, Portugal A. Griest Indiana Hemophilia and Thrombosis Centre and S. Datar Department of Medicine, Indiana University School of Division of Critical Care Neurology, Mayo Clinic, Medicine, Indianapolis, IN, USA Rochester, MN, USA J.F. de Lacerda Department of Hematology and Bone Marrow Transplantation, Hospital de Santa Maria, Lisbon, Portugal T. Dias Endocrinology, Diabetes and Metabolism Unit, Hospital de Santa Maria, Lisbon, Portugal S. Dublis Division of Hematology and Oncology, Department of Medicine, Loyola University Chicago, Stritch School of Medicine, Maywood, IL, USA A.P. Duker Department of Neurology, University of Cincinnati, Cincinnati, OH, USA P.J.B. Dyck Mayo Clinic, Department of Neurology, Rochester, MN, USA M.A. Emanuele Division of Endocrinology and Metabolism, Loyola University Chicago, Stritch School of Medicine, Maywood, IL, USA N.V. Emanuele Division of Endocrinology and Metabolism, Loyola University Medical Center, Maywood, and Endocrinology Section, Edward Hines Jr. VA Hospital, Hines, IL, USA J.M. Ferro Neurology Service, Department of Neurosciences, Hospital de Santa Maria, University of Lisbon, Lisbon, Portugal P. Foy Department of Hematology, Medical College of Wisconsin, Milwaukee, WI, USA R.L. Gamelli Department of Surgery, Loyola University Medical Center, Maywood, IL, USA

G. Gruener Department of Neurology and Leischner Institute for Medical Education, Loyola University Medical Center, Maywood, IL, USA Z. Habib Division of Endocrinology and Metabolism, Loyola University Medical Center, Maywood, IL, USA M. Hadjivassiliou Department of Neurology, Royal Hallamshire Hospital, Sheffield, UK J.C. Kattah Department of Neurology, University of Illinois College of Medicine, Peoria, IL, USA W.C. Kattah Endocrinology Department, University of the Andes, Bogota, Colombia R.E. Kelley Department of Neurology, Tulane University School of Medicine, New Orleans, LA, USA M. Komoroski Loyola University Osteoporosis and Metabolic Bone Disease Center, Loyola University Medical Center, Maywood, IL, USA S.A.M. Kularatne Department of Medicine, Faculty of Medicine, University of Peradeniya, Kandy, Sri Lanka N. Kumar Department of Neurology, Mayo Clinic, Rochester, MN, USA D. Kvarnberg Department of Neurology, Loyola University Medical Center, Maywood, IL, USA S. Lanthier Department of Neurology, Centre Hospitalier de l’Universite´ de Montre´al, and Faculty of Medicine, Universite´ de Montre´al, Montreal, QC, Canada

CONTRIBUTORS xiii H. Lee A. Perez Division of Endocrinology and Metabolism, Loyola Department of Clinical Neurophysiology, University University Stritch School of Medicine, Maywood, of Tennessee Health Science Center, Memphis, IL, USA TN, USA N. Lopez Division of Endocrinology and Metabolism, Loyola University Chicago, Stritch School of Medicine, Maywood, IL, USA I. Martinelli A. Bianchi Bonomi Hemophilia and Thrombosis Center, Department of Internal Medicine and Medical Specialties, Fondazione IRCCS Ca’ Granda, Ospedale Maggiore Policlinico, University of Milan, Milan, Italy E.W. Massey Department of Neurology, Duke University Medical Center, Durham, NC, USA A. Mazhari Division of Endocrinology and Metabolism, Loyola University Chicago, Stritch School of Medicine, Maywood, IL, USA E.M. Manno Neurological Intensive Care Unit, Cleveland Clinic, Cleveland, OH, USA M. McCoyd Department of Neurology, Loyola University Medical Center, Maywood, IL, USA R.E. Moon Departments of Anesthesiology and Medicine, Duke University Medical Center, Durham, NC, USA S. Nand Division of Hematology and Oncology, Department of Medicine, Loyola University Chicago, Stritch School of Medicine, Maywood, IL, USA S.N. Oliveira Department of Neurology, Hospital da Luz, Lisbon, Portugal S.M. Passamonti A. Bianchi Bonomi Hemophilia and Thrombosis Center, Department of Internal Medicine and Medical Specialties, Fondazione IRCCS Ca’ Granda, Ospedale Maggiore Policlinico, University of Milan, Milan, Italy S. Patel Department of Internal Medicine, Conemaugh Health System, Jonestown, PA, USA

R.F. Pfeiffer Department of Neurology, University of Tennessee Health Science Center, Memphis, TN, USA E.H. Reynolds Department of Clinical Neurosciences, King’s College, London, UK M.M. Rosa Neurology Department, Hospital de Santa Maria, Lisbon, Portugal S. Samarasinghe Division of Endocrinology and Metabolism, Loyola University Chicago, Stritch School of Medicine, Maywood, IL, USA D.S. Sanders Department of Gastroenterology, Royal Hallamshire Hospital, Sheffield, UK A. Sanford Department of Surgery, Loyola University Medical Center, Maywood, IL, USA E. Schnitzler Department of Neurology, Loyola University Medical Center, Maywood, IL, USA N. Senanayake Department of Medicine, Faculty of Medicine, University of Peradeniya, Kandy, Sri Lanka S. Shah Division of Hematology and Oncology, Department of Medicine, Loyola University Chicago, Stritch School of Medicine, Maywood, IL, USA A. Sharathkumar Indiana Hemophilia and Thrombosis Centre and Department of Pediatrics, Riley Whitcomb Hospital for Children, Indiana University School of Medicine, Indianapolis, IN, USA P.J. Shaw Sheffield Institute for Translational Neuroscience, University of Sheffield and Department of Neurology, Royal Hallamshire Hospital, Sheffield, UK

xiv

CONTRIBUTORS

D. Singhal Department of Neurology, University of Kentucky College of Medicine, Lexington, KY, USA

K. Webster Department of Pediatrics, Loyola University Medical Center, Maywood, IL, USA

U. Sobol Department of Hematology and Oncology, Cardinal Bernardin Cancer Center, Loyola University Medical Center, Maywood, IL, USA

K. Weissenborn Department of Neurology, Hanover Medical School, Hanover, Germany

P. Stiff Department of Hematology and Oncology, Cardinal Bernardin Cancer Center, Loyola University Medical Center, Maywood, IL, USA

E.F.M. Wijdicks Division of Critical Care Neurology, Mayo Clinic, Rochester, MN, USA

J.A. Tracy Mayo Clinic, Department of Neurology, Rochester, MN, USA J-M. Trocello French National Wilson’s Disease Centre (CNR Wilson), Lariboisie`re Hospital, Paris, France A. Venkataraman Pediatric Neurology and Epilepsy Division, Lutheran Medical Center, Brooklyn, NY, USA

F. Woimant French National Wilson’s Disease Centre (CNR Wilson), Lariboisie`re Hospital, Paris, France C.A. Wood-Allum Sheffield Institute for Translational Neuroscience, University of Sheffield and Department of Neurology, Royal Hallamshire Hospital, Sheffield, UK

Handbook of Clinical Neurology, Vol. 120 (3rd series) Neurologic Aspects of Systemic Disease Part II Jose Biller and Jose M. Ferro, Editors © 2014 Elsevier B.V. All rights reserved

Chapter 39

The neurologic complications of bariatric surgery JOSEPH R. BERGER* AND DIVYA SINGHAL Department of Neurology, University of Kentucky College of Medicine, Lexington, KY, USA

OBESITY: AN OVERVIEW Weight is a national obsession. In 1999, Americans spent over $300 million on prescription medications for obesity (Wilhelm, 2000) and at the end of the 20th century 2.5% of the adult population reported using such preparations (Khan et al., 2001). By some estimates, the medical costs of obesity have risen to $147 billion per year as of 2008. Obesity is now officially recognized by the US Surgeon General as a significant health risk factor. Current prevalence of obesity is estimated to be over 30% in the US (Flegal et al., 2010), and another third are overweight. Obesity increases the risk for numerous medical illnesses, among them diabetes mellitus, hypercholesterolemia, hypertension and other cardiovascular disorders, pulmonary disease, chronic diseases, including osteoarthritis, liver and kidney disease, asthma, chronic back pain, sleep apnea, and depression and some forms of cancer (Fisher and Schauer, 2002; Mokdad et al., 2003). Obesity also increases the risk of death from all causes; it is estimated that 300 000 adults in the US die from obesityrelated causes annually (Allison et al., 1999). Given the increasing prevalence of obesity, it has been postulated that obesity may soon become the leading cause of death in the US (Mokdad et al., 2004). Furthermore, the average obese person costs society more than $7000 a year in lost productivity and as much as $30 000 in added medical costs over a lifetime (Freedman, 2011). The problem of obesity is a global phenomenon consequent on the ready availability of food with high caloric content and the reduction of daily energy expenditure. While the percentage of overweight adults in most Western European countries has not surpassed that of the US, their numbers are increasing rapidly. The number of obese children in many of these countries, such as, England, is growing rapidly (Lobstein et al., 2003) and

has outstripped that in the US. In major population centers of developing countries, obesity is also seen with increasing frequency. For medical purposes, obesity is defined by body mass index (BMI), which is derived by dividing an individual’s weight in kilograms by their height in meters square. Normal weight is defined as a BMI of 18.5–24.9 (National Heart, Lung, and Blood Institute, 1998). A BMI exceeding this value is regarded as overweight. BMIs of 30 or greater are considered obese and those equal to or exceeding 40 as morbidly obese (National Heart, Lung, and Blood Institute, 1998). Prevalence data from the CDC (US Centers for Disease Control and Prevention) indicates that in 1999–2000, 64.5% of US adults were overweight, 30.5% were obese, and 4.7% were morbidly obese (Flegal et al., 2002). Despite the billions of dollars spent on diets, dietary supplements, exercise programs, and other nonsurgical modalities for weight reduction, long-term success rates have been quite variable. Increasing numbers of people are, therefore, resorting to bariatric surgery for control of their weight problem. The increasing popularity of these procedures may, in part, be attributable to media attention provided by television personalities who have undergone surgery for their own weight reduction. From 1990 to 2000, the national annual rate of bariatric surgery increased nearly sixfold, from 2.4 to 14.1 per 100 000 adults (Trus et al., 2005). Population-adjusted rates of bariatric surgery in the overall sample increased more than sevenfold in the study period, from 3.5 per 100 000 US population in 1996 to 24.0 per 100 000 in 2002 (Davis et al., 2006).

BARIATRIC SURGERY National guidelines reserves bariatric surgery for individuals who have failed attempts of nonsurgical weight

*Correspondence to Dr. Joseph R. Berger, Department of Neurology, University of Kentucky College of Medicine, Kentucky Clinic L-445, 740 S. Limestone Street, Lexington, KY 40536-0284, USA. Tel: þ1-859 218-5039, Fax: þ1-859 323-5943, E-mail: [email protected]

588

J.R. BERGER AND D. SINGHAL

loss and have a BMI  35 with an obesity-related comorbidity or a BMI > 40 with or without a comorbidity (National Institutes of Health, 1992). A number of different surgical procedures have been employed for achieving weight reduction (Deitel and Shikora, 2002). Gastroplasties rely on the mechanical restriction of food passage through the stomach, whereas gastric bypass is considered to result in weight loss by a more physiologic mechanism (Livingston, 2002). Gastric bypass, typically the Roux-enY gastric bypass, is now performed more commonly than gastroplasties (Pope et al., 2002). Many of these surgical procedures can be performed laparoscopically (Azagra et al., 1999; Higa et al., 2000; Schauer et al., 2000). Average excess weight loss following laparoscopic Roux-en-Y gastric bypass has been reported to approach 70% at 12 months (Higa et al., 2000) and 83% at 24 months (Schauer et al., 2000), with excellent control of comorbidities (Buchwald et al., 2004). Although success rates have been variable, persistent, long-term weight loss of 10 or more years following these procedures has been documented (Yale, 1989; Pories et al., 1992). Studies of costeffectiveness support the value of bariatric surgery (Clegg et al., 2003; Fang, 2003). The complications of bariatric surgery are not insignificant, in part related to the problems inherent in operating on the obese person. In one series (Schauer et al., 2000), major complications occurred in 3.3% and the in-hospital mortality was 0.4%. Reoperation may be necessitated by bleeding, abscess, and wound dehiscence. A variety of late complications are recognized, generally the consequence of nutritional deficiency (Table 39.1). Mineral deficiencies include iron, calcium, phosphate, and magnesium, and vitamin deficiencies include thiamine (B1), cyanocobalamin (B12), folate, vitamin D, vitamin E. Iron, folate, and vitamin B12 deficiency have been reported to be the most common nutritional deficiencies observed following gastric bypass (Halverson, 1992; Skroubis et al., 2002; Bal et al., 2010). Table 39.1 Micronutrient deficiencies following bariatric surgery ● ● ● ● ● ● ● ● ● ● ●

Thiamine (B1) Cyanocobalamin (B12) Vitamin D Vitamin E Folate Iron Calcium Magnesium Phosphate Copper Selenium

Neurologic complications of bariatric surgery A broad spectrum of neurologic complications has been reported to occur in association with bariatric surgery (Table 39.2). No part of the neuraxis is exempt from these complications. To date, all studies addressing the neurologic complications occurring in the setting of bariatric surgery have been retrospective in nature. In 1987, Abarbanel and colleagues reported that 23 of 500 (4.6%) patients undergoing bariatric surgery experienced neurologic complications (Abarbanel et al., 1987). Of the 500 patients, 457 had a Roux-en-Y procedure and 43 had gastroplasty (Abarbanel et al., 1987). The neurologic complications became manifest 3–20 months after surgery and all affected patients experienced protracted vomiting, a symptom that may occur in up to one-third of all patients undergoing gastric bypass (Halverson, 1992; Bal et al., 2010). The constellation of neurologic complications included chronic and subacute peripheral neuropathy (52%), acute peripheral neuropathy (4%), burning feet (9%), meralgia paresthetica (9%), myotonic syndrome (4%), posterolateral myelopathy (9%), and Wernicke’s encephalopathy (9%) (Abarbanel et al., 1987). In this series, individuals with burning feet and Wernicke’s encephalopathy responded to thiamine administration (Abarbanel et al., 1987). In a retrospective review of 556 patients undergoing bariatric surgery at the Mayo Clinic from 1980 through 2003, Thaisetthawatkul and colleagues observed 48 patients (8.6%) with complications affecting the peripheral nervous system (Thaisetthawatkul, 2003). Of these 48 patients, 23 (48%) developed mononeuropathies with carpal tunnel syndrome, the most common accounting for 74% of the total (Thaisetthawatkul, Table 39.2 Neurologic complications of bariatric surgery ● ● ● ● ● ● ● ● ●

● ● ● ●

Encephalopathy Behavioral abnormalities Seizures Cranial nerve palsies Ataxia Myelopathy Plexopathies Peripheral neuropathy Mononeuropathies ● Carpal tunnel syndrome ● Meralgia paresthetica Compartment syndromes Myopathy Myotonia Restless legs syndrome

THE NEUROLOGIC COMPLICATIONS OF BARIATRIC SURGERY 589 2003). Peripheral neuropathies were observed in 20 Ranne, 1984; Oczkowski and Kertesz, 1985). Although (41.7%), plexopathies in 4 (17.4%), and myopathy in 1 some authors had previously considered Wernicke’s (4.3%) (Thaisetthawatkul, 2003). Perhaps the most debilencephalopathy a “very rare complication” of gastric itating of the neurologic consequences of bariatric sursurgery for morbid obesity (Bozbora et al., 2000), there gery are those occurring from vitamin deficiency, are at least 104 cases in the world’s literature and the particularly vitamins B1 and B12, as these may result in number of cases after bariatric surgery is substantially permanent neurologic disability. higher than previously reported (Aasheim et al., 2009). This complication is almost always associated with severe intractable vomiting. It may be seen relatively Thiamine deficiency (vitamin B1) soon after surgery, usually within 4–12 weeks (Kramer Thiamine deficiency alters mitochondrial function, and Locke, 1987) but occasionally as late as 18 months impairs oxidative metabolism, and causes selective after the procedure. neuronal death by diminishing thiamine-dependent Peripheral neuropathy often, but not invariably, enzymes (Ke et al., 2003). A deficiency of thiamine accompanies Wernicke’s encephalopathy following surresults in peripheral neuropathy, ophthalmoplegia and gery for morbid obesity. Peripheral neuropathy with or nystagmus, ataxia, encephalopathy and may lead to perwithout Wernicke’s encephalopathy that is attributable manent impairment of recent memory. This constellato thiamine deficiency and has occurred following surtion is referred to as Wernicke’s encephalopathy. The gery for morbid obesity has been referred to as “bariatric diagnostic criteria of Wernicke’s encephalopathy require beriberi” (Gollobin and Marcus, 2002). It appears to be two of the following four features: (1) dietary deficiency; more common than encephalopathy and may also occur (2) oculomotor abnormality; (3) cerebellar dysfunction; within 6 weeks of surgery (Chaves et al., 2002), although (4) confusion or mild memory impairment. These criteria intervals exceeding 3 years have been reported (Koike have a very high inter-rater reliability for the diagnosis et al., 2001). The neuropathy predominantly affects (Caine et al., 1997). This condition is most commonly the lower limbs and is both sensory and motor, with described in the setting of alcohol abuse when the individvariable involvement of each. While it may progress ual has had insufficient dietary intake of thiamine over a over years when untreated, rapid progression over long period of time. However, it is important to recall one intervals as short as 3 days may mimic Guillain–Barre´ of the three cases in the initial description in 1881 by Carl syndrome (Koike et al., 2001; Chang et al., 2002). ElecWernicke was the consequence of esophageal damage trophysiologic studies reveal that this neuropathy is axofrom sulfuric acid ingestion with associated refractory nal in nature with markedly reduced amplitudes of emesis (Wernicke, 1881). The mechanism by which compound motor action potential and sensory nerve bariatric surgery leads to thiamine deficiency, whether action potentials, especially in the lower extremities resulting in an encephalopathy or peripheral neuropathy, (Koike et al., 2001). is almost certainly inadequate vitamin repletion attending Thiamine deficiency can be confirmed by assessing persistent, intractable vomiting. the thiamine pyrophosphate effect in erythrocyte transIn 1977, Printen and Mason reported four patients ketolase studies (Boni et al., 1980). The clinical constelwho developed peripheral neuropathy and protracted lation coupled with the response to parenteral thiamine, emesis following gastric operations for obesity especially with the features of the encephalopathy, may (Printen and Mason, 1977). In this and other early reports prove sufficiently diagnostic. Additionally, magnetic of the neurologic complications occurring with gastric resonance imaging (MRI) of the brain may show characbypass, the specific cause of the neurologic disorder teristic abnormalities, in particular, hyperintense signal remained uncertain. In 1981, Ayub and colleagues abnormalities on T2-weighted images in the dorsomedial described confusion, slurred speech, and unsteadiness thalamic nuclei, periaqueductal gray matter, and mamin seven of 110 patients undergoing bariatric surgery millary bodies (Cirignotta et al., 2000; Toth and Voll, (Ayub et al., 1981). The authors listed several potential 2001). Both the encephalopathy and peripheral neuropaetiologies, including nutritional disturbances, metabolic thy of thiamine deficiency may occur despite oral supabnormalities, medication side-effects, lactic acidosis, plementation with thiamine as emesis may preclude or potential “toxic” bowel product attributable to a effective absorption. Substantial functional recovery micro-organism (Ayub et al., 1981). Some, if not all, of typically occurs within 3–6 months of the initiation of these cases were likely due to thiamine deficiency. Over therapy (Koike et al., 2001); however, neurologic recovthe past three decades, several reports linked the appearery may be incomplete (Salas-Salvado et al., 2000), parance of Wernicke’s encephalopathy with surgery for ticularly if the nature of the disorder is not recognized morbid obesity (Rothrock and Smith, 1981; Haid et al., promptly. Physical therapy for the peripheral neuropathy 1982; MacLean, 1982; Milius et al., 1982; Villar and is also recommended (Chaves et al., 2002).

590

J.R. BERGER AND D. SINGHAL hematologic manifestations may not accompany the Copper deficiency myelopathy neurologic syndrome (Kumar et al., 2003, 2004b). Copper deficiency myelopathy (CDM) was first reported Serum copper and serum ceruloplasmin levels have in 2001 and over the last decade, it has become an consistently been found to be low and of statistical sigincreasingly recognized myelopathy that most comnificance. Urinary copper level excretion is frequently monly occurs as a consequence of prior gastrointestinal low (when dietary copper is low). For assessment of surgery. The clinical and radiologic picture is usually metabolically active copper stores, activity of copper indistinguishable from subacute combined degeneration enzymes such as erythrocyte superoxide dismutase (SCD) (Kumar et al., 2004) due to vitamin B12 (cobalamin) and platelet or leukocyte cytochrome c oxidase may be deficiency, and the two may coexist (Juhasz-Pocsine et al., utilized (Kumar and Weimar, 2010). Electrophysiologic 2007). Other, less frequently reported and less clearly tests reveal an axonal sensorimotor peripheral causally related neurologic associations of acquired copneuropathy (to varying degrees). Somatosensory per deficiency include isolated peripheral neuropathy, potentials are abnormal with impaired central conducmotor neuron disease, myopathy, cerebral demyelination, tion being a key finding (Jaiser and Winston, 2010). cognitive dysfunction, and optic neuropathy. Spinal MRI shows an augmented T2 signal involving Jaiser and Winston (2010) reviewed 55 cases in the the dorsal column in posterior cervical or thoracic literature – previous upper gastrointestinal surgery was cord in nearly half of the cases with no contrast enhancethe commonest reported cause, being implicated in almost ment (22 cases) (Kumar, 2006); rarely, signal changes half the cases. Of these, nine cases were attributable to involving the lateral column may be seen (Kumar bariatric surgery. The interval between upper gastrointeset al., 2003). tinal surgery and symptom onset ranged from 5 to 26 Although there have been no studies that address the years in the bariatric group (mean 11.4 years). Likely cause most appropriate dose and duration of copper supplefor copper deficiency is greater reduction in the effective mentation, based on a review of all articles addressing absorption area for copper (postoperatively, food this subject, oral copper supplementation is the preferred bypasses most of the stomach and the entire duodenum). route of supplementation. Oral supplementation equivCopper is a component of numerous metalloenzymes alent to doses ranging from 2 to 8 mg of elemental copand proteins that have a key role in maintaining the per per day have been recommended by different structure and function of the nervous system. It is a constitauthors (Kumar, 2006; Jaiser and Winston, 2010). Pracuent of cytochrome oxidase (oxidative phosphorylation), tice at Mayo Clinic involves 6 mg/day of elemental copsuperoxide dismutase (antioxidant defense), ceruloplasmin per orally for a week, 4 mg/day for the second week, and (iron metabolism), tyrosinase (melanin synthesis), and 2 mg/day thereafter. Periodic assessment of serum copdopamine b-monooxygenase (catecholamine synthesis) per is essential to determine adequacy of replacement (Kumar et al., 2004a). Based on similarities to SCD due and the most appropriate long-term administration stratto vitamin B12 deficiency and copper deficiency myelopaegy is recommended (Winston and Jaiser, 2008). Since thy, it has been hypothesized that methionine synthase zinc can interfere with copper absorption, care must be and S-adenosylhomocysteine hydrolase (another enzyme taken to avoid copper preparations containing significant involved in the cycle) may depend on copper. Dysfunction quantities of zinc, such as multivitamin tablets. As the in the methylation cycle and associated failure of myelin underlying risk factor of decreased copper absorption maintenance could therefore explain the clinical and cannot be eliminated in cases of bariatric surgery, a short radiologic congruence between CDM and SCD, but it 5 day course of parenteral therapy (to facilitate rapid norremains to be proven with direct biochemical evidence malization of body copper stores) followed by indefinite (Kumar et al., 2006; Kumar and Weimar, 2010). oral copper supplementation has been recommended. Most common neurologic manifestation in adults is a Whilst treatment causes prompt and full resolution of myeloneuropathy with gait difficulties (primarily due to hematologic abnormalities, the neurologic deficits merely sensory ataxia due to dorsal column dysfunction), spasstabilize (51% of cases) or partially improve (49% cases). tic gait and lower limb paresthesias; urinary symptoms Hence, it is crucial to avoid delays in diagnosis and treatare infrequent. Examination usually shows a spastic ment to avert potentially irreversible neurologic deterioraparaparesis or tetraparesis with a truncal sensory level tion (Jaiser and Winston, 2010). for dorsal column modalities. A sensory/motor neuropImprovement when present is often subjective and athy frequently coexists and manifests as depression of preferentially involves sensory symptoms. There are distal reflexes and superimposed sensory impairment in some reports of definite improvement in the neurologic a glove and stocking distribution. Although the hematodeficits, nerve conduction studies, evoked potential logic hallmark of copper deficiency is anemia and neustudies, and MRI T2 cord signal changes with normalitropenia, it is being increasingly recognized that zation of serum copper (Kumar, 2006).

THE NEUROLOGIC COMPLICATIONS OF BARIATRIC SURGERY 591 colleagues (2009) have reported that the risk of vitamin Other nutritional and metabolic disorders deficiencies differs significantly based on the type of Absorption of vitamin B12 is complex and requires the bariatric surgery – compared with gastric bypass, duodepresence of intrinsic factor derived from gastric parietal nal switch may be associated with a greater risk of thiacells, acid gastric pH, and absorption in the ileum. mine, vitamin A and D deficiencies in the first year after Bariatric surgery may interfere with several of these the procedure. Hence, patients who undergo these two mechanisms. As liver stores of cyanocobalamin are sufsurgical interventions may require different monitoring ficient to allow for years of dietary insufficiency, these and supplementation regimens in the first year after surfeatures may not appear for long periods of time. A low gery. In addition to vitamin D, patients undergoing gasserum vitamin B12 level has been observed in as many as tric bypass may also be at risk of depletion of another 70% patients undergoing gastric bypass, and vitamin B12 fat-soluble vitamin, vitamin E (Provenzale et al., 1992), deficiency in more than 30% (Amaral et al., 1985). which is also associated with neurologic manifestations The prototypical neurologic disorder occurring with (Satya-Murti et al., 1986). cyanocobalamin deficiency is subacute combined degenBoldery and colleagues have reported two cases eration in which the peripheral nerves and posterior colimplicating selenium deficiency as a cause of heart failumns of the spinal cord are chiefly affected. A large ure in patients after gastric bypass surgery; selenium number of neurologic symptoms and signs have been deficiency is recognized as a cause of Keshan associated with cyanocobalamin deficiency, including, disease, also known as nutritional cardiomyopathy. paresthesias, loss of cutaneous sensation, weakness, However, in postbypass surgery patients with heart faildecreased reflexes, spasticity, ataxia, incontinence, loss ure, obstructive sleep apnea and thiamine deficiency of vision, dementia, psychoses, and altered mood must also be considered as potential explanations (Healton et al., 1991). Subacute combined degeneration (Boldery et al., 2007). has been reported after partial gastrectomy (Weir and Neurologic disorders consequent to rapid fat metabGatenby, 1963; Williams et al., 1969). The infrequency olism or the result of multiple nutritional and metabolic of disorders related to vitamin B12 has suggested to some factors have been reported, but remain unproven. Feit investigators that vitamin B12 deficiency is seldom clinand colleagues reported two patients who developed a ically relevant in the postgastric bypass patient (Brolin severe polyneuropathy chiefly affecting position et al., 1998); however, Bloomberg and colleagues sense associated with ataxia and pseudochorea within (2005) reviewed the literature and reflected on the need 3 months of gastric partitioning for morbid obesity for further studies to evaluate the clinical significance of (Feit et al., 1982). In one patient who died, autopsy nutritional deficiencies and establish guidelines for revealed extensive demyelination associated with extensupplementation. sive accumulation of lipofuscin in anterior horn cells Low plasma folate is seen in up to 42% of persons and dorsal root ganglia and lipid in Schwann cells (Feit undergoing gastric bypass followed for 3 years et al., 1982). The authors suggested that a toxin from (Halverson, 1986). Folate deficiency with an attendant rapid fat catabolism or loss of carnitine were responsible peripheral neuropathy would not be unexpected. Howrather than thiamine or another vitamin deficiency ever, the literature does not suggest that it is a common (Feit et al., 1982). Similarly, Paulson and colleagues problem and some investigators argue that it is not clinireported six patients with a clinical picture characterized cally relevant (Brolin et al., 1998). As with vitamin B12, oral by confusion, abnormal behavior, profound leg weakfolate supplementation appears effective in maintaining ness, diminished or absent muscle stretch reflexes, and levels within the normal range (Brolin et al., 1991). in three, ophthalmoplegia, or nystagmus (Paulson Niacin deficiency and pellagra has occurred after et al., 1985). While thiamine deficiency was considered gastroplasty (Lopez et al., 2000). This syndrome is charin the differential diagnosis, the authors believed that acterized by a symmetrical rash on sun-exposed areas this disorder was likely the consequence of rapid with hyperkeratosis, hyperpigmentation, and desquamametabolism of fat in obesity (Paulson et al., 1985). tion. Glossitis, diarrhea, fatigue, hallucinations, and Other investigators have proposed that some of the neuencephalopathy are also features of the disorder. rologic complications which follow bariatric surgery, Symptomatic hypocalcemia secondary to vitamin D such as the peripheral neuropathy or psychosis, arise deficiency after gastric bypass has been described from multifactorial causes (Seehra et al., 1996; (Marinella, 1999). Marinella reported a patient who Thaisetthawatkul et al., 2004). developed carpopedal spasms, intermittent facial twitchRecurrent spells of encephalopathy characterized by ing and ophthalmoplegia in association with hypocalceconfusion, behavioral abnormalities, weakness, lethargy, mia years after gastric bypass who responded to calcium ataxia, and dysarthria occurring with lactic acidosis may repletion (Marinella, 1999). Interestingly, Aasheim and occur after jejunoileostomy for morbid obesity (Dahlquist

592

J.R. BERGER AND D. SINGHAL

et al., 1984). This same disorder has been described in individuals with short bowel syndrome (Stolberg et al., 1982). It is precipitated by high carbohydrate diets. The neurologic symptoms occur in association with elevated concentrations of D-lactate in blood, urine, and stool (Dahlquist et al., 1984). The elevated levels of D-lactate are believed to result from fermentation of carbohydrates in the colon or bypassed segment of the small bowel (Dahlquist et al., 1984). Minerals (calcium, phosphorus, and magnesium) and trace elements (zinc, iodine, copper, manganese, fluoride, chromium, molybdenum, selenium, and iron) have seldom been studied in patients following gastric bypass (Bal et al., 2010).

Miscellaneous disorders Some of neurologic complications of bariatric surgery that are not ascribable to micronutrient insufficiency, although not exclusive to this setting, remain relatively unique. Unilateral lower compartment syndrome occurring in the immediate postoperative period may be observed (Gorecki et al., 2002). This syndrome arises from ischemic injury to tissues in the anterior compartment with progressive increase in pressure and ultimately nerve injury. Prompt recognition and fasciotomy are essential for a favorable outcome (Gorecki et al., 2002). Lumbosacral plexopathy with an asymmetric peripheral neuropathy has been reported following gastric partitioning (Harwood et al., 1987). The specific pathogenesis of this disorder have not been addressed (Harwood et al., 1987) and whether it is due to a micronutrient deficiency or other cause remains uncertain. Similarly, Thaisetthawatkul noted plexopathy in five of his 435 patients (Thaisetthawatkul, 2003). A wide variety of musculoskeletal symptoms have been reported to occur in association with gastric bypass (Ginsberg et al., 1979). Ginsberg and colleagues reported 13 patients who developed a constellation of arthritis, polyarthalgias, myalgias, and morning stiffness 3 weeks to 48 months after undergoing jejunoileal shunt surgery (Ginsberg et al., 1979). These symptoms tended to be transient in nature and the demonstration of circulating immune complexes suggested an autoimmune process (Ginsberg et al., 1979).

CONCLUSIONS In summary, neurologic complications occurring in the setting of bariatric surgery are not uncommon. These complications have been reported in as many as 5–10% of patients undergoing surgery for obesity (Berger, 2004). Any part of the neuraxis, including brain, cerebellum, spinal cord, peripheral nerve, and muscle, may

be involved by these complications. Most of these neurologic complications are the consequence of micronutrient deficiency. Following bariatric surgery, at 6 month intervals during the first 3 years, then once yearly ferritin, zinc, copper, magnesium, total 25-hydroxyvitamin D, folate, whole blood thiamine, vitamin B12 and 24 h urinary calcium levels should be checked (Bal et al., 2010). Physicians need to be particularly alert to Wernicke’s encephalopathy developing after bariatric surgery as it is a medical emergency and demands rapid diagnosis and intervention. Copper deficiency is an increasingly recognized complication (Jaiser and Winston, 2010) (and expected to become even more frequent with increase in number of bariatric surgeries) – early recognition and treatment is of utmost importance because it can prevent neurologic deterioration (VidetGibou et al., 2010). Physicians caring for patients who have undergone bariatric surgery should be familiar with the constellation of neurologic disorders that may occur.

REFERENCES Aasheim ET, Bjorkman S, Sevik TT et al. (2009). Vitamin status after bariatric surgery: a randomized study of gastric bypass and duodenal switch. Am J Clin Nutr 90: 15–22. Abarbanel JM, Berginer VM, Osimani A et al. (1987). Neurologic complications after gastric restriction surgery for morbid obesity. Neurology 37: 196–200. Allison DB, Fontaine KR, Manson JE et al. (1999). Annual deaths attributable to obesity in the United States. JAMA 282: 1530–1538. Amaral JF, Thompson WR, Caldwell MD et al. (1985). Prospective hematologic evaluation of gastric exclusion surgery for morbid obesity. Ann Surg 201: 186–193. Ayub A, Faloon WW, Heinig RE (1981). Encephalopathy following jejunoileostomy. JAMA 246: 970–973. Azagra JS, Goergen M, Ansay J et al. (1999). Laparoscopic gastric reduction surgery. Preliminary results of a randomized prospective trial of laparoscopic vs open vertical banded gastroplasty. Surg Endosc 13: 555–558. Bal B, Koch TR, Finelli FC et al. (2010). Managing medical and surgical disorders after divided Roux-en-Y gastric bypass surgery. Nat Rev Gastroenterol Hepatol 7: 320–334. Berger JR (2004). The neurological complications of bariatric surgery. Arch Neurol 61: 1185–1189. Bloomberg RD, Fleishman A, Nalle JE et al. (2005). Nutritional deficiencies following bariatric surgery: what have we learned? Obes Surg 15: 145–154. Boldery R, Fielding G, Rafter T et al. (2007). Nutritional deficiency of selenium secondary to weight loss (bariatric) surgery associated with life-threatening cardiomyopathy. Heart Lung Circ 16: 123–126. Boni L, Kieckens L, Hendrikx A (1980). An evaluation of a modified erythrocyte transketolase assay for assessing thiamine nutritional adequacy. J Nutr Sci Vitaminol (Tokyo) 26: 507–514.

THE NEUROLOGIC COMPLICATIONS OF BARIATRIC SURGERY Bozbora A, Coskun H, Ozarmagan S et al. (2000). A rare complication of adjustable gastric banding: Wernicke’s encephalopathy. Obes Surg 10: 274–275. Brolin RE, Gorman RC, Milgrim LM et al. (1991). Multivitamin prophylaxis in prevention of post-gastric bypass vitamin and mineral deficiencies. Int J Obes 15: 661–667. Brolin RE, Gorman JH, Gorman RC et al. (1998). Are vitamin B12 and folate deficiency clinically important after Rouxen-Y gastric bypass? J Gastrointest Surg 2: 436–442. Buchwald H, Avidor Y, Braunwald E et al. (2004). Bariatric surgery: a systematic review and meta-analysis. JAMA 292: 1724–1737. Caine D, Halliday GM, Kril JJ et al. (1997). Operational criteria for the classification of chronic alcoholics: identification of Wernicke’s encephalopathy. J Neurol Neurosurg Psychiatry 62: 51–60. Chang CG, Helling TS, Black WE et al. (2002). Weakness after gastric bypass. Obes Surg 12: 592–597. Chaves LC, Faintuch J, Kahwage S et al. (2002). A cluster of polyneuropathy and Wernicke–Korsakoff syndrome in a bariatric unit. Obes Surg 12: 328–334. Cirignotta F, Manconi M, Mondini S et al. (2000). Wernicke– Korsakoff encephalopathy and polyneuropathy after gastroplasty for morbid obesity: report of a case. Arch Neurol 57: 1356–1359. Clegg AJ, Colquitt J, Sidhu MK et al. (2003). Clinical and cost effectiveness of surgery for morbid obesity: a systematic review and economic evaluation. Int J Obes Relat Metab Disord 27: 1167–1177. Dahlquist NR, Perrault J, Callaway CW et al. (1984). D-Lactic acidosis and encephalopathy after jejunoileostomy: response to overfeeding and to fasting in humans. Mayo Clin Proc 59: 141–145. Davis MM, Slish K, Chao C et al. (2006). National trends in bariatric surgery 1996–2002. Arch Surg 141: 71–74, discussion 75. Deitel M, Shikora SA (2002). The development of the surgical treatment of morbid obesity. J Am Coll Nutr 21: 365–371. Fang J (2003). The cost-effectiveness of bariatric surgery. Am J Gastroenterol 98: 2097–2098. Feit H, Glasberg M, Ireton C et al. (1982). Peripheral neuropathy and starvation after gastric partitioning for morbid obesity. Ann Intern Med 96: 453–455. Fisher BL, Schauer P (2002). Medical and surgical options in the treatment of severe obesity. Am J Surg 184: 9S–16S. Flegal KM, Carroll MD, Ogden CL et al. (2002). Prevalence and trends in obesity among US adults 1999–2000. JAMA 288: 1723–1727. Flegal KM, Carroll MD, Ogden CL et al. (2010). Prevalence and trends in obesity among US adults 1999–2008. JAMA 303: 235–241. Freedman DH (2011). How to fix the obesity crisis. Sci Am 304: 40–47. Ginsberg J, Quismorio FP Jr, DeWind LT et al. (1979). Musculoskeletal symptoms after jejunoileal shunt surgery for intractable obesity. Clinical and immunologic studies. Am J Med 67: 443–448.

593

Gollobin C, Marcus WY (2002). Bariatric beriberi. Obes Surg 12: 309–311. Gorecki PJ, Cottam D, Ger R et al. (2002). Lower extremity compartment syndrome following a laparoscopic Rouxen-Y gastric bypass. Obes Surg 12: 289–291. Haid RW, Gutmann L, Crosby TW (1982). Wernicke– Korsakoff encephalopathy after gastric plication. JAMA 247: 2566–2567. Halverson J (1986). Micronutrient deficiencies after gastric bypass for morbid obesity. Am Surg 52: 594–598. Halverson JD (1992). Metabolic risk of obesity surgery and longterm follow-up. Am J Clin Nutr 55 (2 Suppl): 602S–605S. Harwood SC, Chodoroff G, Ellenberg MR (1987). Gastric partitioning complicated by peripheral neuropathy with lumbosacral plexopathy. Arch Phys Med Rehabil 68: 310–312. Healton EB, Savage DG, Brust JC et al. (1991). Neurologic aspects of cobalamin deficiency. Medicine (Baltimore) 70: 229–245. Higa KD, Boone KB, Ho T et al. (2000). Laparoscopic Roux-en-Y gastric bypass for morbid obesity: technique and preliminary results of our first 400 patients. Arch Surg 135: 1029–1033, discussion 1033–1034. Jaiser SR, Winston GP (2010). Copper deficiency myelopathy. J Neurol 257: 869–881. Juhasz-Pocsine K, Rudnicki SA, Archer RL et al. (2007). Neurologic complications of gastric bypass surgery for morbid obesity. Neurology 68: 1843–1850. Ke ZJ, DeGiorgio LA, Volpe BT et al. (2003). Reversal of thiamine deficiency-induced neurodegeneration. J Neuropathol Exp Neurol 62: 195–207. Khan LK, Serdula MK, Bowman BA et al. (2001). Use of prescription weight loss pills among US adults in 1996–1998. Ann Intern Med 134: 282–286. Koike H, Misu K, Hattori N et al. (2001). Postgastrectomy polyneuropathy with thiamine deficiency. J Neurol Neurosurg Psychiatry 71: 357–362. Kramer LD, Locke GE (1987). Wernicke’s encephalopathy. Complication of gastric plication. J Clin Gastroenterol 9: 549–552. Kumar N (2006). Copper deficiency myelopathy (human swayback). Mayo Clin Proc 81: 1371–1384. Kumar N, Weimar LH (2013). Copper deficiency myeloneuropathy. S. Gilman (Ed.). Neurology Medlink, www. medlink.com/medlinkcontent.asp. Accessed August 10, 2013. Kumar N, Gross Jr JB, Ahlskog JE (2003). Myelopathy due to copper deficiency. Neurology 61: 273–274. Kumar N, Crum B, Petersen RC et al. (2004a). Copper deficiency myelopathy. Arch Neurol 61: 762–766. Kumar N, Gross Jr JB, Ahlskog JE (2004b). Copper deficiency myelopathy produces a clinical picture like subacute combined degeneration. Neurology 63: 33–39. Kumar N, Ahlskog JE, Klein CJ et al. (2006). Imaging features of copper deficiency myelopathy: a study of 25 cases. Neuroradiology 48: 78–83. Livingston EH (2002). Obesity and its surgical management. Am J Surg 184: 103–113.

594

J.R. BERGER AND D. SINGHAL

Lobstein TJ, James WP, Cole TJ (2003). Increasing levels of excess weight among children in England. Int J Obes Relat Metab Disord 27: 1136–1138. Lopez JF, Halimi S, Perillat Y (2000). Pellagra-like erythema following vertical banded gastroplasty for morbid obesity. Ann Chir 125: 297–298. MacLean JB (1982). Wernicke’s encephalopathy after gastric plication. JAMA 248: 1311. Marinella MA (1999). Ophthalmoplegia: an unusual manifestation of hypocalcemia. Am J Emerg Med 17: 105–106. Milius G, Rose S, Owen DR et al. (1982). Probable acute thiamine deficiency secondary to gastric partition for morbid obesity. Nebr Med J 67: 147–150. Mokdad AH, Ford ES, Bowman BA et al. (2003). Prevalence of obesity diabetes and obesity-related health risk factors 2001. JAMA 289: 76–79. Mokdad AH, Marks JS, Stroup DF et al. (2004). Actual causes of death in the United States 2000. JAMA 291: 1238–1245. National Heart, Lung, and Blood Institute (1998). Clinical Guidelines on the Identification Evaluation and Treatment of Overweight and Obesity in Adults: The Evidence Report. National Heart Lung and Blood Institute, Rockville, MD. National Institutes of Health [no authors listed] (1992). Gastrointestinal surgery for severe obesity: National Institutes of Health Consensus Development Conference Statement. Am J Clin Nutr 55 (Suppl 2): S615–S619. Oczkowski WJ, Kertesz A (1985). Wernicke’s encephalopathy after gastroplasty for morbid obesity. Neurology 35: 99–101. Paulson GW, Martin EW, Mojzisik C et al. (1985). Neurologic complications of gastric partitioning. Arch Neurol 42: 675–677. Pope GD, Birkmeyer JD, Finlayson SR (2002). National trends in utilization and in-hospital outcomes of bariatric surgery. J Gastrointest Surg 6: 855–860, discussion 861. Pories WJ, MacDonald Jr KG, Morgan EJ et al. (1992). Surgical treatment of obesity and its effect on diabetes: 10-y follow-up. Am J Clin Nutr 55: 582S–585S. Printen KJ, Mason EE (1977). Gastric bypass for morbid obesity in patients more than fifty years of age. Surg Gynecol Obstet 144: 192–194. Provenzale D, Reinhold RB, Golner B et al. (1992). Evidence for diminished B12 absorption after gastric bypass: oral supplementation does not prevent low plasma B12 levels in bypass patients. J Am Coll Nutr 11: 29–35. Rothrock JF, Smith MS (1981). Wernicke’s disease complicating surgical therapy for morbid obesity. J Clin Neuroophthalmol 1: 195–199.

Salas-Salvado J, Garcia-Lorda P, Cuatrecasas G et al. (2000). Wernicke’s syndrome after bariatric surgery. Clin Nutr 19: 371–373. Satya-Murti S, Howard L, Krohel G et al. (1986). The spectrum of neurologic disorder from vitamin E deficiency. Neurology 36: 917–921. Schauer PR, Ikramuddin S, Gourash W et al. (2000). Outcomes after laparoscopic Roux-en-Y gastric bypass for morbid obesity. Ann Surg 232: 515–529. Seehra H, MacDermott N, Lascelles RG et al. (1996). Wernicke’s encephalopathy after vertical banded gastroplasty for morbid obesity. BMJ 312: 434. Skroubis G, Sakellaropoulos G, Pouggouras K et al. (2002). Comparison of nutritional deficiencies after Roux-en-Y gastric bypass and after biliopancreatic diversion with Roux-en-Y gastric bypass. Obes Surg 12: 551–558. Stolberg L, Rolfe R, Gitlin N et al. (1982). d-Lactic acidosis due to abnormal gut flora: diagnosis and treatment of two cases. N Engl J Med 306: 1344–1348. Thaisetthawatkul P (2003). Peripheral neuropathy following gastric bypass surgery. In: 55th Annual Meeting of the American Academy of Neurology, Honolulu, Hawaii. Thaisetthawatkul P, Collazo-Clavell ML, Sarr MG et al. (2004). A controlled study of peripheral neuropathy after bariatric surgery. Neurology 63: 1462–1470. Toth C, Voll C (2001). Wernicke’s encephalopathy following gastroplasty for morbid obesity. Can J Neurol Sci 28: 89–92. Trus TL, Pope GD, Finlayson SR (2005). National trends in utilization and outcomes of bariatric surgery. Surg Endosc 19: 616–620. Videt-Gibou D, Belliard S, Rivalan J et al. (2010). Acquired copper deficiency myelopathy. Rev Neurol (Paris) 166: 639–643. Villar HV, Ranne RD (1984). Neurologic deficit following gastric partitioning: possible role of thiamine. JPEN J Parenter Enteral Nutr 8: 575–578. Weir D, Gatenby P (1963). Subacute combined degeneration of cord after partial gastrectomy. Br Med J 2: 1175–1176. Wernicke C (1881). Lehrbuch der Gehirnkrankheiten fur Aerzte und Studirende. Theodor Fischer, Kassel. Wilhelm C (2000). Growing the market for anti-obesity drugs. Chem Market Rep, FR23–FR24. Williams JA, Hall GS, Thompson AG et al. (1969). Neurological disease after partial gastrectomy. Br Med J 3: 210–212. Winston GP, Jaiser SR (2008). Copper deficiency myelopathy and subacute combined degeneration of the cord – why is the phenotype so similar? Med Hypotheses 71: 229–236. Yale C (1989). Gastric surgery for morbid obesity. Complications and long-term weight control. Arch Surg 124: 941–946.

Handbook of Clinical Neurology, Vol. 120 (3rd series) Neurologic Aspects of Systemic Disease Part II Jose Biller and Jose M. Ferro, Editors © 2014 Elsevier B.V. All rights reserved

Chapter 40

Neurologic manifestations of inflammatory bowel diseases JOSE´ M. FERRO1*, SOFIA N. OLIVEIRA2, AND LUIS CORREIA3 Neurology Service, Department of Neurosciences, Hospital de Santa Maria, University of Lisbon, Lisbon, Portugal

1

2 3

Department of Neurology, Hospital da Luz, Lisbon, Portugal

Department of Gastroenterology and Hepatology, Hospital de Santa Maria, University of Lisbon, Lisbon, Portugal

INTRODUCTION Inflammatory bowel diseases (IBD) are chronic, relapsing and remitting inflammatory conditions affecting the digestive system, idiopathic in their etiology, comprising two main distinctive diseases, ulcerative colitis (UC) and Crohn’s disease (CD). In UC the inflammation is restricted to the mucosa of the colon, with an almost invariable involvement of the rectum and a variable continuous proximal extension. CD is a condition that involves transmurally any segment of the digestive tract, from mouth to anus, with spared segments and skip lesions. In CD aphthoid ulcers become transmural fissures and, after a variable time interval, inflammation evolves to cause destruction and disability settles in, with fistula or fibrotic stenosis formation, as well as the potential for abscess formation or secondary occlusion (Cosnes et al., 2002). About onethird of CD patients develop perianal disease (Schwartz et al., 2002), from anal fissures to simple or complex perianal fistulae or secondary abscesses. Both diseases can be phenotypically classified according to age, location, extension, behavior, and activity, by several clinical and endoscopic classifications. The most important of them is the Montreal classification, which allows for prognostic validation (Silverberg et al., 2005). Between 10% and 15% of colitis patients resist classification after applying strict clinical, endoscopic, radiologic, and histologic criteria, defining an intermediate condition known as unclassified colitis, or, as named by the pathologist in possession of a colectomy specimen, indeterminate colitis (Lennard-Jones, 1989). The incidence of IBD varies between different areas of the globe, though with a north–south gradient, well documented in Europe and North America. In general,

it is recognized that there is a stable incidence of UC and a rising incidence of CD, with the former maintaining a higher incidence. In a systematic review (Molodecky et al., 2012), the highest reported incidence of UC was 24 and 19.2 per 100 000 person-years and 12.7 and 20.2 per 100 000 patient-years in CD, in Europe and the US, respectively. The pathogenesis of IBD remains unclear. It is generally accepted that the disease appears in non-Mendelian genetically susceptible persons (Satsangi et al., 1997). The genetic predisposition can be demonstrated by an elevated concordance in twins (Orholm et al., 2000) and firstdegree relatives (Lahari et al., 2001), mutations in the NOD2/CARD15 gene (Kugathasan et al., 2004), or association with certain characteristic major histocompatibility complex elements (Bouma et al., 1997). Exposure to environmental risk factors such as infections (acute gastroenteritis, dysbiosis) (Porter et al., 2008), antibiotics, food antigens (breastfeeding, sugar or fat excess, fiber deficits, milk proteins, etc.) or nonsteroidal anti-inflammatory drugs, particularly in infancy, produces pathologic changes in the innate and adaptive immune system; these include changes in the mucosal barrier function, against luminal bacteria or antigens, eliciting an inappropriate immune response against microorganisms that generally would not generate that kind of response. Besides the classic gastrointestinal manifestations, namely bloody diarrhea in UC or abdominal pain, fever, bowel habit change, or perianal disease in CD, a variable number of IBD patients present with complaints located outside the gastrointestinal tract, such as joints, mouth, eyes, skin, and liver, with some more rare manifestations. These are called the IBD extraintestinal manifestations (EIM) (Peyrin-Biroulet et al., 2011).

*Correspondence to: Jose´ M. Ferro, Department of Neurosciences, Hospital de Santa Maria, Av. Prof. Egas Moniz, 1649-035 Lisbo, Portugal. Tel: þ351-21-795-7474, Fax: þ351-21-795-7474, E-mail: [email protected]

596

J.M. FERRO ET AL.

EIM can have multiple mechanisms, such as metabolic complications of the disease (e.g., osteoporosis, nephrolithiasis in ileal CD), adverse effects of the medications (e.g., corticosteroid myopathy or anti-TNF demyelination), but mainly immune-mediated mechanisms. This group, classic EIM, has a phenotypic preference for colonic disease (UC and colonic CD) and can be divided in two subgroups: EIM related to intestinal activity (aphthous ulcers, peripheral arthritis or erythema nodosum) or EIM independent of the activity of the intestinal disease (axial arthritis, pyoderma gangrenosum, primary sclerosing cholangitis). Finally, it is necessary to recognize that there is an IBD-independent group of immune-mediated diseases that can coexist with greater prevalence in IBD, such as celiac or thyroid disease, vitiligo, diabetes, or, more rarely, lupus. In general, as represented by a cohort of 950 prospectively studied Swiss patients (Vavricka et al., 2011), EIM occur more frequently in CD, both globally (43%, versus 31% in UC) and specific to the different manifestations (arthritis 33% versus 21%; aphthous stomatitis 10% versus 4%; uveitis 6% versus 4%; erythema nodosum 6% versus 3%; ankylosing spondyloarthropathy 6% versus 2%) with the exception of primary sclerosing cholangitis (CD 1% versus UC 4%).

NEUROLOGIC MANIFESTATIONS IN INFLAMMATORY BOWEL DISEASE (Tables 40.1 and 40.2)

Table 40.1 Peripheral nervous system manifestations of inflammatory bowel disease Peripheral neuropathy Axonal, large fibers Axonal, small fibers Demyelinating, acute Demyelinating, chronic Mononeuropathy Multifocal motor Myopathy Dermatomyositis Polymyositis Rimmed vascular myopathy Granulomatosis myositis Myasthenia Cranial neuropathy Melkersson Rosenthal syndrome Optic neuritis Hearing loss Sixth nerve palsy

Table 40.2 Central nervous system manifestations of inflammatory bowel diseases Cerebrovascular Ischemic arterial stroke Large artery disease Small vessel disease Cardioembolism, patent foramen ovale Cardioembolism, endocarditis Vasculitis Associated with anti-TNF-a therapy Cerebral venous thrombosis Demyelinating Multiple sclerosis Asymptomatic white matter lesions Related to anti-TNF-a therapy Spinal cord Myelopathy Spinal empyema Other Seizures Headache Encephalopathy

Prevalence Neurologic and psychiatric involvement in inflammatory bowel disease (IBD) is rare though probably underreported. The true incidence is unknown, with values varying from 0.25% to 47.5% (Lossos et al., 1995; Elsehety and Bertorini, 1997; Oliveira et al., 2008; Sassi et al., 2009). In a series of 638 patients with IBD, Lossos et al. (1995) reported 19 patients (3%) with neurolgic involvement that preceded the onset of intestinal symptoms by up to 10 years in 26% and started up to 12 years after IBD presentation in the remaining patients. In 10% it was associated with IBD exacerbation and most patients (53%) exhibited other extraintestinal manifestations and complications. Elsehety and Bertorini (1997) reported a 33.2% incidence of neurologic and neuropsychiatric complications in 253 patients with pathologically confirmed Crohn’s disease, a much higher prevalence than that reported in other series. Two prospective cohort studies (Oliveira et al., 2008; Sassi et al., 2009) looked at the prevalence of peripheral neuropathy and found 13.4% and 8.8% respectively. Asymptomatic focal brain white matter lesions have also been found in MRI studies of patients with IBD as compared to healthy age-matched controls (43.1% versus 16.0%; RR 2.6, 95% CI 1.3–5.3) but their clinical significance is unclear (Geissler et al., 1995).

Pathophysiology Both the central and peripheral nervous systems can be affected in IBD. Several mechanisms are possibly

NEUROLOGIC MANIFESTATIONS OF INFLAMMATORY BOWEL DISEASES involved: malabsorption and nutritional deficiencies, metabolic agents, infections induced by immunosuppresion, side-effects of medication and iatrogenic complications of surgery, thromboembolism, immunologic abnormalities, and disturbances of the so-called “brain–gut axis,” referring to the neuronal influence on enteric disease and vice versa (Derbyshire, 2003; Konturek et al., 2004) via neuroendocrine pathways such as the hypothalamic– pituitary–adrenal axis, release of corticotropin and adrenal corticoid secretion, the autonomic nervous system and its effects on immune functions (Zois et al., 2010). It is important to separate immune- and nonimmunemediated mechanisms directly related to the disease itself from other causes, such as side-effects of medication and nutritional deficiencies.

PERIPHERAL NERVOUS SYSTEM The peripheral nervous system (PNS) is frequently affected in IBD and peripheral neuropathy is one of the most common complications (Lossos et al., 1995; Elsehety and Bertorini, 1997; Oliveira et al., 2008), reported in up to 31.5% of patients. However, wellknown causes of peripheral neuropathy such as vitamin deficiencies and secondary effects of medication should be differentiated from a primary involvement of the PNS in IBD, and prospective data are notably absent. Oliveira et al. (2008) identified peripheral neuropathy as the most common neurolgic complication in their cohort of IBD patients (36.6%), probably immune-mediated and therefore directly disease-related in 13.4%, the remainder being related to vitamin deficiency or metronidazole toxicity. Peripheral neuropathy appeared to be more frequent in UC (Lossos et al., 1995; Gondim et al., 2005) but recent studies have shown comparable incidence in UC and CD patients (Gondim et al., 2005; Oliveira et al., 2008). Several different phenotypes have been described including sensory, motor, autonomic, and mixed, both axonal and demyelinating, acute and chronic (Gondim et al., 2005). In the largest retrospective series (Gondim et al., 2005), demyelinating chronic inflammatory polyneuropathy, small and large-fiber axonal neuropathies were found in UC patients. An acute inflammatory demyelinating polyradiculopathy has also been described in UC (Lossos et al., 1995). Axonal polyneuropathy presents with sensory loss and dysestesias in glove and stocking distribution with decreased or absent ankle jerks. Small fiber sensory neuropathy presents with subjective numbness in the absence of objective abnormalities on electromyography and nerve conduction studies. Patients with IBD have also been found to have mononeuropathies or multifocal motor neuropathies (Gondim et al., 2005).

597

Patients with demyelinating neuropathies (particularly chronic inflammatory demyelinating polyneuropathy) respond better to immunomodulatory therapy that those with axonal neuropathy (Lossos et al., 1995; Gondim et al., 2005). Peripheral neuropathies are not related to disease activity and do not respond to treatment of the underlying IBD. An inflammatory myopathy has also been found in association with IBD (Gendelman et al., 1982; Bhigjee et al., 1987; Sowa, 1991). It is probably more frequent in CD than in UC, and in the series by Lossos et al. (1995) was responsible for 16% of cases of neurolgic dysfunction. Dermatomyositis, polymyositis, rimmed vacuole myopathy, and granulomatous myositis have all been described and may also be completely asymptomatic (Hayashi et al., 1991). Myositis in CD is probably an immune-mediated disease (Shimoyama et al., 2009) as the same CD68 þ macrophages that are found in the diseased colon are also present in the muscle. Involvement of the neuromuscular junction has only rarely been described in the context of IBD (Martin and Shah, 1991; Finnie et al., 1994). It is important to report that at least one patient with CD and myasthenia had significant improvement of gastrointestinal symptoms following thymectomy (Finnie et al., 1994).

CRANIAL NEUROPATHIES Melkersson–Rosenthal syndrome (recurrent facial nerve palsy with facial edema, tongue fissuring, and granulomas) has been described by several authors in IBD (Lloyd et al., 1994; Lossos et al., 1995). Other cranial neuropathies, such as optic neuritis (Sedwick et al., 1984; Lossos et al., 1995), can also be found in single case reports. Sensorineural hearing loss has also been described (Kumar et al., 2000; Akbayir et al., 2005), and is probably an under-recognized, immunologic manifestation of IBD. Of relevance also is the possibility of recovery with early treatment with steroids and immunosuppressive agents (Bachmeyer et al., 1998).

CEREBROVASCULAR COMPLICATIONS Patients with inflammatory bowel disease (IBD) have a remarkable thromboembolic tendency and are at increased risk of both venous and arterial thrombotic complications. In hospital series of IBD, the prevalence of arterial and venous thrombosis is around 4%, while in autopsy studies, this percentage may be more than 30%. The incidence of thrombotic complications ranges from 0.5% to 6.7% per year (Bernstein et al., 2001; Papa et al., 2003). IBD is also a risk factor for recurrent venous thromboembolism (Novacek et al., 2010).

598

J.M. FERRO ET AL.

In a cohort of 49 799 Danish patients with IBD compared with 477 504 members of the general population, patients with IBD had twice the incidence of deep venous thrombosis and pulmonary embolism. Relative risks were higher at young ages (hazard ratio 6.0 below age 20), though actual incidence increased with age (Kappelman et al., 2011). In a retrospective case control study including 17 487 IBD patients and 69 948 controls had an increased risk of arterial thrombotic events. In particular, women under the age of 40 exhibited a twofold higher risk for stroke (Ha et al., 2009). Thromboembolism is more frequent in IBD than in other chronic inflammatory or chronic bowel diseases (Miehsler et al., 2004). Strokes in patients with IBD were already reported in the 1930s. In 1986, Talbot and colleagues (1986) described a 1.3% prevalence of thromboembolic complications among 7199 patients with IBD observed during a 10 year period in a single institution. Among the 92 patients with thromboembolic complications, 61 had deep vein thrombosis or pulmonary embolism. Nine patients had cerebrovascular thrombotic events. There were no subarachnoid or intracerebral hemorrhages. Several other case series (Lossos et al., 1995; Elsehety and Bertorini, 1997; Barclay et al., 2010; Benavente and Morı´s, 2011; Cognat et al., 2011) and case reports of arterial ischemic or cerebral venous thrombosis have been published since then, with a frequency ranging from 0.6% to 4.7%. Cerebrovascular complications are somewhat more frequent in Crohn’s disease than in ulcerative colitis. They are not related to the duration or the severity of IBD, but cerebrovascular events are more frequent during bouts of inflammation. Rarely, they can antedate other manifestations of IBD.

complications. Vitamin deficiencies due to malabsorption, namely B12 and folate, cause hyperhomocysteinemia, in particular when combined with a MTHFR deficit. Cytokines, such as interleukin 1 and 6 and TNF-a can activate the coagulation cascade during periods of active inflammation. Other immune mechanisms include the coexistence of prothrombotic antibodies such as lupus anticoagulant, anticardiolipin antibodies, and atypical (non-MPO, non-PR3) ANCA antibodies, the latter in ulcerative colitis.

Arterial ischemic stroke Ischemic events can be both cerebral and ocular. Ischemic stroke occurs through several mechanisms: (1) large artery disease, including even a case of common carotid occlusion; (2) small vessel disease, e.g., corona radiata (Ogawa et al., 2011) or pontine lacunar infarcts; (3) cardioembolism, related to (a) paradoxical embolism though a patent foramen ovale in patients with lower limb, pelvic, or mesenteric venous thrombosis, either symptomatic or not, (b) endocarditis (Kreuzpaintner et al., 1992); (4) vasculitis.

Stroke and anti-TNF-a therapy A few cases of arterial ischemic complications have been reported as a complication of anti-TNF-a therapy (Vannucchi et al., 2011; Cohen et al., 2012). One of the authors (JMF) has observed two cases of deep intracerebral hemorrhage in young, nonhypertensive males with IBD treated with anti-TNF drugs. No underlying arteriovenous malformation, venous thrombosis, or other lesion was identified (Fig. 40.1).

Vasculitis Pathophysiology The prothrombotic state in IBD has multiple contributors, namely blood coagulation, platelets, endothelium, prothrombotic mutations, vitamin deficiencies, inflammation, and other immune mechanisms (Santos et al., 2001; Bermejo and Burgos, 2008. The hypercoagulation state is related to raised levels of factor V and VIII, fibrinogen, fibrinopeptide A and PAI-1, and decreased levels of protein S and antithrombin. Thrombocytosis is common in IDB, secondary both to anemia and inflammation. Platelet function is disturbed and Von Willebrand factor, a potent mediator of platelet adhesion and aggregation, is increased. In some patients endothelial dysfunction was reported. Prothrombotic mutations such as factor V Leiden and methylenetetrahydrofolate reductase (MTHFR) were identified in some IBD patients with cerebrovascular

Systemic and organ-specific vasculitis, namely cerebral, has been reported in association with IBD, especially with ulcerative colitis (Scheid and Teich, 2007). However, cerebral vasculitis is very rare and only a handful of cases, small case series, and a few nonsystematic reviews have been published (Martı´n de Carpi et al., 2007; Scheid and Teich, 2007). Vasculitis in IBD is immune-mediated, via genetic susceptibility and HLA status, T lymphocyte-mediated cytoxicity, or immune complex deposition (Scheid and Teich, 2007). In other cases IBD is associated with primary vasculitides (giant cell arteritis, Wegener, pANCA vasculitis) (Jacob et al., 1990; Ronchetto and Pistono, 1993; Gobron et al., 2010) or with other immune diseases (Cogan syndrome, thrombotic thrombocytopenic purpura, and lupus) (Chebli et al., 2000; Baron et al., 2002; Hisada et al., 2006; Ullrich et al., 2009).

NEUROLOGIC MANIFESTATIONS OF INFLAMMATORY BOWEL DISEASES

599

Fig. 40.1. MRI showing frontoparietal hemorrhagic stroke in a patient with inflammatory bowel disease, treated with anti-TNF-a agents.

The clinical manifestations of cerebral vasculities in IBD are protean and include headache (Holzer et al., 2009), cranial nerve palsies, focal deficits such as hemiparesis, sensory disturbances, aphasia and visual defects, multifocal signs and vigilance or cognitive troubles, isolated or in combination (Scheid and Teich, 2007). The onset can be acute or subacute. Presentation as a single stroke syndrome is infrequent. Levels of nonspecific inflammatory markers such as ESR or CRP may be elevated. Cerebrospinal fluid examination may reveal increased proteins or a mild pleiocytosis. Magnetic resonance imaging of the brain rarely shows large territorial infarcts. Acute lesions may be detected on diffusionweighted imaging (DWI). The most common findings are deep white matter or periventricular white matter lesions, detectable in FLAIR or T2 sequences (Scheid and Teich, 2007). These lesions are not specific and per se do not allow a definite diagnosis of vasculitis, which relies on the demonstration of inflammation on a vessel wall. In several cases reported in the literature no imaging or biopsy of the vessels was obtained and therefore the diagnosis of vasculitis is only probable. Intraarterial, MR (Schluter et al., 2004), or CT angiography can reveal multiple arterial stenoses or other aspects suggestive of vasculitis, but many of them can also be seen in noninflammatory vasculopathies and in the reversible cerebral vasoconstriction syndrome, in which the arterial stenoses usually regress in a follow-up angiography performed 6–8 weeks later. Moreover, angiography can be normal in vasculitis affecting only small arteries. In large artery vasculitis (Takayasu and giant cell arteritis), the temporal superficial, the carotid, and the axillary arteries are accessible to ultrasound which may demonstrate

inflammatory “halos.” Recently, appropriate MR sequences were reported to depict wall inflammation in medium-size intracerebral vessels, such as the proximal segments of the middle cerebral artery (Mandell et al., 2012). In a few cases of vasculitis associated with IBD, namely UC, necropsy of brain biopsy was obtained. In three cases necrotizing vasculitis was found (Glotzer et al., 1964; Nelson et al., 1986; Carmona et al., 2000), although in one of them hemorrhagic acute disseminated encephalomyelitis could not be ruled out (Glotzer et al., 1964). In other cases only a lymphocyte infiltrate was found (Kraus et al., 1996). Although the outcome is general favorable, a few cases were rapidly fatal. The majority improved with steroids, but in some patients symptoms developed while they were already on steroids. In these patients ciclosporin, plasma exchange, and more recently biologic anti-TNF agents (Ullrich et al., 2009) were used with variable success.

CEREBRALVENOUS THROMBOSIS Thrombosis of the dural sinus and cerebral veins is at least as frequent as arterial stroke in IBD. Thrombosis appears to be more common in ulcerative colitis than in Crohn’s disease, but in some series the opposite was found (Cognat et al., 2011). Cognat and coworkers (2011) recently published eight cases from two centers in Paris and reviewed 49 other cases of cerebral venous thrombosis associated with IBD confirmed by MR. Other cases not included in this review have been published (Milandre et al., 1992; Lossos et al., 1995;

600

J.M. FERRO ET AL.

Papi et al., 1995; Barclay et al., 2010; Nudelman et al., 2010; Casella et al., 2011; Kothur et al., 2012). The publications of Milandre et al. (1992), Nudelman et al. (2010), and Casella et al. (2011) also provide a review of previous reports. When compared to patients with cerebral venous thrombosis with other causes, patients with IBD-related cerebral venous thrombosis are younger and more often male. A more recent review of 65 cases published in English was performed by Katsanos and coworkers (2013). The clinical presentation, consisting of headaches, focal signs, seizures, or encephalopathy, and the sites of the venous occlusions are similar to the usual cerebral venous thromboses. They can occur from 2 months to 17 years after the first attack of IBD. Occasionally, the diagnosis of IBD is established only when cerebral venous thrombosis occurs (Cognat et al., 2011; Katsanos et al., 2013). Although IBD may be asymptomatic when the venous thrombosis occurs, almost all patients had biologic markers of inflammation such as elevated leukocyte count, CRP, or ESR. When concomitant causes of cerebral venous thrombosis are systematically searched for, the majority had other risk factors, such as oral contraceptives, severe iron deficiency anemia, anti-TNF-a treatment, thrombocytosis, hyperhomocystenemia, folate and B12 deficiencies, infection, lupus anticoagulant, and inherited thrombophilia (Milandre et al., 1992; Papi et al., 1995; Nudelman et al., 2010; Casella et al., 2011; Cognat et al., 2011). Despite the potential risk of intestinal bleeding, treatment of acute cerebral venous thrombosis with full dose intravenous heparin or low molecular weight heparin is effective and safe (Tsujikawa et al., 2000; Cognat et al., 2011; Katsanos et al., 2013). Endovascular thrombolysis was tried in a few cases, with favorable and safe outcomes (Philips et al., 1999; Kothur et al., 2012). This limited evidence supports the use of approved guidelines for cerebral venous thrombosis management (Einha¨upl et al., 2006; Saposnik et al., 2011) also when associated with IBD. The prognosis is usually good, but a few cases were fatal (Cognat et al., 2011; Katsanos et al., 2013).

DEMYELINATING DISEASE Multiple sclerosis (MS) has been frequently associated with IBD, particularly with UC (Rang et al., 1982; Sadovnick et al., 1989; Purrmann et al., 1992; Buccino et al., 1994; Kimura et al., 2000; Pandian et al., 2004; Bernstein et al., 2005; Gupta et al., 2005), although evolving diagnostic criteria for MS could mean that some patients actually have an MS-like disease instead of true MS. Both patients with IBD who later developed MS and patients with MS who developed IBD have been described. Kimura et al. (2000) found a 1% prevalence of

clinically definite MS among 400 patients with IBD. Gupta et al. (2005) estimated a relative risk for MS and optic neuritis in CD of 1.54 and of 1.74 in UC when compared with the general population. Between 40% and 50% of IBD presented asymptomatic white matter lesions in a study by Perkin and Murray-Lyon (1998). MS and IBD share similar epidemiology, age of presentation, clinical course, and geographic distribution. However, despite all evidence, the physiopathology of this relationship between diseases is still unknown, although some animal studies may shed some light on the issue. Extensive perivenular demyelination and astrocytosis has been found in monkeys suffering from IBD and cerebral venous thrombosis, possibly due to perivenular edema (Sheffield et al., 1981). Also, IBD may be viewed as a chronic variant of a predemyelinating state that can trigger demyelinating episodes. IBD has also been associated with other chronic inflammatory diseases suggesting a common immunologic etiology (Bernstein et al., 2005). Antitumor necrosis factor-a therapy is contraindicated in the treatment of patients with IBS and multiple sclerosis and there is evidence of onset of a demyelinating process with institution of this therapy (Thomas et al., 2004; Freeman and Flak, 2005).

OTHER CENTRAL NERVOUS SYSTEM COMPLICATIONS A slowly progressive spastic paraparesis due to myelopathy has been reported in association with IBD (Gibb et al., 1987; Ray et al., 1993; Suzuki et al., 1994; Lossos et al., 1995) and in the series by Lossos et al. was present in 26% of patients. An immune-mediated inflammatory origin has been suggested but vitamin B12 deficiency with resulting subacute combined degeneration of the spinal cord may also be responsible in patients with CD following terminal ileum resection. Epidural and subdural spinal empyemas secondary to fistulous extension from the rectum are rare complications of CD and can result in pain and progressive leg weakness (Sacher et al., 1989; Hershkowitz et al., 1990; Heidemann et al., 2003; Gelfenbeyn et al., 2006). In CD patients, Elsehety and Bertorini (1997) reported seizures (5.9%), headaches (4.3%), major depression (4.3%), anxiety disorder (2%), and other less frequent complications (less than 1%) such as Parkinson-like syndrome, cerebellar syndrome, ischemic optic neuritis, orbital pseudotumor, sixth nerve palsy, organic brain syndrome, and chronic fatigue syndrome. A single case of inflammatory pseudotumor of the cerebellum in a patient with CD was reported by Derrey et al. (2012). An association of epilepsy and IBD appears to be preferentially with CD (Elsehety and Bertorini, 1997) but

NEUROLOGIC MANIFESTATIONS OF INFLAMMATORY BOWEL DISEASES secondary seizures related to metabolic and structural causes have been described in UC (Schneidermann et al., 1979; Johns, 1991; Lossos et al., 1995). Diffuse encephalopathy may also develop in patients with CD, possibly related to vitamin deficiency including Wernicke encephalopathy, selenium toxicity due to parenteral nutrition, or precipitated by treatment with sulfasalazine (Schoonjans et al., 1993; Kawakubo et al., 1994; Hahn et al., 1998; Eggsp€ uhler et al., 2003).

PSYCHIATRIC SYNDROMES The most frequent psychiatric syndromes associated with IBD are depression and anxiety. In two Canadian national surveys with 3076 and 1438 respondents with IBD, 16.3% and 14.7%, respectively, reported depressive symptoms (Fuller-Thomson and Sulman, 2006). Depression was more frequent in women and younger respondents and in patients with pain or functional limitations.

MEDICATION-INDUCED NEUROLGIC COMPLICATIONS Steroids In the 1950s, Bunim et al. (1955) described glucocorticoidrelated myopathy, a well known side-effect of this therapy due to direct catabolic effects on the skeletal muscle. It can occur with any of the glucocorticoid preparations, in both initiation of treatment and chronic use. Patients typically present with painless proximal muscle wasting and weakness, normal muscle enzymes (Askari et al., 1976), and improve after decrease in drug dosage.

Metronidazole Metronidazole is an antimicrobial agent with bactericidal effects against anaerobic agents used in the treatment of IBD. Patients treated with metronidazole, particularly with high doses, have presented seizures, dizziness and vertigo, ataxia, confusion, irritability, insomnia, headache, tremors and peripheral neuropathy (Frytak et al., 1978; Kusumi et al., 1980; Halloran, 1982). Peripheral neuropathy is usually pure sensory or autonomic with occasional ataxia; it is mostly transient and resolves completely with discontinuation.

Ciclosporin Mild tremor, but also headache, visual abnormalities, and seizures in the context of a syndrome resembling posterior leukoencephalopathy have been described in patients treated with ciclosporin (Schwartz et al., 1995; Wijdicks et al., 1995; Hinchey et al., 1996).

601

Anti-TNF-a agents (infliximab, adalimumab, certolizumab) A possible link between TNF-a and demyelinating disease has been suggested although a causal relationship has not been established. In 2001, Mohan et al. described 19 patients with brain and spinal cord demyelination on magnetic resonance imaging, presenting with confusion, ataxia, dysesthesia, and paresthesia, who improved after discontinuation of therapy (Mohan et al., 2001). In postmarketing reports both optic neuritis and demyelinating polyneuropathy have also been reported with infliximab and adalimumab (Shin et al., 2006; Simsek et al., 2007; Eguren et al., 2009). Progressive multifocal leukoencephalopathy (PML), a severe demyelinating disease of the central nervous system caused by reactivation of the polyomavirus JC (JC virus) has been reported in patients treated with infliximab (Kumar et al., 2010) and glucocorticoids (Newton et al., 1986), alone or in combination with immune suppressive agents. The risk of PML is associated with duration of treatment, prior use of immune suppressant medication, and JC virus antibody status, being minimal in JC virus antibody-negative patients (Biogen Idec). PML typically presents with cognitive impairment and behavioral changes and can progress to cause motor weakness, visual and language disturbances, and seizures. Diagnosis can be confirmed by quantitative detection of JC virus DNA in the cerebrospinal fluid (Aksamit, 2008).

Anti-a4 integrin (natalizumab) Natalizumab is a humanized monoclonal antibody to a4 integrin recently approved for treatment of CD (Sandborn et al., 2005) outside Europe. PML has been described in association with natalizumab in CD patients (Van Assche et al., 2005).

REFERENCES Akbayir N, Calis AB, Alkim C et al. (2005). Sensorineural hearing loss in patients with inflammatory bowel: a subclinical extraintestinal manifestation. Dig Dis Sci 50: 1938–1945. Aksamit AJ (2008). Progressive multifocal leukoencephalopathy. Curr Treat Options Neurol 10: 178–185. Askari A, Vignos PJ Jr, Moskowitz RW (1976). Steroid myopathy in connective tissue disease. Am J Med 61: 485–492. Bachmeyer C, Leclerc-Landgraf N, Laurette F et al. (1998). Acute autoimmune sensorineural hearing loss associated with Crohn’s disease. Am J Gastroenterol 93: 2565–2567. Barclay AR, Keightley JM, Horrocks I et al. (2010). Cerebral thromboembolic events in pediatric patients with inflammatory bowel disease. Inflamm Bowel Dis 16: 677–683.

602

J.M. FERRO ET AL.

Baron BW, Jeon HR, Glunz C et al. (2002). First two patients with ulcerative colitis who developed classic thrombotic thrombocytopenic purpura successfully treated with medical therapy and plasma exchange. J Clin Apher 17: 204–206. Benavente L, Morı´s G (2011). Neurologic disorders associated with inflammatory bowel disease. Eur J Neurol 18: 138–143. Bermejo PE, Burgos A (2008). Neurological complications of inflammatory bowel disease. Med Clin (Barc) 130: 666–675. Bernstein CN, Blanchard JF, Houston DS et al. (2001). The incidence of deep venous thrombosis and pulmonary embolism among patients with inflammatory bowel disease: a population-based cohort study. Thromb Haemost 85: 430–434. Bernstein CN, Wajda A, Blanchard JF (2005). The clustering of other chronic inflammatory diseases in inflammatory bowel disease: a population-based study. Gastroenterology 129: 827–836. Bhigjee AI, Bill PL, Cosnett JE (1987). Ulcerative colitis and interstitial myositis. Clin Neurol Neurosurg 89: 261–263. Biogen Idec Medical Information website. https://medinfo. biogenidec.com/medinfo/registration/reguser.do. Bouma G, Oudkerk Pool M, Crusius JB et al. (1997). Evidence for genetic heterogeneity in inflammatory bowel disease (IBD); HLA genes in the predisposition to suffer from ulcerative colitis (UC) and Crohn’s disease (CD). Clin Exp Immunol 109: 175–179. Buccino GP, Corrente G, Visintini D (1994). Crohn’s disease and multiple sclerosis: a single case report. Ital J Neurol Sci 15: 303–306. Bunim JJ, Ziff M, McEwen C (1955). Evaluation of prolonged cortisone therapy in rheumatoid arthritis: a four-year study. Am J Med 18: 27–40. Carmona MA, Jaume Anselmi F, Ramı´rez Rivera J (2000). Cerebral thrombosis and vasculitis: an uncommon complication of ulcerative colitis. Bol Asoc Med P R 92: 9–11. Casella G, Spreafico C, Costantini M et al. (2011). Cerebral sinus thrombosis in ulcerative colitis. Inflamm Bowel Dis 17: 2214–2216. Chebli JM, Gaburri PD, de Souza AF et al. (2000). Fatal evolution of systemic lupus erythematosus associated with Crohn’s disease. Arq Gastroenterol 37: 224–226. Cognat E, Crassard I, Denier C et al. (2011). Cerebral venous thrombosis in inflammatory bowel diseases: eight cases and literature review. Int J Stroke 6: 487–492. Cohen M, Baldin B, Thomas P et al. (2012). Neurological adverse events under anti-TNF alpha therapy. Rev Neurol (Paris) 168: 33–39. Cosnes J, Blain A, Cattan et al. (2002). Long-term evolution of disease behavior of Crohn’s disease. Inflamm Bowel Dis 8: 244–250. Derbyshire SWG (2003). A systematic review of neuroimaging data during visceral stimulation. Am J Gastroenterol 98: 12–20. Derrey S, Charpentier C, Ge´rardin E et al. (2012). Inflammatory pseudotumor of the cerebellum in a patient with Crohn’s disease. World Neurosurg 77: 201.e13–201.e16. Eggsp€ uhler AW, Bauerfeind P, Dorn T et al. (2003). Wernicke encephalopathy – a severe neurological complication in a clinically inactive Crohn’s disease. Eur Neurol 50: 184–185.

Eguren C, Dı´az Ley B, Daude´n E et al. (2009). Peripheral neuropathy in two patients with psoriasis in treatment with infliximab. Muscle Nerve 40: 488–489. Einha¨upl K, Bousser MG, de Bruijn SF et al. (2006). EFNS guideline on the treatment of cerebral venous and sinus thrombosis. Eur J Neurol 13: 553–559. Elsehety A, Bertorini TE (1997). Neurologic and neuropsychiatric complications of Crohn’s disease. South Med J 90: 606–610. Finnie IA, Shields R, Sutton R et al. (1994). Crohn’s disease and myasthenia gravis: a possible role for thymectomy. Gut 35: 278–279. Freeman HJ, Flak B (2005). Demyelination-like syndrome in Crohn’s disease after infliximab therapy. Can J Gastroenterol 19: 313–316. Frytak S, Moertel CH, Childs DS (1978). Neurologic toxicity associated with high-dose metronidazole therapy. Ann Intern Med 88: 361–362. Fuller-Thomson E, Sulman J (2006). Depression and inflammatory bowel disease: findings from two nationally representative Canadian surveys. Inflamm Bowel Dis 12: 697–707. Geissler A, Andus T, Roth M et al. (1995). Focal white-matter lesions in brain of patients with inflammatory bowel disease. Lancet 345: 897–898. Gelfenbeyn M, Goodkin R, Kliot M (2006). Sterile recurrent spinal epidural abscess in a patient with Crohn’s disease: a case report. Surg Neurol 65: 178–184. Gendelman S, Present D, Janowitz HD (1982). Neurological complications of inflammatory bowel disease. Gastroenterology 82: 1065. Gibb WRG, Dhillon DP, Zilkha KJ et al. (1987). Bronchiectasis with ulcerative colitis and myelopathy. Thorax 42: 155–156. Glotzer DJ, Yuan RH, Patterson JF (1964). Ulcerative colitis complicated by toxic megacolon, polyserositis and hemorrhagic leukoencephalitis with recovery. Ann Surg 159: 445–450. Gobron C, Kaci R, Sokol H et al. (2010). Unilateral carotid granulomatous arteritis and Crohn’s disease. Rev Neurol (Paris) 166: 542–546. Gondim FA, Brannagan TH, Sander HW et al. (2005). Peripheral neuropathy in patients with inflammatory bowel disease. Brain 128: 867–879. Gupta G, Gelfand JM, Lewis JD (2005). Increased risk for demyelinating diseases in patients with inflammatory bowel disease. Gastroenterology 129: 819–826. Ha C, Magowan S, Accortt NA et al. (2009). Risk of arterial thrombotic events in inflammatory bowel disease. Am J Gastroenterol 104: 1445–1451. Hahn JS, Berquist W, Alcorn DM et al. (1998). Wernicke encephalopathy and beriberi during total parenteral nutrition attributable to multivitamin infusion shortage. Pediatrics 101: E10. Halloran TJ (1982). Convulsions associated with high cumulative doses of metronidazole. Drug Intell Clin Pharm 16: 409. Hayashi K, Kurisu Y, Ohshiba J et al. (1991). Report of a case of Crohn’s disease associated with hyper-creatine phosphokinasemia. Jpn J Med 30: 441–445.

NEUROLOGIC MANIFESTATIONS OF INFLAMMATORY BOWEL DISEASES Heidemann J, Spinelli KS, Otterson MF et al. (2003). Case report: magnetic resonance imaging in the diagnosis of epidural abscess perirectal fistulizing Crohn’s disease. Inflamm Bowel Dis 9: 122–124. Hershkowitz S, Link R, Ravden M et al. (1990). Spinal empyema in Crohn’s disease. J Clin Gastroenterol 12: 67–69. Hinchey J, Chaves C, Appignani B et al. (1996). A reversible posterior leukoencephalopathy syndrome. N Eng J Med 334: 494–500. Hisada T, Miyamae Y, Mizuide M et al. (2006). Acute thrombocytopenia associated with preexisting ulcerative colitis successfully treated with colectomy. Intern Med 45: 87–91. Holzer K, Esposito L, Stimmer H et al. (2009). Cerebral vasculitis mimicking migraine with aura in a patient with Crohn’s disease. Acta Neurol Belg 109: 44–48. Jacob A, Ledingham JG, Kerr AI et al. (1990). Ulcerative colitis and giant cell arteritis associated with sensorineural deafness. J Laryngol Otol 104: 889–890. Johns DR (1991). Cerebrovascular complications of inflammatory bowel disease. Am J Gastroenterol 86: 367–370. Kappelman MD, Horvath-Puho E, Sandler RS et al. (2011). Thromboembolic risk among Danish children and adults with inflammatory bowel diseases: a population-based nationwide study. Gut 60: 937–943. Katsanos AH, Katsanos KH, Kosmidou M et al. (2013). Cerebral sinus venous thrombosis in inflammatory bowel diseases. QJM 106: 401–413. Kawakubo K, Iida M, Matsumoto T et al. (1994). Progressive encephalopathy in a Crohn’s disease patient on long-term total parenteral nutrition: possible relationship to selenium deficiency. Postgrad Med J 70: 215–219. Kimura K, Hunter SF, Thollander MS et al. (2000). Concurrence of inflammatory bowel disease and multiple sclerosis. Mayo Clin Proc 75: 802–806. Konturek SJ, Konturek JW, Pawlik T et al. (2004). Brain-gut axis and its role in the control of food intake. J Physiol Pharmacol 55: 137–154. Kothur K, Kaul S, Rammurthi S et al. (2012). Use of thrombolytic therapy in cerebral venous sinus thrombosis with ulcerative colitis. Ann Indian Acad Neurol 15: 35–38. Kraus JA, Nahser HC, Berlit P (1996). Lymphocytic encephalomyeloneuritis as a neurologic complication of ulcerative colitis. J Neurol Sci 141: 117–119. Kreuzpaintner G, Horstkotte D, Heyll A et al. (1992). Increased risk of bacterial endocarditis in inflammatory bowel disease. Am J Med 92: 391–395. Kugathasan S, Collins N, Maresso K et al. (2004). CARD15 gene mutations and risk for early surgery in pediatric-onset Crohn’s disease. Clin Gastroenterol Hepatol 2: 1003–1009. Kumar BN, Smith MSH, Walsh RM et al. (2000). Sensorineural hearing loss in ulcerative colitis. Clin Otolaryngol 25: 143–145. Kumar D, Bouldin TW, Berger RG (2010). A case of progressive multifocal leukoencephalopathy in a patient treated with infliximab. Arthritis Rheum 62: 3191–3195. Kusumi RK, Plouffe JF, Wyatt RH et al. (1980). Central nervous system toxicity associated with metronidazole therapy. Ann Intern Med 93: 59–60.

603

Lahari D, Debeugny S, Peeters M et al. (2001). Inflammatory bowel disease in spouses and their offspring. Gastroenterology 120: 816–819. Lennard-Jones JE (1989). Classification of inflammatory bowel disease. Scand J Gastroenterol 170 (Suppl): 2–6. Lloyd DA, Payton KB, Guenther L et al. (1994). Melkersson– Rosenthal syndrome and Crohn’s disease: one disease or two? Report of a case and discussion of the literature. J Clin Gastroenterol 18: 213–217. Lossos A, River Y, Eliakim A et al. (1995). Neurologic aspects of inflammatory bowel disease. Neurology 45: 416–421. Mandell DM, Matouk CC, Farb RI et al. (2012). Vessel wall MRI to differentiate between reversible cerebral vasoconstriction syndrome and central nervous system vasculitis: preliminary results. Stroke 43: 860–862. Martı´n de Carpi J, Ribo´ Cruz JM, Anto´n Lo´pez J et al. (2007). Cerebral vasculitis associated with ulcerative colitis. An Pediatr (Barc) 67: 177–178. Martin RW, Shah A (1991). Myasthenia gravis coexistent with Crohn’s disease. J Clin Gastroenterol 13: 112–113. Miehsler W, Reinisch W, Valic E et al. (2004). Is inflammatory bowel disease an independent and disease specific risk factor for thromboembolism? Gut 53: 542–548. Milandre L, Monges D, Dor V et al. (1992). Cerebral phlebitis and Crohn disease. Rev Neurol (Paris) 148: 139–144. Mohan N, Edwards ET, Cupps TR et al. (2001). Demyelination occurring during anti-tumor necrosis factor alpha therapy for inflammatory arthritides. Arthritis Rheum 44: 2862–2869. Molodecky NA, Soon IS, Rabi DM et al. (2012). Increasing incidence and prevalence of the inflammatory bowel diseases with time, based on systematic review. Gastroenterology 142: 46–54. Nelson J, Barron MM, Riggs JE et al. (1986). Cerebral vasculitis and ulcerative colitis. Neurology 36: 719–721. Newton P, Aldridge RD, Lesseells AM et al. (1986). Progressive multifocal leukoencephalopathy complicating systemic lupus erythematous. Arthritis Rheum 29: 337–343. Novacek G, Weltermann A, Sobala A et al. (2010). Inflammatory bowel disease is a risk factor for recurrent venous thromboembolism. Gastroenterology 139: 779–787. Nudelman RJ, Rosen DG, Rouah E et al. (2010). Cerebral sinus thrombosis: a fatal neurological complication of ulcerative colitis. Pathol Res Int 2010: 132754. Ogawa E, Sakakibara R, Yoshimatsu Y et al. (2011). Crohn’s disease and stroke in a young adult. Intern Med 50: 2407–2408. Oliveira GR, Teles BC, Brasil EF et al. (2008). Peripheral neuropathy and neurological disorders in an unselected Brazilian population-based cohort of IBD patients. Inflamm Bowel Dis 14: 389–395. Orholm M, Binder V, Srensen TI et al. (2000). Concordance of inflammatory bowel disease among Danish twins. Results of a nationwide study. Scand J Gastroenterol 35: 1075–1081. Pandian JD, Pawar G, Singh GS et al. (2004). Multiple sclerosis in a patient with chronic ulcerative colitis. Neurol India 52: 282–283.

604

J.M. FERRO ET AL.

Papa A, Danese S, Grillo A et al. (2003). Review article: inherited thrombophilia in inflammatory bowel disease. Am J Gastroenterol 98: 1247–1251. Papi C, Ciaco A, Acierno G et al. (1995). Severe ulcerative colitis, dural sinus thrombosis, and the lupus anticoagulant. Am J Gastroenterol 90: 1514–1517. Perkin GD, Murray-Lyon I (1998). Neurology and the gastrointestinal system. J Neurol Neurosurg Psychiatry 65: 291–300. Peyrin-Biroulet L, Loftus EV Jr, Colombel JF et al. (2011). Long-term complications, extraintestinal manifestations, and mortality in adult Crohn’s disease in population-based cohorts. Inflamm Bowel Dis 17: 471–478. Philips MF, Bagley LJ, Sinson GP et al. (1999). Endovascular thrombolysis for symptomatic cerebral venous thrombosis. J Neurosurg 90: 65–71. Porter CK, Tribble DR, Aliaga PA et al. (2008). Infectious gastroenteritis and risk of developing inflammatory bowel disease. Gastroenterology 135: 781–786. Purrmann J, Arendt G, Cleveland S et al. (1992). Association of Crohn’s disease and multiple sclerosis. Is there a common background? J Clin Gastroenterol 14: 43–46. Rang EH, Brooke BN, Hermon-Taylor J (1982). Association of ulcerative colitis with multiple sclerosis. Lancet 2: 555. Ray DW, Bridger J, Hawnaur J et al. (1993). Transverse myelitis as the presentation of Jo-1 antibody syndrome (myositis and fibrosing alveolitis) in long-standing ulcerative colitis. Br J Rheumatol 32: 1105–1108. Ronchetto F, Pistono PG (1993). Temporal arteritis in a patient with ulcerative colitis. Coincidental association or (immuno) pathogenetic link? Recenti Prog Med 84: 54–56. Sacher M, G€opfrich H, Hochberger O (1989). Crohn’s disease penetrating into the spinal canal. Acta Paediatr Scand 78: 647–649. Sadovnick AD, Paty DW, Yannakoulias G (1989). Concurrence of multiple sclerosis and inflammatory bowel disease. N Eng J Med 321: 762–763. Sandborn WJ, Colombel JF, Enns R et al. (2005). Natalizumab induction and maintenance therapy for Crohn’s disease. N Eng J Med 353: 1912–1925. Santos S, Casadevall T, Pascual LF et al. (2001). Neurological alterations related to Crohn’s disease. Rev Neurol 32: 1158–1162. Saposnik G, Barinagarrementeria F, Brown RD Jr et al. (2011). Diagnosis and management of cerebral venous thrombosis: a statement for healthcare professionals from the American Heart Association/American Stroke Association. Stroke 42: 1158–1192. Sassi SB, Kallel L, Ben Romdhane S et al. (2009). Peripheral neuropathy in inflammatory bowel disease patients: a prospective cohort study. Scand J Gastroenterol 44: 1268–1269. Satsangi J, Jewell DP, Bell JL (1997). The genetics of inflammatory bowel disease. Gut 40: 572. Scheid R, Teich N (2007). Neurologic manifestations of ulcerative colitis. Eur J Neurol 14: 483–493. Schluter A, Krasnianski M, Krivokuca M et al. (2004). Magnetic resonance angiography in a patient with

Crohn’s disease associated cerebral vasculitis. Clin Neurol Neurosurg 106: 110–113. Schneidermann JH, Sharpe JA, Sutton DM (1979). Cerebral and retinal vascular complications of inflammatory bowel disease. Ann Neurol 5: 331–337. Schoonjans R, Mast A, Van Den Abeele G et al. (1993). Sulfasalazine-associated encephalopathy in a patient with Crohn’s disease. Am J Gastroenterol 88: 1416–1420. Schwartz RB, Bravo SM, Klufas RA et al. (1995). Cyclosporine neurotoxicity and its relationship to hypertensive encephalopathy: CT and MR findings in 16 cases. AJR Am J Roentgenol 165: 627–631. Schwartz DA, Loftus EV Jr, Tremaine WJ et al. (2002). The natural history of fistulizing Crohn’s disease in Olmested County, Minnesota. Gastroenterology 122: 875–880. Sedwick LA, Klingele TG, Burde RM et al. (1984). Optic neuritis in inflammatory bowel disease. J Clin Neuroophthalmol 4: 3–6. Sheffield WD, Squire RA, Strandberg JD (1981). Cerebral venous thrombosis in the rhesus monkey. Vet Pathol 18: 326–334. Shimoyama T, Tamura Y, Sakamoto T et al. (2009). Immunemediated myositis in Crohn’s disease. Muscle Nerve 39: 101–105. Shin IS, Baer AN, Kwon HJ et al. (2006). Guillain–Barre´ and Miller Fisher syndromes occurring with tumor necrosis factor alpha antagonist therapy. Arthritis Rheum 54: 1429–1434. Silverberg MS, Satsangi J, Ahmad T et al. (2005). Toward an integrated clinical, molecular, and serological classification of inflammatory bowel disease: report of a working party of the 2005 Montreal World Congress of Gastrenterology. Can J Gastroenterol 19 (Suppl A): 5–36. Simsek I, Erdem H, Pay S et al. (2007). Optic neuritis occurring with anti-tumour necrosis factor alpha therapy. Ann Rheum Dis 66: 1255–1258. Sowa JM (1991). Overlapping polymyositis and ulcerative colitis: HTLV-1 infection as an alternative explanation. J Rheumatol 18: 1939. Suzuki I, Watanabe N, Suzuki J et al. (1994). A case of bronchiectasis accompanied by ulcerative colitis and HTLV-1 associated myelopathy (HAM). Jpn J Thorac Dis 32: 358–363. Talbot RW, Heppell J, Dozois RR et al. (1986). Vascular complications of inflammatory bowel disease. Mayo Clin Proc 61: 140–145. Thomas CW Jr, Weinshenker BG, Sandborn WJ (2004). Demyelination during anti-tumor necrosis factor therapy with infliximab for Crohn’s disease. Inflamm Bowel Dis 10: 28–31. Tsujikawa T, Urabe M, Bamba H et al. (2000). Haemorrhagic cerebral sinus thrombosis associated with ulcerative colitis: a case report of successful treatment by anticoagulant therapy. J Gastroenterol Hepatol 15: 688–692. Ullrich S, Schinke S, Both M et al. (2009). Refractory central nervous system vasculitis and gastrocnemius myalgia syndrome in Crohn’s disease successfully treated with anti-tumor necrosis factor-alpha antibody. Semin Arthritis Rheum 38: 337–347.

NEUROLOGIC MANIFESTATIONS OF INFLAMMATORY BOWEL DISEASES Van Assche G, Van Ranst M, Sciot R et al. (2005). Progressive multifocal leukoencephalopathy after natalizumab therapy for Crohn’s disease. N Eng J Med 353: 362–368. Vannucchi V, Grazzini M, Pieralli F et al. (2011). Adalimumab-induced lupus erythematosus with central nervous system involvement in a patient with Crohn’s disease. J Gastrointestin Liver Dis 20: 201–203. Vavricka SR, Brun L, Ballabini P et al. (2011). Frequency and risk factors of extraintestinal manifestations in the Swiss

605

inflammatory bowel disease cohort. Am J Gastroenterol 106: 110–119. Wijdicks EF, Wiesner RH, Krom RA (1995). Neurotoxicity in liver transplant recipients with cyclosporine immunosuppression. Neurology 45: 1962–1964. Zois CD, Katsanos K, Kosmidou M et al. (2010). Neurological manifestation of inflammatory bowel diseases: current knowledge and novel insights. J Crohns Colitis 4: 115–124.

Handbook of Clinical Neurology, Vol. 120 (3rd series) Neurologic Aspects of Systemic Disease Part II Jose Biller and Jose M. Ferro, Editors © 2014 Elsevier B.V. All rights reserved

Chapter 41

Gluten-related neurologic dysfunction MARIOS HADJIVASSILIOU1*, ANDREW P. DUKER2, AND DAVID S. SANDERS3 1 Department of Neurology, Royal Hallamshire Hospital, Sheffield, UK 2 3

Department of Neurology, University of Cincinnati, Cincinnati, OH, USA

Department of Gastroenterology, Royal Hallamshire Hospital, Sheffield, UK

HISTORICAL PERSPECTIVE Celiac disease (CD) was first described by the Greek doctor Aretaeus the Cappadocian, in AD 100, only to be forgotten and then rediscovered by Samuel Gee in 1888 (Gee, 1888). In a lecture on “the coeliac affection,” Gee described the classic pediatric presentation of the disease. Whilst clinicians began to recognize this disease entity, the etiologic agent remained obscure until the observations of Willem Dicke, a Dutch pediatrician, in 1953 of “the presence in wheat, of a factor having a deleterious effect in cases of celiac disease” (Dicke et al., 1953). As gastrointestinal symptoms (diarrhea, abdominal pain, bloating, weight loss) were dominant in patients with this disease, it was not surprising that CD was thought to be a disease of the gut. Indeed the introduction of endoscopy and small bowel biopsy in the 1950s confirmed the presence of an enteropathy (Paulley, 1954). In 1963 a group of dermatologists made the interesting observation that dermatitis herpetiformis (DH), an itchy vesicular rash, was a form of gluten-related dermatopathy sharing the same small bowel pathology, but not the gastrointestinal symptoms seen in patients with CD (Marks et al., 1966). This was the first evidence of extraintestinal manifestations. Only a small number of case reports of patients with CD and neurologic manifestations (Elders, 1925; Reed and Ash, 1927; Woltman and Heck, 1937) were published prior to the discovery of the etiologic agent and the introduction of small bowel biopsy, demonstrating the typical histologic features that define CD. Such reports need to be treated with caution given that a diagnosis of CD in those patients was speculative. The first comprehensive case series of neurologic manifestations in the context of histologically confirmed

CD was published in 1966 (Cooke and Thomas-Smith, 1966). This detailed work described the range of neurologic manifestations seen in 16 patients with established CD. Of interest was the fact that all patients had gait ataxia and some had severe peripheral neuropathy as well. The assumption was that such manifestations were nutritional as a result of malabsorption. Indeed all of these patients were grossly malnourished and cachectic. Postmortem data from the same report, however, demonstrated an inflammatory process primarily affecting the cerebellum, but also involving other parts of the central and peripheral nervous systems, a finding that was in favor of an immune-mediated pathogenesis. Single and multiple case reports of patients with established CD who then developed neurologic dysfunction continued to be published (Binder et al., 1967; Bundey, 1967; Morris et al., 1970; Coers et al., 1971; Kepes et al., 1975; Finelli et al., 1980; Kinney et al., 1982; Ward et al., 1985; Lu et al., 1986; Kristoferitsch and Pointer, 1987; Kaplan et al., 1988; Tison et al., 1989; Collin et al., 1991; Hermaszewski et al., 1991; Bhatia et al., 1995; Dick et al., 1995; Muller et al., 1996). The key findings from these reports were as follows: ● ●



Ataxia (with and without myoclonus) and neuropathy were the commonest manifestations. Neurologic manifestations were usually reported in the context of established CD and almost always attributed to nutritional deficiencies. In those reports where the effect of the dietary restriction was reported, the results were mixed. None of these reports, however, documented any attempts to monitor adherence to the diet with repeat serologic testing.

*Correspondence to: Marios Hadjivassiliou, Department of Neurology, Royal Hallamshire Hospital, Glossop Road, Sheffield, S10 2JF, UK. E-mail: [email protected]

608

M. HADJIVASSILIOU ET AL.

Thirty years after the first comprehensive case series on neurologic manifestations of CD saw the publication of an original study (Hadjivassiliou et al., 1996) approaching the issue from a neurologic perspective by investigating the prevalence of serologic markers of gluten-related dysfunction (GRD) in patients presenting with neurologic dysfunction of unknown etiology. The results demonstrated that there was a high prevalence of IgG and/or IgA antigliadin antibodies (AGA) in this group of patients compared to controls. Based on duodenal biopsies the same study showed that the prevalence of CD in this group of patients with neurologic dysfunction was 16 times higher than the prevalence of CD in the healthy population. This study rekindled the interest of neurologists in a possible link between GRD and neurologic disease.

EPIDEMIOLOGY OF NEUROLOGIC MANIFESTATIONS The prevalence of CD in the healthy population has been shown to be at least 1% in both European and North American studies (Sanders et al., 2003). There are no accurate figures of the prevalence of the neurologic manifestations of gluten sensitivity in the general population. Figures of between 10% and 22.5% have been reported amongst patients with established CD attending gastrointestinal clinics (Holmes, 1997; Briani et al., 2008). These are unlikely to be accurate because such figures are usually retrospective, derived solely from gastrointestinal clinics, concentrating exclusively on patients with classic CD presentation, and often include neurologic dysfunctions that are unlikely to be gluten related (e.g., carpal tunnel syndrome, idiopathic Parkinson’s disease, etc.). Some estimates of prevalence can be made from patient populations attending specialist clinics although caution must be exercised in extrapolating these as they are inevitably affected by referral bias. Data collected from the Sheffield dedicated CD and gluten sensitivity/neurology clinics suggest that for every seven patients presenting to the gastroenterologists who are then diagnosed with CD, there are two patients presenting to the neurologists who will then be diagnosed as having CD (Hadjivassiliou et al., 2010a). This is likely to be an underestimate because this ratio does not take into account those patients with neurologic manifestations due to GRD that do not have an enteropathy (approximately two-thirds of the whole number of patients presenting with neurologic dysfunction). The authors believe that the prevalence of neurologic dysfunction even within patients with CD presenting to gastroenterologists is likely to be much higher than what has been published if such patients undergo rigorous neurologic workup including magnetic resonance (MR) spectroscopy of the cerebellum. Preliminary work on patients with CD presenting to gastroenterologists with minor

neurologic complaints demonstrates that up to 80% have abnormal MR spectroscopy (low NAA/Cr ratios) of the cerebellum (Hadjivassiliou et al., 2011).

THE DIAGNOSIS OF GLUTEN-RELATED DISEASES CD is characterized by the presence of an enteropathy, a reliable gold standard. It is now accepted, however, that an enteropathy is not a prerequisite for the diagnosis of GRD with predominantly neurologic or other extraintestinal manifestations. Furthermore, the small bowel mucosal changes in the context of GRD represent a spectrum, from histologically normal mucosa to full-blown enteropathy to a pre-lymphomatous state also referred to as the Marsh classification (Marsh, 1992). Most pathology departments have now adopted the Marsh classification when reporting the histologic findings of small bowel biopsies. Given that the histology can be normal, a definition of GRD based solely on histology becomes problematic. Furthermore the diagnosis currently has to rely on serologic tests that are not 100% specific or sensitive. For example, endomysial antibody (EMA) and antitransglutaminase-2 (TG2) IgA antibody detection are specific for the presence of an enteropathy. However, these markers are frequently not detectable in patients with neurologic manifestations, particularly in those who do not have an enteropathy (Table 41.1). Table 41.1 Type of neurologic presentation in gluten sensitivity* Neurologic presentation

No

Total number of patients Ataxia (4 patients with myoclonus, 2 with palatal tremor) Peripheral neuropathy Sensorimotor axonal neuropathy Mononeuropathy multiplex Sensory neuronopathy Small fiber neuropathy Motor neuropathy Encephalopathy Myopathy Myelopathy Stiff man syndrome Chorea (often with ataxia) Neuromyotonia Epilepsy and occipital calcifications

500 233 (93) 182 (48) 135 19 14 8 8 77 (45) 18 (10) 9 (4) 7 (2) 3 (2) 1 (1) 1 (0)

*Based on 500 patients with gluten sensitivity, presenting with neurologic dysfunction and seen in the gluten sensitivity/neurology clinic, Sheffield, UK, from 1994 to 2011. The number of patients from each group who had enteropathy on biopsy is shown in parentheses. Some patients had more than one type of neurologic presentation.

GLUTEN-RELATED NEUROLOGIC DYSFUNCTION 609 GRD cannot be diagnosed on clinical grounds alone. had serologic evidence of GRD. Therefore gluten ataxia The majority of patients presenting with neurologic had a prevalence of 22% amongst sporadic ataxias but as manifestations have no gastrointestinal symptoms. high as 45% amongst idiopathic sporadic ataxias. Using Patients with CD can also have no gastrointestinal sympthe same AGA assay the prevalence of positive AGA in toms. In patients without overt gastrointestinal involvegenetically confirmed ataxias was 8/82 (10%), in familial ment, serum antibodies to TG2 may be absent. Such ataxias (not genetically confirmed) 8/49 (16%), and in patients typically have antibodies primarily reacting with healthy volunteers 149/1200 (12%). A number of studies different TG isozymes, TG3 in DH and TG6 in patients looking at the prevalence of antigliadin antibodies in with gluten ataxia (Hadjivassiliou et al., 2008a). Reacataxias have been published: The original publication tion of such antibodies with TG2 in the intestinal mucosa by the authors (Hadjivassiliou et al., 2003a) reported occurs prior to overt changes in small intestinal morpholthe incidence of IgG and/or IgA AGA in a large cohort ogy and sometimes even before the antibodies are of 224 patients seen in Sheffield, UK, with idiopathic and detectable in serum (Korponay-Szabo´ et al., 2003). Such hereditary ataxia. Antibodies were present in 41% of antibody deposits seem to be present in patients with patients with sporadic ataxia (54/132), compared to neurologic manifestations as well, and may therefore 12% (149/1200) of normal controls from the same popube diagnostically useful (Hadjivassiliou et al., 2006a). lation. Positive antibodies were also found in 14% of However, this test is not readily available and requires patients with familial ataxia (8/59) and 15% of patients experience in its interpretation. In practice, for suswith clinically probable multiple system atrophy pected neurologic manifestations of GRD, it is best to (MSA)-C (5/33). Among AGA-positive individuals with perform serologic tests for both IgA and IgG antibodies sporadic ataxia, evidence of celiac disease was present to TG2 (and if available anti-TG6 and anti-TG3) as well in 24% (12/51) of those patients who underwent duodenal as IgG and IgA antibodies to gliadin. Endomysium antibiopsy. In this same study, a separate group of patients bodies are very specific for the detection of enteropathy, from a second center in London with sporadic ataxia but they detect the same antigen (transglutaminase 2) showed positive antibodies in 32% (14/44). Another study and have thus largely been replaced by TG2 antibody (B€ urk et al., 2001a) found positive antibodies in 11.5% testing. Any differences between the two tests, however, (12/104) of patients with idiopathic ataxia and negative are likely to be related to the different methodologies genetic testing, compared to 5% of 600 blood donors used (ELISA for TG2 versus immunofluorescence for from the same country. An Italian study (Pellecchia the detection of EMA). et al., 1999a) confirmed a higher incidence of positive GRD have a strong genetic predisposition whereby antigliadin antibodies and celiac disease on intestinal 40% of the genetic load comes from MHC class II assobiopsy in idiopathic ataxia (3/24, 12.5%) compared to ciation (Hunt, 2008). In Caucasian populations more than patients with known hereditary ataxia (0/23, 0%). Further 90% of CD patients carry the HLA DQ2, with the remaining confirmatory studies with smaller numbers from having the HLA DQ8. A small number of CD patients do Finland, Japan, and France have also been published not belong into either of these groups but these have been (Luostarinen et al., 2001; Anheim et al., 2006; Ihara shown to carry just one chain of the DQ2 heterodimer. HLA et al., 2006). However, other studies have not shown genetic testing is therefore another useful diagnostic tool, as clear a distinction in antibody prevalence between idiparticularly as, unlike other serologic tests, it is not depenopathic and other causes of ataxia. Abele et al. (2002) dent on an immunologic trigger. However, the HLA DQ found positive antibodies in 15% (10/65) of patients with genotype can be used only as a test of exclusion as the risk idiopathic ataxia compared to 9% (3/32) of patients with genotype DQ2 is common in Caucasian and Asian populaMSA and 7% (1/15) with genetic ataxia. In a separate tions and many carriers will never develop GRD. study, Abele et al. (2003) found no statistically significant difference between the prevalence of positive IgG THE SPECTRUM OF GLUTEN-RELATED and/or IgA AGA in sporadic ataxia (19% of 32 patients), NEUROLOGIC MANIFESTATIONS recessive ataxia (8% of 24 patients), dominant ataxia (15% of 39 patients), and controls (8% of 73 patients). Gluten ataxia Analyzing IgG and IgA subtypes separately also did Gluten ataxia (GA) was originally defined as otherwise not show significant differences between groups. idiopathic sporadic ataxia with positive AGA Bushara et al. (2001) noted similar rates of positive (Hadjivassiliou et al., 2003a). This original definition IgG and/or IgA AGA in sporadic ataxia (27%, 7/26) was based on the serologic tests available at the time. and autosomal dominant ataxia (37%, 9/24). Some In a series of 853 patients with progressive ataxia evaluauthors believe the elevated incidence of AGA in heredated over a period of 15 years in Sheffield, UK, there itary ataxia may be driven in part by spinocerebellar were 152 patients out of 681 with sporadic ataxia who ataxia type 2 (SCA2). In one study, 23% of SCA2

610

M. HADJIVASSILIOU ET AL.

patients had positive AGA, significantly higher compared to 9% of controls (Almaguer-Mederos et al., 2008). The variations in prevalence may relate to geographical differences in the prevalence of CD, referral bias, variability in the AGA assays used, patient selection (some studies included as idiopathic sporadic ataxia patients with cerebellar variant of multisystem atrophy (Combarros et al., 2000)), the small number of patients studied, and no controls. The common theme in most of these studies is the consistently high prevalence of AGA antibodies in sporadic ataxias when compared to healthy controls. GA usually presents with pure cerebellar ataxia or rarely ataxia in combination with myoclonus, palatal tremor (Hadjivassiliou et al., 2008b), opsoclonus (Deconinck et al., 2006), or rarely, chorea (Pereira et al., 2004). GA is usually of insidious onset with a mean age at onset of 53 years. Rarely the ataxia can be rapidly progressive mimicking paraneoplastic cerebellar degeneration. Gaze-evoked nystagmus and other ocular signs of cerebellar dysfunction are seen in up to 80% of cases. All patients have gait ataxia and the majority have limb ataxia. Less than 10% of patients with GA will have any gastrointestinal symptoms but a third will have evidence of enteropathy on biopsy. Up to 60% of patients have neurophysiologic evidence of sensorimotor, lengthdependent axonal neuropathy. This is usually mild and does not contribute to the ataxia. Anti-TG2 IgA antibodies are present in up to 38% of patients, but often at lower titers than those seen in patients with CD. However, unlike CD, IgG class antibodies to TG2 are more frequent than IgA. Antibodies against TG2 and TG6 combined can be found in 85% of patients with ataxia who are positive for AGA antibodies (Hadjivassiliou et al., 2009). Some patients also test positive for anti-TG3 antibodies although the prevalence of such antibodies in patients with gluten ataxia is low when compared to DH. It is unclear at present whether combined detection of TG2 and TG6 IgA/IgG without the use of antigliadin antibodies identifies all patients with gluten ataxia. Up to 60% of patients with gluten ataxia have evidence of cerebellar atrophy on MR imaging (Fig. 41.1). Investigation of the metabolic status of the cerebellum in 15 patients with gluten ataxia and 10 controls using proton MR spectroscopy demonstrated significant differences in mean N-acetylaspartate/creatine levels between patients with GA and healthy controls, suggesting that cerebellar neuronal physiology in these patients is abnormal (Wilkinson et al., 2005). Even in those patients without cerebellar atrophy, proton MR spectroscopy of the cerebellum was abnormal. There is emerging evidence that MR spectroscopy is often abnormal in patients with newly diagnosed CD with minimal or

Fig. 41.1. Severe cerebellar atrophy on axial MR imaging in a 40-year-old woman with a 10 year history of progressive ataxia diagnosed eventually with gluten sensitivity. She remains stable on a gluten-free diet but the ataxia is still present as a result of permanent damage to the cerebellum.

no neurologic complaints and that such abnormalities improve with the introduction of a gluten-free diet. The clinical improvement manifests after 1 year on the diet but continues for at least 2 years. The response to treatment with a gluten-free diet depends on the duration of the ataxia prior to the diagnosis of GRD. Loss of Purkinje cells in the cerebellum, the end result of prolonged gluten exposure in patients with GA, is irreversible, and prompt treatment is more likely to result in improvement or stabilization of the ataxia. Whilst the benefits of a gluten-free diet in the treatment of patients with CD and DH have long been established, there are very few studies, mainly case reports, of the effect of a gluten-free diet on the neurologic manifestations. Most of these reports primarily concern patients with established CD who then develop neurologic symptoms (Beversdorf et al., 1996; Hahn et al., 1998; Pellecchia et al., 1999). These studies suggest variable but overall favorable responsiveness to a gluten-free diet. A small, uncontrolled study looked at the use of intravenous immunoglobulins in the treatment of four patients with

GLUTEN-RELATED NEUROLOGIC DYSFUNCTION 611 GA without enteropathy (B€ urk et al., 2001b; Sander et al., peripheral neuropathy (Luostarinen et al., 2003). A large 2003). All patients improved. In all of these reports, population-based study of over 84 000 subjects in Sweden strict adherence to the gluten-free diet was assumed examined the risk of neurologic disease in patients with and no serologic evidence was provided. The best marker CD and found that polyneuropathy had a significant assoof strict adherence to a gluten-free diet is serologic eviciation with CD (Ludvigsson et al., 2007). In our own dence of elimination of circulating GRD-related antiUK-based study, 34% of patients with idiopathic sporadic bodies. Only one systematic study of the effect of a sensorimotor axonal neuropathy were found to have gluten-free diet on a cohort of patients presenting with circulating AGA (Hadjivassiliou et al., 2006b). Using ataxia, with or without an enteropathy, has been published anti-TG2 antibody detection an Italian study also showed (Hadjivassiliou et al., 2003b). This study also reported a significantly number of patients (21%) with peripheral serologic evidence of elimination of the antigliadin antineuropathy to be positive (Mata et al., 2006). Finally, in a bodies as a confirmation of strict adherence to the diet. tertiary referral centre in the US, retrospective evaluation A total of 43 patients with gluten ataxia were enrolled. of patients with neuropathy showed the prevalence of CD Of these, 26 adhered strictly to the gluten-free diet, had to be between 2.5% and 8% as compared to 1% in the serologic evidence of elimination of antibodies, and comhealthy population (Chin et al., 2003). prised the treatment group; 14 patients refused the diet Gluten neuropathy is defined as otherwise idiopathic and comprised the control group and 3 patients were sporadic neuropathy with serologic evidence of GRD. excluded as despite the diet their antibodies were still posThe commonest types are symmetric sensorimotor axonal itive. Patient and control groups were matched at baseline peripheral neuropathy and sensory ganglionopathy for all variables (age, duration of ataxia, etc.). There was (Hadjivassiliou et al., 2010b). Other types of neuropathies no significant difference in the baseline performance for have also been reported including asymmetric neuropathy each ataxia test between the two groups. There was signif(Kelkar et al., 1996; Hadjivassiliou et al., 1997; Chin et al., icant improvement in performance in test scores and in 2006), small fiber neuropathy (Brannagan et al., 2005), the subjective global clinical impression scale in the treatand rarely, pure motor neuropathy (Hadjivassiliou ment group when compared to the control group. The et al., 1997) or autonomic neuropathy (Gibbons and improvement was apparent even after excluding patients Freeman, 2005). Gluten neuropathy is a slowly progreswith an enteropathy. The study concluded that gluten-free sive disease with a mean age at onset of the neuropathy diet can be an effective treatment for GA. of 55 years (range 24–77 years) and a mean duration of There are no published randomized, placebo9 years (range 1–33 years). A third of the patients will have controlled studies on the subject, perhaps reflecting evidence of enteropathy on biopsy but the presence or the difficulties associated with such a study when the absence of an enteropathy does not predetermine the intervention is dietary elimination of gluten and the etheffect of a gluten-free diet (Hadjivassiliou et al., 2006b). ical considerations of randomizing patients with GA who Limited pathologic data available from postmortems have enteropathy. The current recommendation is that and nerve biopsies are consistent with an inflammatory patients presenting with progressive cerebellar ataxia etiology (perivascular lymphocytic infiltration). The evishould be screened for gluten sensitivity using antigliadin dence of effectiveness of gluten-free diet has largely IgG and IgA, anti-TG2 antibodies and if available antibeen derived from single or multiple case reports most TG6 antibodies. Patients positive for any of these antiof which suggest improvement of the neuropathy. The bodies with no alternative cause for their ataxia should only systematic, controlled study of the effect of a be offered a strict gluten-free diet with regular followgluten-free diet on 35 patients with gluten neuropathy, up to ensure that the antibodies are eliminated (usually with close serologic monitoring of the adherence to takes 6–12 months). Stabilization or even improvement the gluten-free diet, found significant improvement in of the ataxia at 1 year would be a strong indicator that the treated compared with the control group after 1 year the patient suffers from gluten ataxia. The commonest on the diet (Hadjivassiliou et al., 2006b). The improvereason for lack of response is lack of compliance with ment took the form of an increase in the sural sensory the diet. If patients on strict gluten-free diet continue to action potential, the predefined primary endpoint, and progress, with or without elimination of antibodies, the subjective improvement of the neuropathic symptoms. use of immunosuppressive medication (mycophenolate) Subgroup analysis suggested that the capacity for should be considered. Such cases are rare. recovery of the peripheral nerves may be less when the neuropathy is severe or that more time may be needed for such recovery to manifest. As there was a correlation Gluten neuropathy between disease severity and longer duration, gluten Up to 23% of patients with established CD on gluten-free neuropathy may be considered a progressive disease if diet have neurophysiologic evidence of a untreated. In the context of sensory ganglionopathy

612

M. HADJIVASSILIOU ET AL.

GRD has been shown to be as common a cause as Sj€ogren’s syndrome. Dorsal root ganglia demonstrate evidence of inflammatory infiltrates. The disease progresses slowly if untreated. Strict adherence to a gluten-free diet may result in stabilization or even improvement of the neuropathy irrespective of the presence of enteropathy (Hadjivassiliou et al., 2010b).

Gluten encephalopathy (headache and white matter abnormalities) Headache is a common feature in patients with GRD. In 2001 we reported a series of 10 patients with GRD and headache who in addition had CNS white matter abnormalities on MRI scan suggesting the term “gluten encephalopathy” to describe them (Hadjivassiliou et al., 2001). The headaches are usually episodic, similar to migraines, may be associated with focal neurologic deficits, and characteristically resolve with the introduction of a gluten-free diet. The white matter abnormalities can be diffuse or focal and do not resolve following a gluten-free diet, which simply arrests progression of these changes (Fig. 41.2). Their distribution is more suggestive of a vascular rather than demyelinating etiology. Multiple sclerosis does not seem to be associated with GRD on the basis

of numerous studies (Pengiran Tengah et al., 2004; Hadjivassiliou et al., 2005; Borhani Haghighi et al, 2007; Nicoletti et al, 2008). In patients with migraine there is an overrepresentation of CD with a prevalence of 4.4% versus 0.4% in the control population (Gabrielli et al., 2003). Using positron emission tomography (PET) brain imaging, a study on regional cerebral perfusion demonstrated that 73% of patients with CD not on a gluten-free diet, had at least one hypoperfused brain region as compared to 7% in healthy controls and in patients with CD on a gluten-free diet (Addolorato et al., 2004). Another study investigated the prevalence of white matter abnormalities in children with CD and found that 20% of patients had such abnormalities (Kieslich et al., 2001). Over the last 14 years we have encountered 70 patients with gluten encephalopathy, a figure that includes the initial 10 patients reported in the 2001 series. Gluten encephalopathy does not always occur in isolation and such patients will often have additional neurologic features such as ataxia, neuropathy, and cognitive deficits. A study from the Mayo Clinic emphasized the significant cognitive deficits encountered in 13 such patients (Hu et al., 2006). In comparison to gluten ataxia and gluten neuropathy there is a higher prevalence of enteropathy in patients with gluten encephalopathy (40/70), but the age at onset is the same. The observed improvement of the headaches and arrest of progression in the MRI brain abnormalities, suggest a causal link with gluten ingestion. Gluten encephalopathy represents a spectrum of clinical presentations, from episodic headaches responsive to a gluten-free diet at one end to severe debilitating headaches associated with focal neurologic deficits and abnormal white matter on MRI at the other.

Myoclonic ataxia

Fig. 41.2. Marked diffuse white matter abnormalities on MR imaging in a 55-year-old man with celiac disease presenting with headaches and mild ataxia. There was complete resolution of the headaches following the introduction of a glutenfree diet. Neurologically he remains stable.

This form of ataxia is much less common in comparison to gluten ataxia. It was first described in 1986 (Lu et al., 1986). It has been shown that the myoclonus is of cortical origin despite the presence of cerebellar atrophy (Bhatia et al., 1995). Some myoclonus can be seen in a number of such patients but it is not usually troublesome. In our series of patients (over 500) with neurologic manifestations of GRD we have encountered four patients with what appears to be focal disabling myoclonus. All patients had evidence of enteropathy on biopsy. In two patients, despite a strict gluten-free diet, their condition progressed. Both have been treated with mycophenolate resulting in some stabilization. In the remaining patients the ataxia responded to the gluten-free diet but the myoclonus persists. In some of these patients the apparent focal myoclonus resembles epilepsia

GLUTEN-RELATED NEUROLOGIC DYSFUNCTION partialis continua in neurophysiologic terms. The apparent refractoriness of this neurologic manifestation is mirrored by evidence of ongoing enteropathy despite a strict adherence to the gluten-free diet. The treatment of such patients remains problematic but the limited evidence from these small series suggests that mycophenolate may be a useful therapeutic intervention for those patients who appear to progress neurologically despite strict gluten-free diet.

Epilepsy A number of reports have suggested a link between epilepsy and CD (Chapman et al., 1978; Fois et al., 1994; Cronin et al., 1998). There is a particular type of focal epilepsy associated with occipital calcifications that appears to have a strong link with CD (Gobbi et al., 1992). This entity is common in Italy but rare in other countries (Fig. 41.3). It tends to affect young patients (mean age 16 years) and in the majority the seizures are resistant to antiepileptic drugs. The pathogenesis of the cerebral calcifications remains unclear. An autopsy study showed the depositions consisted of both calcium and silica, and microscopically were found in three main types: psammoma-like bodies without any identifiable relationship to cells, vessels, or other structures; small granular deposition along small vessels; and

Fig. 41.3. CT head scan on a 52-year-old patient with epilepsy and gluten sensitivity demonstrating occipital calcifications. This condition is rare outside Italy and it primarily affects children. This patient presented with loss of consciousness ataxia and cognitive decline.

613

focal scanty areas of calcium within neurons (Toti et al., 1996). As most of the reported cases are from Italy, Spain, and Argentina, it has been hypothesized that the syndrome of celiac disease, epilepsy, and cerebral calcifications is “a genetic, non-inherited, ethnically and geographically restricted syndrome associated with environmental factors” (Gobbi, 2005). Whilst studies examining the prevalence of CD amongst patients with epilepsy have suggested a prevalence of 1.2– 2.3%, larger more recent studies failed to demonstrate such an increased prevalence (Ranua et al., 2005). However, most studies on the subject suffer from the same methodologic problem of treating epilepsy as a homogeneous disorder. The only study that attempted to look at the prevalence of GRD in well characterized subgroups of patients with epilepsy found a significant association between AGA and temporal lobe epilepsy with hippocampal sclerosis (Paltola et al., 2009). Of interest are some case reports on patients with CD and epilepsy whose epilepsy improves following the introduction of gluten-free diet (Mavroudi et al., 2005; Harper et al., 2007).

Myopathy This is a relatively rare neurologic manifestation of GRD, first described by Henriksson et al. (1982). This study from Sweden reported that out of 76 patients with suspected polymyositis investigated at a neuromuscular unit, 17 had a history of gastrointestinal symptoms with evidence of malabsorption. Fourteen of these fulfilled the diagnostic criteria for polymyositis and of those, five were diagnosed with CD. A more recent study from Spain (Selva-O’Callaghan et al., 2007) demonstrated the prevalence of AGA antibodies amongst patients with inflammatory myopathies to be 31%. This was accompanied by a higher prevalence of CD within the same population when compared to healthy controls. The clinical data from our series of patients are based on 18 cases encountered over the last 14 years, 13 of which have been reported previously (Hadjivassiliou et al., 2007). Enteropathy was identified following duodenal biopsy in 10 of these patients. The mean age at onset of the myopathic symptoms was 54 years. Ten patients had predominantly proximal weakness, five patients had both proximal and distal weakness, and four patients had primarily distal weakness. Two patients had ataxia and neuropathy, and one patient had just neuropathy in addition to the myopathy. Serum creatine kinase (CK) level ranged from normal (25–190 IU/L) to 4380 IU/L at presentation. Inflammatory myopathy was the most common finding on neuropathologic examination (Fig. 41.4). Six patients received immunosuppressive treatment in addition to starting a glutenfree diet whereas the others went on a gluten-free diet

614

M. HADJIVASSILIOU ET AL.

Fig. 41.4. Muscle biopsy demonstrating focal inflammatory infiltration in a 60-year-old man with celiac disease who became profoundly weak after inadvertently eating a cake made out of rye flour. His weakness was profound and necessitated admission. He made a full recovery just with reintroduction of a strict gluten-free diet.

only. In the majority of those patients who did not receive immunosuppressive treatment there was clinical improvement of the myopathy with gluten-free diet, suggesting that the myopathy was etiologically linked to the GRD. One patient developed a profound myopathy after inadvertently eating rye flour. He made a full recovery by re-establishing a strict gluten-free diet. Two patients had histologic evidence of inclusion body myositis. It is interesting to note that inclusion body myositis shares the same HLA genetic predisposition with CD.

Fig. 41.5. Spinal cord atrophy on MR imaging of the thoracic cord of a 55-year-old man with celiac disease and sensory ataxia. Whilst the patient had celiac disease for many years, he never adhered to the diet strictly and went on to develop neurologic manifestations.

Myelopathy Clinical evidence of a myelopathy in the absence of vitamin and other deficiencies (particularly copper) can be a rare manifestation of CD (Fig. 41.5). It is usually associated with normal imaging of the spinal cord. However, there have been some recent reports of patients with neuromyelitis optica (Devic’s disease) and GRD who have antibodies to aquaporin-4 (Jacob et al., 2005; Jarius et al., 2008). Such patients clearly had abnormal MRI of the spinal cord but the diagnosis of CD was only made at the time of their neurologic presentation. Neuromyelitis optica and CD share the same HLA genetic susceptibility (HLA DQ2). There are very limited data on the effect of the diet on the likelihood of relapse of the disease, particularly given the fact that most patients with Devic’s disease end up on long-term immunosuppressive medication.

positivity for anti-GAD or anti-amphiphysin antibodies. It has a strong association with other autoimmune diseases (e.g., insulin-dependent diabetes mellitus (IDDM), hypothyroidism, etc.). We have found a high prevalence of gluten-related antibodies in patients with this condition over and above that expected from an association of two autoimmune diseases. The relapsing remitting nature of the condition makes a study of any responsiveness to gluten-free diet difficult. There is, however, evidence of reduction of the anti-GAD antibody titer following the introduction of a gluten-free diet suggesting that the diet may be beneficial in treating the condition (Hadjivassiliou et al., 2010c). This finding also supports the concept of prevention of autoimmunity in patients with GRD if the gluten-free diet is introduced early enough.

Stiff man syndrome

PATHOGENESIS

Stiff man syndrome (SMS) is a rare autoimmune disease characterized by axial stiffness, painful spasms, and

Current evidence suggests that neurologic manifestations are immune mediated. Postmortem examination

GLUTEN-RELATED NEUROLOGIC DYSFUNCTION from patients with gluten ataxia demonstrate patchy loss of Purkinje cells throughout the cerebellar cortex, a nonspecific finding in many cerebellar disorders. However, additional findings supporting an immune-mediated pathogenesis include diffuse infiltration mainly of T lymphocytes within the cerebellar white matter as well as marked perivascular cuffing with inflammatory cells (Hadjivassiliou et al., 1998). The peripheral nervous system also shows sparse lymphocytic infiltrates with perivascular cuffing being observed in sural nerve biopsy of patients with gluten neuropathy (Hadjivassiliou et al., 2006a), in dorsal root ganglia in patients with sensory neuronopathy (Hadjivassiliou et al., 2010a) and in patients with gluten myopathy due to GRD (Hadjivassiliou et al., 2007). There is evidence to suggest that there is antibody cross-reactivity between antigenic epitopes on Purkinje cells and gluten proteins. Serum from patients with GA and from patients with CD but no neurologic symptoms, display cross-reactivity with epitopes on Purkinje cells of both human and rat cerebellum (Hadjivassiliou et al., 2002). This reactivity can also be seen using polyclonal AGA and the reactivity eliminated by absorption with crude gliadin. When using sera from patients with GA there is evidence of additional antibodies targeting Purkinje cell epitopes since elimination of AGA alone is

Fig. 41.6. Duodenal biopsy from a patient presenting with progressive ataxia and positive antigliadin antibodies. The biopsy demonstrates the triad of crypt hyperplasia, villous atrophy, and increase in the intraepithelial lymphocytes. Only a third of patients with gluten ataxia will have enteropathy.

615

not sufficient to eliminate such reactivity (Fig. 41.6). There is some evidence that additional antibodies that may be causing such reactivity, including antibodies against one or more transglutaminase isoenzymes. TG2 belongs to a family of enzymes that covalently crosslink or modify proteins by formation of an isopeptide bond between a peptide-bound glutamine residue and a primary amine. However, in some instances TG2 may react with water in preference over an amine leading to the deamidation of glutamine residues. Gluten proteins (from wheat, barley, and rye), the immunologic trigger of GRD, are glutamine-rich donor substrates amenable to deamidation. Activation of TG2 and deamidation of gluten peptides appears to be central to disease development and is now well understood at a molecular level. However, events leading to the formation of autoantibodies against TG2 are still unclear. Questions also remain as to the contribution of these autoantibodies to organ-specific deficits. Anti-TG2 antibodies have been shown to be deposited in the small bowel mucosa of patients with GRD even in the absence of enteropathy. Furthermore such deposits have been found in extraintestinal sites, such as muscle and liver (Korponay-Szabo´ et al., 2004). Widespread deposition of transglutaminase antibodies has also been found around brain vessels in GA (Hadjivassiliou et al., 2006a). The deposition was most pronounced in the cerebellum, pons, and medulla. This finding suggests that such autoantibodies could play a role in the pathogenesis of the whole spectrum of manifestations seen in GRD. However, it is not clear whether these antibodies are derived from the circulation or if their production is mediated within target organs after stimulation of gutprimed gliadin-reactive CD4þ T cells. Variations in the specificity of antibodies produced in individual patients could explain the wide spectrum of manifestations. Whilst TG2 has been shown to be the autoantigen in CD (Dietrich et al., 1997), the epidermal transglutaminase TG3 has been shown to be the autoantigen in DH (Sa´rdy et al., 2002). More recently, antibodies against TG6, a primarily brain expressed transglutaminase, have been shown to be present in patients with GA (Hadjivassiliou et al., 2008a). In GA and DH, IgA deposits of TG6 and TG3 respectively seem to accumulate in the periphery of blood vessels. This could indicate that either the deposits originate from immune complexes formed elsewhere, and are accumulating as a consequence of enhanced vascular leaking, or that TG6/TG3 are derived from perivascular infiltrating inflammatory cells preceding deposit formation. Indeed perivascular cuffing with lymphocytes is a common finding in brain tissue from patients with GA but is also seen in peripheral nerve and muscle in patients with gluten neuropathy or myopathy. In most sera reactive

616

M. HADJIVASSILIOU ET AL.

with more than one TG isoenzyme, distinct antibody populations are responsible for such reactivity rather than this being a result of cross-reactivity with different TG isozymes. This makes shared epitopes less likely to be the cause for immune responses to other TGs and points to the possibility that TG isozymes other than TG2 can be the primary antigen in GRD. IgA deposition in brain vessels and the pathologic finding of perivascular cuffing with inflammatory cells, may indicate that vasculature-centered inflammation may compromise the blood–brain barrier, allowing exposure of the CNS to pathogenic antibodies, and therefore be the trigger of nervous system involvement. Indeed, TG2 is expressed by smooth muscle and endothelial cells in noninflamed brain, is an abundant component of the blood–brain barrier and autoantibody binding could initiate an inflammatory response. Anti-TG2 and other autoantibodies (e.g., AGA) may directly cause selective neuronal degeneration. It is possible that neuronal degeneration is a consequence of the anti-TG antibody spectrum, i.e., occurs in those patients with antibodies reactive with a neuronal TG. IgG class antibodies have been shown to be present in only 60% of CD patients whereas in GA patients positive for anti-TG, the prevalence was 90%. This shift from IgA to IgG may reflect the target organ involved (central nervous system rather than the small bowel). It could be argued that development and deposition of antibodies is an epiphenomenon rather than being pathogenic. One method to demonstrate the pathologic effect of an antibody is the passive transfer of the disease through antibody injection into a naı¨ve animal. While such experimental evidence exists for only very few antibody-mediated diseases, IgG fractions of patients with anti-GAD ataxia and stiff man syndrome have been shown to compromise motor function and impair learning in rodents, an effect possibly ascribed to antibodies against GAD (Manto et al., 2007). A common problem in such studies is to be able to demonstrate whether it is these specific antibodies or other autoantibodies in the IgG-fraction of patient sera that cause neuronal damage. Using a mouse model we have recently shown that serum from GA patients, as well as clonal monovalent anti-TG immunoglobulins derived using phage display, cause ataxia when injected intraventricularly in mice (Boscolo et al., 2010). The fact that not only Ig fractions but also monospecific scFvs mediate functional deficits shows that there is no requirement for complement activation or for the engagement of Fc receptors on Fc receptor-bearing cells in the brain. These data therefore provide evidence that anti-TG immunoglobulins (derived from patients) compromise neuronal function in selected areas of the brain once exposed to the CNS and suggest that this involves an immune system

independent mode of action. While these data implicate anti-TG antibodies in ataxia they do not explain the spectrum of distinct neurologic deficits currently ascribed to gluten sensitivity, nor why only a fraction of patients with circulating anti-TG antibodies are affected.

CONCLUSIONS GRD include immune-mediated diseases triggered by ingestion of gluten proteins. While celiac disease has been the most comprehensively studied of all GRD, dermatitis herpetiformis and neurologic manifestations are the commonest extraintestinal manifestations. To fully understand the immunologic insults resulting from gluten ingestion, the emphasis should perhaps shift toward the study of extraintestinal manifestations. In addition there is a need for the early identification of those patients that are specifically at risk of irreversible complications (e.g., gluten ataxia). To that effect, new diagnostic tools are now becoming available (e.g., antibodies against TG6) which may make a more reliable identification of those patients with neurologic manifestations a reality. Up to 40% of patients presenting to the gastroenterologist who are ultimately diagnosed with CD also have antibodies against TG6 in addition to antibodies against TG2. This subgroup of patients with classic CD presentation may well be the ones susceptible to the development of neurologic dysfunction if they continue to consume gluten, although this remains to be shown in longitudinal studies. The presence of gastrointestinal symptoms, however, offers a major potential advantage to this group, as it substantially increases their chances of being diagnosed with, and treated for, CD, whereas the diagnosis of those patients presenting purely with extraintestinal manifestations may be more difficult. The only way that this can be improved upon is by changing the perception of physicians that glutenrelated diseases are solely diseases of the gut.

REFERENCES Abele M, B€ urk K, Sch€ ols L et al. (2002). The aetiology of sporadic adult-onset ataxia. Brain 125: 961–968. Abele M, Schols L, Schwartz S et al. (2003). Prevalence of antigliadin antibodies in ataxia patients. Neurology 60: 1674–1675. Addolorato G, Di Giuda D, De Rossi G et al. (2004). Regional cerebral hypoperfusion in patients with celiac disease. Am J Med 116: 312–317. Almaguer-Mederos LE, Almira YR, Gongora EM et al. (2008). Antigliadin antibodies in Cuban patients with spinocerebellar ataxia type 2. J Neurol Neurosurg Psychiatry 79: 315–317. Anheim M, Degos B, Echaniz-Laguna A et al. (2006). Ataxia associated with gluten sensitivity, myth or reality? Rev Neurol 162: 214–221.

GLUTEN-RELATED NEUROLOGIC DYSFUNCTION Beversdorf D, Moses P, Reeves A et al. (1996). A man with weight loss, ataxia, and confusion for 3 months. Lancet 347: 448. Bhatia KP, Brown P, Gregory R et al. (1995). Progressive myoclonic ataxia associated with celiac disease. Brain 18: 1087–1093. Binder H, Solitaire G, Spiro H (1967). Neuromuscular disease in patients with steatorrhoea. Gut 8: 605–611. Borhani Haghighi A, Ansari N, Mokhtari M et al. (2007). Multiple sclerosis and gluten sensitivity. Clin Neurol Neurosurg 109: 651–653. Boscolo S, Lorenzon A, Sblattero D et al. (2010). Anti transglutaminase antibodies cause ataxia in mice. PLoS One 5: e9698. Brannagan TH, Hays AP, Chin SS et al. (2005). Small-fiber neuropathy/neuronopathy associated with celiac disease: skin biopsy findings. Arch Neurol 62: 1574–1578. Briani C, Zara G, Alaedini A et al. (2008). Neurological complications of coeliac disease and autoimmune mechanisms: a prospective study. J Neuroimmunol 195: 171–175. Bundey S (1967). Adult coeliac disease and neuropathy. Lancet 1: 851–852. B€ urk K, B€osch S, M€ uller CA et al. (2001a). Sporadic cerebellar ataxia associated with gluten sensitivity. Brain 124: 1013–1019. B€ urk K, Melms A, Schulz JB et al. (2001b). Effectiveness of intravenous immunoglobulin therapy in cerebellar ataxia associated with gluten sensitivity. Ann Neurol 50: 827–828. Bushara KO, Goebel SU, Shill H et al. (2001). Gluten sensitivity in sporadic and hereditary ataxia. Ann Neurol 49: 540–543. Chapman RWG, Laidlow JM, Colin-Jones D et al. (1978). Increased prevalence of epilepsy in coeliac disease. BMJ 2: 250–251. Chin RL, Sander HW, Brannagan TH et al. (2003). Celiac neuropathy. Neurology 60: 1581–1585. Chin RL, Tseng VG, Green PHR et al. (2006). Multifocal axonal polyneuropathy in celiac disease. Neurology 66: 1923–1925. Coers C, Telerman-Toppet N, Cremer M (1971). Regressive vacuolar myopathy in steatorrhea. Arch Neurol 24: 217–227. Collin P, Pirttila T, Nurmikko T et al. (1991). Celiac disease, brain atrophy and dementia. Neurology 41: 372–375. Combarros O, Infante J, Lo´pez-Hoyos M et al. (2000). Celiac disease and idiopathic cerebellar ataxia. Neurology 54: 2346. Cooke WT, Thomas-Smith W (1966). Neurological disorders associated with adult coeliac disease. Brain 89: 683–722. Cronin CC, Jackson LM, Feighery C et al. (1998). Coeliac disease and epilepsy. QJM 91: 303–308. Deconinck N, Scaillon M, Segers V et al. (2006). Opsoclonusmyoclonus associated with celiac disease. Pediatr Neurol 34: 312–314. Dick DJ, Abraham D, Falkous G et al. (1995). Cerebellar ataxia in coeliac disease – no evidence of a humoral aetiology. Postgrad Med J 71: 186. Dicke WK, Weijers HA, Van De Kamer JH (1953). Coeliac disease II The presence in wheat of a factor having a deleterious effect in cases of coeliac disease. Acta Paediatr 42: 34–42.

617

Dietrich W, Ehnis T, Bauer M et al. (1997). Identification of tissue transglutaminase as the autoantigen of celiac disease. Nat Med 3: 797–801. Elders C (1925). Tropical sprue and pernicious anaemia, aetiology and treatment. Lancet 1: 75–77. Finelli P, McEntee W, Ambler M et al. (1980). Adult celiac disease presenting as cerebellar syndrome. Neurology 30: 245–249. Fois A, Vascotto M, Di Bartolo RM et al. (1994). Celiac disease and epilepsy in pediatric patients. Childs Nerv Syst 10: 450–454. Gabrielli M, Cremonini F, Fiore G et al. (2003). Association between migraine and celiac disease: results from a preliminary case-control and therapeutic study. Am J Gastroenterol 98: 625–629. Gee S (1888). On the coeliac affection. St Bartholomew’s Hospital Reports 24: 17–20. Gibbons CH, Freeman R (2005). Autonomic neuropathy and celiac disease. J Neurol Neurosurg Psychiatry 76: 579–581. Gobbi G (2005). Coeliac disease, epilepsy and cerebral calcifications. Brain Dev 27: 189–200. Gobbi G, Bouquet F, Greco L et al. (1992). Coeliac disease, epilepsy and cerebral calcifications. Lancet 340: 439–443. Hadjivassiliou M, Gibson A, Davies-Jones GAB et al. (1996). Is cryptic gluten sensitivity an important cause of neurological illness? Lancet 347: 369–371. Hadjivassiliou M, Chattopadhyay AK, Davies-Jones GAB et al. (1997). Neuromuscular disorder as a presenting feature of celiac disease. J Neurol Neurosurg Psychiatry 63: 770–775. Hadjivassiliou M, Grunewald RA, Chattopadhyay AK et al. (1998). Clinical, radiological, neurophysiological and neuropathological characteristics of gluten ataxia. Lancet 352: 1582–1585. Hadjivassiliou M, Gr€ unewald RAG, Lawden M et al. (2001). Headache and CNS white matter abnormalities associated with gluten sensitivity. Neurology 56: 385–388. Hadjivassiliou M, Boscolo S, Davies-Jones GAB et al. (2002). The humoral response in the pathogenesis of gluten ataxia. Neurology 58: 1221–1226. Hadjivassiliou M, Gr€ unewald RA, Sharrack B et al. (2003a). Gluten ataxia in perspective: epidemiology, genetic susceptibility and clinical characteristics. Brain 126: 685–691. Hadjivassiliou M, Davies-Jones GAB, Sanders DS et al. (2003b). Dietary treatment of gluten ataxia. J Neurol Neurosurg Psychiatry 74: 1221–1224. Hadjivassiliou M, Sanders DS, Grunewald RAG (2005). Multiple sclerosis and occult gluten sensitivity. Neurology 64: 933–934. Hadjivassiliou M, Maki M, Sanders DS et al. (2006a). Autoantibody targeting of brain and intestinal transglutaminase in gluten ataxia. Neurology 66: 373–377. Hadjivassiliou M, Grunewald RA, Kandler RH et al. (2006b). Neuropathy associated with gluten sensitivity. J Neurol Neurosurg Psychiatry 77: 1262–1266. Hadjivassiliou M, Chattopadhyay AK, Gr€ unewald RA et al. (2007). Myopathy associated with gluten sensitivity. Muscle Nerve 35: 443–450.

618

M. HADJIVASSILIOU ET AL.

Hadjivassiliou M, Aeschlimann P, Strigun A et al. (2008a). Autoantibodies in gluten ataxia recognise a novel neuronal transglutaminase. Ann Neurol 64: 332–343. Hadjivassiliou M, Sanders DS, Woodroofe N et al. (2008b). Gluten ataxia. Cerebellum 7: 494–498. Hadjivassiliou M, Aeschlimann P, Sanders DS et al. (2009). Antibodies against TG6 as the only serological marker of gluten ataxia. Proceedings of the 13th International Coeliac Disease Symposium, p. 75, Amsterdam, April 09.1. Hadjivassiliou M, Sanders DS, Grunewald RA et al. (2010a). Gluten sensitivity: from gut to brain. Lancet Neurol 9: 318–330. Hadjivassiliou M, Rao DS, Wharton SB et al. (2010b). Sensory ganglionopathy due to gluten sensitivity. Neurology 75: 1003–1008. Hadjivassiliou M, Aeschlimann D, Grunewald RA et al. (2010c). GAD antibody associated neurological illness and its relationship to gluten sensitivity. Acta Neurol Scand 123: 175–180. http://dx.doi.org/10.1111/J.1600-0404.2010.01356.x. Hadjivassiliou M, Sanders DS, Hoggard N (2011). Magnetic resonance imaging and spectroscopy of the cerebellum in patients with celiac disease and minor neurological complains. Proceedings of the 14th International Coeliac Disease Symposium 2011, p. 30, Oslo, Norway, June 2011. Hahn JS, Sum JM, Bass D et al. (1998). Celiac disease presenting as gait disturbance and ataxia in infancy. J Child Neurol 13: 351–353. Harper E, Moses H, Lagrange A (2007). Occult celiac disease presenting as epilepsy and MRI changes that responded to gluten-free diet. Neurology 68: 533. Henriksson KG, Hallert C, Norrby K et al. (1982). Polymyositis and adult celiac disease. Acta Neurol Scand 65: 301–319. Hermaszewski RA, Rigby S, Dalgleish AG (1991). Coeliac disease presenting with cerebellar degeneration. Postgrad Med J 67: 1023–1024. Holmes GKT (1997). Neurological and psychiatric complications in coeliac disease. In: G Gobbi, F Anderman, S Naccarato et al. (Eds.), Epilepsy and Other Neurological Disorders in Coeliac Disease. John Libbey, London, pp. 251–264. Hu WT, Murray JA, Greenway MC et al. (2006). Cognitive impairment and celiac disease. Arch Neurol 63: 1440–1446. Hunt KA (2008). Newly identified genetic risk variants for coeliac disease related immune response. Nat Genet 40: 395–402. Ihara M, Makino F, Sawada H et al. (2006). Gluten sensitivity in Japanese patients with adult-onset cerebellar ataxia. Intern Med 45: 135–140. Jacob S, Zarei M, Kenton A et al. (2005). Gluten sensitivity and neuromyelitis optica: two case reports. J Neurol Neurosurg Psychiatry 76: 1028–1030. Jarius S, Jacob S, Waters P et al. (2008). Neuromyelitis optica in patients with gluten sensitivity associated with antibodies to aquaporin-4. J Neurol Neurosurg Psychiatry 79: 1084. Kaplan JG, Pack D, Horoupian D et al. (1988). Distal axonopathy associated with chronic gluten enteropathy: a treatable disorder. Neurology 38: 642–645.

Kelkar P, Ross M, Murray J (1996). Mononeuropathy multiplex associated with celiac disease. Muscle Nerve 19: 234–236. Kepes JJ, Chou SM, Price LW (1975). Progressive multifocal leukoencephalopathy with 10-year survival in a patient with nontropical sprue. Neurology 25: 1006–1012. Kieslich M, Errazuriz G, Rosselt HG et al. (2001). Brain white matter lesions in celiac disease: a prospective study in diet treated patients. Paediatrics 108: E21. Kinney HC, Burger PC, Hurwitz BJ et al. (1982). Degeneration of the central nervous system associated with celiac disease. J Neurol Sci 53: 9–22. Korponay-Szabo´ IR, Laurila K, Szondy Z et al. (2003). Missing endomysial and reticulin binding of celiac antibodies in transglutaminase 2 knockout tissues. Gut 52: 199–204. Korponay-Szabo´ IR, Halttunen T, Szalai Z et al. (2004). In vivo targeting of intestinal and extraintestinal transglutaminase 2 by coeliac autoantibodies. Gut 53: 641–648. Kristoferitsch W, Pointer H (1987). Progressive cerebellar syndrome in adult coeliac disease. J Neurol 234: 116–118. Lu CS, Thompson PD, Quin NP et al. (1986). Ramsay Hunt syndrome and coeliac disease: a new association. Mov Disord 1: 209–219. Ludvigsson JF, Olsson T, Ekbom A et al. (2007). A population based study of celiac disease, neurodegenerative and neuroinflammatory diseases. Aliment Pharmacol Ther 25: 1317–1327. Luostarinen LK, Collin PO, Paraaho MJ et al. (2001). Coeliac disease in patients with cerebellar ataxia of unknown origin. Ann Med 33: 445–449. Luostarinen L, Himanen SL, Luostarinen M et al. (2003). Neuromuscular and sensory disturbances in patients with well treated celiac disease. J Neurol Neurosurg Psychiatry 74: 490–494. Manto MU, Laute MA, Aguera M et al. (2007). Effects of antiglutamic acid decarboxylase antibodies associated with neurological diseases. Ann Neurol 61: 544–551. Marks J, Shuster S, Watson AJ (1966). Small bowel changes in dermatitis herpetiformis. Lancet ii: 1280–1282. Marsh M (1992). Gluten, major histocompatibility complex and the small intestine. Gastroenterology 102: 330–354. Mata S, Renzi D, Pinto F et al. (2006). Anti-tissue transglutaminase IgA antibodies in peripheral neuropathy and motor neuronopathy. Acta Neurol Scand 114: 54–58. Mavroudi A, Karatza E, Papastavrou T et al. (2005). Succesful treatment of epilepsy and celiac disease with a gluten-free diet. Pediatr Neurol 33: 292–295. Morris JS, Ajdukiewicz AB, Read AE (1970). Neurological disorders and adult celiac disease. Gut 11: 549–554. Muller AF, Donnelly MT, Smith CML et al. (1996). Neurological complications of coeliac disease – a rare but continuing problem. Am J Gastroenterol 91: 1430–1435. Nicoletti A, Patti F, Lo Fermo S et al. (2008). Frequency of celiac disease is not increased among multiple sclerosis patients. Mult Scler 14: 698–700. Paltola M, Kaukinen K, Dastidar P et al. (2009). Hippocampal sclerosis in refractory temporal lobe epilepsy is associated with gluten sensitivity. J Neurol Neurosurg Psychiatry 80: 626–630.

GLUTEN-RELATED NEUROLOGIC DYSFUNCTION Paulley JW (1954). Observation on the aetiology of idiopathic steatorrhoea, jejunal and lymph node biopsies. Br Med J 2: 1318–1321. Pellecchia MT, Scala R, Filla A et al. (1999a). Idiopathic cerebellar ataxia associated with celiac disease: lack of distinctive neurological features. J Neurol Neurosurg Psychiatry 66: 32–35. Pengiran Tengah CD, Lock RJ, Unsworth DJ et al. (2004). Multiple sclerosis and occult gluten sensitivity. Neurology 62: 2326–2327. Pereira AC, Edwards MJ, Buttery PC et al. (2004). Choreic syndrome and coeliac disease: a hitherto unrecognised association. Mov Disord 19: 478–482. Ranua J, Luoma K, Auvinen A et al. (2005). Celiac diseaserelated antibodies in an epilepsy cohort and matched reference population. Epilepsy Behav 6: 388–392. Reed AC, Ash JE (1927). Atypical sprue. Arch Intern Med 40: 786–799. Sander HW, Magda P, Chin RL et al. (2003). Cerebellar ataxia and celiac disease. Lancet 362: 1548. Sanders DS, Patel D, Stephenson TJ et al. (2003). A primary care cross-sectional study of undiagnosed adult celiac disease. Eur J Gastroenterol Hepatol 15: 407–413.

619

Sa´rdy M, Ka´rpa´ti S, Merkl B et al. (2002). Epidermal transglutaminase (TGase3) is the autoantigen of dermatitis herpetiformis. J Exp Med 195: 747–757. Selva-O’Callaghan A, Casellas F, De Torres I et al. (2007). Celiac disease and antibodies associated with celiac disease in patients with inflammatory myopathy. Muscle Nerve 35: 49. Tison F, Arne P, Henry P (1989). Myoclonus and adult celiac disease. J Neurol 236: 307–308. Toti P, Balestri P, Cano M et al. (1996). Celiac disease with cerebral calcium and silica deposits: X-ray spectroscopic findings, an autopsy study. Neurology 46: 1088–1092. Ward ME, Murphy JT, Greenberg GR (1985). Celiac disease and spinocerebellar degeneration with normal vitamin E status. Neurology 35: 1199–1201. Wilkinson ID, Hadjivassiliou M, Dickson JM et al. (2005). Cerebellar abnormalities on proton MR spectroscopy in gluten ataxia. J Neurol Neurosurg Psychiatry 76: 1011–1013. Woltman HW, Heck FJ (1937). Funicular degeneration of the spinal cord without pernicious anemia. Arch Intern Med 60: 272–300.

Handbook of Clinical Neurology, Vol. 120 (3rd series) Neurologic Aspects of Systemic Disease Part II Jose Biller and Jose M. Ferro, Editors © 2014 Elsevier B.V. All rights reserved

Chapter 42

Neurologic manifestations of malabsorption syndromes RONALD F. PFEIFFER* Department of Neurology, University of Tennessee Health Science Center, Memphis, TN, USA

INTRODUCTION Any engine requires fuel to run and the human body is no different. However, fueling the body is not quite as simple as placing the nozzle from the gas pump into the automobile gas tank and pumping in the gasoline. For the human body, the required fuel is complex, and multiple steps are required to ensure that the fuel is converted into usable form and actually is taken into the body. Impaired function at any of these steps can result in failure to properly absorb the fuel. The fuel itself consists of three basic types of nutrients: carbohydrates, fats, and proteins. Intermixed with these three elements are other substances or additives – vitamins and minerals – that are also required by the body to function properly and must be absorbed along with the essential nutrients. Digestion actually begins in the mouth, where food is chewed and intermixed with salivary enzymes such as amylase. After transfer to the stomach, further mechanical disruption of the food into smaller and smaller particles takes place until the now semi-liquid chyme is ready to be disgorged from the stomach into the duodenum in an orderly fashion, which is controlled by the pyloric sphincter. Particles generally must be smaller than 0.5 mm to be allowed egress from the stomach into the duodenum through the pyloric sphincter (Meyer, 1980). Once in the small intestine, the chyme is exposed to additional enzymes secreted from the pancreas, to bile salts released from the gall bladder, and to still more enzymes found on the brush border membrane and within the mucosal surface of the small intestine itself, all of which promote digestion and ready the nutrients for absorption (Farrell, 2002). The small intestine then continues to mix and propel its contents, with the mixing ensuring maximum exposure of its contents to the intestinal mucosa, where actual absorption occurs. Although

the small intestine appears to be tucked into a relatively small compartment of the body, it is actually 22–23 feet long in adults and has an absorptive surface of approximately 300 square yards (250 square meters), which is the approximate size of a tennis court (Insel et al., 2010). This is possible because of the huge number of folds, villi, and microvilli that constitute and markedly expand the absorptive surface.

ABSORPTION AND MALABSORPTION Absorption of nutrients A full description of the intricacies of gastrointestinal absorption is beyond the scope of this chapter (and the expertise of this neurologist), but a very brief summary of the mechanisms of absorption of fat, carbohydrate, protein, vitamins, minerals, and trace elements will be undertaken. Approximately 35% of adult food energy intake consists of lipids, predominantly triglyceride (Pot et al., 2012). Most dietary lipid is absorbed in the jejunum (Borel et al., 1989). Because fat is insoluble in water, intricate mechanisms exist to assist in its absorption. Dietary fat is first broken down into emulsified droplets that are stabilized by coating with phospholipid and then acted upon by lipases, initially in the stomach and then more extensively in the duodenum, which break down triglyceride into fatty acids and monoglyceride. These products of lipolysis then are formed into micelles by bile salts, which then travel to the enterocyte bush border membrane for absorption (Maldonado-Valderrama et al., 2011). Other dietary lipids, such as phospholipids and cholesterol esters, are handled in a slightly different fashion but are also transported to the enterocyte brush membrane in micelles (Farrell, 2002). Within the enterocyte, triglyceride is resynthesized and

*Correspondence to: Ronald F. Pfeiffer, M.D., Professor and Vice Chair, Department of Neurology, University of Tennessee Health Science Center, 855 Monroe Avenue, Memphis, TN 38163, USA. Tel: þ1-901-448-5209, Fax: þ1-901-448-7440, E-mail: [email protected]

622 R.F. PFEIFFER (Farrell, 2002). Intricate systems are present to control packaged, along with cholesterol and phospholipids, into and regulate the absorption of minerals and trace elechylomicrons and very low-density lipoproteins for export ments, which takes place primarily in the small intestine. into the lymphatic circulation (Thomson et al., 1993). An important component of the surface of chylomicrons is apolipoprotein, which is necessary for both their formaMalabsorption tion within and secretion from the enterocyte (Ros, 2000). Carbohydrate accounts for approximately 48% of If the stomach is unable to properly carry out its mixing function, either because prior disease has impaired its food energy intake in adults (Whitton et al., 2011). Digesmuscular function or prior surgery, such as gastric resectable dietary carbohydrate consists primarily of starch in the form of the polysaccharides amylose and amylopection or bariatric surgery, has diminished its holding tin, which are made up of long chains of glucose molecapacity, inadequately mixed and osmotically active cules, or of sugars in the form of disaccharides such as material may be dumped rapidly into the duodenum. lactose and sucrose, or monosaccharides such as fructose This, in turn, may result in inadequate mixing of bile and glucose. First salivary amylase and then pancreatic salts and pancreatic enzymes with the chyme while at amylase break starch down into short oligosaccharides, the same time the increased osmotic pressure draws additional fluid into the intestine, which increases the such as maltose and maltriose. These oligosaccharides bulk of the ingested material and causes it to move and the dietary disaccharides cannot themselves be absorbed, but are further hydrolyzed to monosaccharides rapidly through the intestine, limiting both time and (glucose, fructose, galactose) by hydrolases (e.g., maltase, extent of contact of the ingested material with the inteslactase, sucrase) within the brush border membrane of the tinal mucosa, where absorption would normally occur. enterocytes, where they are then absorbed via saturable The net result of this mad dash of partially digested carrier-mediated transport systems. food through the intestine can be reduced absorption Protein provides approximately 10–15% of energy of nutrients and associated diarrhea, the hallmarks of malabsorption. intake in the average Western diet (Farrell, 2002) and Malabsorption may also occur via other mechanisms. is the primary source of amino acids. It can be derived both from animal and plant sources. Protein digestion Impaired micelle formation due to reduced luminal conis initiated within the stomach by the actions of proteocentrations of bile salts, as a consequence of hepatic or lytic enzymes called pepsins, which break protein down gall bladder dysfunction, can result in fat malabsorption into smaller peptides that are then released into the (Van Deest et al., 1968). Pancreatic insufficiency from a small intestine. There the peptides encounter pancreatic variety of causes can result in decreased lipase secretion proteases such as trypsin, chymotrypsin, elastin, and with consequent fat malabsorption and decreased secretion of trypsin and chymotrypsin with resultant protein carboxypeptidase A and B, which work together to reduce malabsorption (Owens and Greenson, 2007). Disease the peptides further into oligopeptides and individual amino acids. At the brush border membrane there are processes affecting the enterocytes can impair both fat still more peptidases. Within the brush border there are absorption and chylomicron formation, with subsequent separate systems for absorbing neutral, basic, and acimalabsorption of fat. Poor mixing of gastric and intestidic amino acids. nal contents and mucosal disease processes also can Vitamins can be divided into those that are waterimpair protein and carbohydrate absorption. Loss or soluble and those that are fat-soluble. Water-soluble vitareduction of the absorptive surface, whether due to disease processes or to surgical removal, may also result in mins include ascorbic acid (vitamin C), thiamine (vitamin malabsorption. B1), riboflavin (vitamin B2), niacin (vitamin B3), pyridoxine (vitamin B6), cobalamin (vitamin B12), folic acid, The clinical signs and symptoms of malabsorption pantothenic acid, and biotin. Although in the past it classically involve the gastrointestinal system, with was assumed that most of these substances were absorabdominal distension, abdominal pain, flatulence, diarbed by passive diffusion, it is now recognized that active, rhea, weight loss, and even ascites. However, systemic carrier-mediated processes are responsible (Said, 2011). signs also may appear. Abnormalities of the skin and Vitamins A, D, E, and K are all polar lipids and mucous membranes can become evident, as can muscuthus, are fat-soluble rather than water-soluble. Thereloskeletal, renal, and hematologic dysfunction. fore the initial steps in their digestion probably consist Neurologic involvement may develop in some malabof their transfer from the food matrix in which they sorptive disorders and can assume a variety of appearare embedded to micelles or perhaps to smaller lipid vesances. It is not possible in this chapter to fully detail icles (Borel et al., 2001). They are then absorbed in the all disorders of gastrointestinal absorption that may small intestine, either by passive diffusion (vitamins result in neurologic dysfunction. Some of these disorA, D, E) or by a carrier-mediated process (vitamin K1) ders are discussed in detail in their own chapters in this

NEUROLOGIC MANIFESTATIONS OF MALABSORPTION SYNDROMES 623 compendium and will be treated lightly here. Others will limb dysmetria, and even myoclonic dystonia have also be discussed more fully, but the disease processes disbeen described (Angelini et al., 2002; Aslam et al., cussed in the following paragraphs might best be consid2004; Hammond and Wang, 2008). ered a sampling of disorders that may be of most interest Abnormalities consistent with peripheral neuropathy to the adult neurologist. may be evident on electromyography and nerve conduction studies (EMG/NCV) but are not universally present NEUROLOGIC DYSFUNCTION IN (Ko and Park-Ko, 1999; Hammond and Wang, 2008). MALABSORPTION SYNDROMES Somatosensory evoked potentials may demonstrate abnormalities indicative of posterior column dysfuncThe list of congenital disorders of the gastric and intestion (Puri et al., 2005). Diffuse white matter changes tinal mucosa that result in malabsorption is very long and have also been described in individuals with vitamin E includes disorders that are characterized by malabsorpdeficiency, both in the cerebrum (Aslam et al., 2004) tion of amino acids (e.g., Hartnup disorder, cystinuria, and in the spinal cord (Vorgerd et al., 1996). blue diaper syndrome, Lowe’s syndrome, oasthouse synThe appearance of symptoms of vitamin E deficiency drome, and others), carbohydrates (e.g., lactase defican be strikingly delayed. In patients post gastrectomy, it ciency, trehalase deficiency, glucose galactose may take up to 50 months for evidence of vitamin E defimalabsorption, and others), fat (see below), and also varciency to appear (Ueda et al., 2009). The same investigaious vitamins and minerals (H€ ogenauer and Hammer, tors reported that replacement doses of vitamin E 2002). Many of these present during infancy and not needed to be 300 mg/day or more. all produce neurologic dysfunction. All cannot be covered here. The following paragraphs will focus on disorFamilial hypocholesterolemia ders that do produce neurologic dysfunction; both acquired and inherited disorders will be addressed and Three distinct genetic disorders, familial hypobetalipothose that may appear in adult life will be preferentially proteinemia (FHBL), abetalipoproteinemia (ABL), and described. chylomicron retention disease (CRD), have been identified as causes of chronic diarrhea, malabsorption, malVitamin E deficiency nutrition, growth retardation, and vitamin E deficiency. Neurologic dysfunction in the setting of vitamin E defiOf the three, neurologists are most familiar with ABL, ciency can be genetic in origin, with an autosomal recespreviously known as the Bassen–Kornzweig syndrome sive inheritance pattern and a clinical presentation that (Bassen and Kornzweig, 1950; Sturman, 1968), but can mimic Friedreich’s ataxia, due to a mutation in the awareness of the other two is of value. a-tocopherol transfer protein gene (Ben Hamida et al., ABL is an autosomal recessive disorder due to a 1993; Ouahchi et al., 1995; Fogel and Perlman, 2007; Di mutation in the microsomal triglyceride transfer protein Donato et al., 2010). In most instances, however, it is (MTP) gene on chromosome 4 (Shoulders et al., 1993). the consequence of fat malabsorption (Laplante et al., MTP acts as a chaperone that transfers lipids such as tri1984; Ayuso Blanco et al., 1994). This can occur followglycerides, cholesterol esters and phospholipids onto ing both partial and complete gastrectomy (Rino et al., apolipoprotein B (APOB), thus promoting the secretion 2007; Ueda et al., 2009), in the setting of primary biliary of chylomicrons from the enterocytes and very lowcirrhosis (Sokol et al., 1989) or other biliary diseases density lipoproteins (VLDLs) from hepatocytes (Zamel (Ayuso Blanco et al., 1994), in individuals with pancreet al., 2008; Zeissig et al., 2010). Mutations lead to a atic dysfunction (Yokota et al., 1990), in patients with nonfunctional MTP, with resultant impaired biogenesis common variable immunodeficiency with associated of chylomicrons and VLDL and inability to absorb enteropathy (Aslam et al., 2004; Malamut et al., 2010), fats and fat-soluble vitamins, perhaps most importantly in persons with inflammatory bowel disease (Howard vitamin E. The clinical features of ABL include steatoret al., 1982; Vorgerd et al., 1996), or with cystic fibrosis rhea, diarrhea, retinitis pigmentosa, acanthocytosis, (Bye et al., 1985; Willison et al., 1985). and a variety of neurologic features; hepatic manifestaThe neurologic symptoms and signs in individuals with tions due to hepatic steatosis, occasionally leading to vitamin E deficiency can be quite varied. Ataxia is frecirrhosis, may also be present (Braegger et al., 1998). quently present. Dysarthria and nystagmus may occur. Blood lipid analysis demonstrates extremely low plasma Symptoms and signs of peripheral neuropathy, including levels of total cholesterol, VLDL, and low-density lipoparesthesias, impaired proprioception, impaired vibratory proteins (LDL); APOB, triglycerides, and chylomicrons perception, and hyporeflexia are also common. Proximal are virtually absent (Stevenson and Hardie, 2001; muscle weakness, myopathy, hyperreflexia, extensor Palau and Espino´s, 2006; Tarugi et al., 2007). Gastroinplantar responses, pigmentary retinopathy, action tremor, testinal symptoms are usually evident during infancy,

624

R.F. PFEIFFER

but neurologic dysfunction may not appear until individuals are in their teens or even later (Fogel and Perlman, 2007). Neurologic dysfunction typically consists of progressive cerebellar ataxia. Peripheral neuropathy may also develop. The neuropathy is sensorimotor (but predominantly sensory) in character and primarily associated with impairment of position and vibratory sensation, along with reduced or absent muscle stretch reflexes; both demyelinating (Wichman et al., 1985) and axonal (Iannaccone and Sokol, 1986) pathology have been described. Both the ataxia and the peripheral neuropathy have most often been attributed to vitamin E deficiency, but investigators describing the demyelinating features of the neuropathy have questioned its relationship to vitamin E deficiency. Additional neurologic abnormalities have been described in individuals with ABL. Upper motor neuron signs, such as hyperreflexia and Babinski signs, have been reported (Zamel et al., 2008), as have resting and postural tremor (Soejima et al., 2006). Treatment of neurologic dysfunction with both vitamin E and vitamin A has been advocated, but results have been mixed (Grant and Berson, 2001; Zamel et al., 2008). In contrast to ABL, FHBL is an autosomal codominant disorder in which heterozygotes may have mild symptoms or be asymptomatic except for low plasma cholesterol, whereas homozygotes may be clinically indistinguishable from individuals with ABL (Noto et al., 2009; Peretti et al., 2010). In approximately 50% of individuals, FHBL is due to a mutation in the APOB gene, which is on chromosome 2 (Whitfield et al., 2003; Tarugi et al., 2007). This results in the formation of a truncated APOB protein. The lipid profile of individuals with homozygous FHBL is similar to that of individuals with ABL; in contrast, heterozygotes have reduced but not absent total cholesterol, triglyceride, LDL, and APOB levels and high-density lipoproteins (HDL) may actually be elevated (Linton et al., 1993; Peretti et al., 2010). Homozygous individuals frequently develop retinitis pigmentosa and virtually always have acanthocytosis; neurologic dysfunction is similar to that present in ABL, but may be somewhat less severe (Linton et al., 1993). Neurologic dysfunction is unusual in heterozygotes. CRD (also called Anderson’s disease) is a very rare autosomal recessive disorder due to mutation in the SAR1B gene on chromosome 5, which encodes the SAR1B protein (Georges et al., 2011). SAR1B is involved with the transport of prechylomicron transport vesicles from the endoplasmic reticulum to the Golgi apparatus in enterocytes; impaired function of the protein results in failure to release chylomicrons following a fatcontaining meal and the accumulation of lipids within the enterocytes (Shoulders et al., 2004; Tarugi et al.,

2007; Peretti et al., 2010). In CRD, total cholesterol, LDL, HDL, and APOB are all reduced, but triglycerides are normal. Gastrointestinal dysfunction is evident during infancy, but neurologic dysfunction typically does not appear until the teenage years or adulthood (Peretti et al., 2010). Symptoms suggestive of a peripheral polyneuropathy are the most common neurologic presentation (Peretti et al., 2009), but in adults ataxia, myopathy, and action tremor have all been described (Gauthier and Sniderman, 1983; Peretti et al., 2010). Vitamin E deficiency is the presumed etiology for the neurologic deficits; Peretti et al. (2010) noted that individuals with the more pronounced abnormalities had the lowest vitamin E levels at the time of diagnosis. Treatment with vitamins E, A, and D are recommended to prevent neurologic, ophthalmologic, and osteopenic complications of CRD (Gauthier and Sniderman, 1983; Peretti et al., 2010).

Celiac disease The topic of gluten sensitivity is covered extensively in another chapter in this volume. Celiac disease (CD) will be covered briefly here to emphasize that patients with the classic gastrointestinal symptoms and pathology of CD may also develop neurologic dysfunction. However, it is not at all certain that the neurologic abnormalities of classic CD are the result of malabsorption; immunologic mechanisms may be a more probable explanation (B€ urk et al., 2009). The prevalence of CD, at least in American and European populations, has been estimated to be approximately 1% (Green and Cellier, 2007; Tjon et al., 2010), but recent studies suggest that the number of undiagnosed patients may be considerable and the prevalence much higher than previously proposed (Vilppula et al., 2008). The classic gastrointestinal symptoms of CD primarily are due to fat malabsorption and consist of diarrhea, weight loss, and gassy distension that develop as a consequence of damage to the mucosa of the small intestine, triggered by an immune-mediated response to gluten, the protein fraction of wheat. Malabsorption develops as a consequence of the mucosal injury, which results in blunting and atrophy of the villi, along with crypt hyperplasia. Individuals with classic CD display the presence antigliadin antibodies, both IgG and IgA. They also display the presence of additional gliadinrelated antibodies, such as antiendomysial and antitransglutaminase antibodies. Neurologic dysfunction has been reported to develop in 6–12% of individuals with CD (Pellecchia et al., 1999; Lagerqvist et al., 2001; Vaknin et al., 2004). A broad array of neurologic manifestations has been described, including peripheral neuropathy (Chin et al., 2003;

NEUROLOGIC MANIFESTATIONS OF MALABSORPTION SYNDROMES Briani et al., 2005; Bushara, 2005), myopathy (UygurBayramicli and Ozel, 2011), epilepsy (Bushara, 2005), myelopathy (Cooke and Smith, 1966), neuromyelitis optica (Jacob et al., 2005), headache (Cicarelli et al., 2003; Morello et al., 2003; Bushara, 2005), restless legs (Manchanda et al., 2009; Weinstock et al., 2010; UygurBayramicli and Ozel, 2011), acute inflammatory demyelinating neuropathy (Midha et al., 2007; Gupta and Kohli, 2010), chorea (Pereira et al., 2004), paroxysmal nonkinesigenic dystonia (Hall et al., 2007), autonomic imbalance (Barbato et al., 2010), and others. Some of the reports may simply reflect coincidence and in most of the others no clear connection to malabsorption has been suggested. The two neurologic disorders described most often in the setting of celiac disease are ataxia and peripheral neuropathy. With regard to peripheral neuropathy, one review of the existing literature prompted a conclusion that an association of celiac disease and peripheral neuropathy is unlikely and that celiac disease should not be considered in the workup of patients with chronic peripheral neuropathy (Rosenberg and Vermeulen, 2005).

Tropical sprue Tropical malabsorption is a syndrome that may affect both indigenous residents of tropical countries and travelers visiting or residing in the tropics (Ramakrishna et al., 2006). Both secondary forms, in which an etiology has been identified, and primary (idiopathic) forms have been described. Small intestine mucosal damage inflicted by protozoa (e.g., Giardia intestinalis, Cryptosporidium parvum, Isospora belli, Cyclospora cayetanensis), helminths (e.g., Strongyloides stercoralis, Capillaria philippinensis), bacteria (e.g., Mycobacterium tuberculosis) and viruses (possibly human immunodeficiency virus) may all produce a malabsorption syndrome, as can a variety of other disease processes of inflammatory, autoimmune, neoplastic, or pancreatic origin (Ramakrishna et al., 2006). It is in individuals in whom no etiology can be ascertained that the name tropical sprue (TS) has been applied. Although currently infrequently encountered in North America, TS has been reported to account for approximately 40% of malabsorption in children and adults in some portions of South Asia (Ranjan et al., 2004), although others have found it to be a rare cause of small bowel diarrhea (Thakur et al., 2006). Gastrointestinal symptoms of TS include chronic nonbloody diarrhea, bloating, weight loss, and abdominal cramping (Batheja et al., 2010). The mucosal changes in tropical sprue are sometimes indistinguishable from those of CD, although TS typically involves the entire length of the small intestine, whereas CD typically

625

spares the terminal ileum (Batheja et al., 2010). The mucosal damage in TS results in malabsorption of fat, carbohydrates, and multiple vitamins, including folate and vitamins A, E, and B12 (Ramirez et al., 1973; Glynn, 1986; Ramakrishna et al., 2006). Neurologic dysfunction may develop in the setting of TS. In one study, neurologic symptoms were documented in 67% (16/24) of individuals with TS (Iyer et al., 1973). In this study, proximal muscle weakness was present in 15 of the 16 individuals with neurologic symptoms, but on electrophysiologic testing only 10 had evidence of myopathy; peripheral neuropathy was noted in eight. Night blindness, presumably due to vitamin A deficiency, and combined system degeneration, presumably the result of vitamin B12 deficiency, have been described in TS (Ramakrishna et al., 2006). Peripheral neuropathy in patients with TS has been attributed to vitamin E deficiency (Ghalaut et al., 1995). Periodic paralysis has been reported in an individual with TS (Ghosh et al., 1994). Antibiotic therapy, typically with tetracycline or doxycycline for several months, and vitamin replacement therapy are the standard treatments for TS, but abnormal small intestine permeability may remain evident following treatment and some stool frequency and weight loss may persist (Kumar et al., 2011).

Wernicke’s encephalopathy Neurologists are well acquainted with Wernicke’s encephalopathy (WE) in the setting of chronic alcoholism with nutritional thiamine deficiency, but it can also be the result of malabsorption of thiamine. Considering the possibility of WE in patients who are not alcoholics can be especially problematic since the full classic triad of neurologic features of WE, mental status changes, ophthalmoplegia (nystagmus is actually more common than ophthalmoplegia), and gait ataxia, develops in only 10–16% of affected individuals (Harper et al., 1986; Weathers and Lewis, 2009). Thiamine is primarily absorbed in the duodenum, but the stomach may also play a role (Uruha et al., 2011). In keeping with this, the development of WE in a patient with peptic ulcer disease was attributed to gastric malabsorption of thiamine due to severe gastric mucosal lesions (Uruha et al., 2011). WE has also been documented following bariatric surgery, including techniques such as Roux-en-Y gastric bypass, vertical banded gastroplasty, and gastric partitioning (Rothrock and Smith, 1981; Seehra et al., 1996; Cirignotta et al., 2000; Salas-Salvado´ et al., 2000; Toth and Voll, 2001; Koffman et al., 2006; Singh and Kumar, 2007). It may develop anywhere between 2 and 78 weeks following surgery, although 4–12 weeks

626 R.F. PFEIFFER postoperatively is the most frequent timeframe (Singh symptoms may also be evident (Serdaru et al., 1988; and Kumar, 2007). In a recent review, Aasheim (2008) Weathers and Lewis, 2009). catalogued 84 cases of WE following bariatric surgery; Pellagra has been documented in individuals with in 95%, gastric bypass or a restrictive procedure had malabsorption due to a variety of causes. Several case been performed. The appearance of WE is more frereports describe the development of pellagra in people quent in individuals who experience repeated vomiting, with Crohn’s disease, in which both niacin deficiency presumably with decreased thiamine absorption because due to malabsorption and tryptophan wastage with of the vomiting (Cirignotta et al., 2000; Aasheim, 2008). increased urinary excretion of 5-hydroxyindoleacetic However, individuals undergoing Roux-en-Y gastric acid have been suggested to occur (Pollack et al., 1982; bypass have an additional risk with regard to thiamine, Zaki and Millard, 1995; Abu-Qurshin et al., 1997). Pellasince thiamine is predominantly absorbed in the duodegra has been reported in an immunocompromised num, which is bypassed in the Roux-en-Y procedure patient with colitis due to cytomegalovirus (Lu et al., (Escalona et al., 2004; Al-Fahad et al., 2006; Iannelli 2001). Small intestinal bacterial overgrowth with conseet al., 2010). quent malabsorption and development of pellagra has We has also been described in individuals with other also been described (Wierzbicka et al., 2005). Malabsorpcauses for malabsorption. In one woman with a history tion secondary to amyloidosis in an individual with mulof premature birth and neonatal necrotizing enterocolitis tiple myeloma is yet another reported cause of pellagra with subsequent bowel resection, WE developed during (Itami et al., 1997). pregnancy and was attributed to longstanding chronic malabsorption exacerbated by her pregnancy (Williams Copper deficiency myelopathy et al., 2009). Another individual with colon cancer and an enterocutaneous fistula developed WE following Copper is an essential trace metal and micronutrient that administration of 5-fluorouracil; the role of malabsorpis important for many biological functions (Stern, 2010; de Roman˜a et al., 2011). It is incorporated into at least 30 tion in this patient is uncertain (Papila et al., 2010). Either metalloenzymes and involved with catecholamine synmalabsorption or consumption by the tumor was considered to be responsible for the development of WE in a thesis, brain peptide synthesis, oxidative defenses, and terminally ill cancer patient who was maintaining a reanumerous other metabolic processes (Tapiero et al., sonable caloric intake (Yae et al., 2005). WE has also 2003; Zara et al., 2009). Neurologists are most familiar been reported in the setting of pancreatic encephalopawith the damage that can be caused by excessive copper, thy, but in these cases prolonged fasting and inadequate as in Wilson’s disease, but copper deficiency also prothiamine supplementation were deemed responsible duces neurologic dysfunction. This is, perhaps, best recognized in Menkes disease, in which there is a genetically (Zhang and Tian, 2007). based inability to transport copper across the intestinal barrier due to a mutation in the ATP7A gene (de Bie Pellagra et al., 2007; T€ umer and Mller, 2010; Kodama et al., Pellagra is a disease that is often considered to have died 2011). However, impairment of intestinal copper absorpout in the US and other developed countries; however, tion may also occur in the setting of other malabsorptive although rare, it still occurs (Ishii and Nishihara, 1981; processes. Weathers and Lewis, 2009). Pellagra is due to niacin defiCopper, along with zinc, is absorbed in the proximal ciency, although it can also develop in the setting of defismall intestine, primarily in the duodenum but also to a ciency of the essential amino acid tryptophan, which is a lesser extent in the stomach and more distal small intesprecursor of niacin (Lanska, 2010). As with WE, pellagra tine (Mason, 1979; Tan et al., 2006). Processes that is most often diagnosed in individuals with chronic alcoremove these sites or otherwise impair absorption from holism and inadequate nutritional intake, but it can also them result in eventual copper deficiency. However, it is develop in other conditions, including malabsorption only within the past decade that neurologic dysfunction syndromes. as a consequence of copper deficiency due to copper The classic clinical features of pellagra consist of the malabsorption has been identified in individuals who triad of dermatitis, diarrhea, and dementia. All three are had previously undergone gastric and/or intestinal surnot present in every individual; in one study the entire gery (Schleper and Stuerenburg, 2001; Kumar et al., triad was present in only 22% (Spivak and Jackson, 2003b; Kumar, 2006; Tan et al., 2006; Bellance et al., 1977). In addition to dementia, neurologic abnormalities 2010; Jaiser and Winston, 2010; Pineles et al., 2010). that have been described in pellagra include headache, Schleper and Stuerenburg (2001) described a 46-yearvertigo, myoclonus, tremor, rigidity, weakness, dysphaold woman who developed progressive spastic tetrapargia, seizures, and still others; a variety of psychiatric esis, sensory impairment, and sensory gait ataxia and

NEUROLOGIC MANIFESTATIONS OF MALABSORPTION SYNDROMES 627 was subsequently diagnosed with copper deficiencyNeuroradiologic changes, most typically in the form of associated myelopathy. She had undergone partial gasincreased T2 signal activity within the dorsal trectomy approximately 20 years earlier for treatment columns in the cervical cord, are often present in of gastric ulcers and a second gastric resection proceboth CDM and subacute combined degeneration dure along with resection of the transverse colon approx(Kumar et al., 2006). imately 5 years earlier because of complications from The response to copper replacement therapy in CDM the original procedure. Serum copper and ceruloplasmin is inconsistent. Although the hematologic abnormalities levels were markedly diminished; cerebrospinal fluid typically respond promptly, neurologic dysfunction does copper was also diminished, excluding a diagnosis of not always do so. Progression of dysfunction is often Wilson’s disease (24 hour urinary copper was not halted, but resolution of neurologic dysfunction is often reported). Kumar et al. (2003b) subsequently described incomplete (Kumar, 2006). two individuals, again both women, who also developed progressive myelopathy and sensory gait ataxia followWhipple’s disease ing gastrointestinal surgical procedures. The first woman had undergone an intestinal bypass procedure Whipple’s disease (WhD) is an example of a disease process in which, although characterized by both gastrointesfor obesity 24 years previously; the second woman had tinal malabsorption and neurologic dysfunction, the undergone two procedures, partial small bowel resection for Crohn’s disease 30 years previously and partial gasneurologic dysfunction is not the result of malabsorption trectomy and vagotomy for refractory ulcers 15 years but rather due to central nervous system involvement of previously. In both individuals, serum copper and ceruthe primary disease process itself. Nevertheless, it will be loplasmin levels were markedly reduced and 24 hour uribriefly detailed here because of the presence of both malnary copper excretion was normal. Kumar (2006) absorption and neurologic dysfunction. subsequently reviewed the case records of 25 persons Although originally described as a gastrointestinal disease, it has become abundantly clear that WhD is a multiwho were diagnosed with copper deficiency myelopathy system disorder that may also demonstrate joint, (CDM); 10 of them had a history of prior gastric surgery. More recently, Jaiser and Winston (2010) reviewed 55 dermatologic, lymphatic, cardiac, pulmonary, ocular, case reports of copper deficiency myelopathy collected and neurologic dysfunction (Dutly and Altwegg, 2001). from the literature and confirmed prior upper gastroinThus, in addition to diarrhea, weight loss, and abdominal testinal surgery as an important (though not the only) pain, individuals with WhD may display migratory polyrisk factor. They noted a striking female predominance arthritis, generalized lymphadenopathy, anemia, fever, (F:M ¼ 3.6:1) and attributed the copper deficiency to generalized malaise, chronic cough, pseudo-addisonian skin pigmentation, congestive heart failure, hypotension, impaired absorption in the upper gastrointestinal tract. pericardial friction rub, splenomegaly, focal glomerulitis, Although prior gastric or intestinal surgery may be the most frequent cause of CDM, it has also been visual changes, uveitis, retinitis, and a variety of neuroreported in individuals with excessive zinc ingestion logic manifestations (Weiner and Utsinger, 1986; Dutly (Kumar et al., 2003a) and in individuals with other reaand Altwegg, 2001; Ojeda et al., 2010). sons for malabsorption, such as celiac disease (Kumar The average age of symptom onset in WhD is approxand Low, 2004; Kumar, 2006; Jung and Marziniak, imately 50 years. Males are affected much more fre2008; Jaiser and Winston, 2010). quently than females; in the past the male-to-female ratio was 8:1 but in recent years this may have dropped The clinical features of CDM closely mimic those of to 4–5:1 (Dutly and Altwegg, 2001). Farmers have an subacute combined degeneration due to vitamin B12 deficiency (Kumar et al., 2004). The combination of posincreased risk for developing WhD (Weiner and terior column dysfunction with sensory ataxia and assoUtsinger, 1986). The organism responsible for WhD, ciated corticospinal tract dysfunction are common to Tropheryma whipplei, has been identified and characterboth; peripheral neuropathy may also be present in both, ized as a member of the actinomycete family; it has been although it is not a predominant feature of CDM. Hemasuggested that Tropheryma whipplei may be a soiltologic manifestations are frequently, though not invaridwelling organism, which might explain the increased ably, present in both; anemia and neutropenia are incidence of infection in farmers (Dutly and Altwegg, characteristic in CDM and the anemia may be micro2001). It has also been found in the influxes to sewage cytic, macrocytic, or normocytic (Kumar, 2006). plants, particularly those from agricultural communities Although myelopathy, often with associated peripheral (Sch€oniger-Hekele et al., 2007; Schneider et al., 2008). neuropathy, is the most frequent clinical presentation Neurologic dysfunction may be the presenting feaof CDM, optic neuropathy may also be part of the clinture in approximately 5% of persons with WhD (Peters ical picture (Spinazzi et al., 2007; Pineles et al., 2010). et al., 2002). Clinical central nervous system involvement

628

R.F. PFEIFFER

eventually develops in 10–43% of patients with WhD; postmortem examinations demonstrate central nervous system lesions in over 90% of both symptomatic and asymptomatic individuals (Dutly and Altwegg, 2001; Peters et al., 2002). Cognitive changes appear in 71% of individuals; and may be accompanied by psychiatric symptoms such as depression and personality or behavioral changes (Louis et al., 1996; Franca et al., 2004). Insomnia, hypersomnia, hyperphagia, polyuria, and polydipsia are uncommon, but do occur (Louis et al., 1996; Perkin and Murray-Lyon, 1998; Dutly and Altwegg, 2001). Cerebellar dysfunction with gait and balance impairment and pyramidal tract abnormalities may also develop (Louis et al., 1996; Franca et al., 2004). A variety of ocular and extraocular abnormalities, such as vertical gaze impairment, extraocular muscle dysfunction, internuclear ophthalmoplegia, ptosis, and pupillary abnormalities may also occur (Chan et al., 2001; Franca et al., 2004). Oculomasticatory myorhythmia, consisting of the combination of pendular convergence nystagmus and concurrent slow, rhythmic synchronous contractions of the masticatory muscles, invariably accompanied by a supranuclear vertical gaze paresis, develops in approximately 20% of individuals with central nervous system involvement (Schwartz et al., 1986; Louis et al., 1996). Peripheral neuropathy is a neurologic feature of WhD that actually may be the direct result of nutritional deficiency due to malabsorption (Topper et al., 2002; Franca et al., 2004). PCR analysis appears to be a more sensitive method of diagnosis than identification of PAS-positive inclusions in macrophages present in duodenal biopsy specimens, but there is some evidence that Tropheryma whipplei DNA may be present in healthy individuals without WhD (Dutly and Altwegg, 2001; Peters et al., 2002). In individuals with central nervous system symptomatology, brain biopsy is positive over 80% of the time; cerebrospinal fluid analysis, including PCR, may also be useful (Louis et al., 1996; Perkin and MurrayLyon, 1998; Schijf et al., 2008). Prompt diagnosis of WhD is important because effective treatment is available. An initial 2 week course of parenteral therapy with either a combination of penicillin G and streptomycin or with a third generation cephalosporin (e.g., ceftriaxone), followed by a 1 year course of oral trimethoprim-sulfamethoxazole has been recommended as an effective treatment approach (Dutly and Altwegg, 2001). The prolonged course of trimethoprim-sulfamethoxazole is considered by some to be important to prevent central nervous system relapses, which have a poor prognosis and high mortality rate, but not all investigators agree that this is the optimum treatment approach and instead recommend a combination of doxycycline and hydroxychloroquine,

supplemented by sulfadiazine in patients with neurologic involvement (Lagier et al., 2010).

CONCLUSION Gastrointestinal diseases resulting in malabsorption and consequent nutritional deficiencies can be accompanied by neurologic dysfunction that can manifest itself in a broad and confusing array of symptoms and signs. It is important for internists and gastroenterologists to be aware of this possibility and equally important for neurologists to think about the possibility of underlying nutritional deficiency due to malabsorption when evaluating patients for neurologic dysfunction. Prompt and appropriate testing can lead to diagnosis and treatment, which may, at a minimum, forestall further neurologic progression and, in the best of circumstances, result in complete neurologic recovery.

REFERENCES Aasheim ET (2008). Wernicke encephalopathy after bariatric surgery: a systematic review. Ann Surg 248: 714–720. Abu-Qurshin R, Naschitz JE, Zuckermann E et al. (1997). Crohn’s disease associated with pellagra and increased excretion of 5-hydroxyindolacetic acid. Am J Med Sci 313: 111–113. Al-Fahad T, Ismael A, Soliman MO et al. (2006). Very early onset of Wernicke’s encephalopathy after gastric bypass. Obes Surg 16: 671–672. Angelini L, Erba A, Mariotti C et al. (2002). Myoclonic dystonia as unique presentation of isolated vitamin E deficiency in a young patient. Mov Disord 17: 612–614. Aslam A, Misbah SA, Talbot K et al. (2004). Vitamin E deficiency induced neurological disease in common variable immunodeficiency: two cases and a review of the literature of vitamin E deficiency. Clin Immunol 112: 24–29. Ayuso Blanco T, Martin Martinez J, Figueras P et al. (1994). Chronic polyneuropathy due to vitamin E deficiency. Neurologia 9: 300–302 (Article in Spanish). Barbato M, Curione M, Amato S et al. (2010). Autonomic imbalance in celiac disease. Minerva Pediatr 62: 333–338. Bassen FA, Kornzweig AL (1950). Malformation of the erythrocytes in a case of atypical retinitis pigmentosa. Blood 5: 381–387. Batheja MJ, Leighton J, Azueta A et al. (2010). The face of tropical sprue in 2010. Case Rep Gastroenterol 4: 168–172. Bellance R, Edimo Nana M, Kone M et al. (2010). Sensory ataxic neuromyelopathy in acquired copper deficiency. Rev Neurol (Paris) 166: 734–736 (Article in French). Ben Hamida C, Doerflinger N, Belal S et al. (1993). Localization of Friedreich ataxia phenotype with selective vitamin E deficiency to chromosome 8q by homozygosity mapping. Nat Genet 5: 195–200. Borel P, Lairon D, Senft M et al. (1989). Wheat bran and wheat germ: effect on digestion and intestinal absorption of dietary lipids in the rat. Am J Clin Nutr 49: 1192–1202.

NEUROLOGIC MANIFESTATIONS OF MALABSORPTION SYNDROMES Borel P, Pasquier B, Armand M et al. (2001). Processing of vitamin A and E in the human gastrointestinal tract. Am J Physiol Gastrointest Liver Physiol 280: G95–G103. Braegger CP, Belli DC, Mentha G et al. (1998). Persistence of the intestinal defect in abetalipoproteinemia after liver transplantation. Eur J Pediatr 157: 576–578. Briani C, Zara G, Toffanin E et al. (2005). Neurological complications of celiac disease and autoimmune mechanisms: preliminary data of a prospective study in adult patients. Ann N Y Acad Sci 1051: 148–155. B€ urk K, Farecki ML, Lamprecht G et al. (2009). Neurological symptoms in patients with biopsy proven celiac disease. Mov Disord 24: 2358–2362. Bushara KO (2005). Neurologic presentation of celiac disease. Gastroenterology 128 (4 Suppl 1): S92–S97. Bye AM, Muller DP, Wilson J et al. (1985). Symptomatic vitamin E deficiency in cystic fibrosis. Arch Dis Child 60: 162–164. Chan RY, Yannuzzi LA, Foster CS (2001). Ocular Whipple’s disease: earlier definitive diagnosis. Ophthalmology 108: 2225–2231. Chin RL, Sander HW, Brannagan TH et al. (2003). Celiac neuropathy. Neurology 60: 1581–1585. Cicarelli G, Della Rocca G, Amboni M et al. (2003). Clinical and neurological abnormalities in adult celiac disease. Neurol Sci 24: 311–317. Cirignotta F, Manconi M, Mondini S et al. (2000). Wernicke– Korsakoff encephalopathy and polyneuropathy after gastroplasty for morbid obesity: report of a case. Arch Neurol 57: 1356–1359. Cooke WT, Smith WT (1966). Neurological disorders associated with adult coeliac disease. Brain 89: 683–722. de Bie P, Muller P, Wijmenga C et al. (2007). Molecular pathogenesis of Wilson and Menkes disease: correlation of mutations with molecular defects and disease phenotypes. J Med Genet 44: 673–688. de Roman˜a DL, Olivares M, Uauy R et al. (2011). Risks and benefits of copper in light of new insights of copper homeostasis. J Trace Elem Med Biol 25: 3–13. Di Donato I, Bianchi S, Federico A (2010). Ataxia with vitamin E deficiency: update of molecular diagnosis. Neurol Sci 31: 511–515. Dutly F, Altwegg M (2001). Whipple’s disease and Tropheryma whippelii. Clin Microbiol Rev 14: 561–583. Escalona A, Pe´rez G, Leo´n F et al. (2004). Wernicke’s encephalopathy after Roux-en-Y gastric bypass. Obes Surg 14: 1135–1137. Farrell JJ (2002). Digestion and absorption of nutrients and vitamins. In: M Feldman, LS Friedman, MH Sleisenger (Eds.), Gastrointestinal and Liver Disease, 7th edn. Saunders, Philadelphia, pp. 1715–1750. Fogel BL, Perlman S (2007). Clinical features and molecular genetics of autosomal recessive cerebellar ataxias. Lancet Neurol 6: 245–257. Franca MC Jr, de Castro R, Balthazar MLF et al. (2004). Whipple’s disease with neurological manifestations. Arq Neuropsiquiatr 62: 342–346. Gauthier S, Sniderman A (1983). Action tremor as a manifestation of chylomicron retention disease. Ann Neurol 14: 591. Georges A, Bonneau J, Bonnefont-Rousselot D et al. (2011). Molecular analysis and intestinal expression of SAR1 genes

629

and proteins in Anderson’s disease (chylomicron retention disease). Orphanet J Rare Dis 6: 1. Ghalaut VS, Ghalaut PS, Kharb S et al. (1995). Vitamin E in intestinal fat malabsorption. Ann Nutr Metab 39: 296–301. Ghosh D, Dhiman RK, Kohli A et al. (1994). Hypokalemic periodic paralysis in association with tropical sprue: a case report. Acta Neurol Scand 90: 371–373. Glynn J (1986). Tropical sprue – its aetiology and pathogenesis. J R Soc Med 79: 599–606. Grant CA, Berson EL (2001). Treatable forms of retinitis pigmentosa associated with systemic neurological disorders. Int Ophthalmol Clin 41: 103–110. Green PHR, Cellier C (2007). Celiac disease. N Engl J Med 357: 1731–1743. Gupta V, Kohli A (2010). Celiac disease associated with recurrent Guillain Barre syndrome. Indian Pediatr 47: 797–798. Hall DA, Parsons J, Benke T (2007). Paroxysmal nonkinesigenic dystonia and celiac disease. Mov Disord 22: 708–710. Hammond N, Wang Y (2008). Fat soluble vitamins. In: J Biller (Ed.), The Interface of Neurology and Internal Medicine. Lippincott Williams and Wilkins, Philadelphia, pp. 449–452. Harper CG, Giles M, Finlay-Jones R (1986). Clinical signs in the Wernicke–Korsakoff complex: a retrospective analysis of 131 cases diagnosed at necropsy. J Neurol Neurosurg Psychiatry 49: 341–345. H€ ogenauer C, Hammer HF (2002). Maldigestion and malabsorption. In: M Feldman, LS Friedman, MH Sleisenger (Eds.), Gastrointestinal and Liver Disease, 7th edn. Saunders, Philadelphia, pp. 1751–1782. Howard L, Ovesen L, Satya-Murti S et al. (1982). Reversible neurological symptoms caused by vitamin E deficiency in a patient with short bowel syndrome. Am J Clin Nutr 36: 1243–1249. Iannaccone ST, Sokol RJ (1986). Vitamin E deficiency in neuropathy of abetalipoproteinemia. Neurology 36: 1009. Iannelli A, Addeo P, Novellas S et al. (2010). Wernicke’s encephalopathy after laparoscopic Roux-en-Y gastric bypass: a misdiagnosed complication. Obes Surg 20: 1594–1596. Insel P, Ross D, McMahon K et al. (2010). Nutrition. Jones and Bartlett, Burlington, VA, pp. 121–124. Ishii N, Nishihara Y (1981). Pellagra among chronic alcoholics: clinical and pathological study of 20 necropsy cases. J Neurol Neurosurg Psychiatry 44: 209–215. Itami A, Ando I, Kukita A et al. (1997). Pellagra associated with amyloidosis secondary to multiple myeloma. Br J Dermatol 137: 829. Iyer GV, Taori GM, Kapadia CR et al. (1973). Neurologic manifestations in tropical sprue A clinical and electrodiagnostic study. Neurology 23: 959–966. Jacob S, Zarei M, Kenton A et al. (2005). Gluten sensitivity and neuromyelitis optica: two case reports. J Neurol Neurosurg Psychiatry 76: 1028–1030. Jaiser SR, Winston GP (2010). Copper deficiency myelopathy. J Neurol 257: 869–881. Jung A, Marziniak M (2008). Copper deficiency as a treatable cause of myelopathy. Nervenarzt 79: 421–425, (Article in German).

630

R.F. PFEIFFER

Ko HY, Park-Ko I (1999). Electrophysiologic recovery after vitamin E-deficient neuropathy. Arch Phys Med Rehabil 80: 964–967. Kodama H, Fujusawa C, Bhadhprasit W (2011). Pathology, clinical features and treatments of congenital copper metabolic disorders – focus on neurologic aspects. Brain Dev 33: 243–251. Koffman BM, Greenfield LJ, Ali II et al. (2006). Neurologic complications after surgery for obesity. Muscle Nerve 33: 166–176. Kumar N (2006). Copper deficiency myelopathy (human swayback). Mayo Clin Proc 81: 1371–1384. Kumar N, Low PA (2004). Myeloneuropathy and anemia due to copper malabsorption. J Neurol 251: 747–749. Kumar N, Gross JB Jr, Ahlskog JE (2003a). Myelopathy due to copper deficiency. Neurology 61: 273–274. Kumar N, McEvoy KM, Ahlskog JE (2003b). Myelopathy due to copper deficiency following gastrointestinal surgery. Arch Neurol 60: 1782–1785. Kumar N, Gross JB Jr, Ahlskog JE (2004). Copper deficiency myelopathy produces a clinical picture like subacute combined degeneration. Neurology 63: 33–39. Kumar N, Ahlskog JE, Klein CJ et al. (2006). Imaging features of copper deficiency myelopathy: a study of 25 cases. Neuroradiology 48: 78–83. Kumar S, Ghoshal UC, Jayalakshmi K et al. (2011). Abnormal small intestinal permeability in patients with idiopathic malabsorption in tropics (tropical sprue) does not change even after successful treatment. Dig Dis Sci 56: 161–169. Lagerqvist C, Ivarsson A, Juto P et al. (2001). Screening for adult coeliac disease – which serological marker(s) to use? J Intern Med 250: 241–248. Lagier J-C, Fenollar F, Lepidi H et al. (2010). Failure and relapse after treatment with trimethoprim/sulfamethoxazole in classic Whipple’s disease. J Antimicrob Chemother 65: 2005–2012. Lanska DJ (2010). Chapter 30: Historical aspects of the major neurological vitamin deficiency disorders: the watersoluble B vitamins. Handb Clin Neurol 95: 445–476. Laplante P, Vanasse M, Michaud J et al. (1984). A progressive neurological syndrome associated with an isolated vitamin E deficiency. Can J Neurol Sci 11 (4 Suppl): 561–564. Linton MF, Farese RV Jr, Young SG (1993). Familial hypobetalipoproteinemia. J Lipid Res 34: 521–541. Louis ED, Lynch T, Kaufmann P et al. (1996). Diagnostic guidelines in central nervous system Whipple’s disease. Ann Neurol 40: 561–568. Lu JY, Yu CL, Wu MZ (2001). Pellagra in an immunocompetent patient with cytomegalovirus colitis. Am J Gastroenterol 96: 932–934. Malamut G, Verkarre V, Suarez F et al. (2010). The enteropathy associated with common variable immunodeficiency: the delineated frontiers with celiac disease. Am J Gastroenterol 105: 2262–2275. Maldonado-Valderrama J, Wilde P, Macierzanka A et al. (2011). The role of bile salts in digestion. Adv Colloid Interface Sci 165: 36–46.

Manchanda S, Davies CR, Picchietti D (2009). Celiac disease as a possible cause for low serum ferritin in patients with restless legs syndrome. Sleep Med 10: 763–765. Mason KE (1979). A conspectus of research on copper metabolism and requirements of man. J Nutr 109: 1979–2066. Meyer JH (1980). Gastric emptying of ordinary food: effect of antrum on particle size. Am J Physiol 239: G133–G135. Midha V, Jain NP, Sood A et al. (2007). Landry–Guillaine– Barre´ syndrome as presentation of celiac disease. Indian J Gastroenterol 26: 42–43. Morello F, Ronzani G, Cappellari F (2003). Migraine, cortical blindness, multiple cerebral infarctions and hypocoagulopathy in celiac disease. Neurol Sci 24: 85–89. Noto D, Cefalu` AB, Cannizzaro A et al. (2009). Familial hypobetalipoproteinemia due to apolipoprotein B R463W mutation causes intestinal fat accumulation and low postprandial lipemia. Atherosclerosis 206: 193–198. Ojeda E, Cosme A, Lapaza J et al. (2010). Whipple’s disease in Spain: a clinical review of 91 patients diagnosed between 1947 and 2001. Rev Esp Enferm Dig 102: 108–123. Ouahchi K, Arita M, Kayden H et al. (1995). Ataxia with isolated vitamin E deficiency is caused by mutations in the alpha-tocopherol transfer protein. Nat Genet 9: 141–145. Owens SR, Greenson JK (2007). The pathology of malabsorption: current concepts. Histopathology 50: 64–82. Palau F, Espino´s C (2006). Autosomal recessive cerebellar ataxias. Orphanet J Rare Dis 1: 47. Papila B, Yildiz O, Tural D et al. (2010). Wernicke’s encephalopathy in colon cancer. Case Rep Oncol 3: 362–367. Pellecchia MT, Scala R, Filla A et al. (1999). Idiopathic cerebellar ataxia associated with celiac disease: lack of distinctive neurological features. J Neurol Neurosurg Psychiatry 66: 32–35. Pereira AC, Edwards MJ, Buttery PC et al. (2004). Choreic syndrome and coeliac disease: a hitherto unrecognised association. Mov Disord 19: 478–482. Peretti N, Roy CC, Sassolas A et al. (2009). Chylomicron retention disease: a long term study of two cohorts. Mol Genet Metab 97: 136–142. Peretti N, Sassolas A, Roy CC et al. (2010). Guidelines for the diagnosis and management of chylomicron retention disease based on a review of the literature and the experiences of two centers. Orphanet J Rare Dis 5: 24. Perkin GD, Murray-Lyon I (1998). Neurology and the gastrointestinal system. J Neurol Neurosurg Psychiatry 65: 291–300. Peters G, du Plessis DG, Humphrey PR (2002). Cerebral Whipple’s disease with a stroke-like presentation and cerebrovascular pathology. J Neurol Neurosurg Psychiatry 73: 336–339. Pineles SL, Wilson CA, Balcer LJ et al. (2010). Combined optic neuropathy and myelopathy secondary to copper deficiency. Surv Ophthalmol 55: 386–392. Pollack S, Enat R, Haim S et al. (1982). Pellagra as the presenting manifestation of Crohn’s disease. Gastroenterology 82: 948–952.

NEUROLOGIC MANIFESTATIONS OF MALABSORPTION SYNDROMES Pot GK, Prynne CJ, Roberts C et al. (2012). National Diet and Nutrition Survey: fat and fatty acid intake from the first year of the rolling programme and comparison with previous surveys. Br J Nutr 107: 405–415. Puri V, Chaudhry N, Tatke M et al. (2005). Isolated vitamin E deficiency with demyelinating neuropathy. Muscle Nerve 32: 230–235. Ramakrishna BS, Venkataraman S, Mukhopadhya A (2006). Tropical malabsorption. Postgrad Med J 82: 779–787. Ramirez I, Santini R, Corcino J et al. (1973). Serum vitamin E levels in children and adults with tropical sprue in Puerto Rico. Am J Clin Nutr 26: 1045. Ranjan P, Ghoshal UC, Aggarwal R et al. (2004). Etiological spectrum of sporadic malabsorption syndrome in northern Indian adults at a tertiary hospital. Indian J Gastroenterol 23: 94–98. Rino Y, Suzuki Y, Kuroiwa Y et al. (2007). Vitamin E malabsorption and neurological consequences after gastrectomy for gastric cancer. Hepatogastroenterology 54: 1858–1861. Ros E (2000). Intestinal absorption of triglyceride and cholesterol Dietary and pharmacological inhibition to reduce cardiovascular risk. Atherosclerosis 151: 357–379. Rosenberg NR, Vermeulen M (2005). Should coeliac disease be considered in the work up of patients with chronic peripheral neuropathy? J Neurol Neurosurg Psychiatry 76: 1415–1419. Rothrock JF, Smith MS (1981). Wernicke’s disease complicating surgical therapy for morbid obesity. J Clin Neuroophthalmol 1: 195–199. Said HM (2011). Intestinal absorption of water-soluble vitamins in health and disease. Biochem J 437: 357–372. Salas-Salvado´ J, Garcia-Lorda P, Cuatrecasas G et al. (2000). Wernicke’s syndrome after bariatric surgery. Clin Nutr 19: 371–373. Schijf LJ, Becx MC, de Bruin PC et al. (2008). Whipple’s disease: easily diagnosed, if considered. Neth J Med 66: 392–395. Schleper B, Stuerenburg HJ (2001). Copper deficiencyassociated myelopathy in a 46-year-old woman. J Neurol 248: 705–706. Schneider T, Moos V, Loddenkemper C et al. (2008). Whipple’s disease: new aspects of pathogenesis and treatment. Lancet Infect Dis 8: 179–190. Sch€ oniger-Hekele M, Petermann D, Weber B et al. (2007). Tropheryma whipplei in the environment: survey of sewage plant influxes and sewage plant workers. Appl Environ Microbiol 73: 2033–2035. Schwartz MA, Selhorst JB, Ochs AL et al. (1986). Oculomasticatory myorhythmia: a unique movement disorder occurring in Whipple’s disease. Ann Neurol 20: 677–683. Seehra H, MacDermott N, Lascelles RG et al. (1996). Wernicke’s encephalopathy after vertical banded gastroplasty for morbid obesity. BMJ 312: 434. Serdaru M, Hausser-Hauw C, Laplane D et al. (1988). The clinical spectrum of alcoholic pellagra encephalopathy: a retrospective analysis of 22 cases studied pathologically. Brain 111: 829–842.

631

Shoulders CC, Brett DJ, Bayliss JD et al. (1993). Abetalipoproteinemia is caused by defects of the gene encoding the 97kDa subunit of a microsomal triglyceride transfer protein. Hum Mol Genet 2: 2109–2116. Shoulders CC, Stephens DJ, Jones B (2004). The intracellular transport of chylomicrons requires the small GTPase, Sar1b. Curr Opin Lipidol 15: 191–197. Singh S, Kumar A (2007). Wernicke encephalopathy after obesity surgery: a systematic review. Neurology 68: 807–811. Soejima N, Ohyagi Y, Kikuchi H et al. (2006). An adult case of probable Bassen–Kornzweig syndrome, presenting resting tremor. Rinsho Shinkeigaku 46: 702–706, (Article in Japanese). Sokol RJ, Kim YS, Hoofnagle JH et al. (1989). Intestinal malabsorption of vitamin E in primary biliary cirrhosis. Gastroenterology 96: 479–486. Spinazzi M, De Lazzari F, Tavolato B et al. (2007). Myelooptico-neuropathy in copper deficiency occurring after partial gastrectomy Do small bowel bacterial overgrowth syndrome and occult zinc ingestion tip the balance? J Neurol 254: 1012–1017. Spivak JL, Jackson DL (1977). Pellagra: an analysis of 18 patients and a review of the literature. Johns Hopkins Med J 140: 295–309. Stern BR (2010). Essentiality and toxicity in copper health risk assessment: overview, update and regulatory considerations. J Toxicol Environ Health A 73: 114–127. Stevenson VL, Hardie RJ (2001). Acanthocytosis and neurological disorders. J Neurol 248: 87–94. Sturman RM (1968). The Bassen–Kornzweig syndrome: 18 years in evolution. J Mt Sinai Hosp N Y 35: 489–517. Tan JC, Burns DL, Jones HR (2006). Severe ataxia, myelopathy, and peripheral neuropathy due to acquired copper deficiency in a patient with history of gastrectomy. J Parenter Enteral Nutr 30: 446–450. Tapiero H, Townsend DM, Tew KD (2003). Trace elements in human physiology and pathology Copper. Biomed Pharmacother 57: 386–398. Tarugi P, Averna M, Di Leo E et al. (2007). Molecular diagnosis of hypobetalipoproteinemia: an ENID review. Atherosclerosis 195: e19–e27. Thakur B, Mishra P, Desai N et al. (2006). Profile of chronic small-bowel diarrhea in adults in western India: a hospitalbased study. Trop Gastroenterol 27: 84–86. Thomson AB, Schoeller C, Keelan M et al. (1993). Lipid absorption: passing through the unstirred layers, brushborder membrane, and beyond. Can J Physiol Pharmacol 71: 531–555. Tjon JM, van Bergen J, Koning F (2010). Celiac disease: how complicated can it get? Immunogenetics 62: 641–651. Topper R, Gartung C, Block F (2002). Neurologic complications in inflammatory bowel diseases. Nervenarzt 73: 489–499, (Article in German). Toth C, Voll C (2001). Wernicke’s encephalopathy following gastroplasty for morbid obesity. Can J Neurol Sci 28: 89–92. T€ umer Z, Mller LB (2010). Menkes disease. Eur J Hum Genet 18: 511–518.

632

R.F. PFEIFFER

Ueda N, Suzuki Y, Rino Y et al. (2009). Correlation between neurological dysfunction with vitamin E deficiency and gastrectomy. J Neurol Sci 287: 216–220. Uruha A, Shimizu T, Katoh T et al. (2011). Wernicke’s encephalopathy in a patient with peptic ulcer disease. Case Rep Med, http://dx.doi.org/10.1155/2011/156104. Uygur-Bayramicli O, Ozel AM (2011). Celiac disease is associated with neurological syndromes. Dig Dis Sci 56: 1587–1588. Vaknin A, Eliakim R, Ackerman Z et al. (2004). Neurological abnormalities associated with celiac disease. J Neurol 251: 1393–1397. Van Deest BW, Fordtran JS, Morawski SG et al. (1968). Bile salt and micellar fat concentration in proximal small bowel contents of ileectomy patients. J Clin Invest 47: 1314–1324. Vilppula A, Collin P, Ma¨ki M et al. (2008). Undetected celiac disease in the elderly: a biopsy-proven population-based study. Dig Liver Dis 40: 809–813. Vorgerd M, Tegenthoff M, K€ uhne D et al. (1996). Spinal MRI in progressive myeloneuropathy associated with vitamin E deficiency. Neuroradiology 38 (Suppl 1): S111–S113. Weathers AL, Lewis SL (2009). Rare and unusual . . . or are they? Less commonly diagnosed encephalopathies associated with systemic disease. Semin Neurol 29: 136–153. Weiner SR, Utsinger P (1986). Whipple disease. Semin Arthritis Rheum 15: 157–167. Weinstock LB, Walters AS, Mullin GE et al. (2010). Celiac disease is associated with restless legs syndrome. Dig Dis Sci 55: 1667–1673. Whitfield AJ, Marais AD, Robertson K et al. (2003). Four novel mutations in APOB causing heterozygous and homozygous familial hypobetalipoproteinemia. Hum Mutat 22: 178. Whitton C, Nicholson SK, Roberts G et al. (2011). National Diet and Nutrition Survey: UK food consumption and nutrient intakes from the first year of the rolling programme and comparisons with previous surveys. Br J Nutr 106: 1899–1914.

Wichman A, Buchthal F, Pezeshkpour GH et al. (1985). Peripheral neuropathy in abetalipoproteinemia. Neurology 35: 1279–1289. Wierzbicka E, Machet L, Karsenti D et al. (2005). Pellagra and panniculitis induced by chronic bacterial colonisation of the small intestine. Ann Dermatol Venereol 132: 140–142, (Article in French). Williams NL, Wiegand S, McKenna DS (2009). Wernicke’s encephalopathy complicating pregnancy in a woman with neonatal necrotizing enterocolitis and resultant chronic malabsorption. Am J Perinatol 26: 519–521. Willison HJ, Muller DP, Matthews S et al. (1985). A study of the relationship between neurological function and serum vitamin E concentrations in patients with cystic fibrosis. J Neurol Neurosurg Psychiatry 48: 1097–1102. Yae S, Okuno S, Onishi H et al. (2005). Development of Wernicke encephalopathy in a terminally ill cancer patient consuming an adequate diet: a case report and review of the literature. Palliat Support Care 3: 333–335. Yokota T, Tsuchiya K, Furukawa T et al. (1990). Vitamin E deficiency in acquired fat malabsorption. J Neurol 237: 103–106. Zaki I, Millard L (1995). Pellagra complicating Crohn’s disease. Postgrad Med J 71: 496–497. Zamel R, Khan R, Pollex RL et al. (2008). Abetalipoproteinemia: two case reports and literature review. Orphanet J Rare Dis 3: 19. Zara G, Grassivaro F, Brocadello F et al. (2009). Case of sensory ataxic ganglionopathy–myelopathy in copper deficiency. J Neurol Sci 277: 184–186. Zeissig S, Dougan SK, Barral DC et al. (2010). Primary deficiency of microsomal triglyceride transfer protein in human abetalipoproteinemia is associated with loss of CD1 function. J Clin Invest 120: 2889–2899. Zhang XP, Tian H (2007). Pathogenesis of pancreatic encephalopathy in severe acute pancreatitis. Hepatobiliary Pancreat Dis Int 6: 134–140.

Handbook of Clinical Neurology, Vol. 120 (3rd series) Neurologic Aspects of Systemic Disease Part II Jose Biller and Jose M. Ferro, Editors © 2014 Elsevier B.V. All rights reserved

Chapter 43

Commonly used gastrointestinal drugs ANNU AGGARWAL AND MOHIT BHATT* Center for Brain and Nervous System, Kokilaben Dhirubhai Ambani Hospital and Medical Research Institute, Mumbai, India

INTRODUCTION Commonly used gastrointestinal drugs, including antiemetics, motility modifying drugs, and drugs for acidrelated disorders (Table 43.1), are extensively prescribed in various outpatient clinics, emergency departments, and intensive care units (Karamanolis and Tack, 2006; Herbert and Holzer, 2008). While, as a group, these drugs are generally considered safe across all age groups and many are available as over-the-counter preparations (Parikh and Howden, 2010), some of these drugs can occasionally lead to serious cardiovascular and neurologic complications (Pasricha et al., 2006). Table 43.2 lists a range of neurologic complications that have been reported following use of these gastrointestinal drugs. For instance, acute neurotoxicities including transient akathisias, oculogyric crises, delirium, seizures, strokes can develop after use of certain gastrointestinal medications (described in more detail below), while disabling and pervasive tardive syndromes are described following long-term, and often unsupervised, use of phenothiazines, metoclopramide, and other drugs. In rare instances, some of the antiemetics can precipitate lifethreatening extrapyramidal reactions, neuroleptic malignant syndrome, or serotonin syndrome. At the extreme, concerns over the cardiovascular toxicity of drugs such as cisapride or tegaserod have been grave enough to lead to their withdrawal from many world markets. However, most often the symptoms of neurotoxicity are innocuous (as in akathisias and various tardive dyskinesias), not readily reported by patients or attributed to a gastrointestinal medication, and the offending drug is continued (Miller and Jankovic, 1989). In this chapter we review the mode of action of the commonly used gastrointestinal drugs as well as the spectrum and mechanism of their neurotoxicity,

(Tables 43.1 and 43.2). This information should help a clinician weigh the benefits of prescribing the particular gastrointestinal drug against the associated risk of adverse effects, and recognise symptoms of neurotoxicity if they occur.

ANTIEMETICS Nausea and vomiting are triggered by stimulation of the medullary chemoreceptor trigger zone located outside the blood–brain barrier, the medullary central pattern generator, and the limbic forebrain regions (Hesketh, 2008). Development of antiemetics has paralleled understanding of neurotransmitters and neuroreceptors responsible for emesis. Early research on antiemetics focused on dopamine 2 (D2) receptor antagonists (phenothiazines, substituted benzamides, and butyrophenones). A recent advance in antiemetic therapy has been the elucidation of the key role of serotonin and tachykinins in stimulating emesis through central and peripheral receptors and development of selective serotonin 3 (5-HT3) receptor antagonists and selective neurokinin 1 (NK1) receptor antagonists (Roila and Fatigoni, 2006; Herrstedt and Dombernowsky, 2007; Hesketh, 2008; Feyer and Jordan, 2011). Since the introduction of cisplatin, a highly emetogenic chemotherapeutic agent, in the late 1970s, the main clinical drive to develop potent antiemetics has been to help prevent or abolish chemotherapy-induced nausea and vomiting (CINV) (Herrstedt, 2008). Currently, the therapeutic usefulness of an antiemetic is classified as high or low (Hesketh, 2008) based on their ability to prevent CINV (Roila and Fatigoni, 2006; Herrstedt and Dombernowsky, 2007; Hesketh, 2008; Feyer and Jordan, 2011).

*Correspondence to: Dr. Mohit Bhatt, Center for Brain and Nervous System, Kokilaben Dhirubhai Ambani Hospital and Medical Research Institute, Four Bungalows, Andheri West, Mumbai 400053, India. Tel: þ91-986-704-0404, E-mail: [email protected]

634

A. AGGARWAL AND M. BHATT

Table 43.1

Table 43.2

Commonly used gastrointestinal drugs

Potential neurologic adverse effects associated with various commonly used gastrointestinal drugs#

Antiemetics

Promotility drugs

Laxatives

Antimotility agents Drugs for acidrelated disorders

Phenothiazines, e.g., chlorpromazine, prochlorperazine, promethazine Substituted benzamides – metoclopramide Butyrophenones – domperidone, droperidol Sertons, e.g., ondansetron, granisetron, tropisetron, dolasetron, palonosetron Neurokinin (NK1) receptor antagonists, e.g., aprepitant and fosaprepitant Substituted benzamides – metoclopramide Butyrophenones – domperidone, droperidol Cisapride Mosapride Renzapride Tegaserod Levosulpiride Stimulant laxatives, e.g., bisacodyl, sodium picosulfate Osmotic laxatives, e.g. (magnesium salts) Bismuth salts Dronobinol Selective H2 blockers, e.g., cimetidine, ranitidine, famotidine, nizatidine Proton pump inhibitors (PPI), e.g., omeprazole, esomeprazole, lansoprazole, pantoprazole, tenatoprazole, rabeprazole

Phenothiazines Antipsychotics, phenothiazines (for example, chlorpromazine, prochlorperazine, promethazine) were the first effective antiemetics. They acted by blocking central D2 receptors. Their antiemetic doses were limited by hypotension, restlessness, and sedation. At tolerable doses phenothiazines had a low therapeutic usefulness as antiemetics and extrapyramidal reactions and depression were a serious concern (Bateman et al., 1989; Weiden et al., 1987; Burke et al., 1989). Over the last three decades the availability of newer antiemetics with safer adverse effect profile (like selective serotonin 3 (5-HT3) receptor antagonists and selective neurokinin 1 (NK1) receptor antagonists) has helped curtail the use of phenothiazines as antiemetics.

Metoclopramide Metoclopramide is a benzamide derivative of procaine that was developed in 1964 to equal the antiemetic

Headache Dizziness Extrapyramidal syndromes Acute

Chronic

Mood disorders

Encephalopathy Seizures Cataplexy Syncopy Strokes Hyperthermic syndromes

Akathisias Dystonia including oculogyric crisis, oromandibular dystonia, retrocollis, opisthotonus posturing, dysphagia,* respiratory spasm,* status dystonicus* Tremor Myoclonus Drug induced parkinsonism Akathisias Tardive dyskinesias* Tardive dystonia Myoclonus Anxiety Psychosis Depression

Neuroleptic malignant syndrome* Serotonin syndrome*

#

Neurologic adverse effects associated with a particular gastrointestinal drug are detailed in the text. *Potentially life-threatening neurotoxicities.

properties of phenothiazines. Like phenothiazines, metoclopramide is a selective D2 receptor blocker, but unlike phenothiazines it has only a weak antipsychotic effect (Schulze-Delrieu, 1981). Additionally, metoclopramide has a partial 5-HT4 receptor agonist activity that enhances release of acetylcholine in the myenteric plexus and is responsible for its gastrointestinal prokinetic action. Metoclopramide is equipotent to chlorpromazine in preventing vomiting, at one-tenth of chlorpromazine doses (Harrington et al., 1983; Ganzini et al., 1993). In high doses, metoclopramide can prevent cisplatin-induced vomiting from antagonism of 5-HT3 receptors (Gralla et al., 1981; Schulze-Delrieu, 1981; Karamanolis and Tack, 2006). Metoclopramide was initially approved for use in diagnostic radiology to facilitate duodenal intubation and barium studies of upper gastrointestinal tract in patients with delayed gastric emptying. Later its use was extended to treat nausea and vomiting, diabetic gastroparesis, refractory gastroesophageal reflux and postoperative ileus. Over time, metoclopramide found application in a range of disorders including nonmigrainous headaches,

COMMONLY USED GASTROINTESTINAL DRUGS 635 Tourette’s syndrome, hiccups, neurogenic bladder, orthomarkets in 2000, following reports of its cardiotoxicity, static hypotension, anorexia nervosa, and select cases led to a surge of metoclopramide use as a prokinetic and of amenorrhea (Schulze-Delrieu, 1981; Tisdale, 1981; an increase in the incidence of metoclopramide-induced Harrington et al., 1983; Miller and Jankovic, 1989; TD (Shaffer et al., 2004; Kenney et al., 2008). Currently, Ellis et al., 1993). Currently, metoclopramide is used metoclopramide accounts for a third of all reported extensively as a prokinetic and to lesser extent as an DIMDs (Pasricha et al., 2006). antiemetic agent. TD are characterized by involuntary, repetitive moveMetoclopramide use can lead to extrapyramidal ments typically involving the oromandibular muscles symptoms (movement disorders or parkinsonism) that and axial muscles, including lip puckering, pursing develop acutely (within minutes or days) to more pervaand smacking, facial grimacing, tongue protrusion, sive disorders that develop after high doses or long-term rapid eye movements or blinking, and choreiform use. The extrapyramidal symptoms comprise acute movements of the limbs. The movements may be akathisia, dystonia, oculogyric crisis, tremor, and parkinaccompanied by tremor, dystonia, or parkinsonism. sonism, to tardive syndromes such as tardive dyskinesias, Metoclopramide-induced TD have been reported to dystonia, tremor or myoclonus, and rarely catalepsy lead to life-threatening dyspnea and dysphagia (Samie (Costall and Naylor, 1973; Miller and Jankovic, 1989; et al., 1987). High cumulative doses and long duraSethi, 2004). Mixed movement disorders may occur and tion of treatment are the major risk factors for may be accompanied by behavioral changes such as metoclopramide-induced TD. Elderly women and those restlessness, anxiety, or frank psychosis. The manufacwith a family history of DIMD and diabetes (Miller and turer’s package inserts (http://dailymed.nlm.nih.gov/daiJankovic, 1989; Sewell and Jeste, 1992; Ganzini et al., lymed) warn of extrapyramidal reactions in 1:500 1993) are vulnerable to metoclopramide-induced TD. patients treated with metoclopramide, but reviews of case In 2009, the US Food and Drug Administration (FDA) series suggest a higher figure (1–15%) (Miller and issued a black box warning for metoclopramide, advisJancovic, 1989; Parkman et al., 2004; Pasricha et al., ing that the drug use be restricted to recommended doses 2006). A prospective study of physician-reported and for not more than 12 weeks. It warned that chronic drug-induced dyskinesias-dystonia estimated the inciuse of metoclopramide therapy should be avoided in all dence of metoclopramide-induced dyskinesias-dystonia but rare cases where the benefits were believed to outto be 1/213 new prescriptions (Bateman et al., 1989). weigh the risks (http://dailymed.nlm.nih.gov/dailymed). Miller and Jankovic (1989) identified 131 patients with In a minority of patients TD can abate or resolve after drug-induced movement disorders (DIMD) from a discontinuing metoclopramide but in 71% of patients database of 3000 patients seen over 12 years, of whom or more TD are persistent despite drug withdrawal 16 (12.2%) had metoclopramide-induced DIMD. The aver(Grimes, 1981; Grimes et al., 1982a, b; Sewell and Jeste, age duration of metoclopramide use prior to DIMD onset 1992; Tarsy and Indorf, 2002). Currently there is no was 12 months (range 1 day to 4 years). Interestingly, the known treatment for TD (Samie et al., 1987; Miller drug was continued for 6 months after developing DIMD, and Jancovic, 1989; Sethi, 2004). TD developing after indicating either failure to diagnose DIMD or failure to discontinuation of long-term metoclopramide is also attribute the DIMD to metoclopramide. described (Lavy et al., 1978). Tardive dyskinesias (TD) are persistent and often Acute dystonic reactions including retrocollis, oculoirreversible involuntary movements that occur following gyric crisis, trismus, facial grimacing, dysarthria, dysphaprolonged neuroleptic therapy (Sethi, 2004). TD is the gia, and opisthotonus spasms are observed in commonest of the metoclopramide-induced movement approximately 1% of patients receiving metoclopramide disorders. In an epidemiologic study in the UK a review (Robinson, 1973). Dystonic spasms are often painful of 15.9 million metoclopramide prescriptions from 1967 and frightening. The resultant disability may vary from to 1982 identified 455 patients with TD (Bateman et al., a slight neck discomfort from cervical dystonia to 1985). Ganzini et al. (1993) examined 51 patients prepotentially life-threatening status dystonicus, dysphagia, scribed metoclopramide over a 4 month period at a respiratory distress, or respiratory arrest. Acute rhabdoveterans hospital medical outpatient clinic with an agemyolysis and myoglobinuria are known to occur (Mark and gender-matched control population for DIMD. and Newton-John, 1988; Mastaglia and Argov, 2007). The authors found relative risk of TD to be 1.67 in the The dystonic reactions commence within minutes to metoclopramide group. A retrospective analysis of 434 hours of drug administration and are often self-remitting, patients followed up for TD at a movement disorder or resolve within minutes of anticholinergic or dopamine clinic revealed that metoclopramide was responsible agonist treatment (Bhatt et al., 2004). Dystonia may for 39.4% of cases, and was the second most common be accompanied by acute parkinsonism, dyskinesia, astermedicine to induce TD following haloperidol (Kenney ixis, and myoclonus (Grimes et al., 1982b; Lu and Chu, et al., 2008). Withdrawal of cisapride from the US 1988; Miller and Jankovic, 1989).

636

A. AGGARWAL AND M. BHATT

Children, young adults, and men are susceptible to developing acute dystonia following normal recommended doses of metoclopramide (Casteels-Van Daele et al., 1970; Robinson, 1973; Reid, 1977; Grimes et al., 1982b; Ganzini et al., 1993; Sethi, 2004). Acute dystonic reactions are also reported following metoclopramide overdose or accidental injection (Sills and Glass, 1978; Kerr, 1996). Metoclopramide doses need to be reduced in patients with renal failure (Bateman and Gokal, 1980; Grimes et al., 1982b). A familial tendency towards metoclopramide-induced dystonia is described (Gatrad and Gatrad, 1979; Miller and Jankovic, 1989; Guala et al., 1992). Poorly functioning or nonfunctioning CYP2D6 alleles, which slow metoclopramide metabolism, are reported in some familial cases of metoclopramide-induced dystonia (Van der Padt et al., 2006). Re-exposure to metoclopramide can cause recurrent dystonic reactions and the drug is best avoided if an extrapyramidal reaction has occurred. Rarely, recurrent dystonic reactions such as oculogyric crisis develop despite complete withdrawal of metoclopramide (Sethi, 2004; Schneider et al., 2009). Metoclopramide-induced parkinsonism or worsening of idiopathic Parkinson disease usually develops in the first 3 months of therapy and resolves within months of discontinuation of the drug. Symptoms persisting for a year after drug withdrawal are described (Miller and Jankovic, 1989). Compared to idiopathic Parkinson disease, patients with drug-induced parkinsonism are younger and have a symmetrical tremor (Indo and Ando, 1982; Yamamoto et al., 1987; Miller and Jankovic, 1989; Bondon-Guitton et al., 2011). People at the extremes of the age spectrum, children and the elderly, are at risk of metoclopramide-induced parkinsonism (Andrejak et al., 1990; Perez-Lloret et al., 2010). Akathisia, or motor restlessness, relieved by movements such as pacing, body rocking, crossing and uncrossing of legs, foot tapping, folding and unfolding of arms, or hand rubbing, is observed in association with metoclopramide use. Acute-onset akathisia is reported following intravenous metoclopramide use. Rapid infusions are associated with earlier onset and more severe akathisia (Parlak et al., 2005). Oral metoclopramide use reaching high peak plasma concentrations (over 100 ng/dL) can also lead to akathisia (Bateman et al., 1978), lasting for days after drug cessation (Poortinga et al., 2001). Acute akathisia was observed generally within the first 3 months of metoclopramide therapy (Lang, 1988). Acute onset akathisia is often selfremitting but may be associated with considerable anxiety, feelings of impending doom, and agitation, leading to refusal of treatment or surgery, violence, and even attempted suicide (Drake and Ehrlich, 1985; Caldwell et al., 1987; Sachdev and Kruk, 1994; Chow et al., 1997;

LaGorio et al., 1998). Tardive akathisia has been observed years after metoclopramide exposure (Burke et al., 1989). High-amplitude resting and postural tremor following chronic metoclopramide use has been reported (Stacy and Jankovic, 1992; Tarsy and Indorf, 2002). Reversible palatopharyngeal tremor with parkinsonism was observed in a woman following oral metoclopramide for 3 weeks (Nampiaparampil and Oruc, 2006). Metoclopramide-induced depression can develop after a few doses or after more protracted use (Anfinson, 2002).

Domperidone Domperidone is a peripheral D2 receptor antagonist and is used as a prokinetic and antiemetic of low therapeutic efficacy. It is the preferred drug to counteract levodopainduced vomiting and constipation in patients with Parkinson disease as in recommended doses domperidone does not block central dopamine receptors (Critchley et al., 1985). There are isolated reports of domperidone-induced akathisia, parkinsonism, depression, and tardive dyskinesias (Franckx and Noel, 1984; Biasini and Alberti, 1985; Leeser and Bateman, 1985; Steinherz et al., 1986; Bondon-Guitton et al., 2011), usually in the context of high doses. Psychosis following domperidone withdrawal after chronic use is described (Roy-Desruisseaux et al., 2011). Overdosage can lead to seizures (Weaving et al., 1984).

Setrons (5-hydroxytryptamine 3 (5-HT3) receptor antagonists, serotonin 3 receptor antagonist) Setrons are antiemetics that selectively block peripheral and central 5-HT3 receptors. Ondansetron and granisetron were the first setrons marketed in 1990s, followed by introduction of tropisetron, dolasetron, and finally the second generation setron, palonosetron, in 2003. Ramosetron and azasetron are currently available only in Japan. The available setrons have high therapeutic usefulness, and can prevent cisplatin-induced nausea and vomiting. The first generation setrons are interchangeable at equivalent doses. The drugs are safe, with the commonest adverse effects being constipation, transient elevation of hepatic aminotransferases, mild headache, and lightheadedness. Extrapyramidal reactions are rare (Kovac, 2003; Feyer and Jordan, 2011). Dramatic acute extrapyramidal syndromes have been observed following intravenous ondansetron. These complex involuntary movements variably include multifocal myoclonus, jerky “seizure-like” movements, tremor, involuntary eye blinking, eye deviation, facial grimacing, tongue protrusion, oromandibular dystonia, generalized dystonia, or opisthotonus spasms (Dobrow

COMMONLY USED GASTROINTESTINAL DRUGS et al., 1991; Tolan et al., 1999; Duncan et al., 2001; Ritter et al., 2003; Sprung et al., 2003; Spiegel et al., 2005; Kumar and Hu, 2009). The movements are focal or generalized, and occasionally voluntarily suppressible for short periods of time. Associated confusion, agitation, pyramidal signs, and hemodynamic instability are described (Ritter et al., 2003). Involuntary movements have also been described following use of oral ondansetron for several days (Lee et al., 2010) or overdosage (Sprung et al., 2003). Dose reduction has been shown to prevent the extrapyramidal reaction (Sprung et al., 2003). Benzodiazepines, diphenhydramine, or procyclidine (Stonell, 1998; Ritter et al., 2003; Sprung et al., 2003; Kumar and Hu, 2009) have helped ameliorate these extrapyramidal symptoms. Ondansetron has no direct effect on dopamine receptors. It is postulated that an overlap of central serotonergic and central dopaminergic systems is responsible for the observed extrapyramidal reaction to ondansetron. This hypothesis is supported by animal studies and benefit of ondansetron in levodopa-induced psychosis and dyskinesias in patients with Parkinson’s disease (Ritter et al., 2003; Sprung et al., 2003; Kumar and Hu, 2009). In a 2 year retrospective analysis of 1521 inpatients who received ondansetron for nausea and vomiting, Singh et al. (2009) identified three patients who developed brief generalized tonic-clonic seizures after intravenous ondansetron. Clinical seizures have also been reported following intravenous ondansetron use with other epileptogenic drugs (Sargent et al., 1993; Sharma and Raina, 2001). However, in the absence of electroencephalographic evidence of seizure, some authors have suggested that the “seizure-like” movements may be involuntary movements of extrapyramidal origin (Kanarek et al., 1992; Sprung et al., 2003; Singh et al., 2009). As yet no extrapyramidal reaction has been reported following granisetron or dolasetron, though crossreactivity with ondansetron is described (Lee et al., 1993; Sorbe et al., 1994). There are isolated reports of palonosetron-induced seizures (Zambelli et al., 2009).

Neurokinin receptor antagonists Aprepitant and its prodrug fosaprepitant are nonpeptide molecules that cross the blood–brain barrier and inhibit both peripheral and central receptors of substance P (neurokinin 1 (NK1) receptors). The drugs were initially developed as potential analgesics and antidepressants and later found to have a beneficial antiemetic effect. Both aprepitant and fosaprepitant are highly efficacious antiemetics and have emerged as the first-line antiemetics for controlling CINV. They are well tolerated and are not known to have any serious neurologic

637

adverse effects (Feyer and Jordan, 2011). Aprepitant is metabolized by P450 (CYP) 3A4 and when coadministered with ifosfamide may aggravate ifosfamideinduced encephalopathy (Aapro and Walko, 2010).

DRUGS AFFECTING GASTROINTESTINAL MOTILITY Gastrointestinal motility is regulated by a complex interaction of the enteric nervous system, interstitial cells of Cajal (gastrointestinal pacemakers), smooth muscle cells (effectors of gastrointestinal motility), mucosal neuroendocrine cells, and the autonomic nervous system. Various neuroendocrine mediators including serotonin, dopamine, acetylcholine, motilin, cholecystokinin, and catecholamines help regulate gastrointestinal motility (Grundy et al., 2006; Herbert and Holzer, 2008).

Promotility drugs These accelerate gastric emptying and colonic transit and are used for treating symptoms associated with gastroparesis, functional dyspepsia, or constipation. Currently, metoclopramide is the most widely used gastrointestinal prokinetic. Development of newer prokinetics has been modeled to stimulate the prokinetic properties of metoclopramide without its extrapyramidal adverse effects. Cisapride is a serotonin 5-HT4 receptor agonist and 5-HT3 receptor antagonist that increases gastrointestinal motility by augmenting cholinergic transmission through the myenteric plexus. In 2000, cisapride was withdrawn from the North American and most European markets because of concerns about its potential to induce serious cardiac arrhythmias. Cisapride interferes with the pore-forming subunits of hERG (human Ether-a-go-go-Related Gene) K þ channels, delaying the ventricular repolarization and prolonging the QTc interval on ECG. Cisapride associated cardiotoxicity is enhanced when cisapride coadministered with drugs that inhibit the CYP3A4 enzyme and slow its metabolism (Karamanolis and Tack, 2006; Toga et al., 2007). Cisapride is available in some markets as a prokinetic for infants and young children (Raschetti et al., 2001; Vandenplas et al., 2001). Neurotoxicity from cisapride use is rare. Cisaprideinduced chorea in an 8-month-old boy (Lucena et al., 1998) and torticollis, dystonia, and myoclonus during infancy (Dieckmann et al., 1996) are described. Cisapride has been associated with persistent akathisia in a 3-yearold child from neonatal life. The movements resolved 2 months after discontinuation of the drug. The authors postulated that children are prone to neurotoxicity because of poorly developed blood–brain barrier

638

A. AGGARWAL AND M. BHATT

and CYP3A4 enzymes (Elzinga-Huttenga et al., 2006). Dystonia, orofacial dyskinesias, and aggravation of parkinsonism following cisapride use in adults is reported (Naito and Kuzuhara, 1994). Renzapride and mosapride are benzapride derivatives, partial 5-HT4 receptor agonists, and 5-HT3 receptor antagonists. They are less efficacious than cisapride but have a good cardiac safety profile (with little or no action on hERG K þ channels). Renzapride has application in constipation-dominant irritable bowel syndrome while mosapride is used for upper gastrointestinal motility disorders (Karamanolis and Tack, 2006; Toga et al., 2007). Neurotoxicity has not been described as yet. Tegaserod is a partial 5-HT4 receptor agonist and 5-HT2b receptor antagonist. Antagonism of the 5-HT2b receptor results in decreased prokinetic efficacy. Postmarketing surveys have shown an increased risk of cardiovascular events (unstable angina, myocardial infarction, and stroke) and death with tegaserod use compared to placebo, leading to tegaserod withdrawal from the US and other markets. However, tegaserod is used in some regions for chronic constipation and constipation-dominant irritable bowel syndrome in women (Pasricha, 2007; Herbert and Holzer, 2008). Levosulpiride is a benzamide derivative, a selective D2 receptor inhibitor, 5-HT4 and partial 5-HT3 receptor stimulator. It is as effective as cisapride, but more effective than metoclopramide and domperidone, in increasing gastric and small intestinal motility (Rossi and Forgione, 1995; Karamanolis and Tack, 2006). Over the past decade there has been a considerable increase in levosulpiride prescriptions, especially in Asia and Europe. In South Korea, levosulpiride prescriptions were almost double of metoclopramide prescriptions (Shin et al., 2009). Extrapyramidal reactions have been reported following levosulpiride use and may be related to its ability to cross the blood–brain barrier (Rossi and Forgione, 1995; Kim et al., 2003; Karamanolis and Tack, 2006; Baik et al., 2008). In a movement disorder clinic in South Korea, 91 of 132 patients diagnosed with DIMD between 2002 and 2008 developed DIMD secondary to levosulpiride use. A majority of patients were elderly. Parkinsonism (n ¼ 85) was the commonest DIMD followed by TD (restricted to the orolingual region) (n ¼ 9) and isolated tremor (n ¼ 3). Levosulpiride was administered for period ranging from a few days to a few weeks (up to 3 years) prior to development of DIMD. Parkinsonism was reversible in 51.9% of patients within months of drug withdrawal. Three of the nine patients with TD and all three patients with isolated tremor recovered after levosulpiride withdrawal (Shin et al., 2009).

Antimotility and antidiarrheal agents Bismuth salts have been used for dyspepsia, peptic ulcer disease, colitis, and parasitic infections for many centuries. In 1970s, there was a series of reports of a potentially fatal myoclonic encephalopathy following use of bismuth salts (Ford et al., 2008). In France, 1000 cases of encephalopathy with 72 deaths were reported within a short period leading to the withdrawal of bismuth. The encephalopathy was characterized by a subacute confusional state, visual and auditory hallucinations, generalized tremulousness, myoclonic jerks, and gait problems. The encephalopathy was associated with high serum and CSF levels of bismuth and periodic complexes on EEG (Morrow, 1973; Burns et al., 1974; Escourelle et al., 1977; Supino-Viterbo et al., 1977; Tillman et al., 1996). Autopsy showed bismuth deposits in the brain, primarily in the gray matter, perivenular lymphocytic infiltration, and intracytoplasmic lipofuscin accumulation. Bismuth withdrawal led to remission. Recently there has been a renewed interest in using bismuth salts in smaller doses and for short periods for Helicobacter pylori eradication (Tillman et al., 1996; Ford et al., 2008). Dronabinol is a nonselective cannabinoid receptor agonist and is used to delay gastric emptying. Neurologic adverse events are uncommon and include headaches, dry mouth, lightheadedness and vasovagal syncopes, and poor concentration (Herbert and Holzer, 2008).

LAXATIVES Stimulant (bisacodyl, sodium picosulfate) and osmotic (magnesium salts) laxatives are safe and free from major adverse effects but can cause electrolyte disturbances (Herbert and Holzer, 2008).

DRUGS FOR ACID-RELATED DISORDERS Selective histamine 2 (H2) blockers (cimetidine, ranitidine, famotidine, nizatidine) and proton pump inhibitors (omeprazole, esomeprazole, lansoprazole, pantoprazole, tenatoprazole, rabeprazole) are widely used to treat acid-related diseases and functional gastrointestinal disorders (Lewis, 1991; Howden and Tytgat, 1996). As a class, both H2 blockers and proton pump inhibitors are safe with very few adverse effects other than diarrhea, headache, and dizziness, even on long-term use (Parikh and Howden, 2010). In fact, pooled data analysis suggested that incidence of adverse effects following ranitidine and famotidine use was no more than seen following placebo use (Lewis, 1991; Howden and Tytgat, 1996). A concern with the use of H2 blockers and proton pump inhibitors is inhibition of cytochrome 450 and the prolongation of the half-life of drugs with low

COMMONLY USED GASTROINTESTINAL DRUGS therapeutic index (such as warfarin, phenytoin, tacrolimus, ciclosporin, theophylline, and others). However, clinically relevant drug-to-drug interactions are not observed on chronic outpatient use of H2 blockers or proton pump inhibitors and these drugs are freely available as over-the-counter medications (Parikh and Howden, 2010). Long-term proton pump inhibitor treatment can lead to hypomagnesemia-induced seizure (Cundy and Dissnayake, 2008). Cimetidine (Edmonds et al., 1979; Flind and Rowley-Jones, 1979; Sharpe and Burland, 1980; Cerra et al., 1982; Handler et al., 1982), ranitidine (Bories et al., 1980; Hughes et al., 1983; Davis, 1984; Silverstone, 1984), famotidine (Henann et al., 1988; Catalano et al., 1996; Rodgers and Brengel, 1998; Yuan et al., 2001) and nizatidine (Galynker and Tendler, 1997; Bhanji and Margolese, 2004) induced mental confusion is described in elderly inpatients with coexistent liver or renal failure, or drug overdose. The confusion may be variable, associated with seizures, and visual hallucinations, cerebellar signs, and mild extrapyramidal features are reported. Symptoms remit with dose reduction or drug withdrawal. Dystonia is reported following acute use of cimetidine (Peiris and Peckler, 2001), acute and long-term use of ranitidine (Wilson et al., 1997), and overdosage of nizatidine (Bhanji and Margolese, 2004). Ranitidine-induced acute hemiballismus and dyskinesias are described (Fouddah et al., 2001; Elzinga-Huttenga et al., 2006). There are isolated reports of cimetidine-induced parkinsonism (Leo et al., 1995), myopathy, and motor neuropathy (Feest and Read, 1980; Walls et al., 1980). Further, both H2 blockers and proton pump inhibitors can theoretically impair neuromuscular transmission (Kounenis et al., 1994).

CENTRAL HYPERTHERMIA SYNDROMES AND GASTROINTESTINAL DRUGS Antiemetic use is associated with two life-threatening hyperthermic syndromes; the neuroleptic malignant syndrome (NMS) and the serotonin syndrome. NMS is characterized by hyperthermia, muscle rigidity, fluctuating sensorium, and autonomic instability. It is caused by abrupt central dopamine blockade and has been associated with prochlorperazine, promethazine, metoclopramide, and droperidol use. Dehydration and concomitant lithium therapy are risk factors. The syndrome can be rapidly fatal from rhabdomyolysis and multiorgan failure. Treatment involves withdrawal of the offending drug, hydration, benzodiazepines, dantrolene (muscle relaxants), dopamine agonists and supportive care (Guze´ and Baxter, 1985; Fisher and Davis, 2002).

639

Serotonin syndrome results from a hyperserotonergic state following therapeutic drug use or inadvertent drugto-drug interactions. Manifestations of the serotonin syndrome range from mild akathisia and tachycardia to severe tremulousness, myoclonus, rigidity, sustained clonus, delirium, autonomic instability, hyperthermia, and cardiovascular shock. Metoclopramide and the setrons have been implicated in causing serotonin syndrome. Their use with serotonin reuptake inhibitors, antidepressants, lithium, triptans, opioid analgesics, valproate, linezolid, and other proserotonergic drugs can heighten the symptoms. Treatment involves withdrawal of the offending drug(s), hydration, benzodiazepines, HT2a antagonists, control of hyperthermia and autonomic dysfunction, and supportive care (Fisher and Davis, 2002; Boyer and Shannon, 2005; George et al., 2008; Patel et al., 2011).

CONCLUSION Antiemetic therapy has evolved from the use of dopamine blockers like the phenothiazines with the potential for serious extrapyramidal reactions to selective serotonin and neurokinin receptor inhibitors that are more potent than the phenothiazines (D2 blockers) and have a better safety profile. While metoclopramide remains the most extensively used prokinetic in most parts of the world, newer prokinetic agents with better adverse effect profiles are under development and review. Postmarketing surveys have been critical in identifying serious adverse effects such as the cardiotoxicity of cisapride, cardiovascular events following tegaserod use, and risk of tardive dyskinesias following long-term use of metoclopramide or levosulpiride. Epidemiologic studies have also helped in defining the spectrum of drug-induced neurotoxicity and at-risk populations. For instance, young men are susceptible to metoclopramide-induced acute dystonic reactions while the elderly are more vulnerable to suffering tardive dyskinesias (Bhatt et al., 2004). The commonly used gastrointestinal drugs, comprising antiemetics, promotility drugs and drugs to treat acidrelated disorders, are used to treat disorders that can lead to morbidity but are not fatal (Parikh and Howden, 2010). Therefore, the benefits of their use should be evaluated taking into account possible adverse effects, even if these are uncommon. As far as possible, drugs such as metoclopramide and others that can lead to tardive dyskinesias should be used for as short a time as possible, with close clinical monitoring and patient education.

ACKNOWLEDGEMENT The authors would like to thank Dr. Amruta Ravan for help with manuscript assembly and proofreading.

640

A. AGGARWAL AND M. BHATT

REFERENCES Aapro MS, Walko CM (2010). Aprepitant: drug-drug interactions in perspective. Ann Oncol 21: 2316–2323. Andrejak M, Masmoudi K, Mizon JP (1990). Acute dyskinesia after the ingestion of antiemetics leading to emergency hospitalization. Therapie 45: 33–35. Anfinson TJ (2002). Akathisia, panic, agoraphobia, and major depression following brief exposure to metoclopramide. Psychopharmacol Bull 36: 82–93. Baik JS, Lyoo CH, Lee JH et al. (2008). Drug-induced and psychogenic resting suprahyoid neck and tongue tremors. Mov Disord 23: 746–748. Bateman DN, Gokal R (1980). Metoclopramide in renal failure. Lancet 1: 982. Bateman DN, Kahn C, Mashiter K et al. (1978). Pharmacokinetic and concentration-effect studies with intravenous metoclopramide. Br J Clin Pharmacol 6: 401–407. Bateman DN, Rawlins MD, Simpson JM (1985). Extrapyramidal reactions with metoclopramide. Br Med J (Clin Res Ed) 291: 930–932. Bateman DN, Darling WM, Boys R et al. (1989). Extrapyramidal reactions to metoclopramide and prochlorperazine. Q J Med 71: 307–311. Bhanji NH, Margolese HC (2004). Extrapyramidal symptoms related to adjunctive nizatidine therapy in an adolescent receiving quetiapine and paroxetine. Pharmacotherapy 24: 923–925. Bhatt M, Sethi K, Bhatia K (2004). Acute and tardive dystonia. In: K Sethi (Ed.), Drug-Induced Movement Disorders. Marcel Dekker, New York, pp. 111–128. Biasini A, Alberti A (1985). Extrapyramidal dysfunction after domperidone. Helv Paediatr Acta 40: 93–94. Bondon-Guitton E, Perez-Lloret S, Bagheri H et al. (2011). Drug-induced parkinsonism: a review of 17 years’ experience in a regional pharmacovigilance center in France. Mov Disord 26: 2226–2231. Bories P, Michel H, Brigitte D et al. (1980). Use of ranitidine, without mental confusion, in patient with renal failure. Lancet 2: 755. Boyer EW, Shannon M (2005). The serotonin syndrome. N Engl J Med 352: 1112–1120. Burke RE, Kang UJ, Jankovic J et al. (1989). Tardive akathisia: an analysis of clinical features and response to open therapeutic trials. Mov Disord 4: 157–175. Burns R, Thomas DW, Barron VJ (1974). Reversible encephalopathy possibly associated with bismuth subgallate ingestion. Br Med J 1: 220–223. Caldwell C, Rains G, McKiterick K (1987). An unusual reaction to preoperative metoclopramide. Anesthesiology 67: 854–855. Casteels-Van Daele M, Jaeken J, Van der Schueren P et al. (1970). Dystonic reactions in children caused by metoclopramide. Arch Dis Child 45: 130–133. Catalano G, Catalano MC, Alberts VA (1996). Famotidineassociated delirium. A series of six cases. Psychosomatics 37: 349–355. Cerra BF, Schentag JJ, Mcmillen M et al. (1982). Mental status, the intensive care unit and cimetidine. Ann Surg 196: 565–570.

Chow LY, Chung D, Leung V et al. (1997). Suicide attempt due to metoclopramide-induced akathisia. Int J Clin Pract 51: 330–331. Costall B, Naylor RJ (1973). Is there a relationship between the involvement of extrapyramidal and mesolimbic brain areas with the cataleptic action of neuroleptic agents and their clinical antipsychotic effect? Psychopharmacologia 32: 161–170. Critchley P, Langdon N, Parkes JD et al. (1985). Domperidone. Br Med J (Clin Res Ed) 290: 788. Cundy T, Dissanayake A (2008). Severe hypomagnesaemia in long-term users of proton-pump inhibitors. Clin Endocrinol (Oxf) 69: 338–341. Davis WA (1984). Mental confusion associated with ranitidine. Med J Aust 140: 478. Dieckmann K, Maurage C, Rolland JC et al. (1996). Torticollis as a side effect of cisapride treatment in an infant. J Pediatr Gastroenterol Nutr 22: 336. Dobrow RB, Coppock MA, Hosenpud JR (1991). Extrapyramidal reaction caused by ondansetron. J Clin Oncol 9: 1921. Drake RE, Ehrlich J (1985). Suicide attempts associated with akathisia. Am J Psychiatry 142: 499–501. Duncan MA, Nikolov NM, O’Kelly B (2001). Acute chorea due to ondansetron in an obstetric patient. Int J Obstet Anesth 10: 309–311. Edmonds ME, Ashford RF, Brenner MK et al. (1979). Cimetidine: does neurotoxicity occur? J R Soc Med 72: 172. Ellis GL, Delaney J, DeHart DA et al. (1993). The efficacy of metoclopramide in the treatment of migraine headache. Ann Emerg Med 22: 191–195. Elzinga-Huttenga J, Hekster Y, Bijl A et al. (2006). Movement disorders induced by gastrointestinal drugs: two paediatric cases. Neuropediatrics 37: 102–106. Escourelle R, Bourdon R, Galli A et al. (1977). Neuropathologic and toxicologic study of 12 cases of bismuth encephalopathy. Rev Neurol 133: 153–163. Feest TG, Read DJ (1980). Myopathy associated with cimetidine? Br Med J 281: 1284–1285. Feyer P, Jordan K (2011). Update and new trends in antiemetic therapy: the continuing need for novel therapies. Ann Oncol 22: 30–38. Fisher AA, Davis MW (2002). Serotonin syndrome caused by selective serotonin reuptake-inhibitors-metoclopramide interaction. Ann Pharmacother 36: 67–71. Flind AC, Rowley-Jones D (1979). Mental confusion and cimetidine. Lancet 1: 379. Ford AC, Malfertheiner P, Giguere M et al. (2008). Adverse events with bismuth salts for Helicobacter pylori eradication: systematic review and meta-analysis. World J Gastroenterol 14: 7361–7370. Fouddah A, Canivet JL, Damas P (2001). Clinical case of the month. Severe dyskinetic syndrome induced by ranitidine. Rev Med Liege 56: 548–551. Franckx J, Noel P (1984). Acute extrapyramidal dysfunction after domperidone administration. Report of a case. Helv Paediatr Acta 39: 285–288. Galynker II, Tendler DS (1997). Nizatidine-induced delirium in a nonagenarian. J Clin Psychiatry 58: 327.

COMMONLY USED GASTROINTESTINAL DRUGS Ganzini L, Casey DE, Hoffman WF et al. (1993). The prevalence of metoclopramide-induced tardive dyskinesia and acute extrapyramidal movement disorders. Arch Intern Med 153: 1469–1475. Gatrad AR, Gatrad AH (1979). Familial incidence of dystonic reactions to metoclopramide (Maxolon). Br J Clin Pract 33: 111–115. George M, Al-Duaij N, O’Donnell KA et al. (2008). Obtundation and seizure following ondansetron overdose in an infant. Clin Toxicol 46: 1064–1066. Gralla RJ, Itri LM, Pisko SE et al. (1981). Antiemetic efficacy of high-dose metoclopramide: randomized trials with placebo and prochlorperazine in patients with chemotherapy-induced nausea and vomiting. N Engl J Med 305: 905–909. Grimes JD (1981). Parkinsonism and tardive dyskinesia associated with long-term metoclopramide therapy. N Engl J Med 305: 1417. Grimes JD, Hassan MN, Krelina M (1982a). Long-term follow-up of tardive dyskinesia due to metoclopramide. Lancet 4: 563. Grimes JD, Hassan MN, Preston DN (1982b). Adverse neurologic effects of metoclopramide. Can Med Assoc J 126: 23–25. Grundy D, Al-Chaer ED, Aziz Q et al. (2006). Fundamentals of neurogastroenterology: basic science. Gastroenterology 130: 1391–1411. Guala A, Mittino D, Fabbrocini P et al. (1992). Familial metoclopramide-induced dystonic reactions. Mov Disord 7: 385–386. Guze´ BH, Baxter LR Jr (1985). Current concepts. Neuroleptic malignant syndrome. N Engl J Med 313: 163–166. Handler CE, Besse CP, Wilson AO (1982). Extrapyramidal and cerebellar syndrome with encephalopathy associated with cimetidine. Postgrad Med J 58: 527–528. Harrington RA, Hamilton CW, Brogden RN et al. (1983). Metoclopramide. An updated review of its pharmacological properties and clinical use. Drugs 25: 451–494. Henann NE, Carpenter DU, Janda SM (1988). Famotidineassociated mental confusion in elderly patients. Drug Intell Clin Pharm 22: 976–978. Herbert MK, Holzer P (2008). Standardized concept for the treatment of gastrointestinal dysmotility in critically ill patients – current status and future options. Clin Nutr 27: 25–41. Herrstedt J (2008). Antiemetics: an update and the MASCC guidelines applied in clinical practice. Nat Clin Pract Oncol 5: 32–43. Herrstedt J, Dombernowsky P (2007). Anti-emetic therapy in cancer chemotherapy: current status. Basic Clin Pharmacol Toxicol 101: 143–150. Hesketh PJ (2008). Chemotherapy-induced nausea and vomiting. N Engl J Med 358: 2482–2494. Howden CW, Tytgat GN (1996). The tolerability and safety profile of famotidine. Clin Ther 18: 36–54. Hughes JD, Reed WD, Serjeant CS (1983). Mental confusion associated with ranitidine. Med J Aust 2: 12–13. Indo T, Ando K (1982). Metoclopramide-induced Parkinsonism. Clinical characteristics of ten cases. Arch Neurol 39: 494–496.

641

Kanarek BB, Curnow R, Palmer J et al. (1992). Ondansetron: confusing documentation surrounding an extrapyramidal reaction. J Clin Oncol 10: 506–507. Karamanolis G, Tack J (2006). Promotility medications – now and in the future. Dig Dis 24: 297–307. Kenney C, Hunter C, Davidson A et al. (2008). Metoclopramide, an increasingly recognized cause of tardive dyskinesia. J Clin Pharmacol 48: 379–384. Kerr GW (1996). Dystonic reactions: two case reports. J Accid Emerg Med 13: 221–222. Kim JS, Ko SB, Han SR et al. (2003). Levosulpiride-induced Parkinsonism. J Korean Neurol Assoc 21: 418–421. Kounenis G, Koutsoviti-Papadopoulou, Elezoglou V (1994). Effect of nizatidine and ranitidine on the D-tubocurarine neuromuscular blockade in the toad rectus abdominis muscle. Pharmacol Res 29: 155–161. Kovac AL (2003). Benefits and risks of newer treatments for chemotherapy-induced and postoperative nausea and vomiting. Drug Saf 26: 227–259. Kumar N, Hu WT (2009). Extrapyramidal reaction to ondansetron and propofol. Mov Disord 24: 312–313. LaGorio J, Thompson VA, Sternberg D et al. (1998). Akathisia and anesthesia: refusal of surgery after the administration of metoclopramide. Anesth Analg 87: 224–227. Lang AE (1988). Akathisia and restless leg syndrome. In: J Jankovic, E Tolosa (Eds.), Parkinson’s Disease and Movement Disorders. Urban and Schwarzenberg, Baltimore, pp. 349–364. Lavy S, Melamed E, Penchas S (1978). Tardive dyskinesia associated with metoclopramide. Br Med J 1: 77–78. Lee CR, Plosker GL, McTavish D (1993). Tropisetron. A review of its pharmacodynamic and pharmacokinetic properties, and therapeutic potential as an antiemetic. Drugs 46: 925–943. Lee CY, Ratnapalan S, Thompson M et al. (2010). Unusual reactions to 5-HT3 receptor antagonists in a child with rhabdomyosarcoma. Can J Clin Pharmacol 17: 1–4. Leeser J, Bateman DN (1985). Domperidone. Br Med J (Clin Res Ed) 290: 241–242. Leo RJ, Lichter DG, Hershey LA (1995). Parkinsonism associated with fluoxetine and cimetidine: a case report. J Geriatr Psychiatry Neurol 8: 231–233. Lewis JH (1991). Safety profile of long-term H2-antagonist therapy. Aliment Pharmacol Ther 5: 49–57. Lu CS, Chu NS (1988). Acute dystonic reaction with asterixis and myoclonus following metoclopramide therapy. J Neurol Neurosurg Psychiatry 51: 1002–1003. Lucena R, Monteiro L, Melo A (1998). Cisapride related movement disorders. J Pediatr 74: 416–418. Mark R, Newton-John H (1988). Acute upper airway obstruction due to supraglottic dystonia induced by a neuroleptic. Br Med J 297: 964–965. Mastaglia FL, Argov Z (2007). Toxic and iatrogenic myopathies. Handb Clin Neurol 86: 321–341. Miller LG, Jankovic J (1989). Metoclopramide-induced movement disorders. Clinical findings with a review of the literature. Arch Intern Med 149: 2486–2492. Morrow AW (1973). Requests for reports; adverse reactions with bismuth subgallate. Med J Aust 1: 912.

642

A. AGGARWAL AND M. BHATT

Naito Y, Kuzuhara S (1994). Parkinsonism induced or worsened by cisapride. Nihon Ronen Igakkai Zasshi 31: 899–902. Nampiaparampil D, Oruc NE (2006). Metodopramideinduced palatopharyngeal myoclonus. Mov Disord 21: 2028–2029. Parikh N, Howden CW (2010). The safety of drugs used in acid-related disorders and functional gastrointestinal disorders. Gastroenterol Clin North Am 39: 529–542. Parkman HP, Hasler WL, Fisher RS et al. (2004). American Gastroenterology Association review on the diagnosis and treatment of gastroparesis. Gastroenterology 127: 1592–1622. Parlak I, Atilla R, Cicek M et al. (2005). Rate of metoclopramide infusion affects the severity and incidence of akathisia. Emerg Med J 22: 621–624. Pasricha PJ (2007). Desperately seeking serotonin. A commentary on the withdrawal of tegaserod and the state of drug development for functional and motility disorders. Gastroenterology 132: 2287–2290. Pasricha PJ, Pehlivanov N, Sugumar A et al. (2006). Drug insight: from disturbed motility to disordered movement – a review of the clinical benefits and medicolegal risks of metoclopramide. Nat Clin Pract Gastroenterol Hepatol 3: 138–148. Patel A, Mittal S, Manchanda S et al. (2011). Ondansetroninduced dystonia, hypoglycemia, and seizures in a child. Ann Pharmacother 45: 7. Peiris RS, Peckler BF (2001). Cimetidine-induced dystonic reaction. J Emerg Med 21: 27–29. Perez-Lloret S, Bondon-Guitton E, Rascol O et al. (2010). Adverse drug reactions to dopamine agonists: a comparative study in the French Pharmacovigilance Database; French Association of Regional Pharmacovigilance Centers. Mov Disord 25: 1876–1880. Poortinga E, Rosenthal D, Bagri S (2001). Metoclopramideinduced akathisia during the second trimester of a 37-yearold woman’s first pregnancy. Psychosomatics 42: 153–156. Raschetti R, Maggini M, Da Cas R et al. (2001). Time trends in the coprescribing of cisapride and contraindicated drugs in Umbria, Italy. JAMA 285: 1840–1841. Reid M (1977). Dystonic reactions to metoclopramide (Maxolon). Ulster Med J 46: 38–40. Ritter MJ, Goodman BP, Sprung J et al. (2003). Ondansetroninduced multifocal encephalopathy. Mayo Clin Proc 78: 1150–1152. Robinson OP (1973). Metoclopramide – side effects and safety. Postgrad Med J 49: 77–80. Rodgers PT, Brengel GR (1998). Famotidine-associated mental status changes. Pharmacotherapy 18: 404–407. Roila F, Fatigoni S (2006). New antiemetic drugs. Ann Oncol 2: ii96–ii100. Rossi F, Forgione A (1995). Pharmacotoxicological aspects of levosulpiride. Pharmacol Res 31: 81–94. Roy-Desruisseaux J, Landry J, Bocti C et al. (2011). Domperidone-induced tardive dyskinesia and withdrawal psychosis in an elderly woman with dementia. Ann Pharmacother 45: 51. Sachdev P, Kruk J (1994). Clinical characteristics and predisposing factors in acute drug-induced akathisia. Arch Gen Psychiatry 51: 963–974.

Samie MR, Dannenhoffer MA, Rozek S (1987). Lifethreatening tardive dyskinesia caused by metoclopramide. Mov Disord 2: 125–129. Sargent AI, Deppe SA, Chan FA (1993). Seizure associated with ondansetron. Clin Pharmacol 12: 613–615. Schneider SA, Udani V, Sankhla CS et al. (2009). Recurrent acute dystonic reaction and oculogyric crisis despite withdrawal of dopamine receptor blocking drugs. Mov Disord 24: 1226–1229. Schulze-Delrieu K (1981). Drug therapy. Metoclopramide. N Engl J Med 305: 28–33. Sethi KD (2004). Drug-Induced Movement Disorders, Marcel Dekker, New York. Sewell DD, Jeste DV (1992). Metoclopramide-associated tardive dyskinesia. An analysis of 67 cases. Arch Fam Med 1: 271–278. Shaffer D, Butterfield M, Pamer C et al. (2004). Tardive dyskinesia risks and metoclopramide use before and after U.S. market withdrawal of cisapride. J Am Pharm Assoc 44: 661–665. Sharma A, Raina V (2001). Generalized seizures following ondansetron. Ann Oncol 12: 131–132. Sharpe PC, Burland WL (1980). Mental confusion and H2receptor blockers. Lancet 2: 924. Shin HW, Kim MJ, Kim JS et al. (2009). Levosulpirideinduced movement disorders. Mov Disord 24: 2249–2253. Sills JA, Glass EJ (1978). Metoclopramide in young children. Br Med J 2: 431. Silverstone PH (1984). Ranitidine and confusion. Lancet 1: 1071. Singh NN, Rai A, Selhorst JB et al. (2009). Ondansetron and seizures. Epilepsia 50: 2663–2666. Sorbe B, Halle´n C, Frankendal B (1994). An open, randomized study to compare the efficacy and tolerability of tropisetron with that of a metoclopramide-containing antiemetic cocktail in the prevention of cisplatin-induced emesis. Cancer Chemother Pharmacol 33: 298–302. Spiegel JE, Kang V, Kunze L et al. (2005). Ondansetroninduced extrapyramidal symptoms during cesarean section. Int J Obstet Anesth 14: 368–369. Sprung J, Choudhry FM, Hall BA (2003). Extrapyramidal reactions to ondansetron: cross-reactivity between ondansetron and prochlorperazine? Anesth Analg 96: 1374–1376. Stacy M, Jankovic J (1992). Tardive tremor. Mov Disord 7: 53–57. Steinherz R, Levy Y, Ban-Amiti D et al. (1986). Extrapyramidal reactions to domperidone. J Pediatr 108: 630–631. Stonell C (1998). An extrapyramidal reaction to ondansetron. Br J Anaesth 81: 658. Supino-Viterbo V, Sicard C, Risvegliato M et al. (1977). Toxic encephalopathy due to ingestion of bismuth salts: clinical and EEG studies of 45 patients. J Neurol Neurosurg Psychiatry 40: 748–752. Tarsy D, Indorf G (2002). Tardive tremor due to metoclopramide. Mov Disord 17: 620–621. Tillman LA, Drake FM, Dixon JS et al. (1996). Review article: safety of bismuth in the treatment of gastrointestinal diseases. Aliment Pharmacol Ther 10: 459–467.

COMMONLY USED GASTROINTESTINAL DRUGS Tisdale SA (1981). Metoclopramide. N Engl J Med 305: 1093. Toga T, Kohmura Y, Kawatsu R (2007). The 5-HT(4) agonists cisapride, mosapride, and CJ-033466, a novel potent compound, exhibit different human ether-a-go-gorelated gene (hERG)-blocking activities. J Pharmacol Sci 105: 207–210. Tolan MM, Fuhrman TM, Tsueda K et al. (1999). Perioperative extrapyramidal reactions associated with ondansetron. Anesthesiology 90: 340–341. Van der Padt A, Van Schaik RH, Sonneveld P (2006). Acute dystonic reaction to metoclopramide in patients carrying homozygous cytochrome P450 2D6 genetic polymorphisms. Neth J Med 64: 160–162. Vandenplas Y, Benatar A, Cools F et al. (2001). Efficacy and tolerability of cisapride in children. Paediatr Drugs 3: 559–573. Walls TJ, Pearce SJ, Venables GS (1980). Motor neuropathy associated with cimetidine. Br Med J 281: 974–975.

643

Weaving A, Bezwoda WR, Derman DP (1984). Seizures after antiemetic treatment with high dose domperidone: report of four cases. Br Med J (Clin Res Ed) 288: 1728. Weiden PJ, Mann JJ, Haas G et al. (1987). Clinical nonrecognition of neuroleptic-induced movement disorders: a cautionary study. Am J Psychiatry 144: 1148–1153. Wilson LB, Woodward AM, Ferrara JJ (1997). An acute dystonic reaction with long-term use of ranitidine in an intensive care unit patient. J La State Med Soc 149: 36–38. Yamamoto M, Ujike H, Ogawa N (1987). Metoclopramideinduced parkinsonism. Clin Neuropharmacol 10: 287–289. Yuan RY, Kao CR, Sheu JJ et al. (2001). Delirium following a switch from cimetidine to famotidine. Ann Pharmacother 35: 1045–1048. Zambelli A, Sagrada P, Pavesi L (2009). Seizure associated with palonosetron. Support Care Cancer 17: 217.

Handbook of Clinical Neurology, Vol. 120 (3rd series) Neurologic Aspects of Systemic Disease Part II Jose Biller and Jose M. Ferro, Editors © 2014 Elsevier B.V. All rights reserved

Chapter 44

Neurologic manifestations of acute liver failure SUDHIR DATAR AND EELCO F.M. WIJDICKS* Division of Critical Care Neurology, Mayo Clinic, Rochester, MN, USA

INTRODUCTION Neurologists have been familiar with the manifestations of chronic liver disease for more than a century but in acute liver failure most have been on less sure ground. Acute liver failure (ALF) (Trey and Davidson, 1970; Lee et al., 2008), best known as fulminant hepatic failure, is a clinical syndrome of acute onset of encephalopathy and coagulopathy (international normalized ratio (INR)  1.5) resulting from massive loss of hepatocyte function in a patient without any pre-existing liver disease, with an illness duration of < 26 weeks (Trey and Davidson, 1970; Lee et al., 2012). Suicide attempt with acetaminophen (paracetamol) remains a common cause. Fortunately, such a catastrophic event is rare and in the US, about 2000 cases are reported annually (Lee et al., 2008). In the pre-transplant era, morbidity and mortality was high and autopsies would show brain edema, but these manifestations were viewed as terminal events in a critically ill mechanically ventilated patient. There has been a paradigm shift in our understanding of acute liver failure over the last 20 years and, as a result of the discovery that cerebral edema is treatable, it has become partly a neurologic disease. Emergency liver transplantation (Bismuth et al., 1987; Emond et al., 1989; Brandsaeter et al., 2002; Fujiwara and Mochida, 2002; Russo et al., 2004; Montalti et al., 2005; Detry et al., 2007; O’Grady, 2007) and management in modern intensive care units (ICUs) equipped to handle the complications have increased survival rates to 65%, far better than the miserable earlier survival rates of < 15% (Ostapowicz et al., 2002; Khashab et al., 2007). It has become apparent that one of the major causes of death in these patients is cerebral edema and intracranial hypertension, eventually leading to brain death if untreated. Technology is now available to monitor intracranial pressure (ICP), which, combined with mean

arterial pressure (MAP), provides an estimation of cerebral perfusion. However, these techniques are invasive, needing burr holes and placement of intracranial monitoring devices in a patient with very poor coagulation. The roles that a neurologist can play when evaluating a patient with ALF are: (1) to rapidly identify patients who are at risk of cerebral edema and intracranial hypertension; (2) to carefully select the patient population who will benefit from invasive monitoring; (3) to select the correct time to start monitoring; (4) to suggest to the neurosurgeon the appropriate tissue compartment (subdural, epidural, or parenchymal) for the placement of ICP monitors; (5) to participate in treatment of cerebral edema and increased ICP; (6) to manage complications such as intracranial hemorrhage or seizures; (7) to assist in selection of candidates for liver transplantation and referral to transplant center. This chapter illustrates the clinical presentation and overall management of ALF, with a focus on the management of cerebral edema.

HISTORICAL PERSPECTIVE The liver plays an indispensable role in homeostasis, playing a major role in synthesis of proteins and removal of waste products of metabolism. With acute liver failure, these mechanisms are compromised with the major consequences being coagulopathy and encephalopathy with or without cerebral edema. Cerebral edema in ALF was described as early as the 1960s. Williams and colleagues examined 92 cases of fulminant hepatic failure over a 7 year period and found the presence of cerebral edema in 31% of the cases (Record et al., 1975). Mortality seems to be associated with brain herniation, which was found in 20 patients. The causes of acute injury to the hepatocytes have changed over the last few decades. In the 1980s, acute

*Correspondence to: Eelco F.M. Wijdicks, M.D., Ph.D., Department of Neurology, Mayo Clinic, 200 First Street SW, 55905, Rochester, MN, USA. E-mail: [email protected]

646

S. DATAR AND E.F.M. WIJDICKS

viral hepatitis played a major role. However, with time, paracetamol (acetaminophen) toxicity started emerging as a major cause and today about half of all cases of ALF are due to paracetamol (acetaminophen) overdose. Other causes of acute loss of hepatocyte function are idiosyncratic drug reaction, hepatic venous thrombosis (Budd–Chiari syndrome), acute fatty liver of pregnancy/HELLP (hemolysis, elevated liver enzymes, low platelet count) syndrome, ingestion of certain types of mushrooms, Wilson’s disease, autoimmune hepatitis, cardiac arrest, or any other severe episode of sufficiently prolonged hypotension leading to “shock liver,” infiltration of the liver by malignancy, etc. Reye’s syndrome (Meythaler and Varma, 1987) associated with the use of aspirin or viral infections (influenza-like illness or varicella) is now extraordinarily rare and any child presenting with a similar clinical picture should undergo extensive investigations to rule out inborn errors of metabolism which can mimic Reye’s syndrome (Belay et al., 1999). Attempts at using artificial livers go back to the 1970s. Williams and colleagues, from King’s College Hospital, London, described the use of temporary liver support using isolated but functioning pig’s liver in an extracorporeal perfusion circuit. Attempts were also made at using human liver or the liver of a baboon. Perfusion could be continued for only up to 48 hours with human or baboon’s liver and for up to 6–8 hours with pig’s liver. This was followed by only temporary improvement in laboratory abnormalities or level of consciousness. However, overall, long-term results were discouraging (Williams, 1975). The main conceptional change has been to connect brain edema with care of acute liver failure and to prioritize liver transplantation. A new liver may be the only effective treatment of brain edema in some patients.

CLINICAL PRESENTATION ALF is commonly seen in critically ill patients already admitted to the hospital in the setting of multiorgan failure, shock or cardiac arrest, and a neurologist may get involved quickly. Often these patients are admitted to the ICU for several reasons which could include difficulty with airway protection and respiratory failure needing mechanical ventilation, severe hypotension, sepsis syndrome, and rapidly worsening “hepatic encephalopathy.” As expected, the initial symptoms are typical of any encephalopathy. Apathy is one of the earliest psychiatric symptoms (Wijdicks, 2009). Other symptoms are hypersomnia and impaired judgment (Rothstein and Herlong, 1989). Surprisingly, patients may present with agitated delirium or even euphoria with a transition to mania (Zacharski et al., 1970).

Spatial disorientation, impairment in thought process and content, fluctuations in attention, slowed cognitive processing and impairment of short-term and immediate memory may be present. Multifocal myoclonus and excessive startle responses can occur. In contrast to chronic liver disease, asterixis is rather uncommon (Wijdicks, 2009). Encephalopathy of any grade may signify the onset of cerebral edema, but the probability increases with worsening encephalopathy (Table 44.1). Pupillary responses are normal initially but may become impaired when cerebral herniation develops, eventually progressing to fixed midposition pupils (4–6 mm) due to displacement and compression of the mesencephalon from the diffuse mass effect. Oculocephalic responses usually remain normal but may become brisk or transiently disappear (Heubi et al., 1984). Dysconjugate gaze occurs rarely (Caplan and Scheiner, 1980) and an equally rare but reported phenomenon is the occurrence of periodic alternating gaze deviation (PAGD) in patients with hepatic encephalopathy without any identifiable structural injury, which can resolve with the treatment of encephalopathy (Averbuch-Heller and Meiner, 1995). In general, however, oculomotor dysfunction is uncommon and its presence should raise suspicion for alternative causes other than hepatic encephalopathy. Decerebrate and decorticate posturing, albeit very dramatic, may be completely reversible after correction of ammonia and improvement of liver function (Conomy and Swash, 1968). Other common findings are increased muscle tone, exaggerated deep tendon reflexes, and Babinski’s signs (Wijdicks, 2009). Table 44.1 Grades of encephalopathy: West Haven criteria (adapted from Ferenci et al., 2002) Grade

Clinical characteristics

I

Trivial lack of awareness Euphoria or anxiety Shortened attention span Impaired performance of addition Lethargy or apathy Minimal disorientation for time or place Subtle personality change Inappropriate behavior Impaired performance of subtraction Somnolence to semistupor, but responsive to verbal stimuli Confusion Gross disorientation Coma (unresponsive to verbal or noxious stimulus)

II

III

IV

NEUROLOGIC MANIFESTATIONS OF ACUTE LIVER FAILURE 647 As the severity of liver failure increases, consciouscreatinine suggests acute renal failure, which also ness becomes impaired, eventually progressing to coma. complicates the clinical picture. These acute metabolic Even if these patients initially present with mild encephabnormalities in theory could contribute toward encephalopathy, they can worsen rapidly from quick developalopathy; they are usually transient as they are easily corment of cerebral edema and hence they need close rectable. However, a worsening encephalopathy should monitoring in the hospital which may be best done in not be readily attributed to these metabolic factors as an ICU setting. Grades of hepatic encephalopathy are any appreciable change can signify the onset of cerebral summarized in Table 44.1. edema with fatal consequences if untreated. Generally any pathologic motor response or abnorElevation of white blood cell count could suggest the mality of mesencephalic or pontine reflexes is suspipresence of an underlying infection, especially if it is cious for diffuse cerebral edema. Classic clinical associated with a left shift. Thrombocytopenia may be signs of increased ICP, such as Cushing’s response of present. Blood gas measurement may reveal acidosis systemic hypertension and bradycardia, are not but more commonly shows alkalosis which could be always present. Computed tomography (CT) scans respiratory or metabolic. Other pertinent laboratory should be repeated even if transport of the patient is studies include tests to identify a cause for the ALF. This far from ideal. may include testing of paracetamol (acetaminophen) Both focal and generalized seizures have been levels in cases of suspected exposure, viral hepatitis reported to occur, even in the absence of radiographipanel testing for A, B, C and, if indicated, D and E cally identifiable focal brain injury which could account viruses. If there is clinical suspicion, testing for autoimfor it. In severely encephalopathic patients a low index of mune hepatitis may be indicated with serum antinuclear suspicion must be maintained for ongoing subclinical antibody, smooth muscle antibody, antibodies to liver seizures. A review of electroencephalograms (EEGs) and kidney microsomes (anti-LKM), antimitochondrial over a 10 year period identified 18 (15%) with epileptiantibody and serum protein electrophoresis. Liver ultraform abnormalities (Ficker et al., 1997). In another study, sound with Doppler studies can identify portal vein or by Ellis and colleagues, 13 out of 42 patients with ALF hepatic vein thrombosis. who underwent continuous EEG monitoring were found Brain CT scan is usually the first neuroimaging study to have electrographic seizures (Ellis et al., 2000). Subperformed as it is quick, readily available, and can identify clinical seizure activity is probably an under-recognized complications of ALF such as cerebral edema with evioccurrence which also potentially contributes towards dence of mass effect (Wijdicks et al., 1995) or, in rare cases, cerebral hypoxia and cerebral edema. acute intracranial hemorrhage. Brain edema on CT scan is As expected, jaundice, hepatic tenderness, and ascites demonstrated by effacement of cortical sulci, blurring of (in cases of acute hepatic vein thrombosis) may be pregray–white differentiation, and obliteration of basal cissent. However, signs of chronic liver failure are usually terns (Fig. 44.1). However, it should be noted that brain absent, as by definition, acute liver failure implies CT scan is not a very sensitive way of excluding early cereabsence of any chronic liver disease. bral edema and early abnormalities can be very subtle, Contrary to liver cirrhosis commonly presenting such as disappearance of some sulci and sylvian fissures with gastrointestinal bleeding, in ALF spontaneous (Fig. 44.2) (Munoz et al., 1991). Any patient with ALF hemorrhage is rather uncommon and any significant and advanced hepatic encephalopathy with a normal CT hemorrhage requiring transfusion is rare (Boks et al., scan can still be at high risk of rapidly developing intracra1986). Acute portal hypertension can develop from nial hypertension (IH) and it makes perfect sense to be proALF, but bleeding from varices seldom occurs. active and consider ICP monitoring. Magnetic resonance (MR) imaging abnormalities have been described in the periventricular white matter, thalami, posterior limb of the internal capsule, dorsal LABORATORY TESTS AND brainstem and cortex. These abnormalities are seen on IMAGING STUDIES both fluid attenuated inversion recovery (FLAIR) as well Laboratory tests show marked elevation of transamias diffusion-weighted imaging (DWI). Serum ammonia nases, increased INR and increased serum ammonia. has been shown to correlate strongly with FLAIR and Phosphorus, magnesium, and potassium are frequently DWI severity and clinical outcome (McKinney et al., low and may need supplementation. Glucose should be 2010). Diffusion restriction is likely explained by cytotoxic monitored and replaced if indicated, as severe hypoglyedema from astrocyte swelling (Ranjan et al., 2005). cemia may occur and markedly confound the clinical Diffuse cortical involvement has a higher potential presentation. Similarly severe hyponatremia may be for neurologic sequelae but these changes are reversible present. Acute elevation of blood urea nitrogen and (Fig. 44.3).

648

S. DATAR AND E.F.M. WIJDICKS

B

A

Fig. 44.1. A 30-year-old woman with acute hepatic failure due to HELLP syndrome. (A and B) CT of the brain without contrast showing cerebral edema with effacement of the sulci, blurring of gray–white differentiation and obliteration of cisterns. Bolt for ICP monitoring can be seen in image (B) (arrow). The patient received liver transplantation but did poorly postoperatively due to high intracranial pressure and died from cerebral herniation.

A

B

Fig. 44.2. A 32-year-old woman with acute hepatic failure from paracetamol (acetaminophen) overdose. (A) Normal noncontrast head CT scan without any evidence of cerebral edema but showing effacement of both sylvian fissures. Intracranial pressure (ICP) was elevated. (B) Placement of bolt for monitoring ICP (arrow). She went on to receive a liver transplant and did well.

NEUROLOGIC MANIFESTATIONS OF ACUTE LIVER FAILURE

A

649

B

Fig. 44.3. MRI of the brain without contrast showing evidence of cerebral edema. (A) T2-weighted image showing diffuse effacement of sulci and increased cortical T2 signal suggestive of cerebral edema. (B) Fluid attenuated inversion recovery (FLAIR) image showing periventricular T2 signal abnormalities in addition to cortical signal changes, suggestive of cerebral edema.

Somatosensory evoked potentials (SSEP) are markedly different in patients with fulminant liver failure (Madl et al., 1994). A study using bilateral median nerve-stimulated short- and long-latency sensory evoked potentials found that nine patients had spontaneous recovery, eight patients underwent emergency liver transplantation, and eight patients died (Madl et al., 1994). Probability of correct outcome prediction by sensory evoked potentials versus that based on clinical criteria alone was 0.96 versus 0.72, respectively. It was concluded that serial recording of sensory evoked potentials may help identify a subgroup of patients who should undergo early liver transplantation even though they do not fulfill the King’s College criteria (Table 44.2). However, experience with SSEPs is very limited and until there are more data, SSEPs should be used cautiously for this purpose.

REFERRAL TO TRANSPLANT CENTER Once a diagnosis of acute liver failure is established, the next step is to quickly determine if the patient is a transplant candidate. Urgent liver transplantation is indicated in ALF where prognostic indicators suggest a high probability of poor outcome (see section on outcome). However, complete reliance on the prognostic scoring

systems is not recommended as they do not adequately predict outcome and candidacy for liver transplantation (Lee et al., 2012). Other factors that also play a role in determining the candidacy of transplantation include overall medical comorbidities. Conditions such as severe coexistent medical disease, progressive inotropic or ventilator support, irreversible brainstem dysfunction, multiple previous episodes of self-harm (>5), especially if nondrug methods were used, refractory or resistant mental illness, incapacitating dementia or mental retardation, active intravenous drug use, or excessive alcohol use may be relative contraindications for transplantation (Simpson et al., 2009). Nonetheless, the threshold to prioritize patients for transplantation is low because the time window for effective treatment is very narrow. Timely referral to a center where liver transplantation can be done is critical. Even if the encephalopathy is mild to begin with, it can progress quickly, with development of cerebral edema.

MEDICAL MANAGEMENT OFACUTE LIVER FAILURE Even if not directly involved with care, it is important for neurologists to have a good understanding of this emerging critical illness and in particular the degree of

650

S. DATAR AND E.F.M. WIJDICKS

Table 44.2 King’s College criteria (adapted from Lee et al., 2012) Acetaminopheninduced ALF

Strongly consider OLT listing if: ● Arterial lactate > 3.5 mmol/L after early fluid resuscitation List for OLT if: ● pH < 7.3 OR ● Arterial lactate > 3.0 mmol/L after adequate fluid resuscitation List for OLT if all three occur within a 24-hour period: ● Presence of grade 3 or 4 hepatic encephalopathy ● INR > 6.5 ● Creatinine > 3.4 mg/dL Nonacetaminophen- List for OLT if: induced ALF INR > 6.5 and encephalopathy present (irrespective of grade) OR Any three of the following (encephalopathy present; irrespective of grade): ● Age < 10 or > 40 years ● Jaundice for > 7 days before development of encephalopathy ● INR  3.5 ● Serum bilirubin  17 mg/dL ● Unfavorable etiology, such as Wilson disease, idiosyncratic drug reaction, seronegative hepatitis ALF, acute liver failure; OLT, orthotopic liver transplantation; INR, international normalized ratio.

coagulopathy in ALF. By definition, patients with ALF are coagulopathic and thrombocytopenic. Correction of INR is not indicated unless the patient is actively bleeding or an invasive procedure – which may include a lumbar puncture in selected cases – is planned. Fresh frozen plasma (FFP) has been used for decades to correct the coagulopathy before invasive procedures. However, it is associated with the drawbacks of transfusion reactions including transfusion-related acute lung injury (TRALI) and volume overload. Plasma infusion alone may not adequately correct severely elevated INR and with the risk of volume overload, plasmapheresis may be an option (Munoz et al., 1989). Recently, factor VIIa has been explored for this purpose. In one study, factor VIIa was used in patients with ALF undergoing procedures and the authors found it to be more effective over FFP in transiently controlling the parameters of coagulation (Shami et al., 2003). There remains a risk of thromboembolic complications with the use of factor VIIa (myocardial infarction and portal vein thrombosis) (Pavese et al., 2005). However, none of these concerns were found in a study of 11 patients who received factor

VIIa for placement of ICP monitors, once again in ALF, reported no hemorrhagic or thrombotic complications (Le et al., 2010). Further randomized studies are needed to prove the safety, efficacy, and cost-effectiveness of factor VIIa in this group of patients; however, it remains an available option. Precise guidelines for safe coagulation parameters for the performance of invasive procedures in patients with ALF do not exist. A common practice is to keep the INR below 1.5. Vitamin K (10 mg intravenously) should be routinely administered since vitamin K deficiency has been reported in patients with ALF (Pereira et al., 2005). Some investigators have used thromboelastography (TEG) and demonstrated that despite elevated INR, most patients with ALF maintain normal hemostasis, mechanisms of which include an increase in clot strength with increasing severity of liver injury, increased factor VIII levels, and a reduction in pro- and anticoagulant proteins (Stravitz et al., 2012). Therefore, the actual bleeding risk based on elevated INR may be less than expected (Lee et al., 2012). Similar to INR, correction of platelets is not indicated unless an invasive procedure is planned or there is evidence of active bleeding. Platelet counts of > 10 000/mm3 are generally well tolerated, with a preference of > 20 000/ mm3 in the setting of active infection/sepsis. Transfusion should be considered for patients with active bleeding and platelet counts < 50 000/mm3. If an invasive procedure is planned, then counts between 50 000 and 70 000/mm3 are acceptable (Munoz et al., 2009). Lactulose has not been formally tested in a randomized controlled fashion in ALF; however, we have extensive experience with its use in chronic liver failure to treat hyperammonemia. Thus, combined with the fact that ammonia likely plays a central role in the pathogenesis of encephalopathy and cerebral edema, in theory lactulose may be beneficial in these patients. The dose should be titrated to produce 2–3 bowel movements, but not to the point of producing diarrhea. A small retrospective study showed no difference in overall outcome, but a small increase in survival time in those who received lactulose was observed (Jalan, 2005; Lee et al., 2012). One concern with its use, however, is distension of the bowel and interference with the surgical field during liver transplantation. Fever due to infection can worsen ICP. ALF increases the risk of infection by bacterial and fungal agents. Routine surveillance cultures should be drawn in all patients and treatment with antibiotics should be instituted at the first sign of infection/systemic inflammatory response syndrome (SIRS). Studies suggest an association between infection/SIRS and progression to deeper stages of encephalopathy (Rolando et al., 2000; Vaquero et al., 2003).

NEUROLOGIC MANIFESTATIONS OF ACUTE LIVER FAILURE 651 Another important factor in management of ICP encephalopathy; nevertheless, it has an important role in is maintenance of adequate mean arterial pressure patients with fulminant hepatic failure who have received (MAP). Patients are often intravascularly volume repeated doses of benzodiazepines. This may help cordepleted, either from poor oral intake or from third spacrectly stage hepatic encephalopathy, which is relevant ing. In addition, they may have low peripheral vascular when emergency transplantation is being considered. resistance from vasodilation. Hypotension can decrease Other potential mechanisms of hepatic encephalopathy cerebral blood flow (CBF) and increase the risk of cereare mercaptans, short-chain fatty acids, and depletion bral ischemia. Hypotensive patients should be volume of neurotransmitters, such as norepinephrine and resuscitated initially with normal saline followed by isodopamine. tonic sodium bicarbonate if acidosis develops. Patients Cerebral edema is a well-known complication of ALF may need norepinephrine for the support of blood pres(Ware et al., 1971), mechanisms of which are partially sure to maintain a MAP of at least 75 mmHg or cerebral understood (Blei and Larsen, 1999; Rao and Norenberg, perfusion pressure (CPP) of at least > 60 mmHg, prefer2001; Butterworth, 2003; Jalan, 2005). Ammonia must ably between 60 and 80 mmHg if that can be achieved. play an important role in the pathogenesis of cerebral Vasopressin, or its analog terlipressin, can be considedema although there may be downstream effects. Durered to reduce the need for high doses of norepinephrine, ing ALF, hyperammonemia has been shown to increase which can cause intense peripheral vasoconstriction and the risk of cerebral edema (Clemmesen et al., 1999). tissue ischemia. Although terlipressin produces systemic Patients rarely develop hepatic encephalopathy with artevasoconstriction, it produces cerebral vasodilatation and rial ammonia concentration less than 75 mmol/L (Bernal may inadvertently increase cerebral blood flow (CBF), et al., 2007). A level > 100 mmol/L at admission is an indethereby having additional benefit. One study comparing pendent risk factor for the development of high grade terlipressin and noradrenaline showed little effect on hepatic encephalopathy (Bernal et al., 2007). Conversely ICP while increasing CPP (Eefsen et al., 2007), but there a level of > 200 mmol/L is strongly associated with cerewas a suggestion of increase in ICP in one study bral edema. (Shawcross et al., 2004). It should be used with extreme Under normal circumstances, ammonia which is procaution in patients with cerebral edema and only if ICP duced in the gut is metabolized by the liver to urea and can be monitored. glutamine. However, when the liver fails, ammonia starts building up and is then metabolized in tissues such as the brain and skeletal muscle. Ammonia crosses the MANAGEMENT OF CEREBRAL EDEMA blood–brain barrier by diffusion. The glutamine hypothAmmonia has been implicated in the pathogenesis of esis proposes that astrocytes which are the sites of hepatic encephalopathy, perhaps through an imbalance ammonia detoxification in the brain metabolize excess between inhibitory and excitatory neurotransmitters. ammonia to glutamine (Blei and Larsen, 1999; Blei, The g-aminobutyric acid (GABA) neurotransmitter 2001; Jalan, 2005). Glutamine, being osmotically active, system, an inhibitory system, possibly plays a prominent causes the astrocytes to swell. This process takes place role (Grimm et al., 1988; Bassett et al., 1987; Jones et al., rapidly during ALF, thereby preventing cellular adapta1989; Jones et al., 1990; Lavoie et al., 1990). GABA tion to the changes in osmolality, a process which does causes hyperpolarization of neuronal membrane through occur with chronic ammonia elevation as seen in liver ciropening of chloride channel and influx of chloride inside rhosis. Electron microscopy has demonstrated marked the cells. Barbiturates and benzodiazepines potentiate this swelling of the perivascular astroglial foot processes action. It has been observed in animal models that hepatic (Kato et al., 1992). encephalopathy has many similarities to encephalopathy Impaired cerebral autoregulation with massive liver induced by drugs that potentiate GABA action. It appears necrosis has been demonstrated using rat models that the degree of hepatic encephalopathy is correlated (Dethloff et al., 2008). Patients with cerebral edema with cerebrospinal fluid (CSF) levels of GABA. Furtherand intracranial hypertension have been shown to have more, flumazenil, a benzodiazepine antagonist, has been higher cerebral blood flow (CBF) (Aggarwal et al., shown to improve the encephalopathy at least in some 1994; Durham et al., 1995). Alterations in cerebral hemocases, if not all (Bassett et al., 1987; Grimm et al., 1988; dynamics result from locally induced cerebral hyperJones et al., 1990). These findings have led to the suggesemia. Other mechanisms that have been implicated tion that an endogenous substance with benzodiazepineinclude production of nitric oxide (Blei and Larsen, like properties contributes to the neuropsychiatric 1999). There is increasing evidence that this increase in manifestations of hepatic encephalopathy (Jones et al., CBF is of importance for the development of cerebral 1989). However, based on limited data, flumazenil edema and intracranial hypertension (Raghavan and cannot be recommended for all patients with hepatic Marik, 2006) (Fig. 44.4).

652

S. DATAR AND E.F.M. WIJDICKS

Fig. 44.4. Illustration of the pathophysiologic mechanisms of cerebral edema in acute liver failure. Gln, glutamine; RNOS, reactive nitrogen oxide species; ICP, intracranial pressure. (Reproduced by permission of the Mayo Foundation for Medical Education and Research. All rights reserved.)

Dysfunction of the sodium-potassium adenosine triphosphate (ATPase) pump also likely plays a role. Inhibition of enzyme a-ketoglutarate dehydrogenase (a key enzyme in the Krebs cycle) by ammonia impairs generation of ATP and increases anaerobic metabolism. Cerebral microdialysis studies in patients with ALF revealed elevated lactate in patients with high ICP, which further increased before surges of ICP (Tofteng et al., 2002). Deficits in the ability of astrocytes to take up glutamate from the extracellular space may lead to excitotoxicity of neuronal and glial glutamate receptors and glial dysfunction (Norenberg, 1998). The observation that hemodynamic status as well as IH improves after removal of native liver during transplantation raises the possibility of necrotic liver being the source of vasoactive cytokines inducing the hemodynamic changes. Finally, aquaporin-4, a molecule which modulates brain water transport, may play a role, and inhibition of this water channel may provide a new therapeutic option for reducing brain edema (Manley et al., 2000). The incidence of cerebral edema increases with increasing grades of encephalopathy (Wijdicks et al., 1995), reaching 75% or more with grade IV encephalopathy (Munoz, 1993). The subsequent course depends upon the volume of brain tissue in relation to the volume of the cranial vault. As alluded to earlier, almost half of the cases of ALF occur with paracetamol (acetaminophen) overdose and these patients are relatively young (in their 30s to 40s). They have less or no cerebral atrophy, which can give rise to rapid increases in ICP with small increases in the volume of intracranial contents

due to cerebral edema (Monro-Kellie hypothesis) (Mokri, 2001). Untreated, this can lead to cerebral herniation, vascular compression, and cerebral infarction, which result in more edema and set up a vicious cycle. The eventual outcome can be fatal. Treatment of cerebral edema follows traditional methods of ICP management. Any underlying factors which can worsen the ICP, such as agitation, coughing, frequent turning, endotracheal suctioning and severe hypertension, need to be eliminated. In the emergency setting, it is important to raise the head of the bed to at least 30 and to keep the neck straight, to facilitate venous return from the brain. If the patient is intubated, then hyperventilation can be used as a temporary measure, maintaining partial pressure of carbon dioxide (pCO2) between 28 and 35 mmHg (Wijdicks, 2009). The resulting hypocapnic alkalosis causes potent cerebral vasoconstriction, thereby reducing cerebral blood volume and consequently ICP (Laffey and Kavanagh, 2002). Intracranial pressure may decrease transiently at the expense of cerebral perfusion. In addition, vasoconstriction induced by hypocapnia can decrease regional cerebral blood flow and inflict secondary ischemic brain injury. Not all patients are pCO2 responders and moreover the effect is short lived. Increasing evidence suggests that hypocapnia can induce substantial adverse effects, including cardiac arrhythmias, myocardial ischemia, and bronchospasm to mention a few. Thus, therapeutic hypocapnia should generally be limited to emergency management of lifethreatening increases in intracranial pressure (Laffey and Kavanagh, 2002). Seizures increase ICP (Gabor et al., 1984; Shah et al., 2007) and when they occur, they must be promptly

NEUROLOGIC MANIFESTATIONS OF ACUTE LIVER FAILURE 653 controlled. Both focal and generalized seizures are edema, especially in early stages (Munoz et al., 1991). known to occur in these patients even in the absence Intracranial hypertension is of particular concern during of radiographically detectable structural brain injury. orthotopic liver transplantation (OLT), when rapid shifts Traditionally phenytoin has been used for the treatment in electrolytes and hemodynamics can cause large fluctuof seizures. As subclinical seizures also occur and can ations in ICP. worsen cerebral edema, there is a suggestion that proHence, a high index of suspicion must be mainphylactic phenytoin may be of benefit. One study indeed tained, with a low threshold to use ICP monitoring in showed benefit with prophylactic phenytoin administrapatients who are at increased risk of cerebral edema. tion (Ellis et al., 2000). However, one small trial failed to According to an update released by the American show any improvement in survival or cerebral edema Association for the Study of Liver Diseases (AASLD) with prophylactic use of phenytoin (Bhatia et al., in 2011, ICP monitoring is recommended in ALF 2004). Hence seizure prophylaxis with phenytoin is curpatients with high grade hepatic encephalopathy, in cenrently not recommended (Lee et al., 2012). ters with expertise in ICP monitoring, in patients awaitICP monitoring in cases of ALF is currently controing and undergoing liver transplantation (Lee et al., versial. Some studies have suggested that ICP monitor2012). Monitoring is usually started around the time ing is clinically useful in ALF despite the risk of of intubation for worsening encephalopathy and should hemorrhage (Lidofsky et al., 1992). be continued at least 1–2 days postoperatively, followOne study in 1993 reported a complication rate ing liver transplantation. approaching 20% (Blei et al., 1993), but the numbers vary Hyperosmolar therapy can be given following or based on the type and location of the ICP monitoring simultaneous to the above measures for the treatment device. Epidural location had the lowest complication of ICP (Bhardwaj and Ulatowski, 2004b; Diringer and rate (3.8%). Subdural and parenchymal locations have Zazulia, 2004; Suarez, 2004; Aiyagari et al., 2006; more benefit in regards to accuracy; however, they also Wakai et al., 2007). These solutions increase osmolality have a higher rate of complications (20% and 22%, of the serum. This facilitates osmotic movement of respectively) (Blei et al., 1993). Ventricular catheters water out of the brain tissue according to Starling’s are rarely, if ever, used. In general, epidural catheters law and thereby helps reduce cerebral edema. Mannitol provide a relatively safe way of monitoring ICP although in addition also results in osmotic diuresis thereby facilthey are not as accurate as subdural and parenchymal itating excretion of excess sodium and water out of the devices. One survey (Vaquero et al., 2005) evaluated body. The usual first dose of mannitol is 1 g/kg. Addi332 patients across 24 centers constituting the US ALF tional doses of 0.5 g/kg can be given as needed while study group. ICP monitoring was used in 92 (28%) of monitoring serum osmolality. The usual trigger for the patients and was strongly associated with referral administration is ICP elevation to > 20 mmHg for for liver transplantation. The rate of intracranial hemor>5 min. Mannitol effectively reduces ICP within rhage was 10.3% in a subset of 52 patients, with half of 10–20 minutes of infusion and the effect may last up the complications being incidental radiologic findings. to 4–6 hours. As mannitol induces diuresis, it is also Survival at 30 days in both monitored and nonmonitored important to maintain relative euvolemia as volume patients post-transplantation was similar. However, this depletion in the setting of elevated ICP can lower CBF study was retrospective and it is possible that monitoring and increase ischemic complications. was undertaken in sicker patients. To date, although Mannitol was used in 17 out of 34 patients with ALF proof of improved survival with ICP monitoring is lackand was found to resolve cerebral edema with greater ing, many liver transplant surgeons, hepatologists, and frequency than in those patients who did not receive it neurointensivists consider it a useful tool and, despite (Canalese et al., 1982). Problems arise with its use in some increased risk of hemorrhage, would strongly the presence of renal failure as mannitol can lead to volfavor its use. ume overload which may necessitate the use of dialysis However, the more difficult question is to decide to remove three times the given volume, 15–30 minutes which patient to select for ICP monitoring. In other words, following infusion. Mannitol has traditionally been used who will benefit most from aggressive treatment of elecautiously when the osmolality reaches > 320 mOsm for vated ICP? As pointed out earlier (Bernal et al., 2007), fear of toxicity, especially in renal failure. This concern it was found that an ammonia level of > 200 mmol/L is comes from a small study in which high doses of mannistrongly associated with cerebral edema. Other factors tol were used as continuous infusions. Crossing the which also increase the risk of cerebral edema are grade 320 mOsm threshold may probably be safe (Diringer III or IV encephalopathy, acute renal failure, and the need and Zazulia, 2004), but caution is advised as patients for vasopressors to maintain MAP. Furthermore, CT scan with ALF are already at risk of acute renal failure from of the brain may not reliably demonstrate evidence of hepatorenal syndrome.

654

S. DATAR AND E.F.M. WIJDICKS

Alternatively, hypertonic saline can be used. Induction and maintenance of hypernatremia can reduce the incidence and severity of intracranial hypertension in patients with ALF (Murphy et al., 2004). The ability of an intact blood–brain barrier (BBB) to exclude a given compound is expressed as reflection coefficient (zero ¼ freely permeable and 1 ¼ completely impermeable) (Schell et al., 1996; Paczynski, 1997). Compounds with reflection coefficient approaching 1 are theoretically better osmotic agents because they are completely excluded by an intact BBB and therefore are less likely to exhibit rebound cerebral edema. Sodium has a reflection coefficient of 1.0 for the BBB, hence in theory, hypertonic saline is an effective dehydrating agent (mannitol has reflection coefficient of 0.9) and reduces brain water content in humans and animal models (Suarez, 2004). Other mechanisms which may possibly play a role include enhanced microvascular blood flow and maintenance of integrity of BBB (Raghavan and Marik, 2006). In ALF patients at high risk of cerebral edema, induced hypernatremia in the range of 145–155 mEq/L with hypertonic saline is currently recommended (Lee et al., 2012). This can be achieved either by bolus doses or continuous infusion of hypertonic saline. The different available concentrations are 3%, 7.5%, 10%, and 23.4%. Potential complications with hypertonic saline include transient hypotension, pulmonary edema and heart failure, hypokalemia, hyperchloremic acidemia, coagulopathy, intravascular hemolysis, myelinolysis (only with rapid overcorrection of pre-existing hyponatremia), and encephalopathy (Worthley et al., 1988; Bhardwaj and Ulatowski, 2004a). Phlebitis can occur if peripheral venous access is used for infusion. Serum sodium level has to be monitored when hyperosmolar therapy is used. It is generally accepted that hypertonic saline should not be used if the serum sodium level is > 160 mEq/L. In a study of 600 patients in the neurointensive care unit, there was an independent increase in mortality in patients with serum sodium > 160 mEq/L (Aiyagari et al., 2006). In the same study, the incidence of renal failure was also high in patients with hypernatremia. Whether to choose mannitol or hypertonic saline depends upon the clinical situation. No study has proven superiority of one therapy over the other. Propofol, a highly lipophilic agent, has been used extensively in the ICU for sedation of mechanically ventilated patients (Marik, 2004). Importantly, its rapid onset and short duration of action (Fulton and Sorkin, 1995) makes it easier to follow neurologic status of comatose patients, as wakefulness returns rapidly following discontinuation of infusion. Propofol has been reported to reduce cerebral metabolic rate, cerebral blood flow, and ICP (Vandesteene et al., 1988;

Newman et al., 1995; Ergun et al., 2002; Jalan, 2005). Its pharmacokinetic properties and protein binding remain largely unaffected even in patients with liver failure (Servin et al., 1988a, b; Veroli et al., 1992; Costela et al., 1996). Propofol may also have a neuroprotective role mediated through GABA receptors (Ito et al., 1999) and potentially through its antioxidant properties (Sagara et al., 1999). In seven patients with ALF treated with an infusion of 50 mg/kg/min, ICP remained within normal limits in six (Wijdicks and Nyberg, 2002). Propofol has favorable pharmacokinetics and along with its potential to reduce ICP (Merlo et al., 1991; Watts et al., 1998; Wijdicks and Nyberg, 2002) in patients with elevated intracranial pressure, it should be considered as an option for this purpose, even perhaps as a first-line agent. The suggested dose is 1–3 mg/kg/hour. A major side-effect is hypotension, which is usually transient following a bolus. Another rare but potentially lifethreatening complication, when used at high doses of 5 mg/kg/hour, is propofol infusion syndrome (PRIS), a state of severe metabolic acidosis and circulatory collapse. However, this complication is rare, predominantly seen at high doses and in younger patients, and should not preclude its use. Besides, patients with ALF would only need it for short period of time. Barbiturates are an option in cases resistant to hyperosmolar therapy. This is usually achieved with pentobarbital or pentothal infusion. Some experience has been published and in one study (Forbes et al., 1989) thiopental coma was induced in 13 cases of fulminant hepatic failure with intracranial hypertension unresponsive to other modes of therapy. All had developed acute renal failure. ICP was measured by extradural transducers. The dosage of thiopental was adjusted incrementally until intracranial pressure was normalized or hypotension developed. ICP was reduced by a median dose of 250 mg of thiopental given over 15 minutes. Eight cases needed continuing infusion. Five patients made complete recovery and there were only three deaths from IH. Side-effects included minor hypotension controlled by dose reduction. It should be recognized that barbiturate clearance is markedly reduced in patients with ALF precluding neurologic assessment for prolonged periods of time, thus making it a less attractive option to treat IH. A common dose of thiopental is 4 mg/kg loading dose followed by infusion starting at 1 mg/kg/hour with gradual upward titration until ICP control is established or hypotension develops. A common method of monitoring these patients is EEG, as patients undergoing barbiturate coma have no clinical examination. Dose can be titrated up until burst suppression is achieved on EEG. Mild hypothermia (35 C) in rat models of acute liver failure was found to prevent the development of brain edema. A proposed mechanism of this action was

NEUROLOGIC MANIFESTATIONS OF ACUTE LIVER FAILURE 655 inhibition of blood–brain transfer of ammonia at least in the native liver until its function recovers. This way part from reduced cerebral blood flow (Butterworth, immunosuppression can be limited to the time period 2001; Chatauret et al., 2001). In humans, several unconfor which the auxiliary liver is in place. In this study, trolled studies suggest effective control of brain edema regeneration of the native liver occurred in 11 of 15 and intracranial hypertension with the induction of hypopatients and in 8 of 10 survivors. Six of these eight thermia (core temperature 32–35 C) (Jacob et al., 2009; patients had permanently stopped immunosuppressant Stravitz and Larsen, 2009). Potential deleterious effects therapy (Jaeck et al., 2002). Similar findings were of hypothermia include increased risk of infection, coagreported in one other study (Chenard-Neu et al., 1996). ulation disturbance, and cardiac arrhythmias. Living donor liver transplantation (LDLT) has also been attempted. Right lobe LDLT improves survival of patients with acute liver failure. In one study 7 out of OUTCOME 10 patients with ALF who received LDLT survived Although liver transplantation may seem a necessary (Campsen et al., 2008). The median period of followstep in most patients, only 25% actually receive it, either up was 5 years. Five of the 10 living donors had a total because of spontaneous recovery from paracetamol of seven post-transplant complications. Currently LDLT (acetaminophen) toxicity or due to failure to find a suitis rarely performed. The transplant team must consider able liver match. Usually patients are grafted within the associated complications related to the donor before 3 days of hospitalization as the window of opportunity proceeding. is small (Lee and Wijdicks, 2008). It is important to quickly and accurately predict which ARTIFICIAL LIVERS patients have a good chance of recovering spontaneously Limited experience in humans supports the use of hypoand which patients will need emergency liver transplanthermia as a bridge to liver transplantation, cooling to a tation. This distinction is important as the patients who core temperature of 32–33 C, in cases of intracranial will eventually recover spontaneously can be spared hypertension which are refractory to mannitol or hyperthe hazards of transplantation and lifelong immunosuptonic saline (Jalan et al., 2004). pression. This will also save precious and scarce organs Artificial liver support systems can work as a bridge for those who will need them the most. In a large study in for patients with severe liver disease, either to give them 2002, etiology of acute liver failure was one of the time to recover or to go to transplant. important predictors of outcome (Ostapowicz et al., Many attempts have been made to develop artificial 2002). Transplant free survival was more than 50% with liver support devices (ALSD). These include nonbiologic paracetamol (acetaminophen), hepatitis A, shock liver, ones such as hemodialysis, charcoal hemoperfusion, or pregnancy-related disease, whereas it was less than selective plasma filtration, plasma exchange, hemodiad25% for all other etiologies. Patients with grade III or sorption, albumin dialysis, and biologic ones such as IV hepatic encephalopathy are less likely to survive withwhole liver perfusion, liver cell transplantation, and out transplantation than those with grade I or grade II. A bioartificial liver support. model for end stage liver disease (MELD) score has been Use of albumin-collodion microencapsulated actitested in ALF from paracetamol (acetaminophen) toxicvated charcoal (ACAC) hemoperfusion in fulminant ity but was not found to be superior to INR or King’s hepatic coma showed promising results in rat models College criteria (KCC) (Schmidt and Larsen, 2007). (Chirito et al., 1977). However, randomized trial using The Clichy criteria developed in a cohort of French charcoal hemoperfusion in fulminant hepatic failure in patients used factor V level measurements to predict humans did not show any benefit (O’Grady et al., 1988). outcome. Serum factor V level of less than 20% in The molecular adsorbent recycling system (MARS) patients younger than 30 years of age or less than 30% used a membrane impermeable to proteins but capable in any patient with grade 3–4 encephalopathy predicted of exchanging water soluble and protein-bound toxins mortality with a positive predictive value of 82% and a and a recycled protein containing dialysate (Stange negative predictive value of 98%. At present, KCC is et al., 1999). A meta-analysis of studies using MARS the most commonly used prognostic tool, with a sensitivdid not show significant survival benefit in patients with ity of 68–69% and specificity of 82–92% (Bailey et al., liver failure when compared with standard medical ther2003; McPhail et al., 2010). apy (Khuroo and Farahat, 2004). Randomized trials are Auxiliary partial orthotopic liver transplantation was needed to define the role of MARS in the treatment of tested in both adults and children in a small study done in these patients. France and was found to be a feasible option. In this A bioartificial liver (BAL) is a bioreactor charged procedure, the native liver is left in place and a graft with liver cells that is connected externally to the (auxiliary liver) is implanted to temporarily support

656

S. DATAR AND E.F.M. WIJDICKS

circulation. In animal models they have been shown to prolong survival significantly in comparison to the standard treatment (Flendrig et al., 1999; Hochleitner et al., 2006; Kawazoe et al., 2006). Five systems have been tested clinically. HepatAssist™ is a BAL based on porcine hepatocytes. In a prospective randomized controlled trial it demonstrated safety and improved survival in patients with fulminant/subfulminant hepatic failure (Demetriou et al., 2004). Another bridging method is hepatocyte infusion. Using advanced femoral artery catheter, cryopreserved human hepatocytes are infused into the splenic artery. These hepatocytes appear to survive and even proliferate in the spleen, resulting in a 40–60% reduction of ammonia levels (Strom et al., 1997). Artificial support systems have not been shown to reliably reduce mortality in the setting of ALF (Kjaergard et al., 2003; McKenzie et al., 2008). The standard of care continues to be urgent cadaveric liver transplantation. The concept of a bioengineered liver is only in a very preliminary and experimental phase (Chan et al., 2004; Diekmann et al., 2006; Linke et al., 2007; Fiegel et al., 2008; Ishii et al., 2008).

REFERENCES Aggarwal S, Kramer D, Yonas H et al. (1994). Cerebral hemodynamic and metabolic changes in fulminant hepatic failure: a retrospective study. Hepatology 19: 80–87. Aiyagari V, Deibert E, Diringer MN (2006). Hypernatremia in the neurologic intensive care unit: how high is too high? J Crit Care 21: 163–172. Averbuch-Heller L, Meiner Z (1995). Reversible periodic alternating gaze deviation in hepatic encephalopathy. Neurology 45: 191–192. Bailey B, Amre DK, Gaudreault P (2003). Fulminant hepatic failure secondary to acetaminophen poisoning: a systematic review and meta-analysis of prognostic criteria determining the need for liver transplantation. Crit Care Med 31: 299–305. Bassett ML, Mullen KD, Skolnick P et al. (1987). Amelioration of hepatic encephalopathy by pharmacologic antagonism of the GABAA-benzodiazepine receptor complex in a rabbit model of fulminant hepatic failure. Gastroenterology 93: 1069–1077. Belay ED, Bresee JS, Holman RC et al. (1999). Reye’s syndrome in the United States from 1981 through 1997. N Engl J Med 340: 1377–1382. Bernal W, Hall C, Karvellas CJ et al. (2007). Arterial ammonia and clinical risk factors for encephalopathy and intracranial hypertension in acute liver failure. Hepatology 46: 1844–1852. Bhardwaj A, Ulatowski JA (2004a). Hypertonic saline solutions in brain injury. Curr Opin Crit Care 10: 126–131. Bhardwaj A, Ulatowski JA (2004b). Hypertonic saline solutions in brain injury. Curr Opin Crit Care 10: 126–131.

Bhatia V, Batra Y, Acharya SK (2004). Prophylactic phenytoin does not improve cerebral edema or survival in acute liver failure – a controlled clinical trial. J Hepatol 41: 89–96. Bismuth H, Samuel D, Gugenheim J et al. (1987). Emergency liver transplantation for fulminant hepatitis. Ann Intern Med 107: 337–341. Blei AT (2001). Pathophysiology of brain edema in fulminant hepatic failure, revisited. Metab Brain Dis 16: 85–94. Blei AT, Larsen FS (1999). Pathophysiology of cerebral edema in fulminant hepatic failure. J Hepatol 31: 771–776. Blei AT, Olafsson S, Webster S et al. (1993). Complications of intracranial pressure monitoring in fulminant hepatic failure. Lancet 341: 157–158. Boks AL, Brommer EJ, Schalm SW et al. (1986). Hemostasis and fibrinolysis in severe liver failure and their relation to hemorrhage. Hepatology 6: 79–86. Brandsaeter B, Hockerstedt K, Friman S et al. (2002). Fulminant hepatic failure: outcome after listing for highly urgent liver transplantation – 12 years experience in the Nordic countries. Liver Transpl 8: 1055–1062. Butterworth RF (2001). Mild hypothermia prevents cerebral edema in acute liver failure. J Hepatobiliary Pancreat Surg 8: 16–19. Butterworth RF (2003). Molecular neurobiology of acute liver failure. Semin Liver Dis 23: 251–258. Campsen J, Blei AT, Emond JC et al. (2008). Outcomes of living donor liver transplantation for acute liver failure: the adult-to-adult living donor liver transplantation cohort study. Liver Transpl 14: 1273–1280. Canalese J, Gimson AE, Davis C et al. (1982). Controlled trial of dexamethasone and mannitol for the cerebral oedema of fulminant hepatic failure. Gut 23: 625–629. Caplan LR, Scheiner D (1980). Dysconjugate gaze in hepatic coma. Ann Neurol 8: 328–329. Chan C, Berthiaume F, Nath BD et al. (2004). Hepatic tissue engineering for adjunct and temporary liver support: critical technologies. Liver Transpl 10: 1331–1342. Chatauret N, Rose C, Therrien G et al. (2001). Mild hypothermia prevents cerebral edema and CSF lactate accumulation in acute liver failure. Metab Brain Dis 16: 95–102. Chenard-Neu MP, Boudjema K, Bernuau J et al. (1996). Auxiliary liver transplantation: regeneration of the native liver and outcome in 30 patients with fulminant hepatic failure – a multicenter European study. Hepatology 23: 1119–1127. Chirito E, Reiter B, Lister C et al. (1977). Artificial liver: the effect of ACAC microencapsulated charcoal hemoperfusion on fulminant hepatic failure. Artif Organs 1: 76–83. Clemmesen JO, Larsen FS, Kondrup J et al. (1999). Cerebral herniation in patients with acute liver failure is correlated with arterial ammonia concentration. Hepatology 29: 648–653. Conomy JP, Swash M (1968). Reversible decerebrate and decorticate postures in hepatic coma. N Engl J Med 278: 876–879. Costela JL, Jimenez R, Calvo R et al. (1996). Serum protein binding of propofol in patients with renal failure or hepatic cirrhosis. Acta Anaesthesiol Scand 40: 741–745.

NEUROLOGIC MANIFESTATIONS OF ACUTE LIVER FAILURE Demetriou AA, Brown RS Jr, Busuttil RW et al. (2004). Prospective, randomized, multicenter, controlled trial of a bioartificial liver in treating acute liver failure. Ann Surg 239: 660–667, discussion 667–670. Dethloff TJ, Knudsen GM, Larsen FS (2008). Cerebral blood flow autoregulation in experimental liver failure. J Cereb Blood Flow Metab 28: 916–926. Detry O, De Roover A, Coimbra C et al. (2007). Cadaveric liver transplantation for non-acetaminophen fulminant hepatic failure: a 20-year experience. World J Gastroenterol 13: 1427–1430. Diekmann S, Bader A, Schmitmeier S (2006). Present and future developments in hepatic tissue engineering for liver support systems: state of the art and future developments of hepatic cell culture techniques for the use in liver support systems. Cytotechnology 50: 163–179. Diringer MN, Zazulia AR (2004). Osmotic therapy: fact and fiction. Neurocrit Care 1: 219–233. Durham S, Yonas H, Aggarwal S et al. (1995). Regional cerebral blood flow and CO2 reactivity in fulminant hepatic failure. J Cereb Blood Flow Metab 15: 329–335. Eefsen M, Dethloff T, Frederiksen HJ et al. (2007). Comparison of terlipressin and noradrenalin on cerebral perfusion, intracranial pressure and cerebral extracellular concentrations of lactate and pyruvate in patients with acute liver failure in need of inotropic support. J Hepatol 47: 381–386. Ellis AJ, Wendon JA, Williams R (2000). Subclinical seizure activity and prophylactic phenytoin infusion in acute liver failure: a controlled clinical trial. Hepatology 32: 536–541. Emond JC, Aran PP, Whitington PF et al. (1989). Liver transplantation in the management of fulminant hepatic failure. Gastroenterology 96: 1583–1588. Ergun R, Akdemir G, Sen S et al. (2002). Neuroprotective effects of propofol following global cerebral ischemia in rats. Neurosurg Rev 25: 95–98. Ferenci P, Lockwood A, Mullen K et al. (2002). Hepatic encephalopathy – definition, nomenclature, diagnosis, and quantification: final report of the working party at the 11th World Congresses of Gastroenterology, Vienna, 1998. Hepatology 35: 716–721. Ficker DM, Westmoreland BF, Sharbrough FW (1997). Epileptiform abnormalities in hepatic encephalopathy. J Clin Neurophysiol 14: 230–234. Fiegel HC, Kaufmann PM, Bruns H et al. (2008). Hepatic tissue engineering: from transplantation to customized cellbased liver directed therapies from the laboratory. J Cell Mol Med 12: 56–66. Flendrig LM, Calise F, Di Florio E et al. (1999). Significantly improved survival time in pigs with complete liver ischemia treated with a novel bioartificial liver. Int J Artif Organs 22: 701–709. Forbes A, Alexander GJ, O’Grady JG et al. (1989). Thiopental infusion in the treatment of intracranial hypertension complicating fulminant hepatic failure. Hepatology 10: 306–310. Fujiwara K, Mochida S (2002). Indications and criteria for liver transplantation for fulminant hepatic failure. J Gastroenterol 37 (Suppl 13): 74–77.

657

Fulton B, Sorkin EM (1995). Propofol. An overview of its pharmacology and a review of its clinical efficacy in intensive care sedation. Drugs 50: 636–657. Gabor AJ, Brooks AG, Scobey RP et al. (1984). Intracranial pressure during epileptic seizures. Electroencephalogr Clin Neurophysiol 57: 497–506. Grimm G, Ferenci P, Katzenschlager R et al. (1988). Improvement of hepatic encephalopathy treated with flumazenil. Lancet 2: 1392–1394. Heubi JE, Daugherty CC, Partin JS et al. (1984). Grade I Reye’s syndrome – outcome and predictors of progression to deeper coma grades. N Engl J Med 311: 1539–1542. Hochleitner B, Hengster P, Bucher H et al. (2006). Significant survival prolongation in pigs with fulminant hepatic failure treated with a novel microgravity-based bioartificial liver. Artif Organs 30: 906–914. Ishii Y, Saito R, Marushima H et al. (2008). Hepatic reconstruction from fetal porcine liver cells using a radial flow bioreactor. World J Gastroenterol 14: 2740–2747. Ito H, Watanabe Y, Isshiki A et al. (1999). Neuroprotective properties of propofol and midazolam, but not pentobarbital, on neuronal damage induced by forebrain ischemia, based on the GABAA receptors. Acta Anaesthesiol Scand 43: 153–162. Jacob S, Khan A, Jacobs ER et al. (2009). Prolonged hypothermia as a bridge to recovery for cerebral edema and intracranial hypertension associated with fulminant hepatic failure. Neurocrit Care 11: 242–246. Jaeck D, Boudjema K, Audet M et al. (2002). Auxiliary partial orthotopic liver transplantation (APOLT) in the treatment of acute liver failure. J Gastroenterol 37 (Suppl 13): 88–91. Jalan R (2005). Pathophysiological basis of therapy of raised intracranial pressure in acute liver failure. Neurochem Int 47: 78–83. Jalan R, Olde Damink SW, Deutz NE et al. (2004). Moderate hypothermia in patients with acute liver failure and uncontrolled intracranial hypertension. Gastroenterology 127: 1338–1346. Jones EA, Skolnick P, Gammal SH et al. (1989). NIH conference. The gamma-aminobutyric acid A (GABAA) receptor complex and hepatic encephalopathy. Some recent advances. Ann Intern Med 110: 532–546. Jones EA, Basile AS, Mullen KD et al. (1990). Flumazenil: potential implications for hepatic encephalopathy. Pharmacol Ther 45: 331–343. Kato M, Hughes RD, Keays RT et al. (1992). Electron microscopic study of brain capillaries in cerebral edema from fulminant hepatic failure. Hepatology 15: 1060–1066. Kawazoe Y, Eguchi S, Sugiyama N et al. (2006). Comparison between bioartificial and artificial liver for the treatment of acute liver failure in pigs. World J Gastroenterol 12: 7503–7507. Khashab M, Tector AJ, Kwo PY (2007). Epidemiology of acute liver failure. Curr Gastroenterol Rep 9: 66–73. Khuroo MS, Farahat KL (2004). Molecular adsorbent recirculating system for acute and acute-on-chronic liver failure: a meta-analysis. Liver Transpl 10: 1099–1106.

658

S. DATAR AND E.F.M. WIJDICKS

Kjaergard LL, Liu J, Als-Nielsen B et al. (2003). Artificial and bioartificial support systems for acute and acute-onchronic liver failure: a systematic review. JAMA 289: 217–222. Laffey JG, Kavanagh BP (2002). Hypocapnia. N Engl J Med 347: 43–53. Lavoie J, Layrargues GP, Butterworth RF (1990). Increased densities of peripheral-type benzodiazepine receptors in brain autopsy samples from cirrhotic patients with hepatic encephalopathy. Hepatology 11: 874–878. Le TV, Rumbak MJ, Liu SS et al. (2010). Insertion of intracranial pressure monitors in fulminant hepatic failure patients: early experience using recombinant factor VII. Neurosurgery 66: 455–458, discussion 458. Lee WM, Wijdicks EF (2008). Fulminant hepatic failure: when the hepatologist meets the neurointensivist. Neurocrit Care 9: 1–2. Lee WM, Squires RH Jr, Nyberg SL et al. (2008). Acute liver failure: Summary of a workshop. Hepatology 47: 1401–1415. Lee WM, Stravitz RT, Larson AM (2012). Introduction to the revised American Association for the Study of Liver Diseases position paper on acute liver failure 2011. Hepatology 55: 965–967. Lidofsky SD, Bass NM, Prager MC et al. (1992). Intracranial pressure monitoring and liver transplantation for fulminant hepatic failure. Hepatology 16: 1–7. Linke K, Schanz J, Hansmann J et al. (2007). Engineered liverlike tissue on a capillarized matrix for applied research. Tissue Eng 13: 2699–2707. McKenzie TJ, Lillegard JB, Nyberg SL (2008). Artificial and bioartificial liver support. Semin. Liver Dis 28: 210–217. McKinney AM, Lohman BD, Sarikaya B et al. (2010). Acute hepatic encephalopathy: diffusion-weighted and fluidattenuated inversion recovery findings, and correlation with plasma ammonia level and clinical outcome. Am J Neuroradiol 31: 1471–1479. McPhail MJ, Wendon JA, Bernal W (2010). Meta-analysis of performance of Kings’s College Hospital Criteria in prediction of outcome in non-paracetamol-induced acute liver failure. J Hepatol 53: 492–499. Madl C, Grimm G, Ferenci P et al. (1994). Serial recording of sensory evoked potentials: a noninvasive prognostic indicator in fulminant liver failure. Hepatology 20: 1487–1494. Manley GT, Fujimura M, Ma T et al. (2000). Aquaporin-4 deletion in mice reduces brain edema after acute water intoxication and ischemic stroke. Nat Med 6: 159–163. Marik PE (2004). Propofol: therapeutic indications and sideeffects. Curr Pharm Des 10: 3639–3649. Merlo F, Demo P, Lacquaniti L et al. (1991). Propofol in single bolus for treatment of elevated intracranial hypertension. Minerva Anestesiol 57: 359–363. Meythaler JM, Varma RR (1987). Reye’s syndrome in adults. Diagnostic considerations. Arch Intern Med 147: 61–64. Mokri B (2001). The Monro-Kellie hypothesis: applications in CSF volume depletion. Neurology 56: 1746–1748. Montalti R, Nardo B, Beltempo P et al. (2005). Liver transplantation in fulminant hepatic failure: experience with 40 adult

patients over a 17-year period. Transplant Proc 37: 1085–1087. Munoz SJ (1993). Difficult management problems in fulminant hepatic failure. Semin Liver Dis 13: 395–413. Munoz SJ, Ballas SK, Moritz MJ et al. (1989). Perioperative management of fulminant and subfulminant hepatic failure with therapeutic plasmapheresis. Transplant Proc 21: 3535–3536. Munoz SJ, Robinson M, Northrup B et al. (1991). Elevated intracranial pressure and computed tomography of the brain in fulminant hepatocellular failure. Hepatology 13: 209–212. Munoz SJ, Stravitz RT, Gabriel DA (2009). Coagulopathy of acute liver failure. Clin Liver Dis 13: 95–107. Murphy N, Auzinger G, Bernel W et al. (2004). The effect of hypertonic sodium chloride on intracranial pressure in patients with acute liver failure. Hepatology 39: 464–470. Newman MF, Murkin JM, Roach G et al. (1995). Cerebral physiologic effects of burst suppression doses of propofol during nonpulsatile cardiopulmonary bypass. CNS Subgroup of McSPI. Anesth Analg 81: 452–457. Norenberg MD (1998). Astroglial dysfunction in hepatic encephalopathy. Metab Brain Dis 13: 319–335. O’Grady J (2007). Modern management of acute liver failure. Clin Liver Dis 11: 291–303. O’Grady JG, Gimson AE, O’Brien CJ et al. (1988). Controlled trials of charcoal hemoperfusion and prognostic factors in fulminant hepatic failure. Gastroenterology 94: 1186–1192. Ostapowicz G, Fontana RJ, Schiodt FV et al. (2002). Results of a prospective study of acute liver failure at 17 tertiary care centers in the United States. Ann Intern Med 137: 947–954. Paczynski RP (1997). Osmotherapy. Basic concepts and controversies. Crit Care Clin 13: 105–129. Pavese P, Bonadona A, Beaubien J et al. (2005). FVIIa corrects the coagulopathy of fulminant hepatic failure but may be associated with thrombosis: a report of four cases. Can J Anaesth 52: 26–29. Pereira SP, Rowbotham D, Fitt S et al. (2005). Pharmacokinetics and efficacy of oral versus intravenous mixed-micellar phylloquinone (vitamin K1) in severe acute liver disease. J Hepatol 42: 365–370. Raghavan M, Marik PE (2006). Therapy of intracranial hypertension in patients with fulminant hepatic failure. Neurocrit Care 4: 179–189. Ranjan P, Mishra AM, Kale R et al. (2005). Cytotoxic edema is responsible for raised intracranial pressure in fulminant hepatic failure: in vivo demonstration using diffusionweighted MRI in human subjects. Metab Brain Dis 20: 181–192. Rao KV, Norenberg MD (2001). Cerebral energy metabolism in hepatic encephalopathy and hyperammonemia. Metab Brain Dis 16: 67–78. Record CO, Chase RA, Hughes RD et al. (1975). Glycerol therapy for cerebral oedema complicating fulminant hepatic failure. Br Med J 2: 540. Rolando N, Wade J, Davalos M et al. (2000). The systemic inflammatory response syndrome in acute liver failure. Hepatology 32: 734–739.

NEUROLOGIC MANIFESTATIONS OF ACUTE LIVER FAILURE Rothstein JD, Herlong HF (1989). Neurologic manifestations of hepatic disease. Neurol Clin 7: 563–578. Russo MW, Galanko JA, Shrestha R et al. (2004). Liver transplantation for acute liver failure from drug induced liver injury in the United States. Liver Transpl 10: 1018–1023. Sagara Y, Hendler S, Khoh-Reiter S et al. (1999). Propofol hemisuccinate protects neuronal cells from oxidative injury. J Neurochem 73: 2524–2530. Schell RM, Applegate RL 2nd, Cole DJ (1996). Salt, starch, and water on the brain. J Neurosurg Anesthesiol 8: 178–182. Schmidt LE, Larsen FS (2007). MELD score as a predictor of liver failure and death in patients with acetaminopheninduced liver injury. Hepatology 45: 789–796. Servin F, Desmonts JM, Farinotti R et al. (1988a). Pharmacokinetics of propofol administered by continuous infusion in patients with cirrhosis. Preliminary results. Anaesthesia 43 (Suppl): 23–24. Servin F, Desmonts JM, Haberer JP et al. (1988b). Pharmacokinetics and protein binding of propofol in patients with cirrhosis. Anesthesiology 69: 887–891. Shah AK, Fuerst D, Sood S et al. (2007). Seizures lead to elevation of intracranial pressure in children undergoing invasive EEG monitoring. Epilepsia 48: 1097–1103. Shami VM, Caldwell SH, Hespenheide EE et al. (2003). Recombinant activated factor VII for coagulopathy in fulminant hepatic failure compared with conventional therapy. Liver Transpl 9: 138–143. Shawcross DL, Davies NA, Mookerjee RP et al. (2004). Worsening of cerebral hyperemia by the administration of terlipressin in acute liver failure with severe encephalopathy. Hepatology 39: 471–475. Simpson KJ, Bates CM, Henderson NC et al. (2009). The utilization of liver transplantation in the management of acute liver failure: comparison between acetaminophen and non-acetaminophen etiologies. Liver Transpl 15: 600–609. Stange J, Mitzner SR, Risler T et al. (1999). Molecular adsorbent recycling system (MARS): clinical results of a new membrane-based blood purification system for bioartificial liver support. Artif Organs 23: 319–330. Stravitz RT, Larsen FS (2009). Therapeutic hypothermia for acute liver failure. Crit Care Med 37: S258–S264. Stravitz RT, Lisman T, Luketic VA et al. (2012). Minimal effects of acute liver injury/acute liver failure on hemostasis as assessed by thromboelastography. J Hepatol 56: 129–136. Strom SC, Fisher RA, Thompson MT et al. (1997). Hepatocyte transplantation as a bridge to orthotopic liver transplantation in terminal liver failure. Transplantation 63: 559–569.

659

Suarez JI (2004). Hypertonic saline for cerebral edema and elevated intracranial pressure. Cleve Clin J Med 71 (Suppl 1): S9–S13. Tofteng F, Jorgensen L, Hansen BA et al. (2002). Cerebral microdialysis in patients with fulminant hepatic failure. Hepatology 36: 1333–1340. Trey C, Davidson CS (1970). The management of fulminant hepatic failure. Prog Liver Dis 3: 282–298. Vandesteene A, Trempont V, Engelman E et al. (1988). Effect of propofol on cerebral blood flow and metabolism in man. Anaesthesia 43 (Suppl): 42–43. Vaquero J, Polson J, Chung C et al. (2003). Infection and the progression of hepatic encephalopathy in acute liver failure. Gastroenterology 125: 755–764. Vaquero J, Fontana RJ, Larson AM et al. (2005). Complications and use of intracranial pressure monitoring in patients with acute liver failure and severe encephalopathy. Liver Transpl 11: 1581–1589. Veroli P, O’Kelly B, Bertrand F et al. (1992). Extrahepatic metabolism of propofol in man during the anhepatic phase of orthotopic liver transplantation. Br J Anaesth 68: 183–186. Wakai A, Roberts I, Schierhout G (2007). Mannitol for acute traumatic brain injury. Cochrane Database Syst Rev CD001049. Ware AJ, D’Agostino AN, Combes B (1971). Cerebral edema: a major complication of massive hepatic necrosis. Gastroenterology 61: 877–884. Watts AD, Eliasziw M, Gelb AW (1998). Propofol and hyperventilation for the treatment of increased intracranial pressure in rabbits. Anesth Analg 87: 564–568. Wijdicks EF (2009). Neurologic complications of acute liver failure. In: Neurologic Complications of Critical Illness, Contemporary Neurology, 3rd edn. Vol. 74. Oxford University Press, New York, pp. 204–217. Wijdicks EF, Nyberg SL (2002). Propofol to control intracranial pressure in fulminant hepatic failure. Transplant Proc 34: 1220–1222. Wijdicks EF, Plevak DJ, Rakela J et al. (1995). Clinical and radiologic features of cerebral edema in fulminant hepatic failure. Mayo Clin Proc 70: 119–124. Williams R (1975). Artificial liver support in fulminant hepatic failure. Bull N Y Acad Med 51: 508–518. Worthley LI, Cooper DJ, Jones N (1988). Treatment of resistant intracranial hypertension with hypertonic saline. Report of two cases. J Neurosurg 68: 478–481. Zacharski LR, Litin EM, Mulder DW et al. (1970). Acute, fatal hepatic failure presenting with psychiatric symptoms. Am J Psychiatry 127: 382–386.

Handbook of Clinical Neurology, Vol. 120 (3rd series) Neurologic Aspects of Systemic Disease Part II Jose Biller and Jose M. Ferro, Editors © 2014 Elsevier B.V. All rights reserved

Chapter 45

Portosystemic encephalopathy KARIN WEISSENBORN* Department of Neurology, Hanover Medical School, Hanover, Germany

HISTORY Scientific interest in the relationship between the liver and the brain dates back to ancient times. The basis for our current understanding of hepatic encephalopathy (HE), however, was provided at the end of the 19th century, when Nencki, Pavlov, and Zaleski observed that dogs developed a behavioral syndrome following the formation of a portocaval shunt (Eck’s fistula). Symptoms worsened after a meat feed and thus the phrase “meat intoxication syndrome” was coined. Nencki and coworkers saw that the arterial ammonia levels in the dogs increased after protein ingestion, accompanied by an increase in urinary ammonia excretion. After ammonia ingestion the dogs died. Analysis of the animals’ brain ammonia content showed a fourfold increase of ammonia compared to normal. Thus, it was concluded that the behavioral changes in the animals were induced by ammonia (Shawcross et al., 2005). At the beginning of the 20th century, clinical observations led to the conclusion that there is a connection between raised ammonia levels in the blood and the development of neuropsychiatric symptoms in patients with severe liver disease (Zieve, 1991). In 1952, Phillips et al. established a regular relationship between raised ammonia levels and the development of HE. Later a detailed analysis of the clinical features of HE was performed by Dame Sheila Sherlock and her coworkers (Sherlock et al., 1954; Sherlock, 1958). It was Sherlock who coined the term “portosystemic encephalopathy” for HE in patients with liver cirrhosis. In 2002, a working party on clinical research in hepatic encephalopathy recommended changes in the terminology and the differentiation of three types of HE (Ferenci et al., 2002): 1.

encephalopathy associated with acute liver failure (type A)

2.

3.

encephalopathy associated with portal-systemic bypass and no intrinsic hepatocellular disease (type B) encephalopathy associated with cirrhosis and portal hypertension (type C).

CLINICAL FINDINGS Portosystemic encephalopathy, or type C hepatic encephalopathy (HE), is characterized by disturbances of cognition, motor abilities, and consciousness. Symptoms range from minimal changes in personality or altered sleep pattern to deep coma (Sherlock et al., 1954). Sleep disturbances and slight behavioral changes are usually overlooked as they are not apparent in the clinical examination, but can be detected only by a directed inquiry. HE may stay in this covert stage for a long time but may also proceed to more severe stages with clinically obvious disturbance of consciousness: mild confusion in grade I, drowsiness, lethargy and disorientation in grade II, somnolence in grade III, and coma, with or without response to painful stimuli, in grade IV (a and b) HE (Fig. 45.1). Cognitive dysfunction is the major problem in minimal HE (mHE) and grades I and II HE. While this is obvious in grades I and II HE, it remains unnoticed with clinical examination in patients with mHE, since the patients’ verbal abilities are preserved (Schomerus and Hamster, 1998). Neuropsychological examinations, however, show deficits in attention and visual perception combined with a reduction in motor speed and accuracy (Weissenborn et al., 2001). The patients are especially impaired in their mechanical skills or in tasks which require sustained attention and quick reaction. Schomerus and Hamster (1998) were able to show that mHE decreases the

*Correspondence to: Karin Weissenborn, M.D., Department of Neurology, Hannover Medical School, Carl-Neuberg-Str. 1, 30625 Hanover, Germany. Tel: þ49-511-532-2339, Fax: þ49-511-532-3115, E-mail: [email protected]

662

K. WEISSENBORN

Fig. 45.1. Classification of hepatic encephalopathy according to alterations in attention and alertness (West Haven Criteria).

working ability of blue collar workers, while white collar workers are not impaired unless HE becomes clinically overt. It has repeatedly been shown that minimal HE also affects driving ability (Schomerus et al., 1981; Watanabe et al., 1995; Wein et al., 2004; Bajaj et al., 2009; Kircheis et al., 2009). In patients with grade 1 HE lack of attention and psychomotor slowing is clinically obvious. In patients with grade 2 HE cognitive dysfunction is accompanied by a further decrease of alertness and disorientation for time and sometimes also for place and situation. Motor symptoms such as asterixis, dysarthria, ataxia, tremor, hypokinesia, muscular rigidity, and posture abnormalities are hallmarks of hepatic encephalopathy. They are usually attributed to grade 2 HE. In practice, however, these symptoms are detectable by subtle neurologic assessment even in patients with mild HE (Krieger et al., 1996; Spahr et al., 1996). Speech disturbances and writing disabilities belong to the first symptoms of HE. Initially speech is monotonous and slow. With increasing ataxia speech becomes slurred, dysarthric. In stuporous patients dysphasia occurs combined with perseveration. Writing disabilities are traditionally used for follow-up examinations. Initially omission of single letters, reversal of order and misspellings are common. Later the patients become unable to sign their names or to move the pencil from left to right. Hyperreflexia and ankle clonus is a characteristic finding in grade 3 HE, but can be observed also in patients with mild HE, in principle.

Personality changes are frequent. Mood fluctuates. While some of the patients appear euphoric the majority present with mild to moderate depression. Confusion, bizarre behavior, and paranoid symptoms are common in grade 2–3 HE. In addition, hallucinations may be observed (Sherlock et al., 1954; Summerskill et al., 1956). Rare symptoms of type C HE are headaches, blurred or double vision, focal neurologic signs such as hemiparesis and epileptic seizures (Sherlock et al., 1954; Cadranel et al., 2001). While type C HE occurs episodically in the majority of patients, some show chronic progressive symptoms. In general, these patients have extensive portosystemic shunts either developed in the course of liver cirrhosis or following shunt operation or TIPSS (transjugular intrahepatic portosystemic stent shunt). Occasionally chronic progressive HE can be observed as a complication of a congenital intrahepatic portosystemic venous shunt despite normal liver function (type B HE) (Da Rocha et al., 2004). Read et al. (1967) observed five different syndromes of chronic persistent HE in patients who had undergone shunt operations: “1. predominant psychiatric disorder, 2. paraplegia, 3. cerebellar and basal ganglia disorder, 4. paroxysmal disorders of consciousness or muscle spasms, 5. localized or generalized cortical disease.” While the psychiatric disorder and paraplegia occurred within several months up to 1½ years after shunt operation in their patients, cerebellar and basal ganglia disorder was observed 7 and 8 years after the procedure.

PORTOSYSTEMIC ENCEPHALOPATHY 663 Nowadays, paraplegia in patients with chronic liver be addressed in every individual case before neuropsychidisease is referred to as hepatic myelopathy (HM) atric symptoms are related to HE (Cordoba et al., 2010). (Campellone et al., 1996; Lewis et al., 2000). In most Kril and Butterworth (1997) noted the high prevalence cases HM will develop after several episodes of HE. of unexpected evidence for thiamine deficiency in Upper extremities are affected minimally, if at all. Brisk patients who died with HE, even in patients with nonaltendon reflexes and increased tone of the lower extremcoholic cirrhosis. Thus, Wernicke’s encephalopathy has ities are the predominant findings. Plantar reflexes to be considered and thiamine should be given in case may be flexor, even in patients with ankle clonus. Disturof doubt. bances of sensation or bladder function are uncommon. After months of progressing disability most patients DIAGNOSIS OF MINIMAL HEPATIC either depend upon an assistive device or are confined ENCEPHALOPATHY to a wheelchair. For unknown reasons, most patients While the pattern of cerebral dysfunction in minimal described are men. hepatic encephalopathy (mHE) has been well known Acquired hepatocerebral degeneration (AHD) is a for decades (e.g., Zeegen et al., 1970; Rehnstr€om further intriguing neurologic syndrome associated with et al., 1977; Rikkers et al., 1978; Gilberstadt et al., extensive portosystemic shunting. While patients with 1980; Tarter et al., 1984), the diagnostic approach is conAHD suffer the whole spectrum of cerebral dysfunction troversial. People working on HE seem to be evenly present in HE, cognitive dysfunction and alteration divided: some plead for the use of a neuropsychological of consciousness are by far less prominent than test battery that is able to represent the different cognimotor dysfunction. Parkinsonism, ataxia, dystonia, tive domains which are affected in mHE, while others chorea, and orobuccolingual dyskinesia can be observed. aim to find a single means as surrogate marker of brain Parkinsonism in AHD is characterized by a rapid dysfunction in HE. Recently the International Society progression and postural instability or cognitive impairfor Hepatic Encephalopathy and Nitrogen Metabolism ment early in the course of disease. The most character(ISHEN) formed a commission to review the available istic motor phenotype of AHD is cranial dyskinesia, data on the role of neuropsychological testing in HE, which is often intermixed with dystonic elements such and to make recommendations regarding the routine as forced grimacing, jaw opening or blepharospasm assessment of patients with liver disease (Randolph (Burkhard et al., 2003; Ferrara and Jankovic, 2009). et al., 2009). They recommended the use of either the Repeatable Battery for the Assessment of Neuropsychological Status (RBANS) (Randolph, 1998) or the PSEDIFFERENTIAL DIAGNOSIS Syndrome-Test (Schomerus et al., 1999). The choice of None of the symptoms present in patients with hepatic which battery to use should be based upon the availability encephalopathy proves the diagnosis of HE. Thus the of local translations and normative data. Moreover the exclusion of other possible causes of brain dysfunction commission recommended future systematic studies is requested. Any patient has to undergo a thorough to compare approaches to diagnosing and monitoring diagnostic workup that includes laboratory investigamHE. The PSE syndrome test is a test battery consisting tions and neuroimaging. Conditions that have to be of five paper and pencil tests: the Number Connection considered are intracranial bleeding, especially chronic Tests (NCT) A and B, the Digit Symbol Test (DST), subdural haematoma, Wernicke’s encephalopathy, alcothe Serial Dotting Test (SDT), and the Line Tracing Test hol or drug withdrawal, intoxication, hypoglycemia, (LTT) (Fig. 45.2). In the NCTA the subject is asked to electrolyte disturbances, and in rare cases encephalitis. connect the numbers from 1 to 13 as quickly as possible. Hyponatremia has gained special interest recently, as In the NCTB subjects are asked to connect numbers from it could be shown that the frequency of HE increased 1 to 13 and letters from A to L in alternating order. In the with the reduction in serum sodium levels (15%, 24%, Digit Symbol Test each of the numbers 1–9 is paired with and 38% in patients with serum sodium > 135, 130–135, a specific symbol in adjacent boxes. Subjects are and < 130 mEq/L, respectively) (Angeli et al., 2006). requested to draw the missing symbol in a row of boxes Guevara et al. (2009) showed that hyponatremia is a strong filled with numbers. In the Serial Dotting Test subjects predictor of the development of overt HE. Hyponatremia are requested to place a dot as quickly as possible in is considered to facilitate astrocytic swelling in patients the center of 100 circles, and in the Line Tracing Test with severe liver disease and thereby to facilitate the the task is to draw a continuous line between two parallel development of HE. Although the clinical presentation (winding) lines. The test battery is able to detect deficits of hyponatremic and hepatic encephalopathy share only in attention, visual perception, visual scanning effithe alteration of consciousness, hyponatremia should ciency, motor speed, and accuracy. The PSE syndrome

664

K. WEISSENBORN

A

B

D

C

E

Fig. 45.2. Subtests of the PSE syndrome test (PSE-Syndrome-Test): Number Connection Test A and B, Digit Symbol Test (upper line from left to right), Serial Dotting Test, and Line Tracing Test (lower line).

test is predominantly used in Europe where normative data have been collected for the German (Schomerus et al., 1999), the Italian (Amodio et al., 2008), the Spanish (Romero-Gomez et al., 2006) and the British (Marks et al., 2008) population. Meanwhile the test has also recently been validated and standardized in Poland (Wunsch et al., 2013); and an altered test version has been standardized in India (Thumburu et al., 2008). The RBANS has been developed and standardized in the US, where it is also used predominantly (Mooney

et al., 2007). This test battery contains measures of verbal and visual anterograde memory, working memory, cognitive processing speed, language (including semantic fluency), and visuospatial function (line orientation and figure copy). The results of 12 subtests are converted into age-adjusted index scores for five domains (immediate memory, visuospatial/constructional, language, attention, and delayed memory). The PSE syndrome test takes about 20 minutes for completion and evaluation, the RBANS 25–35 minutes.

PORTOSYSTEMIC ENCEPHALOPATHY 665 Critics of the test batteries complain about the time studied in patients with HE. They have been proven to needed to apply and evaluate the tests, the psychological be useful for follow-up examinations, as they show expertise needed to interpret the test results, and an increase or decrease of latency in close correlation the impact of learning effects, age, and education on to the clinical course of the disease. Endogenous test results. In addition copyright was mentioned as evoked potentials in addition may provide an insight barrier to using the test batteries. The search for “siminto the alteration of cognitive processes in HE. The ple” diagnostic methods has resulted so far in the recomclinical use of evoked potentials, however, is limited mendation of the Inhibitory Control Test (ICT), a (Weissenborn, 1991). They are less sensitive than psychocomputerized test of attention and response inhibition, metric tests and clinical assessment. The widespread which is freely available (Bajaj et al., 2007), and examiuse of endogenous evoked potentials is in addition hamnation of the critical flicker frequency (CFF) (Kircheis pered by the need for sophisticated technical equipment et al., 2002). The ICT results were repeatedly shown to and the need for trained personnel. be worse in patients with minimal HE as diagnosed by NATURAL HISTORY standard psychometric tests than in patients without mHE (Bajaj et al., 2007; Amodio et al., 2010). The diagThe natural history of hepatic encephalopathy is not well known since long-term follow-up studies are sparse. In nostic use of the test, however, remains to be proven. The most cases HE is observed to occur episodically, generICT results have been shown to depend – similar to other neuropsychological tests – upon age and education, and ally related to a precipitating factor that triggers a toxin to some extent even upon sociocultural factors (Amodio load to the brain. et al., 2010), and the elaboration of normative data has Cirrhotic patients with minimal HE are more prone to still to be undertaken. develop overt HE than those without. (Romero-Gomez CFF was recommended for diagnosing mHE by et al., 2001) identified altered CFF, Child–Pugh score, Kircheis et al. in 2002. CFF decreases with increasing and mHE as diagnosed by psychometric tests to be significantly associated with lower survival and higher risk grade of HE, but there is an overlap between the of overt HE. The percentage of patients who developed normal range and the test results in patients with mHE as defined by psychometric tests. In addition, CFF in overt HE was 45% when either psychometric tests or CFF patients with alcohol-induced cirrhosis differs signifiindicated abnormal scores and 3.3% in patients with cantly from CFF in patients with cirrhosis from other normal values in both tests ( p < 0.0001). causes. The data available at present suggest that Dhiman et al. (2010) applied a variant of the PSE synalterations in CFF go in parallel with alterations in psydrome test and CFF examination to 100 patients without chometric tests in a proportion of the patients, while a clinical signs of HE. Minimal HE was detected in 48% considering the PSE syndrome test sum score PHES pathologic result in one of the two does not predict (psychometric hepatic encephalopathy score). CFF was a pathologic result in the other (Romero-Gomez et al., 2007; Sharma et al., 2007). pathologic in 21%. The prevalence of mHE increased Neurophysiologic methods have been used for diagnowith increasing liver dysfunction. During follow-up of sis and follow-up of HE for decades. The majority of up to 2 years, 39.1% died among those who had mHE patients with clinically overt HE show alterations of the compared to 22.9% of those without HE. The probability EEG with an increasing amount of y and d rhythms with of survival in patients with PHES   6 was 67.7% at 1 increasing grade of HE, an initial increase, followed by a year of follow-up, and 26.4% at 2 years, compared to 88.7% at 1 year and 74.6% at 2 years in patients with decrease, in EEG amplitude, a discontinuous pattern, and PHES >  6 ( p ¼ 0.003). CFF was evaluated according finally an isoelectric EEG (Fig. 45.3) (Parsons-Smith et al., 1957; Davies et al., 1991; Montagnese et al., 2004; Guerit to age-adjusted normal data in this study, and thereby et al., 2009). Characteristic is the appearance of triphasic did not show any predictive value. waves. However, normal EEGs have been found in up to In a recent population-based study, Jepsen et al. 60% of patients with distinct clinical symptoms of HE (2010) showed that overt HE is associated with a high (Penin 1967; Weissenborn et al., 1990; Montagnese et al., mortality rate in patients with alcohol-toxic cirrhosis. 2007). Since EEG slowing was shown also in up to 30% Patients with HE had a median survival time of 2.4 months from its onset; 45% died within 1 month, 64% of patients with liver cirrhosis but no clinical signs of died within 1 year, and 85% died within 5 years. Of note, encephalopathy (van der Rijt et al., 1990; Montagnese et al., 2007), EEG is used for the detection of mHE in addipatients do not die of, but with hepatic encephalopathy. tion to other means by several groups (Quero et al., 1996). The presence of HE is just an indication of severe liver Visual evoked potentials (VEP), somatosensory dysfunction. evoked potentials (SSEP), and the P300 wave elicited Fichet et al. (2009) did a retrospective analysis of the in an auditory oddball paradigm have been intensively case records of 71 patients who were admitted to the

666

K. WEISSENBORN

Fig. 45.3. EEG follow-up in a patient who was admitted with grade 2 hepatic encephalopathy (HE) (A) and swiftly improved after therapy with lactulose and antibiotics was started (B and C).

intensive care unit (ICU) of their hospital with grade III and IV HE. Mortality rate at 1 year was 54%. 35% of the patients died during ICU care. The mortality rate was 8.9% in the patients with isolated HE and 80.7% in the patients with accompanying organ dysfunction.

LABORATORY INVESTIGATIONS Laboratory investigations and neuroimaging do not provide findings specific for HE. An elevated ammonia level hints at a potential hepatic origin of a patient’s cerebral condition, but is nevertheless unspecific. Serum

PORTOSYSTEMIC ENCEPHALOPATHY ammonia levels correlate broadly with stages of encephalopathy; however, there is substantial overlap between values at different stages, and ammonia levels may even be in the normal range in patients with clinical symptoms of HE.

NEUROIMAGING INVESTIGATIONS For a while the bright pallidal hyperintensities found on T1-weighted magnetic resonance images (MRI) in about 90% of the patients with cirrhosis of the liver were considered as representative of HE (Fig. 45.4). Meanwhile it is clear that the T1 alterations are due to the deposition of manganese and depend on the presence of portosystemic shunting, while their extent relates only slightly to HE symptoms (Pujol et al., 1993; Krieger et al., 1996; Spahr et al., 1996; Pomier-Layrargues et al., 1998; Rose et al., 1999a). The MRI alterations usually disappear within 1 year after liver transplantation (Lockwood et al., 1997). Recently MRI has successfully been used to support the pathophysiologic model of a slightly increased brain water content – “a low-grade

667

cerebral edema” – as a milestone in the development of HE in liver cirrhosis (Ha¨ussinger et al., 2000). Methods applied were magnetization transfer ratio (MTR) imaging (Rovira et al., 2001; Balata et al., 2003), diffusion-weighted imaging (DWI) and diffusion tensor imaging (DTI) (Kale et al., 2006; Kumar et al., 2008), water content mapping (Shah et al., 2008) and T2 relaxometry (Singhal et al., 2009). While an increase of brain water content in cirrhotic patients with hepatic encephalopathy is generally agreed on, it has to be clarified by further investigations if the patient develops an intra- or extracellular edema, or both (Shah et al., 2008; Poveda et al., 2010). Proton magnetic resonance spectra (MRS) of the brain in patients with liver cirrhosis and HE shows a characteristic alteration of the metabolite pattern – a decrease in the myoinositol and choline signal intensity combined with an increase in the glutamate/glutamine (Glx) signal intensity (Fig. 45.5). These alterations rise with increasing grade of HE and MR has been shown to be a sensitive means to represent metabolic alterations in these patients, with induced hyperammonemia leading to an immediate and significant alteration of the MR spectra (Balata et al., 2003). The MRS alterations have

Fig. 45.4. T1-weighted magnetic resonance imaging of the brain from a healthy subject on the left compared with a patient with cirrhosis on the right. Coronal slices show bilateral hyperintensities in the pallidum and substantia nigra in the patient (due to manganese deposition).

668

K. WEISSENBORN

Fig. 45.5. Proton magnetic resonance spectra of the white matter in patients with liver cirrhosis but no hepatic encephalopathy) (HE) (top), minimal HE (center), and grade 1 HE (bottom) compared to a control spectrum.

also been studied for their diagnostic use but without satisfying results (K€ ostler, 1998; Poveda et al., 2010). Positron emission tomography (PET) has been applied for the study of some pathophysiologic aspects

of HE, such as cerebral ammonia metabolism. In 1991a, Lockwood et al. suggested a significantly increased blood–brain barrier permeability for ammonia in patients with liver cirrhosis and minimal HE compared to controls based on their findings in a 13 N-ammonia PET study. This assumption was well in line with experimental and clinical observations, and backed up the hypothesis that hyperammonemia and an increase of brain ammonia uptake were the main causes for the development of HE. Recently, their data have been contradicted by two further studies which included patients with different grades of liver disease – from mild fibrosis to cirrhosis – and different grades of HE compared to controls, and which did not find a significant difference of the blood–brain barrier permeability for ammonia between the groups (Keiding et al., 2006; Goldbecker et al., 2010). Nevertheless the metabolic flux of ammonia into glutamine was increased in the brain of cirrhotic patients compared to controls as their plasma ammonia levels were increased and in consequence also their brain ammonia levels. Ahl et al. (2004) showed significant regional differences in cerebral ammonia metabolism with an excess of ammonia supply to the basal ganglia and the cerebellum mainly due to differences in the regional cerebral blood flow (CBF). Cagnin et al. (2006) described an increased PK11195 binding in the basal ganglia, dorsolateral prefrontal regions, and anterior cingulate gyrus in patients with liver cirrhosis and minimal HE (Cagnin et al., 2006). PK11195 is a specific high-affinity ligand to the “peripheral-type” benzodiazepine receptors (PTBR) (recently renamed the 18 kDa translocator protein) which in the brain are localized especially in the outer mitochondrial membrane of astrocytes and activated microglia. PTBR are considered to play a role in the pathophysiology of HE via their role in the neurosteroid synthesis (Cagnin et al., 2006). Activation of PTBR results in increased synthesis of neurosteroids – especially allopregnanolone – a potent modulator of GABA-ergic neurotransmission, which enhances GABA-elicited chloride currents and positively modulates the binding of GABA and benzodiazepines to their respective sites on the GABA-A receptor complex (Ahboucha and Butterworth, 2007). In accordance with the clinical observation that HE especially affects the basal ganglia, the cerebellum and the “attention system,” Lockwood and coworkers were able to show by 18 F-fluorodeoxyglucose PET that the glucose utilization is decreased in the cingulum and frontal and parieto-occipital cortex and increased in the basal ganglia, cerebellum, and temporomesial structures in patients with HE compared to healthy controls and patients without HE (Lockwood et al., 1993, 2002). Glucose metabolism of the cingulate gyrus and

PORTOSYSTEMIC ENCEPHALOPATHY 669 cortical areas mentioned was significantly decreased swelling (Schliess et al., 2006; Norenberg et al., 2009). even in patients with minimal HE and was positively Astrocyte swelling is considered the key factor in correlated to their neuropsychological and motor the pathogenesis of HE as it has been shown to trigger performance (Lockwood et al., 2002; Giewekemeyer multiple alterations of astrocyte function and gene et al., 2007). expression in part through oxidative stress-dependent modifications of proteins and RNA. Ammonia as well as TNF-a, benzodiazepines, and hypo-osmotic swelling PATHOLOGY/PATHOPHYSIOLOGY were shown to induce RNA oxidation in cultured The pathogenesis of HE is far from being fully underastrocytes and brain slices and thereby to compromise stood, but there is a working hypothesis which is continsynaptic plasticity and glutamatergic neurotransmission uously adjusted to new findings, and which at present (G€org et al., 2008; Ha¨ussinger and Schliess, 2008). includes ammonia and systemic inflammation as the Recently, G€org et al. (2010) showed evidence for oxidakey players. Blood ammonia levels may be increased tive/nitrosative stress, protein tyrosine nitration, and about two- to threefold in patients with liver cirrhosis RNA oxidation also in postmortem cortical brain tissue compared to controls and brain–blood concentration from patients with cirrhosis and hepatic encephalopathy. ratios of ammonia increase from 2 in controls to 3–4 Ammonia has also been shown to induce mitochonin patients with chronic liver failure (for comparison: drial permeability transition (mPT), probably mediated the brain–blood ammonia ratio in patients with acute by oxidative stress. Induction of the mPT leads to a liver failure is 8) (Butterworth, 2002). In the brain, collapse of the mitochondrial inner membrane potential, ammonia is detoxified by astrocytes via the synthesis swelling of the mitochondrial matrix, defective oxiof glutamine. Accordingly proton MRS studies of the dative phosphorylation, cessation of ATP synthesis, brain in patients with liver cirrhosis and different grades and finally the generation of reactive oxygen species. of HE show an increase of the glutamate/glutamine Thus induction of the mPT is part of the vicious circle signal. It is hypothesized that the brain osmolyte of oxidative/nitrosative stress and astrocytic dysfuncmyoinositol is extruded from the astrocytes to avoid tion (Norenberg et al., 2009). astrocyte swelling in spite of increasing intracellular The characteristic neuropathologic feature of the glutamine levels and thus to maintain astrocyte function. brain in patients who died with hepatic coma is an Of note, astrocyte swelling may be induced not only by increase in the size and number of protoplasmatic astroammonia detoxification but also by other factors such as cytes, the so-called Alzheimer type II astrocytes (Adams inflammatory cytokines, hyponatremia, or benzodiazeand Foley, 1952). They are characterized by large, pines (Ha¨ussinger et al., 2000), and in fact HE is often chromatin-deficient nuclei with prominent nucleoli surprecipitated in patients with liver cirrhosis by rounded by a small rim of cytoplasm. The Alzheimer concomitant disorders such as electrolyte dysbalance, type II astrocytes were found in all parts of the cerebral sedation with benzodiazepines, or infection. The vulnercortex, the basal ganglia, thalamus, amygdala, midbrain, ability of the brain to these factors is postulated to pons, and cerebellum. The number of Alzheimer type II depend upon the extent of the myoinositol depletion. astrocytes within the pallidum is representative for the Low baseline myoinositol levels were observed to predisdegree of astrocytic changes in each individual case. pose for neuropsychological deterioration by an experiThe extent of Alzheimer type II astrocytosis is signifimental amino acid load (Shawcross et al., 2004a). cantly correlated with blood ammonia levels and severity Comparing the response of patients with liver cirrhosis of encephalopathy (Martin et al., 1987). Neurons appear to an oral amino acid challenge in the presence of a syshealthy in neuropathologic studies of the brain from temic inflammation response syndrome (SIRS) and patients with end stage chronic liver disease, except again after its solution, Shawcross et al. (2004b) were for those cases with “acquired hepatocerebral degeneraable to show that the presence of inflammation increases tion.” Here the brain shows a patchy, spongy degenerathe effect of ammonia on brain function. Unfortunately, tion especially in the deep layers of cerebral cortex and patients with liver cirrhosis carry an increased risk of subcortical white matter, particularly in the parietoinfection, since their neutrophils and macrophages have occipital cortex, basal ganglia, and cerebellum. Microa reduced phagocytosis capacity (Mookerjee et al., scopically these regions are characterized by a diffuse 2007). Thus the synergism between ammonia and increase in the size and number of protoplasmic astroinflammation effects can frequently be observed in cytes with glycogen inclusions and regional parenchymal these patients. degeneration with zones of pseudolaminar microcavitaThere is ample experimental evidence that astrocyte tion, together with shrunken, pyknotic nerve cells swelling induces the formation of reactive oxygen and and some swelling and fragmentation of the myelin nitrogen oxide species, including nitric oxide (NO), while (Victor et al., 1965; Plum and Hindfelt, 1976). on the other hand oxidative stress induces astrocyte Butterworth (2007) hints to the fact that neuronal cell

670 K. WEISSENBORN death in brain in liver failure is considerably less than in the colonic lumen due to environmental changes with would be predicted considering the numerous cell death promotion of the growth of nonurease producing bactemechanisms present in this condition, such as NMDA ria; (4) interference with the uptake of glutamine and receptor-mediated excitotoxicity, oxidative/nitrosative its metabolism into NH3 in the gut wall (Conn and Lieberthal, 1979). Lactulose therapy has been shown to stress, and the presence of proinflammatory cytokines. reduce plasma ammonia levels (Als-Nielsen et al., 2004; Therefore he hypothesizes that the extent of neuronal Poo et al., 2006), to improve cognitive function in patients damage in liver failure may be attenuated by compensawith minimal HE (Prasad et al., 2007), and to reduce intertory mechanisms including downregulation of NMDA stitial brain edema in cirrhotics with mild HE with correreceptors or the presence of neuroprotective steroids sponding improvement in neuropsychological test scores such as allopregnanolone. (Kale et al., 2006). Sharma et al. (2009) showed that lacOf note, histologic alterations of the brain are most tulose was effective in the secondary prevention of HE pronounced in those regions with high blood supply episodes. In practice, side-effects such as diarrhea, flatuand thus the highest ammonia load: the basal ganglia, lence, and abdominal pain reduce patients’ compliance the cerebellum, and the cortex. The above average effect with this therapy. on the basal ganglia can be explained by a synergism of Antibiotics directed at urease-producing bacteria ammonia and manganese. Rose and coworkers (1999a) have similar efficacy in HE to nonabsorbable disacchashowed that the manganese concentration in postmortem rides (Als-Nielsen et al., 2004). In the past, neomycin and brain of patients who died with HE is significantly metronidazole have been used. Currently rifaximin, a increased compared to controls and most evident in the nonabsorbed derivative of rifamycin, is recommended globus pallidus. Manganese is probably actively trans(Leevy and Phillips, 2007; Bass et al., 2010). It has been ported and deposited in the pallidum, thalamic nuclei, shown to be well tolerated, safe and efficacious in both and substantia nigra (Aschner et al., 2007). Manganese short- and long-term use. The most recent study anadeposition per se results in neuronal loss, Alzheimer type lyzed in a double-blind, placebo-controlled design the II astrocytosis, and alteration of dopaminergic neuroprotective effect of rifaximin against breakthrough epitransmission (Butterworth et al., 1995). Manganese has sodes of HE in patients with advanced liver disease, who also been shown to increase ammonia toxicity in astrocyte had suffered at least two episodes of HE  2 during the cultures (Jayakumar et al., 2004) and – like ammonia – to previous 6 months. The study drug was added to an induce the expression of the “peripheral-type” benzodiazongoing lactulose therapy in about 90% of the patients. epine receptor mentioned above. Rifaximin significantly reduced the risk of an epsiode of HE over a 6 month period (HR 0.42, 95% CI 0.28–0.64; MANAGEMENT p < 0.001) and also reduced the frequency of hospitalizaHE episodes are frequently provoked by nutritional protion due to HE by half (Bass et al., 2010). The rifaximin tein overload, gastrointestinal bleeds, infection, medicadose applied was 550 mg twice per day. tion (especially sedatives or diuretics), or constipation. A decrease of plasma ammonia levels can also be Therefore, identification and treatment of the precipitatachieved by the application of L-ornithine-L-aspartate ing factor should be the first step in its management. In (LOLA). Ornithine stimulates both the urea cycle and gluaddition, correction of electrolyte disturbances is essentamine synthesis. The major effect results from stimulatial. Considering the role of ammonia in the pathogenesis tion of glutamine synthesis in skeletal muscle (Rose et al., of HE, at present the main route of HE therapy aims at 1999b). A significant reduction of plasma ammonia levels the reduction of ammonia production and absorption. and improvement of clinical symptoms after intravenous Nonabsorbable disaccharides – lactulose and lactitol – application of 80–120 g LOLA was shown in cirrhotic are an established first-line therapy in HE. The daily dose patients with grade III–IV HE as long ago as 1972 of lactulose should be titrated to result in two to four soft, (Leonhardt and Bungert). With regard to lower grades acidic stools per day (30–60 g per day). In comatose of HE, data are contradictory (Kircheis et al., 1997; patients lactulose-containing enemas (250 mL lactulose Stauch et al., 1998; Poo et al., 2006). Nevertheless LOLA in 750 mL water) can be used instead of the oral prepara(three to six sachets daily in three divided doses) is recomtion (Uribe et al., 1987). Lactitol has been shown to be as mended in addition to or as an alternative to rifaximin if effective as lactulose (Morgan and Hawley, 1987). Lactuthere is not complete remission of encephalopathy within lose is considered to be effective by several mechanisms: 24 hours after lactulose treatment onset (Cash et al., 2010). (1) the cathartic effect; (2) acidification of the intestinal Studies on the use of branched-chain amino acids content resulting in a reduction of ammonia absorption (BCAA) for therapy of HE date back more than 30 years. and net movement from ammonia from the blood into A comprehensive Cochrane review (Als-Nielsen et al., the bowel; (3) reduced bacterial production of ammonia 2004) showed that compared to the control treatment,

PORTOSYSTEMIC ENCEPHALOPATHY BCAA significantly increased the number of patients improving from hepatic encephalopathy (59% versus 41%; RR 1.31; 95% confidence limits 1.04–1.66). Two recent studies of BCAA supplementation showed a significant positive effect compared to placebo with regard to mortality and the progression of liver disease (Marchesini et al., 2003; Muto et al., 2005). Hepatic myelopathy and acquired hepatocerebral degeneration do not respond to conventional HE therapy. Here immediate liver transplantation is recommended (Weissenborn et al., 2003).

CONCLUSIONS Hepatic encephalopathy is a frequent complication of end stage chronic liver disease indicating poor prognosis. Neuropathologic findings indicate that it is a glial disease that results in severe neuronal dysfunction. The characteristic extrapyramidal and cerebellar symptoms distinguish HE from metabolic encephalopathies due to other causes. Diagnosis has to be made with careful exclusion of other possible causes of brain dysfunction. Even mild degrees of HE should be treated, as they significantly impair quality of life and activities of daily living. Further efforts are needed to clarify the pathogenesis of HE, to identify new therapeutic targets, and to harmonize the diagnostic approach.

REFERENCES Adams RD, Foley JM (1952). The neurological disorders associated with liver disease. Proc Assoc Res Nerv Ment Dis 32: 198–237. Ahboucha S, Butterworth RF (2007). The neurosteroid system: implication in the pathophysiology of hepatic encephalopathy. Neurochem Int 52: 575–587. Ahl B, Weissenborn K, van den Hoff J et al. (2004). Regional differences in cerebral blood flow and cerebral ammonia metabolism in patients with cirrhosis. Hepatology 40: 73–79. Als-Nielsen B, Gluud LL, Gluud C (2004). Non-absorbable disaccharides for hepatic encephalopathy: systematic review of randomised trials. BMJ 328: 1046–1051. Amodio P, Campagna F, Olianas S et al. (2008). Detection of minimal hepatic encephalopathy: normalization and optimization of the Psychometric Hepatic Encephalopathy Score. A neuropsychological and quantified EEG study. J Hepatol 49: 346–353. Amodio P, Ridola L, Schiff S et al. (2010). Improving detection of minimal hepatic encephalopathy using the inhibitory control task. Gastroenterology 139: 510–518. Angeli P, Wong F, Watson H et al. (2006). Hyponatremia in cirrhosis: results of a patient population survey. Hepatology 44: 1535–1542. Aschner M, Guilarte TR, Schneider JS et al. (2007). Manganese: recent advances in understanding its transport and neurotoxicity. Toxicol Appl Pharmacol 221: 131–147.

671

Bajaj JS, Saeian K, Verber MD et al. (2007). Inhibitory control test is a simple method to diagnose minimal hepatic encephalopathy and predict development of overt hepatic encephalopathy. Am J Gastroenterol 102: 754–760. Bajaj JS, Saeian K, Schubert CM et al. (2009). Minimal hepatic encephalopathy is associated with motor vehicle crashes: the reality beyond the driving test. Hepatology 50: 1175–1183. Balata S, Olde Damink SWM, Ferguson K et al. (2003). Induced hyperammonemia alters neuropsychology, brain MR spectroscopy and magnetization transfer in cirrhosis. Hepatology 37: 931–939. Bass NM, Mullen KD, Sanyal A et al. (2010). Rifaximin treatment in hepatic encephalopathy. N Engl J Med 362: 1071–1081. Burkhard PR, Delavelle J, Du Pasquier R et al. (2003). Chronic parkinsonism associated with cirrhosis: a distinct subset of acquired hepatocerebral degeneration. Arch Neurol 60: 521–528. Butterworth RF (2002). Pathophysiology of hepatic encephalopathy: a new look at ammonia. Metab Brain Dis 17: 221–227. Butterworth RF (2007). Neuronal cell death in hepatic encephalopathy. Metab Brain Dis 22: 309–320. Butterworth RF, Spahr L, Fontaine S et al. (1995). Manganese toxicity, dopaminergic dysfunction and hepatic encephalopathy. Metab Brain Dis 10: 259–227. Cadranel J-F, Lebiez E, Di Martino V et al. (2001). Focal neurological signs in hepatic encephalopathy in cirrhotic patients: an underestimated entity? Am J Gastroenterol 96: 515–518. Cagnin A, Taylor-Robinson S, Forton DM et al. (2006). In vivo imaging of cerebral “peripheral benzodiazepine binding sites” in patients with hepatic encephalopathy. Gut 55: 547–553. Campellone JV, Lacomis D, Giuliani MJ et al. (1996). Hepatic myelopathy. Case report with review of the literature. Clin Neurol Neurosurg 98: 242–246. Cash WJ, McConville P, McDermott E et al. (2010). Current concepts in the assessment and treatment of hepatic encephalopathy. Q J Med 103: 9–16. Conn HO, Lieberthal MM (1979). The Hepatic Coma Syndromes and Lactulose. Williams and Wilkins, Baltimore. Cordoba J, Garcia-Martinez R, Simon-Talero M (2010). Hyponatremic and hepatic encephalopathies: similarities, differences and coexistence. Metab Brain Dis 25: 73–80. Davies MG, Rowan MJ, Feely J (1991). EEG and event related potentials in hepatic encephalopathy. Metab Brain Dis 6: 175–186. Da Rocha, Braga FT, da Silva CJ et al. (2004). Reversal of parkinsonism and portosystemic encephalopathy following embolization of a congenital intrahepatic venous shunt: brain MR imaging and 1H spectroscopic findings. AJNR 25: 1247–1250. Dhiman RK, Kurmi R, Thumburu KK et al. (2010). Diagnosis and prognostic significance of minimal hepatic encephalopathy in patients with cirrhosis of liver. Dig Dis Sci 55: 2381–2390.

672

K. WEISSENBORN

Ferenci P, Lockwood A, Mullen K et al. (2002). Hepatic encephalopathy – definition, nomenclature, diagnosis, and quantification: final report of the working party at the 11th World Congresses of Gastroenterology, Vienna, 1998. Hepatology 35: 716–721. Ferrara J, Jankovic J (2009). Acquired hepatocerebral degeneration. J Neurol 256: 320–332. Fichet J, Mercier E, Genee O et al. (2009). Prognosis and 1year mortality of intensive care unit patients with severe hepatic encephalopathy. J Crit Care 24: 364–370. Giewekemeyer K, Berding G, Ahl B et al. (2007). Bradykinesia in cirrhotic patients with early hepatic encephalopathy is related to a decreased glucose uptake of frontomesial cortical areas relevant for movement initiation. J Hepatol 46: 1034–1039. Gilberstadt SJ, Gilberstadt H, Zieve L et al. (1980). Psychomotor performance defects in cirrhotic patients without overt encephalopathy. Arch Intern Med 140: 519–521. Goldbecker A, Buchert R, Berding G et al. (2010). Blood brain barrier permeability for ammonia in patients with different grades of liver fibrosis and normal plasma ammonia levels is not different from healthy controls. J Cereb Blood Flow Metab 30: 1384–1393. G€ org B, Qvartskhava N, Keitel V et al. (2008). Ammonia induces RNA oxidation in cultured astrocytes and brain in vivo. Hepatology 48: 567–579. G€ org B, Qvartskhava N, Bidmon H-J et al. (2010). Oxidative stress markers in the brain of patients with cirrhosis and hepatic encephalopathy. Hepatology 52: 256–265. Guerit JM, Amantini A, Fischer C et al. (2009). Neurophysiological investigations of hepatic encephalopathy: ISHEN practice guidelines. Liver Int 29: 789–796. Guevara M, Baccaro ME, Torre A et al. (2009). Hyponatremia is a risk factor of hepatic encephalopathy in patients with cirrhosis: a prospective study with time-dependent analysis. Am J Gastroenterol 104: 1382–1389. Ha¨ussinger D, Schliess F (2008). Pathogenetic mechanisms of hepatic encephalopathy. Gut 57: 1156–1165. Ha¨ussinger D, Kircheis G, Fischer R et al. (2000). Hepatic encephalopathy in chronic liver disease: a clinical manifestation of astrocyte swelling and low-grade cerebral edema? J Hepatol 32: 1035–1038. Jayakumar AR, Rama Rao KV, Kalaiselvi P et al. (2004). Combined effects of ammonia and manganese on astrocytes in culture. Neurochem Res 29: 2051–2056. Jepsen P, Ott P, Andersen PK et al. (2010). Clinical course of alcoholic liver cirrhosis: a Danish population-based cohort study. Hepatology 51: 1675–1682. Kale RA, Gupta RK, Saraswat VA et al. (2006). Demonstration of interstitial cerebral edema with diffusion tensor MR imaging in type C hepatic encephalopathy. Hepatology 43: 698–706. Keiding S, Srensen M, Bender D et al. (2006). Brain metabolism of 13 N-ammonia during acute hepatic encephalopathy in cirrhosis measured by positron emission tomography. Hepatology 43: 42–50. Kircheis G, Nilius R, Held C et al. (1997). Therapeutic efficacy of L-ornithine-L-aspartate infusions in patients with

cirrhosis and hepatic encephalopathy: results of a placebo-controlled, double-blind study. Hepatology 25: 1351–1360. Kircheis G, Wettstein M, Timmermann L et al. (2002). Critical flicker frequency for quantification of low-grade hepatic encephalopathy. Hepatology 35: 357–366. Kircheis G, Knoche A, Hilger N et al. (2009). Hepatic encephalopathy and fitness to drive. Gastroenterology 137: 1706–1715. Krieger S, Jauß M, Jansen O et al. (1996). Neuropsychiatric profile and hyperintense globus pallidus on T1-weighted magnetic resonance images in liver cirrhosis. Gastroenterology 111: 147–155. Kril JJ, Butterworth RF (1997). Diencephalic and cerebellar pathology in alcoholic and nonalcoholic patients with end-stage liver disease. Hepatology 26: 837–841. K€ ostler H (1998). Proton magnetic resonance spectroscopy in portal-systemic encephalopathy. Metab Brain Dis 13: 291–301. Kumar R, Gupta RK, Elderkin-Thompson V et al. (2008). Voxel-based diffusion tensor magnetic resonance imaging evaluation of lowgrade hepatic encephalopathy. J Magn Reson Imaging 27: 1061–1068. Leevy CB, Phillips JA (2007). Hospitalizations during the use of rifaximin versus lactulose for the treatment of hepatic encephalopathy. Dig Dis Sci 52: 737–741. Leonhardt H, Bungert HJ (1972). Therapie der schweren Hyper ammoniamie. Med Klin 67: 1052–1056. Lewis MB, MacQillan G, Bamford JM, Howdle PD (2000). Delayed myelopathic presentation of the acquired hepatocerebral degeneration syndrome. Neurology 54: 1011. Lockwood AH, Yap EWH, Wong W-H (1991a). Cerebral ammonia metabolism in patients with severe liver disease and minimal hepatic encephalopathy. J Cereb Blood Flow Metab 11: 337–341. Lockwood AH, Murphy BW, Donnelly KZ et al. (1993). Positron emission tomographic localization of abnormalities of brain metabolism in patients with minimal hepatic encephalopathy. Hepatology 18: 1061–1068. Lockwood AH, Weissenborn K, Butterworth RF (1997). An image of the brain in patients with liver disease. Curr Opin Neurol 10: 525–533. Lockwood AH, Weissenborn K, Bokemeyer M et al. (2002). Correlations between cerebral glucose metabolism and neuropsychological test performance in non-alcoholic cirrhotics. Metab Brain Dis 17: 29–40. Marchesini G, Bianchi G, Merli M et al. (2003). Nutritional supplementation with branched-chain amino acids in advanced cirrhosis: a double-blind, randomized trial. Gastroenterology 124: 1792–1801. Marks ME, Jackson CD, Montagnese S et al. (2008). Derivation of a normative UK database for the psychometric hepatic encephalopathy score (PHES): confounding effect of ethnicity and test scoring. J Hepatol 48: S119. Martin H, Voss K, Hufnagl P et al. (1987). Morphometric and densitometric investigations of protoplasmic astrocytes and neurons in human hepatic encephalopathy. Exp Pathol 32: 241–250.

PORTOSYSTEMIC ENCEPHALOPATHY Montagnese S, Amodio P, Morgan MY (2004). Methods for diagnosing hepatic encephalopathy in patients with cirrhosis: a multidimensional approach. Metab Brain Dis 19: 281–312. Montagnese S, Jackson C, Morgan MY (2007). Spatio-temporal decomposition of the electroencephalogram in patients with cirrhosis. J Hepatol. 46 (3): 447–458. Mookerjee R, Stadlbauer V, Lidder S et al. (2007). Neutrophil dysfunction in alcoholic hepatitis superimposed on cirrhosis is reversible and predicts outcome. Hepatology 46: 831–840. Mooney S, Hasssanein TI, Hilsabeck RC et al. (2007). The UCSD Hepatology Neurobehavioral Research Program. Utility of the Repeatable Battery for the Assessment of Neuropsychological Status (RBANS) in patients with end-stage liver disease awaiting liver transplant. Arch Clin Neuropsychol 22: 175–186. Morgan MY, Hawley KE (1987). Lactitol versus lactulose in the treatment of acute hepatic encephalopathy in cirrhotic patients: a double-blind, randomized trial. Hepatology 7: 1278–1284. Muto Y, Sato S, Watanabe A et al. (2005). Effects of oral branched-chain amino acid granules on event-free survival in patients with liver cirrhosis. Clin Gastroenterol Hepatol 3: 705–713. Norenberg MD, Rama Rao KV, Jayakumar AR (2009). Signaling factors in the mechanism of ammonia neurotoxicity. Metab Brain Dis 24: 103–117. Parsons-Smith BG, Summerskill WHJ, Dawson AM et al. (1957). The electroencephalograph in liver disease. Lancet ii: 867–871. ¨ ber den diagnostischen Wert des Penin H (1967). U Hirnstrombildes bei der hepato-portalen Encephalopathie. Fortschr Neurol Psychiatr 35: 174–234. Phillips G, Schwartz R, Gabuzda G et al. (1952). The syndrome of impending hepatic coma in patients with cirrhosis of the liver given certain nitrogenous substances. N Engl J Med 247: 239–246. Plum F, Hindfelt B (1976). The neurological complications of liver disease. In: P Vinken, DF Bruyn (Eds.), Metabolic and Deficiency Diseases of the Nervous System. Handbook of Clinical Neurology. vol. 27, Elsevier, Amsterdam, pp. 349–377. Pomier-Layrargues G, Rose C, Spahr L et al. (1998). Role of manganese in the pathogenesis of portal-systemic encephalopathy. Metab Brain Dis 13: 311–317. Poo JL, Gongora J, Sanchez-Avila F et al. (2006). Efficacy of oral L-ornithine-L-aspartate in cirrhotic patients with hyperammonemic hepatic encephalopathy. Results of a randomized, lactulose-controlled study. Ann Hepatol 5: 281–288. Poveda M-J, Bernabeu A, Concepcio´n L et al. (2010). Brain edema dynamics in patients with overt hepatic encephalopathy. A magnetic resonance imaging study. Neuroimage 52: 481–487. Prasad S, Dhiman RK, Duseja A et al. (2007). Lactulose improves cognitive functions and health-related quality of life in patients with cirrhosis who have minimal hepatic encephalopathy. Hepatology 45: 549–559.

673

Pujol A, Pujol J, Graus F et al. (1993). Hyperintense globus pallidus on T1-weighted MRI in cirrhotic patients is associated with severity of liver failure. Neurology 43: 65–69. Quero JC, Hartmann IJ, Meulstee J et al. (1996). The diagnosis of subclinical hepatic encephalopathy in patients with cirrhosis using neuropsychological tests and automated electroencephalogram analysis. Hepatology 24: 556–560. Randolph C (1998). The Repeatable Battery for the Assessment of Neuropsychological Status (RBANS). Psychological Corporation, San Antonio. Randolph C, Hilsabeck R, Kato A et al. (2009). Neuropsychological assessment of hepatic encephalopathy: ISHEN practice guidelines. Liver Int 29: 629–635. Read A, Sherlock S, Laidlaw J et al. (1967). The neuropsychiatric syndromes associated with chronic liver disease and an extensive portal-systemic collateral circulation. Q J Med 36: 135–150. Rehnstr€ om S, Simert G, Hansson JA et al. (1977). Chronic hepatic encephalopathy. A psychometric study. Scand J Gastroenterol 12: 305–311. Rikkers L, Jenko P, Rudman D et al. (1978). Subclinical hepatic encephalopathy: detection, prevalence, and relationship to nitrogen metabolism. Gastroenterology 75: 462–469. Romero-Gomez M, Boza F, Garcia-Valdecasas MS et al. (2001). Subclinical hepatic encephalopathy predicts the development of overt hepatic encephalopathy. Am J Gastroenterol 96: 2718–2723. Romero Gomez M, Cordoba J, Jover R et al. (2006). Normality tables in the Spanish population for psychometric tests used in the diagnosis of minimal hepatic encephalopathy. Med Clin (Barc) 127: 246–249. Romero Gomez M, Cordoba J, Jover R et al. (2007). Value of the critical flicker frequency in patients with minimal hepatic encephalopathy. Hepatology 45: 879–885. Rose C, Butterworth RF, Zayed J et al. (1999a). Manganese deposition in basal ganglia structures results from both portal-systemic shunting and liver dysfunction. Gastroenterology 117: 640–644. Rose C, Michalak A, Rao KV et al. (1999b). L-ornithine-Laspartate lowers plasma and cerebrospinal fluid ammonia and prevents brain edema in rats with acute liver failure. Hepatology 30: 636–640. Rovira A, Grive E, Pedraza S et al. (2001). Magnetization transfer ratio values and proton MR spectroscopy of normal appearing cerebral white matter in patients with liver cirrhosis. AJNR 22: 1137–1142. Schliess F, G€ org B, Ha¨ussinger D (2006). Pathogenetic interplay between osmotic and oxidative stress: the hepatic encephalopathy paradigm. Biol Chem 387: 1363–1370. Schomerus H, Hamster W (1998). Neuropsychological aspects of portal-systemic encephalopathy. Metab Brain Dis 13: 361–377. Schomerus H, Hamster W, Blunck H et al. (1981). Latent portasystemic encephalopathy: I. Nature of cerebral functional defects and their effect on fitness to drive. Dig Dis Sci 26: 622–630.

674

K. WEISSENBORN

Schomerus H, Weissenborn K, Hamster W et al. (1999). PSESyndrom-Test. Swets Test Services. Swets and Zeitlinger B.V., Frankfurt. Shah NJ, Neeb H, Kircheis G et al. (2008). Quantitative cerebral water content mapping in hepatic encephalopathy. Neuroimage 41: 706–717. Sharma P, Sharma BC, Puri V et al. (2007). Critical flicker frequency: diagnostic tool for minimal hepatic encephalopathy. J Hepatol 47: 67–73. Sharma BC, Sharma P, Agarwal A et al. (2009). Secondary prophylaxis of hepatic encephalopathy: an open label randomized controlled trial of lactulose versus placebo. Gastroenterology 137: 885–891. Shawcross DL, Balata S, Olde-Damink SW et al. (2004a). Low myoinositol and high glutamine levels in brain are associated with neuropsychological deterioration after induced hyperammonemia. Am J Physiol 287: G503–G509. Shawcross DL, Davies NA, Williams R et al. (2004b). Systemic inflammatory response exacerbates the neuropsychological effects of induced hyperammonemia in cirrhosis. J Hepatol 40: 247–254. Shawcross DL, Olde Damink SWM, Butterworth RF et al. (2005). Ammonia and hepatic encephalopathy: the more things change, the more they remain the same. Metab Brain Dis 20: 169–179. Sherlock S (1958). Pathogenesis and management of hepatic coma. Am J Med 24: 805–813. Sherlock S, Summerskill WHJ, White LP et al. (1954). Portalsystemic encephalopathy. neurological complications of liver disease. Lancet I: 453–457. Singhal A, Nagarajan R, Kumar R et al. (2009). Magnetic resonance T2-relaxometry and 2D L-correlated spectroscopy in patients with minimal hepatic encephalopathy. J Magn Reson Imaging 30: 1034–1041. Spahr L, Butterworth RF, Fontaine S et al. (1996). Increased blood manganese in cirrhotic patients: relationship to pallidal magnetic resonance signal hyperintensity and neurological symptoms. Hepatology 24: 1116–1120. Stauch S, Kircheis G, Adler G et al. (1998). Oral L-ornithineL-aspartate therapy of chronic hepatic encephalopathy: results of a placebo-controlled double-blind study. J Hepatol 28: 856–864. Summerskill WHJ, Davidson EA, Sherlock S et al. (1956). The neuropsychiatric syndrome associated with hepatic cirrhosis and an extensive portal collateral circulation. Q J Med 25: 245–266. Tarter RE, Hegedus AM, Van Thiel DH et al. (1984). Nonalcoholic cirrhosis associated with neuropsychological dysfunction in the absence of overt evidence of hepatic encephalopathy. Gastroenterology 86: 1421–1427.

Thumburu KK, Kurmi R, Dhiman RK et al. (2008). Psychometric hepatic encephalopathy score, critical flicker frequency and P300 event-related potential for the diagnosis of minimal hepatic encephalopathy: evidence that psychometric hepatic encephalopathy score is enough. 13th ISHEN, Padua, 2008 [abstract]. Uribe M, Campollo O, Vargas F et al. (1987). Acidifying enemas (lactitol and lactose) vs. nonacidifying enemas (tap water) to treat acute portal-systemic encephalopathy: a double-blind randomized clinical trial. Hepatology 7: 639–643. Van der Rijt CDC, Schalm S, De Groot GH et al. (1990). Objective measurement of hepatic encephalopathy by means of automated EEG analysis. Electroencephalogr Clin Neurophysiol 75: 289–295. Victor M, Adams RD, Cole M (1965). The acquired (nonWilsonian) type of chronic hepatocerebral degeneration. Medicine (Baltimore) 44: 345–396. Watanabe A, Tuchida T, Yata Y et al. (1995). Evaluation of neuropsychological function in patients with liver cirrhosis with special reference to their driving ability. Metab Brain Dis 10: 239–248. Wein C, Koch H, Popp B et al. (2004). Minimal hepatic encephalopathy impairs fitness to drive. Hepatology 39: 739–745. Weissenborn K, Scholz M, Hinrichs H, Wiltfang J, Schmidt FW, K€ unkel H (1990). Neurophysiological assessment of early hepatic encephalopathy. Electroencephalogr Clin Neurophysiol. 75 (4): 289–295. Weissenborn K (1991). Neurophysiological methods in the diagnosis of early hepatic encephalopathy. In: F Bengtsson, B Jeppsson, T Almdal et al. (Eds.), Progress in Hepatic Encephalopathy and Metabolic Nitrogen Exchange. CRC Press, Boca Raton, pp. 27–39. Weissenborn K, Ennen J, Schomerus H et al. (2001). Neuropsychological characterisation of hepatic encephalopathy. J Hepatol 34: 768–773. Weissenborn K, Tietge UJ, Bokemeyer M et al. (2003). Liver transplantation improves hepatic myelopathy: evidence by three cases. Gastroenterology 124: 346–351. Wunsch E, Koziarska D, Kotarska K et al. (2013). Normalization of the psychometric hepatic encephalopathy score in polish population. A prospective, quantified electroencephalography study. Liver Int 33: 1332–1340. Zeegen R, Drinkwater JE, Dawson AM (1970). Method for measuring cerebral dysfunction in patients with liver disease. Br Med J 2: 633–636. Zieve L (1991). Historical remarks and recent trends in hepatic encephalopathy. In: F Bengtsson, B Jeppsson, T Almdal et al. (Eds.), Progress in Hepatic Encephalopathy and Metabolic Nitrogen Exchange. CRC Press, Boca Raton, pp. 3–8.

Handbook of Clinical Neurology, Vol. 120 (3rd series) Neurologic Aspects of Systemic Disease Part II Jose Biller and Jose M. Ferro, Editors © 2014 Elsevier B.V. All rights reserved

Chapter 46

Neurotoxicity of commonly used hepatic drugs CHRISTINE L. AHRENS AND EDWARD M. MANNO* Neurological Intensive Care Unit, Cleveland Clinic, Cleveland, OH, USA

INTRODUCTION The increased need for and utility of organ transplantation including liver transplantation has increased the use of immunosuppressive agents to prevent rejection. Neurologic complications are common with many of these medications (Table 46.1). The neurologist, surgeon, or intensivist will often be confronted with a variety of these complications. Similarly, the use of interferons has improved the care of patients with hepatitis C and other hepatic infections. This chapter will review the medications used to treat hepatic infections and to prevent liver rejection posttransplantation. Neurologic complications commonly encountered will be emphasized.

CALCINEURIN INHIBITORS Calcineurin inhibitors are a class of immunosuppressant drugs that decrease lymphocytic proliferation through the inhibition of a phophatase calcineurin. The two commonly employed drugs are ciclosporin and tacrolimus (previously referred to as FK506). These drugs represent the mainstay of immunosuppression after orthotopic liver transplantation (OLT).

MECHANISM OF ACTION The lymphocytic response to an antigen is mediated through antigenic binding of the T-helper membrane receptor. This results in the opening of calcium channels which facilitates calcium influx into the cell. Intracellular calcium subsequently binds to calmodulin. This complex stimulates calcineurin (a phosphatase) which activates various transcription factors (primarily IL-2). These factors lead to a clonal expansion of new lymphocytes while inhibiting the apoptosis of existing lymphocytic lines.

This process of antigen-induced clonal expansion is inhibited by ciclosporin. Ciclosporin binds to a cytosolic protein named cyclophilin. This complex inhibits calcineurin-mediated activation of several transcriptases. Tacrolimus inhibits calcineurin through binding to FK-binding protein which in turn inhibits the function of calcineurin (Rang et al., 2003).

NEUROTOXICITY The range of neurologic signs and symptoms attributable to the use of calcineurin inhibitors is considerable. Symptoms can vary from mild headache, paresthesias, and confusion to psychosis and coma. Neurologic signs include tremor, visual disturbances, and seizures. Other reported neurologic complications include speech apraxia, cortical blindness, hemiparesis, parkinsonism, peripheral neuropathy, and ocular motor difficulties (Muellar et al., 1994; Wijdicks et al., 1994, 1995; Bechstein, 2000). The incidence of neurologic complications reported after liver transplantation ranges between 10% and 47% with most studies limiting the range between 20% and 30% (Saner et al., 2007, 2009, 2010). The exact incidence of side-effects attributable to calcineurin inhibitors is difficult to assess. Pre-existing or posttransplant encephalopathy can complicate the evaluation of immunosuppressant medications. Drug interactions with glucocorticoids can confuse which medication is the source of the problem. Other complications can include issues attributable to electrolyte disturbances, infections, sepsis, or sedative medications. Thus, neurotoxicity attributable to calcineurin inhibitors is often a diagnosis of exclusion (Bechstein, 2000). Neurotoxicity attributable to calcineurin inhibitors is often classified as mild, moderate, or severe. The most common side-effect is tremor, seen in up to 40% of

*Correspondence to: Edward M. Manno, M.D., Head, Neurocritical Care, Cleveland Clinic, 9500 Euclid Ave, HB-105, Cleveland, OH 44195, USA. Tel: þ1-216-445-1624, E-mail: [email protected]

676

C.L. AHRENS AND E.M. MANNO

Table 46.1 Potential central nervous system adverse effects of immunosuppresent medications Medication Immunosuppressants Calcineurin inhibitors (ciclosporin, tacrolimus) Sirolimus Mycophenolate mofetil Antihepatitis therapies Interferons Ribavirin Lamivudine Adefovir dipivoxil Entecavir Telbivudine

Central nervous system adverse effects

Headache, paresthesias, confusion, tremor, visual disturbances, apraxia, parkinsonism, peripheral neuropathy, ocular motor difficulties, psychosis, coma seizures, hemiparesis, cortical blindness, leukoencephalopathy Headache, arthralgias, insomnia, dizziness, neuropathy, confusion, abnormal vision, leukoencephalopathy (rare) Headache, insomnia, dizziness, anxiety, pain, psychosis, seizure Depression, asthenia, myalgia, psychosis, insomnia, irritability Headache, fatigue Headache, fatigue, insomnia, neuropathy Headache, asthenia Headache, fatigue, dizziness Headache, dizziness, insomnia, peripheral neuropathy (occurs in combination with pegylated interferon (peg-IFN))

patients. This can resolve spontaneously but usually requires dose reduction. Paresthesias can respond similarly to dose adjustments. Headache, neuralgia, and peripheral neuropathies are also quite common. Severe complications such as psychosis, coma or a leukoencephalopathy occur in approximately 5% of patients and may not be amenable to dose reduction or discontinuation of the calcineurin inhibitor (Bechstein, 2000; Saner et al., 2009).

MECHANISM OF NEUROTOXICITY The mechanism of neurologic cellular dysfunction after the initiation of calcineurin inhibitors is not completely understood. There are, however, several observations and findings that are common to both tacrolimus and ciclosporin toxicity. Both animal models and clinical studies suggest that calcineurin inhibitors increase sympathetic outflow and nerve activity (Scherrer et al., 1990; Lyson et al., 1993). The exact localization of this process is unknown but probably involves both central and peripheral mechanisms (Sander et al., 1996; Bechstein, 2000). Increased sympathetic activity may also be modulated through both pre- and postsynaptic effects on excitatory and inhibitory amino acid receptors. Ciclosporin may decrease GABA-mediated inhibition. In rat brain slice models a ciclosporin–cyclophilin complex can desensitize GABA receptors (Martina et al., 1996). Similarly, tacrolimus has been shown to increase NMDA-induced transmitter release (Lu et al., 1996). Calcineurin inhibition may also be selectively toxic to white matter. In vitro studies of cells incubated with ciclosporin suggest selective toxicity of glial cells

(Stoltenburg-Didinger and Boegner, 1992). Similarly, ciclosporin induces apoptosis in oligodendrocytes (McDonald et al., 1996). Cell lines with the highest density of calcineurin appear to be most affected. These effects increase with the length of exposure, possibly accounting for the delayed development of a leukoencephalopathy seen in some patients (Bechstein, 2000). The white matter changes commonly seen on computed tomography (CT) and magnetic resonance imaging (MRI) may represent vascular injury. Ciclosporin and tacrolimus induced white matter changes on MRI, typically in the occipital white matter and border zone areas similar to those areas encountered during hypoperfusion (Bartynski et al., 1994). Tacrolimus can also affect the thalami and produce vascular injury in animal models (Frank et al., 1993). Patients after liver transplant will develop cortical hyperintensities involving the cingulate gyrus and the occipital lobes noted on proton density-weighted imaging (Jansen et al., 1996). This may represent ciclosporin-induced changes to the vascular basement membrane (Sloane et al., 1985).

SPECIFIC COMPLICATIONS Mild symptoms of calcineurin inhibition include tremor, sleep disturbances, mood alterations, headaches, and confusion. These are commonly encountered and may even have a higher incidence than reported if a thorough neurologic and mental status examination is performed. Moderate and severe symptoms of cortical blindness, seizures, coma, and encephalopathy are found in a smaller percentage of patients and may be more common with tacrolimus (Muellar et al., 1994). The incidence of

NEUROTOXICITY OF COMMONLY USED HEPATIC DRUGS these complications appears to be higher for liver transplantation compared to other solid organ transplants (Muellar et al., 1994) and may be related to high plasma concentration of tacrolimus. The leukoencephalopathy encountered with calcineurin use is difficult to predict or characterize. Clearly white matter tracts are involved as central pontine myelinolysis can occur with devastating effects (Saner et al., 2007). Calcineurin effects on MRI can occur early but also in a delayed fashion after several weeks or months (Saner et al., 2009). Interestingly, MRI changes in white matter do not necessarily correlate with neurologic symptoms (Wijdicks et al., 1995). Seizures are seen most commonly immediately posttransplant. Early studies had suggested that calcineurin inhibition-induced seizures had a poor outcome (Adams et al., 1987; Estol et al., 1989). Wijdicks, however, in a retrospective analysis of post liver transplant patients, suggested that this may not be accurate. In the Mayo series, most seizures occurred during the time of initiation or adjustment of ciclosporin or tacrolimus. All patients had supratherapeutic levels and did not have further seizures with dose reduction (Wijdicks et al., 1996). Hypertension has been shown to be an independent risk factor for developing seizures post-transplant (Erer et al., 1996), and seizures may be associated with hypomagnesemia (Thompson et al., 1989). The significance of calcineurin-induced neurotoxicity post liver transplant is unclear. Mild symptoms probably have little significance; however, older literature has suggested that patients with late onset or severe neurologic complications have worse outcome (Wszolek et al., 1991; Muellar et al., 1994). It is difficult, however, to discern if late neurologic complications are due to calcineurin inhibitors or a late complication of a failing transplant (Bechstein, 2000). There are a number of predisposing factors that can increase the risk to post-transplant patients of calcineurin inhibition toxicity. Liver failure can lead to blood–brain barrier disruption and increased access of ciclosporin and tacrolimus to the brain (Freise et al., 1993). Intravenous administration of drug, concurrent use of prednisone, and hypocholesterolemia can increase the total and unbound level of calcineurin inhibitors, thus increasing brain uptake of the drug used (Freise et al., 1991, 1993; Bechstein, 2000).

TREATMENT OF COMPLICATIONS The correlation between neurotoxicity and calcineurin inhibitor drug levels is weak and routine monitoring of drug levels is usually not indicated. A simple dose response relationship does not exist and discontinuation

677

of drug does not always reverse symptomatology. Severe toxicity, however, does occur with higher drug levels. Toxicity may be mediated through ciclosporin and tacrolimus metabolites which can cross the blood– brain barrier. Impaired hepatic function can also increase these metabolites. Monitoring of these metabolites to assess for neurotoxicity has been suggested but has not gained wide acceptance (Trull et al., 1989). In some instances maneuvers designed to decrease drug levels can help with symptomatology. These can include lowering the dose, switching to oral therapy (Wijdicks et al., 1999), or treating renal failure. Lipid supplementation with soybean oil has been used in five patients post liver transplant to prevent lipophillic calcineurin inhibtors from crossing the blood–brain barrier (Ide et al., 2007). Several antibiotics can have drug interactions which can increase ciclosporin or tacrolimus levels. The use of combined immunosuppressant regimens can be used to lower the levels of any individual drug (Bechstein, 2000). Treatment of post liver transplant hypertension is important to decrease the incidence of neurologic complications. Drug-induced blood–brain barrier and vascular membrane disruption may impair cerebral autoregulation rendering the brain susceptible to a form of malignant hypertension. This may account for the MRI changes commonly encountered which resemble the posterior reversible encephalopathy syndrome (PRES). Seizure control post liver transplant can be difficult. Management involves dose reduction and the initiation of anticonvulsants. Phenytoin, phenobarbital, and carbamazepine will decrease ciclosporin levels. Valproate or levetiracetam may be preferred. Levetiracetam dosing will need to be adjusted based on renal function. Early post-transplant seizures do not appear to affect long-term outcome; however, status epilepticus and epiletiform activity on EEG monitoring has an ominous prognosis (Wszolek et al., 1991; Wijdicks et al., 1996).

OTHER MEDICATIONS USED FOR IMMUNOSUPPRESION AFTER ORTHOTOPIC LIVER TRANSPLANTATION Sirolimus Sirolimus is the generic name for a drug also known as rapamycin. It has a similar mechanism of action to tacrolimus in that sirolimus binds to the intracellular FK-12 protein but inhibits a regulatory kinase (mTOR) which leads to the suppression of T cell proliferation. No evidence of direct central neurotoxicity could be attributed to sirolimus use in a review of over 200 transplant patients (Maramattom and Wijdicks, 2004). A demyelinating

678

C.L. AHRENS AND E.M. MANNO

sensorimotor polyneuropathy, however, has been described (Bilodeau et al., 2008).

Mycophenolate mofetil (CellCept) Mycophenolate mofetil is a prodrug which is hydrolyzed to mycophenolic acid. Mycophenolic acid is a noncompetitive inhibitor of inosine monophosphate dehydrogenase, an enzyme required for B and T cell proliferation (Krensky et al., 2006). The toxicity for this drug is primarily GI and hematopoietic. Use can lead to hyperlipidemia, thrombocytopenia, and leukopenia. Levels can be increased with ingestion of grapefruit juice. A review of 191 liver transplant patients treated with mycophenolate did not report any neurologic complications (Pfitzmann et al., 2003). Mycophenolate has been used for rescue therapy in patients that have developed neurotoxicty from calcineurin inhibitors (Klupp et al., 1997). Two studies have suggested that mycophenolate is a safe and useful adjuvant for immunosuppression in post liver transplant patients (Freise et al., 1993; Klupp et al., 1997).

INTERFERONS, RIBAVIRIN AND NUCLEOSIDE AND NUCLEOTIDE ANALOGS USED IN THE TREATMENT OF HEPATITIS B AND C VIRUS There are a select number of medications available for the treatment of hepatitis B virus (HBV) and hepatitis C virus (HCV) (Lok and McMahon, 2009). Interferons, ribavirin, lamivudine, entecavir, tenofovir, telbivudine, and adefovir make up the armamentarium of commonly used antivirals considered in the treatment of hepatitis. Interferon monotherapy or in combination with ribavirin are the recommended medications for the treatment of chronic hepatitis C virus (HCV). Not all patients with HBV and/or HCV receive pharmacologic treatment. However, for those appropriately selected candidates who do, there exists a potential for adverse effects related to these therapies. Particular adverse events to some of these therapies are the primary reason patients discontinue therapy. Understanding the potential for these effects to occur and patient counseling may reduce discontinuation or denial of therapy (Ghany et al., 2009).

Interferons Interferons (INF) are a class of cytokines which have antiviral, immunomodulating, and antiproliferative effects (Lok and McMahon, 2009). Three classes of human IFNs exist (a, b, and g) with significant antiviral activity; however, only recombinant a and b IFNs are clinically utilized. Standard IFNs were first available for the treatment of HBV and HCV but have been

replaced by the newer pegylated interferons (peg-IFN). Certain advantages exist with peg-IFNs compared to the standard formulations, including a reduction in the severity of side-effects. Commercially available IFNs used in the treatment of HBV are standard IFN-a2b (Intron®A) and peg-IFN-a2a (PEGASYS®) and IFN-a 2b, peg-IFN-a2a, peg-IFN-a2b (PegIntron®), and IFN alfacon-1 (Infergen®).

MECHANISM OF ACTION IFNs activate the JAK-STAT signal-transduction pathway on the surface of target cells leading to nuclear translocation of a cellular protein complex that binds to genes containing an IFN-specific response element. This leads to synthesis of over two dozen proteins that contribute to viral resistance mediated at a different stage of viral penetration. IFN-induced proteins can inhibit protein synthesis in the presence of double-stranded RNA. IFN also induces a phosphodiesterase which cleaves a portion of transfer RNA thus preventing peptide elongation. IFNs may modify the immune response to infection as well through enhancement of the lytic effects of cytotoxic T lymphocytes (Pegasys, 2011).

NEUROTOXICITY Pegylated interferons have several, undesirable adverse effects. Nearly every patient treated with the combination therapy (peginterferon and ribavirin) will experience one or more adverse effects, potentially resulting in discontinuation of therapy. The two most common adverse effects are influenza-like symptoms (fatigue, headache, fever, rigors), asthenia, myalgia, and psychiatric effects, specifically depression, irritability, and insomnia. The incidence of depression is approximately 30% and occurs typically in the first few months of therapy (Lok and McMahon, 2009). An exact causative mechanism for interferon-induced depression has not been fully elucidated. Risk factors linked to the development of depression include a premorbid presence of mood and anxiety symptoms. To a lesser extent, history of depression, higher doses of interferon, and female gender are also considered risk factors. Two syndromes describe interferon-induced depression: a depression-specific syndrome characterized by mood, anxiety, and cognitive complaints and a neurovegetative syndrome characterized by fatigue, anorexia, pain, and psychomotor slowing. Preassessment for neuropsychiatric conditions should be completed prior to initiating interferon therapy. Selective serotonin reuptake inhibitors (SSRIs) have been studied in both prevention and treatment of symptoms associated with depression-specific syndromes. Some centers

NEUROTOXICITY OF COMMONLY USED HEPATIC DRUGS have a low threshold for initiating SSRIs, even prior to interferon therapy. While debatable, consideration of prophylactic antidepressants use should be given for patients with risk factors for the development of depression. Use of certain antidepressants (citalopram, fluoxetine, imipramine, nortriptyline, paroxetine, sertraline) has over an 85% success rate in treating interferon-induced depression. Consideration should be given to which antidepressant to use, based on the particular adverse effects of these agents (Lok and McMahon, 2009).

Ribavirin Ribavirin is indicated for the treatment of chronic hepatitis C in combination with interferon a-2B (pegylated and nonpegylated) in patients with compensated liver disease. (Rebetol®, 2013) (ribavirin) monotherapy is not effective for the treatment of hepatitis C (37).

MECHANISM OF ACTION Ribavirin is a neucleoside analog and has antiviral activity against multiple RNA viruses. However, its mechanism in the treatment of hepatitis C has not been fully elucidated. It is proposed that ribavirin has effects on the host immune response (Lau et al., 2002).

NEUROTOXICITY The most concerning adverse effect associated with ribavirin use is hemolytic anemia. Other adverse effects include fatigue, leukopenia, pruritis, rash, and gout. Ribavirin is a well known teratogenic drug and is US Food and Drug Administration (FDA) pregnancy category X. Clear recommendations on use in women of childbearing age are outlined within the prescribing information (Epivir-HBV, 2011).

Lamivudine ®

Lamivudine (Epivir-HBV ) is approved for the treatment of chronic hepatitis B and may be considered as initial therapy for patients with compensated liver disease. This indication is supported by 1 year data of histologic and serologic responses in adult patients. Advantages of this therapy over IFN-a or adefovir as first line include lower cost and tolerability. Disadvantages are the potential for increased resistance and worsening of hepatic disease.

MECHANISM OF ACTION Lamivudine is a synthetic nucleoside analog and is incorporated into viral DNA by HBV polymerase resulting in premature DNA termination (Lau et al., 2002).

679

NEUROTOXICITY Lamivudine is generally well tolerated. A black box warning within the prescribing information states lactic acidosis and severe hepatomegaly with steatosis, including fatal cases, have been reported. Female gender, obesity, pregnancy, and prolonged exposure may increase the risk of these occurring. While these adverse effects are rare, there should be heightened consideration of encephalopathy related to these metabolic processes in critically ill patients presenting on lamivudine. In addition, severe acute exacerbations of hepatitis B have been reported in patients who have discontinued antihepatitis B therapy, including Epivir-HBV®. Less severe CNS effects include headache (21%), fatigue and malaise (24%), and insomnia (11%). Musculoskeletal pain (12%), neuropathy (12%), and myalgias (8–14%) also commonly occurred. In clinical trials of lamivudine for HBV, increases in creatine kinase levels were observed in 9% of patients (Lau et al., 2002; Lai et al., 2006).

Adefovir dipivoxil (Hepsera®) Adefovir dipivoxil is a prodrug of adefovir, a nucleotide analog of adenosine monophosphate. Hepsera® is approved for the treatment of chronic hepatitis B in adults with evidence of active viral replication and either evidence of persistent elevations in serum aminotransferases or histologically active disease (Hepsera, 2009).

MECHANISM OF ACTION Adefovir is phosphorylated to its active metabolite, adefovir diphosphate, which inhibits HBV DNA polymerase resulting in inhibition of viral replication.

NEUROTOXICITY Adefovir has multiple black box warnings within its prescribing information, including lactic acidosis and severe hepatomegaly with steatosis; severe, acute exacerbation of hepatitis B upon discontinuation; and cautious use in patients with renal dysfunction since chronic administration may result in nephrotoxicity. CNS toxicities associated with adefovir are minimal. In clinical studies adefovir had a similar side-effect profile to placebo. More commonly occurring central nervous system effects are headache (25%) and asthenia (13%). These adverse effects, however, have not been reported to significantly result in discontinuation of patient therapy. If patients experience headache, it is recommended they consult their healthcare provider for appropriate selection of analgesic therapy (Hadziyannis et al., 2003; Marcellin et al., 2003).

680

C.L. AHRENS AND E.M. MANNO

Entecavir (Baraclude®) Entecavir is approved for the treatment of hepatitis B infection with compensated or decompensated liver disease in adults with evidence of active viral replication and either evidence of persistent transaminase elevations or histologically active disease. Entecavir can also be used for patients with lamivudine-resistant viremia. Adjustment in dosing should be considered for patients with CrCl < 50 mL/min (Baraclude, 2010).

MECHANISM OF ACTION Entecavir is a guanosine nucleoside analog. Intracellularly it is phosphorylated to guanosine triphosphate which competes with natural substrates to inhibit HBV reverse transcriptase in three activities: (1) base priming, (2) reverse transcription of the negative strand DNA from the pregenomic messenger RNA, (3) synthesis of the positive strand of HBV DNA (Baraclude, 2010).

NEUROTOXICITY The adverse effect profile of entecavir is similar to lamivudine, including the same black box warnings (Sherman et al., 2006; Baraclude, 2010). Common CNS effects include pyrexia (14% with decompenstated liver disease), headache (2–4%), fatigue (1–3%), and dizziness. In preclinical animal trials, there was a higher incidence of solid tumors with high-dose, prolonged administration of entecavir (Baraclude, 2010). Studies are ongoing to evaluate long-term treatment effects with entecavir.

Telbivudine (Tyzeka®) Telbivudine is indicated for the treatment of chronic HBV in adult patients. It is well tolerated and has been shown to be more efficacious compared to lamivudine and adefovir in treating compensated chronic HBV (Chan et al., 2007; Lai et al., 2007; Liaw et al., 2009). Its use in special populations, such as patients with renal insufficiency, has not been reported.

MECHANISM OF ACTION Telbivudine is a synthetic thymidine nucleoside analog reverse transcriptase inhibitor. Intracellularly it is phosphorylated to its active triphosphate form which competes with naturally occurring thymidine triphosphate for HBV viral DNA elongation. This incorporation into viral DNA results in DNA chain termination (Bryant et al., 2001).

NEUROTOXICITY The same boxed warning which exists for other nucleoside analogs is included in the prescribing information

for telbivudine. In clinical trials comparing telbivudine to lamivudine, side-effects were similar with the exception of increased creatinine kinase levels in the telbivudine patients (Liaw et al., 2009). During these trials, the most commonly occurring neurologic adverse effects seem with telbivudine included headache (10%), dizziness (4%), and insomnia (3%) (Baraclude, 2010). Peripheral neuropathy has been reported with telbivudine either as monotherapy ( 20 ng/dL and a serum aldosterone level of over 15 ng/dL suggest the diagnosis of primary hyperaldosteronism (Larsen et al., 2003). The second step in diagnosis consists of determining if the cause is an adrenal adenoma or bilateral adrenal hyperplasia. The postural challenge test is useful in this regard (Larsen et al., 2003). A baseline plasma aldosterone of < 20 ng/dL that increases upon standing suggests bilateral hyperplasia. With adenomas, baseline levels are over 20 ng/dL and decrease upon standing as a result of decreased stimulation of ACTH. After this test is performed, an abdominal CT scan or other imaging helps to localize the tumor (Larsen et al., 2003).

Treatment The treatment of hyperaldosteronism consists of removal of the adenoma by laparoscopic adrenalectomy (Duncan et al., 2000). Ablative procedures using percutaneous acetic acid injection may also be effective for unilateral adenomas (Minowada et al., 2003). This approach may cause marked reduction of aldosterone secretion with orthostatic hypotension (Milsom et al., 1986). Hypertension may persist after treatment in as many as 40–65% of patients, likely caused by nephrosclerosis after a prolonged period of uncontrolled hypertension (Milsom et al., 1986; Horito et al., 2001; Larsen et al., 2003). Mineralocorticoid receptor antagonists, such as spironolactone and eplerenone, may be used to normalize

NEUROLOGIC COMPLICATIONS OF DISORDERS OF THE ADRENAL GLANDS blood pressure and serum potassium levels, especially in patients with idiopathic hyperaldosteronism (Brown et al., 1972; Lim et al., 2001). If patients fail to tolerate any of these treatments, the potassium-sparing diuretic amiloride can be used. Amiloride has been shown to reduce the systolic and diastolic blood pressure values and increase plasma potassium levels (Griffing et al., 1982). Hydrochlorothiazide can be added for better control of arterial hypertension.

DISORDERS OF THE ADRENAL MEDULLA The adrenal medulla is the area of the adrenal gland that produces catecholamines from secreting cells called chromaffin cells or pheochromocytes because they stain brown with chromium salts. Tumors affecting the adrenal medulla cause increased secretion of norepinephrine and epinephrine, and are called pheochromocytomas. Pheochromocytomas are rare tumors that can be single or multiple, benign or malignant.







Historical aspects The association between adrenal medullary tumors and their symptom spectrum was first noticed by Fra¨nkel in 1886, when he described a patient who had intermittent attacks of palpitations, anxiety, vertigo, headaches, and chest pain, as well as cold sweats and vomiting. When this patient died, her autopsy showed bilateral adrenal tumors (Frankel, 1984). The name pheochromocytoma was proposed by Pick in 1912 (Larsen et al., 2003). This term comes from the Greek words phaios (“dusky”) and chroma (“color”). In 1926, Roux in Switzerland and Charles Mayo in Rochester performed successful surgical removal of adrenal pheochromocytomas (Larsen et al., 2003). In 1929, it was discovered that pheochromocytomas contained an excess of a pressor substance (Manger and Gifford, 1996); subsequently, catecholamines were isolated from these tumors (Manger and Gifford, 1996).







Etiology Catecholamine-secreting tumors arising from the adrenal medulla are called pheocromocytomas. Those tumors arising in the sympathetic ganglia are called extra-adrenal paragangliomas (Lloyd et al., 2004). Both have similar clinical presentations and are treated similarly. A number of genetic conditions associated with pheochromocytomas have been described: ●

Multiple endocrine neoplasia type 2A (MEN 2A), or Sipple syndrome, is an autosomal dominant disease characterized by primary hyperparathyroidism,

765

medullary carcinoma of the thyroid, and bilateral pheochromocytomas (Marx, 2005; Peczkowska and Januszewic, 2005). Multiple endocrine neoplasia type 2B (MEN 2B) is also an autosomal dominant disorder and is phenotypically characterized by the association of pheochromocytomas, mucosal neuromas, and thickening of the optic nerves. Patients may also have intestinal ganglioneuromas. These tumors produce predominantly epinephrine and metaneprhine (Carling, 2005; Marx, 2005; Peczkowska and Januszewicz, 2005). Many patients with MEN 2A or 2B have mutations of the RET proto-oncogene, and genetic testing for this mutation is now commercially available (Larsen et al., 2003). Von Hippel—Lindau disease is another autosomal dominant disorder. Which is characterized by the presence of paragangliomas, pheochromocytomas, retinal angiomas, cerebellar hemangioblastomas, renal and pancreatic cysts, and renal cell carcinomas. Most of these tumors produce predominantly norepinephrine and normetanephrine (Larsen et al., 2003). Neurofibromatosis type 1 (NF1) is an autosomal dominant disease characterized by neurofibromas, optic nerve gliomas, sphenoid dysplasia, cafe´ au lait spots, iris hamartomas, and axillary/inguinal freckling. This disorder is caused by a mutation in the NF1 gene located on chromosome 17, which causes a decreased production of neurofibromin (Walther et al., 1999). Familial paraganglioma is an autosomal dominant disorder characterized by paragangliomas localized in the head and neck. These tumors are usually benign. Symptoms depend on their local invasion and degree of vascularization. Around 10% have a malignant evolution. Prognosis depends on the extension of the disease at the time of diagnosis (Larsen et al., 2003). Finally, other neurocutaneous disorders associated with catecholamine-secreting tumors include ataxia-telangiectasia, tuberous sclerosis complex, and Sturge–Weber syndrome.

Clinical manifestations Stressful stimuli such as anesthesia, hypoglycemia, or heart attacks increase the secretion of catecholamines, which have a very active role in the cardiovascular system and metabolic processes, increasing heart rate, blood pressure, myocardial contractibility and cardiac conduction abnormalities. Clinical characteristics of pheochromocytomas are related to the catecholamines

766

T.E. BERTORINI AND A. PEREZ

effects. The most common is arterial hypertension, which can be severe and can be accompanied by headaches, palpitations, and diaphoresis (Bravo and Gifford, 1984). Tumors secreting norepinephrine usually produce severe sustained hypertension, whereas those that secrete primarily epinephrine produce episodic hypertensive crises. Other symptoms include increased sweating, tremors, weakness, and anxiety associated with palpitations. The triad of episodic headaches, diaphoresis, and palpitations has a specificity of over 60% for pheochromocytomas (Stein and Black, 1991). Angina, nausea, constipation, hyperglycemia, diabetes, hypercalcemia, Raynaud phenomena, and livedo reticularis may be present in some patients. Hypertensive crisis may be precipitated by minor surgical procedures. Neurologic manifestations of pheochromocytoma are usually caused by changes in blood pressure, and include episodic headaches and sometimes ischemic or hemorrhagic cerebrovascular events (Eclavea et al., 1997; Lehmann et al., 1999; Dagartzikas et al., 2002). Musculoskeletal pain, sometimes with radiculopathies, can be caused by bone metastasis (Lynn et al., 1987). Rarely, patients may have seizures caused by either severe hypertension or strokes (Leiba et al., 2003). Recurrent syncopal episodes may result from hypotension due to downregulation of catecholamine receptors caused by chronic exposure to the neurotransmitter and to volume depletion secondary to inhibition of the renin– angiotensin system (Young, 1993). Differential diagnosis of pheocromocytoma is very extensive and includes hyperthyroidism, anxiety, panic attacks, migraines, and drug abuse. Pheocromocytomas may also produce somatostatin and ACTH, causing symptoms resembling those in Cushing syndrome (Larsen et al., 2003).

Diagnosis Diagnosis of catecholamine-secreting tumors is based on measurement of catecholamines, particularly plasma and urine metanephrines (Leiba et al., 2003). Measurements of other catecholamines, including normetanephrine, is also is useful. Other disorders can raise levels of catecholamines and metanephrines, and they should be considered in the interpretation of the tests. These include withdrawal from medications or drugs (e.g., clonidine, alcohol), subarachnoid hemorrhage, migraine headaches, preeclampsia, and the use of medications such as tricyclic antidepressants, levodopa, buspirone, prochlorperazine, and acetaminophen (Young, 1993).

The clonidine suppression test is highly sensitive to distinguish between pheochromocytoma and other causes of increased plasma catecholamines. Clonidine is a centrally acting a-adrenergic receptor agonist that normally suppresses the release of catecholamines from neurons, but does not affect the secretion from tumors. Catecholamine and metanephrine are measured before and after clonidine (0.3 mg) is administered orally (Sjoberg et al., 1992). In patients with essential hypertension, their concentrations decrease significantly, while in patients with pheochromocytoma the concentrations do not change (Sjoberg et al., 1992; Eisenhofer et al., 2001, 2004). Imaging techniques are used to localize the tumor and include CAT scan, MRI, and particularly the radioactive iodine with metaiodobenzylguanidine scan (Ilias and Pacak, 2004; Brink et al., 2005).

Treatment Treatment of pheocromocytomas consists of complete surgical resection of the tumors. Because sustained hypertension during and after surgery is the most serious complication, a careful preoperative and pharmacological preparation is crucial. Presurgical preparation includes administration of an a-adrenergic blocker (e.g., phenoxybenzamine) for control of blood pressure and prevention of arrhythmias. A b-adrenergic antagonist also is used after a-adrenergic blockade is obtained. Metyrosine (a-methylparatyrosine) is also used in this setting because it is a catecholamine synthesis inhibitor with antihypertensive properties. This should be used with caution and only when other agents have been ineffective. Treatment also includes acute management of hypertensive crisis with short-acting antihypertensive drugs (Larsen et al., 2003).

ACKNOWLEDGEMENTS The authors would like to thank Dr. Joseph Fisher for reviewing the manuscript, Kay Daughterty for editorial help, and Cindy Culver for help in typing the manuscript.

REFERENCES Addison T (1855). On the Constitutional and Local Effects of Disease of the Supra-Renal Capsules. Highley, London. Al Sabri AM, Smith N, Busuttil A (1997). Sudden death due to auto-immune Addison’s disease in a 12-year-old girl. J Legal Med 110: 278–280. Anglin RE, Rosebush PI, Mazurek MF (2006). The neuropsychiatric profile of Addison’s disease: revising a forgotten phenomenon. J Neuropsychiatry Clin Neurosci 18: 450–459. Arlt W, Allolio B (2003). Adrenal insufficiency. Lancet 361: 1881–1893.

NEUROLOGIC COMPLICATIONS OF DISORDERS OF THE ADRENAL GLANDS Aubourg P (1996). X-linked adrenoleukodystrophy. In: HW Moser (Ed.), Neurodystrophies and Neurolipidoses. Handbook of Clinical Neurology, vol. 22 (66). Elsevier, Amsterdam, pp. 447–483. Aubourg P (2007). X-linked adrenoleukodystrophy. Ann Endocrinol (Paris) 68: 403–411. Aubourg P, Adamsbaum C, Lavallard-Rousseau MC et al. (1993). A two-year trial of oleic and erucic acids. (“Lorenzo’s oil”) as treatment for adrenomyeloneuropathy. N Engl J Med 329: 745–752. Bakaeen FG, Walkes JC, Reardon MJ (2005). Heparininduced thrombocytopenia associated with bilateral adrenal hemorrhage after coronary artery bypass surgery. Ann Thorac Surg 79: 1388–1390. Barbieri F, Filla A, Grossi D et al. (1982). Clinical and computerized tomographic study of a case of Schilder’s disease. Acta Neurol (Napoli) 4: 57–61. Bautista J, Gil-Neciga E, Gil-Peralta A (1979). Hypokalemic periodic paralysis in primary hyperaldosteronism: subclinical myopathy with atrophy of the type 2A muscle fibers as the most pronounced alteration. Eur Neurol 18: 415–420. Benvenga S, Rodolico C, Trimarchi F et al. (2001). Endocrine evaluation for muscle pain. J R Soc Med 94: 405–407. Berenbaum SA, Snyder E (1995). Early hormonal influences on childhood sex-typed activity and playmate preferences: implications for the development of sexual orientation. Dev Psychol 31: 31–42. Berneis K, Staub JJ, Gessler A et al. (2002). Combined stimulation of adrenocorticotropin and compound-S by single dose metyrapone test as an outpatient procedure to assess hypothalamic–pituitary–adrenal function. J Clin Endocrinol Metab 87: 5470–5475. Bertorini TE, Perez AS, Tammaa M (2008). Disorders of the adrenal glands and neuroendocrine tumors. In: J Biller (Ed.), The Interface of Neurology and Internal Medicine. Lippincott Williams and Wilkins, Philadelphia, pp. 482–496. Betterle C, Dal Pra C, Mantero F et al. (2002). Autoimmune adrenal insufficiency and autoimmune polyendocrine syndromes: autoantibodies, autoantigens, and their applicability in diagnosis and disease prediction. Endocr Rev 23: 327–364. Bhuwan PG, Omkar NM, William ED et al. (1983). Evoked response studies in patients with adrenoleukodystrophy and heterozygous relatives. Arch Neurol 40: 356–359. Bishop MF (1949). The history of the discovery of Addison’s disease. Proc R Soc Med 43 (October 26). Bolton CF (2005). Neuromuscular manifestations of critical illness. Muscle Nerve 32: 140–163. Bravo EL, Gifford RW (1984). Current concepts Pheochromocytoma: diagnosis, localization and management. N Engl J Med 311: 1298–1303. Brian P, O’Neill MD, Hugo W et al. (1984). Adrenoleukodystrophy: clinical and biochemical manifestations in carriers. Neurology 34: 798–801. Brink I, Hoegerle S, Klisch J et al. (2005). Imaging of pheochromocytoma and paraganglioma. Fam Cancer 4: 61–68.

767

Brown JJ, Davies DL, Ferriss JB et al. (1972). Comparison of surgery and prolonged spironolactone therapy in patients with hypertension, aldosterone excess, and low plasma renin. BMJ 2: 729. Brown-Se´quard CE (1856). Recherches expe´rimentales sur la psysiologie et la pathologie des capsules surre´nales. Arch Gen Med 5: 385–401. Budka H, Sluga E, Heiss WD (1976). Spastic paraplegia associated with Addison’s disease: adult variant of adrenoleukodystrophy. J Neurol 213: 237–250. Carey R (1997). The changing clinical spectrum of adrenal insufficiency. Ann Intern Med 127: 1103–1105. Carling T (2005). Multiple endocrine neoplasia syndrome: genetic basis for clinical management. Curr Opin Oncol 17: 7–12. Choy EH, Corkill MM, Gibson T et al. (1991). Isolated ACTH deficiency presenting with bilateral frozen shoulder. Br J Rheumatol 30: 226–227. Coco G, Dal Pra C, Presotto F et al. (2006). Estimated risk for developing autoimmune Addison’s disease in patients with adrenal cortex autoantibodies. J Clin Endocrinol Metab 91: 1637–1645. Cohen SI, Marks IM (1961). Prolonged organic psychosis with recovery in Addison’s disease. J Neurol Neurosurg Psychiatry 24: 366–368. Conn JW, Knopf RF, Nesbit RM (1964). Clinical characteristics of primary aldosteronism from an analysis of 145 cases. Am J Surg 107: 159–172. Cushing H (1932). The basophil adenomas of the pituitary body and their clinical manifestations (pituitary basophilism). Bull John Hopkins Hosp 50: 137–195. Dagartzikas MI, Sprague K, Carter G et al. (2002). Cerebrovascular event, dilated cardiomyopathy, and pheochromocytoma. Pediatr Emerg Care 18: 33–35. Danon MJ, Carpenter S (1991). Myopathy with thick filament (myosin) loss following prolonged paralysis with vecuronium during steroid treatment. Muscle Nerve 14: 1131–1139. Danon MJ, Edinger J (1999). Steroid induced quadriplegic myopathy with selective thick filament loss: elevated proteasoma content suggestive of increased proteolysis of myosin (abstract). Neurology 52 (Suppl. 2): S123. David N (1995). Cushing’s syndrome. N Engl J Med 332: 791–803. Di Giovanni S, Molon A, Broccolini A et al. (2004). Constitutive activation of MAPK cascade in acute quadriplegic myopathy. Ann Neurol 55: 195–206. Dickstein G, Shechner C, Nicholson WE et al. (1991). Adrenocorticotropin stimulation test: effects of basal cortisol level, time of day, and suggested new sensitive low dose test. J Clin Endocrinol Metab 72: 773–778. Dittmann RW, Kappes MH, Kappes ME et al. (1990). Congenital adrenal hyperplasia II: gender-related behavior and attitudes in female salt-wasting and simple-virilizing patients. Psychoneuroendocrinology 15: 421–434. D€ okmetas HS, Kilicli F, Korkmaz S et al. (2006). Characteristic features of 20 patients with Sheehan’s syndrome. Gynecol Endocrinol 22: 279–283.

768

T.E. BERTORINI AND A. PEREZ

Dorin RI, Qualls CR, Crapo LM (2003). Diagnosis of adrenal insufficiency. Ann Intern Med 139: 194–204. Duncan JL 3rd, Fuhrman GM, Bolton JS et al. (2000). Laparoscopic adrenalectomy is superior to an open approach to treat primary hyperaldosteronism. Am Surg 66: 932–935. Eclavea A, Gagliardi JA, Jezior J et al. (1997). Phaeochromocytoma with central nervous system manifestations. Australas Radiol 41: 373–376. Eekhoff EM, van der Lubbe PA, Breedveld FC (1998). Flexion contractures associated with a malignant neoplasm: “A Paraneoplastic Syndrome?”. Clin Rheumatol 17: 157–159. Eisenhofer G, Hunynh TT, Hiroi M et al. (2001). Understanding catecholamine metabolism as a guide to the biochemical diagnosis of pheochromocytoma. Rev Endocr Metab Disord 2: 297–311. Eisenhofer G, Kipin IJ, Goldstein DS (2004). Catecholamine metabolism: a contemporary view with implications for physiology and medicine. Pharmacol Rev 56: 331–349. Emma F, Pizzini C, Tessa A et al. (2006). “Bartter-like” phenotype in Kearns–Sayre syndrome. Pediatr Nephrol 21: 355–360. Ernst M, Maheu FS, Schroth E et al. (2007). Amygdala function in adolescents with congenital adrenal hyperplasia: a model for the study of early steroid abnormalities. Neuropsychologia 45: 2104–2113. Eustachius B (1774). Tabulae Anatomicae. In: B Lancicius (Ed.), Amsterdam. Falorni A, Laureti S, Nikoshkov A et al. (1997). 21hydroxylase autoantibodies in adult patients with endocrine autoimmune diseases are highly specific for Addison’s disease Belgian Diabetes Registry. Clin Exp Immunol 107: 341–346. Falorni A, Laureti S, De Bellis A et al. (2004). Italian Addison network study: update of diagnostic criteria for the etiological classification of primary adrenal insufficiency. J Clin Endocrinol Metab 89: 1598–1604. Ferguson G, Irvin C, Cherniack R (1990). Effect of corticosteroids on respiratory musclehistopathology. Am Rev Respir Dis 142: 1047–1052. Fiad TM, Kirby JM, Cunningham SK et al. (1994). The overnight single-dose metyrapone test is a simple and reliable index of the hypothalamic–pituitary–adrenal axis. Clin Endocrinol (Oxf) 40: 603–609. Findling JW, Doppman JL (1994). Biochemical and radiologic diagnosis of Cushing’s syndrome. Endocrinol Metab Clin North Am 23: 511–537. Findling JW, Raff H, Aron DC (2004). The low-dose dexamethasone suppression test: a reevaluation in patients with Cushing’s syndrome. J Clin Endocrinol Metab 89: 1222–1226. Finucane FM, Liew A, Thornton E et al. (2008). Clinical insights into the safety and utility of the insulin tolerance test (ITT) in the assessment of the hypothalamo–pituitary–adrenal axis. Clin Endocrinol (Oxf) 69: 603–607. Frankel F (1984). Classics in oncology A case of bilateral completely latent adrenal tumor and concurrent nephritis

with changes in the circulatory system and retinitis, 1886. CA Cancer J Clin 34: 93–106. Fujihara K, Miyoshi T, Yamaguchi Y et al. (1990). Tetany as a sole manifestation in a patient with Bartter’s syndrome and successful treatment with indomethacin. Rinsho Shinkeigaku 30: 519–532. Gambelunghe G, Falorni A, Ghaderi M et al. (1999). Microsatellite polymorphism of the MHC class I chainrelated (MIC-A and MIC-B) genes marks the risk for autoimmune Addison’s disease. J Clin Endocrinol Metab 84: 3701–3707. Gonzalbez J, Villabona C, Ramon J et al. (2000). Establishment of reference values for standard dose short Synacthen test (250 microgram), low dose short Synacthen test (1 microgram) and insulin tolerance test for assessment of the hypothalamo–pituitary–adrenal axis in normal subjects. Clin Endocrinol (Oxford) 53: 199–204. Gould E, Cameron HA, Daniels DC et al. (1992). Adrenal hormones suppress cell division in the adult rat dentate gyrus. J Neurosci 12: 3642–3650. Griffin JW, Goren E, Schaumburg H (1977). Adrenomyeloneuropathy: a probable variant of adrenoleukodystrophy I Clinical and endocrinologic aspects. Neurology 27: 1107–1113. Griffing GT, Aurecchia SA, Sindler BH et al. (1982). The effect of amiloride on the renin– aldosterone system in primary hyperaldosteronism and Bartter’s syndrome. J Clin Pharmacol 22: 505–512. Grinspoon SK, Biller BM (1994). Clinical review 62: laboratory assessment of adrenal insufficiency. J Clin Endocrinol Metab 79: 923–931. Guo W, Burris TP, McCabe ER (1995). Expression of DAX-1, the gene responsible for X- linked adrenal hypoplasia congenital and hypogonadotropic hypogonadism, in the hypothalamic– pituitary–adrenal/gonadal axis. Biochem Mol Med 56: 8–13. Harris GW (1948). Neural control of the pituitary gland. Physiol Rev 28: 139–179. Hayashi Y, Hiyoshi T, Takemura T et al. (1989). Focal lymphocytic infiltration in the adrenal cortex of the elderly: immunohistological analysis of infiltrating lymphocytes. Clin Exp Immunol 77: 101–105. Henkin RI (1970). The effects of corticosteroids and ACTH on sensory systems. Prog Brain Res 32: 270–294. Henkin RU, Gill JR, Warmolts JR et al. (1963). Steroiddependent increase of nerve conduction velocity in adrenal insufficiency. J Clin Invest 42: 941. Henkin RI, Gill JR, Bartter FC (1973). Studies on taste thresholds in normal man and in patients with adrenal cortical insufficiency: the role of adrenal cortical steroids and of serum sodium concentration. J Clin Invest 42: 727–735. Horita Y, Inenaga T, Nakahama H et al. (2001). Cause of residual hypertension after adrenalectomy in patients with primary aldosteronism. Am J Kidney Dis 37 (5): 884–889. Huebner A, Elias L, Clark A (1999). ACTH resistance syndromes. J Pediatr Endocrinol Metab 12 (Suppl. 1): 277–293.

NEUROLOGIC COMPLICATIONS OF DISORDERS OF THE ADRENAL GLANDS Husebye E, Lovas K (2009). Pathogenesis of primary adrenal insufficiency. Best Pract Res Clin Endocrinol Metab 23: 147–157. Igarashi M, Belchis D, Suzuki K (1976). Fatty acid abnormality in adrenoleukodystrophy. J Neurochem 26: 851–860. Ilias I, Pacak K (2004). Current approaches and recommended algorithm for the diagnostic localization of pheochromocytoma. J Clin Endocrinol Metab 89: 479–491. Jennings CE, Owen CJ, Wilson V et al. (2005). A haplotype of the CYP27B1 promoter is associated with autoimmune Addison’s disease but not with Graves’ disease in a UK population. J Mol Endocrinol 34: 859–863. Kaga M, Furushima W, Inagaki M et al. (2009). Early neuropsychological signs of childhood adrenoleukodystrophy (ALD). Brain Dev 31: 558–561. Kelestimur F (2003). Sheehan’s syndrome. Pituitary 6: 181–188. K€ ohler B, Lin L, Ferraz-de-Souza B et al. (2008). Five novel mutations in steroidogenic factor 1 (SF1, NR5A1) in 46, XY patients with severe underandrogenization but without adrenal insufficiency. Hum Mutat 29: 59–64. Korenke GC, Hunneman DH, Kohler J et al. (1995). Glyceroltrioleate/glyceroltrierucate therapy in 16 patients with X-chromosomal adrenoleukodystrophy/adrenomyeloneuropathy: effect on clinical, biochemical and neurophysiological parameters. Eur J Pediatr 154: 64–70. Kriegel MA, Lohmann T, Gabler C et al. (2004). Defective suppressor function of human CD4þCD25þ regulatory T cells in autoimmune polyglandular syndrome type II. J Exp Med 199: 1285–1291. Lacomis D, Giuliani MJ, Van Cott A et al. (1996). Acute myopathy of intensive care: clinical, electrormyographic and pathological aspects. Ann Neurol 40: 645–654. Larsen PR, Kronenberg HM, Melmed S (Eds.), (2003). The Adrenal Cortex, and Endocrine Hypertension. Williams Textbook of Endocrinology. Saunders, Philadelphia, pp. 491–585. Laureti S, Aubourgh P, Calcinaro F et al. (1998). Etiological diagnosis of primary adrenal insufficiency using an origin flowchart of immune and biochemical markers. J Clin Endocrinol Metab 83: 3163–3168. Lehmann H, Nair NPV, Kline NS (1979). Beta-endorphin and naloxone in psychiatric patients: clinical and biological effects. Am J Psychiatry 136: 762–766. Lehmann FS, Weiss P, Ritz R et al. (1999). Reversible cerebral ischemia in patients with pheochromocytoma. J Endocrinol Invest 22: 212–214. Leiba A, Bar-Dayan Y, Leker RR et al. (2003). Seizures as a presenting symptom of pheochromocytoma in a young solider. J Hum Hypertens 17: 73–75. Lever EG, Stansfeld SA (1983). Addison’s disease, psychosis, and the syndrome of inappropriate secretion of ADH. Br J Psychiatry 143: 406–410. Li CH, Simpson ME, Evans HM (1943). Adrenocorticotrophic hormone. J Biol Chem 149: 413–424. Lim PO, Young WF, MacDonald TM (2001). A review of the medical treatment of primary aldosteronism. J Hypertens 19: 353–361.

769

Lindstrom LH, Widerlov E, Gunne LM et al. (1978). CSF: clinical correlations to some psychotic states. Acta Psychiatr Scand 57: 153–164. Lloyd TE, Chaudhry V (2011). Treatment and management of hereditary neuropathies. In: TE Bertorini (Ed.), Neuromuscular Disorders: Treatment and Management. Elsevier, Philadelphia, pp. 191–213. Lloyd RV, Tischer AS, Kimura N et al. (2004). Adrenal tumors: introduction. In: RA DeLellis, RV Lloyd, PU Heitz et al. (Eds.), World Health Organization Classification of Tumours: Pathology and Genetics of Tumours of Endocrine Organs. International Agency for Research on Cancer Press, Lyons, France, pp. 136–138. Loes DJ, Stillman AE, Hite S et al. (1994). Childhood cerebral form of adrenoleukodystrophy: short-term effect of bone marrow transplantation on brain MR observations. AJNR Am J Neuroradiol 15: 1767–1771. Lopez ER, Zwermann O, Segni M et al. (2004). A promoter polymorphism of the CYP27B1 gene is associated with Addison’s disease, Hashimoto’s thyroiditis, Graves’ disease and type 1 diabetes mellitus in Germans. Eur J Endocrinol 151: 193–197. Loriaux LD (2008). Jerome W Conn (1907–1994) Historical note. Endocrinologist 18: 159–160. Lovas K, Husebye ES, Holsten F et al. (2003). Sleep disturbances in patients with Addison’s disease. Eur J Endocrinol 148: 449–456. Lynn MD, Braunstein EM, Shaprio B (1987). Pheochromocytoma presenting as musculoskeletal pain from bone metastases. Skeletal Radiol 16: 552–555. McFarland HR (1963). Addison’s disease and related psychoses. Compr Psychiatry 4: 90–95. McNeill TH, Masters JN, Finch CE (1991). Effect of chronic adrenalectomy on neuron loss and distribution of sulfated glycoprotein-2 in the dentate gyrus of prepubertal rats. Exp Neurol 111: 140–144. Mamoli B, Graf M, Toifl K (1979). EEG, pattern-evoked potentials and nerve conduction velocity in a family with adrenoleukodystrophy. Electroencephalogr Clin Neurophysiol 47: 411–419. Manger WM, Gifford RW (1996). Background and importance and diagnosis. In: WM Manger, RW Gifford (Eds.), Clinical and Experimental Pheochromocytoma. 2nd edn Blackwell Science, Cambridge, pp. 1–7, 205–332. Marx SJ (2005). Molecular genetics of multiple endocrine neoplasia types 1 and 2. Nat Rev Cancer 5: 367–375. Mauri M, Sinforiani E, Bono G et al. (1993). Memory impairment in Cushing’s disease. Acta Neurol Scand 87: 52–55. Mayenknecht J, Diederich S, Bahr V et al. (1998). Comparison of low and high dose corticotropin stimulation tests in patients with pituitary disease. J Clin Endocrinol Metab 83: 1558–1562. Medvei VC (1991). The history of Cushing’s disease: a controversial tale. J R Soc Med 84: 363–366. Meyer-Bahlburg HE (2011). Brain development and cognitive, psychosocial, and psychiatric functioning in classical 21-hydroxylase deficiency. Endocr Dev 20: 88–95.

770

T.E. BERTORINI AND A. PEREZ

Michiels C (2004). Physiological and pathological responses to hypoxia. Am J Pathol 164: 1875–1882. Milsom SR, Espiner EA, Nicholls MG et al. (1986). The blood pressure response to unilateral adrenalectomy in primary hyperaldosteronism. QJM 61: 1141–1151. Minowada S, Fujimura T, Takahashi N et al. (2003). Computed tomography-guided percutaneous acetic acid injection therapy for functioning adrenocortical adenoma. J Clin Endocrinol Metab 88: 5814–5817. Mizoguchi K, Ishige A, Takeda S et al. (2004). Endogenous glucocorticoids are essential for maintaining prefrontal cortical cognitive function. J Neurosci 24: 5492–5499. Momose KJ, Killberg RN, Kilman B (1971). High incidence of cortical atrophy of the cerebral and cerebellar hemispheres in Cushing’s disease. Radiology 99: 341–348. Moritz ML, Ayus CJ (2003). The pathophysiology and treatment of hyponatraemic encephalopathy: an update. Nephrol Dial Transplant 18: 2486–2491. Moser AB, Moser HW (1999). The prenatal diagnosis of Xlinked adrenoleukodystrophy. Prenat Diagn 19: 46–48. Moser HW, Moser A, Powers JM et al. (1982). The prenatal diagnosis of adrenoleukodystrophy Demonstration of increased hexacosanoic acid levels in cultured amniocytes and fetal adrenal gland. Pediatr Res 16: 172–175. Moser HW, Moser AE, Trojak JE et al. (1983). Identification of female carriers of adrenoleukodystrophy. J Pediatr 103: 54–59. Moser HW, Raymond GV, Dubey P (2005a). Adrenoleukodystrophy: new approaches to a neurodegenerative disease. JAMA 294: 3131–3134. Moser HW, Raymond GV, Lu SE et al. (2005b). Follow-up of 89 asymptomatic patients with adrenoleukodystrophy treated with Lorenzo’s oil. Arch Neurol 62: 1073–1080. Mosser J, Douar AM, Sarde CO et al. (1993). Putative Xlinked adrenoleukodystrophy gene shares unexpected homology with ABC transporters. Nature 361: 726–730. Myhre AG, Undlien DE, Lovas K et al. (2002). Autoimmune adrenocortical failure in Norway autoantibodies and human leukocyte antigen class II associations related to clinical features. J Clin Endocrinol Metabol 23: 618–623. Myhre AG, Aarsetoy H, Undlien DE et al. (2003). High frequency of celiac disease among patients with autoimmune adrenocortical failure. Scand J Gastroenterol 38: 511–515. Nakamagoe K, Ohkoshi N, Ishii A et al. (1994). Syndrome of contracture facio-brachio-abdomino-crurale en flexion in a case of isolated ACTH deficiency – biopsy findings of muscle and nerve. Rinsho Shinkeigaku 34: 250–254. Nass R, Heier L, Moshang T et al. (1997). Magnetic resonance imaging in the congenital adrenal hyperplasia population: increased frequency of white-matter abnormalities and temporal lobe atrophy. J Child Neurol 12: 181–186. Nejentsev S, Howson JM (2007). Localization of type 1 diabetes susceptibility to the MHC class I genes HLA-B and HLA-A. Nature 450: 887–892. Nerup J, Andersen V, Bendixen G (1969). Anti-adrenal, cellular hypersensitivity in Addison’s disease. Clin Exp Immunol 4: 355–363.

Nerup J, Andersen V, Bendixen G (1970). Anti-adrenal cellular hypersensitivity in Addison’s disease IV In vivo and in vitro investigations on the mitochondrial fraction. Clin Exp Immunol 6: 733–739. Nieman LK (2013a). Clinical Manifestations of Cushing’s Syndrome. In: UpToDate, BD Rose (Eds.), UpToDate. Copyright @ 2007 UptoDate, Inc, Waltham, MA. Nieman LK (2013b). Establishing the Diagnosis of Cushing’s Syndrome. In: UpTo Date, BD Rose (Eds.), UpToDate. Copyright @ 2007 UptoDate, Inc, Waltham, MA. Nieman LK (2013c). Establishing the Cause of Cushing’s Syndrome. In: UpTo Date, BD Rose (Eds.), UpToDate. Copyright @ 2007 UptoDate, Inc, Waltham, MA. Nieman LK (2013d). Clinical Manifestations of Adrenal Insufficiency in adults. In: A Lacroix, KA Martin (Eds.), Copyright 2013, UptoDate, Inc, Waltham, MA. Nishikawa T (2003). Flexion contractures possibly reflect the existence of hypocortisolism (editorial). Intern Med 42: 629–631. Odagaki T, Noguchi Y, Fukui T (2003). Flexion contractures of the legs as the initial manifestation of adrenocortical insufficiency. Intern Med 42: 710–713. Osler W (1896). On six cases of Addison’s disease with the report of a case greatly benefited by the use of suprarenal extract. Internat Med Mag 5: 3–11. Pani MA, Seissler J, Usadel KH et al. (2002). Vitamin D receptor genotype is associated with Addison’s disease. Eur J Endocrinol 147: 635–640. Peczkowska M, Januszewicz A (2005). Multiple endocrine neoplasia type 2. Fam Cancer 4: 25–36. Piedrola G, Casado JL, Lopez E et al. (1996). Clinical features of adrenal insufficiency in patients with acquired immunodeficiency syndrome. Clin Endocrinol (Oxf) 45: 97–101. Powers JM, Schaumburg HH (1973). The adrenal cortex in adreno-leukodystrophy. Arch Pathol 96: 305–310. Powers JM, Schaumburg HH (1974). Adrenoleukodystrophy: similar ultrastructural changes in adrenal cortical and Schwann cells. Arch Neurol 30: 406–408. Presotto F, Fornasini F, Betterle C et al. (2005). Acute adrenal failure as the heralding symptom of primary antiphospholipid syndrome: report of a case and review of the literature. Eur J Endocrinol 153: 507–514. Rajathurai A, Chazan BI, Jeans JE (1983). Self-mutilation as a feature of Addison’s disease. Br Med J 287: 1027. Raux-Demay MC, Girard F (1985). The physiology of corticotropin-releasing factor (CRF). Reprod Nutr Dev 25: 931–943. Reutens AT, Achermann JC, Ito M et al. (1999). Clinical and functional effects on mutations in the DAX-1 gene in patients with adrenal hypoplasia congenital. J Clin Endocrinol Metab 84: 504–511. Reynolds RM, Stewart PM, Seckl JR et al. (2006). Assessing the HPA axis in patients with pituitary disease: a UK survey. Clin Endocrinol (Oxf) 64: 82–85. Romney SJ, Chik LC (2001). Frozen shoulders: an endocrine disease? A case report of isolated ACTH deficiency. Endocrinologist 11: 429–431.

NEUROLOGIC COMPLICATIONS OF DISORDERS OF THE ADRENAL GLANDS Salvatori R (2005). Adrenal insufficiency. JAMA 294: 2481–2488. Sanaker PS, Husebye ES, Fondenes O et al. (2007). Clinical evolution of Kearns–Sayre syndrome with polyendocrinopathy and respiratory failure. Acta Neurol Scand Suppl 187: 64–67. Sauter NP, Toni R, McLaughlin CD et al. (1990). Isolated adrenocorticotropin deficiency associated with autoantibody to a corticotroph antigen that is not adrenocorticotropin or other proopiomelanocortin-derived peptides. J Clin Endocrinol Metab 70: 1391–1397. Sawczuk IS, Reitelman C, Libby C et al. (1986). CT findings in Addison’s disease caused by tuberculosis. Urol Radiol 8: 44–45. Schaumburg HH, Powers JM, Raine CS et al. (1975). Adrenoleukodystrophy: a clinical and pathological study of 17 cases. Arch Neurol 32: 577–591. Shapiro E, Krivit W, Lockman L et al. (2000). Long-term effect of bone-marrow transplantation for childhood-onset cerebral X-linked adrenoleukodystrophy. Lancet 356: 713–718. Sjarif DR, Ploos van Amstel JK, Duran M et al. (2000). Isolated and contiguous glycerol kinase gene disorders: a review. J Inherit Metab Dis 23: 529–547. Sjoberg RJ, Simcic KJ, Kidd GS (1992). The clonidine suppression test for pheochromocytoma A review of its utility and pitfalls. Arch Intern Med 152: 1193–1197. Skinningsrud B, Husebye ES, Gervin K et al. (2008). Mutation screening of PTPN22: association of the 1858T-allele with Addison’s disease. Eur J Hum Genet 16: 977–982. Sloviter RS, Sollas AL, Dean E et al. (1993). Adrenalectomyinduced granule cell degeneration in the rat hippocampal dentate gyrus: characterization of an in vivo model of controlled neuronal death. J Comp Neurol 330: 324–336. Smith CM (1958). Paranoid behavior in Addison’s disease: report of a case. Can Psychiatr Assoc J 3: 145–154. Speiser PW, White PC (2003). Congenital adrenal hyperplasia. N Engl J Med 349: 776–788. Spoto B, Furlo G, Gervasi A et al. (2004). Familial hyperaldosteronism. G Ital Nefrol 21: 139–143. Spurek M, Taylor-Gjevre R, Van Uum S et al. (2004). Adrenomyeloneuropathy as a cause of primary adrenal insufficiency and spastic paraparesis. CMAJ 171: 1073–1077. Starkman MN, Gebarski SS, Berent S et al. (1992). Hippocampal formation volume, memory dysfunction, and cortisol levels in patients with Cushing’s syndrome. Biol Psychiatry 32: 756–765. Stein PP, Black HR (1991). A simplified diagnostic approach to pheochromocytoma: a review of the literature and report of one institution’s experience. Medicine 70: 46–66. Steinpresis RE (1996). The behavioral and neurochemical effects of phencyclidine in humans and animals: some

771

implications for modeling psychosis. Behav Brain Res 74: 45–55. Stewart PM (2003). The adrenal cortex. In: PR Larsen, HM Kronenberg, S Melmed, KS Polonski (Eds.), Williams Textbook of Endocrinology. Saunders, Philadelphia, pp. 491–585. Syriou V, Moisidis A, Tamouridis N et al. (2008). Isolated adrenocorticotropin deficiency and flexion contractures syndrome. Hormones 7: 320–324. Talib A, Mahmood K, Jairmani KL et al. (2004). Hypokalemic quadriparesis with normotensive primary hyperaldosteronism. J Coll Physicians Surg Pak 14: 492–493. Ten S, New M, Maclaren N (2001). Clinical review 130: Addison’s disease 2001. J Clin Endocrinol Metabol 86: 2909–2922. Thompson WF (1973). Psychiatric aspects of Addison’s disease: report of a case. Med Ann Dist Columbia 42: 62–64. Ubogu EE, Ruff RL, Kaminski HJ (1994). Endocrine myopathy. In: A Engel, C Franzizi-Armstrong (Eds.), Myopathy. McGraw-Hill, New York, pp. 1713–1738. van Geel BM, Assies J, Haverkort E et al. (1999). Progression of abnormalities in adrenomyeloneuropathy and neurologically asymptomatic X-linked adrenoleukodystrophy despite treatment with “Lorenzo’s oil”. J Neurol Neurosurg Psychiatry 67: 290–299. Varadaraj R, Cooper AJ (1986). Addison’s disease presenting with psychiatric symptoms. Am J Psychiatry 143: 553–554. Vendrame F, Segni M, Grassetti D et al. (2006). Impaired caspase-3 expression by peripheral T cells in chronic autoimmune thyroiditis and in autoimmune polyendocrine syndrome-2. J Clin Endocrinol Metabol 91: 5064–5068. Walther MM, Herring JC, Enquist EE et al. (1999). von Recklinghausen’s disease and pheochromocytoma: Literature review. J Urol 162: 1582–1586. Weber KT, Singh KD, Hey JC (2002). Idiopathic intracranial hypertension with primary aldosteronism: report of 2 cases. Am J Med Sci 324: 45–50. Winqvist O, Karlsson FA (1992). 21-Hydroxilase, a major autoantige in idiopathic Addison’s disease. Lancet 339: 1559–1562. Wolfgang O (1996). Adrenal insufficiency. N Engl J Med 335: 1206–1212. Young WF Jr (1993). Pheocromocytoma 1926–1993. Trends Endocrinol Metab 4: 122–127. Yu L (1999). DRB1*04 and DQ alleles: expression of 21-hydroxilase autoantibodies and risk of progression to Addison’s disease. J Clin Endocrinol Metab 84: 328–335. Zucker KJ, Bradley SJ, Oliver G et al. (1996). Psychosexual development of women with congenital adrenal hyperplasia. Horm Behav 30: 300–318.

Handbook of Clinical Neurology, Vol. 120 (3rd series) Neurologic Aspects of Systemic Disease Part II Jose Biller and Jose M. Ferro, Editors © 2014 Elsevier B.V. All rights reserved

Chapter 51

Diabetic neuropathy GERALD CHARNOGURSKY*, HONG LEE, AND NORMA LOPEZ Division of Endocrinology and Metabolism, Loyola University Chicago, Stritch School of Medicine, Maywood, IL, USA

HISTORY AND EPIDEMIOLOGY Diabetes mellitus (DM) is a disorder characterized by impaired carbohydrate, protein, and fat metabolism that is due to either the insufficient production of insulin in type 1 DM or to target tissue insulin resistance and impaired insulin response in type 2 DM. The American Diabetes Association estimates over 8.3% of the US population had diabetes in 2011 including both children and adults. The prevalence of diabetes varies depending on the type of diabetes and the age of the patients. The prevalence at any given time is likely underestimated given that type 2 DM and impaired glucose tolerance often remain undiagnosed for many years. Complications from diabetes include heart disease and stroke, blindness, kidney disease, neuropathy, and limb amputations. The American Diabetes Association estimated that the cost of treating the disease in 2007 was US$174 billion. Peripheral diabetic neuropathy is defined as the presence of symptoms and or signs of peripheral nerve dysfunction in people with diabetes after other causes of the dysfunction have been excluded. Boulton and colleagues describe diabetic neuropathy as consisting of a minimum of two of the following abnormalities: symptoms, signs, nerve conduction abnormalities, quantitative sensory test results, or a quantitative autonomic test result. For clinical studies, they suggest a system requiring that one of the two abnormalities include quantitative test results or electrophysiology findings (Boulton et al., 2004, 2005). Neuropathy resulting from diabetes is estimated to affect 60–70% of people with diabetes if mild to severe forms of nervous system damage are included. The prevalence varies among series depending on the definition used. Dyck and colleagues found that some form of neuropathy – peripheral or autonomic – was present in 66% of all diabetic patients in their series (Dyck et al., 1993) using a definition based on symptoms, signs, nerve conductions studies, sensory testing, and autonomic testing. Prevalence estimates vary due to differences

in age, duration of diabetes, the definition of neuropathy used, presence or absence of pain, and whether or not other forms of neuropathy are excluded. Prevalence of painful diabetic neuropathy is estimated to be 10–20% (Galer et al., 2000). Prospective studies confirming the relationship between impaired glucose tolerance (IGT) and neuropathy are lacking but some studies suggest that IGT may lead to neuropathy. Prevalence of IGT varied from 34% to 56% in two studies of patients with idiopathic peripheral neuropathy, as high as three times the prevalence of age-matched controls with normal glucose tolerance (Sumner et al., 2003; Singleton and Smith, 2007). While some studies do suggest IGT is an important cause of polyneuropathy, other studies do not confirm this relationship, possibly due the disparity in patient populations, controls, and the assessment of glycemic exposure and diabetic complications (Dyck et al., 2007). The MONIKA/ KORA Augsburg surveys S2 and S3 revealed polyneuropathy in 28% of diabetics, 13% of patients with impaired glucose tolerance, 11.3% of patient with impaired fasting glucose and 7.4% of subjects with normal glucose tolerance (Ziegler et al., 2008). Diabetic autonomic neuropathy (DAN) prevalence also varies depending on the study, from as low as 8% in new type 1 diabetics (Ziegler et al., 1992) to as high as 80% and 90% in early type 2 diabetics and in insulindependent potential pancreas transplant recipients (Zhong et al., 1981; Kennedy et al., 1995). Differences in the definition of DAN, testing modalities, duration and type of diabetes, patient cohort and glycemic control are some of the confounders reported. Ziegler studied over 1000 diabetic patients from over 20 centers in Europe and found that 25% of type 1 and 34% of type 2 diabetics had abnormal findings in more than two out of six autonomic function tests (Ziegler et al., 1992). Prevalence studies have found differences in neuropathic abnormalities according to ethnicity. Diabetic neuropathy in non-Hispanic whites compared to

*Correspondence to: Gerald Charnogursky, M.D., Division of Endocrinology and Metabolism, Loyola University Stritch School of Medicine, 2160 S. First Ave., Maywood, IL 60153, USA. Tel: þ1-708-216-6252, Fax: þ1-708-216-5936, E-mail: [email protected]

774 G. CHARNOGURSKY ET AL. African Americans was 47% versus 37%, respectively, in Selective small fiber polyneuropathy one series (p < 0.01) (Cohen et al., 1998). The UKPDS Small fiber neuropathies are characterized by pain and trial showed an increase in prevalence in both male diminished thermal and pain perception. The pain is and female Caucasians compared to Asian and described as burning, pricking, stabbing, jabbing, or a Afro-Caribbean, 13% versus 4% and 6%, respectively, tight, band-like pressure. Patients often experience dysfor males and 6% versus 4% and 2%, esthesias. The dysesthesias are more commonly and inirespectively, for females (p < 0.001) (UKPDS, 1994). tially in the toes but may spread to the legs and upper There was no difference in the prevalence among Hisextremities as well. Strength, vibration, proprioception, panics and non-Hispanic whites in one study and tendon reflexes are preserved in small fiber poly(Hamman et al., 1991). neuropathy. Electromyography and nerve conduction studies are useful for following progression of small CLASSIFICATION AND fiber syndromes but not diagnostically. Nerve conducCLINICAL FINDINGS tion study abnormalities may appear if a large fiber component of the neuropathy develops and progresses. Diabetic peripheral polyneuropathy is the most common Testing of autonomic function and of thermal sensitivity neuropathy in diabetics, with a prevalence as high as 54% is used to test small fiber function. in type 1 DM and 45% in type 2 DM (Dyck et al., 1993). Mixed syndromes of diabetic neuropathy may account for the differences in classifications that have been Autonomic neuropathy developed. Symmetric polyneuropathies can be divided into senAutonomic neuropathy is defined as subclinical or sory or sensorimotor polyneuropathy, selective small clinical depending on the absence or presence of overt fiber polyneuropathy, and autonomic neuropathy. symptoms. Diabetic autonomic neuropathy (DAN) can affect all parts of the autonomic nervous system. Signs and symptoms of DAN may manifest as cardiovascular, Sensory or sensorimotor polyneuropathy gastrointestinal, genitourinary, metabolic, sudomotor, or Distal sensory neuropathy with an insidious onset is the pupillary disturbances. The clinical manifestations of carmost common neuropathy in diabetic patients. The eardiovascular autonomic neuropathy include resting tachyliest symptoms involve the toes and then ascend upward cardia, exercise intolerance, and orthostatic hypotension. in a “stocking and glove” pattern of sensation loss, the Autonomic neuropathy affecting the gastrointestinal distal portions of the longest nerves being the first system may manifest as esophageal dysmotility, gastroaffected. Patients describe numbness or tingling mainly paresis diabeticorum, constipation, diarrhea, or fecal of the toes and feet. Upper extremity involvement occurs incontinence. Neurogenic bladder, erectile dysfunction, less commonly, later, and is usually less severe. Pain may retrograde ejaculation, and female sexual dysfunction be one the dominant features. Bilateral foot pain may be are all some of the clinical manifestations of autonomic described as sharp, stabbing, burning, or aching in charneuropathy affecting the genitourinary system. Metabolic acter. Many patients describe a worsening of the pain at derangements seen with autonomic neuropathy include night. Weakness and gait ataxia due to sensory loss may hypoglycemia unawareness. Anhidrosis, heat intolerance, be the predominant symptoms in some patients. and gustatory sweating may be clinical manifestations of Neurologic examination demonstrates reduced or sudomotor dysfunction due to autonomic neuropathy absent ankle jerks. There may be varying degrees of sen(Vinik et al., 2003). sory loss and distal muscle weakness mainly of toe and Cardiovascular autonomic neuropathy (CAN) can foot dorsiflexion. Distal weakness is typically mild and a contribute to silent myocardial infarctions. Many studies less common presentation. Impairment of vibration and have suggested that cardiovascular autonomic neuropajoint position suggest large fiber neuropathy. Sensorithy may contribute to an increase in silent myocardial motor neuropathy has features of both axon loss and infarcts in diabetics compared to nondiabetics and demyelization on electromyography (EMG). The earliest increased mortality in these patients. A 27% mortality and most sensitive findings are a reduction in conduction in diabetic patients with CAN compared with 8% in those velocity and a decrease in signal amplitude on the without CAN (p > 0.01) was found in one study (Ewing EMG. Complete disappearance of a sensory response et al., 1991). Another study found 40% mortality versus may be seen as the neuropathy becomes more severe. 4% in patients with and without CAN (p < 0.01) Motor nerve studies may be abnormal showing some (Jermendy et al., 1991). Other investigators found slowing of nerve conduction waves even in asymptommortality rates of 23–40% in patients with CAN atic patients. compared to a rate of 3–12% in patients without

DIABETIC NEUROPATHY CAN (Rathmann et al., 1993; Navarro et al., 1996; Chen et al., 2001). Investigations examining the association of CAN and mortality vary in the tests of autonomic function used, the definition of CAN, and the subjects studied, but overall they provide consistent evidence that there is an increase in mortality among diabetic individuals with CAN compared to diabetics without it One study found increased 10 year mortality in type 1 diabetics with CAN only in those with symptoms and not in asymptomatic individuals (Sampson et al., 1990). Meta-analysis of 15 studies with follow-up from 1 to 16 years concluded that the pooled estimate of the relative risk of death based on 2900 subjects was 2.14 for subjects with CAN at baseline compared to those with a normal baseline assessment. On multivariate analysis CAN was the most important independent predictor of mortality, with a cumulative 5 year mortality increased over five fold in type 1 diabetics with cardiovascular autonomic neuropathy (Vinik et al., 2007). The causative relationship between CAN and mortality, however, remains speculative. Studies have given alternative possible explanations for the increased mortality in diabetics with CAN including increased occurrence of silent myocardial ischemia (Niakan et al., 1986), asymptomatic ischemia leading to lethal arrhythmias (Rathmann et al., 1993), impaired hypoglycemic awareness (O’Brien et al., 1991), and an increased occurrence of concomitant disorders and other diabetic complications (Orchard et al., 1996). Other studies have cited prolongation of the QT interval (Sampson et al., 1990) and interrelationships with other cardiovascular risk factors (Maser et al., 1990; Spallone et al., 1997).

Focal and multifocal neuropathies Focal and multifocal neuropathies can be divided into cranial neuropathy, limb mononeuropathy (compression and entrapment neuropathies), trunk mononeuropathy, and asymmetric lower limb motor neuropathy (amyotrophy).

775

overt diabetes (Pecket and Schattner, 1982). Patients with Bell’s palsy may complain of weakness or paralysis of all the muscles on one side of the face. On examination, the facial crease and nasolabial fold and forehead furrow of the affected side disappear, the corner of the mouth droops, the eyelid may not close and the lower lid may sag. Impairment in taste is less common in diabetics compared to patients without diabetes which suggests that the lesion in diabetic facial nerve palsy is distal to the chorda tympani. The treatment of Bell’s palsy in diabetics does not differ from the treatment in patients without diabetes except for caution if steroids are used given the risk of steroid-exacerbated hyperglycemia.

Limb mononeuropathy (compression and entrapment neuropathies) There is an increased frequency of compression neuropathy such as carpal tunnel syndrome (CTS) in diabetic patients. Dyck et al. found that although only 9% of type 1 diabetics and 6% of type 2 diabetics were symptomatic, as many as 27% and 35% of type 1 and type 2 diabetics had electrophysiologic evidence of CTS (Dyck et al., 1993). Data analysis of a large registry in the UK of 2655 diabetic and prediabetic patients found that the incidence of CTS in patients with prediabetes was increased, with a relative risk of 1.63. CTS can precede the diagnosis of diabetes by up to 10 years (Gulliford et al., 2006). A study to investigate frequency of symptomatic mononeuropathies in diabetic patients found that only focal limb neuropathies due to external compression appear to be more common. Of 642 consecutive patients with various acute symptomatic mononeuropathies, patients with radial, ulnar neuropathy and peroneal neuropathy had rates of diabetes significantly higher than those anticipated according to the frequency of diabetes in the general population (Stamboulis et al., 2005). In this study patients with CTS and Bell’s palsy had rates of diabetes similar to the general population.

Trunk mononeuropathy Cranial neuropathy The most common cranial neuropathy in diabetics is a mononeuropathy involving the third cranial nerve presenting as sudden onset of double vision, drooping of one eyelid and/or pain in the head or behind the affected eye. Patients may exhibit dysconjugate gaze, with the affected eye deviating laterally, and impaired movement of the gaze medially and vertically. Pupil reaction may be normal or abnormal. Cranial neuropathies involving other nerves are less common. Bell’s palsy appears to be more common in diabetics than in patients with normoglycemia (Gilden, 2004). A study of 126 patients with Bell’s palsy revealed 39% had evidence of biochemical or

Diabetic truncal neuropathy usually occurs in older patients and presents as dysesthesia in one or more of the thoracic dermatomes or as radicular thoracic pain. Stewart described 17 episodes of diabetic truncal neuropathy among seven diabetic patients and found varying combinations of sensory deficits, including but not limited to dysesthesia of a complete dermatomal band, involvement of adjacent main spinal nerves, and most commonly deficits restricted to the distribution of the ventral and dorsal rami of the spinal nerves or branches of the rami (Stewart, 1989). A series of 15 new cases of diabetic thoracic radiculopathy describes patients presenting with severe abdominal pain or chest pain and

776

G. CHARNOGURSKY ET AL.

often weight loss. This condition generally carried a good prognosis for recovery (Kikta et al., 1982).

Asymmetric lower limb motor neuropathy (amyotrophy) Amyotrophy is an asymmetric lower limb motor neuropathy also known as diabetic lumbosacral plexus neuropathy and Bruns-Garland syndrome. Patients typically present with an asymmetric, painful muscle wasting and weakness affecting the lower limbs and loss of reflexes and objective weakness on examination. Patients may describe a sudden onset of sharp pain in the hip and thigh that can spread to the opposite side over weeks to months, generally in a stepwise and steady progression affecting both proximal and distal muscles (Barohn et al., 1991). Amyotrophy is more likely to affect middle to older aged patients. Electrodiagnostic studies are useful in ruling out other conditions that may account for the symptoms of amytrophy.

LABORATORY INVESTIGATIONS Methodologies used in studies that define diabetic neuropathy vary considerably. Methods range from relying on symptoms to including neurologic exams and using more quantitative sensory testing. Evaluation of sensory function includes thresholds for vibration, assessment of light touch, joint position, and pinprick. Feldman described a population-based study in which 66% of patients with diabetes exhibited a decline in vibratory sense of the feet over time which was associated with diabetic neuropathy morbidity including foot ulcerations, infections, and foot amputations (Feldman et al., 1994). Sequential examination of vibration, touch, joint position, and pinprick is a practical clinical approach for following individual patients. Kincaid and colleagues suggest that vibratory quantitative sensory testing and nerve conduction studies have a low to moderate correlation suggesting the tests do not replace one another but are complementary (Kincaid et al., 2007). Comparisons of the clanging tuning fork (CTF) test to the 10 g Semmes-Weinstein monofilament test found the tuning fork to be reproducible, accurate, and able to detect neuropathy even in the presence of a normal monofilament test (Oyer et al., 2007). These investigators recommended replacing the monofilament test with the CTF technique for detection of DPN. Electrophysiologic tests are a sensitive, objective, and reproducible way of diagnosing and following diabetic neuropathy. A long-term follow-up of 114 patients with diabetic neuropathy reported significant correlations between neuropathy disability scores (NDS) and alterations in motor nerve conduction velocities (Negrin and Zara, 1995). Other studies have confirmed these findings

(Onde et al., 2008). Electrophysiologic tests may be abnormal in asymptomatic patients, revealing early stages of neuropathy and subclinical involvement, and they help provide objective measures of diabetic neuropathy diagnosis and severity (Dyck et al., 1991, 1992). Newer methods of screening for diabetic neuropathy have been reported. A new indicator test based on the measurement of sweat production after exposure to dermal foot perspiration had a sensitivity of 86% for detecting sensorimotor polyneuropathy and 80.9% for detecting autonomic neuropathy, with specificity of 67% and 50%, respectively (Liatis et al., 2007). Researchers have also investigated a new technique in assessing possible nerve fiber repair in type 1 diabetics after pancreas transplant using a noninvasive technique of corneal confocal microscopy and found that small fiber repair can be detected 6 months after transplant using this novel technique (Mehra et al., 2007). In a study using corneal confocal microscopy to evaluate 101 diabetic patients compared to 17 controls, investigators found corneal sensation, nerve fiber density and length, and nerve branch density all decreased with increased neuropathic severity and correlated with neuropathy disability scores (Tavakoli et al., 2010). The investigators conclude that corneal confocal microscopy may be a promising noninvasive tool in the detection of diabetic neuropathy.

RISK FACTORS Epidemiologic studies have identified duration and severity of hyperglycemia as the major risk factors for the development of diabetic neuropathy in patients with both type 1 and type 2 diabetes (Genuth, 2006). The Diabetes Complications and Control Trial (DCCT) showed that intensive glycemic control reduced the prevalence of autonomic dysfunction by 53% and clinical neuropathy by 60-69% (DCCT Research Group, 1995). The European Diabetes (EURODIAB) Prospective Complications Study demonstrated that in addition to glycemic control and duration of diabetes, traditional markers of macrovascular disease appear to be important in the pathogenesis of diabetic peripheral neuropathy (DPN). A total of 1172 patients with type 1 diabetes mellitus and no baseline DPN were enrolled from 31 centers and followed for 7.3 years to study the risk factors for the development of DPN. The cumulative incidence of DPN was 23.5%. After adjusting for duration of diabetes and glycosylated hemoglobin, an association was observed between modifiable cardiovascular risk factors and incidence of DPN. DPN developed more frequently when patients had hypertension (odds ratio (OR), 1.92), elevated triglyceride levels (OR, 1.35), smoking (OR, 1.55), or obesity (OR, 1.4). Microvascular disease at

DIABETIC NEUROPATHY baseline (e.g., albuminuria (OR, 1.48) and retinopathy (OR, 1.7)) was also associated with an increased incidence of DPN. Cardiovascular disease at baseline independently doubled the risk of neuropathy (OR, 2.74).

PATHOPHYSIOLOGY Diabetic neuropathy occurs when there is an imbalance between nerve fiber damage and repair. The nerve damaging process preferentially affects autonomic and distal sensory fibers, leading to the progressive loss of sensation. Besides metabolic factors listed above, ischemic factors and inflammation also contribute to the development of diabetic neuropathies. Metabolic factors seem to prevail in length-dependent diabetic polyneuropathy, whereas inflammation superimposed on ischemic nerve lesions is found in severe forms of focal neuropathies. The thickening and hyalinization of the walls of small blood vessels due to the reduplication of the basal lamina around endothelial cells suggests a role for nerve ischemia in diabetic neuropathy. There is also a reduction in endoneurial oxygen tension in the sural nerves of diabetic patients with advanced polyneuropathy (Newrick, 1986). Possible mechanisms for neuropathy development include oxidative stress, nonenzymatic glycation, the polyol pathway, the hexosamine pathway, protein kinase C pathway, poly (ADP-ribose) polymerase and the reduction of neurotrophic factors (Table 51.1). These various pathogenetic factors may act synergistically to cause DPN

Oxidative stress Elevated glucoses can increase oxidative stress by glucose auto-oxidation and production of advanced glycosylation end products and activation of the polyol pathway. Oxidative stress can also lead to activation of cytokoines, vascular adhesion molecules, endothelium-1 and procoagulant tissue factor . Oxidative stress also reduces endothelial production of nitric oxide which leads to impairment of endothelial function and reduced capillary vasodilation. This ultimately contributes to nerve hypoxia. Table 51.1 Pathogenetic mechanisms of neuropathy Oxidative stress with nitric oxide depletion Advanced glycosylated end products Activation of the polyol pathway Activation of the hexosamine pathway Excessive protein kinase C activity Activation of poly(ADP-ribose) polymerase Diminished neurotrophic peptide factors

777

The AGE pathway Advanced glycosylation end products (AGEs) from chronic hyperglycemia play an important role in diabetic neuropathy and microvascular complications (Thornalley, 2002; Sugimoto et al., 2008). Excess glucose combines with amino acids on circulating or tissue proteins to form AGEs. AGEs do not resolve when hyperglycemia is corrected. These AGE peptides crosslink strongly with collagen in vitro, damaging nerve fibers. AGEs also bind to and activate the cell surface receptor called RAGE(Receptor for Advanced Glycation Endproducts). RAGE proteins are proinflammatory and expressed on endothelial cells, fibroblasts, mesangial cells, and macrophages. Endothelial cells with RAGE internalize AGEs into subepithelium enhancing permeability and endothelium-dependent coagulant activity which can contribute to vascular injury and endoneural hypoxia (Singh et al., 2001).

The polyol pathway Excess glucose is shunted into the polyol pathway and converted to sorbitol by aldose reductase and then to fructose by sorbitol dehydrogenase. Increased activity of this metabolic pathway depletes the nicotinamide adenine dinucleotide phosphate hydrogen (NADPH) needed to regenerate the antioxidant glutathione. Without adequate glutathione, nerves are less able to scavenge reactive oxygen species, thus promoting oxidative stress in the nerve. The excess fructose and sorbitol also decrease expression of the sodium/myoinositol cotransporter, leading to a reduction in cellular uptake of myoinositol. Decreased levels of intracellular myoinositol subsequently lower the levels of its metabolite phosphoinositide. Consequently, the phosphoinositide signaling pathway is impaired, interfering with activation of the transmembrane sodium pump and decreasing nerve sodium/potassium ATPase activity. This results in slowed nerve conduction and with chronic exposure neuronal membrane breakdown ensues.

The hexosamine pathway The glycolytic intermediates of excess glucose are also shunted into the hexosamine pathway. Fructose6-phosphate is converted to N-acetylglucosamine6-phosphate by glutamine: fructose-6-phosphate amidotransferase (GFAT). N-Acetylglucosamine-6-phosphate is then converted to N-acetylglucosamine-1,6-phosphate and to uridine diphosphate-N-acetyl glucosamine (UDPGlcNAc). UD-PGlcNAc modifies gene expression and protein production essential for normal cell function. Many of the proteins produced in this pathway are

778 G. CHARNOGURSKY ET AL. inflammatory intermediates that promote neuropathy TREATMENT OF DIABETIC and include plasminogen-activator inhibitor, which NEUROPATHIES inhibits normal blood clotting and increases vascular Disease state modifiers complications (Brownlee, 2001). The treatment of diabetic peripheral and autonomic neuropathies has largely been directed at control of Protein kinase C (PKC) symptoms rather than treatment of the underlying pathpathway and poly (ADP-ribose) ologic mechanisms with the exception of control of polymerase (PARP) hyperglycemia. The Diabetes Control and Complications Protein kinase C (PKC) is involved in controlling the funcTrial (DCCT) outcomes regarding neuropathy have been tion of proteins through the phosphorylation of hydroxyl discussed above. The Epidemiology of Diabetes Intergroups of serine and threonine amino acid residues on ventions and Complications(EDIC) followed a cohort of these proteins. PKC is responsible for the activation of 1257 DCCT participants for 8 years after completion of essential proteins and lipids in cells that are needed for cell the trial and revealed ongoing limitation of neuropathy, survival (Vincent et al., 2004). Nevertheless, excessive based upon patient questionnaire and structured foot PKC can be harmful to the nervous system. Its contribuexam, even though the glycemic control of the previously tion to diabetic neuropathy is likely through effects on intensively treated group approached that of the prior vascular blood flow and microvascular disease rather DCCT conventional care group (Martin et al., 2006). than directly on neuronal cells. Glucose is converted to Further study of these patients by both clinical examination diacylglycerol which activates PKC. PKC then activates and nerve conduction studies at 13–14 years post close of the mitogen-activated protein kinases (MAPK) which DCCT revealed the former intensive treatment group had phosphorylate transcription of stress genes such as heat a 25% prevalence of neuropathy compared to 35% in the shock proteins and c-Jun kinases that can lead to cell former conventional group, supporting the concept of apoptosis or vascular atherosclerosis (Tomlinson, 1999). metabolic memory in neuropathy development (Albers The inhibition of PKC reduces oxidative stress and noret al., 2010). In combined pancreas and kidney transplant malizes blood flow and nerve conduction deficits in patients with type 1 diabetes, correction of glucose abnordiabetic rats (Ishii et al., 1998; Cameron and cotter, malities by successful transplantation has led to corneal 2002). Poly (ADP-ribose) polymerase (PARP) is activated small fiber nerve repair documented by confocal microsin response to hyperglycemia. Overactivation of PARP copy (Mehra et al., 2007). Significant improvement was results in increased free radical formation, enhanced proseen in corneal nerve fiber density and nerve fiber length tein kinase C activity, and AGE formation (Pacher et al., at 6 months post transplant. 2005). Each promotes nerve damage through the metaThe United Kingdom Prospective Diabetes Study bolic pathways described above. (UKPDS) evaluated newly diagnosed type 2 diabetes patients, comparing intensive treatment with insulin, sulfonylurea, or metformin versus standard care with diet. Neurotrophic factors and nerve repair The intensive care group had a 25% risk reduction in The neurotrophic factors comprise a group of endogemicrovascular end points (neuropathy , retinopathy, nous proteins essential to the health and survival of certain and nephropathy) after 10 years and a 60% relative populations of neurons. These neurotrophic peptides risk reduction in neuropathy at year 15 (UKPDS Study include nerve growth factor, brain-derived neurotrophic Group, 1998). More recent glycemic control studies, factor, neurotrophin-3, the insulin-like growth factors though not having long-term extension arms, have failed (IGF), and vascular endothelial growth factors. They are to show differences in progression of neuropathy with important for the maintenance of nerve structure and tight glycemic control. The Veterans Affairs Diabetes function as well as repair following injury. Impaired Trial (VADT) failed to show any decrease in neuropathic peripheral nerve repair in diabetes may be due to outcomes, including autonomic neuropathy, peripheral diabetes-induced loss of these peptides (Kennedy, 2000, neuropathy, and mononeuropathy, in patients receiving 2005). Insulin also functions as a neurotrophic factor to tight glycemic control versus standard care groups with peripheral neurons, and thus loss of insulin in diabetics the median observation period of 5.6 years (Duckworth may compromise nerve viability and repair. Intrathecal et al., 2009). Similarly, the Action in Diabetes and delivery of low-dose insulin has reversed the slowing of Vascular Disease (ADVANCE) trial failed to show limitamotor and sensory nerve conduction velocity. Insulin tion of new or worsening neuropathy in the setting of tight and IGF-1 have also been shown to reverse atrophy in glycemic control over a median of 5 years observation myelinated sensory axons in the sural nerve (Brussee (Patel et al., 2008). In both of these recent diabetes interet al., 2004). vention studies, other vascular factors were also well

DIABETIC NEUROPATHY managed in both treatment and control groups. Patients in these studies generally had more established diabetic end injury and vascular risk factors in the setting of a greater duration of diabetes at study onset. Patients in the VADT had an average duration of diabetes 11.5 years while those in the ADVANCE trial had been diabetic for 8 years at study onset. This compares to the UKPDS and DCCT wherin patients were either newly diagnosed with DM2 (UKPDS) or young healthy patients with DM1 (DCCT). Attempts to treat other proposed pathogenetic mechanisms including protein kinase C activation via hyperglycemia-induced elevation of diacylglycerol, accumulation of advanced glycation end products, aldose reductase activation through the polyol pathway and increased activity of the hexosamine pathway have shown limited success except for the use of a-lipoic acid (ALA) to treat oxidative stress. ALA, also known as thioctic acid, is an antioxidant scavenger of reactive oxygen species. These free radicals can cause endoneurial hypoxia and impair nerve conduction in experimental diabetic neuropathy models (Low et al., 1997a). The Alpha-Lipoic Acid in Diabetic Neuropathy (ALADIN) studies (Ziegler et al., 1995, 1999; Reljanovic et al., 1999) evaluated parenteral, oral and sequential parenteral followed by oral treatments with a-lipoic acid revealing reduced neuropathic symptoms in ALADIN (parenteral 100–1220 mg daily for 3 weeks) and improved NIS (Neuropathy Impairment Score) in ALADIN III (parenteral followed by oral a-lipoic acid 600 mg orally three times daily). It is noteworthy that there were discrepant results between the reduction in subjective clinical neuropathic symptoms and the objective NIS in ALADIN III, raising concerns by the authors regarding the use of clinical symptoms as primary end points. ALA appears safe when used in daily 600 mg intravenous doses for 3 weeks (Ziegler et al., 2004), but parenteral ALA is not available in the US. The Symptomatic Diabetic Neuropathy(SYDNEY) 2 trial showed improvement in both clinical symptoms and NIS after a 5 week course of daily oral a-lipoic acid in 600– 1800 mg/day doses (Ziegler et al., 2006).Oral ALA was well tolerated, with nausea the most common sideeffect. The nausea appeared dose-dependent and since there was no incremental benefit in neuropathic symptom relief with higher doses, the ALA 600 mg/day regimen appears to be the optimal dose. However, the SYDNEY and ALADIN trials were limited by short study durations. The Neurological Assessment of Thioctic Acid in Diabetic Neuropathy (NATHAN) 1 trial evaluated 460 diabetic patients with mild to moderate distal symmetric sensory motor polyneuropathy, randomized to receive oral ALA 600 mg once daily versus placebo for 4 years (Ziegler et al., 2011). The study showed no significant change in the primary end point composite score

779

(NIS, NIS-Lower Limbs (NIS-LL), and seven neurophysiologic tests) between treatment and control groups group. When evaluating individual tests, patients on ALA did better on NIS, NIS-LLE and muscular weakness subscores when compared to placebo patients. Nerve conduction and QST results did not worsen in a placebo group over the 4 years observation, pointing to the slow progression of peripheral neuropathy even in the control patients. Modifiers of the polyol pathyway including aldose reductase inhibitors (ARIs) have been in development for decades and only epalrestat is now available in Japan and India. Ranirestat is currently in clinical trials with a recent study showing no significant improvement in clinical symptoms compared to placebo as both trial groups had improved symptom profiles. After 52 weeks of therapy, ranirestat showed improvement in summed motor nerve conduction velocities (NCV), but did not show significant changes in summed sensory NCV (Bril et al., 2009). Use of other ARIs has been limited either by toxicity (tolrestat) or lack of clinical efficacy. Treatment with protein kinase C b-inhibitor ruboxistaurin showed no differences versus placebo in vibration detection threshold or neuropathy total symptom scores in a 52 week study, but a subgroup with less severe symptomatic diabetic peripheral neuropathy did show improvement in these study parameters (Vinik et al., 2005). Vascular endothelial growth factor (VEGF) has been found to be reduced in diabetic nerves (Quattrini et al., 2008). Injection of plasmid VEGF into standardized areas around peripheral nerves did improve symptoms but did not improve nerve conduction velocities (Ropper et al., 2009). Medical foods such as folate and B vitamins have been used in the treatment of peripheral neuropathies. L-methylfolate and 5-methyl tetrahydrofolate have been associated with improvement in nitric oxide levels, free radical scavenging, and thusly improved endothelial dysfunction (van Etten et al., 2002; Verhaar et al., 2002). Evaluation for B12 deficiency is of particular importance in patients on metformin as this first-line agent for type 2 diabetes has been associated with low B12 levels, elevated methylmalonic acid and homocysteine levels, and higher scores on neuropathy screening tools than matched patients who had not received metformin (Wile and Toth, 2010). This study, however, did not look at intervention with B12 replacement. While controlled trials using the B vitamins have been limited by small numbers and short study durations with equivocal clinical results, a single noncontrolled and nonblinded study of 20 patients with type 2 diabetes and peripheral neuropathy who received the combination of L-methylfolate, methylcobalamin, and pyridoxal 5-phosphate daily for 52 weeks showed improvement in 1-point tactile and 2point discriminatory testing (Walker et al., 2010).

780 G. CHARNOGURSKY ET AL. In the treatment of painful peripheral diabetic neubeginning with duloxetine 30 mg/day and titrating ropathies, it is important to rule out other causes of neuupward. Venlafaxine extended release showed reduction ropathy as up to 15% of cases can have etiologies other in multiple pain score scales over a 6 week trial than solely diabetes. The evaluation should include com(Rowbotham et al., 2004). Most common side-effects plete blood count, erythrocyte sedimentation rate, comof venlafaxine were nausea and somnolence. prehensive metabolic panel, thyroid function tests, B12 Anticonvulsants which modulate calcium channel and serum protein electrophoresis (England et al., activity include gabapentin and pregabalin. These agents 2009). In addition to simply ameliorating symptoms with bind to the a 2-g subunit of calcium channels and inhibit medications, treatment of other underlying metabolic neurotransmitter release. Side-effects of somnolence issues associated with diabetes and also prediabetes and dizziness are seen in both. Gabapentin decreased may be helpful in controlling neuropathy (Tesfaye scores for daily pain severity and sleep interference et al., 2005). Interventions include cessation of cigarette and improved quality of life scores in a double-blind, smoking and controlling hypertension, weight, and lipids placebo-controlled study (Backonja et al., 1998). This in addition to glycohemoglobin. agent is given three or more times daily with dosing and subsequent pain relief often limited by dizziness and somnolence. Gabapentin doses  1800 mg/day are Pain-controlling agents generally needed for pain relief (Backonja and Medications for diabetic peripheral neuropathic pain Glanzman, 2003). An extended release gabapentin has include tricyclic antidepressants, selective serotonin norbeen shown to be effective compared to placebo in epinephrine reuptake inhibitors, antiseizure agents reducing average daily pain scores in once or twice daily including calcium channel a 2-g ligands and sodium regimens (Sandercock et al., 2009). Pooled data from channel blockers, lidocaine and capsaicin topicals, opiseven randomized controlled trials of pregabalin oids, and tramadol. Pharmacotherapies for pain control (Freeman et al., 2008) revealed significant reductions currently approved in the US include duloxetene and in pain and sleep interference end points. The pregabalin pregabalin. These medications as monotherapies or in 600 mg/day dosing showed efficacy when divided into combinations generally lead to amelioration but not twice daily doses while lower daily doses required three resolution of pain symptoms. Most agents require times daily dosing. In addition to dizziness and somno4–6 week trials to assess efficacy. Formal clinical trials lence, peripheral edema was also a common side-effect. of medications for neuropathic pain frequently have difSymptom improvement was seen in 5 days or less with ficulty showing statistical significance of clinical sympdoses 300 mg/day or greater. tom scores, neurologic standardized examination Among anticonvulsants which block the sodium scores, or electrodiagnostic parameters as placebo channels, lacosamide in doses of 400–600 mg/day in groups can show stabilization or improvement in the setan 18 week study has been shown to decrease average ting of better diabetes, lipid, and hypertension control. daily pain score more than placebo, but statistical signifPain control algorithms generally begin with tricyclics icance was not achieved (Ziegler et al., 2010). The including amitriptyline, imipramine, nortriptyline, and authors suspected improved pain scores in the placebo desipramine .The tricyclics can treat concurrent depresgroup contributed to the lack of significance in this prision along with the neuropathic pain. These agents mary end point. are economical but side-effects can limit effective dosTopical therapies including capsaicin and lidocaine ing. Dry mouth, urinary retention, and orthostatic 5% patches or gel have also been useful in decreasing hypotension are common anticholinergic side-effects. pain. As these agents exert their effects locally and withDrowsiness caused by these agents can be helpful in out significant systemic absorption, they are limited to improving sleep but may be bothersome if somnolence treatment of focal painful areas. Local irritation is the lingers into daytime. The selective dual serotonin and most common adverse effect. Capsaicin acts by depletnorepinephrine reuptake inhibitor antidepressants ing tissue substance P and has shown modest efficacy duloxetine and venlafaxine have shown efficacy in conover short-term use (Zhang and Wan, 1994), but requires trolled trials in painful diabetic neuropathy. Duloxetine four times daily local application. A new high concenin 60–120 mg daily doses has been shown in post hoc tration 8% capsaicin patch has shown efficacy in postanalysis of three double-blind, placebo-controlled herpetic neuralgia and HIV neuropathy with a single 12 week trials to reduce average 24 hour pain scores with 60 minute application in a 12 week study (Backonja efficacy apparent after 1 week of treatment (Kajdasz et al., 2008). et al., 2007). Side-effects including nausea, somnolence, Tramadol and opioids are efficacious in controlling dizziness, and anorexia appear more commonly with diabetic neuropathic pain. Tramadol is a weak opioid the higher 120 mg/day dosing and can be limited by receptor agonist which also inhibits norepinephrine

DIABETIC NEUROPATHY and serotonin reuptake. This agent is less potent than the opioids. Side-effects of both groups include sedation, constipation, and nausea. Due to the potential for dependency with these agents, their use should limited to those patients who have failed trials of the other medications discussed above.

Medications for neuropathic pain See Table 51.2.

Treatment of autonomic neuropathy The clinical manifestations of diabetic autonomic neuropathy (DAN) are listed in Table 51.1. Treatment of hyperglycemia and other metabolic factors including hypertension and hyperlipidemia can help limit DAN. The Steno-2 study followed 160 type 2 DM patients with microalbuminuria for over 13 years of treatment with renin–angiotensin system blockers, aspirin and lipid lowering agents in additional to glucose controlling agents (Gaede et al., 2008). Progression of DAN, measured by electrocardiogram RR intervals during paced breathing and orthostatic hypotension, was noted in 39 patients on intensive therapy and 52 in the conventional treatment group (p ¼ 0.004). Treatments for DAN can be directed at abnormalities in the cardiovascular, gastrointestinal, and genitourinary systems. The major treated cardiovascular complication is orthostatic hypotension. Nonpharmacologic interventions include gradual position changes, 10–20 head of bed elevation, isotonic exercise, graded compression stockings, and abdominal binders. Removal of pharmacotherapies known to cause orthostasis can also be helpful. Unfortunately, many of the agents used to treat the peripheral neuropathic pain including tricyclics, duloxetine and anticonvulsants can also cause orthostasis. The mineralocorticoid fludrocortisone at doses of 0.1– 0.5 mg daily plus high salt diet or salt tablets can assist

781

in volume expansion , but peripheral edema, congestive heart failure, hypokalemia, and supine hypertension can limit dose escalation. Midodrine, an a1 adrenoreceptor agonist, at doses up to 10 mg three times a day, has been effective in increasing standing blood pressure (Low et al., 1997b; Wright et al., 1998) but has adverse effects of supine hypertension and urinary retention. Midodrine and fludrocortisone are frequently used in combination, yet supine hypertension can limit dose escalation. Pyridostigmine, a cholinestesterase inhibitor, increases diastolic blood pressure without causing supine hypertension. The baroreceptor efferent limb synapses at the autonomic ganglion with acetylcholine as the neurotransmitter. Singer et al. postulated that since autonomic ganglion activity is low when supine, this agent should not cause supine hypertension Their randomized four way crossover study evaluated pyridostigmine 60 mg alone and with varied doses of midodrine. The fall in standing diastolic blood pressure and orthostatic symptoms were reduced with treatment with pyridostigmine alone and in combination with midodrine 5 mg (Singer et al., 2006). Yohimbine, an a2 adrenergic receptor antagonist, can increase residual sympathetic tone in patients with autonomic failure by increasing norepinephrine release from sympathetic nerves. Shibao et al. studied 31 patients with severe autonomic failure. They postulated that yohimbine and pyridostigmine, due to their differing mechanisms of actions, might have a synergistic effect in ameliorating orthostatic hypotension. Patients received pyridostigmine 60 mg, yohimbine 5.4 mg, or a combination of both agents in a singleblind, randomized, placebo-controlled crossover study. Yohimbine improved standing diastolic pressure measured 60 minutes after drug administration. Contrary to the Singer study noted above, pyridostigmine did not increase diastolic blood pressure significantly in these patients. No evidence of synergy was found in

Table 51.2 Medications for neuropathic pain Class

Agents

Common side-effects

Tricyclic antidepressants

Nortriptyline, desimipramine, amitriptyline, imipramine Duloxetine Venlafaxine Gabapentin Pregabalin 5% lidocaine patch Capsaicin Tramadol Morphine, oxycodone, codeine

Anticholinergic effects, somnolence, orthostasis Nausea, somnolence, anorexia, dizziness

Selective serotonin and norepinephrine reuptake inhibitors Anticonvulsants Topical medications Opioid agonists

Somnolence, dizziness, leg edema Local irritation Nausea, somnolence, constipation

782

G. CHARNOGURSKY ET AL.

patients receiving both pyridostigmine and yohimbine (Shibao et al., 2010). Authors postulated that patients in the Singer trial may have had less severe autonomic dysfunction function with greater reserve. Nonsteroidal anti-inflammatory drugs can help orthostasis by inducing volume expansion and also by antagonizing vasodilating prostaglandins. Erythropoietin has been helpful in patients with both anemia and orthostatic hypotension (Hoeldtke and Streeten, 1993). Gastrointestinal neuropathy can affect any part of the bowel from esophagus to anus. Esophageal reflux can be treated with histamine-2 blockers and proton pump inhibitors. Gastroparesis can be worsened by acute hyperglycemia and can contribute to glucose lability by causing delayed and unpredictable delivery of nutrients to the small bowel and also by impeding absorption of oral diabetes medications. Incretin-modulating agents including exenatide and liraglutide and the amylin analog pramlintide can slow gastric emptying and should be avoided in patients with gastroparesis. Effective gastric prokinetic agents are limited. Cisapride, a stimulator of 5-HT4 receptors, is no longer readily available in the US due to risk of cardiac rhythm disturbances. Domperidone, a dopamine antagonist which binds to the D2 and D3 dopamine receptors, has both prokinetic and antiemetic properties, but is not available in the US. Intravenous erythromycin lactobionate can be used to stimulate gastric emptying (Janssens et al., 1990) while oral forms of this agent have been less effective (Maganti et al., 2003). This macrolide antibiotic should be used only sporadically due to potential for GI side-effects including nausea and colitis. When used with CYP3A4 inhibitors, the drug can also cause QT interval prolongation with cardiac rhythm disturbances. Metoclopramide, a dopamine D2 receptor antagonist and 5-HT4 receptor agonist, improves gastric emptying and is also antiemetic. This agent is available for both oral and intravenous use and has been associated with extrapyramidal side-effects and tardive dyskinesia. An oral rapidly dissolving form is also available which requires no water or swallowing. Jejunostomy tube placement may be needed for nutritional support in patients who are unresponsive to the above described interventions. Gastrostomy tubes are rarely required for decompression in patients with persistent vomiting or distension. Gastric electrical stimulation has very limited availability and can improve gastroparesis symptoms without increasing gastric emptying. Genitourinary neuropathy can present as bladder and erectile dysfunction. Cystopathy can be treated with timed voiding, intermittent self-catheterization, and cholinergic agents. Erectile dysfunction in diabetic male patients can be multifactorial so evaluation for hypogonadism, elevated prolactin, thyroid disease, and confounding medications should be performed. If no

readily treatable etiologies are found, phosphodiesterase 5 inhibitors can be initiated. Sildenafil, vardenafil and tadalafil limit breakdown of cyclic guanosine monophosphate in the penile corpora cavernosa and increase penile blood flow and thus erection. Side-effects can include headache, impaired color vision, lightheadedness, syncope, hearing and vision loss. Tadalafil has a greater than 24 hour duration of action. While each agent is approved for intermittent use around the time of sexual relations, tadalafil, in doses of 2.5–5 mg, is also approved for scheduled daily dosing. Intraurethral prostaglandin E1 (alprostadil), intracavernosal injections of papaverine, phentolamine and alprostadil and penile suction devices have also shown efficacy in treating erectile dysfunction.

CONCLUSIONS While the pathophysiology of diabetic neuropathies is now reasonably well understood, clinicians are still limited in therapies directed at correcting the underlying etiologies. Other than control of glycemia and other vascular risk factors and antioxidants such as a-lipoic acid, specific effective agents are not available. Treatments for diabetic autonomic neuropathies such as orthostatic hypotension are directed largely at treating the end result of the condition rather than the etiology. Medications directed at neuropathic pain control are able to partially ameliorate symptoms rather than give complete resolution. Treatments are needed both to correct the pathologic mechanisms and to control pain and autonomic consequences in a way that will be safe for long-term use in chronic diabetes mellitus.

REFERENCES Albers J, Herman W, Pop-Busai R et al. (2010). Effect of prior intensive insulin treatment during the Diabetes Control and Complications Trial (DCCT) on peripheral neuropathy in type 1 diabetes during the Epidemiology of Diabetes Interventions and Complications (EDIC) study. Diabetes Care 33: 1090–1096. Backonja M, Glanzman R (2003). Gabapentin dosing for neuropathic pain: evidence from randomized, placebocontrolled clinical trials. Clin Ther 25: 81–104. Backonja M, Beydoun A, Edwards K et al. (1998). Gabapentin for the symptomatic treatment of painful neuropathy in patients with diabetes mellitus: a randomized controlled trial. JAMA 280: 1831–1836. Backonja M, Wallace M, Blonsky E et al. (2008). NGX-4010, a high-concentration capsaicin patch, for the treatment of post herpetic neuralgia: a randomized, double-blind study. Lancet Neurol 7: 1102–1112. Barohn R, Sahenk Z, Warmolts J et al. (1991). The Bruns– Garland syndrome (diabetic amyotrophy). Revisited 100 years later. Arch Neurol 48 (11): 1130–1135.

DIABETIC NEUROPATHY Boulton A, Malik R, Arezzo J et al. (2004). Diabetic somatic neuropathies. Diabetes Care 27: 1458–1486. Boulton A, Vinik A, Arezzo J et al. (2005). Diabetic neuropathies: a statement by the American Diabetes Association. Diabetes Care 28: 956–962. Bril V, Hirose T, Tomioka S et al. (2009). Ranirestat for the management of diabetic sensorimotor polyneuropathy. Diabetes Care 32: 1256–1260. Brownlee M (2001). Biochemistry and molecular cell biology of diabetic complications. Nature 414: 813–820. Brussee V, Cunningham F, Zochodne D (2004). Direct insulin signaling of neurons reverses diabetic neuropathy. Diabetes 53: 1824–1830. Cameron N, Cotter M (2002). Effects of protein kinase C beta inhibition on neurovascular dysfunction in diabetic rats: interaction with oxidative stress and essential fatty acid metabolism. Diabetes Metab Res Rev 18: 315–323. Chen H, Hwu C, Kuo B et al. (2001). Abnormal cardiovascular reflex tests are predictors of mortality in type 2 diabetes mellitus. Diabet Med 18: 268–273. Cohen J, Jeffers B, Faldut D et al. (1998). Risks for sensorimotor peripheral neuropathy and autonomic neuropathy in non-insulin-dependent diabetes mellitus (NIDDM). Muscle Nerve 21: 72–80. Diabetes Control and Complications Trial Research Group (1995). The effect of intensive diabetes therapy on the development and progression of neuropathy. Ann Intern Med 122: 561–568. Duckworth W, Abraira C, Moritz T et al. (2009). Glucose control and vascular complications in veterans with type 2 diabetes. N Engl J Med 360: 129–139. Dyck P, Kratz K, Lehman K et al. (1991). The Rochester Diabetic Neuropathy Study: design, criteria for types of neuropathy, selection bias, and reproducibility of neuropathic tests. Neurology 41: 799–807. Dyck P, Karnes J, O’Brien P et al. (1992). The Rochester Diabetic Neuropathy Study: reassessment of tests and criteria for diagnosis and staged severity. Neurology 42: 1164–1170. Dyck P, Kratz K, Karnes J et al. (1993). The prevalence by staged severity of various types of diabetic neuropathy, retinopathy, and nephropathy in a population-based cohort: the Rochester Diabetic Neuropathy Study. Neurology 43: 817–824. Dyck P, Dyck P, Klein C et al. (2007). Does impaired glucose metabolism cause polyneuropathy? Review of previous studies and design of a prospective controlled population-based study. Muscle Nerve 36: 536–541. England J, Gronseth G, Franklin G et al. (2009). Practice parameter: evaluation of distal symmetric polyneuropathy: role of laboratory and genetic testing (an evidence-based review): report of the American Academy of Neurology, American Association of Neuromuscular and Electrodiagnostic Medicine, and American Academy of Physical Medicine and Rehabilitation. Neurology 72: 185–192. Ewing D, Boland O, Neilson J et al. (1991). Autonomic neuropathy, QT interval lengthening, and unexpected deaths in male diabetic patients. Diabetologia 34: 182–185. Feldman E, Stevens M, Thomas P et al. (1994). A practical two-step quantitative clinical and electrophysiological

783

assessment for the diagnosis and staging of diabetic neuropathy. Diabetes Care 17: 1281–1289. Freeman R, Durso-DeCruz E, Emir B (2008). Efficiacy, safety, and tolerability of pregabalin treatment for painful diabetic peripheral neuropathy. Diabetes Care 31: 1448–1454. Gaede P, Lund-Andersen H, Parving H et al. (2008). Effect of a multifactoral intervention on mortality in type 2 diabetes. N Engl J Med 358: 580–591. Galer B, Gianas A, Jensen M (2000). Painful diabetic polyneuropathy: epidemiology, pain description, and quality of life. Diabetes Res Clin Pract 47: 123–128. Genuth S (2006). Insights from the diabetes control and complications trial/epidemiology of diabetes interventions and complications study on the use of intensive glycemic treatment to reduce the risk of complications of type 1 diabetes. Endocr Pract 12 (Suppl 1): 34–41. Gilden DH (2004). Clinical practice. Bell’s palsy. N Engl J Med 351: 1323–1331. Gulliford M, Latinovic R, Charlton J et al. (2006). Increased incidence of carpal tunnel syndrome up to 10 years before diagnosis of diabetes. Diabetes Care 29: 1929–1930. Hamman R, Franklin G, Mayer E et al. (1991). Microvascular complications of NIDDM in Hispanics and non-Hispanic whites. San Luis Valley Diabetes Study. Diabetes Care 14: 655–664. Hoeldtke R, Streeten D (1993). Treatment of orthostatic hypotension with erythropoietin. N Engl J Med 329: 611–615. Ishii H, Koya D, King G (1998). Proteins kinase C activation and its role in the development of vascular complications in diabetes mellitus. J Mol Med (Berl) 76: 21–31. Janssens J, Peeters T, Vantrappen G et al. (1990). Improvement of gastric emptyingin diabetic gastroparesis by erythromycin. N Engl J Med 32: 1028–1031. Jermendy G, Toth L, Voros P et al. (1991). Cardiac autonomic neuropathy and QT interval length. A follow-up study in diabetic patients. Acta Cardiol 46: 189–200. Kajdasz D, Iyengar S, Desaiah D et al. (2007). Duloxetine for the management of diabetic peripheral neuropathic pain: evidence-based findings from post hoc analysis of three multicenter, randomized, double-blind, placebocontrolled, parallel-group studies. Clin Ther 29: 2536–2546. Kennedy J, Zochodne D (2000). The regenerative deficit of peripheral nerves in experimental diabetes: its extent, timing and possible mechanisms. Brain 123: 2118–2129. Kennedy J, Zochodne DW (2005). Impaired peripheral nerve regeneration in diabetes mellitus. J Peripher Nerv Syst 10: 144–157. Kennedy W, Navarro X, Sutherland D (1995). Neuropathy profile of diabetic patients in a pancreas transplantation program. Neurology 45: 773–780. Kikta DG, Breuer AC, Wilbourn AJ (1982). Thoracic root pain in diabetes: the spectrum of clinical and electromyographic findings. Ann Neurol 11: 80–85. Kincaid J, Price K, Jimenez M et al. (2007). Correlation of vibratory quantitative sensory testing and nerve conduction studies in patients with diabetes. Muscle Nerve 36: 821–827.

784

G. CHARNOGURSKY ET AL.

Liatis S, Marinou K, Tentolouris N et al. (2007). Usefulness of a new indicator test for the diagnosis of peripheral and autonomic neuropathy in patients with diabetes mellitus. Diabet Med 24: 1375–1380. Low P, Nickander K, Tritcshler H (1997a). The role of oxidative stress and antioxidant treatment in experimental diabetic neuropathy. Diabetes 46: S38–S42. Low P, Gilden J, Freeman R et al. (1997b). Efficacy of midodrine vs placebo in neurogenic orthostatic hypotension: a randomized, double-blind multicenter study. JAMA 277: 1046–1051. Maganti K, Onyemere K, Jones M (2003). Oral erythromycin and symptomatic relief in gastroparesis: a systematic review. Am J Gastroenterol 98: 259–263. Martin C, Albers J, Herman W et al. (2006). The DCCT/EDIC research group. Neuropathy among the diabetes control and complications trial cohort 8 years after trial completion. Diabetes Care 29: 340–344. Maser R, Pfeifer M, Dorman J et al. (1990). Diabetic autonomic neuropathy and cardiovascular risk. Pittsburgh Epidemiology of Diabetes Complications Study III. Arch Intern Med 150: 1218–1222. Mehra S, Tavakoli M, Kallinikos P et al. (2007). Corneal confocal microscopy detects early nerve regeneration after pancreas transplantation in patients with type 1 diabetes. Diabetes Care 30: 2608–2612. Navarro X, Kennedy W, Aeppli D et al. (1996). Neuropathy and mortality in diabetes: influence of pancreas transplantation. Muscle Nerve 19: 1009–1016. Negrin P, Zara G (1995). Conduction studies as prognostic parameters in the natural history of diabetic neuropathy: a long-term follow-up of 114 patients. Electromyogr Clin Neurophysiol 35: 341–350. Newrick P, Wilson A, Jakubowski J et al. (1986). Sural nerve oxygen tension in diabetes. Br Med J (Clin Res Ed) 293: 1053–1054. Niakan E, Harati Y, Rolak L et al. (1986). Silent myocardial infarction and diabetic cardiovascular autonomic neuropathy. Arch Intern Med 146: 2229–2230. O’Brien IA, McFadden JP, Corrall RJ (1991). The influence of autonomic neuropathy on mortality in insulin-dependent diabetes. Q J Med 79: 495–502. Onde M, Ozge A, Senol M et al. (2008). The sensitivity of clinical diagnostic methods in the diagnosis of diabetic neuropathy. J Int Med Res 36: 63–70. Orchard T, Lloyd C, Maser R et al. (1996). Why does diabetic autonomic neuropathy predict IDDM mortality? An analysis from the Pittsburgh Epidemiology of Diabetes Complications Study. Diabetes Res Clin Pract 34 (Suppl): S165–S171. Oyer D, Saxon D, Shah A (2007). Quantitative assessment of diabetic peripheral neuropathy with use of the clanging tuning fork test. Endocr Pract 13: 5–10. Pacher P, Obrosova I, Mabley J et al. (2005). Role of nitrosative stress and peroxynitrite in the pathogenesis of diabetic complications. Emerging new therapeutical strategies. Curr Med Chem 12: 267–275. Patel A, MacMahon S, Chalmers J et al. (2008). Intensive blood glucose control and vascular outcomes in patients with type 2 diabetes. N Engl J Med 358: 2560–2572.

Pecket P, Schattner A (1982). Concurrent Bell’s palsy and diabetes mellitus: a diabetic mononeuropathy? J Neurol Neurosurg Psychiatry 45: 652–655. Quattrini C, Jeziorska M, Boulton A et al. (2008). Reduced vascular endothelial growth factor expression and intraepidermal nerve fiber loss in human diabetic neuropathy. Diabetes Care 31: 140–145. Rathmann W, Ziegler D, Jahnke M et al. (1993). Mortality in diabetic patients with cardiovascular autonomic neuropathy. Diabet Med 10: 820–824. Reljanovic M, Reichel G, Rett K et al. (1999). Treatment of diabetic polyneuropathy with antioxidant thioctic acid (a-lipoic acid): a two-year multi-center randomized double-blind placebo-controlled trial (ALADIN II). Alpha Lipoic Acid in Diabetic Neuropathy. Free Radic Res 31: 171–179. Ropper A, Gorson K, Gooch C et al. (2009). Vascular endothelial growth factor gene transfer for diabetic polyneuropathy: a randomized, double-blinded trial. Ann Neurol 65: 386–393. Rowbotham M, Goli V, Kunz N et al. (2004). Venlafaxine extended release in the treatment of painful diabetic neuropathy: a double-blind, placebo- controlled study. Pain 110: 697–706. Sampson M, Wilson S, Karagiannis P et al. (1990). Progression of diabetic autonomic neuropathy over a decade in insulindependent diabetics. Q J Med 75: 635–646. Sandercock D, Cramer M, Wu J et al. (2009). Gabapentin extended release for the treatment of painful diabetic neuropathy. Diabetes Care 32: e20. Shibao C, Okamoto L, Gamboa A et al. (2010). Comparative efficacy of yohimbine against pyridostigmine for the treatment of orthostatic hypotension in autonomic failure. Hypertension 56: 847. Singer W, Sandroni P, Opfer-Gehrking T et al. (2006). Pyridostigmine treatment trial in heneurogenic orthostatic hypotension. Arch Neurol 63: 513. Singh R, Barden A, Mori T et al. (2001). Advanced glycation end products: a review. Diabetologia 44: 129–146. Singleton J, Smith A (2007). Neuropathy associated with prediabetes: what is new in 2007? Curr Diab Rep 7: 420–424. Spallone V, Maiello M, Cicconetti E et al. (1997). Autonomic neuropathy and cardiovascular risk factors in insulindependent and non insulin-dependent diabetes. Diabetes Res Clin Pract 34: 169–179. Stamboulis E, Vassilopoulos D, Kalfakis N (2005). Symptomatic focal mononeuropathies in diabetic patients: increased or not? J Neurol 252: 448–452. Stewart JD (1989). Diabetic truncal neuropathy: topography of the sensory deficit. Ann Neurol 25: 233–238. Sugimoto K, Yasujima M, Yagihashi S (2008). Role of advanced glycation end products in diabetic neuropathy. Curr Pharm Des 14: 953–961. Sumner C, Sheth S, Griffin J et al. (2003). The spectrum of neuropathy in diabetes and impaired glucose tolerance. Neurology 60: 108–111. Tavakoli M, Quattrini C, Abbot D (2010). Corneal confocal microscopy. A novel noninvasive test to diagnose and stratify the severity of human diabetic neuropathy. Diabetes Care 22: 1792–1797.

DIABETIC NEUROPATHY Tesfaye S, Chaturvedi N, Eaton S et al. (2005). Vascular risk factors and diabetic neuropathy. N Engl J Med 352: 341–350. Thornalley P (2002). Glycation in diabetic neuropathy: characteristics, consequences, causes, and therapeutic options. Int Rev Neurobiol 50: 37–57. Tomlinson D (1999). Mitogen-activated protein kinase C as glucose transducers for diabetic complications. Diabetologia 42: 1271–1281. UK Prospective Diabetes Study Group (UKPDS) (1994). XI. Biochemical risk factors in type 2 diabetic patients at diagnosis compared with age-matched normal subjects. Diabet Med 11: 534–544. UK Prospective Diabetes Study (UKPDS) Group (1998). Intensive blood-glucose control with sulphonylureas or insulin compared with conventional treatment and risk of complications in patients with type 2 diabetes (UKPDS 33). Lancet 352: 837–853. Van Etten R, deKoning F, Verhaar M et al. (2002). NO-dependent vasodilation in patients with type II non-insulin-dependent diabetes mellitus is restored by acute administration of folate. Diabetologia 45: 1004–1010. Verhaar M, Stroes E, Rabelink T (2002). Folates and cardiovascular disease. Arterioscler Thromb Vasc Biol 22: 6–11. Vincent A, Russell J, Low P et al. (2004). Oxidative stress in the pathogenesis of diabetic neuropathy. Endocr Rev 25: 612–628. Vinik AI, Ziegler D (2007). Diabetic cardiovascular autonomic neuropathy. Circulation 115: 387–397. Vinik A, Maser R, Mitchell B et al. (2003). Diabetic autonomic neuropathy. Diabetes Care 26: 1553–1579. Vinik A, Bril V, Kempler P et al. (2005). Treatment of symptomatic diabetic peripheral neuropathy with the protein kinase C b-inhibitor ruboxistaurin mesylate during a 1year, randomized, placebo-controlled, double-blind clinical trial. Clin Ther 27: 1164–1180. Walker M, Morris L, Cheng D (2010). Improvement of cutaneous sensitivity in diabetic peripheral neuropathy with combination L-methylfolate, methylcobalamin and pyridoxal 5-phosphate. Rev Neurol Dis 7: 132–139. Wile D, Toth C (2010). Association of metformin, elevated homocysteine, and methylmalonic acid levels and clinically worsened diabetic peripheral neuropathy. Diabetes Care 33: 156–161.

785

Wright R, Kaufmann H, Perera R et al. (1998). A double-blind, dose-response study of midodrine in neurogenic orthostatic hypotension. Neurology 51: 120–124. Zhang W, Wan Po A (1994). The effectiveness of topically applied capsaicin: a meta analysis. Eur J Clin Pharmacol 45: 517–522. Zhong X, Zheng B, Hu G et al. (1981). Peripheral and autonomic nerve function tests in early diagnosis of diabetic neuropathy: correlation between motor nerve conduction velocity and fasting plasma glucose. Chin Med J (Engl) 94: 495–502. Ziegler D, Gries F, Spuler M et al. (1992). The epidemiology of diabetic neuropathy. Diabetic Cardiovascular Autonomic Neuropathy Multicenter Study Group. J Diabetes Complications 6: 49–57. Ziegler D, Hanefeld M, Ruhnau K et al. (1995). Treatment of symptomatic diabetic peripheral neuropathy with the antioxidant a-lipoic acid. A three-week multicentre randomized controlled trial (ALADIN study). Diabetologia 38: 1425–1433. Ziegler D, Hanefeld M, Ruhnau K (1999). Treatment of symptomatic diabetic polyneuropathy with the antioxidant alpha-lipoic acid: a 7-month multicenter randomized controlled trial (ALADIN III study). Diabetes Care 22: 1296–1301. Ziegler D, Nowak H, Kempler P (2004). Treatment of symptomatic diabetic polyneuropathy with the antioxidant alpha-lipoic acid: a meta analysis. Diabet Med 21: 114–121. Ziegler D, Ametov A, Barinov A et al. (2006). Oral treatment with alpha lipoic acid improves symptomatic diabetic polyneuropathy-the SYDNEY 2 trial. Diabetes Care 29: 2365–2370. Ziegler D, Rathmann W, Dickhaus T et al. (2008). Prevalence of polyneuropathy in pre-diabetes and diabetes is associated with abdominal obesity and macroangiopathy: the MONICA/KORA Augsburg surveys S2 and S3. Diabetes Care 31: 464–469. Ziegler D, Hidvegi T, Gurieva I et al. (2010). Efficacy and safety of lacosamide in painful diabetic neuropathy. Diabetes Care 33: 838–841. Ziegler D, Low P, Litchy W et al. (2011). Efficacy and safety of antioxidant treatment with alpha lipoic acid over 4 years in diabetic polyneuropathy. The NATHAN 1 trial. Diabetes Care 34: 2054–2060.

Handbook of Clinical Neurology, Vol. 120 (3rd series) Neurologic Aspects of Systemic Disease Part II Jose Biller and Jose M. Ferro, Editors © 2014 Elsevier B.V. All rights reserved

Chapter 52

Neurologic disorders associated with disease of the ovaries and testis JORGE C. KATTAH1* AND WILLIAM C. KATTAH2 Department of Neurology, University of Illinois College of Medicine, Peoria, IL, USA

1

2

Endocrinology Department, University of the Andes, Bogota, Colombia

INTRODUCTION

HISTORY

Whereas cerebral metastases are a known, albeit uncommon, complication of endothelial ovarian cancer (Pectasides et al., 2006), germ cell tumors of the testis rarely metastasize to the brain. Tumors of the testis and ovaries may also be associated with paraneoplastic encephalitis with well-defined clinical phenotypes and serum and CSF markers. These paraneoplastic neurologic syndromes are frequently the initial symptom of these tumors and may represent a formidable diagnostic and treatment challenge The first reported paraneoplastic syndrome due to ovarian cancer was a progressive cerebellar degeneration associated with the anti-Yo tumor marker, often reported among women in the sixth decade and associated with epithelial carcinoma of the ovaries (Greenlee and Brashear, 1983). Subsequently, the anti-Ma-2 serum marker was identified in patients with testicular germ cell tumors and three clinical phenotypes were recognized: limbic encephalitis, hypothalamic hypersomnia (narcolepsy/cataplexy), and a slowly progressive upper brainstem ophthalmoplegia (Voltz et al., 1999; Dalmau et al., 2004; Tenner and Einhorn, 2009). More recently, an anti-N-methyl-D-aspartate (NMDA) receptor encephalitis associated with ovarian teratoma was described (Dalmau et al., 2007; Dalmau et al., 2008; Florance et al., 2009; Tuzun et al., 2009; Lancaster et al., 2010). Less common manifestations of the anti-NMDA antibody syndrome include status epilepticus, dyskniesias of the trunk and face, and jaw dystonia.

Neurologic complications of tumors of the ovaries and testis represent the most common interface between neurologists, gynecologic oncologists and urologic oncologists. From 1993 to 1996, 4% of all ovarian cancer patients evaluated at Memorial Sloan-Kettering Cancer Center, 83, had 121 neurologic consultations. Causes for consultation were iatrogenic complications (n ¼ 38), metastasic disease (n ¼ 27), cerebrovascular disease (n ¼ 14), and paraneoplastic syndromes (n ¼ 4). More than half of the patients had improvement following neurologic intervention (Abrey and Dalmau, 2000). Non-seminomatous tumors are more likely to be associated with cerebral metastases (Bokemeyer et al., 1997), and a brain hemorrhage may be a complication of metastatic choriocarcinoma of the testis (Salvati and Cervoni, 1994; Fadul, 2008). Following radiotherapy of testicular tumors, lumbosacral plexus and lower spinal cord syndromes may develop with a variable latency. Neurologic complications of radiotherapy of the testis are probably more frequent than other postradiation complications such as de novo pelvic and other neoplasms or accelerated iliofemoral atherosclerosis. Neurologic phenotypes of paraneoplastic syndromes associated with cancer of the ovaries and testis are summarized in Table 52.1. These syndromes may precede tumor diagnosis in more than half of the cases. The neurologic syndrome combined with a lymphocytic cerebrospinal fluid (CSF) reaction, increased IgG synthetic rate,

*Correspondence to: Jorge C. Kattah, M.D., Department of Neurology, University of Illinois College of Medicine at Peoria and the Illinois Neurological Institute at OSF Saint Francis Medical Center, 530 NE Glen Oak Avenue, Peoria, IL 61637, USA. E-mail: [email protected]

Table 52.1 Paraneoplastic syndromes associated with cancer of the ovary and the testis Antibody

Syndrome

Target

CNS pathology

MRI findings

CSF

Common tumors

Treatment

Anti-Yo

Pure cerebellar degeneration

CDR2 protein in cytoplasm Purkinje cells

Loss of Purkinje cells Inflammation

1. Normal early 2. Cerebellar atrophy late

Ovarian cancer Breast cancer

Oncotherapy Immunoglobulin Steroids Cytoxan

Anti-Ma-2

1. Limbic encephalitis 2. Narcolepsy 3. Vertical gaze palsy 4. Ataxia Parkinsonism Progressive encephalopathy Delirium psychosis Involuntary facial movement Hypoventilation Dysautonomia

Unknown cytoplasmic neuronal protein

Perivascular lymphocytic cuffing Variable gliosis Neuronal degeneration

Increased signal in hippocampus diencephalon upper brainstem

Lymphocytic Pleocytosis OCB* High protein Lymphocytic Pleocytosis OCB* High protein

Germ cell tumor of testis Lung cancer

Orchidectomy Oncotherapy Immunotherapy

NR1 þ NR2 NMDA** receptor in hippocampus, forebrain, basal ganglia spinal cord

Loss of neurons in hippocampus Gliosis IgG deposits T cells infrequent

Loss of pyramidal cells in hippocampus Gliosis Cerebellum is spared

Teratoma of the ovary in females over 18

Immunotherapy Oncotherapy Tumor resection

Anti-NMDA

*OCB, oligoclonal bands. **NMDA, N-methyl-D-aspartate receptor.

Lymphocytic Pleocytosis OCB* High Protein

NEUROLOGIC DISORDERS ASSOCIATED WITH DISEASE OF THE OVARIES AND TESTIS and oligoclonal bands may prompt the astute neurologist to order a clinical pelvic examination or testicular examination, serum and CSF ovarian and testis markers, and pertinent imaging studies, thus, neurologists may have a very important role to play in these patients, as often gynecologists, oncologists, and urologists may be skeptical about the potential significance of clinical and serum makers and the need for urgent diagnosis and intervention. In these instances, neurologists must convincingly argue the potential relationship between the observed phenotype and a primary gonadal tumor. The annual incidence of epithelial ovarian carcinoma in the US has been estimated to be 12.9/100 000 among women aged 50–54 years. The annual incidence of newly detected testicular tumors in the US has been estimated at between 7500 and 8000. Over a lifetime, the risk of testicular carcinoma is approximately 1 in 250 (0.4%). Testicular tumors are the most common solid tumors in men between the ages of 15 and 34 years (Bosl and Motzer, 1997).

CLINICAL FINDINGS Local pelvic neurologic symptoms of ovarian tumors. Unilateral lumbar plexus syndromes could be seen as a late complication of ovarian carcinoma (Beatrous et al., 1990). Patients typically develop severe pelvic pain, unilateral painful weakness of the iliopsoas and quadriceps muscles, edema of the lower extremity, and foot drop. Isolated malignant infiltration of the psoas muscle may also occur with ovarian tumors. A late lumbosacral plexopathy secondary to irradiation, characterized by painless weakness and atrophy of the lower extremities, may be seen in patients with ovarian carcinoma; this is often seen several years after initial irradiation (Numaba et al., 1990). Brain metastases of ovarian carcinoma are uncommon and usually associated with poor prognosis (Pectasides et al., 2006). The approximate incidence of cerebral metastases is 1%,(0.4–6 %). Most patients with CNS metastases have stage III and IV disease. Isolated brain relapse may be seen in a third of the patients. Single metastases occur in one-third of cases. Multiple metastases occur in the rest. Most lesions are located in the cerebral hemispheres. The median interval between initial tumor diagnosis and brain metastases ranges from 15 to 70 months. Hemiparesis and altered mental status are the most common finding (Pectasides et al., 2006). Early pelvic complications of tumors of the testis are distinctly uncommon; however, late complications from radiotherapy may occur with greater frequency, particularly if the average total radiation dose exceeds 40 Gy. Following a variable latency, the clinical picture involves progressive lower extremity flaccid weakness and atrophy with sparing of sphincters function

789

(Greenfield and Stark, 1948; Bowen et al., 1996; Knap et al., 2007). Approximately 10% of all nonmetastatsic neurologic complications in patients with cancer are paraneoplastic (Tenner and Einhorn, 2009). In two-third of cases, the neurologic syndrome precedes the diagnosis of ovarian or testicular tumor (Table 52.1). A progressive cerebellar degeneration is typical in cases of the anti-Yo antibody syndrome (Figs 52.1 and 52.2). The majority of anti-Yo cases are characterized by a pure, progressive limb and truncal cerebellar ataxia (Hudson et al., 1993; Abrey and Dalmau, 2000; Keime-Guibert et al., 2000; Dorn et al., 2003). Common paraneoplastic neuroophthalmologic manifestations of gonadal tumors are listed in Table 52.2 and include ophthalmoplegia, opsoclonus, nystagmus, and fixation intrusions, etc. (Ko et al., 2008). Clinical manifestations of the anti-Ma-2 antibody syndrome associated with germ cell tumors of the testicles include three characteristic clinical phenotypes: (1) limbic encephalitis with short-term memory loss, altered mental status, and psychomotor seizures; (2) hypothalamic syndromes with excessive daytime sleepiness, cataplexy, hypnagogic hallucinations, hyperthermia, and endocrinopathies; (3) vertical gaze palsy. Less common presentations include brainstem encephalitic syndromes with encephalopathy, long tract signs, ophthalmoparesis, vestibular dysfunction, nystagmus, ataxia, and cranial neuropathies. Overlap syndromes with simultaneous limbic system, diencephalon, and brainstem structures are less common (Dalmau et al., 2004; Ko et al., 2008). The anti-NMDAR antibody syndrome is characterized by a viral prodrome, prominent psychiatric symptoms, encephalopathy, abnormal facial and jaw movements, autonomic instability, and central hypoventilation requiring frequent intubation (Dalmau et al., 2007, 2008; Florance et al., 2009; Tuzun et al., 2009; Lancaster et al., 2010).

NATURAL HISTORY Pelvic complications of tumors of the ovaries or testis represent late complications of irradiation and are far more common with testicular tumors (Greenfield and Stark, 1948; Numaba et al., 1990; Bowen et al., 1996; Knap et al., 2007). Single metastatic ovarian carcinoma to the brain may transiently respond favorably to surgical resection, irradiation, and chemotherapy, with average survival rates varying from 16 to 27 months (Cormio et al., 2003; D’Andrea et al., 2004; Pectasides et al., 2006). In general, paraneoplastic syndromes without treatment cause death from either oncologic or neurologic causes within a few months from initial symptoms. Whereas the anti-Yo antibody has poor prognosis, favorable response of tumor treatment with human

790

J.C. KATTAH AND W.C. KATTAH

A

B Fig. 52.1. (A) Observe saccadic horizontal (h) pursuit when the patient tracks a target toward her right and to a lesser extent to the left. Tracings were obtained in a 67-year-old woman with an acute vestibular syndrome, saccadic horizontal pursuit and downbeat nystagmus. The workup showed a positive anti-Yo antibody and she was found to have stage 3B ovarian carcinoma. (B) Observe a 10 second recording of central fixation in the same patient. Downbeat nystagmus in the primary position of gaze; frequency: 1.5 Hz, amplitude 3 degrees and velocity 3 degrees/sec is observed in the vertical V tracing.

A

B

Fig. 52.2. (A) Axial CT scan of the abdomen obtained in the patient described in Figures 52.1 (A and B). It demonstrates a cystic mass involving the right ovary (arrow). Histopathologic examination established a diagnosis of carcinoma of the ovary. (B) Ovarian cancer in patient with anti-Yo syndrome described in Figure 52. 1A. H&E 400. Observe large nuclei and multinucleated cells. Mitosis can be observed in several cells.

NEUROLOGIC DISORDERS ASSOCIATED WITH DISEASE OF THE OVARIES AND TESTIS

791

Table 52.2 Neuro-ophthalmologic paraneoplastic syndromes in tumors of the ovary and testis Syndrome

Neurophthalmologic findings

Tumor

Anti-Yo antibody

Downbeat nystagmus Direction changing Horizontal nystagmus Saccade dysmetria Saccadic pursuit Vertical gaze palsy* Complete gaze palsy Hearing loss/vertigo OTR/skew INO/sixth nerve palsy Up and downbeat nystagmus Opsoclonus/ocular flutter Positional central nystagmus** Visual hallucinations Oculogyric crisis Opsoclonus

Epithelial cancer of the ovary

Anti-Ma-2

Anti-NMDA antibody

Germinoma of the testis

Teratoma of the ovary

*Vertical gaze abnormalities are frequent in the anti-Ma-2 antibody syndrome and the differential frequently includes Whipple’s disease (Dalmau et al., 2004; Ko et al., 2008). **Patient with a paraneoplastic cerebellar degeneration and testicular neoplasia presented with episodic vertigo and positional central nystagmus, he had a negative anti-Ma-2 antibody, was found to have a germinoma in undescended testis, and improved following bilateral orchidectomy and immunotherapy (Tafur et al., 2008).

immunoglobulin has been reported (Keime-Guibert et al., 2000; Dorn et al., 2003). The anti-NMDAR antibody syndrome responds well to tumor resection and immunosuppression (Ferioli et al., 2010; Kurlan et al., 2010). Likewise, the anti-Ma-2 antibody syndrome responds to orchidectomy, even in cases of testicular germinoma in situ (Dalmau et al., 2004).

LABORATORY INVESTIGATIONS Ovarian cancer tumor markers are indicated when there is a clinical suspicion that a pelvic lesion is a primary tumor of the ovary. CA 125 may be elevated in 83% of women with ovarian cancer. When a teratoma of the ovary is suspected, a-fetoprotein, human choriogonadotropic hormone (h-CGD), and lactate dehydrogenase can be confirmatory. When suspecting germ cell tumors of the testicle, a-fetoprotein, h-CGD, and lactate dehydrogenase may also be helpful (Bosl and Motzer, 1997). Investigation for paraneoplastic syndromes includes a panel of antibodies ordered on the basis of the clinical phenotype. Anti-Yo (PCA-1) antibodies, anti-Ma-2 protein antibodies and anti-NMDA receptor antibodies are known serum/ CSF markers associated with the tumors under consideration in this chapter. However, since the clinical findings may not be fully defined at the time of the initial workup, a more extensive panel of antibodies is often requested,

including anti-Tr and mGUR1 antibodies, anti-ANNNA-1, ANNA-2, ANNA-3, CV2/CRMP-5, antiphysin and voltage gated calcium channel (VGCC) antibodies. A more recently introduced anti-LGI1 antibody may be added, particularly in cases thought to have anti-voltage gated K-channel antibodies (Meizan et al., 2010). CSF examination includes determination of protein content, glucose, and cell count, including cytology, CSF IgG, IgG synthetic rate, oligoclonal bands, and tumor markers for paraneoplastc syndromes.

NEUROIMAGING INVESTIGATIONS 1.

Tumor diagnosis localized to the pelvis. Ovarian tumors are first investigated with ultrasonography, computed tomography (CT) scan of the abdomen, or magnetic resonance imaging (MRI) with contrast. These investigations may provide an idea of the extent of tumor invasion (Fig. 52.2A) and are helpful in staging and surgery planning. Imaging and tumor markers are also helpful in differentiating benign versus malignant tumors. Testicular tumors are best differentiated from chronic orchitis and epididymitis with ultrasound. The typical germ cell tumor is intratesticular and has microcalcifications (one or more hypoechoic masses). More extensive testicular tumors are usually investigated with

792

2.

3.

J.C. KATTAH AND W.C. KATTAH pre- and postcontrast CT scan, MRI, and positron emission tomography (PET) scans which will be also useful for staging and treatment planning. In the event of lumbar or sacral plexus dysfunction these techniques may also show tumoral invasion of retroperitoneal structures. In cases of delayed postirradiation neurologic syndromes, there may be contrast enhancement of the cauda equina and lumbar and sacral roots (Bowen et al., 1996). Cerebral metastases of ovarian carcinoma. MRI with contrast is the most sensitive test in patients with suspected metastatic ovarian carcinoma. Characteristic imaging findings include multiple lesions, localization at the gray–white matter junction, and small tumor focus with significant vasogenic edema. Neuroimaging findings of paraneoplastic syndromes. The anti-Yo antibody syndrome is usually associated with a normal MRI in the initial phase; cerebellar atrophy develops in later stages. The anti-Ma-2 antibody syndrome may be associated with asymmetric areas of increased signal in the temporal lobes, particularly the hippocampi, hypothalamus, and upper brainstem. These lesions typically enhance following contrast administration. Simultaneous multifocal lesions of the diencephalon and brainstem may also be found. PET scan may show areas of hypometabolism (Dalmau et al., 2004). Patients with the anti-NMDAR antibody syndrome may have areas of increased signal abnormalities in different locations of the cerebrum on long T2 and fluid attenuated inversion recovery (FLAIR) sequences scans (Lancaster et al., 2010).

PATHOLOGY Metastasic tumors of the ovaries to the brain. The histopathologic findings in metastatic tumor of the ovaries to the brain involve the characteristic findings of the primary tumor, associated with congestion and edema. The histopathologic findings in the late, postirradiation lower motor neuron syndrome following testicular and rarely with ovarian tumors has been reported in detail. Gross examination of the cauda equina shows irregular thickening with focal areas of hemorrhage. Microscopically, clusters of dilated vascular channels with thickened hyalinized walls are found with sparing of anterior horn cells (Bowen et al., 1996). On the basis of these neuropathologic findings, the term “postirradiation lumbosacral radiculopathy” has been proposed (Bowen et al., 1996; Knap et al., 2007). Paraneoplastic anti-Yo antibody syndrome. Neuropathologic examination of the cerebellum in patients with epithelial carcinomas of the ovaries show severe loss of Purkinje cells with proliferation of Bergmann glia

and infiltrates of inflammatory cells in the deep cerebellar nuclei. The actual mechanism of neuronal injury in this syndrome is not quite clear. The cytoplasmic Yo protein found in normal Purkinje and in ovarian carcinoma cells represent 34, 52, and 62 kDa cytoplasmic proteins known to interact with c-Myc. The disruption of the normal c-Myc cytoplasmic pathway leads to accelerated neuronal apoptosis. Anti-Yo antibodies may be seen in ovarian cancer, breast cancer, and at least one report of transitional cancer of the bladder. The anti-Yo antibody may also be present in patients with ovarian neoplasm without neurologic symptoms for prolonged follow-up periods. Anti-Ma-2 antibody syndrome. The neuropathology of the anti Ma-2 antibody syndrome associated with germ cell tumors of the testis involves perivascular lymphocytic cuffing, lymphocytic interstitial infiltrates, variable gliosis and neuronal degeneration; The majority of lymphocytes are T cells with a smaller number of B cells, macrophages, plasma cells, and microglia (Dalmau et al., 2004). The predominant pathogenic mechanism, hypothesizes that the neurologic deficit is mediated by cytotoxic T cells attacking neurons. The initial event is triggered by apoptotic tumor cells which are captured by tissue dendritic cells, generating a specific T cell-mediated response. The dendritic cells in turn activate CD8 þ and CD4-helper cells in lymph nodes, targeting cytoplasmic Ma proteins which are presumably involved in neuronal apoptosis prevention. (Tenner and Einhorn, 2009). The Ma-2 antibody has been found in other neoplasms, including lung, and in a recent report of hypothalamic neurosarcoidosis (Desestret et al., 2010). Anti-N-methyl-D-aspartate receptor (anti-NMDAR) antibody encephalitis. Extensive neuronal loss, microgliosis, T lymphocytes, IgG deposits, and rare inflammation are the most distinctive characteristics. Preferential CNS involvement include the hippocampi, forebrain, basal ganglia, and spinal cord (Tuzun et al., 2009). The Purkinje cells are relatively spared, and cells expressing T cell markers of cytotoxicity are infrequent. The histopathology of the ovarian teratomas studied in this syndrome contain nervous tissue (mature and immature neurons) confirmed by their morphologic characteristics and neuronal markers (microtubule-associated protein2). The absence of “encephalitis” on clinical grounds contrasts with the vigorous macrophage and T lymphocyte inflammatory reaction found on pathologic examination (Dalmau et al., 2008; Tuzun et al., 2009).

MANAGEMENT Management of cerebral metastases of carcinoma of the ovaries depends on number and locations of these

NEUROLOGIC DISORDERS ASSOCIATED WITH DISEASE OF THE OVARIES AND TESTIS lesions. Solitary metastases in noneloquent brain areas are best treated with surgical resection followed by whole brain irradiation (Salvati and Cervoni, 1994; Cormio et al., 2003; Pectasides et al., 2006). Surgical resection followed by whole brain irradiation and platinum-based chemotherapy has been advocated (Cormio et al., 2003). However, the role of chemotherapy has not been studied in detail. Platinum chemotherapy may be associated with transient neurologic deficits, including status epilepticus and cortical blindness and posterior reversible encephalopathy syndrome (Kattah et al., 1987). Moreover, platinum treatment is frequently associated with peripheral neuropathy and nephropathy. The management of the paraneoplastic syndromes associated with tumors of the ovaries and testes involve optimal oncologic management with resection of the primary tumor and/or immunotherapy. In the anti-Ma-2 (Dalmau et al., 2004) and the anti-NMDAR syndromes (Dalmau et al., 2007, 2008), neurologic improvement is possible and frequent. The anti-Yo syndrome may be more difficult to treat (Keime-Guibert et al., 2000). However, chemotherapy and, more recently, rituximab have been shown to be helpful (Shams’ili et al., 2006; Esposito et al., 2008; Goret et al., 2008). Successful management of the anti-NMDA antibody syndrome can also be accomplished in pregnant women with ovarian teratomas with a good outcome for the mother and neonate (Kumar et al., 2010).

INFERTILITY, INFECUNDITY, ANDROGEN RECEPTOR INSENSITIVITY SYNDROME, AND PRIMARY OVARIAN FAILURE IN PATIENTS WITH NEUROLOGIC DISORDERS Approximately 5–15% of couples in the reproductive age are unable to either become pregnant or maintain pregnancy to full term. Most cases (56%) relate to female infertility, about 25% to male infertility, and have a mixed cause in the remainder. Infertility deserves expert consultation after a period exceeding 1 year without successful pregnancy. Predictably, individuals in their thirties and forties have higher rates of infertility than the younger counterparts. A number of genetic X-linked neurologic disorders are associated with androgen receptor insensitivity (ARI), leading to testicular atrophy, oligospermia/azoospermia, gynecomastia and infertility in males and primary ovarian failure (POF) in carrier females. Other non-X chromosome genetic neurologic disorders, neuroendocrine syndromes, and commonly acquired neurologic syndromes, such as spinal cord injury and multiple sclerosis, among others, may affect male fertility.

793

Common, non-neurologic causes of infertility in males and females Female “causes” for infertility include sequelae from sexually transmitted diseases, endometriosis, ovulatory dysfunction, poor nutrition, hormone imbalance, ovarian cysts, pelvic infection, tumors, or blockage of transport from the cervix through the fallopian tubes. Common “male” causes for infertility include testicular tumors, varicoceles, testicular atrophy, low sperm volume, low concentration or absence of sperm per milliliter of semen (oligospermia, azoospermia), or low sperm motility. Once preliminary tests have been conducted, when screening men and women for potential infertility causes the cervical mucus penetration assay test is performed. If less than 5–20% of uncovered hamster eggs are penetrated by spermatozoa, infertility is diagnosed (Healy et al., 1999; Montella et al., 2000; Bhasin, 2007). From the aforementioned it is clear to neurologists that only a few neurologic disorders will be associated with AIS or POF. However, awareness of the possible association may be helpful in the patient’s management and may lead to proper gynecologic, urologic, and genetic counseling. Table 52.3 is a list of the most common neurologic entities associated with POF and AIS. These are discussed briefly as they are associated with AIS in males and POF in females.

Fragile X syndrome INTRODUCTION The fragile X syndrome results from an expansion of CGG repeats in the FMR1 gene which encodes the FMRP protein critical for intellectual development. Individuals with CGG repeats from 55 to 200 are considered premutation (carriers). More than 200 CGG repeats are associated with the fragile X syndrome (Hagerman and Hagerman, 2004; Wattendorf and Muenke, 2005).

HISTORY The fragile X syndrome is the most common cause of inherited mental retardation with a variable phenotype depending on number of trinucleotide repeats in the X chromosome. Male adults with low CGG repeat expansion may have late onset ataxia and tremor (fragile Xassociated tremor/ataxia syndrome, FXTAS). Women carriers may present with POF. A high number of CGG repeat expansion causes the full fragile X phenotype. It is possible that the father, and to a lesser extent the mother, of fragile X syndrome patients may develop late-onset FXTAS.

794

J.C. KATTAH AND W.C. KATTAH

Table 52.3 Testicular atrophy, azoospermia, primary ovarian failure in common neurologic disorders Disease and androgen receptor (AR) gene expansion*

Primary ovarian failure (POF)

Testis

Male phenotype

Infertility

þ

Macroorchidism

Normal

Common in males POF in females**

Moderate androgen receptor insensitivity (ARI) Severe ARI

Gynecomastia Micropenis Testicular atrophy

Undervirilization Gynecomastia Female phenotype

Fertility possible Infertility

Possible

Phenotypically female. Testis in abdomen. Ambiguous genitalia Testicular atrophy Azoospermia in > than 18 CAG/CTG repeats

Gynecomastia

Infertility depends on CAG/CTG repeats

Machado Joseph’s spinocerebellar ataxia

Unknown

Depends on CAG repeats Azoospermia > than 29 repeats

Normal

Infertility edepends on CAG repeats

3 CAG repeats Mitochondrial disorders Spinal cord injury þ myelopathy

Possible

Normal

Normal

Infertility due to decreased spermatozoa mobility

Not present

Normal

Normal

Sperm abnormal Spermatozoa normal

Fragile X syndrome CGG repeats Kennedy’s syndrome CAG repeats Mild CAG repeats in AR < 28 repeats Severe >28 CAG repeats Myotonic dystrophy CAG/CTG repeats

*Idiopathic azoospermia ¼ 31 CAG repeats in the AR. **POF in premutation females ¼ 55–200 CAG repeats.

CLINICAL FINDINGS

PATHOLOGY

Mental retardation, autism-like picture, and attention deficit hyperactivity disorder (ADHD), behavioral changes, a typical facies, scoliosis, and significant macrorchidism are common findings in the fragile X syndrome. Seizures in the late-onset form are common. Ataxia and tremor may be observed. Young patients with limited CGG repeats may be fertile and become infertile over time. Females may have POF (Sherman, 2000; Welt et al., 2004; Sullivan, 2005).

Histopathologic studies show no gross structural abnormalities, despite the profound degree of mental retardation (Nelson, 1998). Microscopic examination shows eosinophilic intranuclear inclusions in neuron and astrocytes throughout the cortex, subcortical structures and brainstem (Chaussenot et al., 2008), abnormal dendrite spine morphology with preservation of neuronal density in the neocortex may be found (Hinton et al., 1991).

NATURAL HISTORY Rate of progression of neurologic findings depends on the number of CGG repeats (usually greater than 200). The phenotype of a male with a smaller number of repeats is associated with a more benign clinical course which may manifest as late-onset essential type tremor and ataxia (FXTAS).

LABORATORY INVESTIGATIONS The cause of the fragile X syndrome is a defect in the synthesis of the fragile X mental retardation protein (FMRP), a regulatory protein in neurons and dendrites required for normal neuronal maturation. The presence of expanded CGG repeats interferes with the synthesis and action of FMRP, which has a role in ctytoplasmic mRNA metabolism (Nelson, 1998).

NEUROLOGIC DISORDERS ASSOCIATED WITH DISEASE OF THE OVARIES AND TESTIS 1.

2.

DNA testing is recommended. The exact number of CGG repeats in the FMR1 gene may be determined by Southern blot or polymerase chain reaction (PCR). The normal number of CGG repeats vary from 5 to 54. More than 55 CGG repeats are considered premutation. More than 200 CGG repeats define a full mutation. Mosaic patterns are common. Mothers of fragile X males have a premutation. Females with fragile X are less severely affected. Germane to this topic, premutation females may present with isolated POF. In fragile X-phenotype males and females CGG expansion may not be found, but point mutation in the FMR1 gene may be identified. MRI of the brain in the FXTAS shows cerebral and cerebellar atrophy, and nonspecific increased signal in the white matter, including the symmetric middle cerebellar peduncle sign (Berry-Kravis et al., 2007).

MANAGEMENT Symptomatic treatment of the neurologic manifestations and genetic counseling are the main treatment options (Berry-Kravis et al., 2007).

795

decades (Kennedy et al., 1968; Sperfeld et al., 2002). During puberty, overt signs of undervirilization may be noted, but the more severe forms of ARI are not associated with this disease (Nistche and Hiort, 2000; Wisniewski et al., 2000).

PATHOLOGY Muscle biopsy shows predominant neurogenic and, to a lesser extent, myopathic changes.

LABORATORY INVESTIGATIONS Testosterone levels are decreased, estradiol may be increased, and luteinizing hormone (LH) may be increased. All of these hormonal levels correlate with the amount of CAG repeats in the AR. Men with greater than 28 repeats have increased risk of impaired spermatogenesis. However, if clinical signs of AR develop in the fourth or fifth decade then they may able to procreate; 72% report having children. PCR for CAG repeats in exon 1 of the AR gene will demonstrate a diagnostic expansion (greater than 26 CAG repeats).

MANAGEMENT

Kennedy’s Disease

Other than symptomatic treatment and genetic counseling, there is little else to offer to these patients.

Also known as spinobulbar muscular atrophy (SBMA), this is an X-linked spinal bulbar neuronopathy associated with androgen receptor insensitivity (La Spada et al., 1991).

Myotonic dystrophy

INTRODUCTION

HISTORY Muscle weakness and wasting are usually present between the ages of 15 and 60 years. Muscle weakness is mostly proximal. Fasciculations involving the facial muscles and bulbar musculature are common. Insensitivity to male hormones, gynecomastia, microrchidism, infertility and impotence may be present in the teenage years or develop in the forties or fifties. In some cases with milder forms, fertility may be normal. Diabetes mellitus may also be present (Kennedy et al., 1968).

NATURAL HISTORY The natural history varies with the degree of CAG repeats in the androgen receptor located in chromosome Xq 11-12 which causes progressive loss of motoneurons (normal CAG repeats: 17–26). An unstable expansion of the CAG repeat in exon 1 of the androgen receptor (AR) gene is the pathogenic mechanism. Progressive spinal atrophy may lead to an inability to ambulate. Bulbar musculature compromise may cause slowly progressive swallowing difficulties and dysarthria. In milder forms, the patient may be able to walk even in the fourth to fifth

INTRODUCTION Myotonic dystrophy (MD). The most frequent cause of muscular dystrophy in adults (Harper, 2001) is an autosomal dominant inherited disorder characterized by muscle weakness and wasting (dystrophy) and sustained involuntary muscle contractions (myotonia). Systemic manifestations of MD may be seen chiefly with cardiac, endocrine, and multiorgan compromise.

HISTORY Initial manifestations often occur in teenagers in the form of distal muscle weakness, atrophy, and myotonia. Both males and females are affected. Testicular atrophy, impotence, and loss of sex drive develops among MD males. Females often report loss of sex drive, premature menopause, and habitual abortions. Myotonia may be symptomatic. Cataracts may occasionally be the first manifestation; their features may be quite characteristic by slit lamp examination (Christmas tree, multicolored iridescent). Gynecomastia and lack of male sexual development, impotence, and infertility may be initial complaints (Fig. 52.3). Diabetes mellitus, glucose intolerance, respiratory failure, and cardiac arrhythmias/insufficiency may be late manifestations (Harper, 2001).

796

J.C. KATTAH AND W.C. KATTAH specificity (Shelbourne et al., 1993). EMG and muscle biopsy can offer diagnostic information but genetic testing is the recommended choice if possible. Prenatal and preimplantation diagnosis is possible and genetic intervention may be successful (Sermon et al., 1997; Kakorou et al., 2007).

TREATMENT Symptomatic treatment is the only therapeutic alternative. Management of systemic complications involves multiple specialists and from the aforementioned, genetic counseling is critical in this disorder.

Miscellaneous

Fig. 52.3. Observe gynecomastia in a patient with myotonic dystrophy and a complaint of infertility. He had low serum testosterone levels, testicular atrophy, and erectile dysfunction.

NATURAL HISTORY Clinical signs vary with the number of CTG trinucleotides in the myotonic dystrophy protein kinase (DMPK) gene (normal 5–30 repeats). The gene normally encodes the myotonic dystrophy protein kinase (myotininkinase), which is expressed primarily in muscle. Loss of DMPK results in altered muscle-specific chloride channel function. The gene is located in the short arm of chromosome 19 (19q). MD tends to be more severe with subsequent generations. The cause for infertility in males with MD is not clearly defined. Decreased sperm function has been found in the ejaculates of sterile MD patients (Hortas et al., 2000). However, infertility is not universal in MD males. Females may have higher risk of complications during pregnancy. A relationship between the number of CTG repeats and sterility has been reported (Hsiao, 2002). Increased CTG/CAG repeats in the androgen receptor have been identified in MD and Machado Joseph’s disease (Pan et al., 2002).

LABORATORY INVESTIGATIONS The clinical characteristics are usually diagnostic. However, a genetic diagnosis provides 100% sensitivity and

There are several genetic and acquired disorders of the hypothalamus and pituitary gland that result in secondary gonadal atrophy and infertility in addition to neurologic abnormalities. The main congenital disorders include: (1) Praeder–Labhart–Willi syndrome: congenital deficiency of gonadal releasing hormone (GnRH) with hypogonadism, mental retardation, facial dysmorphism and obesity (Ledbetter et al., 1981); (2) Kallmann syndrome: decreased GnRh and anosmia ataxia, hearing loss (Kallmann et al., 1943); (3) Laurence–Moon–Bardet– Biedl syndrome with hypogonadism, retinitis pigmentosa, obesity, and polydactyly (McLouglin and Shanklin, 1967; Laurence and Moon, 1886; Qureshi et al., 2003). Acquired causes of hypothalamic and pituitary gland dysfunction include pituitary tumors, craniopharyngioma, germinomas, granulomatous disorders, hystiocytosis and s/p therapeutic radiation (Kattah and Kattah, 2008). Infertility may also be a complication of mitochondrial disorders with reduced sperm motility (Folgero et al., 1993). Patients with spinal cord injury besides erectile dysfunction may also have abnormal semen, most likely due to denervation of the seminal vesicles and prostate with diminished sperm motility (Kafetsoulis et al., 2006; Brackett et al., 2010). Although sperm may be obtained without major difficulty, in these patients a successful pregnancy rate is low (Yamamoto et al., 1997).

REFERENCES Abrey LE, Dalmau JO (2000). Neurologic complications of ovarian carcinoma. Cancer 85: 127–133. Beatrous TE, Choyke PL, Frank JA (1990). Diagnostic evaluation of cancer patients with pelvic pain: comparison of scintigraphy, CT and MR imaging. AJR Am J Roentgenol 155: 85–88. Berry-Kravis E, Abrams L, Coffey SM et al. (2007). Fragile Xassociated tremor/ataxia syndrome: clinical features, genetics, and testing guidelines. Mov Disord 22: 2018–2030.

NEUROLOGIC DISORDERS ASSOCIATED WITH DISEASE OF THE OVARIES AND TESTIS Bhasin S (2007). Approach to infertility man. Clin Endocrinol Metab 92: 1995–2004. Bokemeyer C, Nowak P, Haupt A (1997). Treatment of brain metastases in patties with testicular cancer. J Clin Oncol 15: 1449–1454. Bosl GJ, Motzer RJ (1997). Testicular germ cell tumors. N Engl J Med 337: 242–254. Bowen J, Gregory R, Squier M et al. (1996). The postirradiation lower motor neuron syndrome neuropathy or radiculopathy. Brain 119: 1429–1439. Brackett NL, Lynne CM, Ibrahim E et al. (2010). Treatment of infertility in men with spinal cord injury: semen abnormalities. Nat Rev Urol 7: 162–172. Chaussenot A, Borg M, Bayreuther C et al. (2008). Late cerebellar ataxia associated with fragile X premutation. Rev Neurol 164: 957–963. Cormio G, Maneo A, Colamaria A et al. (2003). Surgical resection of solitary brain metastasis from ovarian carcinoma: an analysis of 22 cases. Gynecol Oncol 89: 116–119. D’Andrea GD, Roperto R, Dinia L et al. (2004). Solitary cerebral metastases from ovarian epithelial carcinoma: 11 cases. Neurosurg Rev 28: 120–123. Dalmau J, Graus F, Villarejo A et al. (2004). Clinical analysis of anti-Ma2-associated encephalitis. Brain 127: 1831–1844. Dalmau JO, Tuzun E, Wu HY et al. (2007). Paraneoplastic anti-N-methyl aspartate receptor encephalitis associated with ovarian teratoma. Ann Neurol 61: 25–36. Dalmau JO, Gleichman AJ, Hughes EG et al. (2008). AntiNMDA receptor encephalitis: case series and analysis of the effect of antibodies. Lancet Neurol 7: 1091–1098. Desestret V, Didelot A, Meyronet D et al. (2010). Neurosarcoidosis with diencephalitis and anti-Ma2 antibodies. Neurology 74: 772–774. Dorn C, Knobloch C, Kupka M et al. (2003). Paraneoplastic neurological syndrome: patient with anti-Yo antibody and breast cancer: a case report. Arch Gynecol Obstet 269: 62–65. Esposito M, Penza P, Orefice G et al. (2008). Successful treatment of paraneoplastic cerebellar degeneration with rituximab. J Neurooncol 86: 363–364. Fadul CE (2008). Testis and ovaries. In: J Biller (Ed.), The Interface of Neurology and Internal Medicine. Wolters Kluwer/Lippincott, Williams and Wilkins, Philadelphia, pp. 502–507. Ferioli S, Dalmau J, Kobet CA et al. (2010). Anti-N-methyl-D aspartate receptor encephalitis. Arch Neurol 67: 250–251. Florance NR, Davis RL, Lam C et al. (2009). Anti-N-methylD-aspartate receptor (NMDAR) encephalitis in children and adolescents. Ann Neurol 66: 11–18. Folgero T, Bertheussen K, Lindal S (1993). Andrology: mitochondrial disease and reduced sperm motility. Hum Reprod 11: 1863–1993. Goret F, Bosca I, Fratalia L et al. (2008). Long-lasting remission after rituximab treatment in a case of anti-HU associated neuronopathy and gastric pseudoobstruction. J Neurooncol 93: 421–423. Greenfield MM, Stark FM (1948). Post-irradiation neuropathy. Am J Roentgenol 60: 617–622.

797

Greenlee JE, Brashear HR (1983). Antibodies to cerebellar Purkinje cells in patients with paraneoplastic cerebellar degeneration and ovarian carcinoma. Ann Neurol 14: 609–613. Hagerman PJ, Hagerman RJ (2004). The fragile-X premutation: a maturing perspective. Am J Hum Genet 74: 805–816. Harper PS (2001). Myotonic Dystrophy. 3rd edn WB Saunders, London. Healy DL, Trounson AO, Anderson AN et al. (1999). Female infertility: causes and treatment. Lancet 343: 1539–1544. Hinton VJ, Brown WT, Wisnieswski K et al. (1991). Analysis of neocortex in three males with the fragile x-syndrome. Am J Med Genet 41: 289–294. Hortas ML, Castilla JA, Gil MT et al. (2000). Decreased sperm function of patients with myotonic dystrophy. Hum Reprod 15: 445–448. Hsiao KM (2002). Reported relationship between increased CTG repeat lengths in myotonic dystrophy and azoospermia. Hum Reprod 17: 1578–1583. Hudson CN, Curling M, Potsides P et al. (1993). Paraneoplastic syndromes in patients with ovarian neoplasia. J R Soc Med 86: 202–204. Kafetsoulis A, Brackett NL, Ibrahim E et al. (2006). Current trends of infertility in men with spinal cord injury. Fertil Steril 86: 781–789. Kakorou G, Dhnajal S, Daphni D et al. (2007). Preimplantation genetic diagnosis for myotonic dystrophy type 1: detection of crossover between the gene and the linked marker APOC2. Prenat Diagn 27: 111–116. Kallmann FJ, Schonfeld WA, Barrera SE et al. (1943). The genetic aspects of primary eunuchoidism. Am J Ment Defic 48: 203–206. Kattah JC, Kattah WC (2008). Pituitary and hypothalamus. In: J Biller (Ed.), The Interface of Neurology and Internal Medicine. Wolters Kluwer/Lippincott, Williams and Wilkins, Philadelphia, pp. 465–470. Kattah JC, Potolicchio SJ, Kotz HL et al. (1987). Cortical blindness and occipital lobe seizures induced by cys-platinum. Neurophthalmology 7: 99–104. Keime-Guibert F, Graus F, Fleury A et al. (2000). Treatment of paraneoplastic neurological syndromes with antineuronal antibodies (Anti-Hu, anti-Yo) with a combination of immunoglobulins, cyclophosphamide, and methylprednisolone. J Neurol Neurosurg Psychiatry 68: 479–482. Kennedy WR, Alter M, Sung JH et al. (1968). Progressive proximal spinal and bulbar muscular atrophy of late onset: a sex-linked recessive trait. Neurology 18: 671–680. Knap MM, Bentzen SM, Overgaard J (2007). Late neurological complications after irradiation of malignant tumors of the testis. Acta Oncol 46: 497–503. Ko MW, Dalmau J, Galetta S (2008). Neuro-ophthalmologic maanifestations of paraneoplastic syndromes. J Neuroophthalmol 28: 58–68. Kumar MA, Jain A, Dechant VE et al. (2010). Anti-N-methylD aspartate receptor encephalitis during pregnancy. Arch Neurol 67: 884–887.

798

J.C. KATTAH AND W.C. KATTAH

Kurlan M, Lalive PH, Dalmau JO et al. (2010). Opsoclonusmyoclonus syndrome in anti-methyl-D-aspartate receptor encephalitis. Arch Neurol 67: 118–121. La Spada AR, Wilson EM, Lubahn DB et al. (1991). Androgen receptor gene mutation in X-linked spinal and bulbar muscular atrophy. Nature 352: 77–79. Lancaster E, Lai M, Peng X (2010). Antibodies to the GABAB receptor in limbic enecephalitis with seizures: case series and characterization of the antigen. Lancet Neurol 9: 67–76. Laurence JZ, Moon RC (1886). Four cases of retinitis pigmentosa occurring in the same family and accompanied by general imperfections of metabolism. Ophthalmol Rev 2: 32. Ledbetter DH, Riccardi VM, Airhart SD et al. (1981). Deletion of chromosome 15 as a cause Praeder–Willi syndrome. N Engl J Med 304: 325–329. McLouglin TG, Shanklin JR (1967). Pathology of Laurence– Moon–Bardet–Biedl syndrome. J Pathol 93: 65–79. Meizan L, Maarteje GMH, Lancaster E et al. (2010). Investigation of LGI1 as the antigen in lı´mbic encephalitis previously attributed to potassium channels: a case series. Lancet Neurol 9: 776–785. Montella K, Keekly E, Laifer S et al. (2000). Evaluation and management of infertility in women: the internist role. Ann Intern Med 132: 974–981. Nelson DL (1998). Molecular Basis of Mental Retardation: Fragile X-Syndrome. In: JB Martin (Ed.), Molecular Neurology. Scientific American, New York, pp. 19–35. Nitsche EM, Hiort O (2000). The molecular basis of androgen insensitivity. Horm Res 54: 327–333. Numaba K, Ito M, Uchiyama S (1990). A case of delayed radiation lumbosacral plexopathy. No To Shinkei 42: 629–633. Pan H, Li Y, Li T (2002). Increased (CTG/CAG) lengths in myotonic dystrophy type 1 and Machado–Joseph disease genes in idiopathic azoospermia patients. Hum Reprod 17: 1578–1583. Pectasides D, Pectasides M, Econoopoulos T (2006). Brain metastases from epithelial ovarian cancer: a review of the literature. Oncologist 11: 252–260. Qureshi T, Nasti AR, Ashai M (2003). Laurence, Moon, Bardet, Biedl syndrome. JK Practitioner 10: 217–218. Salvati M, Cervoni L (1994). Solitary metastasis from ovarian carcinoma: report of 4 cases. J Neurooncol 19: 75–77.

Sermon K, Lissens W, Jones S (1997). Clinical application of preimplanatation diagnosis for myotonic dystrophy. Prenat Diagn 17: 925–932. Shams’ili S, Beukelaar J, Willem Gartama J et al. (2006). An uncontrolled trial of rituximab for antibody associated paraneoplastic neurologic syndromes. J Neurol 253: 16–20. Shelbourne P, Davies J, Baxton J et al. (1993). Direct diagnosis of myotonic dystrophy with a disease-specific DNA marker. N Engl J Med 328: 471–475. Sherman SL (2000). Primary ovarian failure in the fragile xsyndrome. Am J Med Genet 3: 189–194. Sperfeld AD, Karitzky J, Brummer D et al. (2002). Xlinked bulbospinal neuronopathy. Arch Neurol 59: 1921–1926. Sullivan AK (2005). Association of FMR1 repeats size with ovarian dysfunction. Hum Reprod 20: 402–412. Tafur AJ, Baumann Kreuzinger LM, Quevedo F et al. (2008). 28-year-old man with severe vertigo. Mayo Clin Proc 83: 1070–1073. Tenner L, Einhorn L (2009). Ma-2 paraneoplastic encephalitis in the presence of bilateral testicular cancer. J Clin Oncol 27: e57–e58. Tuzun E, Zhou L, Baehring JM (2009). Evidence for antibodymediated pathogenesis in anti-NMDAR encephalitis associated with ovarian teratoma. Acta Neuropathol 118: 737–743. Voltz R, Gultekin SH, Rosenfeld MR et al. (1999). A serologic marker of paraneoplastic limbic and brainstem encephalitis in patients with testicular cancer. N Engl J Med 340: 1788–1795. Wattendorf DJ, Muenke M (2005). Diagnosis and management of fragile X-syndrome. Am Fam Physician 2005: 111–113. Welt CK, Smith PC, Taylor AE (2004). Evidence of early ovarian aging in fragile-X premutation carriers. J Clin Endocrinol Metab 9: 4569–4574. Wisniewski AB, Migeon CJ, Meyer-Bahlburg HF et al. (2000). Complete androgen insensitivity syndrome: long term medical, surgical and psychosexual outcome. J Clin Endocrinol Metab 85: 2664–2669. Yamamoto N, Yamada K, Hirata N et al. (1997). Electroejaculation and assisted reproductive techniques in patients with spinal cord injury. Nippon Heikatsukin Gakkai Zasshi 88: 420–426.

Handbook of Clinical Neurology, Vol. 120 (3rd series) Neurologic Aspects of Systemic Disease Part II Jose Biller and Jose M. Ferro, Editors © 2014 Elsevier B.V. All rights reserved

Chapter 53

Neurologic complications of multiple endocrine syndromes JORGE C. KATTAH1* AND WILLIAM C. KATTAH2 Department of Neurology, University of Illinois College of Medicine, Peoria, IL, USA

1

2

Endocrinology Department, University of the Andes, Bogota, Colombia

INTRODUCTION

Introduction

The neurologic manifestations of different endocrine syndromes may be classified according to specific hormone production changes and target tissue receptor response. Central nervous system (CNS) and peripheral nervous system (PNS) abnormalities are common manifestations of endocrine disorders. This chapter reviews the CNS and PNS manifestations of multiple endocrine syndromes (MES). MES account for a group of uncommon, familial disorders with polyglandular abnormalities, occurring in two main clinical contexts. The first involves autoimmune-mediated destruction of glandular tissue with decreased or absent hormone production: polyendocrine autoimmune syndromes (APS) acknowledge clinical phenotypes, type I and II involving adrenal insufficiency and type III sparing the adrenal glands. A second relates to familial oncogene derangements associated with multiple endocrine neoplasia (MEN), types 1 and 2. CNS and PNS abnormalities may be seen with both autoimmune and neoplastic MES and represent the subject of this chapter. Although the pathogenesis of the neurologic abnormalities associated with MES frequently involves endocrine and metabolic-mediated CNS and PNS alterations, independent autoimmune neurologic syndromes due to antineuronal antibodies may also be observed in the autoimmune polyglandular syndrome.

The endocrine characteristics define autoimmune polyglandular syndrome (APS) I. The pathogenesis involves antibody production against glandular surface cellular receptors, intracellular enzymes, and secreted proteins. APS I has no known HLA association. It is inherited as a sporadic, autosomal recessive (AR) trait. The genetic locus resides in the short arm of chromosome 21. A monogenic change of the AIRE-1 (autoimmune regulator-1) gene causes decreased immune tolerance leading to multiple endocrine glandular dysfunctions. The AIRE gene controls the expression of a tissuespecific substance in the thymus that controls autoimmune tolerance. Different mutations, deletions and insertions of the AIRE gene are responsible for the development of APS I (Balazs and Feher, 2009). The disorder is considered very uncommon. Given its AR inheritance, it is clustered in communities with high rates of consanguinity (Neufeld et al., 1981; Heino et al., 1999).

AUTOIMMUNE POLYGLANDULAR SYNDROME TYPE I (Table 53.1)

History Typical initial manifestations are observed among young children beginning from ages 3–5 years to early adolescence. The three cardinal manifestations include: mucocutaneous candidiasis; clinical manifestations consistent with hypoparathyroidism with tetany and encephalopathy in childhood or adolescence; Addison’s disease occurring in the first two decades of life, with encephalopathy, arterial hypotension, hypoglycemia, shock, and other manifestations of Addison’s disease. Additional multiple endocrinopathies may develop in

*Correspondence to: Jorge C. Kattah, M.D., Department of Neurology, University of Illinois College of Medicine at Peoria and the Illinois Neurological Institute at OSF Saint Francis Medical Center, 530 NE Glen Oak Avenue, Peoria, IL 61637, USA. E-mail: [email protected]

800

J.C. KATTAH AND W.C. KATTAH

Table 53.1 Polyendocrine autoimmune syndromes Type

Clinical manifestations

Genetics

Neurologic manifestations

1

Mucocutaneous candidasis Hypoparathyroidism Addison’s disease Addison’s disease Type 1 diabetes mellitus Autoimmune thyroid disease Normal adrenal function Autoimmune thyroiditis Type 1 diabetes mellitus

Autosomal recessive

Movement disorders Ataxia Tetany Related to diabetes mellitus and hypothyroidism

2

3

older patients with APS I. Addison’s disease may be categorized by age of onset (Carey, 1997). The association with recurrent candidiasis and absence of overt neurologic and neuro-ophthalmologic abnormalities is the best way to separate this syndrome from the childhood-onset, sex-linked variant of adrenoleukodystrophy (Moser et al., 2004).

Clinical findings APS I should be suspected in any patient with autoimmune Addison’s disease; genetic testing may be used for confirmation. Evidence of recurrent mucocutaneous candidiasis is a frequent finding in these patients and is present in the first year of life. The defining cardinal manifestations occur within the first 20 years of life. Hypocalcemia results from parathyroid hypoplasia or DiGeorge syndrome. Carpopedal spasm, paresthesias, seizures, laryngospasm, cataracts, papilledema, and encephalopathy may be observed. The electrocardiogram may show prolonged QT interval. Addison’s disease is usually the last endocrinopathy to occur in patients with APS I. Orthostatic intolerance, weakness, and fatigue are early signs. Additional features include hypogonadism, malabsorption, pernicious anemia, alopecia, and vitiligo. Late neurologic abnormalities include a single case report of a Miller Fisher syndrome with spontaneous recovery followed by progressive autoimmune spinocerebellar syndrome (Berger et al., 2008), a case of choreoathetosis and hemibalismus (Baumert et al., 1993), and a familial, limb girdle, muscular dystrophy in three sisters with APS I who had trabecular fibers in muscle biopsy, cataracts, epilepsy, and intracranial basal ganglia calcifications (Gazulla et al., 2005).

Polygenic

Autosomal dominant

Related to diabetes mellitus and hyopothyroidism

Natural history Laboratory investigations To confirm the autoimmune etiology of the endocrinopathies, an investigation for organ-specific antibodies is required. Autoantibodies against the parathyroid glands and/or the calcium sensor may be present. The 21-hydroxylase (OH) is the principal target antigen in cases of autoimmune Addison’s disease, found in 85% of patients with APS I (Kahaly, 2009). Specific AIRE1 genetic testing for symptomatic patients may confirm the clinical suspicion. Patients with APS I should be routinely monitored for the emergence of new endocrine and systemic autoimmune complications. Neurologic abnormalities may be potential manifestations of this syndrome, particularly in patients with circulating antibodies which may precede clinically detectable abnormalities. Common antibodies directed to specific neurotransmitters found in these patients include: aromatic L-amino acid decarboxylase antibodies (AADC), tyrosine hydroxilase (TH), tryptophan hydroxylase (TPH), glutamic acid decarboxylase (GAD), and g-aminobutyric acid (GABA). Antibodies against dopamine, serotonin, and noradrenaline may also be found (Fettisov et al., 2009). Although antibodies against enzymes involved in monoamine neurotransmitter synthesis are found in these patients, they do not always cause clinical manifestations.

Neuroimaging No specific cerebral imaging abnormalities are found in patients with APS I. Cerebral calcinosis may be found in some cases, probably related to hypoparathyroidism (Gass, 1962; Baba et al., 2005).

NEUROLOGIC COMPLICATIONS OF MULTIPLE ENDOCRINE SYNDROMES

Pathology Lymphocytic infiltration of affected endocrine glands is associated with functional loss of epithelial cells and scarring. A dominance of T-helper cells and deficiency of suppressor T cells has been demonstrated in endocrine autoimmunity. No specific report of neuropathologic findings among APS I have been reported to our knowledge (Kahaly, 2009).

Management The individual endocrine syndrome is managed as usual. Antifungal therapy is used as needed for recurrent candidiasis. Ketoconazole should be avoided, however, due to its potential effect on endocrine function. Calcium, vitamin D, and parathyroid hormone (PTH) are provided as needed. A major challenge is to anticipate the future development of additional endocrine failure. In general, antibodies against 21-hydroxylase (OH) represent an indication for treatment. Genetic counseling may be advisable.

AUTOIMMUNE POLYGLANDULAR SYNDROME TYPE II Introduction Also known as Schmidt’s syndrome, APS type II is characterized by the association of Addison’s disease, type 1 diabetes mellitus (DM), and autoimmune thyroid disease. Adrenal insufficiency is present in a high percentage of cases. APS II is more common in women and is far more frequent than APS I. The prevalence of APS II has been estimated at 1.4–2/100 000 and occurs most frequently in the third to fourth decades. Type 1 DM is one of the earliest manifestation of APS II. APS type II is inherited as an autosomal dominant (AD) trait with incomplete penetrance. HLA antigens and their IR genes are decisive in the pathogenesis of this disease (Balazs and Feher, 2010). HLA-DR3 and HLA-DR4 are more common in APS II. The pathogenesis of APS II is unknown. A familial defect of immunoregulation is likely (Dittmar and Kahaly, 2003).

History Clinical manifestations depend on the specific hormone deficiency. Diagnosis is suspected when there is a combination of type 1 DM, adrenal insufficiency, and autoimmune thyroiditis or Graves’ disease. Neurologic abnormalities may result from complications of DM, adrenal disease, or autoimmune thyroiditis. Graves’ disease and ophthalmopathy may occur. A paraneoplastic syndrome with neuropathy and ataxia was reported in a patient with APS II and small cell carcinoma of the lung (Watanabe et al., 2008). An APS II patient with low

801

serum vitamin B12 and antiparietal cell antibodies had fluctuating neurologic symptoms and a radiologic picture resembling multiple sclerosis (Hosseini et al., 1998). In large published series from the University of Gutemberg, concerning 15 000 patients with different endocrine disorders, 360 were diagnosed with APS II and 151 of them were followed for 13 years (Dittmar and Kahaly, 2003). Autoimmune thyroid disease was observed in 65% of cases. Autoimmune thyroiditis and Graves’ disease occurred with the same frequency. Type 1 DM occurred in 60% of cases. Addison’s disease was found in 18.5% and gonadal failure in 5.3% of cases. Nonendocrine manifestations included vitiligo, alopecia, and pernicious anemia. The interval between the different clinical APS II manifestations was variable, but usually no longer than 10 years. In the Gutemberg series, the presence of endocrine autoantibodies generally predicted eventual clinical manifestations of glandular failure.

Clinical findings The systemic and neurologic findings in these patients are related to the underlying single or multiple endocrinopathies present (Heuss et al., 1995). Besides APS-II, common causes of concurrent endocrine and neurologic disorders include the following: (1) multiple endocrine neoplasia; (2) autoimmune thyroid disease (Brain et al., 1966; Flanagan et al., 2010); (3) Graves’ disease; (4) mitochondrial disorders; (5) DM, diabetes insipidus, optic atrophy, and sensorineural deafness in cases of Wolfram’s syndrome; (6) polyglandular endocrine failure may also be seen in association with panhypopituitarism and hypothalamic disorders (Kattah and Kattah, 2008); (7) endocrinopathy associated with plasma cell dyscrasia and circulating cytokines (IL-1b), characterized clinically by organomegaly, adenopathy, monoclonal gammopathy syndrome and known by the acronym POEMS (Ghandi et al., 2007).

Natural history Most of the clinical manifestations of APS II are treated with hormonal replacement. Patients must be monitored for additional endocrine and other systemic autoimmune disorders.

Laboratory investigations An autoimmune etiology should be considered in any patient with type 1 DM, thyroid failure, Graves’ disease, or adrenal failure. Antibodies against thyroid peroxidase (TPO), thyroglobulin (TG), and thyroid stimulating immunoglobulin may be ordered for patients with clinical signs of thyroid dysfunction. Anti-GAD and antiinsulin antibodies may be requested to diagnose type 1

802

J.C. KATTAH AND W.C. KATTAH

DM. The 21-hydroxylase (OH), the 17-a-(OH) and the side chain cleavage enzyme antibodies should be requested to confirm autoimmune Addison’s disease. Genetic testing is not available as this is a polygenic disorder. HLA typing could be requested in selected cases.

Neuroimaging and pathology Neuroimaging or neuropathologic findings in APS II are not available to our knowledge. In general, the pathologic findings in the endocrine glands of these patients are similar in all APS syndromes and overlap with those found in single autoimmune endocrine syndromes. We report in this chapter typical clinical and brain MRI findings in a patient with APS II with acute limbic encephalitis (Fig. 53.1). Immunosuppression improved the clinical and imaging abnormalities.

Management Management of APS II involves hormonal replacement. Therapy for syndromic endocrine failure is similar to isolated endocrine failure. Normalization of adrenal failure must precede thyroid replacement when managing simultaneous adrenal and thyroid dysfunction. Life-threatening

adrenal failure may develop as a result of increased hepatic corticosteroid metabolism due to the replaced thyroid hormone (Majeroni and Patel, 2007).

AUTOIMMUNE POLYGLANDULAR SYNDROME TYPE III Introduction The characteristic phenotype of this syndrome includes polyglandular failure with autoimmune thyroiditis, type 1 DM, and other autoimmune disorders with absence of adrenal insufficiency. Autoimmune thyroiditis or Graves’ disease may occur, usually in the third decade of life. The precise pathogenesis of this disorder is unknown. APS III has been reported in monozygotic twins (Ugur et al., 2004). The HLA DRB-1 is found in this group of patients. The presumed pathogenesis involves a similar mechanism to APS II. The diagnosis of APS III followed treatment with interferon-a. (Sasso et al., 2003). This raises the possibility of viral-mediated mechanisms, molecular mimicry, or permanent autoimmunity. Adrenal failure would be distinctly uncommon in this syndrome in contrast to APS I and APS II. APS III is rare. No epidemiologic data are currently

Fig. 53.1. Axial MRI obtained on a patient with a long-standing history of polyglandular dysfunction. She was initially diagnosed as having Hashimoto’s thyroiditis in her twenties. This was followed a few years later by autoimmune adrenal failure. Positive antibodies against 21-hydroxylase were detected with a 27.3 U/mL serum titer (normal: 10% bone marrow lymphoplasmacytic infiltration with a characteristic immunophenotype (Rajkumar et al., 2006). Treatment for patients with WM is indicated if they have diseaseassociated anemia, thrombocytopenia, constitutional symptoms (weakness/fatigue, weight loss, night sweats), hyperviscosity, symptomatic cryoglobulinemia, significant hepatosplenomegaly, or lymphadenopathy.

Waldenstr€ om macroglobulinemia and peripheral neuropathy PN occurs in about one-third of patients with WM (Katirji and Koontz, 2012). Typically it is chronic, symmetric, predominantly sensory neuropathy, similar to IgM-MGUS

1093

PN. Anti-MAG antibodies can be found in about 50% of WM patients (Katirji and Koontz, 2012). Electrophysiologic studies demonstrate demyelination, similar to IgM-MGUS. Treatment is geared at the underlying disease process with some responses in neuropathy symptoms. WM can also produce cryoglobulinemia (described below), which may manifest with an immune-mediated vasculitis, leading to painful, distal symmetric sensorimotor neuropathy (Drappatz and Batchelor, 2004).

Waldenstr€om macroglobulinemia and the central nervous system HYPERVISCOSITY SYNDROME The IgM antibody that is secreted by the malignant lymphoplasmacytic cells is a multivalent molecule that has the tendency to aggregate, leading to slowing of the cerebral and retinal circulation resulting in symptoms due to transient ischemia (Drappatz and Batchelor, 2004). Symptoms usually occur when IgM levels are > 3 g/dL and/or serum viscosity is  4.0 centipoise (normal 1.4–1.8 centipoise), although this may vary among individual patients (Mehta and Singhal, 2003). The clinical triad of hyperviscosity syndrome includes neurologic symptoms, vision changes, and mucosal bleeding (Ghobrial et al., 2003). Neurologic symptoms include dizziness, headache, vertigo, ataxia, confusion, syncope, and stroke. Vision changes include blurry vision and diplopia with fundoscopic examination revealing papilledema, tortuosity of veins and thrombosis (Ghobrial et al., 2003). Mucosal bleeding in the oropharynx, gastrointestinal tract, ureter, or vagina can occur as the monoclonal IgM interferes with platelet function. Rarely patients can have retinal and intracranial hemorrhage. As a result of increased plasma volume patients may experience dyspnea, chest pain, pulmonary edema, or congestive heart failure. Treatment is aimed at emergently decreasing the IgM level in order to provide rapid relief of symptoms and avoid long-term complications. In severe situations this can be quickly achieved with aggressive intravenous hydration and plasmapheresis. Long-term management consists of systemic chemotherapy in order to decrease the IgM production. Much less frequently, MM can result in hyperviscosity syndrome. This can be seen when IgG levels are > 4 g/ dL and IgA levels > 6 g/dL along with increased serum viscosity (Mehta and Singhal, 2003).

BING–NEEL SYNDROME Bing–Neel syndrome is an extremely rare neurologic complication of WM and usually presents late in the disease course. It occurs when the neoplastic lymphoplasmacytic or plasma cells infiltrate the perivascular

1094

U. SOBOL AND P. STIFF

spaces, leptomeninges, and/or brain parenchyma in the CNS. These infiltrates can be found in other organs as well, including bone marrow, lymph nodes, spleen, and liver. The infiltrate may be diffuse in the CNS, causing symptoms of confusion and lethargy. Alternatively, the cells may coalesce into tumors with symptoms mainly from mass effect including paralysis and seizures (Malkani et al., 2009). Additional symptoms can include headache, blurry vision, psychiatric manifestations, numbness, paresthesias, hearing loss, and weakness. CSF analysis can show leukocytosis (WBC 100–500 cells/mm3), elevated total protein (>100 mg/ dL), normal or decreased glucose, and hypergammaglobulinemia with detectable M protein on protein electrophoresis and IgM confirmation on immunofixation of the CSF (Malkani et al., 2009). These CSF abnormalities are more common in diffuse rather than coalescent CNS disease. Brain imaging can be normal or show regions of enhancement on T2-weighted MRI images and occasionally tumoral or nodular masses on CT scans (Grewal et al., 2009). Treatment includes intrathecal chemotherapy and radiation therapy (focal or cariospinal radiation therapy),

along with systemic chemotherapy. Preliminary data suggest a role for intrathecal rituximab; however, additional studies are needed to evaluate dosing and potential toxicities before it can be recommended as a treatment option (Rubenstein et al., 2007). Due to the rarity of the disease, randomized controlled studies which establish the treatment recommendations in this disease have been difficult to do and will continue to be a challenge. Fortunately, with treatment, patients can have an improvement in their quality of life and remission can be achieved, with some patients having a sustained, long-lasting remission (Grewal et al., 2009).

AL AMYLOIDOSIS (IMMUNOGLOBULIN LIGHT CHAIN AMYLOIDOSIS) Amyloidosis refers to the deposition of insoluble fibrillar proteins in various tissues (Kyle et al., 2005). The deposited protein is detected by Congo red staining based on characteristic apple-green birefringence under polarized light (Gertz et al., 2005). Amyloidosis consists of several distinct types, based on the protein composition of the amyloid fibril, as seen in Table 73.7. Only one

Table 73.7 Classification of amyloidosis Type of amyloidosis

Precursor protein component

Symptoms

AL amyloidosis*

l or k immunoglobulin light chain (l is more common; l to k ratio, 3:1)

AA amyloidosis{

Serum amyloid A protein

Systemic: nephrotic syndrome, restrictive cardiomyopathy, neuropathy Localized: isolated organ involvement (e.g., carpal tunnel syndrome, isolated lesions in ureter, urethra, bladder, lung, bronchus or trachea) Renal presentation most common, associated with chronic inflammatory conditions

ATTR amyloidosis Mutant TTR{

Mutated transthyretin

Normal TTR{

Normal transthyretin

b2-microglobulin amyloidosis Ab amyloidosis Other hereditary amyloidosis A fibrinogen Lysozyme Apolipoprotein A-I

b2-microglobulin Ab protein precursor

Fibrinogen a-chain Lysozyme Apolipoprotein A-I

Hereditary; peripheral neuropathy and/or cardiomyopathy (commonly referred to as familial amyloid polyneuropathy) Restrictive cardiomyopathy; carpal tunnel syndrome (commonly referred to as senile amyloidosis) Carpal tunnel syndrome (associated with long-term dialysis) Alzheimer’s syndrome

Renal presentation (also called familial renal amyloidosis) Renal presentation most common Renal presentation most common

*Previously referred to as primary amyloidosis; the only amyloidosis secondary to a plasma cell disorder. { Previously referred to as secondary amyloidosis. { TTR, transthyretin (prealbumin). (Modified from Rajkumar et al., 2006.)

NEUROLOGIC ASPECTS OF PLASMA CELL DISORDERS form of amyloidosis is secondary to a clonal plasma cell disorder, AL amyloidosis (also referred to as immunoglobulin light chain amyloidosis and previously known as primary systemic amyloidosis). Diagnostic criteria for AL amyloidosis are listed in Table 73.8. Potential areas to biopsy for tissue confirmation of amyloid deposition include the affected organ (e.g., heart, kidney, peripheral nerve, rectum), bone marrow, or abdominal fat. In clinical practice the diagnosis can be quite challenging and often delayed given the vague constellation of symptoms, which mimic more common diseases. AL amyloidosis may be localized or systemic. Localized AL amyloidosis is often benign, can affect isolated organ systems (e.g., carpal tunnel syndrome) and treatment consists primarily of symptom relief. Systemic AL amyloidosis, on the other hand, can have profound multisystem involvement and requires treatment with chemotherapy and stem cell transplantation in transplant-eligible patients. Presentation may be variable depending on dominant organ involvement, with nephrotic syndrome, restrictive cardiomyopathy, peripheral and autonomic neuropathy being most common. Patients may also have macroglossia, carpal tunnel syndrome, organomegaly, weight loss, and periorbital and face purpura (“raccoon eyes sign”) (Rajkumar et al., 2006). AL amyloidosis can coexist with MM in 10% of patients but typically one of the two disorders dominates the clinical picture (Rajkumar et al., 2006). Improvement in organ dysfunction can be seen in about 50% of responding patients and prolonged organ remission can be achieved with stem cell transplantation (Rajkumar et al., 2006). Overall prognosis is poor but improving over the years thanks to the advances in therapy, stem cell transplantation and supportive care. Patients with confirmed amyloidosis need to be referred Table 73.8 Diagnostic criteria for AL amyloidosis Diagnosis (all four required)

Signs and symptoms

1. Amyloid related systemic syndrome* 2. Positive Congo red staining in any tissue 3. Light chain confirmation{ 4. Clonal plasma cell disorder{

Fatigue, weight loss, proteinuria (nephrotic range), CHF, hepatomegaly, peripheral and autonomic neuropathy

*Nephrotic syndrome, restrictive cardiomyopathy, hepatomegaly, malabsorption, peripheral or autonomic neuropathy (axonal neuropathy). { By direct examination of the amyloid using mass spectrometry. { M-protein, abnormal free light chain ratio, or clonal plasma cells. CHF, congestive heart failure. (Modified from Rajkumar, 2012.)

1095

to tertiary medical center for a thorough evaluation and appropriate management.

AL amyloidosis and peripheral neuropathy Amyloid neuropathy can be present in about 17–35% of patients with AL amyloidosis and is the presenting manifestation in 10% (Kyle and Gertz, 1995; Matsuda et al., 2011). It is a progressive, usually painful sensory polyneuropathy, with or without autonomic dysfunction. Neuropathy may be asymmetric, worse distally than proximal, with lower limbs being affected earlier than upper limbs (i.e., length-dependent neuropathy). Pain and temperature sensation are lost before light touch or vibratory sense and motor neuropathy tends to appear after sensory loss (Drappatz and Batchelor, 2004). Patients typically complain of burning, painful electrical sensations as well as symptoms of carpal tunnel syndrome, which may be present in up to 25% of patients and is due to amyloid deposition in the flexor retinaculum (Katirji and Koontz, 2012). Electrophysiologic studies show an axonal, sensory greater than motor neuropathy (Ramchandren and Lewis, 2012). Nerve biopsy can confirm the diagnosis by identifying endoneurial amyloid deposits. This direct toxin effect and amyloid-induced vascular insufficiency have been proposed as possible mechanisms in the pathogenesis of neuropathy. Unfortunately there is no clear evidence that treatment directed at the underlying plasma cell disorder improves symptoms of PN. Autonomic neuropathy is frequently present. It may present with symptoms due to orthostatic hypotension, impotence, bladder dysfunction, or gastrointestinal dysfunction. Symptomatic treatment with elastic stockings, fluorinated steroids, or dihydroergotamine may be helpful in patients with orthostatic hypotension.

AL amyloidosis and the central nervous system A subset of amyloid fibrils can deposit in the CNS; examples include amyloid b (Ab, responsible for some cases of Alzheimer’s disease), transthyretin (TTR), and British amyloid protein (BR12) (Lee and Picken, 2012). Common symptoms include cognitive decline, dementia, stroke, and less frequently, symptoms due to mass effect. AL amyloidosis is extremely rare in the CNS but has been reported in the literature with clinical manifestations related to amyloidoma and leptomeningeal cerebral amyloid angiopathy. CNS amyloidoma contains light chain amyloid which can form nodules or space-occupying lesions within the brain parenchyma or vertebral spinal axis (Tabatabai et al., 2005; Lee and Picken, 2012). CNS amyloidoma can occur in patients with or without evidence of

1096

U. SOBOL AND P. STIFF

systemic amyloidosis or plasma cell dyscrasia. Leptomeningeal cerebral amyloid angiopathy occurs when light chain amyloid fibrils deposit in small- and medium-sized blood vessels, similarly to the more common amyloid angiopathy in Alzheimer’s disease with amyloid b deposition (Lee and Picken, 2012). Focal intracranial hemorrhages have been reported as a complication of amyloid angiopathy. Unfortunately there is no effective treatment for this complication; supportive care and symptom relief are recommended.

CRYOGLOBULINEMIA Cryoglobulins are immunoglobulins that precipitate when cooled and dissolve when heated. Cryoglobulins are classified into three types: type I is monoclonal, type II is mixed (one monoclonal immunoglobulin the other polyclonal, most commonly associated with hepatitis C infection), and type III is strictly polyclonal (without monoclonal immunoglobulins). Type I cryoglobulinemia is most commonly of the IgM or IgG class and is associated with Waldenstr€ om macroglobulinemia, MM, or MGUS (Drappatz and Batchelor, 2004). It is estimated that 5–7% of patients with MM and 20% of patients with WM have an associated cryoglobulinemia (Bloch and Franklin, 1982). Patients with cryoglobulinemia can be asymptomatic or can present with Raynaud’s phenomenon, purpura, acrocyanosis, and skin ulceration. Other manifestations can include arthralgias, renal disease, and neuropathy. Treatment usually consists of rewarming and decreasing the immunoglobulin concentration by plasma exchange, with systemic chemotherapy if cryoglobulinemia is driven by underlying MM or WM (type I cryoglobulinemia). Therapy in type II should be directed at the underlying hepatitis C infection, if present.

Cryoglobulinemia and peripheral neuropathy Peripheral neuropathy has been reported in 17–56% of patients with cryoglobulinemia (Hoffman-Snyder and Smith, 2008). Neuropathy in type I (monoclonal) cryoglobulinemia is infrequent but a lot more common in type II and III cryoglobulinemia. Presentations of neuropathy can range from subacute mononeuritis multiplex to a chronic distal symmetric sensorimotor polyneuropathy, with sensory symptoms usually preceding motor dysfunction (Garcia-Bragado et al., 1988). Neuropathy is most often characterized by axonal degeneration with some reports of demyelination, either primary demyelination or secondary to axonal damage (Ropper and Gorson, 1998). Nerve biopsy can show epineurial vasculitis and epineurial cryoglobulin deposits (which have been described in MM and WM) (Vital

et al., 1991). Improvement in symptoms of neuropathy, even with treatment of the underlying cause, may be slow and very limited.

CONCLUSION Plasma cell disorders range from benign and indolent to malignant and aggressive disease processes. Clinical manifestations are variable and an array of neurologic complications can be present. Although the impact on the peripheral central nervous system is far more common, the clinical implications are typically greater with central nervous system involvement. Peripheral neuropathy is the most common complication and symptoms may vary from mild to debilitating, pure sensory to sensorimotor. Treatment of the underlying plasma cell disorder is often ineffective at controlling or improving neuropathy and, in fact, treatment of the underlying malignancy may cause or exacerbate the neuropathy. Symptomatic relief is necessary, though not always successful or adequate. Central nervous system involvement is rare, can have variable etiologies and symptoms, and usually carries a poor prognosis with limited treatment options. Recognition of the underlying plasma cell disorder is of great importance as it requires a proper diagnostic evaluation, surveillance, and treatment, if indicated.

REFERENCES Altekruse SF, Kosary CL, Krapcho M et al. (Eds.), (2007). SEER Cancer Statistics Review, 1975–2007. National Cancer Institute, Bethesda, MD, http://seer.cancer.gov/ csr/1975_2007/, based on November 2009 SEER data submission, posted to the SEER website, 2010. Arbogast SD, Khanna S, Koontz DW et al. (2007). Chronic ataxic neuropathy mimicking dorsal midbrain syndrome. J Neurol Neurosurg Psychiatry 78: 1276–1277. Barton DL, Wos EJ, Qin R et al. (2011). A double-blind, placebo-controlled trial of a topical treatment for chemotherapy-induced peripheral neuropathy: NCCTG trial N06CA. Support Care Cancer 19: 833–841. Benedetti L, Briani C, Franciotta D et al. (2008). Long-term effect of rituximab in anti-MAG polyneuropathy. Neurology 71: 1742–1744. Bloch KJ, Franklin E (1982). Plasma cell dyscrasias and cryoglobulins. JAMA 248: 2670–2676. Blume G, Pestronk A, Goodnough LT (1995). Anti-MAG antibody-associated polyneuropathies: improvement following immunotherapy with monthly plasma exchange and IV cyclophosphamide. Neurology 45: 1577–1580. Blumenthal DT, Glenn MJ (2002). Neurologic manifestations of hematologic disorders. Neurol Clin 20: 265–281. Bringhen S, Larocca A, Rossi D et al. (2010). Efficacy and safety of once-weekly bortezomib in multiple myeloma. Blood 116: 4745–4753. Byrne TN (1992). Spinal cord compression from epidural metastases. N Engl J Med 327: 614–649.

NEUROLOGIC ASPECTS OF PLASMA CELL DISORDERS Cavaletti G, Bogliun G, Marzorati L et al. (1994). Long-term peripheral neurotoxicity of cisplatin in patients with successfully treated epithelial ovarian cancer. Anticancer Res 14: 1287–1292. Cerase A, Tarantino A, Gozzetti A et al. (2008). Intracranial involvement in plasmacytomas and multiple myeloma: a pictorial essay. Neuroradiology 50: 665–674. Chaudhry V, Cornblath DR, Corse A et al. (2002). Thalidomide-induced neuropathy. Neurology 59: 877–878. Colak A, Cataltepe O, Erbengi A (1989). Spinal cord compression caused by plasmacytomas: a retrospective series of 14 cases. Neurosurg Rev 12: 305–308. Dalakas MC, Rakocevic G, Salajegheh M et al. (2009). Placebo-controlled trial of rituximab in IgM anti-myelinassociated glycoprotein antibody demyelinating neuropathy. Ann Neurol 65: 286–293. Dennis M, Chu P (2000). A case of meningeal myeloma presenting as obstructive hydrocephalus – a therapeutic challenge. Leuk Lymphoma 40: 219–220. Dimopoulos MA, Pouli A, Zervas K et al. (2003). Propsective randomized comparison of vincristine, doxorubicin and dexamethasone (VAD) administered as intravenous bolus injection and VAD with liposomal doxorubicin as first-line treatment in multiple myeloma. Ann Oncol 14: 1039–1044. Dimopoulos MA, Mateos MV, Richardson PG et al. (2011). Risk factors for, and reversibility of, peripheral neuropathy associated with bortezomib-melphalan-prednisone in newly diagnosed patients with multiple myeloma: subanalysis of the phase 3 VISTA study. Eur J Haematol 86: 23–31. Dispenzieri A (2012). POEMS syndrome: update on diagnosis, risk-stratification, and management. Am J Hematol 87: 805–814. Drappatz J, Batchelor T (2004). Neurologic compications of plasma cell disorders. Clin Lymphoma 5: 163–171. Eurelings M, Moons KGM, Notermans NC et al. (2001). Neuropathy and IgM M proteins: prognostic value of antibodies to MAG, SGPD, and sufatide. Neurology 56: 228–233. Garcia-Bragado F, Fernandez JM, Navarro C et al. (1988). Peripheral neuropathy in essential mixed cryoglobulinemia. Arch Neurol 45: 1210–1214. Gedlicka C, Scheithauer W, Schull B et al. (2002). Effective treatment of oxaliplatin-induced cumulative polyneuropathy with alpha-lipoic acid. J Clin Oncol 20: 3359–3361. Gertz MA (2006). Managing monoclonal gammopathyassociated neuropathy. Leuk Lymphoma 47: 785–786. Gertz MA, Lacy MQ, Dispenzieri A et al. (2005). Amyloidosis. Best Pract Res Clin Haematol 18: 709–727. Ghobrial IM, Gertz MA, Fonseca R (2003). Waldenstr€om macroglobulinaemia. Lancet Oncol 4: 679–685. Giannini F, Volpi N, Rossi S et al. (2003). Thalidomideinduced neuropathy: a gangliopathy? Neurology 60: 877–878. Grewal JS, Preetkanwal KB, Sahijdak WM et al. (2009). Bing– Neel syndrome: a case report and systematic review of clinical manifestations, diagnosis, and treatment options. Clin Lymphoma Myeloma 9: 462–466.

1097

Hadden RD, Nobile-Orazio E, Sommer CL et al. (2010). European Federation of Neurological Societies/ Peripheral Nerve Society Guideline on management of paraproteinemic demyelinating neuropathies. Report of a Joint Task Force of the European Federation of Neurological Societies and the Peripheral Nerve Society – first revision. J Peripher Nerv Syst 15: 185–195. Hoffman-Snyder C, Smith BE (2008). Neuromuscular disorders associated with paraproteinemia. Phys Med Rehabil Clin N Am 19: 61–79. Hogan MC, Lee A, Solberg LA et al. (2002). Unusual presentation of multiple myeloma with unilateral visual loss and numb chin syndrome in a young adult. Am J Hematol 70: 55–59. International Myeloma Working Group (2003). Criteria for the classification of monoclonal gammopathies, multiple myeloma and related disorders: a report of the International Myeloma Working Group. Br J Haematol 121: 749–757. Jagannath S, Barlogie B, Berenson J et al. (2004). A phase 2 study of two doses of bortezomib in relapse or refractory myeloma. Br J Haematol 127: 165–172. Katirji B, Koontz D (2012). Disorders of peripheral nerves. In: RB Daroff, GM Fenichel, J Jankovic et al. (Eds.), Bradley’s Neurology in Clinical Practice. Vol. II. Elsevier, Philadelphia, pp. 1971–1976. Kelly JJ, Kyle RA, O’Brien PC et al. (1981). Prevalence of monoclonal protein in peripheral neuropathy. Neurology 31: 1480–1483. Kyle RA, Gertz MA (1995). Primary systemic amyloidosis: clinical and laboratory features in 474 cases. Semin Hematol 32: 45–49. Kyle RA, Rajkumar SV (2009). Treatment of multiple myeloma: a comprehensive review. Clin Lymphoma Myeloma 9: 278–288. Kyle RA, Therneau TM, Rajkumar SV et al. (2002). A long term study of prognosis in monoclonal gammopathy of undetermined significance. N Engl J Med 346: 564–569. Kyle RA, Gertz MA, Witzig TE et al. (2003). Review of 1027 patients with newly diagnosed multiple myeloma. Mayo Clin Proc 78: 21–33. Kyle RA, Kelly JJ, Dyck PJ (2005). Amyloidosis and neuropathy. In: PJ Dyck, PK Thomas (Eds.), Peripheral Neuropathy. 4th edn. Elsevier Saunders, Philadelphia, pp. 2427–2451. Kyle RA, Therneau TM, Rajkumar SV et al. (2006). Prevelance of monoclonal gammopathy of undetermined significance. N Engl J Med 354: 1362–1369. Kyle RA, Remstein ED, Therneau TM et al. (2007). Clinical course and prognosis of smoldering (asymptomatic) multiple myeloma. N Engl J Med 356: 2582–2590. Landowski TH, Megli CJ, Nullmeyer KD et al. (2005). Mitochondrial-mediated disregulation of Ca2 þ is a critical determinant of Velcade (PS-34/bortezomib) cytotoxicity in myeloma cell lines. Cancer Res 65: 3828–3836. Lee JM, Picken MM (2012). Cerebrovascular amyloidosis. In: MM Picken, A Dogan, GA Herrera (Eds.), Amyloid and Related Disorders, Surgical Pathology and Clinical Correlations. Springer Science þ Business Media, New York, pp. 106–109.

1098

U. SOBOL AND P. STIFF

Leger J-M, Viala K, Bombelli F et al. (2010). For the RIMAG trial group (France and Switzerland): randomized controlled trial of rituximab in demyelinating neuropathy associated with anti-MAG IgM gammopathy (RIMAG study). J Peripher Nerv Syst 15: 269. Lunn MP, Nobile-Orazio E (2006). Immunotherapy for IgM anti-myelin associated glycoprotein paraprotein associated neuropathy. Cochrane Database Syst Rev (2): CD002827. Malkani RG, Tallman M, Gottardi-Littell N et al. (2009). Bing–Neel syndrome: an illustrative case and a comprehensive review of the published literature. J Neurooncol 96: 301–312. Maranzano E, Latini P (1995). Effectiveness of radiation therapy without surgery in metastatic spinal cord compression: final results from a prospective trial. Int J Radiat Oncol Biol Phys 32: 959–967. Matsuda M, Gono T, Morita H et al. (2011). Peripheral nerve involvement in primary systemic AL amyloidosis. Eur J Neurol 18: 604–610. McLaughlin DM, Gray WJ, Jones FGC et al. (2004). Plasmacytoma: an unusual cause of a pituitary mass lesion. A case report and review of the literature. Pituitary 7: 179–181. Mehta J, Singhal S (2003). Hyperviscosity syndrome in plasma cell dyscrasias. Semin Thromb Hemost 29: 467–471. Mendez CE, Hwang BJ, Destian S et al. (2010). Intracranial multifocal dural involvement in multiple myeloma: case report and review of the literature. Clin Lymphoma Myeloma Leuk 10: 220–223. Moreau P, Pylypenko H, Grosicki S et al. (2011). Subcuteous versus intravenous administration of bortezomib in patients with relapsed multiple myeloma: a randomised-phase 3 non-inferiority study. Lancet Oncol 12: 431–440. Morgan GJ, Davies FE, Gregory WM et al. (2010). First-line treatment with zoledronic acid as compared with clodronic acid in multiple myeloma (MRC Myeloma IX): a randomised controlled trial. Lancet 376: 1989–1999. Mundy GR (1998). Myeloma bone disease. Eur J Cancer 34: 246–251. Notermans NC, Lokhorst HM, Franssen H et al. (1996). Intermittent cyclophosphamide and prednisone treatment or polyneuropathy associated with monoclonal gammopathy of undetermined significance. Neurology 47: 1227–1233. O’Connor OA, Stewart AK, Vallone M et al. (2009). A phase I dose escalation study of the safety and pharmacokinetics of the novel proteasome inhibitor carfilzomib (PR-171) in patients with hematologic malignancies. Clin Cancer Res 15: 7085–7091. Orlowski RZ, Kuhn DJ (2008). Proteasome inhibitors in cancer therapy: lessons from the first decade. Clin Cancer Res 14: 1649–1657. Poruchynsky MS, Sackett DL, Robey RW et al. (2008). Proteasome inhibitors increase tubulin polymerization and stabilization in tissue culture cells: a possible mechanism contributing to peripheral neuropathy and cellular

toxicity following proteasome inhibition. Cell Cycle 7: 940–949. Rajabally YA (2011). Neuropathy and paraproteins: review of a complex association. Eur J Neurol 18: 1291–1298. Rajkumar SV (2012). Multiple myeloma : 2012 update on diagnosis, risk-stratification, and management. Am J Hematol 87: 78–88. Rajkumar SV, Dispenzieri A, Kyle RA (2006). Monoclonal gammopathy of undetermined significance, Waldenstr€ om macroglobulinemia, AL amyloidosis, and related plasma cell disorders: diagnosis and treatment. Mayo Clin Proc 81: 693–703. Ramchandren S, Lewis RA (2012). An update on monoclonal gammopathy and neuropathy. Curr Neurol Neurosci Rep 12: 102–110. Richardson PG, Sonneveld P, Schuster MW et al. (2009a). Reversibility of symptomatic peripheral neuropathy with bortezomib in the phase III APEX trial in relapsed multiple myeloma: impact of a dose-modification guideline. Br J Haematol 144: 895–903. Richardson P, Hofmeister C, Jakubowiak A et al. (2009b). Phase 1 clinical trial of the novel structure proteasome inhibitor NPI-0052 in patients with relapsed and relapsed/refractory multiple myeloma. Blood (ASH Annual Meeting Abstracts), 114. Abstract 431. Richardson PG, Delforge M, Beksac M et al. (2012). Management of treatment-emergent peripheral neuropathy in multiple myeloma. Leukemia 26: 595–608. Ritz MF, Erne B, Ferracin F et al. (1999). Anti MAG IgM penetration into myelinated fibers correlates with the extent of myelin widening. Muscle Nerve 22: 1030–1037. Ropper AH, Gorson KC (1998). Neuropathies associated with paraproteinemia. N Engl J Med 338: 1601–1607. Rubenstein JL, Fridlyand J, Abrey L et al. (2007). Phase I study of intraventricular administration of rituximab in patients with recurrent CNS and intraocular lymphoma. J Clin Oncol 25: 1350–1356. Sadiq SA, Thomas FP, Kilidireas K et al. (1990). The spectrum of neurologic disease associated with anti-DM1 antibodies. Neurology 40: 1067–1072. Siegel DS, Martin T, Wang M et al. (2012). A phase 2 study of single-agent carfilzomib (PX-171-003-A1) in patients with relapsed and refractory multiple myeloma. Blood 120: 2817–2825. Silberman J, Lonial S (2008). Review of peripheral neuropathy in plasma cell disorders. Hematol Oncol 26: 55–65. Simmons Z, Tivakaran S (1996). Acquired demyelinating polyneuropathy presenting as a clinical sensory syndrome. Muscle Nerve 19: 1174–1176. Simmons Z, Albers JW, Bromberg M et al. (1995). Long-term follow-up of patients with chronic inflammatory demyelinating polyradiculoneuropathy, without and with monoclonal gammopathy. Brain 118: 359–368. Sonneveld P, Jongen JLM (2010). Dealing with neuropathy in plasma-cell dyscrasias. Hematology 423–430. Tabatabai G, Boehring J, Hochberg FH (2005). Primary amyloidoma of the brain parenchyma. Arch Neurol 62: 477–480.

NEUROLOGIC ASPECTS OF PLASMA CELL DISORDERS Vital A (2001). Paraproteinemic neuropathies. Brain Pathol 11: 399–407. Vital A, Vital C, Ragnaud JM et al. (1991). IgM cryoglobulin deposits in the peripheral nerve. Virchows Arch A Pathol Anat Histopathol 418: 83–85. Vrethem M, Cruz M, Wen Xin H et al. (1993). Clinical neurophysiological, and immunological evidence of polyneuropathy in patients with monoclonal gammopathies. J Neurol Sci 114: 193–199.

1099

Willison HJ, O’Leary CP, Veitch J et al. (2001). The clinical and laboratory features of chronic sensory ataxic neuropathy with anti-disialosyl IgM antibodies. Brain 124: 1968–1977. Yeung KB, Thomas PK, King RHM (1991). The clinical spectrum of peripheral neuropathies associated with benign monoclonal IgM, IgG and IgA paraproteinemia: comparative clinical immunological and nerve biopsy findings. J Neurol 238: 383–391.

Handbook of Clinical Neurology, Vol. 120 (3rd series) Neurologic Aspects of Systemic Disease Part II Jose Biller and Jose M. Ferro, Editors © 2014 Elsevier B.V. All rights reserved

Chapter 74

Neurologic manifestations of Henoch–Scho¨nlein purpura MAXIME D. BE´RUBE´1, NORMAND BLAIS2, AND SYLVAIN LANTHIER1* Department of Neurology, Centre Hospitalier de l’Universit de Montral, and Faculty of Medicine, Universit de Montral, Montreal, QC, Canada

1

2

Department of Haematology, Centre Hospitalier de l’Universit de Montral, and Faculty of Medicine, Universit de Montral, Montreal, QC, Canada

Henoch–Scho¨nlein purpura (HSP) is a systemic small vessel vasculitis seen predominantly in children. It usually affects the skin, joints, gastrointestinal tract, and kidneys. While often underappreciated, neurologic complications may occur in one of every 14 patients with the disease. Up to 20% of these patients are left with persistent deficits, highlighting the importance of early recognition and treatment.

GENERAL ASPECTS Historical aspects The first description of the disease is generally credited to the English physician William Heberden, who published his princeps observation in 1801. It was Johann Lukas Scho¨nlein, though, who established the association of joint symptoms and rash as a distinct clinical entity, which he named “peliosis rheumatica.” Abdominal pain and nephritis were later recognized as part of the clinical picture by his former student, Eduard Heinrich Henoch. In 1914, Sir William Osler described the first case of neurologic involvement (Osler, 1914). In the same paper, he also made the initial suggestion that the disease might be caused by an allergic or immunologic derangement (Gairdner, 1948; Roberts et al., 2007). This hypothesis would be confirmed decades later with the demonstration of the essential pathologic feature of the disease, vascular IgA deposition (Urizar et al., 1968).

Systemic disease HSP is largely a disease of childhood, with over 75% of patients having their onset before age 10 (Gedalia, 2004).

Estimates of its annual incidence range from 13.5 to 20.4 per 100 000 children. While rarely affected, adults often have a more severe disease course, especially regarding renal involvement. The male to female ratio is approximately 1.5 to 1. The distribution of vasculitic involvement gives rise to the characteristic association of purpuric rash, arthralgias, abdominal pain, and nephritis. While these manifestations may appear in any order, purpura is usually the first, and its presence at some point in the evolution is mandatory for diagnosis (Ozen et al., 2010). It consists of small (2–10 mm in diameter), palpable, nonblanching lesions found predominantly over buttocks, ankles, and extensor surfaces. Arthralgias affect close to 75% of patients and involve especially the large joints such as knees and ankles (Saulsbury, 2007). Gastrointestinal involvement takes the form of colicky abdominal pain in 60% of patients. It is usually moderate in intensity but occasionally so severe as to mimic an acute abdomen. It may be complicated by bleeding in 30% and, rarely, intussusception. Renal impairment remains by far the main contributor to the long-term morbidity and mortality of this disease (Fervenza, 2003). It is seldom present at onset and may even be delayed for weeks. Its earliest manifestation is usually microscopic hematuria, which can eventually be accompanied by proteinuria in two-thirds of patients (Saulsbury, 1999). The risk of end-stage renal failure in an unselected patient population is around 1–3%, but rises to 50% in those with a nephritic-nephrotic syndrome. Other infrequent complications that have been described include carditis, pulmonary hemorrhage, pancreatitis, cholecystitis, pseudomembranous colitis, orchitis, and urethritis (Gedalia, 2004; Saulsbury, 2007).

*Correspondence to: Sylvain Lanthier, Department of Neurology, Centre Hospitalier de l’Universite´ de Montre´al, CHUM - Hoˆpital Notre-Dame, 1560 est Sherbrooke, Suite GR-1159, Montre´al (Que´), H2L 4 M1, Canada. Tel: þ1-514-890-8000  26268, E-mail: [email protected]

M.D. BE´RUBE´ ET AL.

1102

Diagnostic criteria The first set of formal diagnostic criteria established by the American College of Rheumatology in 1990 required the patient to meet at least two of the following criteria: (1) palpable purpura, not related to thrombocytopenia; (2) age 20 years or younger; (3) abdominal pain usually with gastrointestinal bleeding; (4) biopsy showing granulocytes in the vessel wall (arterioles or venules) (Mills et al., 1990). These criteria yielded a sensitivity of 87.1% and a specificity of 87.7%. However, they did not account for occurrence of the disease in adults or require demonstration of the signature presence of IgA deposits. These issues were addressed in a new set of criteria proposed by the European League Against Rheumatism (EULAR), Paediatric Rheumatology International Trials Organization (PRINTO), and Paediatric Rheumatology European Society (PRES) consensus conference (see Table 74.1) (Ozen et al., 2010). These criteria have now been validated, and were found to have a sensitivity and specificity of 100% and 87%, respectively (Ozen et al., 2010).

Table 74.1 EULAR/PRINTO/PRES diagnostic criteria for Henoch–Scho¨nlein purpura (Ozen et al., 2010) Criterion

Glossary

Purpura (mandatory criterion)

Purpura (commonly palpable and in crops) or petechiae, with lower limb predominance,* not related to thrombocytopenia Diffuse abdominal colicky pain with acute onset assessed by history and physical examination. May include intussusception and gastrointestinal bleeding Typically leukocytoclastic vasculitis with predominant IgA deposit or proliferative glomerulonephritis with predominant IgA deposit Arthritis of acute onset defined as joint swelling or joint pain with limitation on motion Arthralgia of acute onset defined as joint pain without joint swelling or limitation on motion Proteinuria > 0.3 g/24 h or > 30 mmol/mg of urine albumin/creatinine ratio on a spot morning sample Hematuria or red blood cell casts: >5 red blood cells/high power field or red blood cells casts in the urinary sediment or  2 þ on dipstick Purpura or petechiae (mandatory) with lower limb predominance* and at least one of the four following criteria: Abdominal pain Histopathology Arthritis or arthralgia Renal involvement

1. Abdominal pain

2. Histopathology

3. Arthritis or arthralgias

Pathology HSP is classified according to the modified Chapel Hill Consensus Criteria in the group of nongranulomatous small vessel vasculitides. In the skin, the histopathologic pattern is that of a nonspecific leukocytoclastic vasculitis characterized by necrosis of the vessel wall and infiltration of inflammatory cells (mostly polymorphonuclear and mononuclear cells) within the capillaries and postcapillary venules (Mills et al., 1990). Delineation of the disease requires demonstration of perivascular IgA deposition, which can be seen by immunofluorescence along with C3, fibrin, and occasional IgG or IgM (Davin et al., 2001). In the kidney, the main site of IgA deposition is the mesangium, where it forms granular deposits. Renal lesions are diverse in type and severity, but in children are most commonly consistent with a focal and segmental glomerulonephritis (Pillebout and Niaudet, 2008).

4. Renal involvement

HSP EULAR/ PRINTO/PRES Ankara 2008 classification definition

*For purpura with atypical distribution, demonstration of an IgA deposit in a biopsy is required.

Etiology and pathogenesis As in most primary vasculitides, the inciting event in HSP remains unknown. Its predilection for onset in autumn and winter months and the presence of preceding respiratory infections in many cases has led some to suspect a role for infectious agents, although no single pathogen has been consistently identified (Yang et al., 2008). Evidence of group A b-hemolytic streptococcus infection can be found in 20–50% of HSP children. A host of “secondary causes” including exposure to medications such as L-dopa or chlorpromazine and medical conditions such as cancer have been linked to HSP, but in many cases their

relationship remains uncertain (Aram, 1987; Niedermaier and Briner, 1997; Saulsbury, 1999). Variations in ethnic and geographic distribution also occur and may be related to genetic or environmental specificities of affected populations. For instance, skewing towards Caucasian ethnicity has been reported in North American children (Allen et al., 1960). Certain HLA serotypes, familial Mediterranean fever gene mutations, and specific gene polymorphisms have been suggested to modulate the susceptibility to and clinical expression of the disease (Saulsbury, 1999; Brogan, 2007).

¨ NLEIN PURPURA NEUROLOGIC MANIFESTATIONS OF HENOCH–SCHO 1103 Many lines of evidence point to a pivotal role for those with neurologic manifestations, there is a 1.5:1 prehumoral immunity, specifically IgA, in the pathophysiolponderance of male patients, similar to that of the genogy of HSP. In addition to IgA deposition in the vessel eral HSP population. While no systematic data are walls and mesangium, patients also have evidence of available, a seemingly disproportionate number of neuincreased serum levels of IgA, circulating IgArologic cases have been reported in adults (data from containing immune complexes, and IgA-producing B Garzoni et al., 2009). This is in line with the previous sugcells (Davin et al., 2001; Pillebout and Niaudet, 2008; gestion that HSP has a more severe expression in this age Yang et al., 2008). Immunoglobulin deposits in HSP group. have been found to be composed exclusively of one of the two subclasses of IgA, IgA1. IgA1 differs from NERVOUS SYSTEM MANIFESTATIONS IgA2 by the presence of a hinge region containing Neurologic manifestations often occur in patients with N-acetylgalactosamine glycosylation sites. Much attenan unusually severe disease course, comprising frequent tion has been focused on abnormalities in the galactosyrenal impairment and unusual multiorgan involvement lation and sialylation of these sites in the genesis of both (Garzoni et al., 2009). When evaluating these complex HSP nephritis and the closely related IgA nephropathy patients, it is useful to think in terms of the possible (Davin et al., 2001). However, one study found that pathophysiologic mechanisms that may be responsible these abnormalities were restricted to HSP patients of CNS damage, either directly from extension of with nephritis, questioning their role in the genesis of the vasculitis to the CNS or indirectly from systemic the leukocytoclastic vasculitis (Allen et al., 1998). involvement. Antibodies may mediate some of their effect through complement activation. IgA have a known ability to actiCerebrovascular disease vate the indirect and lectin pathways, and C3 deposits can be seen in biopsy specimens. This, however, conVasculitis can extend to cerebral vessels and cause edema, ischemia, infarction and hemorrhage. Pathologtrasts with the generally normal serum complement ically confirmed cases of CNS vasculitis are scarce, howlevels (Saulsbury, 2001; Chen et al., 2010). Some of these antibodies have been shown to directly induce secretion ever (Gairdner, 1948; Allen et al., 1960; Gilbert and Da of IL-8 by the endothelial cells that they bind, stimulating Silva, 1966). Foci of vascular fibrinoid necrosis associrecruitment of polymorphonuclear cells and inflammaated with diffuse cortical petechiae and ischemic neurotion (Yang et al., 2008). nal changes were described in the brain of an 8-year-old boy with a severe course of HSP (de Montis and Turpin, 1971; Laplane et al., 1973). In another case, IgA deposiEPIDEMIOLOGY OF NERVOUS SYSTEM tion could be demonstrated in the cerebral vessels of a MANIFESTATIONS patient with intracerebral hematoma, providing some Although severe neurologic involvement in HSP is evidence of the identity of the vasculitic process in the uncommon, milder manifestations may be underapbrain and other vascular beds (Murakami et al., 2008). preciated. In one series, seizures or confusion were Nonetheless, most reports of cerebral vasculitis rely retrospectively identified in 17 of 244 cases (6.9%) of mainly on imaging findings deemed suggestive thereof. HSP. More recent studies tend to report lower inciOccipital and posterior parietal lobes are preferentially dences. For instance, in a review of 100 patients examinvolved, with a predominance of brain edema and ined over the course of 20 years, Saulsbury (1999) infarct in these regions. Frontal, temporal, and cerebellar described only two cases of seizures. Similarly, involvement is rarer. This peculiar topography has led Trapani et al. (2005) found a 3% incidence of headache some authors to speculate that hemodynamic characterbut no other neurologic manifestations in 150 patients istics of the posterior circulation might favor preferenstudied over 5 years. Other large series make no mention tial IgA deposition in these vessels (Paolini et al., of neurologic symptoms among their patients (Fischer 2003). Brainstem is notably spared but at least one et al., 1990; Sticca et al., 1999; Calvino et al., 2001; patient was described who presented with an isolated Peru et al., 2008). Whether these discrepancies are due lesion of the medulla oblongata that regressed under corto selection bias, improvement in supportive care or ticotherapy (Bulun et al., 2001). The vasculitic process underreporting is hard to ascertain. The latter may be can progress to hemorrhage, with necrosis of the vessel particularly important in the case of headache and wall leading to rupture and extravasation (Paolini et al., subtle behavioral changes which may easily be obscured 2003). Coincident zones of putative vasculitic activity by or attributed to other manifestations of the disease. and cerebral hemorrhage have been demonstrated in In one report, these features were present in a third one case on MRI (Wen et al., 2005). Most strikingly, of HSP patients (Ostergaard and Storm, 1991). Among hemorrhages share the same predominantly posterior

1104

M.D. BE´RUBE´ ET AL.

distribution as vasculitis. They may be life-threatening, and evacuating surgery had to be performed in four of 11 reported cases (Scattarella et al., 1983; Clark and Fitzgerald, 1985; Altinors and Cepoglu, 1991; Ng et al., 1996; Chiaretti et al., 2002; Imai et al., 2002; Paolini et al., 2003; Misra et al., 2004; Wen et al., 2005; Karamadoukis et al., 2008; Murakami et al., 2008). Subarachnoid bleeding has also been reported as an infrequent complication, mostly in the early series and on the basis of hemorrhagic CSF (Belman et al., 1985). A small aneurysm was found in one case of cerebellar hemorrhage, which was not felt to be related to vasculitis (Paolini et al., 2003). Subdural hematomas have also been described, including two children who needed surgical drainage (Belman et al., 1985). One patient suffered from an extensive venous thrombosis extending to the superior sagittal, straight and transverse sinuses (Abend et al., 2007). His prothombotic workup revealed the presence of a lupus anticoagulant. Focal neurologic signs occur in 26–32% of patients with neurologic involvement and, in most cases, they are caused by cerebrovascular disease (Belman et al., 1985; Garzoni et al., 2009). Among 79 HSP patients with neurologic manifestations, focal deficits were hemiparesis (n ¼ 11; 14%), aphasia (n ¼ 6; 8%), cortical blindness (n ¼ 4; 5%), ataxia (n ¼ 2; 3%), as well as paraparesis, quadriplegia, and chorea (n ¼ 1 each; 1%) (Belman et al., 1985). Nystagmus, dysarthria, and hemianesthesia were also reported. One case of cerebellar mutism occurred following drainage of a posterior fossa hemorrhage (Paolini et al., 2003).

Hypertensive encephalopathy and posterior reversible encephalopathy syndrome Hypertension has long been suspected as a cause of neurologic dysfunction in encephalopathic HSP patients (Belman et al., 1985). Posterior reversible encephalopathy syndrome (PRES) cases have provided documented imaging evidence of this association. PRES is a clinical-radiologic syndrome characterized by the occurrence of headache, visual dysfunction, seizures, and altered consciousness in association with reversible, posteriorly predominant white matter hyperintensities (Hinchey et al., 1996). These hyperintensities are thought to represent vasogenic edema caused by disruption of blood–brain barrier integrity. Although its pathophysiology is complex, loss of cerebral autoregulation from uncontrolled hypertension, volume overload, and endothelial injury from a variety of systemic conditions (including inflammatory disorders, sepsis, or exposure to certain drugs) are believed to be paramount (Bartynski, 2008). Patients with vasculitis often present a combination of these risk factors, making them

especially susceptible (Min et al., 2006). Four HSP patients were described that presented with the typical clinical course of PRES (Woolfenden et al., 1998; Ozcakar et al., 2004; Sasayama et al., 2007; Salloum et al., 2009). Cortical blindness accounts for 5.1% of all focal signs associated with HSP, and may be explained by PRES in many cases (Belman et al., 1985). Neither the clinical nor the imaging features of PRES are specific, however, and it may be difficult to distinguish these features from those of vasculitic lesions, especially when no clear predisposing factor is present. In fact, among the four cases that were published, only two had hypertension and only one had renal dysfunction. In addition, some authors have pointed out that cases published as “cerebral vasculitis” had an evolution that closely resembled PRES (Woolfenden et al., 1998). Both conditions seemingly affect the same brain regions. Diffusion-weighted imaging (DWI) and apparent diffusion coefficient (ADC) have been advocated as means to distinguish vasogenic edema from ischemic lesions of vasculitis (Mak et al., 2008). Theoretically, ADC is increased in the former and diminished in the latter. However, many vasculitic lesions do not progress to irreversible ischemia and their DWI and ADC signals may sometimes be consistent with vasogenic edema (Moritani et al., 2001, 2004). Presence of inflammatory changes in the CSF has also been proposed as an additional distinguishing feature (Min et al., 2006). However, while data on CSF parameters in these two conditions are scant, it has frequently been reported as normal or noninflammatory in both, and may not permit adequate discrimination. Further compounding the problem is that the two processes may in fact coexist. Absence of hypertension or renal injury in these and other cases suggests that direct vasculitic involvement of cerebral vessels may indeed play a direct causal role in PRES (Bartynski, 2008; Nishio et al., 2008). CSF pleocytosis in one case of PRES was interpreted as indirect evidence of concomitant CNS inflammation (Woolfenden et al., 1998).

Disorders of hemostasis Brain lesions that result from small vessel CNS vasculitis are frequently reversible, and most patients recover. Nonetheless, vasculitis may be prone to thrombotic complications and thrombosis may be associated with coexistent prothrombotic conditions. A study of 28 HSP and 79 healthy children suggests that markers of hypercoagulability such as fibrinogen, D-dimer, thrombinantithrombin complex (TAT), prothrombin fragments 1 þ 2 (PF1 þ 2), von Willebrand antigen and activity are significantly elevated in the acute phase of HSP compared with the recovery phase of HSP or in controls (Yilmaz et al., 2006). Transient synthesis of

¨ NLEIN PURPURA NEUROLOGIC MANIFESTATIONS OF HENOCH–SCHO antiphosphatidyl-ethanolamine antibodies was documented in a 17-year-old girl with extensive periopercular ischemic stroke (Sokol et al., 2000). Antiphospholipid antibodies were also uncovered in the one documented case of venous sinus thrombosis (Abend et al., 2007). Antiphospholipid antibodies have been described in association with HSP before, with or without thrombosis. Their relationship remains uncertain, but some have hypothesized that endothelial disruption by IgA antibodies may expose hidden phospholipids and promote synthesis of thrombogenic antiphospholipid antibodies (Abend et al., 2007). Low levels of factor XIII were found in two patients with intracerebral hemorrhage (ICH) (Imai et al., 2002; Murakami et al., 2008). Factor XIII is an essential hemostasis enzyme that serves to polymerize fibrin. Patients with an inherited deficiency, who usually have serum levels of less than 1%, are prone to severe bleeding complications including spontaneous ICH (Lorand et al., 1980; Hsieh and Nugent, 2008). A somewhat controversial state of acquired deficiency, with levels generally between 50% and 75%, has been described in many inflammatory disorders including HSP (Henriksson et al., 1977). Some authors speculate that such relative deficiency may enhance bleeding in the presence of a pre-existing diathesis such as vasculitis. Administration of factor XIII concentrate in patients with HSP has been suggested to correct the bleeding diathesis (Imai et al., 2002; Prenzel et al., 2006). Another patient with ICH was found to have prolonged prothrombin and partial thromboplastin time suggesting an associated disseminated intravascular coagulation process (Clark and Fitzgerald, 1985). In this regard, mild decreases in factor XIII is more likely a reflection of a mild consumptive coagulopathy (Zajadacz and Juszkiewicz, 2005).

Metabolic abnormalities and infections The multisystemic nature of HSP exposes patients to many potential disturbances of homeostasis, which may contribute to neurologic dysfunction. In addition to hypertension, renal impairment may lead to uremia, electrolyte disturbances, and volume overload. In two series, hyponatremia was a frequent accompaniment of neurologic symptoms, especially seizures (de Montis and Turpin, 1971; Laplane et al., 1973). Superimposed infections may add to the diagnostic confusion. CMV reactivation was thought to contribute to cerebral vasculitis in one case and concomitant Staphylococcus aureus infection was detected in another (Murakami et al., 2008; Temkiatvises et al., 2008). Corticosteroid or cyclophosphamide use may be responsible for opportunistic infections, PRES, and disturbances of electrolytic balance.

1105

Headache and mental status alterations Headache and mild behavioral alterations are frequently overlooked, and may represent telltale signs of subtle CNS involvement. In the only prospective study documenting neurologic manifestations of HSP, headache accompanied by behavioral changes was found in 31% of patients. Their presence was significantly associated with EEG changes (Ostergaard and Storm, 1991). Severe depression of consciousness may also occur, usually in association with a postictal state, hypertension, hemorrhage, or marked metabolic alterations.

Seizures Seizures occur in 53% of patients with neurologic manifestations, mostly during the acute phase and occasionally as the initial manifestation of HSP (Belman et al., 1985; Mannenbach et al., 2009). Of these, 17% have partial and 83% have generalized convulsions (Belman et al., 1985; Garzoni et al., 2009). Status epilepticus is reported in 4% (Belman et al., 1985). Postictally, the EEG most often shows slow activity, but an epileptic focus can be seen in a minority of cases. In one study, EEG was reported to be abnormal in 46% of HSP children without seizure or overt neurologic dysfunction (Ostergaard and Storm, 1991). However, the authors could not rule out that these anomalies were benign variants. Seizures rarely recur beyond the acute phase, with only a few cases of epilepsy.

Neuro-ophthalmologic manifestations Anterior ischemic optic neuropathy attributed to vasculitis was described in a 54-year-old diabetic male with a chronic course of HSP who presented with painless monocular visual loss (Chuah and Meaney, 2005). His vision improved with prednisolone treatment. A case of bilateral central retinal artery occlusion was also reported in a 6-year-old girl with concomitant cerebral vasculitis (Chen et al., 2000; Wu et al., 2002). Other rare ophthalmologic manifestations of HSP include central retinal vein occlusion, episcleritis, scleritis, keratitis, and anterior uveitis (Wu et al., 2002).

Myelopathy A rare HSP patient was described in whom prolonged treatment with corticosteroids led to epidural lipomatosis and ensuing mild myelopathy (Kano et al., 1998). His symptoms abated completely within 1 month of dose reduction.

Neuromuscular manifestations In contrast to its high prevalence in other primary small vessel vasculitides, peripheral nervous system

1106

M.D. BE´RUBE´ ET AL.

involvement in HSP is infrequent. Despite this, a wide spectrum of lesions has been described. Mononeuropathies account for the majority of cases. In 24 HSP patients with peripheral nervous system manifestations, 14 (58%) had mononeuropathies involving the peroneal (n ¼ 4), sciatic (n ¼ 3), posterior tibial (n ¼ 2), femoral (n ¼ 1), and facial nerves (n ¼ 3), four (17%) had Guillain–Barre´ syndrome, three (13%) had brachial plexopathy, two (8%) had polyradiculopathy, and one (4%) had mononeuritis multiplex (Belman et al., 1985). Interestingly, mononeuritis multiplex, the classic pattern of vasculitic neuropathy, has been reported only rarely (Campbell et al., 1994). The reason is unknown, but may be due to the short course of the disease preventing accumulation of lesions over time. Establishing a causal relationship of vasculitis with these lesions has been hampered by the lack of pathologic confirmation and presence of alternative etiologies in some patients. Temporal association with the onset of HSP and response to corticosteroids have been cited as circumstantial evidence of the presence of a vasculitic process. Other cases have been ascribed to local nerve compression by edema or hematoma (Belman et al., 1985). Four cases of rapidly progressive polyradiculoneuropathy and ascending paralysis consistent with Guillain– Barre´ syndrome have been reported in HSP (Sanghvi and Sharma, 1956; Moreau et al., 1988; Goraya et al., 1998; Mutsukura et al., 2007). CSF analysis revealed albuminocytologic dissociation in all cases. Electromyography was performed in two cases and showed an acute motor and sensory neuropathy (AMSAN) pattern in one (Mutsukura et al., 2007) and acute inflammatory demeyelinating polyneuropathy (AIDP) in the other (Moreau et al., 1988). This latter patient underwent sural nerve biopsy that revealed demyelination without signs of vasculitis or amyloidosis. In addition, there was evidence of intrathecal formation of IgA-containing immune complexes. On this basis, the authors suggested that IgA may have played a role in the initiation of demyelination in their patient, even though there was no evidence of IgA deposition in the pathologic specimen. Although IgA monoclonal spikes remain controversial as a cause of paraproteinemic neuropathy, recent evidence has borne out the notion that it can sometimes accumulate in and damage nerve fibers (Vital et al., 2008). Cases of Guillain–Barre´-like syndrome have been described in patients with IgA MGUS (Ropper and McKee, 1993). Muscle involvement has been reported in the form of intramuscular hemorrhage. It was present in 2% of patients in one series (Fischer et al., 1990). Pathologic examination in one case uncovered evidence of lymphocytic vasculitis in the hemorrhagic zone (Mahevas et al., 2004). Somekh et al., 2008 attributed severe calf and leg pain in three pediatric patients to muscular hemorrhage,

although there was no histological confirmation and muscle enzymes were normal or mildly elevated.

INVESTIGATIONS Nonspecific laboratory findings There is no specific marker for HSP and the purpose of laboratory testing is to eliminate alternative diagnoses and to detect complications. Mild inflammatory changes are commonly seen including anemia, mild leukocytosis, thrombocytosis and slightly elevated erythrocyte sedimentation rate. Elevation in creatinine levels occurs in a minority of patients. Routine coagulation tests are typically unremarkable, but markers of hypercoagulability and inflammation such as fibrinogen, D-dimer, thrombinantithrombin complex (TAT), prothrombin fragments 1 þ 2 (PF1 þ 2), von Willebrand antigen and activity may be increased. Factor XIII levels may be slightly decreased in up to 50% of cases (Tizard and Hamilton-Ayres, 2008). Markers of autoimmunity including antinuclear antibody and rheumatoid factor are typically negative. Testing for the lupus anticoagulant, anticardiolipin, and anti-b2-glycoprotein-I antibodies should be considered in the case of a thrombotic complication in order to rule out a coexistant antiphospholid antibody syndrome.

Antineutrophil cytoplasmic antibodies Antineutrophil cytoplasmic antibodies (ANCA) are autoantibodies associated with a specific group of small vessel vasculitides that includes Wegener granulomatosis (WG), microscopic polyangiitis (MPA), and Churg–Strauss syndrome. In these conditions, ANCA are usually of the IgG or IgM class. In HSP, systematic series have reported either absence (Robson et al., 1994) or very low rates of IgG ANCA positivity (Ozaltin et al., 2004). The existence of ANCAs of the IgA class in HSP is a contentious issue, with a wide range of reported positivity rates, from 0% to 82% (Ozaltin et al., 2004). Furthermore, it is unclear whether the antibodies that have been detected result from true antigen recognition or from nonspecific interactions attributable for instance to abnormal IgA glycosylation (Coppo et al., 1997). A single HSP patient with cerebral vasculitis and IgA positive and IgG negative ANCA was reported (Fanos, 2009). Another rare case of cerebral vasculitis diagnosed as a “Henoch–Scho¨nlein/MPA overlap syndrome” had mixed IgA predominant and pauci-immune lesions in addition to ANCA positivity (Nagasaka et al., 2009). In general, occurrence of IgG ANCA should lead to a diagnosis of one of the ANCA-positive vasculitides (von Scheven et al., 1998).

¨ NLEIN PURPURA NEUROLOGIC MANIFESTATIONS OF HENOCH–SCHO 1107 nonpalpable lesions (Schreiner, 1989). Though in chilCerebrospinal fluid analysis dren its appearance most frequently heralds HSP, palpaAmong HSP patients with an imaging study (computed ble purpura may be seen in a number of other tomography (CT) or magnetic resonance imaging inflammatory disorders, some of which also affect the (MRI)) deemed consistent with cerebral vasculitis, lumnervous system (Kathiresan et al., 2005). These include bar puncture is frequently normal or noninflammatory chiefly other primary vasculitides, but also infections (Benhamou et al., 1991; Goncalves et al., 2004). Abnorand embolic phenomena. Wegener’s granulomatosis malities that have been reported have included slight pro(WG) and microscopic polyangiitis (MPA) in particular teinorachia (Palesse et al., 1989), raised opening pressure are frequently confused with HSP initially (Hall et al., (Elinson et al., 1990), or elevated red blood cells (Chen 1985). Unlike HSP, their neurologic manifestations prefet al., 2000; Temkiatvises et al., 2008). Results of those erentially take the form of peripheral and, in WG, cranial with combined HSP and PRES have also been normal neuropathies; although rarer cases of cerebral vasculitis (Salloum et al., 2009) or showed mild lymphocytosis with have been described (von Scheven et al., 1998; Kono increased red blood cells (Woolfenden et al., 1998). Thus, et al., 2000; Ulinski et al., 2005). Systemic features as already stated, the usefulness of lumbar puncture to unusual for HSP such as rapidly progressive renal failure differentiate vasculitis from PRES seems limited. Its prior pulmonary involvement in the form of upper airway mary interest lies in excluding vasculitis mimickers and disease (WG), cavitary lung lesions (WG), or pulmonary secondary causes of CNS involvement. hemorrhage (WG or MPA) should lead the clinician to suspect these diagnoses. Additional discriminating charImaging studies acteristics include the presence of serum ANCAs and absence of immune complex deposition on biopsy. NecImaging is indicated in all cases of suspected CNS rotizing granulomas may be found in WG. Other vascuinvolvement. CT scan may be normal or show areas of litides affecting the small vessels should also be loss of attenuation. MRI is superior to CT in detecting considered, including Churg–Strauss syndrome, the early ischemic changes, hemorrhages and lesions of hypersensitivity vasculitides as well as those associated the posterior fossa. This superiority has been illustrated with inflammatory disorders such as systemic lupus in many HSP case reports (Chen et al., 2000; Bulun et al., erythematosus, Behc¸et’s disease, rheumatoid arthritis, 2001; Fanos, 2009). It should be considered the study of or Sjo¨gren syndrome. Most of these diseases are rarely choice. In cases of vasculitis, MRI will most often demencountered in children. onstrate confluent areas of cortical and subcortical T2Among infectious causes, meningococcemia and hyperintense signal predominant in the parieto-occipital Rocky Mountain spotted fever may present with extenregions. Enhancement with gadolinium can be seen (Eun sive palpable purpura and CNS signs. They should et al., 2003). One patient with multiple recurrent bouts of always be kept in mind, especially in cases with a suggesHSP over 6 years was found to have moderate cerebral tive epidemiological context, septic features, or atrophy in addition to multiple periventricular hyperindepressed consciousness. Similarly, the presentation of tensities on MRI (Perez et al., 2000). Magnetic resoinfectious endocarditis may combine petechiae as well nance angiography was reported to show irregularities as multifocal CNS signs. Actual cerebral vasculitis from on segments of middle and posterior cerebral arteries immune complex deposition supervenes in some patients in a single case of suspected cerebral vasculitis (Eun (Johnson and Johnson, 2010). et al., 2003). These finding should be taken with caution, By definition, the purpura of HSP is nonthrombocyhowever, as this technique is subject to artifacts and the topenic, eliminating such potential etiologies of CNS nature of the pathology makes it unlikely that anomalies dysfunction as thrombotic thrombocytopenic purpura will be seen with the current resolution achieved. Simiand disseminated intravascular coagulation. larly, conventional cerebral angiography has not shown The diagnosis of HSP can be difficult when it presents anomalies in patients in whom it was performed (Ng with symptoms other than purpura, as it occurs in 25– et al., 1996; Paolini et al., 2003; Temkiatvises et al., 50% of cases (Saulsbury, 1999). In one review, CNS man2008). Its greater resolution may help identifying alterifestations preceded the onset of the rash in 16% native diagnoses in selected cases. (Garzoni et al., 2009). They may even occasionally be the heralding feature of the disorder (Mannenbach DIFFERENTIAL DIAGNOSIS et al., 2009). Abdominal presentations are particularly The hallmark of HSP is purpura. Its palpable nature is a confusing and entail a large differential diagnosis. key characteristic that signals inflammation of the Among vasculitides, polyarteritis nodosa may present small skin vessels and permits distinction from hematowith prominent gastrointestinal complaints. Unlike logical or cutaneous causes, which are associated with HSP, however, there is usually a history of prodromal

M.D. BE´RUBE´ ET AL.

1108

systemic symptoms, cutaneous findings are different, and glomerulonephritis is absent. Again, the PNS is most commonly affected, but focal CNS deficits have been reported in up to 24% of patients (Rosenberg et al., 1990). Abdominal angiography may be required in some case to exclude the diagnosis.

MANAGEMENT HSP is usually a self-limited disease that requires only supportive care. Steroids are used in patients with refractory abdominal pain, and those with severe nephritis may respond to high-dose methylprednisolone, alone or in combination with cyclophosphamide (Tizard and Hamilton-Ayres, 2008). No controlled data exist in cases of cerebral involvement. Most patients have been treated with corticosteroids, usually as intravenous pulses of methylprednisolone followed by an oral tapering regimen. This is reported to have led to resolution of neurologic manifestations in the majority, but whether this represents true treatment effect or natural evolution of the disease is hard to discern. Cyclophosphamide has also been employed in combination with steroids in a few cases but again, it is unknown if it provides additional benefit for CNS manifestations (Goncalves et al., 2004; Ozkaya et al., 2007; Karamadoukis et al., 2008). Although some have proposed that the combination should be considered as the standard treatment regimen (Garzoni et al., 2009), the literature suggests that most patients do well on steroids alone. Therefore, cyclophosphamide seems best reserved for refractory cases or those with concurrent nephritis. Refractory patients may also benefit from plasma exchange. Several case reports as well as a small retrospective series that included patients with HSP provide some support for its efficacy, either as an initial therapy or as a rescue intervention in those who had failed to improve on steroids (Gianviti et al., 1996; Chen et al., 2000; Eun et al., 2003; Wen et al., 2005; Murakami et al., 2008). An effect for intravenous immunoglobulins was suggested in the case of an adult HSP patient with mild encephalopathy and MR hyperintensities who experienced resolution of his symptoms following their administration (Perez et al., 2000). It should again be emphasized that distinguishing vasculitis from PRES is difficult and that, when in doubt, one should address the two conditions simultaneously. In PRES, the mainstay of treatment is control of hypertension. Hemodialysis may occasionally be required to reduce fluid overload and uremia. The use of corticosteroids is more controversial. A causal relationship linking high-dose steroids to PRES has been suggested, but not established, in some patients (Hinchey et al., 1996). Others have pointed out that they may be detrimental

by amplifying hypertension, volume overload, and hypercoagulability. Nonetheless, steroids have been successfully employed in several cases of PRES attributable to different vasculitides, including those reported in association with HSP (Woolfenden et al., 1998; Ozcakar et al., 2004; Min et al., 2006; Sasayama et al., 2007; Salloum et al., 2009). They may have a role in reducing endothelial injury from systemic inflammation and restoring permeability of the blood–brain barrier (Nishio et al., 2008). Their use may be justified in many cases. Patients with hemorrhage should be closely monitored and emergent neurosurgical intervention considered in case of deterioration. Attention should be paid to correction of uncontrolled hypertension and metabolic abnormalities. The role of factor XIII replacement has not been firmly established and cannot be recommended at this time. PNS lesions in HSP generally have a favorable prognosis and most may not warrant immunosuppression (Garzoni et al., 2009). Nonetheless, consideration to treatment should still be given in cases with severe impairment, as illustrated by a report of refractory and persisting brachial plexopathy (Yilmaz et al., 2006).

PROGNOSIS Most patients with neurologic dysfunction make a good recovery although minor deficits occur with some frequency. In one review, 21% of patients with CNS manifestations were left with sequelae, including visual field defects, verbal disabilities, focal neurologic signs and localization-related epilepsy. Half had previously developed ICH. In 17 patients with a PNS lesion, only one (6%) had residual impairment (Garzoni et al., 2009). While three of 17 patients in the de Montis 1971 series had died, only one death has occurred in the 39 HSP cases with CNS dysfunction (3%) reported since 1983 (Garzoni et al., 2009).

REFERENCES Abend NS, Licht DJ, Spencer CH (2007). Lupus anticoagulant and thrombosis following Henoch–Scho¨nlein purpura. Pediatr Neurol 36: 345–347. Allen DM, Diamond LK, Howell DA (1960). Anaphylactoid purpura in children (Scho¨nlein–Henoch syndrome): review with a follow-up of the renal complications. AMA J Dis Child 99: 833–854. Allen AC, Willis FR, Beattie TJ et al. (1998). Abnormal IgA glycosylation in Henoch–Scho¨nlein purpura restricted to patients with clinical nephritis. Nephrol Dial Transplant 13: 930–934. Altinors N, Cepoglu C (1991). Surgically treated intracerebral hematoma in a child with Henoch–Scho¨nlein purpura. J Neurosurg Sci 35: 47–49.

¨ NLEIN PURPURA NEUROLOGIC MANIFESTATIONS OF HENOCH–SCHO Aram H (1987). Henoch–Scho¨nlein purpura induced by chlorpromazine. J Am Acad Dermatol 17: 139–140. Bartynski WS (2008). Posterior reversible encephalopathy syndrome, part 1: fundamental imaging and clinical features. AJNR Am J Neuroradiol 29: 1036–1042. Belman AL, Leicher CR, Moshe SL et al. (1985). Neurologic manifestations of Schoenlein–Henoch purpura: report of three cases and review of the literature. Pediatrics 75: 687–692. Benhamou B, Balafrej A, Jaritz E et al. (1991). Cerebral ischemia and severe digestive manifestations during rheumatoid purpura. Ann Pediatr (Paris) 38: 484–486. Brogan PA (2007). What’s new in the aetiopathogenesis of vasculitis? Pediatr Nephrol 22: 1083–1094. Bulun A, Topaloglu R, Duzova A et al. (2001). Ataxia and peripheral neuropathy: rare manifestations in Henoch– Scho¨nlein purpura. Pediatr Nephrol 16: 1139–1141. Calvino MC, Llorca J, Garcia-Porrua C et al. (2001). Henoch– Scho¨nlein purpura in children from northwestern Spain: a 20-year epidemiologic and clinical study. Medicine (Baltimore) 80: 279–290. Campbell SB, Hawley CM, Staples C (1994). Mononeuritis multiplex complicating Henoch–Scho¨nlein purpura. Aust N Z J Med 24: 580. Chen CL, Chiou YH, Wu CY et al. (2000). Cerebral vasculitis in Henoch–Scho¨nlein purpura: a case report with sequential magnetic resonance imaging changes and treated with plasmapheresis alone. Pediatr Nephrol 15: 276–278. Chen M, Daha MR, Kallenberg CG (2010). The complement system in systemic autoimmune disease. J Autoimmun 34: J276–J286. Chiaretti A, Caresta E, Piastra M et al. (2002). Cerebral hemorrhage in Henoch–Schoenlein syndrome. Childs Nerv Syst 18: 365–367. Chuah J, Meaney T (2005). Anterior ischaemic optic neuropathy secondary to Henoch–Scho¨nlein purpura. Eye (Lond) 19: 1028. Clark JH, Fitzgerald JF (1985). Hemorrhagic complications of Henoch–Scho¨nlein syndrome. J Pediatr Gastroenterol Nutr 4: 311–315. Coppo R, Cirina P, Amore A et al. (1997). Properties of circulating IgA molecules in Henoch–Scho¨nlein purpura nephritis with focus on neutrophil cytoplasmic antigen IgA binding (IgA-ANCA): new insight into a debated issue. Italian Group of Renal Immunopathology Collaborative Study on Henoch–Scho¨nlein purpura in adults and in children. Nephrol Dial Transplant 12: 2269–2276. Davin JC, Ten Berge IJ, Weening JJ (2001). What is the difference between IgA nephropathy and Henoch–Scho¨nlein purpura nephritis? Kidney Int 59: 823–834. de Montis G, Turpin JC (1971). Rheumatoid purpura and neurologic manifestations. Ann Med Interne (Paris) 122: 841–848. Elinson P, Foster KW Jr, Kaufman DB (1990). Magnetic resonance imaging of central nervous system vasculitis. A case report of Henoch–Scho¨nlein purpura. Acta Paediatr Scand 79: 710–713. Eun SH, Kim SJ, Cho DS et al. (2003). Cerebral vasculitis in Henoch–Scho¨nlein purpura: MRI and MRA findings, treated with plasmapheresis alone. Pediatr Int 45: 484–487.

1109

Fanos V (2009). Cerebral vasculitis and nephritis in a child: complicated Henoch–Scho¨nlein purpura or a rare case of Wegener’s granulomatosis? J Paediatr Child Health 45: 163–165. Fervenza FC (2003). Henoch–Scho¨nlein purpura nephritis. Int J Dermatol 42: 170–177. Fischer PJ, Hagge W, Hecker W (1990). Scho¨nlein–Henoch purpura. A clinical study of 119 patients with special reference to unusual complications. Monatsschr Kinderheilkd 138: 128–134. Gairdner D (1948). The Scho¨nlein–Henoch syndrome (anaphylactoid purpura). Q J Med 17: 95–122. Garzoni L, Vanoni F, Rizzi M et al. (2009). Nervous system dysfunction in Henoch–Scho¨nlein syndrome: systematic review of the literature. Rheumatology (Oxford) 48: 1524–1529. Gedalia A (2004). Henoch–Scho¨nlein purpura. Curr Rheumatol Rep 6: 195–202. Gianviti A, Trompeter RS, Barratt TM et al. (1996). Retrospective study of plasma exchange in patients with idiopathic rapidly progressive glomerulonephritis and vasculitis. Arch Dis Child 75: 186–190. Gilbert EF, Da Silva A (1966). Henoch–Schoenlein purpura. Report of a fatal case with gastrointestinal hemorrhage and necrosis. Clin Pediatr (Phila) 5: 181–186. Goncalves C, Ferreira G, Mota C et al. (2004). Cerebral vasculitis in Henoch–Scho¨nlein purpura. An Pediatr (Barc) 60: 188–189. Goraya JS, Jayashree M, Ghosh D et al. (1998). Guillain–Barre´ syndrome in a child with Henoch–Scho¨nlein purpura. Scand J Rheumatol 27: 310–312. Hall SL, Miller LC, Duggan E et al. (1985). Wegener granulomatosis in pediatric patients. J Pediatr 106: 739–744. Henriksson P, Hedner U, Nilsson IM (1977). Factor XIII (fibrin stabilising factor) in Henoch–Scho¨nlein’s purpura. Acta Paediatr Scand 66: 273–277. Hinchey J, Chaves C, Appignani B et al. (1996). A reversible posterior leukoencephalopathy syndrome. N Engl J Med 334: 494–500. Hsieh L, Nugent D (2008). Factor XIII deficiency. Haemophilia 14: 1190–1200. Imai T, Okada H, Nanba M et al. (2002). Henoch–Scho¨nlein purpura with intracerebral hemorrhage. Brain Dev 24: 115–117. Johnson MD, Johnson CD (2010). Neurologic presentations of infective endocarditis. Neurol Clin 28: 311–321. Kano K, Ozawa T, Kuwashima S et al. (1998). Uncommon multisystemic involvement in a case of Henoch– Scho¨nlein purpura. Acta Paediatr Jpn 40: 159–161. Karamadoukis L, Ludeman L, Williams AJ (2008). Henoch– Scho¨nlein purpura with intracerebral haemorrhage in an adult patient: a case report. J Med Case Reports 2: 200. Kathiresan S, Kelsey PB, Steere AC et al. (2005). Case records of the Massachusetts General Hospital. Case 14-2005. A 38-year-old man with fever and blurred vision. N Engl J Med 352: 2003–2012. Kono H, Inokuma S, Nakayama H et al. (2000). Pachymeningitis in microscopic polyangiitis (MPA): a case report and a review of central nervous system involvement in MPA. Clin Exp Rheumatol 18: 397–400.

1110

M.D. BE´RUBE´ ET AL.

Laplane R, Fontaine JL, Escourolle G et al. (1973). The neurological signs of rheumatoid pupura. Pathology and clinical signs in one case. Ann Pediatr (Paris) 20: 525–530. Lorand L, Losowsky MS, Miloszewski KJ (1980). Human factor XIII: fibrin-stabilizing factor. Prog Hemost Thromb 5: 245–290. Mahevas M, Makdassi R, Presne C et al. (2004). Muscular haematoma in Henoch–Scho¨nlein purpura. Rev Med Interne 25: 927–930. Mak A, Chan BP, Yeh IB et al. (2008). Neuropsychiatric lupus and reversible posterior leucoencephalopathy syndrome: a challenging clinical dilemma. Rheumatology (Oxford) 47: 256–262. Mannenbach MS, Reed AM, Moir C (2009). Atypical presentation of Henoch–Scho¨nlein purpura. Pediatr Emerg Care 25: 513–515. Mills JA, Michel BA, Bloch DA et al. (1990). The American College of Rheumatology 1990 criteria for the classification of Henoch–Scho¨nlein purpura. Arthritis Rheum 33: 1114–1121. Min L, Zwerling J, Ocava LC et al. (2006). Reversible posterior leukoencephalopathy in connective tissue diseases. Semin Arthritis Rheum 35: 388–395. Misra AK, Biswas A, Das SK et al. (2004). Henoch–Scho¨nlein purpura with intracerebral haemorrhage. J Assoc Physicians India 52: 833–834. Moreau BA, Schuller E, Georges B (1988). Association of Guillain–Barre´ syndrome and Henoch–Scho¨nlein purpura: is immunoglobulin a responsible for the neurologic syndrome? Am J Med Sci 296: 198–201. Moritani T, Shrier DA, Numaguchi Y et al. (2001). Diffusionweighted echo-planar MR imaging of CNS involvement in systemic lupus erythematosus. Acad Radiol 8: 741–753. Moritani T, Hiwatashi A, Shrier DA et al. (2004). CNS vasculitis and vasculopathy: efficacy and usefulness of diffusionweighted echoplanar MR imaging. Clin Imaging 28: 261–270. Murakami H, Takahashi S, Kawakubo Y et al. (2008). Adolescent with Henoch–Scho¨nlein purpura glomerulonephritis and intracranial hemorrhage possibly secondary to the reactivation of latent CMV. Pediatr Int 50: 112–115. Mutsukura K, Tsuboi Y, Fujiki F et al. (2007). Acute motor sensory axonal neuropathy associated with Henoch– Scho¨nlein purpura. J Neurol Sci 263: 169–173. Nagasaka T, Miyamoto J, Ishibashi M et al. (2009). MPOANCA- and IgA-positive systemic vasculitis: a possibly overlapping syndrome of microscopic polyangiitis and Henoch–Schoenlein purpura. J Cutan Pathol 36: 871–877. Ng CC, Huang SC, Huang LT (1996). Henoch–Scho¨nlein purpura with intracerebral hemorrhage: case report. Pediatr Radiol 26: 276–277. Niedermaier G, Briner V (1997). Henoch–Scho¨nlein syndrome induced by carbidopa/levodopa. Lancet 349: 1071–1072. Nishio M, Yoshioka K, Yamagami K et al. (2008). Reversible posterior leukoencephalopathy syndrome: a possible manifestation of Wegener’s granulomatosis-mediated endothelial injury. Mod Rheumatol 18: 309–314.

Osler W (1914). The visceral lesions of purpura and allied conditions. Br Med J Clin Res J 1: 517–525. Ostergaard JR, Storm K (1991). Neurologic manifestations of Scho¨nlein–Henoch purpura. Acta Paediatr Scand 80: 339–342. Ozaltin F, Bakkaloglu A, Ozen S et al. (2004). The significance of IgA class of antineutrophil cytoplasmic antibodies (ANCA) in childhood Henoch–Scho¨nlein purpura. Clin Rheumatol 23: 426–429. Ozcakar ZB, Ekim M, Fitoz S et al. (2004). Hypertension induced reversible posterior leukoencephalopathy syndrome: a report of two cases. Eur J Pediatr 163: 728–730. Ozen S, Pistorio A, Iusan SM et al. (2010). EULAR/PRINTO/ PRES criteria for Henoch–Scho¨nlein pupura, childhood polyarteritis nodosa, childhood Wegener granulomatosis and childhood Takayasu arteritis: Ankara 2008. Part II: Final classification criteria. Ann Rheum Dis 69: 798–806. Ozkaya O, Bek K, Alaca N et al. (2007). Cerebral vasculitis in a child with Henoch–Scho¨nlein purpura and familial Mediterranean fever. Clin Rheumatol 26: 1729–1732. Palesse N, Marrelli A, Legge MP et al. (1989). Neurological complications of Schoenlein–Henoch syndrome: contribution of MR to the diagnosis. Case report. Ital J Neurol Sci 10: 351–355. Paolini S, Ciappetta P, Piattella MC et al. (2003). Henoch– Scho¨nlein syndrome and cerebellar hemorrhage: report of an adolescent case and literature review. Surg Neurol 60: 339–342. Perez C, Maravi E, Olier J et al. (2000). MR imaging of encephalopathy in adult Henoch–Scho¨nlein purpura. AJR Am J Roentgenol 175: 922–923. Peru H, Soylemezoglu O, Bakkaloglu SA et al. (2008). Henoch Scho¨nlein purpura in childhood: clinical analysis of 254 cases over a 3-year period. Clin Rheumatol 27: 1087–1092. Pillebout E, Niaudet P (2008). Henoch–Scho¨nlein purpura. Rev Prat 58: 507–511. Prenzel F, Pfaffle R, Thiele F et al. (2006). Decreased factor XIII activity during severe Henoch–Schoenlein purpura – does it play a role? Klin Padiatr 218: 174–176. Roberts PF, Waller TA, Brinker TM et al. (2007). Henoch– Scho¨nlein purpura: a review article. South Med J 100: 821–824. Robson WL, Leung AK, Woodman RC (1994). The absence of anti-neutrophil cytoplasmic antibodies in patients with Henoch–Scho¨nlein purpura. Pediatr Nephrol 8: 295–298. Ropper AH, McKee AC (1993). Case records of the Massachusetts General Hospital. Weekly clinicopathological exercises. Case 21-1993. A 71-year-old man with a rash and severe sensorimotor neuropathy. N Engl J Med 328: 1550–1558. Rosenberg MR, Parshley M, Gibson S et al. (1990). Central nervous system polyarteritis nodosa. West J Med 153: 553–556. Salloum AC, Cuisset JM, Vermelle M et al. (2009). Posterior reversible encephalopathy as a complication of rheumatoid purpura: a case study. Arch Pediatr 16: 284–286. Sanghvi LM, Sharma R (1956). Guillain–Barre´ syndrome and presumed allergic purpura. AMA Arch Neurol Psychiatry 76: 497–499.

¨ NLEIN PURPURA NEUROLOGIC MANIFESTATIONS OF HENOCH–SCHO Sasayama D, Shimojima Y, Gono T et al. (2007). Henoch– Scho¨nlein purpura nephritis complicated by reversible posterior leukoencephalopathy syndrome. Clin Rheumatol 26: 1761–1763. Saulsbury FT (1999). Henoch–Scho¨nlein purpura in children. Report of 100 patients and review of the literature. Medicine (Baltimore) 78: 395–409. Saulsbury FT (2001). Henoch–Scho¨nlein purpura. Curr Opin Rheumatol 13: 35–40. Saulsbury FT (2007). Clinical update: Henoch–Scho¨nlein purpura. Lancet 369: 976–978. Scattarella V, Pannarale P, D’Angelo V et al. (1983). Occipital hemorrhage in a child with Scho¨nlein–Henoch syndrome. J Neurosurg Sci 27: 37–39. Schreiner DT (1989). Purpura. Dermatol Clin 7: 481–490. Somekh E, Fried D, Hanukoglu A (2008). Muscle involvement in Scho¨nlein-Henoch syndrome. Arch Dis Child 58: 929–930. Sokol DK, McIntyre JA, Short RA et al. (2000). Henoch– Scho¨nlein purpura and stroke: antiphosphatidylethanolamine antibody in CSF and serum. Neurology 55: 1379–1381. Sticca M, Barca S, Spallino L et al. (1999). Scho¨nlein–Henoch syndrome: clinical-epidemiological analysis of 98 cases. Pediatr Med Chir 21: 9–12. Temkiatvises K, Nilanont Y, Poungvarin N (2008). Stroke in Henoch–Scho¨nlein purpura associated with methicillinresistant Staphylococcus aureus septicemia: report of a case and review of the literature. J Med Assoc Thai 91: 1296–1301. Tizard EJ, Hamilton-Ayres MJ (2008). Henoch Scho¨nlein purpura. Arch Dis Child Educ Pract Ed 93: 1–8. Trapani S, Micheli A, Grisolia F et al. (2005). Henoch Scho¨nlein purpura in childhood: epidemiological and clinical analysis of 150 cases over a 5-year period and review of literature. Semin Arthritis Rheum 35: 143–153.

1111

Ulinski T, Martin H, Mac Gregor B et al. (2005). Fatal neurologic involvement in pediatric Wegener’s granulomatosis. Pediatr Neurol 32: 278–281. Urizar RE, Michael A, Sisson S et al. (1968). Anaphylactoid purpura. II. Immunofluorescent and electron microscopic studies of the glomerular lesions. Lab Invest 19: 437–450. Vital A, Nedelec-Ciceri C, Vital C (2008). Presence of crystalline inclusions in the peripheral nerve of a patient with IgA lambda monoclonal gammopathy of undetermined significance. Neuropathology 28: 526–531. von Scheven E, Lee C, Berg BO (1998). Pediatric Wegener’s granulomatosis complicated by central nervous system vasculitis. Pediatr Neurol 19: 317–319. Wen YK, Yang Y, Chang CC (2005). Cerebral vasculitis and intracerebral hemorrhage in Henoch–Scho¨nlein purpura treated with plasmapheresis. Pediatr Nephrol 20: 223–225. Woolfenden AR, Hukin J, Poskitt KJ et al. (1998). Encephalopathy complicating Henoch–Scho¨nlein purpura: reversible MRI changes. Pediatr Neurol 19: 74–77. Wu TT, Sheu SJ, Chou LC (2002). Henoch–Scho¨nlein purpura with bilateral central retinal artery occlusion. Br J Ophthalmol 86: 351–352. Yang YH, Chuang YH, Wang LC et al. (2008). The immunobiology of Henoch–Scho¨nlein purpura. Autoimmun Rev 7: 179–184. Yilmaz C, Caksen H, Arslan S et al. (2006). Bilateral brachial plexopathy complicating Henoch–Scho¨nlein purpura. Brain Dev 28: 326–328. Zajadacz B, Juszkiewicz A (2005). Increased levels of plasma D-dimer in the course of Henoch–Scho¨nlein purpura. Wiad Lek 58: 581–583.

Handbook of Clinical Neurology, Vol. 120 (3rd series) Neurologic Aspects of Systemic Disease Part II Jose Biller and Jose M. Ferro, Editors © 2014 Elsevier B.V. All rights reserved

Chapter 75

Hemolytic uremic syndrome 1

KATHLEEN WEBSTER1* AND EUGENE SCHNITZLER2 Department of Pediatrics, Loyola University Medical Center, Maywood, IL, USA

2

Department of Neurology, Loyola University Medical Center, Maywood, IL, USA

INTRODUCTION The terms “microangiopathic hemolytic anemia” and “thrombotic microangiopathy” have been used to describe disorders consisting of endothelial cell damage, thrombosis, and resulting thrombocytopenia and red cell destruction. Two important causes of these findings are hemolytic uremic syndrome (HUS) and thrombotic thrombocytopenic purpura (TTP). Both disorders can lead to widespread organ damage and predominantly affect the brain and kidney. In TTP, renal disease is less common and neurologic involvement is more pronounced. Although TTP is more commonly reported in adults and HUS in children, there have been reported cases of each disorder occurring outside of the typical age ranges and both should be considered in the appropriate clinical setting (Moake, 2002).

DEFINITION HUS was first described by Gasser and colleagues in 1955. It is defined by the triad of microangiopathic hemolytic anemia, thrombocytopenia, and acute renal failure (Eriksson et al., 2001; Scheiring et al., 2008; Johnson and Taylor, 2009; Michael et al., 2009). Typical (D þ) HUS follows within 2 weeks of an acute diarrheal illness. Atypical (or D ) HUS has also been reported and accounts for 10–20% of reported cases. Potential etiologies of atypical HUS include complement deficiency, inborn errors of metabolism, medications, infectious agents, and autoimmune disease processes (Table 75.1). The distinction between these two types can be important with regards to etiology, pathogenesis, long-term course, and treatment. Atypical HUS (aHUS) is associated with an increased risk of seizures and other

neurologic complications (Johnson and Taylor, 2009; Michael et al., 2009; Scheiring et al., 2010).

EPIDEMIOLOGYAND ETIOLOGY Although HUS is most commonly known as a leading cause of acute renal failure in children (Cerda et al., 2008), other organ systems have also been reported to be affected, particularly the central nervous system (CNS) (Eriksson et al., 2001; Banerjee, 2009; Scheiring et al., 2010). It is reported that up to half of children with HUS will have CNS involvement, most commonly in the form of seizures, altered mental status, or coma. HUS is estimated to occur with a frequency of 0.3–3.3 per 100 000 children worldwide, although it is noted that the incidence has been increasing in the past decade (Eriksson et al., 2001; Pomajzl et al., 2009). The most commonly affected age groups are children less than 5 years old, accounting for 50–70% of cases, and adolescents. Infants have rarely been reported to be affected. Up to 40% of children may require critical care support. Mortality from HUS is 2– 7%, with higher risk seen in females, children under 5 years of age, the elderly, and patients with atypical or recurrent HUS (Gould et al., 2009; Rust and Worrel, 2009). In both typical HUS and aHUS, early presentation of CNS symptoms is associated with a less favorable outcome (Rust and Worrel, 2009; Scheiring et al., 2010).

Typical hemolytic uremic syndrome The typical presentation of HUS occurs following a diarrheal prodrome. Diarrhea is infectious, usually due to Shiga toxin-producing Escherichia coli (STEC) (Ruggenenti et al., 2001; Michael et al., 2009). Although the 0157:H7 subtype of E. coli is the most commonly identified in the US, other subtypes have been found (Centers for Disease Control, 2009; Melmann et al., 2009). Of

*Correspondence to: Kathleen Webster, M.D., Loyola University Medical Center, 2160 S 1st Avenue, Maywood, IL 60153, USA. Tel: þ1-708-327-9137, Fax: þ1-708-216-5602, E-mail: [email protected]

1114

K. WEBSTER AND E. SCHNITZLER

Table 75.1 Etiologies of atypical hemolytic uremic syndrome Infectious

Noninfectious

STEC infection Diarrheal

Complement deficiency Cobalamin metabolism deficiency Malignancy Transplant SLE Antiphospholipid syndrome Pregnancy HELLP syndrome Medication-induced Quinine Oral contraceptives Immunosuppressants Tacrolimus Ciclosporin Mycophenolate

Urinary Other diarrheal infections Shigella Citrobacter Yersinia Clostridium difficile Giardia Viral infections HIV H1N1 influenza A CMV EBV Other infections Pertussis Malaria Pneumococcal

STEC, Shiga toxin-producing Escherichia coli; HIV, human immunodeficiency virus; CMV, cytomegalovirus; EBV, Epstein–Barr virus; SLE, systemic lupus erythematosus; HELLP, hypertension, elevated liver enzymes, low platelets.

note, although E. coli 0157:H7 is well known as the cause of HUS, only 8% of persons with diarrheal illness due to this organism go on to develop HUS. Other infectious causes of diarrhea have been identified in afflicted patients, including Shigella, Citrobacter, Yersinia, Clostridium difficile and Giardia (Besbas et al., 2006; Pomajzl et al., 2009). STEC associated HUS accounts for up to 90% of cases and follows a seasonal pattern with higher incidence in summer and fall. Food-borne outbreaks have been reported and may be associated with raw ground beef, unpasteurized juice or milk, fresh produce such as lettuce, spinach, and alfalfa sprouts, and contaminated water (Razzaq, 2006; Rivero et al., 2010). The bacteria may be easily spread, and infection may be acquired from contact with animals and their environment, or other infected persons. In cases of multiple family members with symptoms it is important to distinguish concurrent HUS with diarrheal symptoms from asynchronous events, which may be related to genetic causes or complement deficiencies (Centers for Disease Control, 2009; Pomajzl et al., 2009).

Atypical hemolytic uremic syndrome Atypical HUS (aHUS) is being described in a growing body of literature and is more commonly associated with

neurologic sequelae. In addition to diarrheal infections, HUS may be triggered by STEC infection of the urinary tract. Viral infections may also be associated with HUS. Reported cases have involved human immunodeficiency virus (HIV), novel H1N1 influenza A, cytomegalovirus (CMV), Epstein–Barr virus (EBV), and others (Ariceta et al., 2009; Banerjee, 2009; Printza et al., 2011; C¸altik et al., 2011). There are also several case reports of infants with HUS related to pertussis (Chaturvedi et al., 2010). Sharma et al. (1993) reported HUS following malaria. Another notable infectious cause is bacteremia, meningitis, or empyema due to Streptococcus pneumoniae. This particularly virulent form of HUS mainly affects children under 2 years of age and is associated with a much higher mortality (Brandt et al., 2002; Barit and Sakarcan, 2005; Besbas et al., 2006; Lei et al., 2010; Scheiring et al., 2010). Atypical HUS is most commonly related to noninfectious causes. Over 50% of cases are attributed to congenital or acquired complement deficiency, notably disorders of factor H, factor I, factor B, and membrane cofactor protein (MCP) (Quintrec et al., 2010). Abnormal processing of cobalamin (vitamin B12) leading to severe deficiency is an inborn error of metabolism and well described as an etiology of a particularly severe form of HUS with a high rate of neurologic symptoms (Sharma et al., 2007). Other reported associations with atypical HUS are malignancies, chemotherapy, systemic lupus erythematosis (SLE), antiphospholipid syndrome, pregnancy, and HELLP syndrome, and with medications such as quinine or oral contraceptives (Besbas et al., 2006; Michael et al., 2009; Scheiring et al., 2010). Patients who have had solid organ or bone marrow transplant also seem to be more susceptible to development of HUS. It is unclear whether this is due to the transplant state itself or due to immunosuppressive medications, such as tacrolimus, ciclosporin, and mycophenolate (Ariceta et al., 2009; Banerjee, 2009). In many of the atypical cases of HUS, it has been proposed that both a genetic predisposition and an environmental trigger are required to produce disease (Johnson and Taylor, 2008).

Thrombotic thrombocytopenic purpura The presentation of HUS is closely related to thrombotic thrombocytopenic purpura (TTP). Like HUS, TTP is associated with microangiopathic hemolytic anemia and thrombocytopenia. The initial description of TTP by Moschowitz in 1925 reported a “pentad” of these two findings in addition to fever, neurologic impairment, and renal dysfunction. As in HUS, platelet aggregation occurs, although the etiology in TTP is deficiency of a protease known as ADAMTS13. This deficiency

HEMOLYTIC UREMIC SYNDROME may be congenital, known as Upshaw–Schulman disease, with presentation occurring early in childhood, even in infancy (Schneppenheim et al., 2004; Lowe and Werner, 2005). Acquired deficiency may be related to autoantibodies against ADAMTS13 and present later in life, triggered by medications, collagen vascular disease, or infection. Untreated, the mortality of TTP is over 90%, although with plasma therapy, the incidence of mortality drops to 10–20%. In the current literature, there is disagreement as to whether TTP is a distinct syndrome from HUS, or whether these two disorders represent a spectrum of disease. HUS is classically thought of as a renal disease with systemic effects, while TTP is considered as a systemic disease with effects on the central nervous system and kidney (Desch and Motto, 2007). Often the two disorders are difficult to distinguish based on clinical presentation alone and further testing of complement and genetic factors is required to make a definitive diagnosis. Many of the same medications and diseases can trigger TTP and atypical HUS (Nolasco et al., 2005; Desch and Motto, 2007). It remains to be seen whether these two disorders will be found to be more similar or more distinct as the pathophysiology continues to be explored.

PATHOPHYSIOLOGY As described above, a wide range of etiologies of typical and atypical HUS and TTP are reported. Although each of these results in a common clinical picture, the pathophysiology of each is distinct.

Typical hemolytic uremic syndrome In STEC-associated HUS, the infecting organism enters the gastrointestinal tract and binds to and causes inflammation of the intestinal epithelium (Pomajzl et al., 2009). The bacteria produce a toxin, called Shiga toxin (Stx) or verotoxin. The toxin enters the bloodstream, bound to a carrier such as monocytes, neutrophils, or platelets. The toxin binds to the globotriaosyl ceramide (Gb3) receptor. The toxin results in red cell lysis and damage to vascular endothelium. This in turn leads to a cascade of microthrombosis, platelet activation, and thrombotic injury to the vasculature (Lowe and Werner, 2005; Karpman et al., 2010; Scheiring et al., 2010).

Atypical hemolytic uremic syndrome Atypical HUS may be caused by a variety of etiologies, each with a different trigger resulting in the final common pathway of inflammatory cascade causing renal endothelial and vascular injury and resultant thrombotic microangiopathy. Up to 50% of patients may have chromosome 1 mutations or autoantibody production leading

1115

to deficiency and dysregulation of the complement cascade (Loirat et al., 2008; Banerjee, 2009). Another mutation on chromosome 1 may lead to deficient processing of cobalamin (vitamin B12). This leads to a defect in the processing of cobalamin, with associated homocystinuria and methylmalonic academia (Sharma et al., 2007; Banerjee, 2009; Bouts et al., 2010; Martinelli et al., 2011). This form of aHUS usually presents in infancy and is associated with significant neurologic manifestations including hypotonia, lethargy, feeding difficulties and failure to thrive (Ariceta et al., 2009). Finally, pneumococcal infection may lead to production of neuraminidase, which exposes a protein known as Thomsen-Friedenreich antigen (TF-Ag) on renal endothelial cells, and causing immune activation (Banerjee, 2009; Bouts et al., 2010).

CLINICAL MANIFESTATIONS In typical HUS, the history may include exposure to contaminated food. Symptoms generally develop 1–8 days following ingestion of the offending agent. Clinically the child usually presents with bloody diarrhea and cramping abdominal pain. Physical examination may show petechiae and/or mucosal bleeding. Diagnostic criteria include anemia, thrombocytopenia, and elevated creatinine. An “incomplete” HUS has been described in children who present with bloody diarrhea, anemia, and thrombocytopenia but have mild or absent elevation of creatinine and do not go on to develop overt renal failure (Lowe and Werner, 2005; Besbas et al., 2006; George, 2009; Scheiring et al., 2010). The differential diagnosis includes sepsis and disseminated intravascular coagulation (DIC), SLE, and TTP. Of note, in both HUS and TTP, other markers of coagulation including PT and aPTT are normal, thus distinguishing these from DIC. Since the anemia is due to hemolysis, the peripheral blood smear will often reveal schistocytes and helmet cells (Lowe and Werner, 2005). Most HUS will have a negative direct Coombs test, with the notable exception of that following S. pneumoniae infection, in which up to 90% may be positive (Banerjee, 2009; Scheiring et al., 2010).

NEUROLOGIC FINDINGS AND SEQUELAE CNS involvement occurs in 20–50% of patients with hemolytic uremic syndrome and is usually seen within the first week of illness (Bale et al., 1980). CNS symptoms may be mild with many patients demonstrating only irritability and lethargy. More severe CNS presentation may be manifested by seizures, stupor, coma, and hemiparesis. Aphasia, ataxia, chorea, dystonia, and visual disturbances may also occur (Hahn et al., 1989). Cerebral edema with headache, vomiting, papilledema,

1116 K. WEBSTER AND E. SCHNITZLER sixth nerve weakness, and altered mental status is comand hearing loss. Dialysis is frequently used as a treatmonly observed. HUS presenting with early CNS signs ment modality for HUS and is associated with improved and symptoms is associated with higher rates of mortalsurvival and prevention of encephalopathy (Stewart and ity as well as neurologic and non-neurologic sequelae Tina, 1993); however, dialysis itself may cause neuro(Upadhyahya et al., 1980). logic abnormalities. Headache is commonly observed The etiology of CNS pathophysiology in HUS is mulduring dialysis in adults, adolescents, and older children. tifactorial but underlying mechanisms closely parallel Headaches may be migraine variants or secondary to those attributed to acute renal failure (ARF). ARF has water intoxication or hypertension. Headache is also a been known to be associated with altered mental status component of the dialysis disequilibrium syndrome for more than a century. Although a significant degree which also includes mental status changes, fatigue, of renal failure can be tolerated without neurologic and muscle cramps. In more severe cases of dialysis diseffects, rapid deterioration of kidney function such as equilibrium syndrome, seizures and coma may develop. occurs in HUS is more likely to result in precipitous alterDialysis disequilibrium syndrome tends to occur early in ation of neurologic status (Eriksson et al., 2001). the course of dialysis and as a complication of rapid dialThe neurologic deterioration seen in HUS with ARF ysis. The underlying pathophysiology is incompletely relates to uremia, hypertension, and altered fluid and understood but seems to be secondary to cerebral edema electrolyte metabolism. Water intoxication is seen early produced by osmotic gradients of urea and other osmotin the course of ARF and is accompanied by hyponatreically active compounds (Rust and Chun, 1999). mia. Early clinical signs of water intoxication include Although hypertension, dialysis equilibrium synheadache and altered mental status which can progress drome, and the metabolic changes of ARF account for rapidly to seizures and coma. Uremia in HUS is primarily a significant proportion of the CNS pathophysiology due to glomerular injury resulting in failure of excretion seen in HUS, other factors appear to be involved. of toxic catabolites of protein. This is accompanied by Dhuna et al. (1992) reported a series of 11 children with acidosis with an increased ion gap, bicarbonate wasting, HUS and seizures. Generalized seizures occurred in four and elevated serum potassium levels. Early uremic patients and partial seizures in seven patients. The chilencephalopathy is characterized by subtle changes in dren with generalized seizures had diffuse slowing on mental status including irritability, inattentiveness, conEEG and normal CT scans of the brain. All four children fusion, memory lapses, lack of interest, fatigue, speech had a normal outcome. However, in the patients with dyspraxia, and mild cognitive impairment. These symppartial seizures, there was diffuse slowing, but focal toms may be followed by movement disorders, particuslowing, focal spikes, asynchronous slowing, and burst larly tremor, tetany, and asterixis. Primitive reflexes suppression patterns were also noted. Six of the seven such as snouting and rooting sometimes occur. Hemiparpatients with focal seizures had clinical findings of hemiesis and transient loss of vision and hearing have also paresis. CT scans in these six patients demonstrated been noted. Uremia may also produce generalized weakstrokes primarily in the basal ganglia and thalamus. ness secondary to neuropathy (Rust and Chun, 1999). All six patients recovered with residual hemiparesis. Seizures occur in up to 40% of children with uremia Thus, partial seizures in HUS were correlated with focal secondary to HUS. These are usually generalized but EEG changes and structural pathology. Strokes secondmay also be focal or myoclonic. Hypertension due to ary to infarctions and microinfarctions seemed to ARF also typically results in an encephalopathy characaccount for these structural findings. terized by generalized seizures as well as headache and Sheth and associates (1986) reviewed 44 children with visual changes. Focal findings including aphasia, hemiHUS seen between 1972 and 1984. Fifteen patients had paresis and particularly sixth nerve palsies have been neurologic complications including 12 who had seizures. described. Occipital blindness is common with hypertenStatistically significant correlations were noted between sive encephalopathy of various etiologies including CNS involvement and metabolic abnormalities including ARF. This is referred to as “reversible posterior leukoenhypocalcemia, hypocapnia, and elevated serum creaticephalopathy” or “occipital-parietal encephalopathy.” nine levels. CSF protein was elevated in four patients, Correlating magnetic resonance imaging (MRI) and but there were none with CSF pleocytosis or decreased computed tomography (CT) findings of white matter CSF glucose. Only six patients with CNS involvement edema without infarction have been noted to accompany recovered completely. Three patients died and six had the clinical loss of vision (Sebire et al., 1995; Hinchey neurologic and/or renal residuals or hypertension. Postet al., 1996). mortem examination of one patient who died showed Early dialysis is effective in prevention and reduction probable cerebral edema but no findings of any vascular of symptoms of uremic encephalopathy as well as microthrombi. Eriksson et al. (2001) summarized EEG improving outcomes in cases of uremic neuropathy data in 22 patients with HUS. They noted that patients

HEMOLYTIC UREMIC SYNDROME 1117 with periodic activity as well as focal and multifocal epineurologic involvement does not necessarily imply poor leptogenic activity had higher rates of mortality, epiprognosis and that basal ganglia involvement in particulepsy, and neurologic sequelae. In particular, focal lar is often associated with full recovery or only mild occipital and temporal slowing and epileptiform disresiduals. Favorable outcome following CNS involvecharges were correlated with residuals of vision impairment even with thrombotic strokes has also been ments and complex partial seizures. The majority of reported by others (Steinberg et al., 1986; Ogura et al., patients with only generalized slowing on EEG recovered 1998; Nakahata et al., 2001). without neurologic complications. Predilection for basal ganglia involvement in HUS Rooney et al. (1971) reported a series of postmortem was corroborated by Steinborn et al. (2004). They neuropathologic findings in HUS patients and did not reviewed MRI and CT scans in 10 patients with HUS describe any unique markers for the disorder. They who had significant CNS involvement. Some 60% had noted cerebral edema and hypoxic changes. In contrast imaging abnormalities in the basal ganglia. AbnormaliUpadhyaya and associates (1980) reviewed 15 patients ties were also seen in the brainstem, cerebellum, and with HUS, three of whom died. Microthrombi were thalami. These signal changes were suggestive of microfound in the kidneys and lungs in all three patients but infarctions. The authors concluded that the basal ganglia also in the brain in two out of the three deceased patients. are commonly affected in HUS and a direct toxic effect The authors speculated that nonrenal multiple organ of verotoxin was again postulated. Furthermore the microthrombi formation seemed to correlate with a imaging findings were often reversible and did not more fulminant course in patients with HUS. necessarily imply an adverse outcome. The presence In a 1980 series, by Bale et al., of 60 patients with HUS, of hemorrhage in a lesion seemed to correlate with subhalf had encephalopathy and/or seizures. CNS involvesequent gliotic or cystic MRI findings as well as residual ment was correlated with azotemia, hyponatremia, and neurologic dysfunction. Two of the MRIs in this series the need for dialysis. Coma on admission and elevated of patients also included diffusion-weighted imaging CSF protein were associated with subsequent mortality (DWI). A case with DWI demonstrating basal ganglia and neurologic morbidity. Only one patient died and lesions was also described by Toldo and associates was autopsied. No microthrombi were seen in the brain. (2009). DWI may be more sensitive in determining neuOnly edema and anoxic changes were noted. rologic prognosis in that reduced apparent diffusion Neuroimaging with CT and MRI has proven to be coefficient (ADC) may correlate with irreversible valuable in the diagnosis, treatment, and prognosis of lesions. A summary of the clinical EEG and neuroimagHUS. Hahn and associates (1989) reported 78 children ing findings in HUS is found in Table 75.2. with HUS; 16 had neurologic complications. CT scans The neuropsychological prognosis of children survivshowed cerebral edema in four patients, large vessel ing HUS has generally been favorable even in patients infarctions in four patients, and multiple hemorrhages who had coma and seizures. Qamar and associates in one patient. At autopsy of three patients, cerebral (1996) reviewed the long-term neurologic and psychoeedema was found but no microthrombi or large vessel ducational outcomes of seven children who had experithrombi were noted. In one patient there was a large enced at least one seizure during their acute presentation hemorrhagic infarction. with HUS. Three children had normal neurologic examIn 1987, DiMario et al. first reported a lacunar infarcinations but four demonstrated mild neurologic abnortion in the basal ganglia as a complication of HUS in a 5malities such as clumsiness, fine motor deficits, year-old girl. Following initial presentation with mild hyperactivity, and distractibility. However, in-depth psyencephalopathy, she developed a left hemiparesis. chometric studies revealed normal IQ scores and behavMRI demonstrated increased T1- and T2-weighted ioral indices in all seven children. In a Canadian images in the consistent with a subacute hemorrhagic multicenter study of 91 case control pairs, Schlieper infarction in the right caudate and lentiform nuclei. and associates (1999) found no significant impact on Barnett and associates (1995) presented two additional cognitive skills, learning, or academic achievement meacases with HUS complicated by coma and dystonic possured at least 6 months after the acute episode of HUS. A turing. MRI and CT scans showed signal changes in the subtle but significant effect of severity of HUS as meabasal ganglia bilaterally. The authors reviewed the litersured by serum creatinine was correlated with lower ature on eight other patients with basal ganglia involvescores on subtests of verbal abilities. However, the inciment. They postulated possible etiologies including dence of attention deficit disorder was no higher in surmicrothrombi secondary to endothelial cell damage vivors of HUS than in age-matched controls. and/or direct effects of the enteric verotoxin. Since the Cimolai and associates (1992) reviewed the risk facmajority of patients recovered, the authors hypothesized tors associated with CNS involvement in HUS in 91 a reversible process. Furthermore they noted that patients. They concluded that younger age and prior

1118

K. WEBSTER AND E. SCHNITZLER

Table 75.2 Clinical EEG and neuroimaging findings in hemolytic uremic syndrome with central nervous system involvement Clinical findings Mild CNS involvement

Prognosis: favorable for recovery

Irritability, lethargy, headache, altered mental status

EEG

CT, MRI

Normal, mild diffuse slowing

Normal, mild cerebral edema

Clinical findings Severe CNS involvement

Prognosis: variable, increased mortality and neurological sequelae

Seizures, stupor, coma, hemiparesis, aphasia, movement disorders, papilledema, visual disturbances, sixth nerve palsy

EEG

CT, MRI

Diffuse slowing, focal slowing, asynchronous slowing, focal spikes burst suppression

Reversible posterior leukoencephalopathy. Infarction and micro-infarctions, primarily in the basal ganglia and thalamus

treatment with gastrointestinal antimotility drugs led to a higher risk of development of HUS in patients infected with Escherichia coli 0157:H7. Using multivariate analysis significant CNS risk factors included female gender, prolonged use of an antimotility drug, and increased hemoglobin levels. Conversely, prior treatment with blood products was associated with a lower risk of neurologic findings. The increased risk factor of female gender applied only to presentation with encephalopathy but not to seizures. The antimotility drugs included opium, codeine, loperamide, diphenoxylate, and anticholinergics. The association of gastrointestinal antimotility agents suggests several possible etiologic mechanisms including more prolonged gastrointestinal absorption of verotoxin. In addition these agents may have direct CNS toxicity which might be further enhanced by impaired renal excretion. It should also be noted that these agents are no longer routinely prescribed by pediatricians for the treatment of diarrhea in infants and young children (Bell et al., 1997).

DIAGNOSIS Diagnosis of typical HUS is usually made clinically based on history and laboratory findings (Table 75.3) (Levandosky et al., 2008; Johnson and Taylor, 2009). All children with diarrheal HUS should have culture of the

stool performed. E. coli 0157:H7 is identified by growth on selective agar within 24 hours. Further testing with PCR or EIA may be done. Although these tests are rapid, they are best performed on colonies already isolated on a culture plate, or from enriched broth that has been incubated. Therefore the earliest confirmed diagnosis still requires 24–36 hours to complete (Centers for Disease Control, 2009). In addition to stool, urine culture for STEC has been reported to be positive, even in the absence of diarrhea. Abdominal ultrasound may be useful in diagnosis during the prodromal phase by identifying thickening of large bowel wall and echogenicity of renal parenchyma (Glatstein et al., 2010). In atypical HUS, a risk factor or deficiency can be identified in approximately 60% of cases (Ariceta et al., 2009). Bacterial culture may help to identify pneumococcal infection as a trigger. For all other causes, serum testing may be of use. Due to the wide range of testing and expense and length of time to diagnosis, a prioritized and stepwise approach to diagnosis is recommended (Table 75.4); however, if therapy with plasma infusion or pheresis is being considered, it is necessary to obtain serum samples prior to initiation of therapy (Banerjee, 2009) or wait a minimum of 2 weeks following plasma infusion (Kavanagh et al., 2007). The most common etiology of atypical HUS is complement deficiency (Besbas et al., 2006; Ariceta et al., 2009). Serum

HEMOLYTIC UREMIC SYNDROME

1119

Table 75.3 Diagnosis of typical hemolytic uremic syndrome History

Laboratory study

Diarrhea within past 2 weeks Age > 6 months Endemic area Exposure to contaminated food Abrupt onset Single episode Concurrent affected contact

Classic features Chemistry panel CBC Hemolytic studies Peripheral smear

Etiologic studies Stool culture Selective agar for E. coli 0157 Serotyping of isolates PCR for Stx1, Stx2

LDH Haptoglobin Coagulation studies PT, PTT Fibrinogen D-dimer Red cell production Reticulocyte count

Findings

Elevated BUN Elevated creatinine Anemia Thrombocytopenia Schistocytes Helmet cells Burr cells Elevated Decreased

Normal Normal Elevated Elevated

PCR, polymerase chain reaction; Stx, Shiga toxin; CBC, complete blood count; LDH, lactate dehydrogenase; PT, prothrombin time; PTT, partial thromboplastin time; BUN, blood urea nitrogen.

Table 75.4 Approach to diagnostic and etiologic work-up in atypical hemolytic uremic syndrome Clinical factor

Possible etiology

Suggested testing

Meningitis, pneumonia

Pneumococcal infection

Age < 6 months, failure to thrive, neurologic symptoms Other atypical HUS

Deficient cobalamin metabolism

Culture of affected site Direct Coombs test Urine and serum amino acids C3, C4 levels Factor H, B, I levels MCP expression ADAMTS13 activity

Neurologic symptoms, family history, insidious onset

Complement deficiency

TTP

HUS, hemolytic uremic syndrome; TTP, thrombotic thrombocytopenic purpura; MCP, membrane cofactor protein; ADAMTS13, a disintegrin and metalloproteinase with a thrombospondin type 1 motif, member 13.

assays for levels of C3 along with complement factors H, I and B and MCP should be obtained. If a low level is found, genetic testing or autoantibody assays may also be considered (Kavanagh et al., 2007). In children with coexisting failure to thrive and prominent neurologic symptoms, assessment of urine and serum amino acids may reveal congenital cobalamin deficiency (Ariceta et al., 2009; Banerjee, 2009). For patients in whom TTP is suspected, activity of the enzyme ADAMTS13 may be analyzed. Activity < 5% is reported to be specific for TTP (George, 2009). Decreased

activity < 10% is suggestive but not specific for TTP. Gene mutations versus autoantibodies are still a consideration. Finally, both HUS and TTP can be triggered by autoimmune disease, so evaluation of antinuclear antibody, lupus anticoagulant, and antiphospholipid antibody are also important (Ariceta et al., 2009). Renal biopsy is not needed for diagnosis; however, it is sometimes undertaken, especially in severe renal failure or patients with recurrent disease. Disctinctive findings on biopsy may help differentiate HUS from TTP (Banerjee, 2009).

1120

K. WEBSTER AND E. SCHNITZLER

TREATMENT The mainstay of treatment for typical HUS is supportive care. Volume resuscitation, fluid and electrolyte monitoring, and transfusion of blood products are common, as is temporary need for dialysis (Lowe and Werner, 2005; Scheiring et al., 2008; Pomajzl et al., 2009). Treatment of neurologic manifestations is most often associated with control of seizure activity, with care to address any electrolyte imbalance as a potential cause.

Antibiotics Antibiotic therapy is necessary in infections due to Shigella or S. pneumoniae. In other causes of diarrheal HUS, there have been reports of worsened outcomes with antibiotic treatment. A meta-analysis of studies in which antibiotics were used was unable to demonstrate conclusively that harm occurs, although wide variation in treatments was observed (Safdar et al., 2002; Banerjee, 2009; Mohsin et al., 2010).

Cobalamin replacement In cases of cobalamin deficiency, supplementation may help to prevent future episodes (Banerjee, 2009). Unfortunately, despite therapy this disorder is often associated with progression of severe neurologic deficits due to the underlying homocysteine accumulation (Martinielli et al., 2011).

Plasma infusion and plasmapheresis Plasma infusion has been reported to be of benefit in some patients with atypical HUS or TTP. The goal of plasma infusion is to replace complement factors or ADAMTS13 enzyme that are deficient in the afflicted patient. In patients who have deficiencies, marked improvement is reported (Banerjee, 2009). Since many of these deficiencies may be caused not only by decreased production but by destruction via autoantibodies, plasma infusion alone may in some cases be harmful. HUS caused by pneumococcal infection (Banerjee, 2009) and by complement antibodies (Boyer et al., 2010) have both been worsened by plasma infusion. Because the underlying etiology is often unknown at the time of presentation, initial empiric therapy should be plasma exchange to remove the offending antibody. Plasma infusion may be useful in preventing recurrence, once the underlying deficiency has been identified (Kohli and Gulati, 2006; Loirat et al., 2010).

Immunotherapy Plasma exchange is likely to be effective in only 30–50% of patients with atypical HUS (Ariceta et al., 2009; Banerjee, 2009). Some patients may show an initial

response but this response is not sustained and/or repeated lifelong prophylactic treatment is needed. In such patients, reports of treatment with corticosteroids, vincristine, azathioprine, and cyclophosphamide have reported anecdotal success (Michael et al., 2009; Boyer et al., 2010). Often a combination of both immunosuppression and intermittent plasma exchange is necessary to sustain remission in severe relapsing forms of the disease. Inhibition of complement may be effective in aHUS due to complement protein deficiencies. Eculizumab and rituximab have been reported as effective therapies in both atypical HUS (Banerjee, 2009; Mache et al., 2009; Boyer et al., 2010; K€ose et al., 2010) and chronic relapsing TTP (Levandovsky et al., 2008). Currently, investigations are ongoing involving urtoxazumab, a targeted antibody therapy to neutralize Shiga toxin (Lopez et al., 2010). While this may help prevent the cascade of events leading to full HUS, the challenge will be providing therapy within 24–72 hours of exposure (Bitzan et al., 2010). Further investigation of this therapy is needed to provide more conclusive evidence of effect.

Transplantation Patients with aHUS may progress to renal failure and ultimately require transplantation. Unfortunately, as aHUS is most commonly associated with defective or deficient complement protein, the risk of disease recurrence or graft rejection is high (Cheong, 2009).

Complement therapy In variations of HUS associated with deficiency of complement regulatory proteins, replacement of these factors would be beneficial and more specific than plasma infusion alone. While no specific factor is currently available, work is ongoing to develop recombinant or concentrate of these important complement factors (Johnson and Taylor, 2008). Overall, the vast array of etiology and pathophysiology that underlies HUS creates a clinical challenge, with careful attention to presentation, family history, and diagnosis in order to achieve the proper diagnosis and therapy.

REFERENCES Ariceta G, Besbas N, Johnson S et al. (2009). Guideline for the investigation and initial therapy of diarrhea-negative hemolytic uremic syndrome. Pediatr Nephrol 24: 687–696. Bale JF Jr, Brasher C, Siegler RL (1980). CNS manifestations of the hemolytic-uremic syndrome. Relationship to

HEMOLYTIC UREMIC SYNDROME metabolic alterations and prognosis. Am J Dis Child 134: 869–872. Banerjee S (2009). Hemolytic uremic syndrome. Indian Pediatr 46: 1075–1084. Barit G, Sakarcan A (2005). Antibiotic resistant Streptococcus pneumoniae and hemolytic uremic syndrome. Eur J Pediatr 164: 414–416. Barnett ND, Kaplan AM, Bernes SM et al. (1995). Hemolytic uremic syndrome with particular involvement of basal ganglia and favorable outcome. Pediatr Neurol 12: 155–158. Bell BP, Griffin PM, Lozano P et al. (1997). Predictors of hemolytic uremic syndrome in children during a large outbreak of Escherichia coli O157:H7 infections. Pediatrics 100: E12. Besbas N, Karpman D, Landau D et al. (2006). A classification of hemolytic uremic syndrome and thrombotic thrombocytopenic purpura and related disorders. Kidney Int 70: 423–431. Bitzan M, Schaefer F, Reymond D (2010). Treatment of typical (enteropathic) hgemolytic uremic syndrome. Semin Thromb Hemost 36: 594–610. Bouts AH, Roofthooft MTR, Salomous GS et al. (2010). CD46-associated atypical hemolytic uremic syndrome with uncommon course caused by cblC deficiency. Pediatr Nephrol 25: 2547–2548. Boyer O, Balzamo E, Charbit M et al. (2010). Pulse cyclophosphamide therapy and clinical remission in atypical hemolytic uremic syndrome with anti-complement factor H autoantibodies. Am J Kidney Dis 55: 923–927. Brandt J, Wong C, Mihm S et al. (2002). Invasive pneumococcal disease and hemolytic uremic syndrome. Pediatrics 110: 371–376. ¨ et al. (2011). Hemolytic ureC¸altik A, Aky€ uz SG, Erdogan O mic syndrome triggered with a new pandemic virus: influenza A (H1N1). Pediatr Nephrol 26: 147–148. Centers for Disease Control and Prevention (2009). Morbidity and Mortality Weekly Report: Recommendations for diagnosis of Shiga toxin-producing Escherichia coli infections by clinical laboratories. October 16, 2009, Vol. 58, No. RR-12, pp. 1–14. Cerda J, Lameire N, Eggers P et al. (2008). Epidemiology of acute kidney injury. Clin J Am Soc Nephrol 3: 881–886. Chaturvedi S, Licht C, Langlois V (2010). Hemolytic uremic syndrome caused by Bordetella pertussis infection. Pediatr Nephrol 25: 1351–1364. Cheong H (2009). Can liver-kidney transplantation cure aHUS? Nephrology 5: 556–557. Cimolai N, Morrison BJ, Carter JE (1992). Risk factors for the central nervous system manifestations of gastroenteritisassociated hemolytic-uremic syndrome. Pediatrics 90: 616–621. Desch K, Motto D (2007). Is there a shared pathophysiology for thrombotic thrombocytopenic purpura and hemolyticuremic syndrome? J Am Soc Nephrol 18: 2457–2460. Dhuna A, Pascual-Leone A, Talwar D et al. (1992). EEG and seizures in children with hemolytic-uremic syndrome. Epilepsia 33: 482–486. DiMario FJ Jr, Br€onte-Stewart H, Sherbotie J et al. (1987). Lacunar infarction of the basal ganglia as a complication

1121

of hemolytic-uremic syndrome: MRI and clinical correlations. Clin Pediatr 25: 586. Eriksson KJ, Boyd SG, Tasker RC (2001). Acute neurology and neurophysiology of haemolytic-uraemic syndrome. Arch Dis Child 84: 434–435. Gasser C, Gautier E, Steck A et al. (1955). Hemolytic-uremic syndrome: bilateral necrosis of the renal cortex in acute acquired hemolytic anemia. Schweiz Med Wochenschr 85: 905–909. George JN (2009). The thrombotic thrombocytopenic purpura and hemolytic uremic syndromes: overview of pathogenesis (experience of the Oklahoma TTP-HUS Registry, 1989–2007). Kidney Int 75 (Suppl 112): S8–S10. Glatstein M, Miller E, Garcia-Boumissen F et al. (2010). Timing and utility of ultrasound in diarrhea-associated hemolytic uremic syndrome: 7-year experience of a large tertiary care hospital. Clin Pediatr 49: 318. Gould LH, Demma L, Jones TF et al. (2009). Hemolytic uremic syndrome and death in persons with Escherichia coli O157:H7 infection, Foodborne Diseases Active Surveillance Network Sites, 2000–2006. Clin Infect Dis 49: 1480–1485. Hahn JS, Havens PL, Higgins JJ et al. (1989). Neurological complications of hemolytic-uremic syndrome. J Child Neurol 4: 108–113. Hinchey J, Chaves C, Appigani B et al. (1996). A reversible posterior leukoencephalopathy syndrome. N Engl J Med 334: 494. Johnson S, Taylor CM (2008). What’s new in haemolytic uraemic syndrome? Eur J Pediatr 167: 965–971. Johnson S, Taylor CM (2009). In: ED Ainer, D Ellis (Eds.), Pediatric Nephrology. 6th edn. Springer, pp. 1155–1180. Karpman D, Sartz L, Johnson S (2010). Pathophysiology of typical hemolytic uremic syndrome. Semin Thromb Hemost 36: 575–585. Kavanagh D, Richards A, Fremeaux-Bacchi V et al. (2007). Screening for complement system abnormalities in patients with atypical hemolytic uremic syndrome. Clin J Am Soc Nephrol 2: 591–596. Kohli R, Gulati S (2006). Plasma infusion therapy in atypical hemolytic uremic syndrome – long term outcome. Indian Pediatr 43: 164–166. ¨ , Zimmerhackl L-B, Jungraithmayr T et al. (2010). New K€ ose O treatment options for atypical hemolytic uremic syndrome with the complement inhibitor eculizumab. Semin Thromb Hemost 36: 669–672. Lei T-H, Hsia S-H, Wu C-T et al. (2010). Streptococcus pneumoniae-associated haemolytic uremic syndrome following influenza A virus infection. Eur J Pediatr 169: 237–239. Levandosky M, Harvey D, Lara P et al. (2008). Thrombotic thrombocytopenic purpura-hemolytic uremic syndrome (TTP-HUS): a 24-year clinical experience with 178 patients. J Hematol Oncol 1: 23 http://www.jhoonline.org/ content/1/1/23. Loirat C, Noris M, Fremeaux-Bacchi V (2008). Complement and the atypical hemolytic uremic syndrome in children. Pediatr Nephrol 123: 1957–1972.

1122

K. WEBSTER AND E. SCHNITZLER

Loirat C, Garnier A, Sellier-Leclerc A-L et al. (2010). Plasmatherapy in atypical hemolytic uremic syndrome. Semin Thromb Hemost 36: 673–681. Lopez EL, Contrini MM, Glatstein E et al. (2010). Safety and pharmacokinetics of urtoxazumab, a humanized monoclonal antibody, against Shiga-like toxin 2 in healthy adults and in pediatric patients infected with Shiga-like toxin-producing Escherichia coli. Antimicrob Agents Chemother 54: 239–243. Lowe EJ, Werner EJ (2005). Thrombotic thrombocytopenic purpura and hemolytic uremic syndrome in children and adolescents. Semin Thromb Hemost 31: 717–729. Mache CJ, Acham-Roschitz B, Fremeaux-Bacchi V et al. (2009). Complement inhibitor eculizumab in atypical hemolytic uremic syndrome. Clin J Am Soc Nephrol 4: 1312–1316. Martinelli D, Deodato F, Dionisi-Vici C (2011). Cobalamin C defect: natural history, pathophysiology, and treatment. J Inherit Metab Dis 34: 127–135. Melmann A, Fruth A, Friedrich AW et al. (2009). Phylogeny and disease association of Shiga toxin-producing Escherichi coli O91. Emerg Infect Dis 15: 1474–1477. Michael M, Elliott EJ, Craig JC et al. (2009). Interventions for hemolytic uremic syndrome and thrombotic thrombocytopenic purpura: a systematic review of randomized controlled trials. Am J Kidney Dis 53: 259–272. Moake JL (2002). Thrombotic microangiopathies. N Engl J Med 347: 589–600. Mohsin M, Haque A, Ali A et al. (2010). Effects of ampicillin, gentamicin, and cefotaxime on the release of Shiga toxins from Shiga toxin-producing Escherichia coli isolated during a diarrhea episode in Faisalabad, Pakistan. Foodborne Pathog Dis 7: 85–90. Nakahata T, Tanaka H, Tateyama T et al. (2001). Thrombotic stroke in a child with diarrhea-associated hemolytic-uremic syndrome with a good recovery. Tohoku J Exp Med 193: 73–77. Nolasco LH, Turner NA, Bernardo A et al. (2005). Hemolytic uremic syndrome-associated Shiga toxins promote endothelial-cell secretion and impair ADAMTS13 cleavage of unusually large von Willebrand factor multimers. Blood 106: 4199–4209. Ogura H, Takaoka M, Kishi M et al. (1998). Reversible MR findings of hemolytic uremic syndrome with mild encephalopathy. AJNR Am J Neuroradiol 19: 1144–1145. Pomajzl RJ, Varman M, Holst A et al. (2009). Hemolytic uremic syndrome (HUS) – incidence and etiologies at a regional Children’s Hospital in 2001–2006. Eur J Clin Microbiol Infect Dis 28: 1431–1435. Printza N, Roilides E, Kotsiou M et al. (2011). Pandemic influenza A (H1N1) 2009-associated hemolytic uremic syndrome. Pediatr Nephrol 26: 143–144. Qamar IU, Ohali M, MacGregor DL et al. (1996). Long-term neurological sequelae of hemolytic-uremic syndrome: a preliminary report. Pediatr Nephrol 10: 504–506.

Quintrec ML, Roumenina L, Noris M et al. (2010). Atypical hemolytic uremic syndrome associated with mutations in complement regulator genes. Semin Thromb Hemost 36: 641–652. Razzaq S (2006). Hemolytic uremic syndrome: an emerging health risk. Am Fam Physician 74: 991–996. Rivero MA, Passucci JA, Rodriguez EM et al. (2010). Role and clinical course of verotoxigenic Escherichia coli infections in childhood acute diarrhoea in Argentina. J Med Microbiol 59: 345–352. Rooney JC, Anderson RM, Hopkins IJ (1971). Clinical and pathologic aspects of central nervous system involvement in the haemolytic uraemic syndrome. Proc Aust Assoc Neurol 8: 67–75. Ruggenenti P, Noris M, Remuzzi G (2001). Thrombotic microangiopathy, hemolytic uremic syndrome, and thrombotic thrombocytopenic purpura. Kidney Int 60: 831–846. Rust R, Chun R (1999). Interrelationships between renal and neurologic diseases and therapies. In: KF Swaiman, S Ashwal (Eds.), Pediatric Neurology Principles and Practice. 3rd edn. Mosby, St Louis, pp. 1403–1437. Rust RS, Worrel TE (2009). Hemolytic uremic syndrome. eMedicine Neurology 1–30. Safdar N, Said A, Gangon RE et al. (2002). Risk of hemolytic uremic syndrome after antibiotic treatment of Escherichia coli O157:H7 enteritis: a meta-analysis. JAMA 288: 996–1001. Scheiring J, Andreoli SP, Zimmerhackl LB (2008). Treatment and outcome of Shiga-toxin-associated hemolytic uremic syndrome. Pediatr Nephrol 23: 1749–1760. Scheiring J, Rosales A, Zimmerhackl LB (2010). Today’s understanding of the haemolytic uraemic syndrome. Eur J Pediatr 169: 7–13. Schlieper A, Orrbine E, Wells GA et al. (1999). Neuropsychological sequelae of haemolytic uraemic syndrome. Arch Dis Child 80: 214–220. Schneppenheim R, Budde U, Hassenpflug W et al. (2004). Severe ADAMTS-13 deficiency in childhood. Semin Hematol 41: 83–89. Sebire G, Husson B, Lasser C et al. (1995). Encephalopathy induced by arterial hypertension: clinical, radiological and therapeutic aspects. Arch Pediatr 2: 513. Sharma J, Bharadawa K, Shah K et al. (1993). Plasmodium vivax malaria presenting as hemolytic-uremic syndrome. Indian Pediatr 30: 369–371. Sharma AP, Greenberg CR, Prasad AN et al. (2007). Hemolytic uremic syndrome secondary to cobalamin C C (Cb1c) disorder. Pediatr Nephrol 22: 2097–2103. Sheth KJ, Swick HM, Haworth N (1986). Neurological involvement in hemolytic-uremic syndrome. Ann Neurol 19: 90–93. Steinberg A, Ish-Horowitcz M, el-Peleg O et al. (1986). Stroke in a patient with hemolytic-uremic syndrome with a good outcome. Brain Dev 8: 70–72.

HEMOLYTIC UREMIC SYNDROME Steinborn M, Leiz S, R€ udisser K et al. (2004). CT and MRI in haemolytic uraemic syndrome with central nervous system involvement: distribution of lesions and prognostic value of imaging findings. Pediatr Radiol 34: 805–810. Stewart CL, Tina LU (1993). Hemolytic uremic syndrome. Pediatr Rev 14: 218–224.

1123

Toldo I, Manara R, Cogo P et al. (2009). Diffusion-weighted imaging findings in hemolytic uremic syndrome with central nervous system involvement. J Child Neurol 24: 247–250. Upadhyahya K, Barwick K, Fishaut M et al. (1980). The importance of non-renal involvement in hemolytic uremic syndrome. Pediatrics 65: 115–120.

Handbook of Clinical Neurology, Vol. 120 (3rd series) Neurologic Aspects of Systemic Disease Part II Jose Biller and Jose M. Ferro, Editors © 2014 Elsevier B.V. All rights reserved

Chapter 76

Commonly used drugs in hematologic disorders ELISE ANDERES AND SUCHA NAND* Division of Hematology and Oncology, Department of Medicine, Loyola University Chicago, Stritch School of Medicine, Maywood, IL, USA

AGENTS USED TO TREAT BENIGN HEMATOLOGIC DISORDERS Replacement therapies ANEMIAS RESULTING FROM IRON AND VITAMIN DEFICIENCIES

Anemias are commonly encountered in medical practice and are often due to deficiencies of iron and vitamins such as cobalamin (vitamin B12), or folic acid. Iron deficiency is the most common cause of anemia in the US and worldwide (Umbreit, 2005). Iron deficiency can be treated orally, parenterally, or with blood transfusion. Oral iron is the simplest and least expensive option. Ferrous iron salts are preferred due to increased solubility at the pH of the duodenum and jejunum. Approximately 200 mg of elemental iron per day is required to replete iron stores. Each 325 mg tablet of ferrous sulfate contains 66 mg of elemental iron; therefore one tablet three times daily is the recommended replacement dose. The most common side-effects of oral iron sulfate include nausea, heartburn, constipation, or loose stools. Oral iron is best absorbed on an empty stomach, but is best tolerated with foods. The majority of patients will tolerate therapy without significant side-effects. Reducing the frequency of administration or taking iron supplements with meals can alleviate gastrointestinal complaints (Cook, 2005; Killip et al., 2007). Parenteral iron replacement is indicated in patients with iron malabsorption due to resection or disease of the stomach or bowel, conditions with high iron requirements such as chronic gastrointestinal blood loss or endstage renal disease, and failure of oral iron replacement due to poor tolerance or compliance (Cook, 2005). Parenteral replacement is best given intravenously, since the

intramuscular route has been associated with development of soft tissue sarcomas. Formulations available in the US include iron dextran, iron sucrose, and iron gluconate. All are associated with the risk of lifethreatening anaphylaxis, arthralgias, fever, and hypotension. Iron sucrose and iron gluconate have a lower incidence of anaphylaxis and are generally preferred, although they are more expensive (Silverstein, 2004). Headaches and dizziness are associated with intravenous iron preparations. Paresthesias and syncope have also been reported with iron gluconate infusions. Pleocytosis of the cerebrospinal fluid following a febrile reaction to iron dextran has been reported. The patient also developed a peripheral blood leukocytosis (Forristal and Witt, 1968). In another patient, meningismus without increased leukocytes in the spinal fluid but a high spinal fluid iron concentration was documented (Wallerstein, 1968). Iron sucrose and iron gluconate have a lower incidence of anaphylaxis and are generally preferred, although they are more expensive (Silverstein, 2004). Megaloblastic anemias most commonly result from deficiencies of cobalamin or folic acid. Folate deficiency can occur due to decreased intake secondary to poor nutrition, impaired absorption in the duodenum or jejunum due to tropical or nontropical sprue, or increased requirement due to pregnancy or hemolytic anemia. Various drug interactions can also lead to folate deficiency. Decreased dietary intake is the most common cause of folate deficiency and is often seen in alcoholics or elderly or poor patients. The recommended daily intake of dietary folate is 400 mg daily and inadequate consumption leads to anemia within several months. Folate deficient persons are treated with 1–5 mg daily of oral folate. Changes in mental status, sleep disturbances, irritability, and excitability have been reported with higher

*Correspondence to: Sucha A. Nand, MD, Loyola University Medical Center, Cancer Center, Room 345, 2160 S. First Avenue, Maywood, IL 60153, USA. E-mail: [email protected]

1126

E. ANDERES AND S. NAND

doses of folate such as 15 mg/day (Hunter et al., 1970). Folic acid was also reported to exacerbate seizure activity in a young woman who started supplementation while attempting to become pregnant. She had a history of monthly seizures not controlled by medication, but her seizures became more frequent and more severe after starting folate 0.8 mg/day. Substitution of folinic acid 7.5 mg/day resulted in improvement of her seizure activity (Guidolin et al., 1998). Concomitant cobalamin deficiency should be excluded prior to beginning treatment with folate, as anemia may improve but neurologic symptoms due to cobalamin deficiency will progress. Folate replacement is inexpensive and effective even in persons with malabsorption (Krishnaswamy and Nair, 2001). Cobalamin deficiency is most commonly caused by impaired absorption secondary to pernicious anemia. Other causes include gastric or ileal resection, regional enteritis, intestinal lymphoma, bacterial overgrowth in blind intestinal loops, and vegan diet (Babior, 2006). Cobalamin deficiency can result in severe neuropsychiatric complications as well as anemia or pancytopenia. Cobalamin is usually given by intramuscular injections, which saturate tissue stores and compensate for daily losses. Oral and intranasal preparations are also available. In symptomatic patients or those with severe cytopenias, 1000 mg of cobalamin is typically given intramuscularly every day for 2 weeks, then weekly until cytopenias resolve, and then monthly indefinitely. For those with subclinical deficiency (cobalamin levels between 200 and 350 ng/L), replacement with daily injections for 1 week, followed by weekly injections for 4 weeks, then monthly injections indefinitely is acceptable. Oral replacement with 1000–2000 mg daily is equally effective in most cases of cobalamin deficiency (Oh and Brown, 2003), though patients should be monitored closely to ensure that laboratory parameters are correcting and the cobalamin levels are being repleted. For those who respond to oral replacement, lifelong therapy with 1000 mg daily is indicated. For nonresponders, parenteral replacement should be given. Excess cobalamin is excreted in the urine, so toxicity from excess vitamin replacement does not occur. Asthenia, dizziness, and headache have been reported after cobalamin injections in approximately 12% of patients. This may be related to the method of administration. A summary of the agents used to treat the various hematologic disorders discussed in this chapter is given in Table 76.1.

COAGULOPATHIES Fresh frozen plasma Fresh frozen plasma (FFP) is obtained from units of whole blood donation or from plasmapheresis of

Table 76.1 Commonly used drugs in hematologic disorders 1. Agents used to treat benign hematologic disorders A. Replacement therapies Anemias resulting from iron and vitamin deficiencies Coagulopathies B. Antifibrinolytic agents Lysine analogs C. Antiplatelet agents Cyclooxygenase inhibitors Adenosine diphosphate receptor inhibitors Glycoprotein IIb/IIIa antagonists Thromboxane synthase inhibitors D. Antithrombotic agents Unfractionated heparin Low molecular weight heparin Factor Xa inhibitors Warfarin Direct thrombin inhibitors 2. Management of hemorrhagic complications of anticoagulation A. Intracranial hemorrhages due to Warfarin Heparin Direct thrombin inhibitors Antiplatelet agents 3. Agents used to treat hematologic malignancies A. Chronic myeloproliferative disorders Hydroxyurea (hydroxycarbamide) Anagrelide BCR-ABL tyrosine kinase inhibitors B. Myelodysplasia DNA methyltransferase inhibitors C. Leukemias, lymphomas and multiple myeloma Chemotherapeutic agents Targeted therapies a. Monoclonal antibodies b. Radioimmunotherapy c. Immunomodulatory drugs d. Proteosome inhibitors

volunteer donors. FFP can be used for replacement of factors II, V, VII, IX, X, or XI, and protein S, since specific factor replacement are often not available. Thus, FFP is most commonly used in the treatment of multiple factor deficiencies, such as in patients with disseminated intravascular coagulation (DIC), patients on warfarin with significant bleeding, those with vitamin K deficiency requiring urgent correction of factor deficiencies, patients with bleeding associated with acute blood loss, or those requiring plasma exchange for treatment of thrombotic thrombocytopenic purpura (TTP) or hyperviscosity syndrome. A typical unit of plasma derived from a collection of whole blood has a volume of nearly 300 mL, and local and national guidelines for usage

COMMONLY USED DRUGS IN HEMATOLOGIC DISORDERS 1127 generally specify a dose of around 10–20 mL/kg anti-inflammatory drugs (NSAIDs), antihistamines, or (Stanworth, 2007). intravenous glucocorticoids may also be helpful (Reutter and Luger, 2004). Headache is the most comCryoprecipitate mon side-effect of IVIg therapy, with reported incidence ranging from 5–80%. Usually the headaches are mild Cryoprecipitate is a relatively concentrated preparation and can be alleviated by slowing the infusion rate and of procoagulant factors, including fibrinogen, factor giving analgesics or antihistamines. However, in some VIII, von Willebrand factor, factor XIII, and fibronecpatients IVIg therapy must be terminated because of tin. Cryoprecipitate is most commonly used for replacesevere headaches (Orbach et al., 2005). Other adverse ment of acquired or congenital hypofibrinogenemia. An effects associated with IVIg administration include renal adult dose of around 10 single bags of cryoprecipitate failure, arterial and venous thrombosis, and dermatoderived from units of whole blood typically raises the logic toxicity, such as urticaria, rash, and pruritus plasma fibrinogen level by up to 1 g/L (60–100 mg/dL) (Orbach et al., 2005). Thrombotic events occur in 1–13% (Stanworth, 2007). It can also be used in treatment of of patients, with arterial thromboses (stroke, myocardial von Willebrand disease or hemophilia, though specific infarction) being more common than venous (pulmonary replacement with von Willebrand factor or factors VIII embolism, deep venous thrombosis). Arterial events also and IX are usually preferred. Patients with DIC and low tend to occur earlier, with 50% of events occurring fibrinogen are probably best treated with a combination within 4 hours of the IVIg infusion. Older age and carof FFP and cryoprecipitate, to minimize the risk of inducdiovascular risk factors increase the risk of arterial ing thrombosis with transfusion of cryoprecipitate thromboses, while obesity and immobility increase the alone. Adequate transfusion should be given to maintain risk of venous thromboses (Paran et al., 2005). Premedithe fibrinogen level above 100 mg/dL. Cryoprecipitate cation with aspirin may decrease the risk of thromboses can also be used for treatment of qualitative platelet dysin patients with underlying risk factors. Hyperviscosity function in uremia. may also increase the risk of stroke, and a baseline viscosity should be checked in patients at risk for Immunoglobulin hyperviscosity (e.g., monoclonal gammopathy) (Marie Intravenous immunoglobulin (IVIg) is derived from et al., 2006). A significant proportion of patients who large pools of human plasma. Most of the immunoglobreceive IVIg develop a positive direct antigobulin test ulin in commercially available preparations of IVIg is (DAT, or direct Coombs test), due to the presence of IgG, with a subtype distribution of IgG1–IgG4 similar anti-A or anti-B derived from type O individuals in the to that in normal human plasma. Relatively small donor pools. Transient hemolysis has been reported amounts of IgA and IgM are also present. IVIg is used (Copelan et al., 1986). to treat a variety of hematologic disorders, including Aseptic meningitis is a rare complication of IVIg congenital or acquired immunodeficiency syndromes, treatment, occurring in about 1% of patients. Clinical immune thrombocytopenic purpura (ITP), autoimmune manifestations include acute severe headache with neck hemolytic anemia (AIHA), autoimmune neutropenia rigidity, fever, lethargy, photophobia, nausea, and (AIN), and recurrent bacterial infections occurring in vomiting. Cerebrospinal fluid (CSF) examination demassociation with chronic lymphocytic leukemia (CLL) onstrates pleocytosis with high protein content and negor multiple myeloma. IVIg is also used to treat a variety ative culture. Signs and symptoms usually begin 48 hours of autoimmune disorders. In patients with ITP and after the infusion and last for 3–5 days (Orbach et al., AIHA, IVIg is considered the best “emergency” inter2005). Infusing IVIg at a slow rate and ensuring adevention when a rapid, albeit transient, response is quate hydration may prevent or reduce the incidence required (Knezevic-Maramica and Druskall, 2003). of aseptic meningitis. In patients with a history of The neurologic indications for the use of IVIg include IVIg-induced aseptic meningitis, premedication with Guillain–Barre´ syndrome and myasthenia gravis, among acetaminophen or antihistamines may reduce the inciothers. dence of recurrence. Corticosteroid treatment is not conThe immediate adverse reactions following IVIg sidered beneficial in IVIg-induced aseptic meningitis administration are usually mild and transient flu-like (Redman et al., 2002). symptoms, and include headache, facial flushing, malaise, chest tightness, fever and chills, myalgia, fatigue, Transfusion reactions and risks dyspnea, nausea, vomiting, diarrhea, change in blood pressure, and tachycardia. These side-effects usually Transfusion of plasma and plasma fractions can lead to resolve if the infusion rate is decreased (Orbach et al., adverse reactions or events, of which immune-mediated 2005). Premedication with analgesics, nonsteroidal reactions are most common. These include allergic and

1128

E. ANDERES AND S. NAND

anaphylactic reactions, transfusion-related acute lung injury (TRALI) and hemolysis, and can range in severity from mild to fatal. TRALI is a leading cause of transfusion-related morbidity and mortality. It is characterized by acute noncardiogenic pulmonary edema and respiratory compromise in the setting of transfusion. TRALI is caused by passive transfusion of antigranulocyte antibodies that interact with recipient neutrophils, resulting in activation and aggregation in pulmonary capillaries, release of local biologic response modifiers causing capillary leak, and lung injury (Triulzi, 2006). Signs and symptoms include hypoxemia, hypotension, dyspnea, fever, and bilateral infiltrates on chest radiograph. Fluid overload and citrate toxicity can occur after rapid or massive transfusion. In developed countries, microbial transmission rates are low because of donor selection and testing. The risk of viral transmission is very low because of the use of two independent viral inactivation steps. The risk of transmission of variant Creutzfeldt–Jakob disease in both plasma components and pooled plasma products is as yet unknown. The low titer of prion infectivity in the blood of an infected individual (approximately 10 infectious units/mL) would be massively diluted by the thousands of units of plasma in the pool, likely making the risk extremely low. Subsequent manufacturing processes also remove prions from the final product (MacLennan and Barbara, 2006).

Recombinant factor VIIa Recombinant factor VIIa (rfVIIa or NovoSeven®) is a procoagulant protein concentrate that was developed to “bypass” factor VIII or IX inhibitors in patients with hemophilia A or B. It is approved in these patients for the treatment of bleeding episodes, or for prophylaxis prior to invasive procedures or surgery. It is also approved in these settings in patients with congenital factor VII deficiency. RfVIIa is occasionally used off-label in patients with trauma and massive hemorrhage or in patients with platelet disorders or liver disease and uncontrolled bleeding. Recombinant factor VIIa binds directly to activated platelets and activates factor X, which in turn catalyzes the conversion of prothrombin to thrombin. Thrombin generation activates the intrinsic pathway and further promotes thrombin formation. Binding of rfVIIa to activated platelets localizes it to the site of bleeding and helps prevent thrombotic complications (Roberts et al., 2004). The incidence of serious adverse events, including myocardial infarction, stroke and venous thromboembolism, is about 1% in hemophiliacs (Abshire and Kenet, 2004). RfVIIa should be used cautiously in patients with a predisposition to thrombotic complications, such as obesity, cancer, or cardiovascular disease.

Prothrombin complex concentrates Prothrombin complex concentrates (PCCs) are a source of the vitamin K-dependent coagulation factors, including factors II, VII, IX and X and proteins C and S. They are isolated from the cryoprecipitate supernatant of large plasma pools after removal of antithrombin and factor XI. The PCCs are standardized according to their factor IX content. The concentrates are processed to inactivate the clotting factors and treated to inactivate transfusion-transmitted viruses. PCC administration is indicated for emergent reversal of oral anticoagulant therapy, for example in patients on warfarin with intracerebral hemorrhage. PCCs can also be used in the treatment of hemophilia B or congenital factor VII deficiency when specific factor concentrates are not available. Adverse events associated with PCC use include allergic reactions, heparininduced thrombocytopenia (when heparin is added to the PCC to inactivate clotting factors), and DIC (Hellstern, 1999).

Antifibrinolytic agents LYSINE ANALOGS Fibrinolysis occurs when plasmin that is generated by plasminogen activators digests fibrin clots. Both plasmin and plasminogen bind thrombin through lysine-binding sites. The synthetic lysine analogs Eaminocaproic acid (EACA, Amicar®) and tranexamic acid (AMCA) compete with plasmin and plasminogen activators at lysine binding sites. Binding of these agents inhibits fibrinolysis and stabilizes the clot. These agents can be administered orally, intravenously or topically, although optimal dosing has not been established in most clinical settings (Verstraete, 1985). They are used in the treatment of severe mucosal hemorrhage (e.g., upper gastrointestinal bleeding, menorrhagia) or other bleeding conditions associated with increased fibrinolysis. EACA and AMCA are also used to control bleeding in thrombocytopenic conditions such as ITP. Both agents are generally well tolerated, although nausea, vomiting, diarrhea, dizziness, malaise, fever, rash, and transient hypotension or cardiac arrhythmias may occur. EACA can also rarely cause rhabdomyolysis, particularly with prolonged use. Headache is a common side-effect of AMCA and can also occur with EACA. Both agents can cause or aggravate cerebral infarction when used in patients with subarachnoid hemorrhage. EACA and AMCA should not be used in patients with DIC, as excessive thrombosis can occur; they are also relatively contraindicated in patients with urologic bleeding conditions.

COMMONLY USED DRUGS IN HEMATOLOGIC DISORDERS

Antiplatelet agents CYCLOOXYGENASE INHIBITORS Aspirin is a nonselective inhibitor of cyclooxygenase (COX), the enzyme that regulates conversion of arachidonic acid to prostaglandins and thromboxane A2. Aspirin has an irreversible effect on COX-1 that results in inhibition of platelet aggregation and prevention of vasoconstriction. Aspirin has many therapeutic uses, including the prevention and treatment of arterial and venous thromboses. It is most commonly used to prevent or treat coronary or cerebral arterial thromboses. In hematology practice, aspirin is used as thrombosis prophylaxis in patients with primary bone marrow disorders such as multiple myeloma and myeloproliferative disorders. Aspirin can also alleviate microvascular symptoms such as headache, light-headedness, acral paresthesia and erythromelalgia in patients with polycythemia vera (PV) or essential thrombocythemia (ET) (McCarthy et al., 2002; Tefferi, 2003). In patients with arterial thrombophilias such as antiphospholipid antibody syndrome, aspirin is used for prevention of ischemic stroke (APASS Writing Committee, 2004; Albers et al., 2008) and recurrent pregnancy loss (Kutteh, 1996; Rai et al., 1997). The primary complication of aspirin use is bleeding. Bleeding can occur at any site but most commonly manifests as gastrointestinal bleeding or hemorrhagic stroke. Hypertension, older age and higher doses of aspirin appear to increase the risk of CNS hemorrhage. For most patients the benefits of aspirin in preventing cardiovascular, cerebrovascular, and ischemic events significantly outweigh the risk of a major hemorrhage (Gorelick and Weisman, 2005). Aspirin can also be nephrotoxic, causing acute renal failure from renal vasoconstriction or acute interstitial nephritis. Systemic vasoconstriction can cause exacerbation of congestive heart failure.

ADENOSINE DIPHOSPHATE RECEPTOR INHIBITORS Adenosine diphosphate (ADP) is a platelet agonist that is stored in platelet-dense granules. When a platelet is activated, ADP is released and binds to platelet surface receptors, thus recruiting additional platelets to form a platelet plug. ADP receptor inhibitors such as clopidogrel (Plavix®) and ticlopidine (Ticlid®) prevent platelet aggregation by selectively and irreversibly binding the platelet surface receptor P2Y12. Platelet aggregation is inhibited for the remainder of the platelet lifespan (7–10 days). Clopidogrel is indicated for treatment of acute ST and non-ST elevation myocardial infarction, peripheral arterial disease, arteriosclerotic vascular disease, and stroke. Ticlopidine is indicated after placement of coronary stents and after thromboembolic stroke (Varon and Spectre, 2009).

1129

A noteworthy side-effect of ticlopidine and clopidogrel is thrombotic thrombocytopenic purpura (TTP). This was first reported with ticlopidine in the late 1990s, and incidence was estimated at 1 case per 1600–5000 patients treated (Bennett et al., 1998). Clopidogrel became the preferred antiplatelet agent after three phase III trials including more than 20 000 patients reported a more favorable safety profile, with less neutropenia, skin and gastrointestinal toxicities, and no cases of TTP. However, in the year 2000 a report of 11 patients who developed TTP while on clopidogrel was published (Bennett et al., 2000). Ten of 11 patients developed TTP within 2 weeks of starting the drug. The mechanism for development of TTP is not known, but is thought to be nonimmunologic given the short time to onset. Patients required a median of eight plasma exchanges (range, 1–30), and were prone to recurrence of TTP. One patient died shortly after the diagnosis of TTP. Treating physicians should be aware of this rare but serious side-effect of clopidogrel. The newest ADP receptor P2Y12 inhibitors include prasugrel and ticagrelor. Compared to clopidogrel, prasugrel has a more rapid onset of action and its inhibitory effect is stronger (Wallentin et al., 2008). In addition, prasugrel is not affected by genetic variations of cytochrome P450. The effect of prasugrel on platelets is irreversible and appears to be responsible for increased risk of bleeding with this agent (Wiviott et al., 2007). Ticagrelor is different in two respects: Its binding to the P2Y12 is stronger and faster than prasugrel but it is reversible. In a double blind randomized trial, comparing ticagrelor to prasugrel in patients with acute coronary syndromes, ticagrelor was superior in composite death rate, without increasing the risk of major bleeding (Schomig, 2009; Wallentin et al., 2009).

GLYCOPROTEIN IIB/IIIA ANTAGONISTS The primary use of these agents is in the cardiac catheterization laboratory. The representative agents of this group are abciximab, eptifibatide, and tirofiban. The published trial suggests a 9% reduction in death rate at 30 days in patients with acute coronary syndromes. Oral GPIIb/IIIa blockers (xemilofiban, orbofiban, sibrafiban, and lotrafiban) have been disappointing as they have been shown to be no more effective than aspirin and may even increase the risk of mortality (Chew et al., 2001).

THROMBOXANE SYNTHASE INHIBITORS A brief mention also must be made of dipyridamole, which inhibits thromboxane synthase. This results in lower uptake of thromboxane A2 and lowered cellular uptake of adenosine. It acts as an antiplatelet agent

1130

E. ANDERES AND S. NAND

and is also a vasodilator. Dipyridamole has been studied extensively, frequently in combination with aspirin. A review of over 25 trials suggests that while its effects in coronary syndromes are modest at best, it may reduce the risk of stroke (Diener et al., 1996). Newer formulation of dipyridamole with better bioavailability may confirm such an advantage of this old drug.

Antithrombotic agents UNFRACTIONATED HEPARIN Unfractionated heparin is a glycosaminoglycan composed of polysaccharide chains with molecular weights ranging from 5000 to 30 000 kDa (Hirsh et al., 2001). It exerts its anticoagulant effect by binding to antithrombin (AT), causing a conformational change that accelerates the inhibition of thrombin and factor Xa. Inhibition of thrombin and factor Xa prevents the conversion of fibrinogen to fibrin and the activation of other clotting factors, thus minimizing clot formation. Heparin is effective and indicated for the prevention of venous thromboembolism; for the treatment of venous thrombosis and pulmonary embolism (PE); for the early treatment of patients with unstable angina and acute myocardial infarction (MI); for patients who undergo cardiac surgery using cardiac bypass, vascular surgery, and coronary angioplasty; in patients with coronary stents; and in selected patients with disseminated intravascular coagulation. Heparin can cause hyperkalemia, osteoporosis after long-term use, and nonimmune-mediated thrombocytopenia due to platelet agglutination. Heparin can cause bleeding at any site, reported to occur in 5–10% of patients receiving heparin (Kelton and Hirsh, 1980). Epidural or spinal hematomas can occur in patients receiving unfractionated or low molecular weight heparin, particularly in patients who undergo lumbar puncture or epidural catheter placement for analgesia. Careful timing of heparin administration, attention to heparin dose, and avoidance of other medications that can cause bleeding are essential to minimize the risk of epidural or spinal hematomas (Wysowski et al., 1998). Heparin can be inactivated by protamine sulfate to prevent or minimize bleeding complications. The most feared complication of heparin is heparininducted thrombocytopenia and thrombosis (HITT), which occurs in about 1% of patients receiving unfractionated heparin. HITT is caused by antibodies directed against heparin-platelet factor 4 complexes, which activate platelets and stimulate thrombin generation. Onset typically occurs 7–10 days after heparin exposure, unless patients have had prior heparin exposure within the past 3 months, in which case onset can occur within 1–2 days. Patients have a drop in platelet count of 50% and about

half of patients develop venous or arterial thrombosis. Thrombosis can occur even after heparin is discontinued, so patients should be placed on anticoagulation with a direct thrombin inhibitor and eventually transitioned to warfarin for a minimum of 2–3 months (longer if thrombosis occurred). Patients presenting with acute thrombosis 1–2 weeks after heparin exposure should be evaluated for HITT (Hirsh et al., 2001).

LOW MOLECULAR WEIGHT HEPARIN Low molecular weight heparin (LMWH) is derived from depolymerization of unfractionated heparin. It has an average molecular weight of 5000 kDa, with a range of 1000–10 000. The majority of these heparin chains are too short to bind ATIII and thrombin, and the anticoagulant effect is primarily the result of ATIIImediated inactivation of factor Xa. LMWH has a more predictable pharmacokinetic profile than unfractionated heparin and monitoring is usually not necessary. Antifactor Xa levels can be measured if anticoagulant monitoring is desired, since LMWH does not reliably prolong the aPTT. LMWH is primarily cleared by the kidneys and it should be used with caution in patients with renal insufficiency. It is prudent to check factor Xa levels periodically in this patient population, though there are few guidelines for dose modifications. Similarly, there are no guidelines for dose adjustments in morbidly obese patients, and factor Xa levels should be checked a few days after therapy is initiated (Hirsh et al., 2001). LMWH is indicated for prevention of venous thromboembolism (VTE), for treatment of venous thrombosis, for treatment of acute PE, and for the early treatment of patients with unstable angina. Like unfractionated heparin, bleeding can occur with LMWH but is less common. Spinal and epidural hematomas have been reported (see section on unfractionated heparin). HITT can occur with LMWH but the incidence is very low. LMWH is only partially neutralized by protamine sulfate.

FACTOR XA INHIBITORS Fondaparinux is an indirect factor Xa inhibitor, which acts by catalyzing factor Xa inhibition by antithrombin. It is a synthetic analog of the antithrombin-binding pentasaccharide found in heparin or LMWH (Weitz et al., 2008). Fondaparinux is currently approved for treatment of DVT or PE, and VTE prophylaxis after major orthopedic surgery or abdominal surgery. It was shown to be as effective as LMWH in the treatment of acute DVT (Buller et al., 2004), and as effective as intravenous unfractionated heparin in the treatment of acute PE (Buller et al., 2003). The main complication of therapy is bleeding, and protamine sulfate is not effective at neutralizing fondaparinux (Rosenberg, 2001). As with other

COMMONLY USED DRUGS IN HEMATOLOGIC DISORDERS heparinoids, spinal and epidural hematomas have been reported. Isolated cases of HITT have also been reported (Warkentin et al., 2007).

WARFARIN Warfarin (4-hydroxycoumarin) inhibits the synthesis of vitamin K-dependent coagulation factors, including factors II, VII, IX, and X, as well as proteins C and S. It is commonly used for prevention and treatment of VTE, prevention of stroke in atrial fibrillation, anticoagulation for mechanical prosthetic heart valves, and myocardial infarction prevention in coronary artery disease. Hemorrhage is the most common side-effect of anticoagulation with warfarin and can occur at any site. Cases of acute femoral neuropathy have been reported with therapeutic use of warfarin and are secondary to retroperitoneal bleeding (Butterfield et al., 1972). FFP can rapidly, albeit temporarily, reverse the anticoagulant effect in patients with life-threatening bleeding. Otherwise, oral vitamin K should be used to correct the coagulopathy. Warfarin is extensively metabolized by the hepatic cytochrome P450 system and polymorphisms in these enzymes contribute to interpatient variations in dosing (Francis, 2008). Warfarin also interacts with a variety of medications, which can lead to sub- or supratherapeutic INR values in a given patient. Similarly, variations in intake in vitamin K-containing foods can affect warfarin dosing. Warfarin is a teratogen and should not be given to women of child bearing age who may become pregnant. Warfarininduced skin necrosis and venous gangrene can occur in patients with protein C or S deficiency who start warfarin without prior alternate anticoagulation. Anticoagulation with another medication prior to starting warfarin prevents this rare but serious complication of therapy, but is not uniformly required (Brandjes et al., 1992).

DIRECT THROMBIN INHIBITORS Direct thrombin inhibitors inactivate thrombin in an antithrombin-independent fashion. Unlike heparin, they can inactivate thrombin that is bound to fibrin. Examples include hirudin (lepirudin), bivalirudin, and argatroban. These agents do not bind to plasma proteins, and therefore have a more predictable anticoagulant response than unfractionated heparin (Bauer, 2006). Also, they do not cause HITT and all three drugs are approved as treatment of patients with HITT. Argatroban does not require dose adjustment in patients with renal failure, but does for patients with liver disease. Lepirudin is renally excreted and should be dose-reduced in patients with decreased glomerular filtration rates. All three agents are given by continuous intravenous infusion and are monitored using the aPTT. Argatroban can also elevate the PT, making the transition to warfarin more

1131

complicated. All direct thrombin inhibitors can cause bleeding, including intracranial hemorrhage. Dabigatran (Pradaxa ®) is a reversible oral thrombin inhibitor. Its absorption is pH sensitive and is decreased by approximately 30% in the presence of proton pump inhibitors. Unlike warfarin, its metabolism is not dependent on cytochrome P450 enzymes and there are fewer drug interactions. Dabigatran can accumulate in patients with renal failure. A recent phase III trial of anticoagulation in patients with atrial fibrillation demonstrated rates of stroke and systemic embolism similar to those observed with warfarin, but lower rates of major hemorrhage (Connolly et al., 2009). Another trial (Schulman et al., 2009) shows that dabigatran is as effective as warfarin in the management of acute venous thromboembolism. This drug was recently FDA approved for use in patients with atrial fibrillation. Bleeding is the most common side-effect of dabigatran, including intracranial hemorrhage in 0.3% of patients (Connolly et al., 2009).

MANAGEMENT OF HEMORRHAGIC COMPLICATIONS OFANTICOAGULATION (Table 76.2)

Intracranial hemorrhages WARFARIN Warfarin use accounts for 10–15% of intracranial hemorrhages (ICH) (SPIRIT Study Group, 1997). The frequency of events is increasing as more elderly patients receive anticoagulation. Warfarin increases the risk of Table 76.2 Management of coagulopathic intracranial hemorrhage Anticoagulant

Treatment

Dose

Warfarin

FFP or PCC and Vitamin K Protamine sulfate

15 mL/kg 15–30 U/kg 10 mg

Heparin (unfractionated or low molecular weight) Direct thrombin inhibitors Antiplatelet agents

No antidote available Platelet transfusion and/or desmopressin (DDAVP)

1 mg per 100 U of heparin or 1 mg of enoxaparin

Transfuse to > 100 000 platelets 0.3 mg/kg

1132

E. ANDERES AND S. NAND

ICH two to five times, with the risk of bleeding being proportional to the intensity of anticoagulation (Aguilar et al., 2007). There are no randomized trials addressing management of warfarin-induced ICH. Prompt, emergent reversal of anticoagulation is indicated and should be initiated prior to obtaining results of coagulation studies (Rincon et al., 2007). Reversal is usually accomplished with vitamin K and FFP; however, this takes several hours to accomplish. The volume of FFP required to reverse the INR can be > 2 liters, which is problematic in older patient populations. PCCs normalize the INR more rapidly and can be given in smaller volumes than FFP; however, they carry an increased risk of thrombosis and DIC in patients with severe brain injury. There are few data on PCCs in ICH, and different PCCs vary in their coagulation factor components, making comparison between trials difficult. Dosing is dependent on the specific PCC available, but the goal of treatment is reduction of the INR to < 1.2 (Aguilar et al., 2007). Vitamin K should also be administered. Recombinant FVIIa has been used off-label to reverse anticoagulation in patients with ICH on warfarin. RFVIIa can reverse the INR within minutes, though the effect only lasts several hours and vitamin K and FFP should also be administered (Freeman et al., 2004). The FAST trial investigated two different doses of rFVIIa in ICH. Although rFVIIa reduced growth of the hematoma, there was no survival benefit or improvement in functional outcome at 90 days for either dose compared to placebo. Additionally, treatment increased the frequency of arterial thromboembolic serious adverse events in the high dose arm as compared to placebo (Mayer et al., 2008). Though this trial does not specifically address patients on warfarin with ICH, it does provide safety information and dosing guidelines. RFVIIa is currently not approved for treatment of ICH. Restarting anticoagulation in patients with atrial fibrillation or mechanical prosthetic valves poses a therapeutic dilemma. There are no large randomized trials, and recommendations in the literature for holding anticoagulation vary from 1–6 weeks. The risk of embolic stroke occurring within 7–14 days after stopping warfarin appears to be low. If the INR is reversed with PCCs, subcutaneous heparin can be considered to reduce the risk of thrombosis. In most patients, warfarin can safely be restarted 7–14 days after the ICH (Aguilar et al., 2007).

UNFRACTIONATED AND LOW MOLECULAR WEIGHT HEPARIN

The anticoagulant effect of unfractionated heparin can be reversed with protamine sulfate. Protamine can partially reverse the effect of low molecular weight heparin.

The recommended dose of protamine is 1 mg per 100 U of heparin, or 1 mg of enoxaparin. Side-effects include flushing, bradycardia, and hypotension.

DIRECT THROMBIN INHIBITORS The direct thrombin inhibitors lipirudin, argatroban, and bivalirudin directly inhibit thrombin. Unfortunately no direct antidote is available for their reversal.

ANTIPLATELET AGENTS Aspirin, nonsteroidal anti-inflammatory agents, and ADP receptor inhibitors irreversibly inhibit platelet function. Treatment of hemorrhage associated with these agents involves stopping the drugs and transfusing platelets. Platelet levels should be maintained above 100 000/mL. Desmopressin (DDAVP) 0.3 mg/kg can be considered. This treatment promotes release of von Willebrand factor and enhances platelet function. However, there are few data to support this approach (Rincon et al., 2007).

AGENTS USED TO TREAT HEMATOLOGIC MALIGNANCIES Chronic myeloproliferative neoplasms The classic myeloproliferative neoplasms include polycythemia vera (PV), essential thrombocythemia (ET), idiopathic myelofibrosis (IMF), and chronic myeloid leukemia (CML). They are characterized by clonal bone marrow expansion of the myeloid series, with clinical features of hepatosplenomegaly, hypercatabolism, and increased numbers of circulating mature blood cells. The focus of treatment is to reduce cell proliferation and prevent sequelae of thrombosis and hemorrhage. In CML, small molecule tyrosine kinases target the disease defining genetic abnormality, the t(9;22)(q34;q11).

HYDROXYUREA Hydroxyurea (Hydrea; hydroxycarbamide) is classified as an antimetabolite. It is thought to be cell cycle-specific for the S phase of cell division. Hydrea inhibits the enzyme ribonucleotide reductase, which converts ribonucleotides to deoxyribonucleotides, critical precursors for de novo DNA biosynthesis and DNA repair. There does not appear to be an effect on synthesis of RNA or protein (Chu and DeVita, 2009). In PV and ET, hydroxyurea is used as cytoreductive therapy in patients over the age of 60 years or those with a prior thrombotic event. Treatment with hydroxyurea reduces platelet counts as well as the thrombosis rate in these patients (Cortelazzo et al., 1995; Fruchtman et al., 1997). The most common side-effect is myelosuppression. Mucocutaneous and skin ulcers, usually in the lower

COMMONLY USED DRUGS IN HEMATOLOGIC DISORDERS extremities, have been reported. Peripheral neuropathy has been reported in HIV-infected patients who receive hydroxyurea in combination with the antiretroviral agents didanosine (ddI) and stavudine (d4T). ddI and d4T are well known to cause neuropathy, but the addition of hydroxyurea to these medications significantly increases the risk of developing neuropathy. In this setting, hydroxyurea is used to potentiate the effect of these drugs (Moore et al., 2000). The most concerning potential side-effect of hydrea is development of secondary leukemia or myelodysplasia; however, data are not conclusive. Hydrea is teratogenic and is not given to women of child bearing potential. Interferon-a is used in place of hydroxyurea in this patient population.

ANAGRELIDE Anagrelide is thought to reduce platelet production by decreasing megakaryocyte hypermaturation. It is used as a second-line agent in patients with ET who are intolerant of hydroxyurea. Anagrelide can cause arterial thrombosis including myocardial infarction, stroke, and transient ischemic attack (TIA). Serious hemorrhage, cardiomyopathy, and edema can also occur. Headache is a common side-effect and can lead to drug discontinuation (Harrison et al., 2005). There are no controlled studies on anagrelide in pregnant women, therefore its use is not recommended in this patient population.

BCR-ABL TYROSINE KINASE INHIBITORS This class of drugs was rationally designed to target the disease-defining genetic abnormality that defines CML: the t(9;22)(q34;q11) or Philadelphia chromosome and its molecular equivalent, the bcr-abl fusion tyrosine kinase. Imatinib mesylate (Gleevec™) was the first drug in this class and is approved for front-line therapy in patients with CML. Imatinib binds to the ATP pocket of the bcr-abl protein and inhibits substrate phosphorylation, thereby inducing apoptosis (Chu and DeVita, 2009). Imatinib also inhibits other receptor tyrosine kinases such as platelet-derived growth factor receptors (PDGFR), stem cell factor (SCF) and c-kit. It is well tolerated and common side-effects include edema and fluid retention, fatigue, rash, nausea and vomiting, diarrhea, myelosuppression, and cough. Congestive heart failure is the most serious reported side-effect. Headaches, insomnia, paresthesias, dizziness and asthenia have been reported in up to approximately 10–20% of patients. Second-generation bcr-abl tyrosine kinase inhibitors have been developed to overcome resistance to imatinib. These agents were recently approved for first-line treatment of CML and are also used to treat accelerated or blast phase CML. Dasatinib (Sprycel®) is a potent

1133

inhibitor of the bcr-abl kinase as well as the SRC family of kinases, c-kit, and PDGFR-b. It binds to both the active and inactive conformations of the abl kinase domain, thereby overcoming imatinib resistance resulting from bcr-abl mutations (Chu and DeVita, 2009). Common side-effects include fluid retention, rash, hypocalcemia, and hypophosphatemia, diarrhea, nausea and vomiting, headache, dyspnea, and fatigue. Serious side-effects include myelosuppression, pleural effusions, QT prolongation, and hemorrhage (gastrointestinal and cerebral) secondary to platelet dysfunction. Nilotinib (Tasigna®) is another second-generation bcr-abl kinase inhibitor, which also inhibits c-kit and PDGFR-b kinases. Nilotinib has a higher binding affinity and selectivity for the abl kinase domain when compared to imatinib, and is able to overcome imatinib resistance resulting from bcr-abl mutations (Chu and DeVita, 2009). Common side-effects include edema, pruritis, rash, constipation or diarrhea, myelosuppression, headache, nausea, fatigue, electrolyte disturbances, and elevations in serum lipase. QT prolongation and sudden death have been reported. Imatinib is predominantly metabolized in the liver by the cytochrome P450 enzymes CYP3A4 and CYP3A5, while dasatinib and nilotinib are primarily metabolized in the liver by CYP3A4. Drug interactions are common and should be considered when caring for a patient on these medications.

Myelodysplasia DNA METHYLTRANSFERASE INHIBITORS AND NUCLEOSIDE ANALOGS

5-azacitidine (Vidaza®) and 5-aza-20 -deoxycitidine (decitabine) are hypomethylating agents that inhibit DNA meythltransferase by incorporation of either azacitidine triphosphate or decitabine triphosphate into DNA. This leads to a loss of DNA methylation and reactivation of aberrantly silenced genes. They are cell cycle-specific with activity in the S phase. Azacitidine triphosphate is also incorporated into RNA, resulting in inhibition of RNA processing and function. A phase III trial demonstrated a survival advantage for azacitidine over conventional care, and established it as the preferred therapy for patients with high-risk myelodysplasia (MDS) (Fenaux et al., 2009). A similar trial conducted with decitabine did not demonstrate a survival advantage (WijerMans et al., 2008), though many feel the drugs are clinically equivalent and biologically similar. Common side-effects of these agents include myelosuppression, fatigue and anorexia, nausea, vomiting, constipation and abdominal pain, and peripheral edema. Azacitidine can cause renal toxicity with elevations in serum creatinine, renal tubular acidosis and

1134 E. ANDERES AND S. NAND hypokalemia, while decitabine can cause hyperbilirubiincludes dose-limiting myelosuppression, immunosupnemia. Both medications can also cause dizziness and pression, pulmonary toxicity, and severe neurotoxicity, headache (Chu and DeVita, 2009). CNS toxicity includalthough this complication has occurred primarily at siging coma was reported in a small number of patients on a nificantly greater doses than currently recommended for clinical trial who were treated with doses of azacitidine clinical use. Cladribine can cause a dose-dependent that are higher than those used in current clinical practice delayed sensorimotor peripheral neuropathy character(Saiki et al., 1981). Similarly, acute CNS toxicity with ized by axonal degeneration and secondary demyelinsevere myalgias and altered mental status was reported ation (Vahdat et al., 1994). Irreversible paraparesis and in a child receiving high doses of azacitidine (Weisman quadraparesis were reported in patients with refractory et al., 1985). acute leukemia and non-Hodgkin lymphoma who were treated with high doses of cladribine (Beutler et al., Leukemias, lymphomas, and 1991). High doses of fludarabine and pentostatin are also multiple myeloma neurotoxic (Cheson et al., 1994). At standard doses of fludarabine, somnolence and peripheral neuropathy CHEMOTHERAPEUTIC AGENTS have been reported (Cheson et al., 1994). Chemotherapeutic agents are classified according to The 6-thiopurines are primarily used in treatment of their mechanism of action. Broad categories include acute leukemia, while fludarabine, cladribine and penalkylating agents, platinum agents, antimetabolites, tostatin are active in both leukemias and lymphomas. topoisomerase inhibitors, antimicrotubule agents, and Pyrimidine analogs include cytarabine, gemcitabine, antibiotics. 5-fluorouracil and capecitabine. Cytarabine is critical Alkylating agents form covalent bonds with DNA in the treatment of acute myeloid leukemia (AML). It bases. Bifunctional alkylators interact with two strands is also used in treatment of acute lymphoblastic leukeof DNA and form a “cross-link” that covalently links mia (ALL) and lymphomas, including primary central the two strands of the DNA double helix. This prevents nervous system lymphoma. At high doses, cytarabine the cell from replicating effectively. Common sidecrosses the blood–brain barrier and can be effective as effects of alkylating agents include myelosuppression, prophylaxis for CNS leukemia. High-dose cytarabine is nausea and vomiting, and alopecia. They are teratogenic neurotoxic and can cause seizures, cerebral and cerebeland leukemogenic, with secondary myelodysplasia and lar dysfunction, peripheral neuropathy, bilateral rectus acute leukemia occurring 7–10 years after therapy muscle palsy, optic neuritis, aphasia, and parkinsonian (Colvin and Friedman, 2005). Examples of alkylating symptoms. Cerebral dysfunction manifests as generalagents used in hematologic malignancies include cycloized encephalopathy, with somnolence, confusion, disphosphamide, ifosfamide, melphalan, and chlorambucil. orientation, memory loss, cognitive dysfunction, Ifosfamide can cause CNS toxicity including confusion, psychosis, and frontal lobe release signs. Rapid infusion somnolence and hallucinations, as well as encephalopaof high doses of cytarabine increases the risk of cerebral thy and seizures. toxicity (Baker et al., 1991). Clinical signs of cerebellar Platinum agents interact with guanine and adenine dysfunction occur in up to 15% of patients within 8 days residues to form DNA adducts and cross-link DNA of treatment and include dysarthria, dysdiadochokinestrands. If the DNA damage is not repaired or tolerated, sia, dysmetria, and ataxia. EEG will often demonstrate apoptosis occurs (Johnson and O’Dwyer, 2005). Examdiffuse slow wave activity. Even when therapy is ples of platinum agents include cisplatin, carboplatin, discontinued, up to 30% of patients with cerebellar and oxaliplatin. Carboplatin is used for second-line treattoxicity do not fully recover. Peripheral neuropathy ment of leukemia and lymphoma. Cisplatin can cause is a rare complication of cytarabine therapy, occurring peripheral neuropathy and ototoxicity. Oxaliplatin comin < 1% of patients. The severity of symptoms monly causes both acute and delayed sensory neuropaincreases with higher cumulative doses of cytarabine thy. Less commonly, it can cause pharyngolaryngeal and can range from a pure sensory neuropathy to a rapdysesthesia and Lhermitte’s sign. idly progressive ascending polyneuropathy (Baker et al., Antimetabolites can be subclassified as purine ana1991). Neurotoxicity can be reduced by increasing logs, pyrimidine analogs, and antifolates. They are cell the duration of the infusion to more than 3 hours. cycle-specific and are therefore active on replicating Patients older than age 50 and those with elevated cells. Antimetabolites interfere with DNA production serum creatinine are particularly susceptible to neuroand replication, causing apoptosis. Purine analogs logic toxicity from high-dose cytarabine (Kummar include the 6-thiopurines 6-mercaptopurine and 6et al., 2005). Prior to each dose, patients are evaluated thioguanine, fludarabine, 2-chlorodeoxyadenosine (2for neurotoxicity and doses are held or adjusted as necCdA or cladribine) and pentostatin. Their toxicity profile essary to minimize toxicity.

COMMONLY USED DRUGS IN HEMATOLOGIC DISORDERS Cytarabine is also given intrathecally for prophylaxis of CNS leukemia in patients with acute lymphoblastic leukemia (ALL), and to treat leptomeningeal disease in both leukemias and solid tumors. A depot formulation, in which the cytarabine is encapsulated in multivesicular liposomes for sustained release in the CSF, is also available. Doses can be administered through an Ommaya reservoir or via lumbar puncture. For treatment of CNS leukemia, doses are typically given twice weekly until the leukemia is no longer detectable in the CSF. This is followed by weekly doses for 4 weeks, and monthly doses for up to 1 year. The depot formulation is typically given every 2 weeks. Dose-limiting toxicities include headache and arachnoiditis and are more common with the depot formulation. Myelopathy, paraplegia, papilledema, and seizures can also occur (Baker et al., 1991). Occasionally systemic toxicities such as nausea and myelosuppression, are seen with intrathecal cytarabine (Kummar et al., 2005). Antifolate analogs include methotrexate, raltitrexed and pemetrexed. Methotrexate inhibits dihydrofolate reductase and prevents de novo thymidylate and purine nucleotide biosynthesis (Kummar et al., 2005). High dose methotrexate, often used in CNS lymphoma, is occasionally associated with an acute, transient cerebral dysfunction manifesting as paresis, aphasia, and behavioral abnormalities. Seizures have been described in 4– 15% of patients who receive high-dose methotrexate. Symptoms generally occur within 6 days of treatment and completely resolve within 48–72 hours. Chronic neurotoxicity with encephalopathy, dementia and motor paresis can occur 2–3 months after administration of high-dose methotrexate (Kummar et al., 2005). Intrathecal methotrexate is used for CNS prophylaxis in ALL, as well as treatment of CNS leukemia and leptomeningeal involvement by solid tumors. There are three distinct neurotoxic syndromes associated with intrathecal methotrexate (Walker et al., 1984). Acute chemical arachnoiditis can occur immediately after administration and is the most common toxicity. This syndrome is characterized by severe headaches, nuchal rigidity, vomiting, fever, and an inflammatory cell infiltrate in the CSF. A subacute form of neurotoxicity is seen in approximately 10% of patients and usually occurs after the third or fourth course of intrathecal therapy. This most commonly occurs in patients with active meningeal leukemia and consists of motor paralysis, cranial nerve palsies, seizures, and/or coma. Continued intrathecal therapy with methotrexate can result in death, therefore a change in therapy is mandatory. The third syndrome is a chronic demyelinating encephalopathy, which typically occurs in children several months to years after treatment. Patients present with dementia, limb spasticity, and in advanced cases, coma. Ventricular

1135

enlargement, cortical thinning, and diffuse intracerebral calcifications are noted on computed tomographic (CT) scan (Kummar et al., 2005). Inhibitors of topoisomerase I and II include irinotecan, topotecan, etoposide, and anthracyclines. DNA topoisomerases modify the tertiary structure of DNA without altering the primary nucleotide sequence. Topoisomerase inhibitors generate single and double strand breaks in DNA, resulting in apoptosis and cell death. They are cell cycle-specific. Etoposide and anthracyclines are used in treatment of acute leukemia and lymphomas. Antimicrotubule agents include the vinca alkaloids (vincristine, vinblastine, vindesine) and the taxanes (paclitaxel and docetaxel). These agents interfere with microtubule function, particularly within the mitotic spindle apparatus. Vinca alkaloids are used in the treatment of ALL and lymphomas. Vincristine is particularly known for causing neurotoxicity, which is characterized by a symmetric, mixed sensorimotor and autonomic polyneuropathy. Pathologically, vincristine interferes with axonal microtubule function and causes axonal degeneration and decreased axonal transport. Symptoms start with a distal symmetric sensory impairment. With continued treatment, neuritic pain and loss of deep tendon reflexes can develop. This may be followed by foot drop, wrist drop, motor dysfunction, ataxia, and paralysis. Cranial nerves are rarely affected, but can manifest as hoarseness, diplopia, jaw pain, and facial palsies. Central toxicity from vincristine is also very rare because of minimal uptake in the CNS. Central effects can include confusion, mental status changes, hallucinations, insomnia, seizures, and coma. Acute, severe autonomic toxicity may be seen with high doses (greater than 2 mg/m2) or in patients with altered hepatic function. Autonomic toxicities can include constipation and abdominal cramps, ileus, urinary retention, and orthostatic hypotension. In adults, neurotoxicity can occur after treatment with cumulative doses of 5–6 mg, and toxicity can become severe after 15–20 mg. In routine treatment for lymphoma, adult patients receive 2 mg per treatment, with cumulative doses of 12–16 mg being given over 4–6 months. Children appear to be less sensitive, and older patients are particularly susceptible. Patients with preexisting neurologic disorders and hepatic dysfunction are at especially increased risk. The only treatment for vincristine neurotoxicity is discontinuation of the drug. Vinblastine and vindesine rarely cause neurotoxicity (Rowinsky and Tolcher, 2005). Taxanes, particularly paclitaxel, can also cause neurotoxicity. This is most commonly a peripheral sensory neuropathy in a symmetric stocking-glove distribution. Neurologic examination reveals sensory loss and loss of deep tendon reflexes. Most patients experience mild

1136

E. ANDERES AND S. NAND

to moderate effects, but patients with pre-existing neuropathy are more prone to development of taxaneinduced neuropathy. Symptoms usually begin after several courses of standard-dose therapy, but can occur as early as 24–72 hours after the first treatment. Motor and autonomic neuropathy has also been reported with paclitaxel. Transient myalgia and arthralgia are also observed and most commonly occur within 24–48 hours after treatment. Treatment with prednisone can reduce myalgia and arthralgia. Taxanes are commonly used in the treatment of solid tumors.

TARGETED THERAPIES Monoclonal antibodies Rituximab (Rituxan®) is a chimeric (murine/human) monoclonal antibody against CD20, which is expressed on B lymphocytes. It is used in combination with chemotherapy to treat chronic lymphocytic leukemia, and can be given as a single agent or with chemotherapy to treat CD20 expressing non-Hodgkin’s lymphomas. Rituxan is also approved for use in rheumatoid arthritis, and is sometimes used off label to treat autoimmune cytopenias and other autoimmune diseases. Rituxan causes cell lysis through complement-dependent cytotoxicity (CDC) and antibody-dependent cellular cytotoxicity (ADCC). Common side-effects include infusion reactions (hypotension, bronchospasm, rigors, angioedema), fatigue, and myelosuppression. Tumor lysis syndrome can occur in patients with significant tumor burden. Patients with prior hepatitis B infection can have reactivation of the virus, and should be screened for prior infection before the drug is administered. Similarly, reactivation of the JC virus can cause progressive multifocal leukoencephalopathy (PML) in patients who receive Rituxan in combination with chemotherapy, immunosuppression, or stem cell transplant. The largest series reported 57 HIVnegative patients with hematologic malignancies and autoimmune disorders (Carson et al., 2009). Patients commonly presented with confusion/disorientation, motor weakness or hemiparesis, poor motor coordination, and changes in speech or vision. Symptoms progressed over weeks to months. The diagnosis was primarily confirmed by magnetic resonance imaging and JC virus detection in the CSF, or by brain biopsy or autopsy. Mortality was very high: all patients diagnosed with PML within 3 months of the last rituximab dose died, compared with 84% of patients who were diagnosed more than 3 months after the last rituximab dose. There is no standard effective therapy for this condition. Gemtuzumab ozogamicin (GO or Mylotarg®) is a recombinant humanized IgG4 k antibody that is conjugated with calicheamicin, a cytotoxic antitumor

antibiotic. The antibody portion of GO targets CD33, which is commonly expressed on leukemic blasts. After the antibody binds, a complex is formed and internalized, calicheamicin is released within the myeloid cell lysosomes, and the cell dies. GO is approved for treatment of relapsed leukemia. Common side-effects include infusion reactions, myelosuppression, and hepatotoxicity. Radioimmunotherapy Radioimmunotherapy allows the targeted delivery of ionizing radiation to the tumor site, while minimizing toxicity on normal tissue. This is accomplished by conjugating radioactive isotopes to monoclonal antibodies that target tumor cells. Currently, two products are approved for treatment of relapsed and refractory non-Hodgkin lymphoma. Tositumomab (Bexxar®) and ibritumomab (Zevalin®) are monoclonal antibodies to CD20 that are conjugated with the radionuclides iodine-131 (I-131) and yttrium-90 (Y-90), respectively. Side-effects include infusion reactions, asthenia and fatigue, and myelosuppression. There is also a risk of myelodysplasia and/or acute leukemia (Davies, 2007). Immunomodulatory drugs Thalidomide and lenalidomide are immunomodulatory drugs (IMiDs) primarily used in the treatment of multiple myeloma. Lenalidomide is also being studied for treatment of relapsed lymphomas. They have direct cytotoxic effects and induce apoptosis or growth arrest of myeloma cells. They also have potent antiangiogenic and anti-inflammatory effects that appear to inhibit cell growth (Kumar and Rajkumar, 2006). In the 1950s thalidomide was used to treat morning sickness in pregnant women. Several years later it was identified as a teratogen and was not used again until the 1990s, when it was investigated in cancer patients. Common sideeffects include constipation, drowsiness and somnolence, neutropenia, and VTE when used in combination with steroids or chemotherapy. Thalidomide also causes a debilitating peripheral neuropathy as well as orthostatic hypotension and dizziness. Lenalidomide, a second-generation IMiD, is much better tolerated, with myelosuppression being the most common side-effect. Lenalidomide can also cause VTE when used in combination with steroids or chemotherapy, but it is not associated with peripheral neuropathy. Proteosome inhibitors Bortezomib (Velcade®) is a small molecule that inhibits the activity of the 26S proteasome, which degrades

COMMONLY USED DRUGS IN HEMATOLOGIC DISORDERS ubiquitinated proteins. The ubiquitin-proteasome pathway is critical for regulating concentrations of intracellular proteins and maintaining intracellular homeostasis. Inhibition of the proteasome prevents this targeted proteolysis and can affect multiple signaling cascades within the cell. Disruption of the normal homeostatic mechanisms can lead to cell death. Bortezomib is active in multiple myeloma as well as non-Hodgkin lymphomas. Common side-effects include constipation or diarrhea, nausea and vomiting, myelosuppression, dyspnea and cough, and peripheral neuropathy. The neuropathy is predominantly sensory, but motor neuropathy has also been reported.

REFERENCES Abshire T, Kenet G (2004). Recombinant factor VIIa: review of efficacy, dosing regimens and safety in patients with congenital and acquired factor VIII or IX inhibitors. J Thromb Haemost 2: 899–909. Aguilar MI, Hart RG, Kase CS et al. (2007). Treatment of warfarin-associated intracerebral hemorrhage: literature review and expert opinion. Mayo Clin Proc 82: 82–92. Albers GW, Amarenco P, Easton JD et al. (2008). Antithrombotic and thrombolytic therapy for ischemic stroke: American College of Chest Physicians evidencebased clinical practice guidelines (8th edition). Chest 133: 630S–669S. APASS Writing Committee (2004). Antiphospholipid antibodies and subsequent thrombo-occlusive events in patients with ischemic stroke. JAMA 291: 576–584. Babior BM (2006). Folate, cobalamin and megaloblastic anemias. In: E Beutler, K Kaushansky, TJ Kipps et al. (Eds.), Williams Hematology, McGraw-Hill, United States, pp. 477–509. Baker JW, Royer GL, Weiss RB (1991). Cytarabine and neurologic toxicity. J Clin Oncol 9: 679–693. Bauer KA (2006). New anticoagulants. Hematology Am Soc Hematol Educ Program 2006: 450–456. Bennett CL, Weinberg PD, Rozenberg-Ben-Dror K et al. (1998). Thrombotic thrombocytopenic purpura associated with ticlopidine: a review of 60 cases. Ann Intern Med 128: 541–544. Bennett CL, Connors JM, Carwile JM et al. (2000). Thrombotic thrombocytopenic purpura associated with clopidogrel. N Engl J Med 342: 1773–1777. Beutler E, Piro LD, Saven A et al. (1991). 2Chlorodeoxyadenosine (2-CdA): a potent chemotherapeutic and immunosuppressive nucleoside. Leuk Lymphoma 5: 1–8. Brandjes DP, Heijboer H, Buller HR et al. (1992). Acenocoumarol and heparin compared with acenocoumarol alone in the initial management of proximal vein thrombosis. N Engl J Med 327: 1485–1489. Buller HR, Davidson BL, Decousus H et al. (2003). Subcutaneous fondaparinux versus intravenous unfractionated heparin in the initial treatment of pulmonary embolism. N Engl J Med 349: 1695–1702.

1137

Buller HR, Davidson BL, Decousus H et al. (2004). Fondaparinux or enoxaparin for the initial treatment of symptomatic deep venous thrombosis: a randomized trial. Ann Intern Med 140: 867–873. Butterfield WC, Neviaser RJ, Roberts MP (1972). Femoral neuropathy and anticoagulants. Ann Surg 176: 58–61. Carson KR, Evens AM, Richey EA et al. (2009). Progressive multifocal leukoencephalopathy after rituximab therapy in HIV-negative patients: a report of 57 cases from the Research on Adverse Drug Events and Reports project. Blood 113: 4834–4840. Cheson BD, Vena DA, Foss FM et al. (1994). Neurotoxicity of purine analogs: a review. J Clin Oncol 10: 2216–2228. Chew DP, Bhatt DL, Sapp S et al. (2001). Increased mortality with oral platelet glycoprotein IIb/IIIa antagonists: a metaanalysis of phase II multicenter trials. Circulation 103: 201–206. Chu E, DeVita VT (2009). Physicians’ Cancer Chemotherapy Drug Manual, Jones and Bartlett, Sudbury, MA. Colvin MO, Friedman HS (2005). Alkylating agents. In: VT DeVita, S Hellman, SA Rosenberg (Eds.), Cancer: Principles and Practice of Oncology, Lipincott Williams and Wilkins, Philadelphia, PA. Connolly SJ, Ezekowitz MD, Yusuf S et al. (2009). Dabigatran versus warfarin in patients with atrial fibrillation. N Engl J Med 361: 1–11. Cook JD (2005). Diagnosis and management of iron deficiency anemia. Best Pract Res Clin Haematol 18: 319–332. Copelan EA, Strohm PL, Kennedy MS et al. (1986). Hemolysis following intravenous immune globulin. Transfusion 26: 410–412. Cortelazzo S, Finazzi G, Ruggeri M et al. (1995). Hydroxyurea for patients with essential thrombocythemia and a high risk of thrombosis. N Engl J Med 332: 1132–1136. Davies AJ (2007). Radioimmunotherapy for B-cell lymphoma: Y90 ibritumomab tiuxetan and I(131) tositumomab. Oncogene 26: 3614–3628. Diener HC, Cunha L, Forbes C (1996). European Stroke Study II Dypiridamole and acetylsalicylic acid in the secondary prevention of stroke. J Neurol Sci 143: 1–13. Fenaux P, Mufti GJ, Hellstrom-Lindberg E et al. (2009). Efficacy of azacitidine compared with that of conventional care regimens in the treatment of higher-risk myelodysplastic syndromes: a randomised, open-label, phase III study. Lancet Oncol 10: 223–232. Forristal T, Witt M (1968). Pleocytosis after iron dextran injection. Lancet 1: 1428. Francis CW (2008). New issues in oral anticoagulants. Hematology Am Soc Hematol Educ Program 2008: 259–265. Freeman WD, Brott TG, Barrett KM et al. (2004). Recombinant factor IVVa for rapid reversal of warfarin anticoagulation in acute intracranial hemorrhage. Mayo Clin Proc 79: 1495–1500. Fruchtman SM, Mack K, Kaplan ME et al. (1997). From efficacy to safety: a Polycythemia Vera Study Group report on hydroxyurea in patients with polycythemia vera. Semin Hematol 34: 17–23.

1138

E. ANDERES AND S. NAND

Gorelick PB, Weisman SM (2005). Risk of hemorrhagic stroke with aspirin use – an update. Stroke 36: 1801–1807. Guidolin L, Vignoli A, Canger R (1998). Worsening in seizure frequency and severity in relation to folic acid administration. Eur J Neurol 5: 301–303. Harrison CN, Campbell PJ, Buck G et al. (2005). Hydroxyurea compared with anagrelide in high-risk essential thrombocythemia. N Engl J Med 353: 33–45. Hellstern P (1999). Production and composition of prothrombin complex concentrates: correlation between composition and therapeutic efficiency. Thromb Res 95 (4 Suppl 1): S3–S6. Hirsh J, Warkentin TE, Shaughnessy SG et al. (2001). Heparin and low-molecular-weight heparin mechanisms of action, pharmacokinetics, dosing, monitoring, efficacy, and safety. Chest 119: 64S–94S. Hunter R, Barnes J, Oakeley HF et al. (1970). Toxicity of folic acid given in pharmacological doses to healthy volunteers. Lancet 1: 61–63. Johnson SW, O’Dwyer PJ (2005). Cisplatin and its analogues. In: VT DeVita, S Hellman, SA Rosenberg (Eds.), Cancer: Principles and Practice of Oncology. Lipincott Williams and Wilkins, Philadelphia, PA. Kelton JG, Hirsh J (1980). Bleeding associated with antithrombotic therapy. Semin Hematol 17: 259–291. Killip S, Bennett JM, Chambers MD (2007). Iron deficiency anemia. Am Fam Physician 75: 671–678. Knezevic-Maramica I, Druskall MS (2003). Intravenous immune globulines: an update for clinicians. Transfusion 43: 1460–1480. Krishnaswamy K, Nair KM (2001). Importance of folate in human nutrition. Br J Nutr 85 (Suppl 2): S115–S124. Kumar S, Rajkumar SV (2006). Thalidomide and lenalidomide in the treatment of multiple myeloma. Eur J Cancer 42: 1612–1622. Kummar S, Noronha V, Chu E (2005). Antimetabolites. In: VT DeVita, S Hellman, SA Rosenberg (Eds.), Cancer: Principles and Practice of Oncology. Lipincott Williams and Wilkins, Philadelphia, PA. Kutteh WH (1996). Antiphospholipid antibody-associated recurrent pregnancy loss: treatment with heparin and low-dose aspirin is superior to low-dose aspirin alone. Am J Obstet Gynecol 174: 1584–1589. MacLennan S, Barbara JA (2006). Risks and side effects of therapy with plasma and plasma fractions. Best Pract Res Clin Haematol 19: 169–189. Marie I, Maurey G, Herve´ F et al. (2006). Intravenous immunoglobulin-related thrombotic complications. Br J Dermatol 155: 714–721. Mayer SA, Brun NC, Begtrup K et al. (2008). Efficacy and safety of recombinant activated factor VII for acute intracerebral hemorrhage. N Engl J Med 358: 2127–2137. McCarthy L, Eichelberger L, Skipworth E et al. (2002). Erythromelalgia due to essential thrombocythemia. Transfusion 42: 1245. Moore RD, Wong WE, Keruly JC et al. (2000). Incidence of neuropathy in HIV-infected patients on monotherapy

versus those on combination therapy with didanosine, stavudine and hydroxyurea. AIDS 14: 273–278. Stroke Prevention in Reversible Ischemia Trial (SPIRIT) Study Group: a randomized trial of anticoagulants versus aspirin after cerebral ischemia of presumed arterial origin (1997). Ann Neurol 42: 857–865. Oh RC, Brown DL (2003). Vitamin B12 deficiency. Am Fam Physician 67: 979–986. Orbach H, Katz U, Sherer Y et al. (2005). Intravenous immunoglobulin: adverse effects and safe administration. Clin Rev Allergy Immunol 29: 173–184. Paran D, Herishanu Y, Elkayam O et al. (2005). Venous and arterial thrombosis following administration of intravenous immunoglobulins. Blood Coagul Fibrinolysis 16: 313–318. Rai R, Cohen H, Dave M et al. (1997). Randomised controlled trial of aspirin and aspirin plus heparin in pregnant women with recurrent miscarriage associated with phospholipid antibodies (or antiphospholipid antibodies). BMJ 314: 257. Redman RC, Miller JB, Hood M et al. (2002). Trimethopriminduced aseptic meningitis in an adolescent male. Pediatrics 110: 1–2. Reutter A, Luger TA (2004). Efficacy and safety of intravenous immunoglobulin for immune-mediated skin disease: current view. Am J Clin Dermatol 5: 153–160. Rincon F, Fernandez A, Mayer SA (2007). Hematology interventions for acute central nervous system disease. In: CS Kitchens, BM Alving, CM Kessler (Eds.), Consultative Hemostasis and Thrombosis, Saunders Elsevier, Philadelphia, PA. Roberts HR, Monroe DM, White GC (2004). The use of recombinant factor VIIa in the treatment of bleeding disorders. Blood 104: 3858–3864. Rosenberg RD (2001). Redesigning heparin. N Engl J Med 344: 673. Rowinsky EK, Tolcher AW (2005). Antimicrotubule agents. In: VT DeVita, S Hellman, SA Rosenberg (Eds.), Cancer: Principles and Practice of Oncology, Lipincott Williams and Wilkins, Philadelphia, PA. Saiki JH, Bodey GP, Hewlett JS et al. (1981). Effect of schedule on activity and toxicity of 5-azacytidine in acute leukemia. Cancer 47: 1739–1742. Schomig A (2009). Ticagrelor – is there need for a new player in the antiplatelet therapy field? N Engl J Med 361: 1108–1110. Schulman S, Kearon C, Kakkar AK et al. (2009). Dabigatran versus warfarin in the treatment of acute venous thromboembolism. N Engl J Med 361: 2342–2352. Silverstein SB (2004). Parenteral iron therapy options. Am J Hematol 76: 74–78. Stanworth SJ (2007). The evidence-based use of FFP and cryoprecipitate for abnormalities of coagulation tests and clinical coagulopathy. Hematology Am Soc Hematol Educ Program 2007: 179–186. Tefferi A (2003). Polycythemia vera: a comprehensive review and clinical recommendations. Mayo Clin Proc 78: 174–194. Triulzi DJ (2006). Transfusion-related acute lung injury: an update. Hematology Am Soc Hematol Educ Program 2006: 497–501.

COMMONLY USED DRUGS IN HEMATOLOGIC DISORDERS Umbreit J (2005). Iron deficiency: a concise review. Am J Hematol 78: 225–231. Vahdat L, Wong ET, Wile MJ et al. (1994). Therapeutic and neurotoxic effects of 2-chlorodeoxyadenosine in adults with acute myeloid leukemia. Blood 84: 3429–3434. Varon D, Spectre G (2009). Antiplatelet agents. Hematology Am Soc Hematol Educ Program 2009: 267–262. Verstraete M (1985). Clinical application of inhibitors of fibrinolysis. Drugs 29: 236–261. Walker RW, Allen JC, Rosen G et al. (1984). Transient cerebral dysfunction secondary to high dose methotrexate. Cancer 53: 1849. Wallentin L, Varenhorst C, James S (2008). Prasugrel achieves greater and faster P2Y12 receptor mediated platelet inhibition than clopidogrel due to more efficient generation of its active metabolite in aspirin treated patients with coronary artery disease. Eur Heart J 29: 21–30. Wallentin L, Becker RC, Budaj A et al. (2009). Ticagrelor versus clopidogrel in patients with acute coronary syndromes. N Engl J Med 361: 1045–1057. Wallerstein RO (1968). Intravenous iron-dextran complex. Blood 32: 690.

1139

Warkentin TE, Maurer BT, Aster RH et al. (2007). Heparininduced thrombocytopenia associated with fondaparinux. N Engl J Med 356: 2653–2655. Weisman SJ, Berkow RL, Weetman RM et al. (1985). 5Azacytidine: acute central nervous system toxicity. Am J Pediatr Hematol Oncol 7: 86–88. Weitz JI, Hirsh J, Samama MM (2008). New antithrombotic drugs: American College of Chest Physicians evidencebased clinical practice guidelines (8th edition). Chest 133: 234S–256S. WijerMans P, Suciu S, Baila L et al. (2008). Low dose decitabine versus best supportive care in elderly patients with intermediate or high risk MDS not eligible for intensive chemotherapy: final results of the randomized phase III study (06011) of the EORTC Leukemia and German MDS Study Groups. Blood (ASH Annual Meeting Abstracts) 112: 226. Wiviott SD, Braunwald E, McCabe CH et al. (2007). Prasugrel versus clopidogrel in patients with acute coronary syndrome. N Engl J Med 361: 1045–1057. Wysowski DK, Talarico L, Bacsanyi J et al. (1998). Spinal and epidural hematoma and low molecular weight heparin. N Engl J Med 338: 1774.

Index NB: Page numbers in italics refer to figures and tables.

A Abciximab 1639 Abdominal aortic aneurysm (AAA) 225–226 Abducens nerve (cranial nerve VI) 307 Abetalipoproteinemia (ABL) 623–624 ABO compatibility 1238 Abscess brain see under Brain conditions spinal epidural 1532 tuberculous 1494, 1494 ‘Absences’(‘absent-mindedness’) 184 Absent cough reflex 1240 Absidia 1383, 1396 Acanthamoeba spp 1287–1288, 1411 Acanthophis spp 992 Acarbose 817, 818–819 Accessory nerve (cranial nerve XI) 308 Acebutolol 1636 Acenocumarol 1638–1639 Aceruloplasminemia 853, 860 Acetaminophen 953 Acetylcholinesterase inhibitors 1628 Acetyl-L-carnitine 1090 Achondroplasia 551–564 craniovertebral dislocation 444 homozygous 560 management 553, 560–561, 561 neurologic complications 552–560, 552 Aciclovir 388, 1246, 1380–1381, 1380 Aciclovir-resistant herpes simplex virus type 1 (HSV-1) 1380 Acid-base disorders 366, 375–377, 376 clinical findings 375–376 gastrointestinal drugs 634, 638–639 laboratory investigations 376 management 376–377 pathophysiology 376 see also specific conditions terminology 375 Acidemia, defined 375 Acidosis, defined 375 Acoustic neuromas 1188 Acquired hemophilia 1052–1053 Acquired hepatocerebral degeneration (AHD) 663 Acquired immunodeficiency syndrome see AIDS Acquired phosphorus disorders 874 Acquired rubella infection 1349–1350 Acquired thrombophilia 1063, 1065, 1067

Acrodermatitis chronica atrophicans (ACA) 1576 Acromegaly 691–692, 697–698 Activated partial thromboplastin time (APTT) 86–87 ACTIVE (Atrial Fibrillation Clopidogrel Trial with Irbesartan for Prevention of Vascular Events) 121, 138 Acupuncture, fibromyalgia 523 Acute adrenal crisis (Addisonian crisis) 757 Acute areflexic paralysis 378 Acute cellular rejection (ACR) 1239, 1239 Acute coronary syndrome (ACS) 93, 94, 95 anticoagulation 101–103 antiplatelet therapy 101, 102 atherosclerotic plaques 99–100 etiology 96 nonischemic chest pain 96–97 thrombolysis 103–104, 104 Acute disseminated encephalomyelitis (ADEM) 1779 Acute dystonic reactions 635, 636 Acute encephalomyelitis 1346 Acute encephalopathy 79 Acute inflammatory demyelinating polyneuropathy (polyradiculoneuropathy) (AIDP) see Guillain–Barre´ syndrome (GBS) Acute intermittent porphyria (AIP) 841–842, 843, 844–846 seizures 846–847 treatment 846–847 Acute ischemic stroke 5 anticoagulation 66 thrombolysis 86 vascular/intensive care neurology 5 Acute liver failure (ALF) 645–659 artificial liver support systems 655–656 clinical presentation 646–647, 646 historical perspective 645–646 laboratory test/imaging 647–649, 648, 649, 650 management 649–651 cerebral edema 651–655, 652 outcome 655 transplants 649 Acute lymphoblastic leukemia (ALL) 1031, 1038, 1039 central nervous system (CNS) 1040

Acute lymphoblastic leukemia (ALL) (Continued ) clinical presentation 1039 leukostasis 1040–1041 treatment 1134, 1135 venous sinus thrombosis (VST) 1041 Acute motor axonal neuropathy (AMAN) 845 Acute myeloid leukemia (AML) 1038–1039 central nervous system (CNS) 1040 treatment 1134 Acute myelomonocytic leukemia (AAML), clinical presentation 1039 Acute myelopathy 1524 causes 1522, 1524–1525 Acute myocardial infarction (AMI) 93–111 clinical findings/diagnostic criteria 96 epidemiology 93–94 etiology 96 future directions 106 historical perspective 93 laboratory investigations 96–97, 97 management 100–104 intensive care 45–46 neuroimaging 97–99, 98, 99 pathology 99–100 prevention 104–106 prognosis 106 risk factors 94–96 thrombolysis 44–45, 45 Acute myopathy of intensive care 1234–1235 Acute painful neuropathy (APN) (insulin neuritis) 815 Acute promyelocytic leukemia (APL) 1040–1041 Acute renal disease see Renal disease, acute/chronic Acute renal failure (ARF) 1116 Acute respiratory distress syndrome (ARDS) 280, 1676 Acute respiratory failure see Respiratory failure (RF), acute/chronic Acute reversible cerebral angiopathy, diagnosis criteria 1729 Acute spinal fractures 455 Acute syphilitic meningitis 1533 Acute transverse myelitis 1525, 1540 Acute uremic encephalopathy 383–384 Adalimumab 601

I2 Adamkiewicz artery 224–225 ADAMTS13 protease 1114–1115, 1119, 1120 Addisonian crisis (acute adrenal crisis) 757 Addison’s disease (AD) 755–756, 799–800, 801 pathophysiology 755–756 Adefovir dipivoxil (Hepsera®) 679 mechanism of action 679 neurotoxicity 676, 679 Adenine deaminase (ADA) deficiency 828, 832 Adenomas gonadotropin-producing 693–694 nonfunctional 694 pituitary 685, 687–688, 1186–1187, 1187 thyroid-stimulating hormone (TSH)producing 693 Adenosine 132–133, 132 Adenosine diphosphate (ADP) receptor inhibitors 1129 Adenylsuccinate lyase (ADSL) deficiency 828, 831 ADEPT (Alzheimer’s Disease AntiInflammatory Prevention Trial) 580 Adrenal gland 749–772 disorders see Adrenal insufficiency (AI); Adrenal medulla disorders; Adrenoleukodystrophy (ALD)/ adrenomyeloneuropathy (ALM); Cushing syndrome (CS); Hyperaldosteronism function 749–750 historical perspective 621–623 Adrenal insufficiency (AI) 754–759 clinical manifestations 757–758, 757 destructive lesions 755–756 developmental defects 756 diagnosis 758–759 etiology/pathogenesis 754–757 historical perspective 754 secondary 689 steroidogenesis, impaired 756–757 treatment 759 Adrenal medulla disorders 765–766 clinical manifestations 765–766 diagnosis 766 etiology 765 historical perspective 765 treatment 766 Adrenalectomy, neurocardiogenic injury 20 Adrenocortical autoantibodies (ACA) 759 Adrenocorticotrophic hormone (ACTH) 686, 749 acromegaly 691–692 adrenal insufficiency (AI) 759 Allgrove syndrome 756 Cushing syndrome (CS) 750, 752–754 -dependent Cushing’s disease 692–693 excess 692 Adrenoleukodystrophy (ALD)/ adrenomyeloneuropathy (ALM) 759–762 clinical manifestations 760, 761

INDEX Adrenoleukodystrophy (ALD)/ adrenomyeloneuropathy (ALM) (Continued ) diagnosis 760–762, 762, 763 historical perspective 760 treatment 762 Adrenomyeloneuropathy (ALM) see Adrenoleukodystrophy (ALD)/ adrenomyeloneuropathy (ALM) ADVANCE (Action in Diabetes and Vascular Disease) trial 778–779 Advance care planning (ACP) 1222–1223 Adverse event, defined 1549 Advisory Committee on Immunization Practices (ACIP) 1554, 1555 African American ethnicity 93, 94, 95 African vipers (Bitis) 992–993 bush (Atheris) 992–993 AGE (advanced glycosylation end product) pathway 777 Agency for Health Care Research in Quality (USA) 1603 Aging comorbidities, hemophilia 1054 folic acid deficiency 931–932 premature 881 Aging Study Amsterdam 881–882 Agkistrodon spp 992–993 Agkistrodon halys 992–993 Agomelatine 1642 AIDS 755, 923 developing world 1776–1777 tropics 1527 tuberculosis (TB) 1485 vacuolar myelopathy (VM) 1529, 1530 Air stacking concept 284 AIRE gene 799 Akathisia 636 Akinetic mutism 1272 AL (immunoglobulin light chain) amyloidosis 1033, 1083, 1084, 1085, 1094–1096 central nervous system (CNS) 1085, 1095–1096 classification 1083, 1094 diagnostic criteria 1086, 1095 peripheral nervous system (PNS) 1084, 1095 ALADIN (Alpha-Lipinoic Acid in Diabetic Neuropathy) studies 779 Alagille syndrome, genetics 53 Albendazole 1426, 1433, 1434, 1436 gnathostomiasis 1431–1432, 1539 neurocysticercosis 1455, 1537 Albraxane 1202 Albumin 1758–1759 Albumin-collodion microencapsulated activated charcoal (ACAC) hemoperfusion 655 Alcohol 257 Aldose reductase inhibitors (ARIs) 779, 821 Alemtuzumab 507 Alendronate 878 ‘A-lines’ 41–42 Alitretinoin 1212–1213

Alkalemia, defined 375 Alkalosis defined 375 treatment 377 Alkylating agents 1134 adverse effects 1199–1201, 1208–1210, 1652 Allgrove (triple A) syndrome 756 Allograft procurement, lung 1238 Allograft recipients, intestinal 1287 Allopurinol 1654 Allylamines 1649 Alma-Ata Declaration (1978) 1774 Alpha-2d ligands 519, 521 a-Lipinoic acid (ALA) 779 Alteplase 104 Aluminum, excessive exposure 852 Alveolar echinococcosis 1426–1427 clinical manifestations 1426 diagnosis 1426–1427 pathogen 1426 prevention 1427 treatment 1427 Alzheimer-characteristic proteins 497–498 Alzheimer’s disease (AD) 9 breathing 247 developing world 1778–1779 folic acid deficiency 931–932 iron disorders 861 NSAIDs 581 vitamin B12 931–932 Alzheimer’s Disease International 1778–1779 Amantadine 1504 adverse effects 1641, 1641 American Academy of Neurology (AAN) 34, 52, 1221–1222 American Academy of Pediatrics (AAP) 1551 Cardiology and Cardiac Surgery Section 12 American Association of Poison Control Centers 898 American Association of Sleep Medicine (AASM) 261 American Association for the Study of Liver Diseases (AASLD) 653 American College of Cardiology (ACC) 83, 103, 157 Clinical Competence Statement on Invasive Electrophysiology Studies, Catheter Ablation and Cardioversion 139–140 American College of Chest Physicians (ACCP) 86–87, 290, 298, 300, 1638 Conference on Antithrombotic and Thrombolytic Therapy 118–119, 119 American College of Rheumatology (ACR) 450, 463, 465, 513, 514 American Diabetes Association 817 American Heart Association (AHA) 26–27, 28, 103, 111, 202–203, 1600, 1635 Cardiopulmonary Resuscitation and Emergency Care 30

INDEX American Heart Association (AHA) (Continued ) Clinical Competence Statement on Invasive Electrophysiology Studies, Catheter Ablation and Cardioversion 139–140 Heart Disease and Stroke Statistics 93 infective endocarditis 56, 83, 85–86 American Pain Society (APS) 518 American Porphyria Foundation 846 American Society for Testing and Materials International 97 American Spinal Injury Association (ASIA) 1763, 1764, 1766 American Stroke Association (ASA) 1635 Secondary Stroke Prevention Guidelines 1020 American Thoracic Society (ATS) 1338, 1490 Amfetamines 1644 Amikacin 1492, 1532 Amine, precursor uptake and decarboxylase (APUD) cells 803–804 5-Amino-4-imidazolecarboxamide ribotide transformylase/IMP cyclohydrolase (ATIC) deficiency 828, 831–832 Aminoglutethimide 754 Aminoglycosides 1490, 1492, 1646 Aminolevulinic acid (ALA) 843, 845 d-Aminolevulinic acid synthase (ALAS) 839–841, 843, 844, 846 Amiodarone 132, 133, 1637 Amisulpride 1642 Amitriptyline 515, 1090 Amlodipine 1636 Amoebiasis 1407, 1410–1411, 1410, 1410 Amoxicillin 1470, 1480 AMPAr-Abs (antibodies) 1162 limbic encephalitis (LE) 1168 Amphiphysin-Abs (antibodies) 1163 limbic encephalitis (LE) 1166, 1169 Amphotericin B 1248–1249, 1329–1330, 1329 Amphotericin B deoxycholate (AmBd) 1388–1389, 1395 Ampicillin 1247, 1379–1380 Amylin analog 820–821 Amyloid-b-related angiitis (ABRA) 487–488 Amyloidosis 1257 see also AL (immunoglobulin light chain) amyloidosis Amyotrophic lateral sclerosis (ALS) 247, 275–276 motor disability 1221 palliative care 1220, 1222–1223 Amyotrophy 776 Anagrelide 1075, 1133 Anakinra 459, 507 Analgesic drugs 1645–1651 Anaplasma spp 1403 Anaplastic astrocytoma 1184 Ancylostoma brasiliense 1432 Ancylostoma duodenale 1432

Anderson’s disease (chylomicron retention disease (CRD)) 623, 624 Androctonus 993 Androgen receptor insensitivity syndrome 793–796 Anemias 1003–1014 bone marrow failure syndromes 1009–1011 aplastic anemia 1010 background 1009 Fanconi’s anemia (FA) 1010 lead poisoning 1010–1011 pure red cell aplasia (PRCA) 1010 of chronic disease (AOCD) 1008 clinical consequences 1006–1007 definition/pathophysiology 1005–1006 diagnostic approach 1006 hemolytic 1011–1013 autoimmune (AIHA) 1012 background 1011 diagnosis/clinical findings 1011 glucose 6-phosphate deaminase (G6PD) deficiency 1012 hemoglobinopathies 1013 hereditary spherocytosis 1012 paroxysmal nocturnal hemoglobinuria (PNH) 1012–1013 treatment 1011–1012 iron deficiency 1007–1008, 1125–1126 causes 1007 clinical findings 1007–1008 laboratory findings 1008 treatment 1008 megaloblastic 927, 929–930, 930, 1009, 1125–1126 myelodysplastic syndromes (MDS) 1009 pernicious see Pernicious anemia (PA) vitamin deficiency see Vitamin B12 deficiency see also Folic acid deficiency Aneurysmal subarachnoid hemorrhage (aSAH) 1603, 1604 Angioma, cutaneous 1569–1573 Angiostrongylus cantonensis/ costaricensis 1432–1433, 1537–1538, 1538 clinical manifestations 1433 diagnosis 1433 epidemiology 1432 pathogen 1432–1433 therapy 1433 Angiostrongylus costaricensis see Angiostrongylus cantonensis/ costaricensis Angiostrongylus mackerras 1432–1433 Angiostrongylus malayensis 1432–1433 Angiotensin-converting enzyme (ACE) inhibitors 12, 165 Angiotensin-receptor blockers (ARBs) 165 Anion gap 375 acidosis 375 metabolic acidosis 376 Anisakiasis 1435 clinical manifestations 1435

I3 Anisakiasis (Continued ) diagnosis 1435 epidemiology 1435 pathogen 1435 therapy 1435 Anisakis spp 1431 Anisakis simplex 1435 Ankylosing spondylitis (AS) 440–441, 457 Annals of Surgery (Bigelow) 29 Anopheles spp 1429, 1514 Ant stings 995 Anterior cord syndrome 1763 Anthrax vaccine 1555–1556 Anthrax Vaccine Adsorbed (AVA) 1556 Anthrax Vaccine Expert Committee (AVEC) 1556 Antiallergic drugs 1651–1654 Anti-a4 integrin (natalizumab) 507, 601, 1607–1608 Antiandrogens 1655 Antiangiogenic drugs 1205, 1213 Antiarrhythmic drugs 132–136, 132, 1636, 1637 Antibacterial agents see Antibiotics Antiberiberi factor 891 Antibiotics 1120, 1247, 1479–1480, 1575–1576 adverse effects 388, 1203, 1211, 1644–1645, 1646 bacterial meningitis 1367–1369, 1368 infective endocarditis 83–84, 84 Antibodies 1160–1163, 1161 Antibody-mediated rejection (AMR) 1239, 1239 Anticholinergic drugs 1641, 1641 Anticoagulants 56, 86, 101–103, 1153 adverse effects 65–66, 1126, 1131–1132, 1638–1639 Anticoagulation system 1061, 1062 Anticonvulsants see Antiepileptic drugs (AEDs) entries Antidepressants 519–523, 519, 1332–1333, 1642 see also Tricyclic antidepressants (TCA) Antidiarrheal drugs 638 Antidiuretic hormone (ADH) 365, 367–369, 371, 750, 812 Antiemetic drugs 633–637, 634, 1655, 1656 Antiepileptic drugs (AEDs) 711–712, 1221 adverse effects 1639–1640, 1640 brain metastases 337, 1153 diabetic neuropathy 780, 781, 781 traumatic brain injury (TBI) 1628, 1758 Antiepileptic drugs (AEDs), hepatic/renal disease 417–432 dose adjustments 425 elimination 417, 418 specific drugs 417–429, 418 hemodialysis 427 Antiestrogens 1655 Antifolate analogs 1135 Antifungal agents 1388–1391, 1395 adverse effects 1645, 1649 Antigonadotrophin-releasing agents 1655 Antihelminthics (antihelmintics) 1539, 1650

I4 Antihistamines 1651–1654 Antihypertensive agents 9, 165, 165 adverse effects 1635–1636, 1636 Anti-inflammatory drugs 1645–1651 Antilipemic drugs 1636–1637, 1638 Anti-Ma-2 antibody syndrome 788, 789–792, 791, 793 Anti-Ma-2 protein antibodies 791 Antimalarials 1650 Antimetabolites 1134 adverse effects 1203–1204, 1205–1208, 1652 Antimicrobial therapy see Antibiotics Antimicrotubule agents 1135 Antimigraine agents 1650–1651 Antineoplastic drugs 1645, 1652 Antineutrophil cytoplasmic antibodies (ANCA) 1106 Anti-N-methyl-D-aspartate receptor (NMDAR) antibodies 791 antibody encephalitis 787, 792 antibody syndrome 787, 788, 789–792, 791, 793 Anti-osteoporotic agents 1655 Antiparasitic agents 1645, 1650 Antiparkinsonian drugs 1641, 1641 Antiphospholipid antibodies (APLA) 465, 466, 1063 Antiphospholipid syndrome (APLS) 289–290, 466, 1566–1567, 1600–1602 antibody positivity 1600 anticoagulant considerations, pregnancy 1602, 1603 catastrophic 1601–1602 diagnosis/treatment 1600 geoepidemiology 1600 manifestations CNS, treatment 1602 criteria 1601 non-criteria 1601 seronegative 1602 a2-Antiplasmin deficiency 1056 Antiplatelet agents 65, 86, 101, 479, 1020 adverse effects 1639 classification 101, 102 hematologic disorders 1129–1130, 1131, 1132 Antipsychotics 1642–1643 Antiretroviral therapy (ART) see Highly active antiretroviral therapy (HAART) Antiseizure medications 1231 Antispastic drugs 1640–1641 Antithrombotic agents 86–87, 1130–1131, 1635–1639 Antithrombotic Trialists’ Collaboration 101 Antithymocyte globulin (ATG) 1278–1279 Antithyroid agents 813–814, 813, 1655 Antithyroid antibodies 725 Anti-TNF (tumor necrosis factor) agents 718–719, 1718–1719 anti-TNF-a 598, 599, 601 Antitumor antibiotics 1652

INDEX Antitumor necrosis factor-a agents 479, 485–486 Antivenom 995, 997 Antiviral agents 1645, 1648 Anti-Yo antibody syndrome 788, 789–792, 791, 793 Anti-Yo (PCA-1) antibodies 791 Anton syndrome 35 Anxiety 465 Aortic bioprosthetic heart valves 64 Aortic diseases 223–238 calcific stenosis 62–63 neurologic complications 225–233 aortic aneurysm 225–226 aortic coarctation 54, 231–232, 231 aortic dissection 226–227 aortic surgery 229–231, 233–234, 233 giant cell arteritis (GCA) 227–229, 228 syphilis aortitis 232–233 Takayasu’s arteritis (TA) 229–231, 229, 230 spinal cord blood supply 223–225, 224 Apical ballooning syndrome see Stress cardiomyopathy Apis dorsata 995 Apis mellifera scutellata 995 Apixaban 122, 139, 295, 1639 Aplastic anemia 1010 Apnea-hypopnea index (AHI) 253, 261, 262 obstructive sleep apnea syndrome (OSAS) 257, 258, 260 Apolipoprotein B (APOB) 623–624 Apoplexy parathyroid 744 pituitary 686–687, 744 Aprepitant 637 Aquaporin (AQP) 396–397 Arachidonic acid pathway 577, 578 Arachnoiditis 1747 Araneomorphae 994 Arctic Sun® system 954 Argatroban 1131, 1132 Arginine vasopressin (antidiuretic hormone) 365, 367–369, 371, 750, 812 Armillifer spp 1436 Armillifer armillatus 1436 Arrhythmia 129–150 direct complications 129–131 cognition disorders 130–131 obstructive sleep apnea syndrome (OSAS) 258 stroke/cerebrovascular disease 129–130 epilepsy 175 syncope 171 treatment complications cardiology procedures 139–143 cardiopulmonary resuscitation (CPR) 131–132 cardiovascular surgery 143–144 drugs 132–139 see also Catheter ablation Arsenic trioxide 1204

Arsphenamine 1461–1462 Artemisinin 1517 Arterial gas embolism (AGE) see Decompression illness (DCI) Arterial hypertension 258 Arterial ischemic stroke (AIS) 405–407, 408–409 essential thrombocythemia (ET) 1076 inflammatory bowel diseases (IBD) 598 pre-eclampsia 1598–1602 risks 1066–1068 Arterial neuro-Behc¸et syndrome (NBS) 1710 Arterial tortuosity syndrome (ATS) 571 description 571 neurologic complications 571 Arteriovenous malformations (AVMs) hereditary hemorrhagic telangiectasia (HHT) 1572 pregnancy 1604 radiotherapy (RT) 1189, 1193 Artesunate 1517 Arthrogryposis multiplex congenita (AMC) 1610 Arthropod-borne viruses 1378, 1379, 1381 Artificial liver support devices (ALSD) 655 Arts syndrome 828, 830 Ascaridia 1433 Ascaris lumbricoides 1433, 1539 Ascorbic acid see Vitamin C deficiency Aseptic meningitis 322, 579–580, 579 ibuprofen-induced meningitis 1648 Asiatic arboreal pit viper (Trimeresurus complex) 992–993 Aspart 815 Aspergillosis 1385, 1390, 1395–1396 Aspergillus spp 1041, 1383 clinical syndromes 1301–1302, 1385, 1386, 1387, 1535 aspergillosis 1385, 1390, 1395–1396 endocarditis 67–68 echinocandins 1389 imaging 1387, 1390 investigation 1386, 1387 transplantation 1248, 1248, 1278–1279, 1287–1288 Aspergillus flavus 1386, 1395–1396 Aspergillus fumigatus 1232, 1241, 1248, 1395–1396, 1532 liver transplantation 1261, 1262 Aspergillus niger 1395–1396 Aspergillus terreus 1395–1396 Aspirin 101, 113, 199, 953, 1129, 1571 adverse effects 1639, 1647, 1648 infective endocarditis 69, 86 intracranial hemorrhage 138 stroke 122, 578–579, 580, 581 Astasia/astasia-abasia 183 Asymmetric lower limb neuropathy 776 Asymptomatic neurosyphilis 1464, 1533 Atalimuma 507 Ataxia cerebellar see Cerebellar ataxia degenerative 9

INDEX Ataxia (Continued ) fragile X-associated ataxia syndrome (FXTAS) 793 Friedreich’s (FA) 9 gait 183–184 gluten (GA) 609–611, 610 myoclonic 612–613 Atenolol 1636 ATHENA (A Trial with Dronedarone to Prevent Hospitalization or Death in Patients with Atrial Fibrillation) trial 134 Atheris 992–993 Atherosclerosis Risk in Communities (ARIC) study 130 Atlantoaxial subluxation 442 Atlas: Country Resources for Neurological Disorders 2004 (WHO) 1773 Atlas: Epilepsy Care in the World (WHO) 1775–1776 Atlas: Multiple Sclerosis Resources in the World (WHO/MSIF) 1779 Atomoxetine 1644, 1645 Atorvastatin 9 Atovaquone 1325 ATP7A gene 626, 853 ATP7A-related conditions copper transport disorders 853–854, 853 distal motor neuropathy (DMN) 853, 854 neuropathy 853 ATP7B-related copper transport disorders 854–857 Atractaspididae 987 ATRIA study (atrial fibrillation-related thromboembolism) 119, 119 Atrial aneurysm surgery 194 Atrial fibrillation (AF) 117–118 arrhythmia 129 catheter ablation 153, 155 internal cardioversion 140 external cardioversion 139 intracranial hemorrhage 138 preoperative 144 rheumatic valvular heart disease (RVHD) 61–62 warfarin 122–123, 123 Atrial flutter 62 Atrial myxoma 211–216, 211 natural history 214–215, 214 pathology 215–216 structural neurologic complications 212–214 systemic effects 212 Atrial septal aneurysms 52 surgery 200 Atrial tachyarrhythmias 118 Atrial vein isolation 143–144 Attention deficit hyperactivity disorder (ADHD) 1644 Attenuated Streptococcus 75–76 Atypical neuroaxonal dystrophy (ANAD) 859–860 Australian elapids 992

Australian National Polio Reference Library 1527 Autoantibodies 501–502 Autoimmune diseases 1694 Autoimmune hemolytic anemias (AIHA) 1012 Autoimmune polyendocrine syndromes (APS) 799 Autoimmune polyglandular syndrome, type I (APSI) 799–801, 800 clinical findings 800 historical perspective 799–800 laboratory investigations 800 management 801 neuroimaging 800 pathology 801 Autoimmune polyglandular syndrome, type II (APSII) (Schmidt’s syndrome) 801–802 clinical findings 801 historical perspective 801 laboratory investigations 801–802 management 802 natural history 801 neuroimaging/pathology 802, 802 Autoimmune polyglandular syndrome, type III (APSIII) 802–803 clinical findings 803 historical perspective 803 laboratory investigations 803 management 803 natural history 803 neuroimaging/pathology 803 Autoimmune-mediated destruction of glandular tissue 799 Autonomic dysreflexia (AD) 1766 Autonomic nervous system (ANS) autonomic failure 172, 174 diabetic neuropathy (DAN) 773, 774–775, 781–782 diphtheria 1357 dysautonomia 517 insufficiency 353 pancreas transplantation 1283–1285, 1284 paraneoplastic disorders (PNS) 1171–1172 with pseudo-obstruction 1172 renal disease 390–391 rheumatoid arthritis (RA) 452 scleroderma (systemic sclerosis) (SSc) 469 Sj€ ogren’s syndrome (SS) 471 tetanus 1509 Autosomal dominant polycystic kidney disease (ADPKD) 1251 Avermectins 1650 Axillary artery catheterization 6–7 Axonal and demyelinating neuropathy 1273 5-Azacitidine (Vidaza®) 1133 5-Aza-2´deoxycitidine (decitabine) 1133 Azasetron 636

I5 Azathioprine 354, 459, 485–486, 507, 1231, 1613 giant cell arteritis (GCA) 229, 481 neuro-Behc¸et syndrome (NBS) 1718–1719, 1720 neurosarcoidosis 325, 326, 327 Azithromycin 1403–1404 Azole drugs 1389, 1395 Azoospermia 794

B B cell lymphomas 1263 Babesia spp 1406–1410 Babinski sign 1545 Bacillus anthracis 1556 Baclofen 1090, 1332, 1640 Bacterial infection 1247–1248, 1554–1556 Bacterial meningitis 68, 1361–1375 clinical presentation 1363–1365 cerebrospinal fluid (CSF) shunts, infection 1365 community-acquired 1363–1365 post-traumatic 1365 epidemiology 1361–1362 genetics 1362 management 1365–1373, 1366, 1367 adjunctive therapies 1366, 1369–1372 antimicrobial therapy 1367–1369, 1368 cerebrospinal fluid (CSF) analysis 1365–1367 lumbar puncture, repeat 1373 recurrent 1372–1373 outcome 1372, 1373 pathophysiology/pathology 1362–1363, 1364 Bacterial myelopathy 1530–1532 Bacterial toxins 988 Bacteroides spp 80–81 Bair Hugger™ rewarming technique 953–954 Balamuthia mandrillaris 1411 Balloon kyphoplasty, complications 1747–1748, 1748 Banana spiders 994 Banded sea snake (Hydrophis fasciatus atriceps) 992 Barbiturate coma, traumatic brain injury (TBI) 1628 Barbiturates 1624, 1630, 1631 ‘Barefoot doctors’ 1774 Bariatric beriberi 589 Bariatric surgery 587–592 copper deficiency myelopathy (CDM) 590 metabolic disorders 591–592 miscellaneous disorders 592 neurologic complications 588–589, 588 nutritional deficiency 588, 588, 591–592 vitamin B1 deficiency 589 see also Obesity Bartonella spp 80–81, 1403 Bartonella henselae 1530 Bartter’s syndrome 872 Basal ganglia calcifications 377 Basal-bolus regimen 814

I6 Bath Ankylosing Spondylitis Disease Activity Index (BASDAI) 454 Bath Ankylosing Spondylitis Functional Index (BASFI) 454 Baylisascariasis 1539–1540 Baylisascaris procyonis 1537–1538, 1538, 1539–1540 Beaked sea snake (Enhydrina schistose) 992 Becker muscular dystrophy (BMD) 10–12, 11, 125 Bee stings 995 Behavioral problems brain metastases 1145 fibromyalgia 517 management 263 neurosarcoidosis 311 Behc¸et syndrome (BS) 1703–1723 diagnosis/manifestations 1703–1705, 1704 laboratory investigations 1705 epidemiology 1703 nervous system see Neuro-Behc¸et syndrome (NBS) pathology/pathogenesis 1705–1707 Bell, Sir Charles 435 Bell’s palsy 775 Benign angiopathy of central nervous system (BACNS) 1725–1726, 1726 Benzimidazoles 1426, 1650 Benznidazole 1414 Benzodiazepines 636–637, 1508–1509, 1625, 1631, 1640 adverse effects 1640, 1644 Benzylpenicillin see Penicillin G Bereavement care 1223 Beriberi 891 bariatric 589 dry 896–897 infantile 897 b-Blockers 12, 165 Betamethasone 1613 Betrixaban 139 Bevacizumab 339–340, 1152–1153, 1184, 1584 adverse effects 1205, 1213 Bicarbonate 375 Biguanides 816–818, 1654 Bilharziasis (schistosomiasis) 1419–1421, 1535–1536 diagnosis 1420, 1535–1536 differential diagnosis 1536 epidemiology 1419, 1535 features 1420, 1535 microbiology 1535 neuroimaging 1535, 1536 pathogen 1419–1420, 1419 pathogenesis 1535 treatment 1421, 1536 Bing–Neel syndrome 1033, 1093–1094 Bioartificial liver (BAL) 655–656 BIOMED Study of Stroke Care Group 106 Bioprosthetic heart valves 64–65 Biotin see Vitamin B7 deficiency; Vitamin B8 deficiency

INDEX Bismuth 638 Bisphosphonates 536–537, 878, 1090–1091 Bites see Venomous bites Bitis 992–993 Bivalirudin 1131, 1132 Black box warning 326–327 Black snake (Pseudechis) 992 Blastocystis spp 1410 Blastomyces 1535 Blastomyces dermatitidis 1383, 1384, 1385, 1397 Blastomycosis 1385, 1397 Blastomycosis spp 1386 BCR-ABL tyrosine kinase inhibitors 1133 Bleeding disorders see Congenital bleeding disorders; Hematologic disorders, treatment Bleeding risk, warfarin 122–123 Bleomycin 1211 Blind-loop syndrome 1541 Bloch–Sulzberger syndrome see Incontinentia pigmenti (IP) Blood culture, cerebrospinal fluid (CSF) analysis 1367 Blue spotted sea snake (Hydrophis cyanocinctus) 992 Body mass index (BMI) 587 Body temperature 945–958 heat exchange, mechanisms 947 heat-related illness 948–949 clinical features 948, 948 laboratory studies 948–949 management 949 therapeutic manipulation 954–955 internal methods 954 external methods 954 see also Fever; Hyperthermia; Hypothermia Bone marrow failure syndromes see under Anemias Bone marrow transplantation (BMT) 1022, 1295–1305 historical perspective 1295–1296 Bone mineral density (BMD) 741–742, 869, 876 Border zone cerebral infarction 131 Borderline lepromatous leprosy 1573–1575 Borderline tuberculoid leprosy 1573–1575 Bordetella parapertussis 1358 Bordetella pertussis 1358, 1555–1556 Borrelia 1532, 1533, 1534–1535 Borrelia afzelii 1476 Borrelia burgdorferi 1532–1533, 1576–1577 see also Lyme disease Borrelia burgdorferi sensu lato 1476 Borrelia burgdorferi sensu stricto 1476 Borrelia garinii 1476 Borrelia recurrentis 1534–1535 Borreliosis see Lyme disease Bortezomib (Velcade®) 1032, 1089, 1090, 1136–1137 adverse effects 1200, 1203, 1652 Bortezomib-induced peripheral neuropathy (BiPN) 1089 Boston Circulatory Arrest Trial 55

Boston Collaborative Drug Surveillance Program 134–135 Bothrops 992–993 Botulinum toxin 1332, 1640–1641 Boyle’s Law 959, 965 Brachial artery catheterization 5–6 Brachial plexopathy 199, 199, 345–346, 1192–1193 Bradycardia 140 Brain conditions abscess 68, 79, 81, 83, 1147, 1149, 1248 amoebic 1407, 1410–1411, 1410, 1410 anomalies, congenital heart disease (CHD) 53–54 biopsy 1326 edema 365, 1261–1262, 1262 embolism 67–68 global injury 1191–1192 metastases see Brain metastases necrosis 1191 trauma see Traumatic brain injury (TBI) tumors see Brain tumors see also Inherited neurodegeneration with brain iron accumulation (NBIA) Brain metastases 335–340, 792, 1141–1157 clinical findings 336, 1144–1145, 1145 diagnostic procedures 1146–1149 known cancer patients 1145, 1146–1149, 1147, 1148 suspected cancer patients 1149 differential diagnosis 1149–1150 incidence 335–336, 336, 1143–1144 pathology/pathophysiology 1145–1146 primary tumors 726, 1143–1144 treatment 336–340, 1150–1153, 1150 chemotherapy 339, 1152–1153 hormone therapy 1152–1153 interstitial brachytherapy 339 molecular targeted agents 339–340, 1152–1153 stereotactic radiosurgery (SRS) 338–339, 1151–1152 supportive drugs 1153 surgery 337–338, 1151 whole-brain radiation therapy (WBRT) 337, 340, 1150–1151, 1152 Brain Tumor Study Group 1184 Brain tumors 726, 1580–1584 neurocutaneous disorders 1580–1584 pregnancy 1614–1616, 1615 Brain-heart connection 20, 26 Brainstem auditory evoked responses (BAERs) 315, 316 breathing control 241–243, 242 disease 245–247, 247 encephalitis 351 Brainstem auditory evoked response (BAER) 761 Branch retinal artery occlusion 198 Branched-chain amino acids (BCAA) 670–671 Bratton–Marshall reaction 831

INDEX BRAVO (Blockade of the IIb/IIIa Receptor to Avoid Vascular Occlusion) trial 101 Breast cancer 1144 Breast feeding 1606, 1608 Breath holding spells 175 Breathing 239–250 Cheyne–Stokes respiration pattern (CSRP) 263–264, 264, 278 neuroanatomy 241–243, 251–252 cortical control 241, 242 subcortical/brainstem control 241–243, 242 neurochemical control 244–245 neurologic conditions 245–248, 246 central (CNS)/peripheral nervous system (PNS) 246, 248 cortex/subcortical structures/ brainstem 245–247, 247 neuromuscular junction disorders 248 spinal cord diseases 247–248 physiologic control 243–244 pacemaker neurons 243–244, 244 rhythm generation 243 Brentuximab 1205 British Infection Society 1490 British Medical Journal 75–76 British Thoracic Society 1338 Bromides 846 Bromocriptine 697–698, 810, 811, 1655 Bronchiolitis obliterans syndrome (BOS) 1239, 1239 Bronchodilators 1655 Brown snake (Pseudonaja) 992 Brucella spp 80–81, 1531, 1532 Brucella abortus 1531 Brucella melitensis 1531 Brucella ovis 1531 Brucellosis 1531–1532, 1531 Brugia malayi 1429 Brugia timori 1429 Bruns–Garland syndrome 776 Bruxism 257 Buerger disease 488 clinical features 488, 488 Buformin 816 Bulbar dysfunction 1356 ‘Bull neck’ 1356 Bungarus spp 991–992 Bungarus caeruleus 991–992 Bungarus candidus 991–992 Bungarus fasciatus 991–992 Bungarus multicinctus 989, 990, 991–992 Bungarus niger 991–992 Bupivacaine 994 Bupropion 1644, 1645 Burkitt lymphoma (BL) 1030–1031 ‘Burning feet’ symptom 1542–1543 ‘Burning hands syndrome’ 1762 Busulfan 1075, 1210, 1299 Buthus 993 Buthus matensi Karsch 993–994

C Cabergoline 697–698, 810, 811, 1655 Cadmium, excessive exposure 852 Cafe´ au lait spots 1581–1582, 1581 Calcific valvular heart disease 62–63 Calcineurin inhibitors (CNI) 675–677 heart transplantation 1231 liver transplantation 1260 mechanisms of action 675 neurotoxicity 675–676, 676, 1245–1246, 1246 specific complications 676–677 treatment 677 Calcitonin (CT) 737, 866, 878 Calcium 737–738, 865, 868 malabsorption 1286 Calcium channel blockers 31, 165 Calcium glutonate 870 California Encephalitis Project 1377 Call syndrome see Reversible cerebral vasoconstriction syndrome (RCVS) Call–Fleming syndrome see Reversible cerebral vasoconstriction syndrome (RCVS) Callitroga (screwworms) 1436 Calloselasma rhodostoma 992–993 Campylobacter jejuni 9–10, 1552, 1611 Cancer, radiation 1745 Candida spp 1383, 1384, 1385, 1386, 1389 central nervous system (CNS) 1394–1395 endocarditis 67–68 imaging 1388, 1389 investigation 1386, 1387 meningitis 1385, 1386 transplantation 1232, 1278–1279, 1301–1302 Candida albicans 1301–1302, 1385, 1386, 1394 Candida glabrata 1301–1302, 1394–1395 Candida krusei 1301–1302 Candidal infections 1389, 1394–1395 Candidiasis 1385 Cannabinoids 519, 522–523 Cannon, Walter B 19 Capecitabine 1204, 1207 Capreomycin 1490, 1491, 1492 Capsaicin 780 Carbamazepine 677, 847, 1153, 1231, 1234 adverse effects 1640, 1640, 1644 elimination 418, 422, 425, 427 Carbapenems 1646 Carbimazole 813 Carbohydrates, absorption 622 Carbon dioxide 375 Carbon ions 1183 Carbon monoxide (CO) intoxication 971–980 clinical findings 973–974, 973 acute intoxication 974 chronic intoxication 974 delayed neuropsychiatric syndrome 974, 975

I7 Carbon monoxide (CO) intoxication (Continued ) diagnosis 974–975 differential diagnosis 976 laboratory tests 975–976 neuroimaging 976 pathology 972, 973 pathophysiology 971–973, 972 prognosis 977–978 treatment 976–977 Carboplatin 339, 1134, 1200 Carcinoma, ovarian 791–792 Carcinoma-associated retinopathy 351 Cardiac arrest 25–39 epidemiology 26–28 in-hospital 26–27 out-of-hospital 27–28 imaging 34 long-term complications 34–35 neurologic complications 25–26 prognosis 31–34 treatment 28–31 basic measures 28–29 neuroprotective pharmacology 31 sedation/neuromuscular blockade 30–31 seizure control/prevention 31 therapeutic hypothermia 29–30 Cardiac arrhythmia see Arrhythmia Cardiac asystole 175 Cardiac autonomic neuropathy (CAN), diabetic 10 Cardiac catheterization 194, 196–197 -related stroke 95–96 Cardiac involvement, Fabry disease (FD) 1564 Cardiac lesions 52 Cardiac myxomas 1566 Cardiac surgery/interventions 193–208 clinical findings 193–196 historical perspective 193 investigations 202–203 management 203–204 natural history 201–202 pathology 196–201 Cardiac syncope 171, 179 clinical features 171 etiology 171 pathophysiology 171 treatment 186 Cardiac tests/procedures 41–48 acute myocardial infarction, management 45–46 future directions 46 historical perspective 41 laboratory investigations 45 neurologic complications 41–45 central nervous system (CNS) 43–45 peripheral nervous system (PNS) 41–43 Cardiac Transplant Research Database (CTRD) 1233 Cardiac tumors 209–222 clinical triad 209–211

I8 Cardiac tumors (Continued ) cardiac dysfunction 209–210 constitutional symptoms 210–211 embolic phenomena 211 diagnosis 217–218 echocardiography 217 imaging 217 pathology 218 neurologic complications 209–222 primary 209, 210, 216 secondary 216–217 specific 211–217 surgery 194–195, 200–201 treatment/prognosis 218–219 Cardiocerebral resuscitation (CCR) 26 Cardiogenic embolism 1598 Cardiology arrhythmia 139–143 interventional see Cardiac surgery/ interventions movement disorders/ neurodegenerative diseases 8–9 neuromuscular disorders 9–13 paroxysmal events 7–8 vascular/intensive care neurology 3–7 management 7 pathophysiology 3–5, 4 Cardiomyopathy 111–129 atrial fibrillation (AF) 117–118, 122–123, 123 bleeding risk, warfarin 122–123 classification 111, 112 definition 111 left ventricular dysfunction 113–115, 115 mitochondrial dysfunction 125–126 patent foramen ovale (PFO) 112–113 risk stratification schemes 118–122, 119, 120, 122 stress cardiomyopathy 115–117 Cardiopulmonary bypass (CPB) 1238 Cardiopulmonary resuscitation (CPR) arrhythmia 131–132 survival rates 27–28 see also Cardiac arrest Cardiovascular disorders autonomic neuropathy (CAN) 774–775 Behc¸et’s syndrome 1705 Obstructive sleep apnea syndrome (OSAS) 258 Paget’s disease 878 primary hyperparathyroidism (PHPT) 867 respiratory failure (RF) 280 surgery 143–144 see also Cardiology Cardiovascular drugs 1635–1639 Cardiovascular implantable electronic devices 85–86 Carfilzomib 1089–1090, 1203 Carmustine (BCNU) 1208–1209 Carney’s complex (CC) 211, 1562, 1566 Carotid artery dissection 964 Carotid cavernous fistula (CCF) 1604

INDEX Carotid sinus massage 186 Carotid sinus syndrome (CSS) 175, 176 Carotid-cavernous fistula 567–568 characteristics/management 567–568 mechanisms/frequency 567 spontaneous 567, 574 Carpal tunnel syndrome (CTS) 390, 401, 449, 450 diabetes mellitus (DM) 775 pregnancy 1608–1609 Carpet vipers 992–993 CASCADE (Concerted Action on Seroconversion to AIDS and Death in Europe) cohort 1339 Caspofungin 1248 CASPR2-Abs (antibodies) 1161 limbic encephalitis (LE) 1167 Cataplexy/cataplexy-like episodes 182, 183 Cataracts, radiation 1745 Catastrophic antiphospholipid syndrome (CAPS) 466 Catastrophic life events 26 Catastrophic thinking 517 Catecholamines 20 Catechol-O-methyl transferase (COMT) inhibitors 1641 Catheter ablation 151–160 clinical history 151–154 genetics 155 historical perspective 151 laboratory investigations 155 management 156 natural history 154–155 neuroimaging 155 pathology 156 Catheter angiography 1692, 1735 complications 1743–1745, 1744 Catheterization, complications 5, 43–44 Cauda equina syndrome 456, 1763 Cauliflower ear 458 Cefepime 1379–1380, 1644 Cefotaxime 1369, 1479, 1480 Ceftazidime 1369 Ceftriaxone 88, 1379–1380, 1532 bacterial meningitis 1369 Lyme disease 1479, 1480, 1533 neurosyphilis 1337, 1468–1469 Cefuroxime axetil 1480 Celecoxib 580 Celiac disease (CD) 624–625 see also Gluten-related diseases (GRD) CellCept see Mycophenolate mofetil (MMF) Cement extravasation 1747 Cement leakage 1747 Center for Epidemiologic Studies Depression (CES-D) scale 881–882 Centers for Disease Control and Prevention (CDC) 587, 1324–1325, 1465–1466 neurosyphilis 1468, 1469–1470, 1534 tuberculosis (TB) 1338, 1490 vaccines 1549, 1553 Centers for Medicare and Medicaid Services 283

Central American mountain pit vipers (Cerrophidion) 992–993 Central Brain Tumor Registry in the United States (CBTRUS) 1187–1188 Central cord syndrome 1762 Central diabetes insipidus (DI) 690 Central hyperthermia syndromes 639 Central hypothyroidism (CH) 689 Central nervous system (CNS) AL amyloidosis 1095–1096 angiitis see Primary angiitis of central nervous system (CNS) bleeding, treatment 1051 breathing 246, 248 cardiac tests 43–45 chemotherapy complications 1205–1213 dialysis patients 395–400, 396 Ehlers–Danlos syndromes (EDS) 570 fibromyalgia 516–517 fungal infections see Fungal infections HAART penetration effectiveness 1340 heart transplantation 1229, 1230, 1232, 1233 helminthic infestation 1414–1436 hemolytic uremic syndrome (HUS) 1113, 1115–1118, 1118 hemophilia 1053 immunosuppressants 676 infections, developing world 1776–1777 infective endocarditis 68, 79 inflammatory bowel diseases (IBD) 596, 600–601 intestinal transplantation (ITx) 1272, 1273 leukemias 1040 liver transplantation 1262–1263 Lyme disease 1475 lymphoma 1035 see also Primary central nervous system lymphoma (PCNSL), HIVassociated malignancies 1278–1279 metastases 1143 monoclonal gammopathy of undetermined significance (MGUS) 1088 multiple myeloma (MM) 1090–1092 neuro-Behc¸et’s syndrome (NBS) 1708–1711, 1712 pancreas transplantation 1278–1279, 1281–1282 paraneoplastic neurologic syndromes (PNS) 1160, 1163–1170 parasympathetic outflow tracts 4 parenchymal disease 309–312, 322–323 plasma cell disorders 1085 renal disease 383–388 renal transplantation 1247–1251, 1252 scleroderma (SSc) 468, 469 sickle cell disease (SCD) 1015 Sj€ ogren’s syndrome (SS) 470 small bowel transplantation 1278–1279, 1287–1288 sympathetic outflow tracts 4

INDEX Central nervous system (CNS) (Continued ) thrombosis 1075 toxoplasmosis see Toxoplasmosis entries tuberculosis (TB) see under HIVinfected patients; Tuberculosis, central nervous system (CNS) vitamin D 879–882 Waldenstr€om macroglobulinemia (WM) 1093–1094 Central neurogenic hyperventilation 375–376 Central pontine (extrapontine) myelinolysis (CPM) 368, 1278–1279 Central retinal artery occlusion 198 Central sensitization, fibromyalgia 516 Central sleep apnea (CSA) 263–266 classification 278 primary 263, 263 respiratory failure (RF) 278 Central venous catheterization 43 access catheters 44, 45 Centruroides 988, 993, 994 Centruroides exilicauda 993 Cephalosporins 1247, 1369, 1379–1380, 1403–1404, 1533 adverse effects 1646 Cerastes 992–993 Cerebellar ataxia 708 see also Subacute cerebellar degeneration (SCD) Cerebellar degeneration 347, 349–351 see also Subacute cerebellar degeneration (SCD) Cerebral amyloid angiopathy (CAA) 487–488 Cerebral aneurysms 54, 54, 213, 1719 Cerebral angiography 1732–1733, 1735, 1736 Cerebral autosomal dominant arteriopathy with subcortical infarcts and leukoencephalopathy (CADASIL) 123, 1614 Cerebral edema 371–372 management 651–655, 652 Cerebral effects, neuroanesthesia 1625, 1627 Cerebral embolism 51, 51 air 153–155, 156 cardiac catheterization 43–44 Cerebral hemorrhage 1778 Cerebral hypoperfusion 170–176 Cerebral imaging 217 Cerebral infarctions patent foramen ovale (PFO) 113 ‘silent’ 1016 Cerebral malaria 1514–1515, 1517 Cerebral metastases 791–792 Cerebral performance categories (CPC) 28 Cerebral perfusion pressure (CPP) 651, 1753 Cerebral schistosomiasis 1777 Cerebral vasculitis 475–494, 1104 diagnosis 476

Cerebral vasculitis (Continued ) differential diagnosis 475, 476 infectious origin 476–477 inflammatory bowel diseases (IBD) 598–599 noninfectious inflammatory 477–488, 477 patient evaluation 475–476, 476 rheumatoid arthritis (RA) 451–452, 452 Cerebral vasospasm 1725 Cerebral venous sinus thrombosis (CVST) 407–408, 408 essential thrombocythemia (ET) 1076 inflammatory bowel diseases (IBD) 599–600 inherited thrombophilia 1064–1065 neuro-Behc¸et syndrome (NBS) 1708–1709, 1710–1711, 1715, 1717, 1718 polycythemia vera (PV) 1075 pregnancy 1605–1606 primary myelofibrosis (PMF) 1078 risks 1064–1066 hyperthyroidism 723 treatment 1719–1720 Cerebral venous thrombosis (CVT) see Cerebral venous sinus thrombosis (CVST) Cerebral/spinal cord ischemia 233–234, 569–570 characteristics/management 569–570 mechanisms/frequency 569–570 Cerebrospinal fluid (CSF) analysis 1365–1367 blood culture 1367 fungal infections 1386 Hashimoto’s encephalopathy (HE) 725 Henoch–Sch€ onlein purpura (HSP) 1107 neuro-Behc¸et syndrome (NBS) 1715 sarcoidosis 315 serum markers, inflammation 1367 shunts, infection 1365 skin biopsy 1367 Cerebrotendinous xanthomatosis (CTX) 1562, 1586–1587 clinical symptoms 1586, 1587 diagnosis/testing 1587 radiology 1586–1587 treatment 1587 Cerebrovascular disorders arrhythmia 129–130 congenital heart disease (CHD) 51–52, 51 Ehlers–Danlos syndromes (EDS) 570 Henoch–Sch€ onlein purpura (HSP) 1103–1104 HIV 1322 homocystinuria 572 characteristics/management 572 mechanisms/frequency 572 infective endocarditis 77–79 inflammatory bowel diseases (IBD) 597–599

I9 Cerebrovascular disorders (Continued ) intestinal transplantation (ITx) 1272, 1274–1275 Loeys–Dietz syndrome (LDS) 571 multiple organ transplantation 1311 neuropsychiatric systemic lupus erythematosus (NPSLE) 465 osteogenesis imperfecta (OI) 573 pseudoxanthoma elasticum (PXE) 571–572 radiotherapy (RT) 1193 renal transplantation 1251 Sneddon syndrome (SS) 1566–1567 Cerebrovascular phenomenon, intracranial hemorrhage (ICH) 1016 Cerrophidion 992–993 Certolizumab 601 Cervical artery dissection (CAD) 567, 568, 569, 574 characteristics/management 568 mechanisms/frequency 568 Cervical spinal cord injury (SCI) 276–277, 1629 Cervical spine stenosis, surgery 547, 547, 548 Cervical thoracic orthotic (CTO) device 1767 Cestodes (tapeworms) 1414, 1422–1428 Cetirizine 1654 CHADS score 118–122, 120, 122, 123, 142 Chagas disease 1413–1414 Chalastic attacks 182 Charcot–Marie–Tooth (CMT) disease pregnancy 1612 respiratory failure (RF) 277 Charcot–Marie–Tooth inherited neuropathy (CMTX5) 830 CHARGE (coloboma, heart disease, choanal atresia, retarded growth, genital/urinary anomalies, ear anomalies and deafness) syndrome 52, 53 CHARISMA (Clopidogrel for High Atherothrombotic Risk and Ischemic Stabilization, Management and Avoidance) trial 101 Chemokines 507 Chemotherapy 1199–1218 adverse effects 388 agents inducing neuropathy 1200 brain metastases 339, 1152–1153 central nervous system (CNS) 1205–1213 alkylating agents 1208–1210 antibiotics 1211 antimetabolites 1205–1208 biologic/targeted therapies 1212–1213 mitotic spindle inhibitors 1210–1211 chemobrain 1213 Hodgkin lymphoma (HL) 1035 leukemia/lymphoma/multiple myeloma (MM) 1134–1136 peripheral nervous system (PNS) complications 1199–1205

I10 Chemotherapy (Continued ) alkylating agents 1199–1201 antibiotics 1203 antimetabolites 1203–1204 biologic/targeted therapies 1204–1205 mytotic spindle inhibitors 1201–1203 prevention/rehabilitation 1205 proteasome inhibitors 1203 Chenodeoxycholic acid (CDCA), cerebrotendinous xanthomatosis (CTX) 1587 Chest wall disorders 265, 266 Cheyne–Stokes respiration pattern (CSRP) 263–264, 264, 278 Chickenpox see Varicella zoster virus (VZV) Child–Pugh score 665 Children achondroplasia 552, 554 arterial ischemic stroke (AIS) 1067–1068 bacterial meningitis 1369 cerebral venous sinus thrombosis (CVST) 1065–1066 congenital heart disease (CHD) 52–53, 55, 56 excessive sleepiness 254 heart transplantation 1234–1235 Lyme disease 1474 obstructive sleep apnea (OSA) 251, 254, 261 percutaneous coronary intervention (PCI) 197 respiratory problems 561 sickle cell disease (SCD) 1020 sudden death 556 tuberculous meningitis (TBM) 1487, 1490–1492 ChimeriVax-JE vaccine 1554 Chinese krait (Bungarus multicinctus) 989 Chlamydia spp 80–81 Chlorambucil 1209, 1718–1719 Chloramphenicol 1646 Chloromas (granulocytic sarcomas) 1040 Chloroquine 325, 326 Chlorpromazine 634, 951 ‘Chokes’ 962 Choking 254 Cholestatic hepatic failure 1286 Cholesterol emboli syndrome (CES) 1562, 1564–1566 clinical presentation 1564–1565 cutaneous 1565, 1565 neurologic 1565 diagnosis 1565–1566, 1565 treatment 1566 Cholesterol-lowering agents 9 Chordoma 445, 446 Chorea 715–716 polycythemic 1075 Choreoathetosis 56 Choriocarcinoma 217 Chronic gastrointestinal pseudoobstruction 1160

INDEX Chronic graft-versus-host disease 1302 Chronic inflammatory demyelinating polyneuropathy (polyradiculoneuropathy) (CIDP) 841, 845, 1088, 1333, 1611 Chronic kidney disease (CKD) see Renal disease, acute/chronic Chronic lymphoblastic leukemia (CLL) 1040–1041 Chronic morning headache (CMH) 254 Chronic myelogenous (myeloid) leukemia (CML) 1039, 1040–1041 treatment 1132, 1133 Chronic myelopathy 1524 Chronic myeloproliferative diseases 1073–1081 definitions 1073–1074 neoplasms (MPN) 1038–1039, 1132–1133 Chronic obstructive pulmonary disease (COPD) 279–280, 281 Chronic respiratory failure see Respiratory failure (RF), acute/chronic Chronic retroviral myelopathy 1527–1530 Chronic uremic encephalopathy 383–384 Churg–Strauss angiitis 484–486 clinical features 485, 485 treatment 485–486 Chvostek’s sign 377, 743 Chylomicron retention disease (CRD) (Anderson’s disease) 623, 624 Ciclosporin 326, 354, 485–486, 507, 1613, 1718–1719 adverse effects 601, 675, 676, 1201, 1245, 1278–1279 transplantation 1231, 1260, 1281–1282 Ciguatera 964, 988 Ciguatoxin 996 Cilindrocarpum lucidum 1299–1300 Cimetidine 638–639 Cinarizine 1642 Ciprofloxacin 1403–1404 Cisapride 637, 782, 1656 Cisplatin 1089, 1090, 1134 adverse effects 1200, 1200, 1208 Citalopram 520 Citrobacter spp 1113–1114, 1114 Cladophialophora bantiana 1383 Cladosporiosis 1385 Cladribine 1134, 1204, 1207–1208 Classic dermatomyositis (CDM) see Idiopathic inflammatory myopathy (IIM) Claudication, spinal stenosis 542–543 Clebopride 1656 Cleveland Family Study 253 Clindamycin 1324–1325, 1357, 1517 Clinical Immunization Safety Assessment (CISA) Network 1549 Clinical target volume (CTV) 1181–1182 Clinically amyopathic dermatomyositis (CADM) see Idiopathic inflammatory myopathy (IIM) Clodronate 536 Clofarabine 1208

Clofazimine 1575–1576 Clofibrate 1638 Clonazepam 846, 1640 Clonidine 1636, 1640 Clopidogrel (Plavix®) 101, 581, 1129, 1639 Clostridium spp 80–81 Clostridium botulinum 988, 996, 1640–1641 Clostridium difficile 1113–1114, 1114 Clostridium tetani 988, 1506, 1507 CLOTS (Clots in Legs Or sTockings after Stroke) trials 294, 296 Clotting function 1595–1596 c-Myc gene 1031 Clozapine 1642 Cluster headache 259 Coagulation factors VII/IX inhibition 1052 scheme 1062 tests 1050 Coagulopathies 1126–1128 ‘Coasting’ phenomenon 923 Cobalamin (Cbl) see Vitamin B12 deficiency Cobb syndrome 1562, 1572–1573, 1572 Cobras (Naja) 989, 991 Coccidioides spp 1386, 1397–1398, 1535 Coccidioides immitis 1383, 1384, 1385, 1397–1398 Coccidioidomycosis 1385, 1397–1398 Cochrane meta-analysis 294, 296 Cochrane reviews 507, 670–671, 1575–1576, 1759–1760 Cochrane Stroke Group Trials 118 CODACS (Consciousness Disorders After Cardiac Surgery) trial 144 Code of Federal Regulations (Regulations) 1549 Coenurosis 1428 diagnosis 1428 epidemiology 1428 pathogen 1428 signs/symptoms 1428 treatment 1428 Coenurus spp 1422 Coenzyme Q10 31 Cogan’s syndrome (CS) 483, 1717 clinical features 483 Cognition drugs 1641–1642, 1642 Cognitive behavioral therapy (CBT) 518 Cognitive dysfunction arrhythmia 130–131 atrial fibrillation 117–118 brain metastases 1145 congenital heart disease (CHD) 52–53, 53 dialysis patients 387, 397, 399–400 end of life (EOL) phase 1220 folic acid deficiency 934 neuro-Behc¸et syndrome (NBS) 1710 neuropsychiatric systemic lupus erythematosus (NPSLE) 464–465 neurosarcoidosis 311 obstructive sleep apnea syndrome (OSAS) 255

INDEX Cognitive dysfunction (Continued ) postoperative (POCD) 1630 treatment 1221 see also Neurocognition COL3A1 gene 565–567, 1567–1568 Colchicine 1650, 1718–1719 Cold pressure test 391 Collagenopathy 1567 Colubridae 987 Coma barbiturate 1628 hypothyroidism 705–706 Common pathway 1045–1046 Community Aging and Dementia Project 931 Community-acquired bacterial meningitis 1361, 1363–1365 Compartment syndrome 1053 Complement therapy 1120 Complex sleep apnea syndrome (cSAS) 264 Comprehensive psychopathologic rating scale (CPRS) 868–869 Compression neuropathy 775 Computed tomography (CT) brain metastases 1148, 1149 cardiac arrest 34 complications 1745–1746 hemolytic uremic syndrome (HUS) 1116, 1117 myelography 544–545 reversible cerebral vasoconstriction syndrome (RCVS) 1731–1732, 1732, 1733 single-photon emission (SPECT) 515, 1715 Concussive convulsions 179 Conduction 947 Cone snail venom 988 Confusion/delirium end of life (EOL) phase 1220 treatment 1221 Congenital adrenal hyperplasia 756 Congenital bleeding disorders 1045–1046 fibrinolytic pathway deficiencies 1056 homeostasis, pathophysiology 1045–1046, 1046 management, treatment centers 1058 overview 1047 rare 1054–1056 diagnosis 1054–1055 genetic testing 1055 management 1055–1056, 1055 prophylaxis 1056 symptoms 1054 see also Hemophilia; Platelet function disorders; von Willebrand disease (VWD) Congenital central hypoventilation syndrome (CCHS) (Ondine’s curse) 245–246, 278 alveolar (CCAHS) 265 Congenital heart disease (CHD) 49–60 brain anomalies 53–54

Congenital heart disease (CHD) (Continued ) cerebral aneurysm/aortic coarctation 54, 54 cerebrovascular complications 51–52, 51 clinical features 49, 50 cognitive disturbances 52–53, 53 genetics 53 historical perspective 49 incidence 49–50 infective embolism 52 laboratory investigations 53, 56 management 56 complications 54–56 signs/symptoms 51 surgical procedures 49, 50 Congenital hypothyroidism (CH) 703–705 clinical/radiologic features 704 etiology 704 pathogenesis 704 treatment/prognosis 704–705 Congenital phosphorus disorders 874 Congenital rubella syndrome (CRS) 1349, 1350 clinical manifestations 1350 Congestive heart failure (CHF) 258 Conivaptan 810, 812 Conjugate vaccines 1550 Connective tissue disorders (CTDs) 1567–1569 idiopathic inflammatory myopathy (IIM) 496, 499–500, 501 inherited 565–576 neurologic manifestations 565–574, 566 see also Scleroderma; Sj€ ogren’s syndrome (SS); Systemic lupus erythematosus (SLE) Conscious state, abnormal 1259–1261, 1260 Consciousness, impaired 1287–1288 Continuous chest compressions (CCC) 26 Continuous positive airway pressure (CPAP) 254, 260–261, 262 Continuous renal replacement therapy (CRRT) 400 Contrast media 1746 Contrast-induced nephropathy (CIN) 1745 Conus medullaris syndrome 1763 Convection 947 Coomb’s test 1011 Cooperative Cardiovascular Project 104–105 Cooperative Study of Sickle Cell Disease (CSSCD) 1016 Copper disorders 851–858 acquired copper deficiency 853, 857 acquired copper toxicosis 857–858 ATP7A-related transport disorders 853–854, 853 ATP7B-related transport disorders 854–857 deficiency myelopathy (CDM), bariatric surgery 590 malabsorption 626–627 metabolism 851–852, 852

I11 Coral snakes (Micrurus) 992 Cordylobia anthropophaga 1436 Cornea verticillata 1564 Coronary artery bypass grafting (CABG) 194, 195, 197–199, 199 stroke and 93, 198 Coronary artery dilators 1636, 1637 Coronary artery disease (CAD) 111, 906 Coronary artery stents (CAS) 99, 99 Coronary heart disease (CHD) 93 Cortex, disease 245–247, 247 Cortical blindness 35 transient 44, 44, 197 Cortical cerebellar degeneration 897–898 Cortical control, breathing 241, 242 Corticospinal tract disorders 715 Corticosteroids (CS) adverse effects 1269–1270, 1278–1279, 1645–1647 bacterial meningitis 1370 brain tumor 337, 1150, 1153, 1220–1221 breast feeding 1608 cerebral vasculitis 481, 483, 485–486, 487 connective tissue disorders (CTDs) 469–470, 471 gnathostomiasis 1539 helminthic infection 1429, 1433, 1434, 1435 hemolytic uremic syndrome (HUS) 1120 Henoch–Sch€ onlein purpura (HSP) 1108 idiopathic inflammatory myopathy (IIM) 506–507 intracranial pressure (ICP) 1759 leprosy 1513–1514 lung cancer 341–342, 354 Lyme disease 1480 mononeuropathy 1333–1334 multiple myeloma (MM) 1090–1091 neurocysticercosis 1456 neuromuscular conditions 1275, 1613 paraneoplastic neurologic syndromes (PNS) 1173 sarcoidosis 321, 322, 323, 327 spinal stenosis 546 tuberculosis (TB) 1338, 1490–1492 Corticotrophin-releasing hormone (CRH) 749, 750 ACTH levels 692 adrenal insufficiency (AI) 759 Cortrosyn test 759 Corynebacterium diphtheriae 1355, 1356 Co-trimoxazole (Bactrim®) see Trimethoprimsulfamethoxazole (TMP-SMX, TMP-SMZ) Cough reflex, absent 1240 Cough syncope 175, 177 Coumarins 1638–1639 COX-2 inhibitors 580 Coxiella spp 1403 Coxiella burnetii 79, 80–81, 1403–1404 Cranial nerve disorders diphtheria 1356 hypothyroidism 708–709

I12 Cranial nerve disorders (Continued ) palsies 322 radiotherapy (RT) 1192 Cranial neuritis 1474 Cranial neuropathy 307–308 diabetic mellitus (DM) 775 inflammatory bowel diseases (IBD) 597 Paget’s disease of bone (PDB) 530 Cranial vault lymphomas, clinical presentation 1036–1037 Craniocervical junction anomalies 554–556, 555 dislocation 435 Craniopharyngiomas, radiotherapy (RT) 1188–1189 Craniotomy 1760 Craniovertebral dislocation 433–448 anatomic concepts 436–437 causes 436 clinical features 437–438 progressive craniovertebral subluxation 437–438 traumatic craniovertebral subluxation 438, 438, 439 congenital malformations 441, 445 historical perspective 435 neuroimaging 438–440, 440 neurologic complications 436, 438, 440–446 craniovertebral junction 441, 445, 445, 446, 446 Cranium, fractures 1752 Creatine kinase (CK), raised 714 Creatine monohydrate 506 Cremophor 1202 CREST (calcinosis, Raynaud’s phenomenon, esophageal dysmotility, sclerodactyly, teleangiectasia) syndrome 468 Creutzfeldt–Jakob disease (CJD) 811 Critical flicker frequency (CFF) 665 Critical illness myopathy (CIM) 751–752, 754, 1674–1675 lung transplantation 1240 outcome/prognosis 1680 pathophysiology 1675–1676 risk factors 1675 Critical illness myopathy-polyneuropathy 1278–1279 Critical illness neuromyopathy (CINM) 1675 Critical illness polyneuropathy (CIP) 1674 outcome/prognosis 1680 pathophysiology 1675–1676 risk factors 1675 Crohn’s disease (CD) see Inflammatory bowel diseases (IBD) Crotalinae 992–993 Crotalus 992–993 Crotalus atros 993 Crotalus durissus durissus 993 Crotalus durissus terrificus 990, 992–993 Crotalus horridu 993 Crotalus scutulatus 993

INDEX Crow–Fukase syndrome see POEMS syndrome Cruciate paralysis 1763 Cryoglobulinemia 1083, 1084, 1087, 1096 peripheral nervous system (PNS) 1096 Cryoprecipitate 1127 Cryothermy 157 Cryptococcal meningitis, HIV-associated 1248–1249, 1328–1330, 1391–1394, 1393, 1394 clinical features 1328–1329 diagnosis 1329 epidemiology 1328 imaging 1329 intracranial pressure, raised 1330 pathogenesis 1328 treatment 1329–1330, 1329 antiretroviral therapy (HAART) 1330 prophylaxis 1330 Cryptococcosis 1385 Cryptococcus spp 1252, 1328–1329, 1387, 1535 Cryptococcus neoformans 1328, 1329–1330, 1383, 1384, 1386, 1389–1390, 1391 investigation 1386 transplantation 1232, 1241, 1262 Cryptococcus neoformans var gattii 1328, 1391 Cryptococcus neoformans var neoformans 1328, 1385, 1391 Cryptogenetic drop attacks, women 182–183, 183 Cryptosporidium spp 1410 Crystalline aqueous penicillin 1468, 1470 Cuba Neuropathy Field Investigation Team 1543 Cuban epidemic myeloneuropathy 1542–1543 clinical manifestations 1543–1544 epidemiology 1543 etiology 1544 treatment/prevention 1544 Cucurbita pepo 1445 Culex spp 1429 Cunninghamella 1383, 1396 CURE (Clopidogrel in Unstable Angina to prevent Recurrent Events) trial 101 percutaneous coronary intervention subset (PCI-CURE) 103 Cushing syndrome (CS) 750–754 clinical manifestations 750–752, 752, 753, 754 diagnosis 752–754 etiology 750, 751 historical perspective 750 treatment 754 Cushing’s disease ACTH-dependent 692–693 management 698 Cutaneomeningospinal angiomatosis (CMA) see Cobb syndrome Cutaneous manifestations angioma 1569–1573 Carney complex (CC) 1566, 1566

Cutaneous manifestations (Continued ) neurofibromas 1581–1582 see also Neurocutaneous disorders CV2/CRMP5-Abs (antibodies) limbic encephalitis (LE) 1166, 1169 subacute cerebellar degeneration (SCD) 1164 Cyanide 1544–1545 Cyanocobalamin deficiency 591 Cyclooxygenase (COX) 577–578, 579 inhibitors 580, 1129 Cyclophosphamide (CTX) 459, 471, 507, 1108, 1173, 1718–1719 adverse effects 1210 cerebral vasculitis 479, 483, 485–486, 487 neurosarcoidosis 325, 326, 327 Cyclops spp 1427–1428, 1429 Cycloserine 1491, 1492 CYP27A1 gene 1586 Cyproheptadine 951, 1654 Cystic echinococcosis see Hydatid disease Cystic fibrosis 1280 Cysticerci characteristics 1447 immune response 1448 involution stages 1447, 1447 tissue reaction 1447–1448, 1448 Cysticercosis 1445, 1777 Cysticercus cellulosae 1423–1424 Cysticidal drugs 1455 Cytarabine see Cytosine arabinoside (ARa-C) Cytomegalovirus (CMV) heart transplantation 1232, 1233 hemolytic uremic syndrome (HUS) 1114, 1114 liver transplantation 1263 mononeuropathy 1333–1334 progressive polyradiculopathy 1334 renal transplantation 1249 serology mismatch 1238 treatment 1380 Cytomegalovirus (CMV) encephalitis 1377, 1379 diagnosis 1380 focal 1232 treatment 1381 Cytomegalovirus (CMV) encephalitis, HIV-associated 1330–1331 clinical features 1330–1331 diagnosis 1331 prevention 1331 treatment 1331 Cytosine arabinoside (cytarabine) (ARa-C) 1134, 1135, 1263 adverse effects 1200, 1203, 1206–1207 Cytotoxic drugs 1694

D

Dabigatran (Pradaxa®) 121–122, 139, 295, 1131 Dabigatran etexilate, adverse effects 1639 Daboia palaestinae 993 Daboia russelii 988, 990, 992–993

INDEX Daboia siamensis 993 Dacarbazine 1209 Dantrolene 951 Dapsone 1575–1576 Dasatinib (Sprycel®) 1133 Data and Safety Monitoring Board (DSMB) 122, 1021–1022 Daunorubicin 1211 Death adder (Acanthophis) 992 Decompression illness (DCI) 959–970 arterial gas embolism (AGE) 959–960, 967 case examples 966–967 clinical manifestations 960–962, 961 decompression sickness (DCS) 959, 966–967 diagnosis 960–962, 963, 964 epidemiology 960, 960, 961 long-term consequences 965–966 prevention 962–963 treatment 963–965 adjunctive therapy 965 first aid 963–965 recompression 965 Decompression sickness (DCS) see Decompression illness (DCI) Decompressive craniectomy 1759–1760 DECRA study (Australia) 1759–1760 Deep hypothermic circulatory arrest (DHCA) 55 Deep venous thrombosis (DVT) 1767 brain metastases 1153 spinal cord injury (SCI) 1766 see also Venous thromboembolism (VTE) Deer tick virus, encephalitis 1378 Deferasirox 1020–1022 Deferoxamine 1020–1021 Defibrillators, implantable 141 complications 157 Degenerative ataxias 9 Degos disease (malignant atrophic papulosis/Kohlmeier-Degos disease) 1562, 1567 Deinagkistrodon 992–993 Delirium see Confusion/delirium Deltaretrovirus 1527 Dementia arrhythmia 131 cardiology 9 dialysis 397, 399–400 folic acid deficiency 931–932 HIV 1322 vascular (VaD) 9, 931–932, 1778 see also Alzheimer’s disease (AD) Demyelinating disorder 466, 600 and axonal neuropathy 1273 Dendroaspis spp 990, 992 Dendroaspis angusticeps 990, 992 Dendroaspis jamesoni 992 Dendroaspis polylepis 992 Dendroaspis viridis 992 Dendrobatidae 996 Deoxyguanosine kinase (dGK) deficiency 828, 832–833

Department of Health and Human Services (USA) 1338 Depot Ara-C (DepoCyt®) 1203 Depressed skull fracture 1760 repair 1760 Depression 254 folic acid deficiency 931–932, 933–934, 933 hypothyroidism 706–707 obstructive sleep apnea syndrome (OSAS) 255–256 vitamin D 881–882 Dermatobia hominis 1436 Dermatomyositis (DM) see Idiopathic inflammatory myopathy (IIM) Desferrioxamine 397 Desflurane 1626, 1627, 1630–1631 Desirudin 1639 Desmopressin acetate (DDAVP) 810, 812, 1051–1052, 1057, 1132 Developing world 1773–1782 healthcare systems 1773–1774 primary care 1774 secondary care 1774 tertiary care 1774 human resources/training/education 1774–1775 neurologic diseases, common 1775–1780 see also Tropical myeloneuropathy; Tropical myelopathy Developmental defects 756 Developmental delay 557–558 Device-associated meningitis 1369 Devic’s disease 1540 Dexamethasone 341, 506, 1379–1380, 1492–1493, 1613 bacterial meningitis 1369–1371 brain tumors 337, 1153, 1220–1221 tropical myelopathy 1545, 1537, 1539 Dextromethorphan 1655 Dextrose 819 DGUOK gene 832–833 Diabetes Complications and Control Trial (DCCT) 776–777, 778–779 Diabetes insipidus (DI) 370, 370 central 690 nephrogenic 372 Diabetes mellitus (DM) drugs 814–821 neuropathy see Diabetic neuropathy pancreas transplantation 1280 Diabetes mellitus (DM) type 1 (T1DM) 773, 801 drugs 814, 817, 820–821 Diabetes mellitus (DM) type 2 (T2DM) 773, 774 drugs 779, 814, 815, 816, 817, 817, 820–821 Diabetic autonomic neuropathy (DAN) 773, 774–775, 781–782 Diabetic cardiac autonomic neuropathy (CAN) 10 Diabetic ketoacidosis (DKA) 377 Diabetic neuropathy 773–785 classification/clinical findings 774–776

I13 Diabetic neuropathy (Continued ) asymmetric lower limb neuropathy 776 autonomic neuropathy (DAN) 773, 774–775 cranial neuropathy 775 focal/multifocal neuropathies 775 limb mononeuropathy 775 selective small fiber polyneuropathy 774 sensory/sensory motor polyneuropathy 774 trunk mononeuropathy 775–776 historical perspective/epidemiology 773–774 laboratory investigations 776 pathophysiology 777–778, 777 AGE pathway 777 hexosamine pathway 777–778 neurotrophic factors/nerve repair 778 oxidative stress 777 polyol pathway 777 protein kinase C (PKC) pathway 778 risk factors 776–777 treatment 778–782 autonomic neuropathy (DAN) 781–782 disease state modifiers 778–780 neuropathic pain 781 pain-controlling agents 780–781 Diabetic peripheral neuropathy (DPN) 776–777 Diagnostic biopsy 1148 Diagnostic and Statistical Manual of Mental Disorders (DSM-IV) 144 Dialysis dementia 397, 399–400 Dialysis disequilibrium syndrome (DDS) 386, 396–397 clinical presentation 386 pathogenesis 386 therapy 386, 386 Dialysis encephalopathy syndrome 387–388 clinical symptoms 388 diagnosis 387–388 treatment 388 Dialysis patients 395–404 central nervous system (CNS) 395–400, 396 cognitive impairment 387, 397, 399–400 peripheral nervous system (PNS) complications 396, 400–401 3,4-Diaminopyridine 354 Diazepam 31, 1508–1509 Diclofenac 580, 1648–1649 Dicumarol 1638–1639 Diethylcarbamacin 1429 Diffuse axonal injury (DAI) 1753, 1755 Diffuse head injury 1755 Diffuse leukoencephalopathy 1150 Diffuse noxious inhibitory control (DNIC) 516 Diffuse peripheral neuropathy 707–708 DiGeorge syndrome 53

I14 Digestive tract drugs 1655–1656, 1656 Digit Symbol Test (DST) 663–664, 664 Digitalis purpurea 133, 133 Digoxin 132, 133 Dihydroergotamine 1651 Dihydropyrimidinase (DHP) deficiency (dihydropyrimidinuria) 834, 834 Dihydropyrimidine dehydrogenase (DPD) deficiency 834 Diltiazem 132, 133–134, 1636 Dipeptidyl peptidase (DPP-4) inhibitors 819–820 Diphenhydramine 636–637, 1651–1654 Diphosphonates 1655 Diphtheria 1355–1357 clinical features 1356–1357 epidemiology 1355–1356 immunization 1358 diphtheria-tetanus-pertussis (DTP) vaccine 1358, 1550, 1555–1556, 1651 laboratory features 1357 outcome 1357 pathogenesis/pathology 1356 treatment 1357 Diphyllobothrium dendriticum 1428–1429 Diphyllobothrium latum (fish tapeworm) 918, 1422, 1428–1429, 1541 clinical manifestations 1429 diagnosis 1429 epidemiology 1428–1429 pathogen 1429 therapy 1429 Dipyridamole 1129–1130, 1639 Direct thrombin inhibitors (DTIs) 1131, 1131, 1132, 1639 Disc herniation, acute myelopathy 1524–1525 Discovertebral destruction 456 Disease-modifying therapies (DMTs) 1607–1608 Disopyramide 132, 134, 187 Distal motor neuropathy (DMN), ATP7Arelated 853, 854 Distal symmetric polyneuropathy (DSP) 1321, 1332–1333, 1335 clinical features 1332 diagnosis 1332 treatment 1332–1333 Diuretics 12, 165, 866 Dive tables 962 Divers Alert Network (DAN) 960, 961 Diving see Decompression illness (DCI) DMPK gene 796 DNA methyltransferase inhibitors 1133–1134 Docetaxel 1202, 1210 Dofetilide 132, 134 Dolasetron 636, 637 Dolichovespula 995 Domperidone 185, 636, 1642, 1656 Donepezil 1642 Dopamine 517, 811, 1637 Dopamine agonists (DA) 519, 522, 697, 811, 1641

INDEX Dopamine dysregulation syndrome 1641 Dopamine receptor-blocking drugs (DRBDs) 1642–1643, 1643 Dorsolateral myelopathy 1544 ‘Doughnut appearance’ 1679–1680 Down syndrome craniovertebral dislocation 435, 443 genetics 52, 53 Doxorubicin 1203, 1211 Doxycycline 625, 628, 1379–1380, 1403–1404, 1429, 1517 Lyme disease 1479–1480, 1480, 1533 neurosyphilis 1337, 1468–1469 D-penicillamine 856–857 Dreaming, excessive 254 Dronabinol 638 Dronedarone 132, 134 ‘Drop attacks’ 181–184 cryptogenetic, women 182–183, 183 Droperidol 639, 1642, 1656 Drug-induced conditions central sleep apnea (CSA) 264 encephalopathy 1299 hyperthermia see under Hyperthermia hypomagnesemia 871 inflammatory bowel diseases (IBD) 601 movement disorders (DIMD) 635, 638 multiple myeloma (MM) 1089–1090 neurotoxicity 1245–1247 posterior reversible encephalopathy syndrome (PRES) 1692, 1695 renal failure (RF) 388 see also Iatrogenic neurology Dual antiplatelet therapy 581 Duchenne muscular dystrophy (DMD) 10–12, 11, 125 respiratory failure (RF) 277 Duke criteria infective endocarditis 81, 82 stroke 67 Duloxetine 520, 780, 781 Dural metastases 345 Dwarfism see Achondroplasia Dystonic spasms 635 Dystrophinopathies 10–12, 11, 124–125 Dystrophy, hereditary 1612

E Eastern Association for the Surgery of Trauma (EAST) 298, 1763 Eastern equine encephalitis virus 1378 diagnosis 1380 Echinocandins 1389, 1395, 1649 Echinococcosis 1425–1427 cystic see Hydatid disease Echinococcus spp 1287–1288, 1422 Echinococcus granulosus 1425–1427 Echinococcus multilocularis 1425, 1426–1427 Echis 992–993 Echocardiography 80, 80, 185, 217 transesophageal (TEE) 7–8 Eclampsia defined 1597 long-term effects 1597–1598

Eclampsia (Continued ) posterior reversible encephalopathy syndrome (PRES) 1692, 1693–1694, 1696 treatment 1604 Eculizumab 507, 1120 Edema, traumatic brain injury (TBI) 1753 Education developing world 1774–1775 fibromyalgia 518 Eflornithine 1412 Egyptian cobra (Naja haje) 987, 989 Ehlers–Danlos syndrome (EDS) 570–571 description 570 neurologic complications 570–571 type IV 1562, 1567–1568, 1567 type IX (occipital horn syndrome (OHS)) 853, 853 vascular see Vascular Ehlers–Danlos syndrome (vEDS) Ehrlichia spp 1403 Elapidae 987, 991–992 Elastinopathy 1567 Electrical cardioversion 139–140 Electrical injuries see Thermal injuries Electrocardiography (ECG) monitoring 185 thermal injuries 982, 983 Electrocution, defined 981 Electroencephalography (EEG) brain metastases 1149 cardiac arrest 32–33 Hashimoto’s encephalopathy (HE) 725 hemolytic uremic syndrome (HUS) 1116–1117, 1118 Electrolyte disturbances 366 see also specific conditions Electromyography (EMG) 502, 1679 Electrophysiologic studies 185 procedures 142 Embolism anticoagulation 66 arterial gas (AGE) see Decompression illness (DCI) cardiac tumors 212–213 cerebral see Cerebral embolism congenital heart disease (CHD) 52 defined 93 infective endocarditis 67–68, 85, 85 saddle 296 stroke 95 see also Thromboembolism, catheter ablation; Venous thromboembolism (VTE) Emery–Dreifuss muscular dystrophy (EDMD) 11, 12, 125 Enalapril 1636 Enalaprilat 165, 165 Encainide 132, 134 Encephalitic (furious) rabies 1501–1502 Encephalitis 1377–1381 clinical presentation 1378 diagnosis 1378–1379, 1380 etiology 1377–1378 granulomatous 1411

INDEX Encephalitis (Continued ) HIV 1322 Japanese (JE) 1554 Toxoplasma (TE) 1323, 1324 treatment 1379–1381, 1380 Encephaloduroarteriosynangiosis (EDAS) 1022 Encephalomyelitis 1475–1476 Encephalopathy acute 79 coronary artery bypass grafting (CABG) 198 dialysis see Dialysis encephalopathy syndrome gluten 612, 612 Hashimoto’s see Hashimoto’s encephalopathy (HE) heart transplantation 1232 hematopoietic stem cell transplantation (HSCT) 1297–1299 hepatic see Hepatic encephalopathy (HE) hypertensive 1104 hypothyroidism 705–706 infective endocarditis 68–69 intestinal transplantation (ITx) 1271–1272, 1271, 1274 lung transplantation 1241 multifocal 348 neurosarcoidosis 310–311, 311 portosystemic see Portosystemic encephalopathy small bowel transplantation 1287–1288 uremic see Uremic encephalopathy Wernicke’s see Wernicke’s encephalopathy (WE) see also Hypertension/hypertensive encephalopathy; Posterior reversible encephalopathy (leukoencephalopathy) syndrome (PRES/PRLS) End of life (EOL) phase 1219–1227 bereavement care 1223 decision-making 1221–1222 advance care planning (ACP) 1222–1223 euthanasia/physician-assisted suicide (PAS) 1222 nontreatment 1222 palliative sedation 1222 neurologic signs/symptoms 1219–1220 cognitive dysfunction/delirium 1220 impaired motor function/immobility 1220 raised intracranial pressure (ICP) 1219 seizures 1219–1220 supportive treatment 1220–1221 cognitive dysfunction/delirium 1221 impaired motor function/immobility 1221 raised intracranial pressure (ICP) 1220–1221 seizures 1221

Endemic ataxic polyneuropathy (tropical ataxic myeloneuropathy (TAN)) 1542–1543 Endocarditis infective see Infective endocarditis noninfective 69–70 Endocrine disorders 310 Carney complex (CC) 1566, 1566 fibromyalgia 517 obstructive sleep apnea syndrome (OSAS) 257–258 primary brain tumors 726 Endocrine therapy 809–824 adverse effects 1204 brain metastases 1152–1153 diabetes mellitus (DM) 814–821 hormone-related drugs 1654, 1655 hypothalamic/pituitary hormones/ analogs 809–812, 810 thyroid/parathyroid agents 812–814 End-stage renal disease (ESRD) 384, 385, 395, 1245 dementia 399–400 periodic limb movement disorder (PLMD) 398–399 polyneuropathy 400–401 sleep disorders 398 stroke 400 Engel, George L 19 Enhydrina schistose 992 Enoxaparin 1132 Entamoeba histolytica 1407, 1410–1411, 1410, 1410 Entecavir (Baraclude®) 676, 680 Enterococcus spp 52, 68, 1532 Enteroviruses 1378, 1380 Entrapment neuropathy 450–451, 450, 707, 775 Ependymomas, radiotherapy (RT) 1185 Ephedrine 187 Epicardial radiofrequency vein isolation 143–144 Epidemiology of Diabetes Interventions and Complications (EDIC) 778 Epidermal growth factor receptor (EGFR) inhibitors 1152–1153 Epidermoid spinal tumors 1747 Epidural disease, multiple myeloma (MM) 1090–1091, 1090, 1091 Epidural hematomas (EDH) 530, 1752, 1760, 1761 Epilepsy cardiology 8, 8 developing world 1775–1776 folic acid deficiency 932–933 gluten-related diseases (GRD) 613, 613 multiple organ transplantation 1310 neurocutaneous disorders 1584–1590 pregnancy 1606–1607 sudden unexplained death (SUDEP) 8, 22–23 vitamin D 882

I15 Epileptic transient loss of consciousness (TLOC) 175, 177–178, 179, 184 clinical features 177–178, 178 falls 182 pathophysiology 178 Eplerenone 764–765 E-Aminocaproic acid (EACA) 1128 Epstein–Barr virus (EBV) Burkitt lymphoma (BL) 1030–1031 encephalitis 1378, 1379 diagnosis 1380 treatment 1380 heart transplantation 1232, 1233 hemolytic uremic syndrome (HUS) 1114, 1114 Hodgkin lymphoma (HL) 1034 intestinal transplantation (ITx) 1270 serology mismatch 1238 Eptifibatide 1639 Epworth Sleepiness Scale 260 Ergocalciferol 745 Ergotamine 1650, 1654 Ergotism 1650 Eribaxaban 139 Eribulin 1203 Erythromycin 1337, 1357, 1468–1469 Erythromycin lactobionate 782 Erythropoiesis-stimulating agents (ESAs) 1009 Erythropoietin 782, 1006, 1205 Escherichia coli (E. coli) 1369, 1532, 1540–1541 hemolytic uremic syndrome (HUS) 1113–1114, 1117–1118 Eslicarbazepine, elimination 418, 422–423, 425, 427 Esmolol 132, 134, 165, 165 Esomeprazole 638–639 Essential thrombocythemia (ET) 1075–1077 clinical characteristics 1075–1076 defined 1073 diagnostic criteria 1076 JAK2 gene 1073–1074 neurologic manifestations 1076–1077, 1077 treatment 1077, 1132 Estramustine 1209 Etanercept 507, 718–719, 1719 Ethambutol 1490, 1491 Ethanol 376–377 Ethionamide 1490, 1491, 1492 Ethosuximide 1640, 1640 elimination 418, 425, 427, 428 Ethylene glycol toxicity 375–376 Etomidate 698, 754, 1625, 1625, 1631 Etoposide 1203, 1210–1211 EURAP Epilepsy Pregnancy Registry 1606–1607 European Association for the Study of Diabetes 817 European Diabetes (EURODIAB) Prospective Complications Study 776–777

I16 European Federation of Neurological Societies (EFNS) Task Force 1173 European Group on Graves’ Orbitopathy 718–719 European Headache Federation 1776 European Heart Survey 119, 122–123 European League Against Rheumatism (EULAR) 450, 466–467 fibromyalgia 518, 521–522 Henoch–Sch€onlein purpura (HSP) 1102, 1102 European Medicines Agency (EMA) 809, 813 European Neuromuscular Centre (ENMC) 496 European Society of Cardiology 80 infective endocarditis 83–84, 84, 85, 86–87 Task Force for the Diagnosis and Management of Syncope 174 Task Force on Prevention, Diagnosis and Treatment of Infective Endocarditis 75 Euthanasia 1222 Evaporation 947 Everolimus 1231, 1289–1290 Excessive daytime sleepiness (EDS) 254, 398 Exenatide 817, 820 Exercise 185, 518 Expanded Disability Status Scale (EDSS) 1717 Expert Consensus Panel, cardiomyopathy definition 111 Extended criteria donors (ECD) 1237 External cardioversion 139–140 Extracorporeal membrane oxygenation (ECMO) 55, 201–202, 1238 Extrapyramidal disorders 183–184, 351 Eye involvement 1568, 1704 ‘Eye of the tiger’ image 860 Ezetimibe 1638 Ezogabine 418, 425, 427, 429

F Fabry disease (FD) 123, 1561–1564, 1562 clinical features 1561–1564 cardiac involvement 1564 cutaneous 1561 neurologic 1561–1564 ophthalmologic 1564 renal manifestations 1564 diagnosis/counseling 1564 treatment 1564 ‘Face of the giant panda’ sign 856, 856 Facial baroparesis 964 Facial cutaneous angioma 1570 Facial nerve (cranial nerve VII) 307–308 Facial palsies 964 Facial paralysis 1533 ‘Facies lactrodectismica’ 994–995 Factor Xa antagonists 139 Factor Xa inhibitors 295, 1130–1131, 1639 Fainting larks 175

INDEX Falls accidental 184 epileptic 182 gait ataxia/extrapyramidal disorders 183–184 muscle weakness 184 psychogenic 184 Famciclovir 1380–1381 Familial expansive osteolysis, clinical characteristics 534 Familial glucocorticoid deficiency (PGD) 756 Familial hypobetalipoproteinemia (FHBL) 623, 624 Familial hypocholesterolemia 623–624 Familial oncogene derangements 799 Familial Paget’s disease, clinical characteristics 534 Familial paraganglioma 765 Famotidine 638–639 Fanconi’s anemia (FA) 1010 Fasciculins 990 Fascioscapulohumeral muscular dystrophy (FSHD) 1612 FAST trial 1132 Fat metabolism 591 Fatigue, sarcoidosis 328 Favaloro, Rene´ 41 FBN1 gene 569 Felbamate 846 elimination 418, 425, 427, 428 Femoral artery catheterization 7 Femoral nerve, cardiac surgery 197 Fenoldopam 165, 165 Fentanyl 1626, 1758 Fever 951–953 causes 952 clinical features 951–952 laboratory findings 952 management 953 Fibrinolysis 1595–1596 Fibrinolytic pathway deficiencies 1047, 1056 Fibro fog 513–514, 521 Fibroelastomas 218 papillary 216 Fibromyalgia 513–528 clinical presentation 513–514 diagnostic criteria 514 epidemiology 514 genetics 516 historical perspective 513 laboratory investigations 515 management 517–523 complementary 523 pharmacologic 518–523, 519 nonpharmacologic 518 natural course 514–515 neuroimaging 515–516 functional magnetic resonance imaging (fMRI) 101 positron emission tomography (PET) 515–516 proton magnetic resonance spectroscopy (H-MRS) 516

Fibromyalgia (Continued ) single-photon emission computed tomography (SPECT) 515 obstructive sleep apnea syndrome (OSAS) 259–260 pathophysiology 516–517 autonomic dysautonomia 517 central nervous system (CNS) 516–517 endocrine influences 517 peripheral influences 516 Fibromyalgia Impact Questionnaire (FIQ) 514, 518 ‘Final Push’ strategy (WHO on leprosy) 1512–1513 Fingolimod 507, 1607–1608 Flatworms (platyhelminths) 1414, 1419–1429 Flecainide 132, 134, 1637 Flexion contractures syndrome 758 Flucloxacillin 1369 Fluconazole 1329–1330, 1329 Fluctuating vigilance 177–178, 184 Flucytosine 1248–1249, 1329–1330, 1329, 1389 Fludarabine 1134, 1201, 1207 Fludrocortisone 185, 187, 781 Flukes 1419–1422 Flunarizine 1642 Fluorinate pyrimidines 1652 Fluoroquinolones 1490, 1492, 1646 Fluorosis 1547–1546 5-Fluorouracil (5-FU) 834, 1200, 1201, 1204, 1207 Fluoxetine 520 Flupirtine 1649–1650 Fluprednisolone 1647 Flutter VRPI® 283 FMR1 gene 793, 794–795 Focal deficits amyotrophy 1681 brain metastases 1145 mass lesions, fungal infection 1385 neurocysticercosis (NCC) 1449 neuropathies, diabetes mellitus (DM) 775 radionecrosis 1150 reversible cerebral vasoconstriction syndrome (RCVS) 1728 Folate see Folic acid deficiency Folic acid (folate) deficiency 927–943, 1125–1126 bariatric surgery 591 clinical dissociation 935–936 disorders 937 epilepsy 932–933 folate metabolism 928–929, 928 inborn errors 935, 935 historical perspective 927–928 homocysteine/depression/dementia/ aging 931–932 megaloblastic anemia 927, 929–930, 930, 1007 metabolic mechanisms 928, 936–938, 937

INDEX Folic acid (folate) deficiency (Continued ) neural tube defects (NTDs) 932, 934–935 neuropsychiatric disorders 930–931 short bowel syndrome 1286 treatment 933–934, 933 cognitive function 934 depression 933–934, 933 Follicle-stimulating hormone (FSH) 686 acromegaly 691–692 excess 693–694 hyperprolactinemia 690–691 secondary hypogonadism 689–690 Fomepizole 376–377 Fondaparinux 1130–1131 Food and Drug Administration (FDA) acute stroke 156 dabigatran 121 deferasirox 1020–1021 endocrine therapy 809 implantable devices 1746 metoclopramide 635 multiple sclerosis (MS) 1607–1608 mycophenolate mofetil 326–327 nephrogenic systemic fibrosis (NSF) 97–99 pregabalin 1332–1333 pregnancy 1603 ribavirin 679 serotonin syndrome (SS) 1651 stroke 1639 sunitinib 1213 thionamides 813 vaccines 1549, 1552, 1554 Foreign accent syndrome 1260 Forssmann, Werner 41 Fosaprepitant 637 Foscarnet 1331, 1380, 1381 Fosphenytoin, malaria 1517 Fotemustine 1208–1209 Fractionated heparin 1719–1720 Fractionated radiation therapy (RT) 341–342 Fractures, cranium 1752 Fragile X syndrome 793–795, 794 clinical findings 794 historical perspective 793 laboratory investigations 794–795 management 795 natural history 794 pathology 794 Fragile X-associated tremor/ataxia syndrome (FXTAS) 793 Framingham Heart Study 118–119, 161, 904, 931 Framingham risk score 111 Frascati classification, neurocognitive disorders 1338–1339, 1339 French Vasculitis Study 482–483 Fresh frozen plasma (FFP), coagulopathies 1126–1127, 1131–1132 Friedreich’s ataxia (FA) 9 Frog venom 988 ‘Frozen bone’ 536

FTL (ferritin light polypeptide) gene 860 Fulminant hepatic failure 1258, 1261, 1262, 1262 Functional magnetic resonance imaging (fMRI) 101 Fungal infections 1383–1401 central nervous system (CNS) 1248–1249 clinical syndromes 1385–1386, 1385 epidemiology 1383 imaging 1387–1388, 1388, 1389, 1390, 1391, 1392, 1393 laboratory diagnosis 1386–1387 cerebrospinal fluid (CSF) 1386 markers 1387 PCR-based assays 1387 serum antigens/antibodies 1386 myelopathy 1535 pathogenesis 1384–1385, 1384 pathogens 1383–1384 predisposing factors 1384 specific infections 1391–1398 therapy 1388–1391, 1395 antifungal agents 1388–1391, 1395 surgery 1390–1391 Furosemide 1628–1629, 1758–1759

G GABAB-Abs (antibodies) 1162 limbic encephalitis (LE) 1166, 1168 Gabapentin 521, 780, 846–847, 1332–1333, 1640 adverse effects 1640, 1640 elimination 418, 425, 426, 427 Gain-of-sodium disorders 370 Gait disorders 183–184, 1145 Galantamine 1642 Galen 241 Gallium nitrate 878 Gambierdiscus toxicus 996 Gamma knife 1152, 1189 g-Aminobutyric acid (GABA) 843 Ganciclovir 1331, 1333–1334, 1380, 1381 Ganglionated plexus ablation 143–144 Garin–Boujadoux–Bannwarth syndrome 1475, 1480 Gastric H2-receptor inhibitors 1655 Gastric outlet obstruction (GOO) 1241 Gastric protonic pump inhibitors 1655–1656 Gastroesophageal reflux (GER) 260 Gastrointestinal complications Behc¸et’s syndrome (BS) 1705, 1717 primary hyperparathyroidism (PHPT) 867 respiratory failure (RF) 281 treatment 782 Gastrointestinal drugs 633–643, 634 acid-related disorders 634, 638–639 antiemetics 633–637 central hyperthermia syndromes 639 laxatives 634, 638 motility 634, 637–638 antimotility 634, 638 promotility 634, 637–638 neurologic adverse effects 634

I17 Gastroprokinetic drugs 1655 Gatifloxacin 1491 Gemcitabine 1203, 1208 Gemfibrozil 1638 Gemtuzumab ozogamicin (GO or Mylotarg®) 1136 Genital ulcers 1704 Genitourinary drugs 1655–1656, 1656 Genitourinary neuropathy 782 Genitourinary tract cancers 1144 Germ cell tumors, radiotherapy (RT) 1185 Germinomas 1185 Giant cell arteritis (GCA) 227–229, 228, 479–481 clinical features 480, 480 hypothyroidism 711 Giant cell vasculitis 477 Giardia spp 1113–1114, 1114 Gitelman’s syndrome 872 Glandular tissue, autoimmune-mediated destruction 799 Glasgow Coma Scale (GCS) 29, 116, 1365, 1366, 1373, 1756 traumatic brain injury (TBI) 1754, 1754, 1755, 1758, 1760 Glatiramer acetate (GA) 1607–1608 Glia cells, fibromyalgia 517 Glibenclamide (glyburide) 817, 818 Gliclazide 817, 818 Glimepiride 817, 818 Glioblastoma multiforme (GBM) 299, 1263 Glioblastoma, radiotherapy (RT) 1184 Gliomas, low-grade 1184 Glipizide 818 Global brain injury 1191–1192 Global Polio Eradication Initiative (WHO) 1777 Global Stroke Initiative (WHO/ISS/WFN) 1778 Glossopharyngeal nerve (cranial nerve IX) 308 ‘Gloves and socks’ syndrome 1351 Gloydius 992–993 Glucagon-like peptide-1 (GLP-1) receptor analog 820 Glucocorticoids 31, 866, 1357, 1719–1720, 1737–1738 Glucose-6-phosphatase (G6Pase) deficiency 828, 833 Glucose-6-phosphate deaminase (G6PD) deficiency 1012 a-Glucosidase inhibitors 818–819 Glutamine 1261 Gluten-related diseases (GRD) 607–619 diagnosis 608–609, 608 epidemiology 608 neurologic manifestations, spectrum 609–614 epilepsy 613, 613 gluten ataxia (GA) 609–611, 610 gluten encephalopathy 612, 612 gluten neuropathy 611–612 myelopathy 614, 614 myoclonic ataxia 612–613 myopathy 613–614, 614

I18 Gluten-related diseases (GRD) (Continued ) stiff man syndrome (SMS) 614 pathogenesis 614–616, 615 Glyburide 817, 818 Glycerol 1371 Glycogen storage disease (GSD) type I (von Gierke disease) 833 Glycopeptides 1646 Glycoprotein IIB/IIIA antagonists 1129 Glycosides (digitalis) 1637 Glycyrrhetinic acid 373 Glycyrrhiza glabra 373, 374 Gnathostoma spp 1430–1432 Gnathostoma doloresi 1430–1431 Gnathostoma hispidum 1430–1431 Gnathostoma malaysiae 1430 Gnathostoma nipponicum 1430–1431 Gnathostoma spinigerum 1430–1431, 1432, 1433, 1537–1539, 1538, 1539 Gnathostomiasis 1430–1432, 1432, 1538–1539, 1539 clinical manifestations 1431 diagnosis 1431 epidemiology 1430 pathogen 1430–1431 treatment 1431–1432 Gold salts 1650 Gonadotropin-releasing hormone (GnRH) 689–691 Gout 827–829, 828 GRACE (Global Registry of Acute Coronary Events) trial 101–103 Graduated compression stockings (GCS) 296 Graft rejection 1278–1279 Graft-versus-host disease (GVHD) 1313, 1314 Gram-negative bacteria (bacilli) 67, 81–83 Granisetron 636, 637 Granulocytic sarcomas (chloromas) 1040 Granulomatosis see Wegener disease Granulomatous encephalitis 1411 Gravely ill, neuropathy 751–752 see also Critical illness entries Graves’ disease 714, 716–717 seizures 722 Graves’ ophthalmopathy 716–719 clinical features 716–717, 717 diagnostic tools 717–718 pathogenesis 718 prognosis 719 treatment 718–719 Gray-out 172 Green mamba 990, 992 Grisel’s syndrome 442–443 Gr€onblad–Strandberg syndrome see Pseudoxanthoma elasticum (PXE) Gross tumor volumes (GTV) 1181–1182 Growth hormone (GH) 686 acromegaly 691–692 deficiency 690 excess 691 fibromyalgia 517

INDEX Growth hormone-releasing hormone (GHRH) 691 Guanylic monophosphate (GMP) 830 Guidant PRIZM AVT 141 Guillain–Barre´ syndrome (GBS) (AIDP) 841, 842, 845, 1333 cardiology 9–10 decompression illness (DCI) 964 inflammatory demyelinating polyneuropathy (IDP) 1333 influenza vaccine 1552, 1553 nephrotic syndrome 410 pregnancy 1611 renal transplantation 1251 respiratory failure (RF) 276 Gulstonian lectures (1885), malignant endocarditis 75–76 Gummas 1465, 1467–1468 GUSTO-1 (Global Utilization of Streptokinase and TissuePlasminogen Activator (tPA) for Occluded Coronary Arteries) trial 45, 103–105, 106 GUSTO-V (Global Use of Strategies To Open Coronary Arteries) trial 100

H H1N1 influenza A 1114, 1114 H1-receptor antagonists 1651–1654 HACEK group gram-negative bacilli 75, 80–81 Hadju–Cheney syndrome 444 Haemophilus influenzae 1367, 1369, 1611 Haemophilus influenzae type B (Hib) 1361–1362, 1370 vaccine 1550, 1554–1555, 1556–1557, 1651 Halothane 1625, 1627 Handbuch der Neurologie 1445 Hand-foot syndrome 1204 Hardwick’s sea snake (Lapemis curtus) 992 HARP syndrome (hypo-blipoproteinemia, acanthocyosis, retinitis pigmentosa, pallidal degeneration) 859 Hartnup disease 902–904 HAS-BLED score 122–123, 123 Hashimoto’s encephalopathy (HE) 724–725 clinical features 724 diagnosis 725 antithyroid antibodies 725 differential 725 tests 725 neuropathology 725 pathophysiology 724–725 treatment 725 Hashimoto’s thyroiditis (HT) 724–725 Head trauma, cardiology 6–7 Headache brain metastases 1144 cerebral venous thrombosis (CVT) 1605–1606 chronic morning (CMH) 254 cluster 259

Headache (Continued ) developing world 1776 giant cell arteritis (GCA) 228 gluten-related diseases (GRD) 612 Henoch–Sch€ onlein purpura (HSP) 1105 hyperthyroidism 723 intestinal transplantation (ITx) 1270–1271, 1274 intracranial pressure 1220–1221 medication overuse (MOH) 582 migraine 7–8, 964, 1736–1737, 1776 multiple organ transplantation 1312 neuro-Behc¸et syndrome (NBS) 1710–1711, 1710 neurocysticercosis (NCC) 1450 neuropsychiatric systemic lupus erythematosus (NPSLE) 465 obstructive sleep apnea syndrome (OSAS) 254, 259 Paget’s disease of bone (PDB) 530 pancreas/small bowel transplantation 1278–1279 pituitary adenomas 688 polycythemia vera (PV) 1074–1075 pregnancy 1613–1614 small bowel transplantation 1287–1288 thunderclap see Reversible cerebral vasoconstriction syndrome (RCVS) Head-up tilt test 185–186 Healthcare systems, developing world 1773–1774 Hearing loss 530, 878 Heart Disease and Stroke Statistics (AHA) 93 Heart transplantation 1227–1236 heart-lung transplantation 1313 long-term aspects 1230–1235 central nervous system (CNS) 1232, 1233 children 1234–1235 encephalopathy 1232 immunosuppression 1231–1232 peripheral nervous system (PNS), complications 1234 seizures 1233–1234, 1235 stroke 1233, 1235 postoperative complications 1230 central nervous system (CNS) 1230 peripheral nervous system (CNS) 1230 preoperative evaluation 1229–1230 central nervous system (CNS) 1229 peripheral nervous system (PNS) 1230 Heart valves, prosthetic 63–65 Heat-related illness see under Body temperature Heavy metals, disorders of 852 see also Copper disorders; Iron disorders Helicobacter pylori 638, 917, 918, 1007, 1541 HELLP syndrome, pre-eclampsia-induced 1596, 1597 Helminthic infestation 1414–1436, 1415

INDEX Hemangiomas, vertebral 1616 Hematin 846 Hematocrit (HCT) 1005–1006 Hematologic agents 136–138, 137, 138 Hematologic disorders, treatment 1125–1141 anticoagulation 1126, 1131–1132 intracranial hemorrhage (ICH) 1131–1132, 1131 benign disorders 1125–1131, 1126 antifibrinolytic agents 1128 antiplatelet agents 1129–1130 antithrombotic agents 1130–1131 replacement therapies 1125–1128 leukemias 1040–1041 lymphomas 1037–1038 malignancies 1126, 1132–1137 chronic myeloproliferative neoplasms 1132–1133 leukemias/lymphomas/multiple myeloma (MM) 1134–1137 myelodysplasia 1133–1134 Hematopoietic progenitors 1078 Hematopoietic stem cell therapy 762 Hematopoietic stem cell transplantation (HSCT) chronic graft-versus-host disease 1302 encephalopathy 1297–1299 metabolic 1297–1299 treatment-induced 1299 historical perspective 1295–1296 immunosuppression 1299–1301 infection 1301–1302, 1302 multiple organ, combined 1314 neurologic complications 1296–1297 allogeneic HLA-matched transplantation 1296 reduced-intensity allogeneic transplantation 1296–1297 Heme biosynthetic pathway 839–840, 840 Hemiscorpion lepturus 988, 994 Hemodialysis, antiepileptic drugs (AEDs) 427 Hemoglobin (HGB) 1005–1006, 1008, 1012 Hemoglobinopathies 1013 Hemolytic anemias see under Anemias Hemolytic uremic syndrome (HUS) 1113–1123 clinical manifestations 1115 definition 1113 diagnosis 1118–1119, 1119 epidemiology/etiology 1113–1115, 1114 atypical (aHUS) 1114 thrombotic thrombocytopenic purpura (TTP) 1114–1115 typical 1113–1114 neurologic findings/sequelae 1115–1118, 1118 pathophysiology 1115 atypical (aHUS) 1115 typical 1115 treatment 1120 antibiotics 1120 cobalamin replacement 1120

Hemolytic uremic syndrome (HUS) (Continued ) complement therapy 1120 immunotherapy 1120 plasma infusion/plasmapheresis 1120 transplantation 1120 Hemophilia 1045, 1046–1052 acquired 1052–1053 classification 1049 clinical manifestations 1047, 1049–1050 frequency, bleeding 1049 symptoms, bleeding 1049–1050 coagulation factors VII/IX inhibition 1052 diagnosis 1050 coagulation tests 1050 genetic testing 1050 epidemiology 1046 genetics 1046–1049 management 1050–1052, 1051, 1055 neurologic manifestations 1053–1054 age-related comorbidities 1054 central nervous system (CNS) 1053 peripheral nervous system (PNS) 1053–1054 treatment centers 1058 HEMORR2HAGES scheme 122–123, 123 Hemorrhage brain metastases 1149 intracerebral (ICH) see Intracerebral hemorrhage (ICH) intracranial see Intracranial hemorrhage (ICH); Intracranial hemorrhage/aneurysms intraparenchymal 5 polycythemia vera (PV) 1075 subarachnoid (SAH) see Subarachnoid hemorrhage (SAH) Hemorrhagic complications 570 characteristics/management 570 mechanisms/frequency 570 Hemorrhagic stroke decompression illness (DCI) 964 pregnancy 1602–1605 Henderson–Hasselbalch formula 375 Henoch–Sch€ onlein purpura (HSP) 1101–1112 diagnostic criteria 1102, 1102 differential diagnosis 1107–1108 etiology/pathogenesis 1102–1103 historical perspective 1101 investigations 1106–1107 antineutrophil cytoplasmic antibodies (ANCA) 1106 cerebrospinal fluid (CSF) analysis 1107 imaging 1107 nonspecific laboratory findings 1106 management 1108 nervous system manifestations 1103–1106 epidemiology 1103 pathology 1102 prognosis 1108

I19 Henoch–Sch€ onlein purpura (HSP) (Continued ) systemic disease 1101 Heparin 65, 105, 299, 1638 fractionated 1719–1720 unfractionated see Unfractionated heparin(UFH) see also Low molecular weight heparin (LMWH) Heparin-induced thrombocytopenia (HIT) 137, 295, 1638 and thrombosis (HITT) 1130–1131 Heparinoids 1638 Hepatic disease see Antiepileptic drugs (AEDs), hepatic/renal disease Hepatic drugs 675–682, 676 Hepatic encephalopathy (HE) acute liver failure (ALF) 646, 646, 661 classification 661–662, 662 portosystemic see Portosystemic encephalopathy types 661 Hepatic myelopathy (HM) 663 Hepatitis A virus (HAV) 1651 Hepatitis B virus (HBV) 678–680, 1333–1334 vaccine 1550, 1553, 1651 Hepatitis C virus (HCV) 678–680, 1333–1334 Hepatocyte infusion 656 Hereditary hemochromatosis (HH) 860 Hereditary hemorrhagic telangiectasia (HHT) 1562, 1571–1572 clinical manifestations 1571–1572 arteriovenous malformations (AVMs) 1572 mucocutaneous telangiectasias 1571–1572, 1571 neurologic 1572 diagnosis/genetic testing 1572 Hereditary motor and sensory neuropathy IV (HMSN IV) 1579 Hereditary neuropathy/dystrophy/ myopathy, pregnancy 1612 Hereditary spherocytosis 1012 Hereditary xanthinuria 828, 833 Herpes simplex encephalitis (HSE) 1149 Herpes simplex virus (HSV), encephalitis 1232, 1252, 1378 Herpes simplex virus type 1 (HSV-1) encephalitis 1377, 1378–1379 diagnosis 1380 treatment 1380–1381, 1380 heart transplantation 1232 renal transplantation 1249 Herpes simplex virus type 2 (HSV-2) 1232 Herpes simplex virus type 6 (HSV-6) 1232 Herpes viruses 1249, 1527 Herpes zoster ophthalmicus (HZO) 1562, 1569, 1569 Herpes zoster (zona/shingles) 1577–1578 clinical manifestations 1577–1578 cutaneous 1577 neurologic 1577–1578 diagnosis 1578

I20 Heterometrus fulvipes 993 Heterometrus longimanus 993–994 Heterometrus scaber 993 Heterometrus spinifer 993–994 Hexamethylmelamine 1210 Hexosamine pathway 777–778 High density lipoproteins (HDL) 624 High-altitude periodic breathing 264 High-grade glioma (HGG) 1222 Highly active antiretroviral therapy (HAART) cryptococcal meningitis 1330 HIV 1321, 1323 -associated neurocognitive disorder (HAND) 1340 /tuberculosis (TB) coinfection 1338, 1338 Hodgkin lymphoma (HL) 1034 primary central nervous system lymphoma (PCNSL) 1326 progressive multifocal leukoencephalopathy (PML) 1328 Hippocrates 1506 Hirudin (lepirudin) 1131 Histamine 2 (H2) blockers 638–639 Histoplasma antigen 1386, 1398 Histoplasma capsulatum 1383, 1384, 1389–1390, 1398 Histoplasmosis 1385, 1398 HIV Medicine Association (IDSA) 1324–1325, 1329, 1331 HIV-infected patients 755, 1319–1344 developing world 1776–1777 etiology/diagnostic approach 1321, 1322 hemolytic uremic syndrome (HUS) 1114, 1114 HIV-associated dementia (HAD) 1339–1340, 1340, 1778 HIV-associated neurocognitive disorder (HAND) 1321, 1338–1340 classification 1338–1339, 1339 clinical features 1339, 1340 diagnosis 1339–1340 prevalence 1339 treatment 1340 HIV-associated vacuolar myelopathy (VM) 1321, 1326, 1331–1332 clinical features 1331–1332, 1529 diagnosis 1332 epidemiology 1331 incidence/prevalence 1331 pathology 1331 treatment 1332 Hodgkin lymphoma (HL) 1034 myelopathy 1322, 1529–1530, 1529 neuromuscular disorders 1332–1334 neurosyphilis 1334–1337, 1533–1534 clinical features 1334–1337, 1336, 1534 diagnosis 1334–1336, 1467, 1467, 1534 epidemiology 1334–1337, 1462, 1533–1534 laboratory findings 1336 natural history 1465–1466

INDEX HIV-infected patients (Continued ) neuropathology 1534, 1534 stages 1336 treatment 1336–1337, 1337, 1469–1470 opportunistic infection 1323–1331 pathogenesis, neurologic complications 1321–1323, 1322 primary central nervous system lymphoma (PCL) 1028–1029 spinal cord disorders 1331–1332 tuberculosis (TB), central nervous system (CNS) 1337–1338, 1485, 1488, 1494–1495 clinical features 1337 diagnosis 1337–1338 neuroimaging 1338, 1489 pathogenesis 1337 treatment 1338, 1338, 1490–1492 HMG-CoA reductase inhibitors 1636–1637, 1638 Hodgkin lymphoma (HL) 1034–1035 Hodgkin Reed–Sternberg cells (HRS) 1034 ‘Hole-in-donut’ appearance 1537 Holt–Oram syndrome, genetics 53 Home mechanical ventilation (HMV) 285 Homeostasis, disorders of 1104–1105 Homocysteine (Hcy) 917, 919–921, 923, 931–932 Homocystinuria 572–573 description 572 neurologic complications 572–573 Homozygous achondroplasia 560 Hormonal disorders 410 hyperfunction 690–694 Hormone therapy see Endocrine therapy Horned vipers (Cerastes) 992–993 Horner’s syndrome 7 Hornet stings 995 Hu-Abs (antibodies) 1161, 1162–1163 limbic encephalitis (LE) 1166, 1168 subacute cerebellar degeneration (SCD) 1164 Human herpes virus-6 (HHV-6) encephalitis 1377, 1379 diagnosis 1380 treatment 1381 focal encephalitis 1232 liver transplantation 1262–1263 pancreas/small bowel transplantation 1278–1279 renal transplantation 1249 treatment 1380 Human immunodeficiency virus see HIV-infected patients Human papilloma virus (HPV) 1554 Human parvovirus B19 1350–1351 clinical manifestations 1351 diagnosis 1351 epidemiology 1350 treatment/prognosis/prevention 1351 Human rabies immunoglobulin (HTIG) 1504

Human resources, developing world 1774–1775 Human T cell lymphotropic virus-1 (HTLV-1) 1527–1529, 1540 clinical features 1527–1528 differential diagnosis 1529 epidemiology 1527 imaging 1528 laboratory findings 1528 neuropathology 1528–1529 public health measures 1529 treatment 1529 virology 1527 Human T cell lymphotropic virus-2 (HTLV-2) 1527, 1529 Human tetanus immunoglobulin (TIG) 1508, 1509 Huntington’s disease (HD) 1780 Hydatid disease 1425–1426 clinical manifestations 1425 diagnosis 1425 epidemiology 1425 pathogen 1425 therapy 1425–1426 Hydralazine 165, 165, 1697 Hydrocephalus 308–309, 309, 322 achondroplasia 444, 552–554, 553, 554 Hydrochlorothiazide 764–765 Hydrophiinae 992 Hydrophis cyanocinctus 989, 992 Hydrophis fasciatus atriceps 992 Hydrosoluble vitamins 889–914 ascorbic acid see Vitamin C deficiency future trends 910 historical perspective 891–895 investigations 894 niacin (nicotinic acid) see Vitamin B3 deficiency overview 892 pantothenic acid see Vitamin B5 deficiency pyridoxine (pyridoxal) see Vitamin B6 deficiency riboflavin see Vitamin B2 deficiency thiamin see Vitamin B1 deficiency HydroxoCbl 923 Hydroxycarbamide 1075 Hydroxychloroquine 325, 326, 628 5-Hydroxytryptamine 3 (5-HT3) receptor antagonists 636–637 Hydroxyurea (HU) 1021, 1132–1133, 1208 Hymenoptera venom 988 Hyperaldosteronism 763–765 clinical manifestations 764 diagnosis 764 etiology 764 primary hyperaldosteronism 764 secondary hyperaldosteronism 764 historical perspective 763–764 treatment 764–765 Hyperbaric oxygen (HBO) 977 Hypercalcemia 378, 865–866 associated disorders 738–742 causes 739

INDEX Hypercalcemia (Continued ) classification 865–866 clinical features 866 renal transplantation 1251–1252 treatment 741–742, 866 Hypercapnia 273 Hyperchloremic acidosis 375 Hypercoagulability 405–407, 406 Hyperekplexia 182 Hyperglycemia 328, 1251, 1286 Hyperkalemia 374–375 clinical findings 374 laboratory investigations 374, 375 management 374–375 terminology 374 Hyperkinetic movement disorders 1780 Hyperlipidemia 407 Hypermagnesemia 378, 873–874 clinical manifestations 873–874 treatment 874 Hypernatremia 369–372 clinical findings 369–370 historical perspective/terminology 369 laboratory investigations 370, 370, 371 management 370, 371–372 pathophysiology 370–371 Hyperosmolar therapy, traumatic brain injury (TBI) 1628 Hyperparathyroidism primary see Primary hyperparathyroidism (PHPT) pseudopseudohyperparathyroidism 745 secondary 742–743, 744 tertiary 742–743, 744 Hyperperfusion syndrome (HS) 234, 1695 Hyperphosphatemia 378, 379, 875 classification 875 clinical manifestations 875 treatment 875 Hyperplasia, pituitary 686 Hyperprolactinemia 690–691, 691 Hypertension (HTN) arterial 258 nephrotic syndrome 407 primary hyperparathyroidism (PHPT) 867, 869 pulmonary 258 see also Hypertension/hypertensive encephalopathy Hypertension/hypertensive encephalopathy 161–168 clinical findings 162 genetics 163 Guillain–Barre´ syndrome (GBS) 9–10 historical perspective/terminology 161 laboratory investigations 162 management 164–166 natural history 162 neuroimaging 162–163, 163, 164 pathology 163–164, 164 Hypertensive encephalopathy 1104 posterior reversible encephalopathy syndrome (PRES) 1692, 1693

Hypertensive encephalopathy (Continued ) treatment 1697 see also Hypertension/hypertensive encephalopathy Hyperthermia drug-induced 949–951 clinical features 949–951 laboratory studies 951 management 951 malignant 1612 pathogenesis 947 Hyperthyroid myopathy 712–714 clinical features 712–713 inflammatory 714 investigations 713 skeletal muscle changes 713–714 treatment/prognosis 714 raised creatine kinase (CK) 714 Hyperthyroidism 712–725 associated conditions 723–724 corticospinal tract disorders 715 encephalopathy see Hashimoto’s encephalopathy (HE) mental/psychiatric disorders 722–723 movement disorders 715–716 myopathy see Hyperthyroid myopathy ophthalmopathy see Graves’ ophthalmopathy paralysis see Thyrotoxic hypokalemic periodic paralysis (TPP) peripheral neuropathy 714–715 seizures 722 thyroid storm 721–722 Hypertonic saline 1757–1759 Hypertrophic cardiomyopathy 124 Hyperventilation, bacterial meningitis 1371–1372 Hyperviscosity syndrome 1093 Hypervolemic hypernatremia 370 Hypervolemic hyponatremia 367, 368–369 Hypnale 992–993 Hypnotics 1644 Hypoalbuminemia 377 Hypocalcemia 377–378, 377, 742–745, 870 causes 743 classification 870 clinical features 743–745, 870 pathophysiology 742–743 treatment 745, 870 Hypoglossal nerve (cranial nerve XII) 308 Hypoglycemia 177, 818 insulin treatment 815 total parenteral nutrition (TPN) 1286 Hypogonadism 328 secondary 689–690 Hypokalemia 372–374 clinical findings 372 laboratory investigations 373, 373 management 373–374 pathophysiology 373, 374 periodic paralysis 372 terminology 372, 373 Hypomagnesemia 377–378, 739 classification 872 clinical manifestations 872–873

I21 Hypomagnesemia (Continued ) drug-induced 871 etiologies 741, 742 functional hypoparathyroidism 871 neurologic manifestations 741 renal transplantation 1252 treatment 873 Hypomelanosis of Ito (HI) 1562, 1588 cutaneous manifestations 1588 neurologic manifestations 1588 Hyponatremia 365–369 clinical findings 366–367, 367 historical perspective/terminology 365, 367 laboratory investigations 367–368, 368 management 367, 368–369, 369 pathophysiology 368 renal transplantation 1252 Hypoparathyroidism/ pseudohypoparathyroidism 377, 871–872, 874 clinical manifestations 871 treatment 871–872 Hypophosphatasia 878–879 clinical manifestations 879 diagnosis 879 treatment 879 Hypophosphatemia 378, 739, 874–875 classification 874 clinical manifestations 874 etiologies 741, 741 neurologic manifestations 741 treatment 875 Hypopituitarism 688–689 Hypoplastic left heart syndrome (HLHS) 1234 Hypothalamic hormones/analogs 809–812, 810 Hypothalamic ‘osmostat’ 310 Hypothalamic-pituitary-adrenal (HPA) axis 517 evaluation 686, 690 Hypothermia accidental 953–954 clinical features 953, 953 laboratory studies 953, 954 management 953–954 effects 1631 intracranial pressure (ICP) 1759 pathogenesis 947–948 therapeutic 29–30, 954–955, 955 traumatic brain injury (TBI) 1628 Hypothermia After Cardiac Arrest (HACA) Study Group 26, 29–30 Hypothermic circulatory arrest (HCA) 234 Hypothyroid myopathy 709–711 clinical features 709–710, 710 investigations 710 pathology 710–711 pathophysiology 711 treatment/prognosis 711 Hypothyroidism 328, 703–711 associated neurologic conditions 711–712 cerebellar ataxia 708

I22 Hypothyroidism (Continued ) congenital see Congenital hypothyroidism (CH) cranial nerve disorders 708–709 encephalopathy/coma/seizures 705–706 mental changes 706–707 myopathy see Hypothyroid myopathy peripheral neuropathy 707–708 secondary 689 sleep disorders 708 Hypotonic hyporesponsive episode (HHE) 1555–1556 Hypoventilation 263–266, 280, 281 Hypovolemic hypernatremia 370 Hypovolemic hyponatremia 367–368, 367 Hypoxanthine-guanine phosphoribosyltransferase deficiency (HPRT) 828, 830–831, 830 Hypoxemia 265–266, 273, 280, 281 Hypoxia 274 Hypoxic-ischemic encephalopathy 197, 274

I Iatrogenic neurology 1635–1671 antiallergic drugs 1651–1654 antiarrythmics/inotropes/coronary artery dilators 1636, 1637 antibiotics 1644–1645, 1646 anticoagulants 1638–1639 antidepressant drugs 1642 antiepileptic drugs (AEDs) 1639–1640, 1640 antifungal agents 1645, 1649 antihypertensive agents 1635–1636, 1636 anti-inflammatory/analgesic drugs 1645–1651 antilipemic drugs 1636–1637, 1638 antineoplastic drugs 1645, 1652 antiparasitic agents 1645, 1650 antiparkinsonian drugs 1641, 1641 antiplatelet agents 1639 antipsychotics/dopamine receptorblocking drugs (DRBDs) 1642–1643, 1643 antispastic drugs 1640–1641 antiviral agents 1645, 1648 cognition drugs 1641–1642, 1642 genitourinary/digestive tract drugs 1655–1656, 1656 hormone-related drugs 1654, 1655 immunomodulatory drugs 1645, 1653 metabolism drugs 1654–1655 psychostimulants 1644, 1645 respiratory tract drugs 1655 sedatives/hypnotics 1644 thrombolytics 1637–1638 vaccines 1645, 1651 IBMPFD (inclusion body myopathy, Paget disease of bone, frontotemporal dementia) disorder 533, 534 Ibritumomab (Zevalin®) 1136 Ibuprofen 1571 Ibuprofen-induced meningitis 1648

INDEX Ibutilide 132, 134 Idebenone 9 Idiopathic inflammatory myopathy (IIM) 495–512, 1160, 1160, 1172 classification 496 clinical features 498–499, 498, 500 epidemiology 497 etiopathogenesis 497–498 laboratory investigations 501–505 autoantibodies 501–502 electromyography (EMG) 502 muscle biopsy 502–505, 503, 504 serum creatine kinase (sCK) activity 501 skin biopsy 502 management 505–507 corticosteroids 506–507 future therapeutic prospects 507 second-line treatment 507 natural history 499–501 neuroimaging 505, 506 Idiopathic myelofibrosis (IMF) 1132 Idiopathic reversible cerebral vasoconstriction syndrome (RCVS) 1726 Ifosfamide 1200, 1201, 1201, 1210 IFRT (involved-field radiotherapy) 1035 IGG- and IGA-related neuropathy 1088 IGM-related peripheral neuropathy anti-MAG negative and/or no detectable antibody 1084, 1087–1088 anti-MAG positive 1084, 1085–1087 Imatinib 1133, 1205 Imatinib mesylate (Gleevec™) 1133 Imipenem 1247, 1644 Immobility, end of life (EOL) phase 1220 Immune reconstitution inflammatory syndrome (IRIS) 1330 antifungal agents 1389–1390 antiretroviral therapy induced- 1325 cytomegalovirus encephalitis (CMV) 1331 syphilis 1466 tuberculosis 1338 Immune-inflammatory disorders 1540 Immunity, pregnancy 1596 Immunoglobulins 1173 Immunomodulatory drugs (IMiDs) 1136, 1645, 1653 Immunosuppressants 1120, 1613 adverse effects 675, 676, 1246, 1278–1279 posterior reversible encephalopathy syndrome (PRES) 1692, 1694, 1698 transplantation 677–678, 1231–1232, 1246, 1268, 1281–1282, 1299–1301 Impaired glucose tolerance (IGT) 773 Implants cardiac defibrillators (ICDs) 141, 202–203 complications 157 cardiovascular electronic devices 85–86 risks 1746 Impotence 254

Inactive sclerotic phase, defined 533–535 Inclusion body myositis (IBM) see Idiopathic inflammatory myopathy (IIM) Incontinentia pigmenti (IP) (Bloch–Sulzberger syndrome) 1562, 1587–1588 clinical symptoms 1587–1588 central nervous system (CNS) 1587–1588 cutaneous 1587–1588, 1588 ophthalmologic 1587–1588 diagnosis 1588 radiology 1588 treatment 1588 Incretin mimetics 820 Indometacin 1274 Induction agents, neuroanesthesia 1624–1625, 1625 Infantile beriberi 897 Infantile neuroaxonal dystrophy (INAD) 859–860 Infectious Diseases Society of America (IDSA) 1338, 1393 HIV Medicine Association 1324–1325, 1329, 1331 Infectious myelopathy 1522, 1525–1540 Infective (bacterial) endocarditis 56, 67–69, 75–92 antithrombotic therapy 69 cardiac surgery 69 classification 75 historical aspects 75–76 clinical aspects 76–79 central nervous system (CNS) 79 cerebrovascular manifestations 77–79 neurologic manifestations 77, 79 systemic/cardiac/multiorgan manifestations 76–77 complications cerebrovascular 67–68 noncerebrovascular 68–69 definition 75 diagnosis 79–81 diagnostic criteria 81, 82 echocardiography 80, 80 electrocardiography 80 laboratory 80 neurologic complications 81, 83 pathogenic agent 80–81 epidemiology 76 pathophysiology 76 prevention 83 prognosis 81–83 treatment 83–88 antimicrobial 83–84, 84 antithrombotic 86–87 cardiac surgery 84–85, 85, 85, 87 cardiovascular implantable electronic devices 85–86 intracranial infections 88 mycotic aneurysms 87–88 neurologic complications 86, 87

INDEX Infective (bacterial) endocarditis (Continued ) thrombolysis, stroke 86 Infective embolism 52 Infecundity 793–796 Inferior vena cava (IVC) filters 296, 298 Infertility 793–796 causes 793, 794 Inflammatory bowel diseases (IBD) 595–605 cerebrovascular complications 597–599 anti-TNF-a therapy 598, 599 arterial ischemic stroke (AIS) 598 cerebral venous thrombosis (CVT) 599–600 pathophysiology 598 vasculitis 598–599 neurologic manifestations 596–597, 596 central nervous system (CNS) 596, 600–601 cranial neuropathy 597 demyelinating disease 600 medication-induced 601 pathophysiology 596–597 peripheral nervous system (PNS) 596, 597 prevalence 596 psychiatric syndromes 601 Inflammatory demyelinating polyneuropathy (IDP) 1333, 1335 treatment 1333 Inflammatory joint disorders 440–442 see also Rheumatoid arthritis (RA) Inflammatory myopathy, pregnancy 1612 Inflammatory neuropathy/myopathy 278–279 Inflammatory spondyloarthropathy 454–457 acute spinal fractures 455 cauda equina syndrome 456 clinical features 454 discovertebral destruction 456 laboratory investigations 455 neurologic aspects 455 pathogenesis 454–455 Infliximab 479, 507, 601, 1719 neurosarcoidosis 325, 326, 327 Influenza vaccine 1550, 1552 influenza A (HIN1) 1553 trivalent inactivated vaccine (TIV) 1552–1553 Inhalation agents, neuroanesthesia 1625–1626, 1627 Inherited neurodegeneration with brain iron accumulation (NBIA) 858–860 type 1: pantothenate kinase-associated neurodegeneration 859, 859 type 2: classic infantile neuroaxonal dystrophy (INAD)/atypical neuroaxonal dystrophy (ANAD) 859–860 aceruloplasminemia 853, 860

Inherited neurodegeneration with brain iron accumulation (NBIA) (Continued ) idiopathic neurodegeneration with brain iron accumulation 860 neuroferritinopathy 860 Inherited thrombophilia 1061–1063, 1064–1065, 1064, 1066–1067 Inhibitory Control Test (ICT) 665 Inner ear barotrauma 964 Inner ear decompression sickness (IEDCS) 963 Inosine monophosphate (IMP) 827, 830 Inotropes 1636, 1637 Institute of Medicine (IOM) 1551 Insufflation-exsufflation devices 284 Insulin tolerance test (ITT) 759 Insulin/analogs 814–816, 815, 816 age constraints 814, 815 diabetic ketoacidosis (DKA) 377 intermediate and long-acting 811 neuritis 815 short- and rapid-acting 811 structure/bioavailability 815 Intensity-modulated radiotherapy (IMRT) 342, 1182–1183, 1186, 1187–1188 Intensive care unit (ICU) tetanus 1509 see also Cardiology, vascular/intensive care neurology Intensive care unit-acquired weakness (ICUAW), focal 1681 Intensive care unit-acquired weakness (ICUAW), generalized 1673–1685 clinical presentation 1673–1674 critical illness myopathy (CIM) 1674–1676, 1680 critical illness neuromyopathy (CINM) 1675 critical illness polyneuropathy (CIP) 1674, 1675–1676, 1680 definitions 1673 diagnostic methods 1678–1680 electromyography (EMG) 1679 laboratory tests 1680 muscle/nerve biopsy 1679–1680 nerve stimulation 1678–1679 differential diagnosis 1677–1678, 1678 incidence 1674 myasthenia gravis (MG) 1676 outcome/prognosis 1680 pathophysiology 1675 prolonged neuromuscular junction (NMJ) block 1676, 1677 rhabdomyolysis 1676–1677 Interferon-a (IFN-a) 485–486, 1504, 1718–1719 adverse effects 1201, 1204 Interferons (INF) 678–679, 1075, 1381, 1607–1608, 1719 adverse effects 676, 678–679, 1201, 1212, 1653 mechanisms of action 678 Interleukins (IL) 1201, 1212, 1653 Intermediate leprosy 1573–1575

I23 Intermittent hemodialysis (IHD) 400 Intermittent pneumatic compression (IPC) 294–295 Internal cardioversion 140 Internal target volume (ITV) 1181–1182 International Classification of Headache Disorders (ICHD-II) 254, 1726 International Classification of Sleep Disorders (ICSD2) 251, 261 International Collaboration on Endocarditis-Prospective Cohort Study 76, 77, 81–83 International Committee for Standardization in Hematology 1016–1017 International Headache Society 1729, 1776 International HIV dementia scale 1339–1340 International League Against Epilepsy 1775 International Liaison Committee on Resuscitation (ILCOR) 28, 30 Advanced Life Support Task Force 30 International Myositis Assessment and Clinical Studies Group (IMACS) 507 International Normalized Ratio (INR) 113, 120, 140 International Pancreas Transplant Registry 1280 International Prognostic Scoring System (IPSS) 1009 International Society for Heart and Lung Transplantation (ISHLT) 1238 International Society for Hepatic Encephalopathy and Nitrogen Metabolism (ISHEN) 663–664 International Stroke Society (ISS) 1778 International Stroke Trial 296–297 International Subarachnoid Aneurysm Trial 1604 International Verapamil SR-Trandolapril Study (INVEST) 93 International Workshop on Primary Hyperparathyroidism (Third) 869 Interstitial brachytherapy 339 Interstitial lung disease (ILD) 499, 500, 501 Interventional cardiology see Cardiac surgery/interventions Intestinal allograft recipients 1287 Intestinal failure 1306 defined 1305–1307 Intestinal transplantation (ITx) 1267–1276 immunosuppression 1268 indications/contraindications 1267, 1268 isolated 1308 -liver 1308–1309 neurologic complications 1268–1275 clinical presentation 1270–1273 diagnostic approach/management 1273–1275 epidemiology 1269 etiology 1269–1270, 1270 surgical procedures 1267–1268

I24 Intra-aortic balloon pump (IABP) 201 Intra-arterial digital angiography 79, 81 Intracerebral hemorrhage (ICH) 5, 198 management 103–104, 105 pregnancy 1604 renal disease 387 Intracranial hemorrhage (ICH) 1760 management 1759 pancreas/small bowel transplantation 1278–1279 sickle cell disease (SCD) 1016 treatment 1131–1132, 1131 Intracranial hemorrhage (ICH)/aneurysms characteristics 568 infective endocarditis 68, 78 management 103–104, 105, 568 mechanisms/frequency 568 venous thromboembolism (VTE) 296–297, 298, 299–300, 568 warfarin 138, 138 Intracranial hypertension 1449–1450 Intracranial infections, treatment 88 Intracranial masses 1628–1629 Intracranial plasmacytoma 1091 Intracranial pressure (ICP), increased 177, 400, 1219, 1753–1754, 1758 acute liver failure (ALF) 645, 650, 651–655 anesthesia 1623–1625, 1626–1628 Lund concept 1371 management 1330, 1758–1760 monitors 649–650 treatment 1220–1221 Intraocular cysticerci 1450 Intraparenchymal hemorrhage 5 Intraparenchymal mass lesions 310, 310, 315, 322 Intravascular large B cell lymphoma 1033, 1034 Intravenous immunoglobulin (IVIg) 485–486, 1127, 1333–1334, 1381 123 Iodine-metaiodobenzylguanidine (IMIBG) scintigraphy 8–10 Irbesartan 138 Irinotecan 1212 Iron disorders 858–861 acquired diseases, excess/deficiency 861 acquired neurodegenerative disorders 861 anemia see under Anemias deficiency, anemias 1125–1126 genetic systemic iron accumulation with neurologic features 860–861 metabolism 858, 858 restless leg syndrome (RLS) 861 see also Inherited neurodegeneration with brain iron accumulation (NBIA) Iron-induced organ injury 1020–1021 Irradiation see Radiotherapy (RT) Ischemic infarction 1278–1279, 1287–1288 Ischemic lesions, brain metastases 1149 Ischemic optic neuropathy 198 Ischemic stroke acute see Acute ischemic stroke

INDEX Ischemic stroke (Continued ) cardiac catheterization 43–44 infective endocarditis 67–68, 77–78, 78 large artery atherosclerosis 574 Marfan syndrome (MFS) 574 neurosarcoidosis 311 transient see Transient ischemic attacks (TIAs) Isoflurane 1625, 1627, 1630–1631 Isolated adrenocorticotrophic hormone (ACTH) deficiency 756 Isolated amnesia 35 Isoniazid 1247–1248, 1495, 1646 tuberculous meningitis (TBM) 1490, 1491, 1492 Isoprenaline 132, 134 Isoprinosine 1348–1349 Isoproterenol 132, 134 Italian Society for Haemostasis and Thrombosis (SISET) 296, 297 Itraconazole 1532 Ivermectin 1429, 1431–1432, 1435, 1436 Ivory vertebrae 535 Ixabepilone 1200, 1203 Ixodes ticks 1473–1474, 1479, 1576

J JAK2 gene 1073–1074, 1076, 1077–1078 Japanese encephalitis (JE) 1378, 1379 diagnosis 1380 vaccines 1554 Jarisch–Herxheimer reaction 1469 Jaw claudication 228 JC polyomavirus 1263, 1378 diagnosis 1380 heart transplantation 1232 progressive multifocal leukoencephalopathy (PML) 1232 renal transplantation 1250 JE-VAX vaccine 1554 Jewel AF 141 ‘Jumping Frenchmen of Maine’ 180 Juvenile dermatomyositis (JDM) see Idiopathic inflammatory myopathy (IIM) Juvenile Paget’s disease, clinical characteristics 534

K Kallmann syndrome 689–690, 796 Kanamycin 1491, 1492 Kaplan–Meier event rate 114 Katayama fever 1419–1420 Kayser–Fleischer (KF) rings 854–855, 855 Kearns–Sayre syndrome (KSS) 11, 13 Kennedy’s disease 794, 795 historical perspective 795 laboratory investigations 795 management 795 natural history 795 pathology 795 Kernig’s sign 1514–1515 Ketamine 1090, 1504, 1624–1625, 1625, 1631 Ketoconazole 698, 754

Ketolides 1646 Kidney-pancreas transplantation 1313–1314 King’s college criteria, acute liver failure (ALF) 649, 650, 655 Kinky hair disease (Menkes disease) 626, 851, 853, 853 Klebsiella spp 1301 spinal epidural abscess 1532 Klebsiella pneumoniae 1369 Klippel–Trenaunay–Weber syndrome 1573 Kohlmeier-Degos disease 1562, 1567 Konzo 1540, 1543 Korsakoff psychosis 897, 900 Kraits (Bungarus) 991–992 Kyphoplasty 342–343 Kyphosis 559

L La Crosse virus 1378, 1380 Labetalol 165, 165, 1636, 1697 Lacosamide 780 elimination 418, 424–426, 425, 427 beta-Lactam antibiotics 1247 Lactate dehydrogenase (LDH) 1011 Lactic acidosis 591–592 Lactitol 670 Lactulose 670 Lacunar infarcts 115, 115 Lambert–Eaton myasthenic syndrome (LEMS) 10, 353–354, 1160, 1160, 1171 antibodies 1161 subacute cerebellar degeneration (SCD) 1164 treatment 1173 Lamivudine (Epivir-HBV®) 676, 679 Lamotrigine 846, 1332–1333, 1640, 1640 elimination 418, 423, 425, 427 Lance–Adams syndrome 35 Lance-heads (Bothrops) 992–993 The Lancet 1551 Lanreotide 697, 810, 810, 811 Lansoprazole 638–639 Lapemis curtus 992 Large artery atherosclerosis 574 Large-vessel encephalitis 1577–1578 Laryngeal nerve injury, recurrent 1240 Laser assisted uvulopalatoplasty (LAUP) 262 L-Asparaginase 1201, 1211, 1652 Late-delayed toxicity, radiation therapy 1150 Lathyrism 1547–1546 Lathyrus 1547–1546 Lathyrus sativus (chickling pea) 1547–1546 Laticauda spp 992 Laticauda colubrine 992 3L. laticauda 989 Laticauda semifasciatus 992 Latrodectus spp 988, 994–995 Latrodectus geometricus 994 Latrodectus mactans 994 ‘Laughing gas’ see Nitrous oxide (N2O)

INDEX Laurence–Moon–Bardet–Biedl syndrome 796 Laxatives 634, 638 Lead, excessive exposure 852, 1010–1011 Left atrial appendage, percutaneous closure 142–143 Left ventricular assist devices (LVADs) 201, 202 Left ventricular dysfunction 113–115, 115 Left ventricular noncompaction 124 Legionella spp 80–81 Leigh’s disease 898 Leiurus quinquestriatus quinquestriatus 993–994 Lenalidomide 1032, 1090, 1136, 1204, 1652 Leonine facies 1573–1575, 1574 Lepirudin (hirudin) 1131 Leprosy 1509–1514, 1562, 1573–1576 classification 1509–1510, 1510 clinical manifestations 1573–1575 cutaneous 1574 developing world 1777 diagnosis 1575 investigations/diagnosis 1511–1512 biopsy 1511–1512, 1513 electrophysiology 1511 radiology 1511, 1512 serology 1511 lepromatous 1573–1575 neurology 1510–1511 pathophysiology 1511 treatment 1512–1514, 1513, 1575–1576 Leptomeningeal (LM) manifestations leukemias 1039–1040 lymphomas 1036–1037 metastases 343–345, 344 multiple myeloma (MM) 1091 rheumatoid arthritis (RA) 451 Leptospira 1532 Lesch–Nyhan disease 828, 830–831, 830, 832, 835 variants 831, 835 Letrozole 1152–1153 Leucovorin 1324–1325, 1325 Leukemias 1030, 1038–1041 classification 1027, 1038 clinical presentation 1039–1041 central nervous system (CNS) 1040 leptomeningeal (LM) 1039–1040 peripheral nervous system (PNS) 1041 spinal cord 1040 vascular/hematologic 1040–1041 hematopoietic stem cells 1027, 1028 treatment 1134–1137 Leukoencephalopathy progressive see Progressive multifocal leukoencephalopathy (PML) entries renal transplantation 1252 see also Posterior reversible encephalopathy (leukoencephalopathy) syndrome (PRES/PRLS) Leuprolide 1655 Levamisole 1433, 1653

Levetiracetam 677, 846–847, 1153, 1274, 1640, 1640 elimination 418, 423–424, 425, 427 Levodopa 1641, 1641 Levofloxacin 1491, 1492 Levosulpiride 638 Levothyroxine sodium 812, 813 Lgi1-Abs (antibodies) 1161 limbic encephalitis (LE) 1167 Lhermitte’s sign 559, 1090–1091, 1190–1191, 1200, 1542 Libman–Sacks endocarditis 70 Lidocaine 156, 780 adverse effects 132, 134–135, 1637 Lidoflazine 31 ‘Life settings’, sudden cardiac death (SCD) 19 ‘Lifting the Burden’ (WHO campaign) 1776 Lightning see Thermal injuries Lille scoring system 1078 Limb symptoms 1356–1357 mononeuropathy 775 Limbic encephalitis (LE) 1160, 1160, 1165–1169, 1166 antibodies 1161, 1162 lung cancer 347, 348–349, 350, 354–355 neuronal cell surface antigen antibodies (NSA-Abs) 1165–1166 AMPAr-Abs 1168 GABABr-Abs 1166, 1168 neurophil-Abs 1167 NMDAr-Abs 1167–1168 VGKC-Abs/Lgi1-Abs/CASPR2-Abs 1166, 1167 onconeuronal antibodies (ON-Abs) 1168–1169 Amphiphysin-Abs 1166, 1169 CV2/CRMP5-Abs 1160, 1166, 1169 Hu-Abs 1166, 1168 Ma2-Abs 1166, 1169 seronegative 1169 subacute cerebellar degeneration (SCD) 1164 treatment 1173 Limited cutaneous scleroderma (ISSc) 468 Lincosamides 1646 Line Tracing Test (LTT) 663–664, 664 Linear accelerators (Linac) 1152, 1183, 1183 Linguatula serrata 1436 Liothyronine sodium 812, 813 Lipids 621–622 Lipirudin 1132 a-Lipoic acid 1090 Lipopeptides 1646 Liraglutide 817, 820 Lispro 815 Listeria 1252 Listeria meningitis 1247 Listeria monocytogenes 1232, 1241, 1361–1362, 1369 Lithium 388 Live attenuated influenza vaccine (LAIV) 1550, 1552

I25 Live attenuated monovalent vaccine (LAMV) 1553 Livedo racemosa 1566–1567 Liver disorders see Acute liver failure (ALF) Liver transplantation 1257–1266 abnormal conscious state 1259–1261, 1260 brain edema 1261–1262, 1262 central nervous system (CNS) infection 1262–1263 malignant tumors 1263 historical perspective 1257–1258, 1258, 1259 immunosuppressants 677–678 neurologic features 1258–1259, 1259 neuromuscular complications 1263–1264 seizures 1261 Liver-intestine transplantation 1308–1309 Living donor liver transplantation (LDLT) 655 L-methionine 923 Locked-in syndrome 245–246 Loeys–Dietz syndrome (LDS) 571 description 571 neurologic complications 571 Lomustine (CCNU) 1184–1185, 1208–1209 Long bones, Paget’s disease of bone (PDB) 531 Lorazepam 1517 Lorenzo’s oil 762 L-ornithine-L-aspartate (LOLA) 670 Losartan 1636 Lovastatin 762, 1638 Low back pain, pregnancy 1610 Low density lipoproteins (LDLs) 623–624, 749–750 Low molecular weight heparin (LMWH) 297, 298, 1605–1606 adverse effects 137, 137, 1638 hematologic disorders 1130, 1131, 1132 venous thromboembolism (VTE) 295–297, 298–300 Lower airway obstruction 265–266 Lower extremity mononeuropathy/ radiculopathy 1609 Lower jaw protrusion prosthesis 262 Lower limb neuropathy, asymmetric 776 Low-grade gliomas, radiotherapy (RT) 1184 Loxosceles spp 994–995 Lubag’s disease 1780 Lumbar puncture 81 complications 1747 meningitis 1365, 1366, 1367 repeat 1373 Lumbar spine stenosis, surgery 546 Lumbosacral plexopathy 592, 776 Lund concept 1371, 1753 Lung Allocation Scoring (LAS) system (UNOS) 1238 Lung cancer 335–362 brain metastases 1143–1144 metastases 335–346 paraneoplastic disorders 346–355

I26 Lung cancer (Continued ) autoimmunity 346–348, 347 clinical syndromes 347, 348–354, 350 overview 346, 346 treatment/outcomes 354–355 Lung transplantation 1237–1243 allograft procurement 1238 background 1237 candidate evaluation 1238 heart-lung transplantation 1313 neurologic complications 1239–1241 absent cough reflex 1240 critical illness myopathy/neuropathy 1240 encephalopathy 1241 gastric outlet obstruction (GOO) 1241 metabolic deficits 1241 neuromuscular deficits 1240 perioperative 1239 peroneal nerve injury 1240 postoperative 1240 recurrent laryngeal nerve injury 1240 sedation/ventilator weaning 1240 neuropsychological effects 1241 outcomes 1238–1239, 1239 procedure 1238 Luteinizing hormone (LH) 686 acromegaly 691–692 excess 693–694 hyperprolactinemia 690–691 secondary hypogonadism 689–690 Lyme disease (borreliosis) 1473–1483, 1532–1533, 1533, 1540 clinical findings 1473–1476, 1474, 1475, 1576, 1576 neurocutaneous 1562, 1576–1577 diagnosis 1576–1577 differential diagnosis 1479 laboratory investigations 1476–1478, 1477 management 1479–1480, 1480 neuroimaging 1478 pathology/pathogenesis 1478–1479 treatment 1577 Lymphatic filarioses 1429 Lymphocyte predominant (LP) cells 1034 Lymphocytic leukemia 1038 Lymphocytic meningitis 1533 Lymphomas 1027–1038, 1030 B cell 1263 classification systems 1027, 1029 clinical presentation 1035–1038 central nervous system (CNS) 1035 infectious disease 1037 leptomeningeal (LM) 1036–1037 paraneoplastic 1038 peripheral nervous system (PNS) 1037 spinal cord 1035–1036 vascular/hematologic 1037–1038 hematopoietic stem cells 1027, 1028 Hodgkin (HL) 1034–1035 non-Hodgkin’s 1028–1034, 1029 renal transplantation 1250, 1252 treatment 1134–1137 Lymphomatosis cerebri 1030

INDEX Lymphoplasmacytic lymphoma see Waldenstr€ om’s macroglobulinemia (WM) Lymphoproliferative disorders 1282 Lysine analogs 1128 Lyssavirus 1501

M Ma2-Abs (antibodies), limbic encephalitis (LE) 1166, 1169 McArdle’s disease 833 McConnell’s sign 292 Machado Joseph’s spinocerebellar ataxia 794 Macrocephaly 552–554, 553, 554 Macrolides 1646 Magnesium 31, 737–738, 846–847, 872–873, 1286 Magnesium sulfate 1508–1509, 1697, 1737–1738 Magnetic resonance angiography 1692 Magnetic resonance imaging (MRI) brain metastases 1145, 1146–1149 cardiac arrest 34 complications 1746–1747 coronary artery stents (CAS) 99, 99 diffusion-weighted (DW-MRI) 1117, 1147, 1147 functional (fMRI) 101 hemolytic uremic syndrome (HUS) 1116, 1117 myocardial infarction (MI) 97, 98, 99 perfusion 1147 reversible cerebral vasoconstriction syndrome (RCVS) 1731–1732, 1734, 1735 spectroscopy 1147, 1148 spinal stenosis 545, 545 Maintenance of Wakefulness Test (MWT) 261 Malabsorption syndromes 621–632 causes/symptoms 622–623 neurologic dysfunction 623–628 nutrient absorption 621–622 Maladaptive behavior, fibromyalgia 517 Malaria 1514–1517 developing world 1777 diagnosis 1515, 1515 future research 1517 hemolytic uremic syndrome (HUS) 1114, 1114 management 1517 neuroimaging 1515 neurologic manifestations 1514–1515 pathogenesis 1516, 1516 vaccine 1517 Malayan pit viper (Calloselasma rhodostoma) 992–993 Malignant atrophic papulosis 1562, 1567 Malignant endocarditis, Gulstonian lectures (1885) 75–76 Malignant hyperthermia 949–951, 1612 management 951 Mambas (Dendroaspis) 990, 992 Manganese, excessive exposure 852

Mannitol 1262, 1628–1629, 1757–1759 Marantic endocarditis 69–70 Marfan syndrome (MFS) 569–570 description 569 diagnostic criteria 569 ischemic stroke 574 neurologic complications 569–570 Maroteaux–Lamy syndrome (MPS type VI) 444–445, 573 Marshall criteria, traumatic brain injury (TBI) 1756, 1757 Mass lesions fungal infection 1385 HIV 1322 Maxillomandibular advancement (MMA) 262 Maze procedure 143 Mean arterial pressure (MAP) 31, 645, 651 Mean corpuscular volume (MCV) 1006 Measles encephalomyelitis 1348 Measles virus 1345–1349 clinical manifestations 1346, 1346 diagnosis 1347, 1347, 1348 epidemiology 1345 measles-mumps-rubella (MMR) vaccine 1349, 1350, 1550–1551, 1550, 1651 treatment/prognosis/prevention 1348–1349 Meat intoxication syndrome 661 Mebendazole 1426, 1433 Mecasermin 810, 812 Mechanical circulatory devices 196, 196, 201 Mechanical heart valves 65 Mechanical ventilation 279 chronic airflow obstruction 279–280 complications 280–281, 280, 281 home (HMV) 285 prolonged (PMV) 283 weaning from 281–283, 281, 282 modes/protocol-driven 282–283 parameters 282 Mechlorethamine 1209 Medical Outcomes Study Short-Form Health Survey (SF-36) 507, 514, 536 Medical Research Council (UK) 1487–1488 Medicare 94, 104–105, 122 Medication overuse headache (MOH) 582 Medline 117, 118 Medtronic GEM III AT 141 Medullary carcinoma of the thyroid (MCT) 805, 806 Mefloquine 1517 Megaloblastic anemias 927, 929–930, 930, 1009, 1125–1126 Megestrol 1152–1153 Meglitinides 820 Melano brain metastasis 1580–1581, 1580 Melanoma 1144, 1146 Melarsoprol 1412 MELAS (mitochondrial encephalomyopathy, lactic acidosis and stroke-like episodes) syndrome 130, 410–411

INDEX Memantine 1642 MEN1 gene 804 Meningeal disease 232–233, 308, 308, 309 Meningiomas, radiotherapy (RT) 1187–1188 Meningitis 1577–1578 aseptic 322, 579–580, 579 bacterial see Bacterial meningitis belt 1361–1362 cryptococcal 1248–1249 HIV 1322 leukemic 1039–1040 Listeria 1247 multidrug-resistant (MDR) 1491, 1492 neonatal 1369 nosocomial 1362, 1369 oesinophilic 1537–1538, 1538 physician-associated 1362 purulent 1411 shunt-associated 1369 spontaneous 1369 tuberculous see Tuberculous meningitis (TBM) Meningococcal vaccine 1550, 1554–1555, 1651 Meningomyelitis 1534 syphilitic 1464–1465, 1534 Meningoradiculopathy 1533, 1533 Meningovascular syphilis 1464, 1533 Menkes disease (MD) (kinky hair disease) 626, 851, 853, 853 Mental disturbances Henoch–Sch€onlein purpura (HSP) 1105 hyperthyroidism 722–723 hypothyroidism 706–707 see also Psychiatric manifestations Mercury, excessive exposure 852 Meropenem 1369 Mesobuthus tamulus 988, 993–994 Metabolic disorders acid-base 375 acidosis 375–376 bariatric surgery 591–592 bone disease 443–445 encephalopathy 1278, 1297–1299 Henoch–Sch€onlein purpura (HSP) 1105 lung transplantation 1241 multiple myeloma (MM) 1091–1092 posterior reversible encephalopathy syndrome (PRES) 1694 renal transplantation 1251–1252 transient loss of consciousness (TLOC) 184 Metabolism drugs 1654–1655 Metachronous presentation 1144 Metastatic disease 213–214 brain see Brain metastases lung cancer 335–346 spinal see Spinal epidural metastases Metazoa (opisthokonta) 1404–1414, 1436 Metformin 1541, 1746 diabetes mellitus (DM) 816, 817–818, 817, 820 Metformin-associated lactic acidosis 1746 Methadone 1649

Methanol 375–376, 1090 Methimazole 813–814, 1655 Methohexital 1624 Methotrexate (MTX) 459, 507, 1613, 1718–1719 adverse effects 1201, 1203, 1205–1206 acute/subacute 1205–1206 chronic 1206 cerebral vasculitis 479, 486 giant cell arteritis (GCA) 229, 481 leukemia/lymphomas 1135 sarcoidosis 324–326, 325, 327 Methyl alcohol intoxication 1543 Methylcobalamin 779 Methyldopa 1636 Methylenetetrahydrofolate reductase (MTHFR) 598, 929, 932, 935 5,10-Methylene-THF 928 L-Methylfolate 779 Methylmalonic acid (MMA) 917, 919–921, 923 Methylmalonic aciduria 1541 Methylphenidate 1644, 1645 Methylprednisolone 323, 325, 506, 1608, 1718, 1719 5-Methyltetrahydrofolate (5-MTHF) 779, 928–929 Methylthiouracil 813 Metoclopramide 634–636, 639, 782 adverse effects 388, 1642, 1656 Metoprolol 132, 135 Metriphonate 1546 Metrix system 141 Metronidazole 88, 601, 1212, 1508, 1646 Metyrapone 698, 754, 759 Metyrosine 766 Mexiletine 132, 135, 1637 Microadenomas 1186–1187 Microalbuminuria 407 Microangiopathic hemolytic anemia 1113 Microcephalophis gracilis gracilis 992 Microscopic polyangiitis 486 clinical features 486 Microsomal triglyceride transfer protein (MTP) gene 623–624 Microtubule-stabilizing agents 1210–1211 Micrurus spp 992 Micrurus nigrocinctus 992 Midazolam 1260–1261, 1504, 1625, 1625, 1631, 1758 Mid-borderline leprosy 1573–1575 Midodrine 185, 187, 781 Miglitol 817, 818, 819 Migraine 7–8, 964, 1736–1737, 1776 Migraine Intervention with STARflex Technology (MIST-1) study 7–8 Milnacipran 520–521 Milrinone 1738 Mineral metabolism disorders 737–748 see also specific disorders Mineralizing microangiopathy 1041 Mini Mental State Examination (MMSE) 130–131, 707, 1191–1192 Minimal hepatic encephalopathy (mHE) 661–662, 663–665, 664, 666

I27 Minocycline 1512–1513 Misonidazole 1204, 1205, 1212 Misoprostol 1649 Mitiglinide 820 Mitochondrial disorders 756, 794, 796 cytopathies 12–13 pregnancy 1612 Mitomycin C 1201, 1211 Mitotane 698 Mitotic spindle inhibitors 1201–1203, 1210–1211 Mitoxantrone 1212, 1607–1608 Mitral annulus calcification (MAC) 63 Mitral bioprosthetic heart valves 64–65 Mitral stenosis 62 Mitral valve prolapse (MVP) 63 Mixed connective tissue disease (MCTD) 442, 468 Mixed thrombophilia 1063–1064, 1065, 1067 MMR (measles-mumps-rubella) vaccine 1349, 1350, 1550–1551, 1550, 1651 Modafinil 1644, 1645 Model for end stage liver disease (MELD) 655 Model Spinal Cord Injury Systems 1761 Modified neurosyphilis 1465 Molecular adsorbent recycling system (MARS) 655 Molecular targeted agents 339–340, 1152–1153 Molluscum contagiosum 1328–1329 Molybdenum cofactor deficiency 828, 833 Monoamine oxidase (MAO) A inhibitors 1642 Monoamine oxidase (MAO) B inhibitors 1641, 1641 Monobactams 1646 Monoclonal antibodies 1136 adverse effects 1205, 1213, 1653 Monoclonal gammopathy of undetermined significance (MGUS) 1031–1032, 1031, 1083–1088, 1086, 1087 central nervous system (CNS) 1088 clinical presentation 1037 peripheral neuropathy 1084, 1085–1088 Monogenea 1414 Mononegavirales order 1501 Mononeuritis multiplex (MM) HIV-infected patients 1333–1334, 1335 leprosy 1510 Mononeuropathy dialysis patients 401 HIV-infected patients 1333–1334, 1335 infective endocarditis 79 limb 775 lower extremity 1609 upper extremity 1608–1609 renal disease 390 trunk 775–776 ‘Montauk knee’ 1473 Montreal Cognitive Assessment test (MoCA) 1191–1192, 1339–1340 Mood disorder 465, 706–707

I28 Morphine 1626 Morquio–Brailsford syndrome (type V) 444–445 Mosapride 638 Motor function, impaired end of life (EOL) phase 1220 treatment 1221 Motor neuron disease 352 Mouth breathing, children 254 Movement Disorder Society 1775, 1780 Movement disorders 56 cardiology 8–9 developing world 1779–1780 drug-induced 635, 638 hyperthyroidism 715–716 Moxifloxacin 1491, 1492 Moyamoya 1022 MPL gene 1076, 1077–1078 Mucocutaneous telangiectasias 1571–1572, 1571 Mucopolysaccharidoses (MPS) 444–445, 573–574 description 573 neurologic complications 573–574 types 573 Mucor 1383, 1396 Multicenter AIDS Cohort study 1339 Multicenter Study of Hydroxyurea (MSH) 1021 Multidisciplinary approach 518, 552 Multidrug-resistant (MDR) meningitis 1491, 1492 Multifocal encephalopathy 348 Multifocal motor neuropathy (MMN), pregnancy 1611–1612 Multiple acyl-CoA dehydrogenation deficiency (MADD) 901–902 Multiple endocrine neoplasia (MEN) 799, 803–806, 803 Multiple endocrine neoplasia (MEN), type 1 (Wermer’s syndrome) 685, 803, 804–805 clinical findings 804 historical perspective 804 laboratory investigations 804–805 management 805 natural history 804 neuroimaging 805 neuropathology 805 Multiple endocrine neoplasia (MEN), type 2 803, 804 Multiple endocrine neoplasia (MEN), type 2A (Sipple syndrome) 765, 803, 805–806 historical perspective 805 laboratory investigations 805 management 806 natural history 805 neuroimaging 805–806 Multiple endocrine neoplasia (MEN), type 2B 765, 803, 804, 806 historical perspective 806 laboratory investigations 806 management 806 natural history 806

INDEX Multiple endocrine neoplasia (MEN), type 2B (Continued ) neuroimaging 806 neuropathology 806 Multiple endocrine syndromes (MES) 799–808 Multiple myeloma (MM) 1031–1034, 1031 central nervous system (CNS) 1090–1092 classification 1089 clinical presentation 1037 peripheral nervous system (PNS) 1089 plasma cells 1083, 1084, 1085, 1086, 1087, 1088–1092 smoldering 1031–1032, 1083, 1086, 1088 staging system 1088 treatment 1134–1137 -related neuropathy 1089–1090 Multiple organ transplantation 1305–1317 heart-lung 1313 hematopoietic stem cell (HSCT), combined 1314 kidney-pancreas 1313–1314 neurologic complications 1309–1313, 1309 alterations of consciousness/ behavior 1310 cerebrovascular complications 1311 epilepsy 1310 headache 1312 infection 1311–1312 neuromuscular 1312–1313 see also Multivisceral/intestinal (MVI) transplantation Multiple Sclerosis International Federation (MSIF) 1779 Multiple sclerosis (MS) ankylosing spondylitis (AS) 457 brain metastases 1150 decompression illness (DCI) 964 developing world 1779 nephrotic syndrome 410–411 vs neuro-Behc¸et syndrome (NBS) 1716 pregnancy see under Pregnancy respiratory failure (RF) 277–278 thyroid disease 726 tropics 1540 vitamin D 882 Multiple sleep latency test (MLST) 180–181, 260–261, 398 Multisystem atrophy (MSA) 8–9, 246–247 Multivisceral/intestinal (MVI) transplantation 1305–1309, 1306, 1308, 1308 isolated intestine 1308 liver-intestine 1308–1309 pediatric 1309 Mumps virus 1345–1349 clinical manifestations 1346–1347 diagnosis 1348 epidemiology 1345–1346 measles-mumps-rubella (MMR) vaccine 1349, 1350, 1550–1551, 1550, 1651

Mumps virus (Continued ) treatment/prognosis/prevention 1349 Muromonab (OKT3) 1246, 1268, 1269–1270, 1274, 1278–1279 Murray Valley encephalitis virus 1378 Muscle biopsy 1679–1680 Muscle disorders 10–12, 11 Ehlers–Danlos syndromes (EDS) 570–571 primary hyperparathyroidism (PHPT) 739–741 Sj€ ogren’s syndrome (SS) 471 weakness 184, 1252, 1357 see also Intensive care unit-acquired weakness (ICUAW), generalized Muscle relaxants 1626–1628 Musculoskeletal involvement Behc¸et’s syndrome 1705 gastric bypass 592 scleroderma (systemic sclerosis) (SSc) 468 Myasthenia Gravis Foundation of America 279 Myasthenia gravis (MG) 10, 410 hypothyroidism 711 intensive care unit (ICU) 1676 neuroanesthesia 1628 pregnancy 1610–1611 thyroid disease 726 Mycobacterium africanum 1485 Mycobacterium avium 755, 1485 Mycobacterium bovis 1485 Mycobacterium leprae see Leprosy Mycobacterium tuberculosis 318, 1232, 1247–1248, 1273, 1532 see also Tuberculosis (TB), central nervous system (CNS) Mycophenolate mofetil (MMF) 459, 507, 1613 adverse effects 676, 678 cerebral vasculitis 479, 485–486 sarcoidosis 324, 325, 326–327 transplantation 1231, 1260, 1280 Mycoplasma spp 80–81 Mycoplasma pneumoniae 1530 Mycoses Study Group (USA) 1394 Mycotic aneurysms 68, 78–79, 79 treatment 87–88 Myelitis acute transverse 1540 with oesinophilic meningitis 1537–1538, 1538 tuberculous 1495 varicella zoster 1577–1578 Myelodysplastic syndrome (MDS) 1009, 1038–1039 treatment 1133–1134 Myelography complications 1747 computed tomography (CT) 544–545 spinal stenosis 544 Myeloma see Multiple myeloma (MM) Myelomatosis 446, 446 Myeloneuropathy see Tropical myeloneuropathy

INDEX Myelopathy 351–352 achondroplasia 557 gluten-related diseases (GRD) 614, 614 Henoch–Sch€onlein purpura (HSP) 1105 HIV see under HIV-infected patients necrotizing 1040 neurosarcoidosis 312, 312 Paget’s disease of bone (PDB) 535 spinal stenosis 543 tropical see Tropical myelopathy vacuolar (VM) see under HIV-infected patients Myeloproliferative neoplasm (MPN) see Chronic myeloproliferative diseases Mygalomorphae 994 MYH9 gene 1057 Myiasis 1436 Myocardial infarction (MI) 111, 115 see also Acute myocardial infarction (AMI) Myocardial injury 5, 6–7 Myoclonic ataxia 612–613 Myoclonic encephalopathy 410–411 Myodenylate deaminase deficiency 828, 833 Myoglobinuria, thermal injuries 982–983, 984 Myopathy 354 critical illness see Critical illness myopathy (CIM) dialysis patients 401 gluten-related diseases (GRD) 613–614, 614 hereditary 1612 idiopathic see Idiopathic inflammatory myopathy (IIM) inflammatory 1612 intestinal transplantation (ITx) 1273, 1273 sarcoidosis 313, 323 Myositis disease activity (MYOACT) 507 damage (MYODAM) 507 Myotonic (muscular) dystrophy (MMD) 11, 12–13, 794, 795–796 historical perspective 795, 796 laboratory investigations 796 natural history 796 neuroimaging 790, 791–792 treatment 796 type 1 12, 1612 Myxedema coma 705–706 clinical features 705 laboratory investigations 705–706 management 706 Myxedematous madness 706 Myxoma 200–201, 218 see also Atrial myxoma Myxosarcoma 216

N Nabilone 522 Naegleria spp 1411 Naegleria fowleri 1411 Naja spp 989, 991

Naja haje 987, 989 Naja kaouthia 989, 991 Naja mandalayensis 991 Naja melanoleuca 991 Naja mossambica 991 Naja naja 989, 991 Naja nigricollis 989, 991 Naja oxiana 991 Naja philippinensis 991 Naja sagittifera 991 Naja siamensis 991 Naja sputatrix 991 Naja sumatrana 991 Naloxone 1649 Naproxen 580, 1649 Narcolepsy 257 Natalizumab 507, 601, 1607–1608 Nateglinide 817, 820 NATHAN 1 (Neurological Assessment of Thioctic Acid In Diabetic Neuropathy) trial 779 National Arthritis Data Workgroup 514 National CPR (NCPR) registry 26–27 National Heart, Lung and Blood Institute (NHLBI) 1018, 1021–1022 National Hospital Discharge Survey 1229 National Institute for Health and Clinical Excellence (NICE) 83 National Institutes of Health (NIH) 868, 869–870, 1015, 1324–1325, 1480 Alzheimer’s Disease AntiInflammatory Prevention Trial (ADEPT) 580 Stroke Scale (NIHSS) 66, 156, 296–297, 962 National Multiple Sclerosis Society 1607–1608 National Registry of Atrial Fibrillation 122 National Registry of Cardiopulmonary Resuscitation (NRCPR) 27 National Registry of Myocardial Infarction 105 National Spinal Cord Injury Database 1761 Nationwide Inpatient Sample of Health Care Cost and Utilization Project (USA) 1603 Necator americanus 1432 Necrotizing autoimmune myopathy (NAM) see Idiopathic inflammatory myopathy (IIM) Necrotizing vasculitis 481–482 Needle damage 1681 Nefazodone 1642 Neisseria gonorrhea 1461 Neisseria meningitidis 1367, 1369, 1777 bacterial meningitis 1361–1362, 1369 Nelarabine 1204, 1208 Nelson’s syndrome 698 Nematodes (roundworms) 1429–1436 Neonatal meningitis 1369 Neonatal myasthenia (NMG) 1610 Neoplastic diseases, renal transplantation 1250–1251 Nephrogenic systemic fibrosis (NSF) 97–99, 1746–1747

I29 Nephrotic syndrome 405–416 causes 405, 406 infection 407 neurologic manifestations 405, 406, 407–411, 407 diagnosis/therapy/prognosis 411 pathophysiology 405–407, 406 Nerve entrapment 707, 775 rheumatoid arthritis (RA) 450–451, 450 Nerves biopsy 1679–1680 deafness 1543 repair 778 root compression 555, 559–560 stimulation 1678–1679 Neural tube defects (NTDs) 932, 934–935 Neurally-mediated (reflex) syncope (NMS) 174–176, 179 clinical features 174 etiology 175–176, 175, 176 pathophysiology 174–175 treatment 187 Neurasthenia 513 Neuroanesthesia 1623–1633 cerebral effects 1625, 1627 disease states 1628–1629 fundamentals 1624 historical perspective 1623–1624 induction agents 1624–1625, 1625 inhalation agents 1625–1626, 1627 neurophysiologic monitoring 1630–1632, 1631 neuroprotection 1630 opioids/muscle relaxants 1626–1628 peripheral nervous system (PNS)/ positioning injuries 1629–1630 postoperative cognitive dysfunction (POCD) 1630 Neuro-Behc¸et syndrome (NBS) 1704, 1707–1712, 1707 arterial 1710 cognitive changes 1710 diagnostic criteria 1708 diagnostic studies 1712–1715 cerebrospinal fluid (CSF) 1715 neuroimaging 1712–1715, 1713, 1714 SPECT studies 1715 differential diagnosis 1715–1717 headache 1710–1711, 1710 intra-axial 1709–1710, 1715–1717, 1716 extra-axial 1708–1709, 1716 neuro-psycho-Behc¸et syndrome 1710 peripheral nervous system (PNS) 1711, 1712 prognosis 1718 secondary neurologic involvement 1712 subclinical 1711–1712 treatment 1718–1720 intra-axial 1718–1719 extra-axial 1719–1720 Neuroborreliosis 1474, 1533, 1533 Neurocognition Frascati classification 1338–1339, 1339 see also under HIV-infected patients vitamin D 881

I30 Neurocutaneous disorders 1559–1594, 1562 brain/peripheral nerve tumors 1580–1584 cutaneous angioma 1569–1573 epilepsy 1584–1590 peripheral nervous system (PNS) 1573–1580 stroke 1561–1569 vascular 1569 Neurocysticercosis (NCC) 1149, 1422–1425, 1445–1459, 1536–1537 clinical features 1422–1423, 1448–1450, 1449, 1537 control measures 1456 diagnosis 1423, 1423, 1424, 1450–1454 criteria 1454, 1454 immunologic 1453–1454 epidemiology 1445–1446, 1537 etiopathogenesis 1446–1448, 1447 cysticerci 1447–1448, 1448 Taenia solium, life cycle 1446–1447, 1446 historical perspective 1445 imaging 1450–1453, 1537, 1538 laboratory findings 1450, 1537 pathogen 1422 pathogenesis 1537 prevention 1424 treatment 1424, 1454–1456, 1537 cysticidal drugs 1455 surgery 1456 symptomatic 1455–1456 Neurodegeneration with brain iron accumulation see Inherited neurodegeneration with brain iron accumulation (NBIA) Neurodegenerative diseases, cardiology 8–9 Neurodermatology see Neurocutaneous disorders Neuroendocrine disorders 184 Neuroferritinopathy 860 Neurofibromatosis type 1 (NF1) (von Recklinghausen’s neurofibromatosis) 765, 1562, 1581–1582 clinical symptoms 1581–1582 cutaneous 1581–1582, 1581 ophthalmologic 1581–1582 orthopedic 1581–1582 diagnosis 1582 Neurofibromatosis type 2 (NF2) 1562, 1582–1584 clinical symptoms 1582–1583 cutaneous 1582–1583 neurologic tumors 1582–1583, 1583 ocular 1582–1583 diagnosis 1583 radiology 1583 treatment 1584 Neurofilarioses 1429 clinical manifestations 1429 diagnosis 1429 epidemiology 1429

INDEX Neurofilarioses (Continued ) pathogen 1429 therapy 1429 Neurogenic cardiac events, management 7 Neurogenic claudication 543 Neurogenic compression theory 543 Neurogenic pulmonary edema (NPE) 248 Neurogenic stunned cardiomyopathy 6, 6, 9–10 Neuroimaging, complications 1743–1750 catheter angiography 1743–1745, 1744 computed tomography (CT) 1745–1746 magnetic resonance imaging (MRI) 1746–1747 myelography/lumbar puncture 1747 vertebroplasty 1747–1748, 1748 Neurokinin receptor antagonists 636–637 Neuroleptic malignant syndrome (NMS) 639, 949–951, 950 hypothyroidism 712 management 951 Neurological Disorders: Public Health Challenges (WHO) 1773 Neurolymphomatosis 1037 Neuromas, radiotherapy (RT) 1188 Neuromuscular blocking agents (NMBA) 30–31, 1676 Neuromuscular disorders (NMD) Ar-Abs (antibodies) 1161–1162, 1161 limbic encephalitis (LE) 1167–1168 cardiology 9–13 Henoch–Sch€ onlein purpura (HSP) 1105–1106 HIV-infected patients 1332–1334 intestinal transplantation (ITx) 1273, 1273, 1275 lung transplantation 1240 multiple organ transplantation 1312–1313 pancreas transplantation 1282 pregnancy 1608–1613 primary hyperparathyroidism (PHPT) 867–869 respiratory failure (RF), acute/chronic 273–275, 284–285 sleep-related hypoventilation 265, 266 Neuromuscular junction (NMJ) block 1676, 1677 Neuromuscular junction (NMJ) disorders 10, 248 Neuromyelitis optica (NMO) 1779 Neuromyopathy (CINM), critical illness 1675 Neuromyotonia 352 Neuronal cell surface antigen antibodies (NSA-Abs) limbic encephalitis (LE) 1165–1166 paraneoplastic neurologic syndromes (PNS) 1161–1162 Neuronal degeneration 581 Neuron-specific enolase (NSE) 33–34 Neuro-onchocercosis 1429–1430 Neuro-ophthalmologic problems Henoch–Sch€onlein purpura (HSP) 1105 obstructive sleep apnea syndrome (OSAS) 259

Neuropathy Impairment Score (NIS) 779 Lower Limbs (NIS-LL) 779 Neurophil-Abs (antibodies), limbic encephalitis (LE) 1167 Neuroprotective pharmacology 31 Neuropsychiatric manifestations carbon monoxide (CO) intoxication 974, 975 folic acid deficiency 930–931 primary hyperparathyroidism (PHPT) 867–869 Neuropsychiatric systemic lupus erythematosus (NPSLE) 463, 464–466, 465 cerebrovascular disease 465 cognitive dysfunction 464–465 demyelinating lesions 466 diagnosis 466–467 headache 465 incidence 464 management 467–468 pathogenesis 466 psychosis/mood changes/anxiety 465 seizures 465 Neuro-psycho-Behc¸et syndrome 1710 Neuropsychological effects, lung transplantation 1241 Neurosarcoidosis 305 clinical findings 306–308, 307 definitions 313–314 differential diagnosis 314–316, 315 treatment 325 ‘Neuro-Sweet disease’ (NSD) 1717 Neurosyphilis 232–233, 1461–1472 clinical manifestations, syphilis 1464 diagnosis 1466–1468, 1467 epidemiology 1462 historical perspective/nomenclature 1461–1462 HIV see under HIV-infected patients pathogenesis 1462–1464 treatment 1468–1470 types 1464–1466, 1465 Neurotraumatology 1751–1772 brain injury see Traumatic brain injury (TBI) historical perspective 1743–1745 spinal cord see Spinal cord injury (SCI) Neurotrophic factors/nerve repair 778 Neutral protamine Hagedorn (NPH) 814, 815 New England Journal of Medicine 28, 29–30 New Orleans criteria, minor head trauma 1756 New York Heart Association (NYHA) 62, 819 Niacin see Vitamin B3 deficiency Nicardipine 165, 165, 1697 Nicotinamide adenine dinucleotide phosphate (NADPH) 1012 Nicotinic acid see Vitamin B3 deficiency Nifedipine 1636 Nifurtimox 1412, 1414 Nilotinib (Tasigna®) 1133

INDEX Nimodipine 31, 1737–1738 Nimustine (ACNU) 1208–1209 NINDS (National Institute of Neurological Disorders and Stroke) intravenous thrombolysis trial 203 Nissi–Alzheimer arteritis 1463 Nitrates 1637 Nitroglycerine 165 Nitrosoureas 1201, 1208–1209 Nitrous oxide (N2O) (‘laughing gas’) 918, 937, 1541, 1626, 1631 effects 1627, 1630–1631 Nizatidine 638–639 Nocardia 1232, 1247, 1252, 1278–1279 Nocardia asteroides 1037, 1532 Nocturia 254 Nocturnal sweats 254 Nonanion gap metabolic acidosis 375, 376 Nonarteritic optic neuropathy (NAON) 259 Nonbacterial thrombotic endocarditis (NBTE) 69 Nonclassic paraneoplastic neuropathies 1170–1171 Nonclassic rabies 1502 Nongerminomatous tumors 1185 Non-Hodgkin’s lymphomas 1028–1034, 1029 Noninsulin blood glucose lowering drugs 816–821, 817 Noninvasive ventilation (NIV) 275–276, 277, 281, 284–285 Non-nucleoside reverse-transcriptase inhibitors (NNRTIs) 1324–1325 Nonretroviral agents 1648 Nonsecretory myeloma 1032, 1083, 1092 Non-selective COX inhibitors 1647–1649 Non-small-cell lung cancer (NSCLC) brain metastases 1143–1144, 1151, 1152 dipeptidyl peptidase (DPP-4) inhibitors 819–820 see also Lung cancer Nonspecific or overlap myositis see Idiopathic inflammatory myopathy (IIM) Non-ST elevation MI (NSTEMI) 93, 101–103 Nonsteroidal anti-inflammatory drugs (NSAIDs) 577–584 adverse effects 582, 583, 1647–1649 arachidonic acid pathway 577, 578 aseptic meningitis 579–580, 579 diabetic neuropathy 782 enzyme cyclooxygenase (COX) 577–578, 579 fever 953 hypophosphatasia 879 medication overuse headache (MOH) 582 neuronal degeneration 581 overdose/intoxication 582–583 Reye’s syndrome 582 spinal stenosis 546 stroke, risks 580–581 Non-valvular atrial fibrillation (NVAF) 62

Noonan syndrome 53 Norepinephrine reuptake inhibitors (NRI) 1642 Norepinephrine-dopamine reuptake inhibitors (NDRI) 1642 Normovolemic hyponatremia 367–369, 367 North American Spine Society (NASS) 298 Norwegian Medical Birth Registry 1610 Nosocomial meningitis 1362 post-traumatic 1369 Notechis 992 NovoSeven® 1128 Nucleoside analogs 1133–1134 5’-Nucleotidase associated pervasive development disorder (NAPDD) 828, 832 Nucleus pulposus embolism 1525 clinical features 1525 differential diagnosis 1525 epidemiology 1525 imaging 1525, 1526 neuropathology 1525 pathogenesis 1525, 1526 treatment 1525 ‘Numb chin’ phenomenon 1039–1040 Number Connection Tests (NCT) 663–664, 664 Nutrient absorption 621–622 Nutritional disorders 1306 bariatric surgery 591–592 deficiency, bariatric surgery 588, 588 myeloneuropathy 1540–1544 Nutritional polyneuropathy (dry beriberi) 896–897

O Obesity 587 obstructive sleep apnea syndrome (OSAS) 253, 255, 256, 257 Obesity hypoventilation syndrome (OHS) 278 Obstructive sleep apnea (OSA)/syndrome (OSAS) 251–263 breathing control 251–252 clinical observation 260 clinical presentation 253–255 comorbidities 257–260 definitions 251 diagnostic criteria 261 gender differences 256 genetics 253 imaging 256 neuropsychiatric symptoms 255–256 pathophysiology 252, 253 prevalence 253 respiratory failure (RF) 278 risks/predisposing factors 257 severity, estimation 260 sleepiness, assessment 260–261 treatment 261–263 behavior management 263 oral appliances 262 pharmacologic 262

I31 Obstructive sleep apnea (OSA)/syndrome (OSAS) (Continued ) positive airway pressure (PAP) therapy 261–262 surgical procedures 262 Occipital horn syndrome (OHS) 853, 853 Octreotide 697, 810–811, 810 Ocular symptoms neurofibromatosis type 2 (NF2) 1582–1583 see also Ophthalmologic manifestations Oculomotor nerve (cranial nerve III) 307 Oesinophilic meningitis, with myelitis 1537–1538, 1538 Oestrus ovis (sheep botfly) 1436 Ofloxacin 1512–1513 Ohm’s Law 983 OKT3 see Muromonab-OKT3 Olanzapine 1642 ‘Old world vipers’ 992–993 Olfactory nerve (cranial nerve I) 307 Oligodendrogliomas, radiotherapy (RT) 1184–1185 Omalizumab 485–486 Omeprazole 638–639 On a form of chronic inflammation of the bones (osteitis deformans) (Paget) 529 Onchocerca volvulus 1429–1430 Onconeuronal antibodies (ON-Abs) limbic encephalitis (LE) 1168–1169 opsoclonus myoclonus (OM) 1170 paraneoplastic neurologic syndromes (PNS) 1159, 1162–1163 Ondansetron 636–637, 1656 Ondine’s curse see Congenital central hypoventilation syndrome (CCHS) ‘Onion bulb’ formation 1579 Ontario Prehospital Advanced Life Support Study 27–28 Open mouth ventilation 185 Ophiophagus hannah 991 Ophthalmologic manifestations Fabry disease (FD) 1564 incontinentia pigmenti (IP) 1587–1588 neurofibromatosis type 1 (NF1) 1581–1582 Sj€ ogren–Larsson syndrome (SLS) 1589 Sturge–Weber syndrome (SWS) 1571 see also Ocular symptoms Opiates 1649 Opioids 1260–1261, 1626–1628, 1631, 1649 diabetic neuropathy 780–781, 781 fibromyalgia 519, 521–522 Opisthokonta (metazoa) 1404–1414, 1436 Opisthotonos 377 Opsoclonus 1278–1279 Opsoclonus-myoclonus (OM) 351, 354, 1160, 1160, 1169–1170 onconeuronal antibodies (ON-Abs) 1170 Optic chiasm injury, radiotherapy (RT) 1192 Optic nerve (cranial nerve II) 307 decompression 1760 injury, radiotherapy (RT) 1192

I32 Optic neuritis 351 Optic neuropathy 1544 Oral aphthae, Behc¸et syndrome (BS) 1704 Oral appliances 262 Organization to Assess Strategies for Ischemic Syndromes (OASIS) program 93 Orienta 1403 Ornithoctoninae 994 Oropharyngeal secretions 283 Orotic acid phosphoribosyltransferase (OPRT) 827, 833, 834 Orotic aciduria 833–834, 834 Orotidine monophosphate decarboxylase (OPD) 827 Orthomyxoviridae 1345 Orthopedic manifestations, neurofibromatosis type 1 (NF1) 1581–1582 Orthoretrovirinae subfamily 1527 Orthostatic (postural) hypotension (OH) 171–174, 179 clinical features 171–172 etiology 172–174, 173, 174 pancreas transplantation 1282 pathophysiology 172 initial 172 delayed 172 treatment 186–187 Ortoidine-5’-monophosphate (OMP) 833, 834 Osler, William 75–76 Osler–Weber–Rendu syndrome see Hereditary hemorrhagic telangiectasia (HHT) Osmolytes 368 Osmoprotection 370–371 Osteitis deformans 529 Osteoarthritis 537 Osteoblastic phase, defined 533–535 Osteogenesis imperfecta (OI) 444, 573 description 573 neurologic complications 573 Osteolytic disorder 535 Osteolytic phase, defined 533–535 Osteomalacia 444 Osteoporosis 875–876 clinical manifestations 876 etiology 875 sarcoidosis 328 treatment 876 Osteosclerotic myeloma see POEMS syndrome Outcome Measures in Rheumatology Clinical Trials (OMERACT) 524 Ovaries/testis, disease of 787–798 clinical findings 788, 789, 790, 791 historical perspective 787–789, 788 laboratory investigations 791 management 792–793 natural history 789–791 neuroimaging 791–792 neurologic disorders 793–796, 794 pathology 792 OVID databases 118

INDEX Oxaliplatin 1134, 1200–1201, 1200 Oxamniquine, bilharziasis 1545 Oxazolidinone 1646 Oxcarbazepine 847, 1153, 1640, 1640 elimination 418, 422, 425, 427 Oxidative stress 777 Oxybarbiturates 1624 Oxygen parameters 274, 274 ‘Oxygen window’ 965 Oxyuranus spp 992

P Pacemaker neurons, breathing 243–244, 244 Pacemakers 140–141 complications 157 Paclitaxel 1135–1136, 1202, 1210 Paediatric Rheumatology European Society (PRES) 1102, 1102 Paediatric Rheumatology International Trials Organization (PRINTO) 1102, 1102 Paediatric Sleepiness Scale 261 Paget’s disease of bone (PDB) 529–540, 876–878 clinical characteristics 530–531, 534, 877–878 craniovertebral abnormalities 435, 443–444, 443 diagnosis 878 genetics 532–533 historical perspective 529 laboratory investigations 532 management 535–537 natural history 531–532 neuroimaging 532 pathology 533–535 sarcoma 531–532, 536 treatment 878 Pain-controlling agents 780–781 PAIR technique 1425–1426 Palla’s pit viper (Agkistrodon halys) 992–993 Palliative care see End of life (EOL) phase Palmar-plantar erythrodysesthesia (PPE) 1204 Palonosetron 636, 637 Pamidronate 536, 878 Pancreas transplantation 1279–1285 diabetes 1280 islet 1280 kidney-pancreas transplantation 1313–1314 neurologic benefit 1282–1285 autonomic neuropathy 1283–1285, 1284 peripheral neuropathy 1283 neurologic complications 1280–1281, 1281 central nervous system (CNS), infection 1281 first postoperative month 1278 immunosuppression 1281–1282 neuromuscular complications 1282 orthostatic hypotension 1282

Pancreas transplantation (Continued ) perioperative 1277–1278 post-transplant lymphoproliferative disorders (PTLD) 1282 six months after 1278–1279 period preceding 1277 Pancuronium 1509 Pantoprazole 638–639 Pantothenate kinase-associated neurodegeneration (PKAN) 859, 859, 860 Pantothenic acid see Vitamin B5 deficiency Papillary fibroelastoma 216 Paracetamol 1371 Paracoccidioides brasiliensis 1384 Paradoxical embolism 1068 Paragonimiasis 1421–1422 clinical manifestations 1421 diagnosis 1421–1422 epidemiology 1421 pathogen 1421 treatment 1422 Paragonimus 1421 Paragonimus westermani 1421 Paralysis see Thyrotoxic hypokalemic periodic paralysis (TPP) Paralytic (dumb) rabies 1502 Paralytic shellfish poisoning (PSP) 964, 996 Paramyxoviridae 1345 Paranasal sinus overpressurization 964 Paraneoplastic encephalitis 787 Paraneoplastic encephalomyelitis (PEM) 1160, 1160, 1165 Paraneoplastic neurologic syndromes (PNS) 346–355, 1159–1179 central nervous system (CNS), specific disorders 1160, 1163–1170 clinical management 1172–1173 treatment 1173 tumor identification 1172 diagnosis 1160, 1160, 1161 epidemiology 1159–1160 pathogenesis/antibodies 1160–1163, 1161 neuronal cell surface antigen antibodies (NSA-Abs) 1161–1162 onconeuronal antibodies (ON-Abs) 1159, 1162–1163 peripheral nervous system (PNS), specific disorders 1170–1172 Paraneoplastic neuro-ophthalmologic manifestations 789, 791 Paraneoplastic presentation, lymphomas 1038 Paraneoplastic progressive necrotizing myelopathy 1040 Paraneoplastic syndromes 787–789, 788, 791–792 Parasites central nervous system (CNS) 1248 see also Metazoa (opisthokonta); Protozoa Parasitic myelopathy 1535–1540 Parasympathetic outflow tracts 4 Parathyroid apoplexy 744

INDEX Parathyroid hormone (PTH) 813, 814, 872 analogs 813, 814 disorders 737–748, 813, 865 Parathyroidectomy (PTX) 868, 871 Parenchymal CNS disease 1492–1494 Parenchymal neurocysticercosis 1451, 1452, 1453 Parenchymatous neurosyphilis 1465 Parkinsonian disorders 8–9 Parkinsonism, metoclopramide-induced 636 Parkinson’s disease (PD) 636 breathing 246 cardiology 8–9 developing world 1779–1780 iron disorders 861 nonsteroidal anti-inflammatory drugs (NSAIDs) 581 respiratory failure (RF) 278 venous thromboembolism (VTE) 292 vitamin D 881 Paroxetine 520 Paroxysmal events, cardiology 7–8 Paroxysmal nocturnal hemoglobinuria (PNH) 1012–1013 Pasireotide 810 Patent foramen ovale (PFO) 7–8, 51–52, 112–113, 194 surgery 200 Pathergy phenomenon 1704–1705 ‘Pavor mortis’ 994–995 Pediatric intracranial pressure (ICP), increased 1759 Pediatric multivisceral/intestinal (MVI) transplantation 1309 Pediatric spine trauma 1767 Pediculus humanus (human body louse) 1534–1535 Pegvisomant 697–698, 810, 812 Pelamis platurus 992 Peliosis rheumatica 1101 Pellagra 626, 891, 902–903 bariatric surgery 591 Pelvic bone, Paget’s disease of bone (PDB) 531 Pelvic neurologic symptoms, ovarian tumors 789, 791–792 Pemoline 1644 Penicillin 1357, 1369, 1508, 1646 Lyme disease 1479, 1480, 1533, 1577 neurosyphilis 1336–1337, 1465–1466, 1468 Penicillin G 1337, 1468, 1534 benzathine 1468, 1469–1470, 1508, 1534 Pentastomiasis 1436 Pentobarbital 1262 Pentostatin 1134, 1208 Perchlorates 813, 814 Percutaneous coronary intervention (PCI) 194, 194, 196–197 Percutaneous vertebroplasty 342–343 Perfluorocarbon emulsions (PFCs) 965 Pergolide 811

Periodic alternating gaze deviation (PAGD) 646 Periodic limb movement disorder (PLMD) 257, 398–399 Peripartum cardiomyopathy (PPCM) 1598 Peripheral facial nerve palsy 321–322 Peripheral nervous system (PNS) 9–10 AL amyloidosis 1095 bortezomib-induced (BiPN) 1089 breathing 246, 248 cardiac surgery complications 197 cardiac tests 41–43 chemotherapy complications 1199–1205 cryoglobulinemia 1096 Cuban epidemic myeloneuropathy (CEM) 1544 diabetic neuropathy (DPN) 776–777 dialysis patients 396, 400–401 Ehlers–Danlos syndromes (EDS) 570–571 fibromyalgia 516 heart transplantation 1230, 1234 HIV 1322 hyperthyroidism 714–715 hypothyroidism 707–708 inflammatory bowel diseases (IBD) 596, 597 injuries 200 intestinal transplantation (ITx) 1273 leukemias 1041 lymphomas 1037 monoclonal gammopathy of undetermined significance (MGUS) 1084, 1085–1088 multiple myeloma (MM) 1089 neuro-Behc¸et syndrome (NBS) 1711, 1712 neurocutaneous disorders 1573–1580 neuropsychiatric systemic lupus erythematosus (NPSLE) 466 obstructive sleep apnea syndrome (OSAS) 259 pancreas transplantation 1283 paraneoplastic neurologic syndromes (PNS) 1170–1172 plasma cell disorders 1084 POEMS syndrome 1092, 1093, 1578 positioning injuries 1629–1630 renal disease 388–391 renal transplantation 1251 sarcoidosis 312–313, 323 scleroderma (systemic sclerosis) (SSc) 468–469 sickle cell disease (SCD) 1016 Sj€ ogren’s syndrome (SS) 470–471 thalidomide-induced (TiPN) 1090 Waldenstr€ om macroglobulinemia (WM) 1093 Permetrexed 339 Pernicious anemia (PA) 915, 917, 921, 922, 923 folic acid deficiency 927, 929–930, 930, 936–937

I33 Pernicious anemia (PA) (Continued ) progressive 927 tropics 1541 Peroneal nerve injury 1240 Persistent intracranial pressure (ICP), increased 1759–1760 Pertussis 1358 hemolytic uremic syndrome (HUS) 1114, 1114 immunization 1358 diphtheria-tetanus-pertussis (DTP) vaccine 1358, 1550, 1555–1556, 1651 Phaeohypomycosis spp 1385 PHASE (Pre-Hospital Survival Evaluation) study 27 Phaseolus vulgaris 1544 Phenformin 816 Phenobarbital 677, 1153, 1231, 1504, 1640 elimination 418, 421, 425, 427 seizures 846, 1234 Phenothiazines 634 Phenoxybenzamine 766 Phenprocoumon 1638–1639 Phenylbutazone 1649 Phenylbuterate 762 Phenytoin 677, 1153, 1231, 1517, 1628 adverse effects 1640, 1640 elimination 417–420, 418, 425, 427 seizures 846, 847, 1234, 1274 Pheochromocytomas 805–806 PHES (psychometric hepatic encephalopathy score) 665 Phosphoribosylpyrophosphate synthase (PRPS) 828, 829–830 Phosphorus disorders, congenital/acquired 874 homeostasis 737–738 Phosphorus-32 1189 Photons 1183 Phrenic nerve injury 199 catheter ablation 154 electrophysiologic procedures 142 Phrenic neuropathy 1681 Physician-assisted suicide (PAS) 1222 Physician-associated meningitis 1362 Pica 1007 Pickwick morphotype 257 Pickwickian syndrome (obesity hypoventilation syndrome (OHS)) 278 Picture-frame sign 535 PIGA gene 1012–1013 Pioglitazone 817, 819, 820 Pipecuronium 1509 ‘Pit vipers’ 992–993 Pituitary hormones/analogs 809, 810 anterior 811–812 posterior 812 Pituitary lesions 410, 683–702 adenomas 685, 687–688 radiotherapy (RT) 1186–1187, 1187 apoplexy (Sheehan syndrome) 756–757, 1604–1605 central diabetes insipidus (DI) 690

I34 Pituitary lesions (Continued ) future directions 699 growth hormone (GH) deficiency 690 hormonal hyperfunction 690–694 hypopituitarism 688–689 hypothalamic-pituitary axis, evaluation 686, 690 imaging 694–695, 694, 695 metastases 345 neurologic manifestations 685–688, 686 pituitary/target hormones 686 prolactin deficiency 690 secondary conditions adrenal insufficiency (AI) 689 hypogonadism 689–690 hypothyroidism 689 tumors see Pituitary tumors Pituitary tumors 686, 687, 695–699 growth predictors 695 medical management 697–698 radiotherapy 698–699 surgical management 695–696, 696 postoperative complications 696–697 PLA2G6 gene 859, 860 PLAATO (Percutaneous Left Atrial Appendage Transcatheter Occlusion study) device 142–143 Planning target volume (PTV) 1181–1182 Plant alkaloids 1652 Plasma cell disorders 1083–1100 cryoglobulinemia see Cryoglobulinemia macroglobulinemia see Waldenstr€ om’s macroglobulinemia (WM) myeloma, nonsecretory 1083, 1092 see also Multiple myeloma (MM) POEMS syndrome see POEMS syndrome solitary plasmacytoma 1083, 1084, 1089, 1092 see also Monoclonal gammopathy of undetermined significance (MGUS) Plasma infusion/plasmapheresis 1120 Plasmacytoma intracranial 1091 solitary 1083, 1084, 1089, 1092 Plasminogen activator inhibitor-1 (PAL-1) deficiency 1056 Plasmodium falciparum 1514, 1515, 1516, 1517 Plasmodium knowlesi 1514 Plasmodium malariae 1514 Plasmodium ovale 1514 Plasmodium vivax 1514 Platelet function disorders 1057–1058 anatomy/physiology 1057 diagnosis 1057–1058 symptoms, bleeding 1047, 1057 treatment 1055, 1058 Platinum-based agents 1134, 1652 adverse effects 1199–1200, 1201, 1208 PLATO (Platelet Inhibition and Patient Outcomes) trial 101 Platyhelminths (flatworms) 1414, 1419–1429

INDEX Platyzoa 1414 p-aminosalicylic acid (PAS) 1491, 1492 Plexiform neurofibromas 1581–1582 Plexopathy brachial 199, 199, 345–346, 1192–1193 lumbrosacral 592, 776 pregnancy 1610 Plicamycin 878 Pneumococcal vaccines 1550, 1554–1555, 1651 Pneumococcus spp 1532 Pneumocystis carinii 1232, 1246, 1248 Pneumocystis jiroveci 1323, 1325, 1328–1329 PNM system (WHO) 1426–1427 Podophyllin 1203 Podophyllotoxins 1152 Poecilotheriinae 994 POEMS (polyneuropathy, organomegaly, endocrinopathy, M-protein, skin changes) (Crow–Fukase) syndrome 845, 1032, 1083, 1084, 1087, 1092–1093, 1578–1579 clinical features 1037, 1578 laboratory studies/diagnosis 1578, 1579 peripheral nervous system (PNS) 1092, 1093 treatment 1578–1579 Poiseuille’s Law 41 Poliomyelitis 1527, 1553 Poliovirus vaccine 1550, 1553, 1651 Polyarteritis nodosa (PAN) 482–483 clinical features 482, 482 Polycythemia 51 Polycythemia vera (PV) 1074–1075 clinical characteristics 1074 defined 1073 diagnostic criteria/risk assessment 1074 JAK2 gene 1073–1074 neurologic symptoms 1074–1075 thrombosis/hemorrhage 1075 treatment 1075, 1132 Polyenes 1388–1389, 1395, 1649 Polyglandular autoimmune syndrome 750 type I 871 Polymixin 1646 Polymyalgia rheumatica 711 Polymyositis (PM) see Idiopathic inflammatory myopathy (IIM) Polyneuropathy critical illness see Critical illness polyneuropathy (CIP) dialysis patients 400–401 distal see Distal symmetric polyneuropathy (DSP) endemic ataxic 1542–1543 infective endocarditis 79 inflammatory chronic demyelinating polyneuropathy (CIDP) 841, 845, 1088, 1333, 1611 see also Guillain–Barre´ syndrome (GBS); Inflammatory demyelinating polyneuropathy (IDP)

Polyneuropathy (Continued ) nutritional (dry beriberi) 896–897 sensory/sensory motor 353, 774 small fiber 774 see also POEMS syndrome Polyol pathway 777 Polysomnography (PSG) 260, 261 Pomalidomide 1032, 1090 Porocephalalosis 1436 Porocephalus spp 1436 Porphyria 839–849 background 839–841, 840 clinical presentation 841–842 decompression illness (DCI) 964 diagnosis 845–846 nervous system dysfunction, mechanisms 843–845 risk factors 842–843 treatment 846–847 Portogonyaulax spp 996 Portosystemic encephalopathy 661–674 clinical findings 661–663, 662 differential diagnosis 663 historical perspective 661 laboratory investigations 666–667 management 670–671 minimal hepatic encephalopathy (mHE) 661–662, 663–665, 664, 666 natural history 665–666 neuroimaging 667–669, 667, 668 pathology/pathophysiology 669–670 Posaconazole 1329–1330 Positioning injuries 1629–1630 Positive airway pressure (PAP) therapy 261–262 Positron emission tomography (PET) brain metastases 1148, 1149 fibromyalgia 515–516 Post Lyme disease syndrome 1476 Post-cardiac arrest syndrome 26 Posterior ischemic optic neuropathy (PION) 1629–1630 Posterior reversible encephalopathy (leukoencephalopathy) syndrome (PRES/PRLS) 161, 384–385, 452, 1108, 1213, 1245–1246, 1687–1701 causes 1692–1695, 1692 clinical features 1687–1688, 1688 dialysis patients 397–398 Guillain–Barre´ syndrome (GBS) 9–10 heart transplantation 1230 Henoch–Sch€ onlein purpura (HSP) 1104 infection 1694 intestinal transplantation (ITx) 1271–1272 nephrotic syndrome 409–410, 410 neuroradiologic features 1688–1692, 1688 additional sequences 1690–1692 catheter/magnetic resonance angiography 1692 conventional imaging 1688–1690, 1689, 1690 pancreas/small bowel transplantation 1278–1279

INDEX Posterior reversible encephalopathy (leukoencephalopathy) syndrome (PRES/PRLS) (Continued ) pathological features 1696–1697 pathophysiology 1695–1696 Rapamune (sirolimus) 1246 reversible cerebral vasoconstriction syndrome (RCVS), overlap 1727 total body irradiation (TBI) 1299 treatment 1697–1698 Postherpetic neuralgia (PHN) 1577–1578 Posthypoxic myoclonus (PHM) 35 Postpartum angiopathy see Reversible cerebral vasoconstriction syndrome (RCVS) Post-resuscitation disease 26 Postsynaptic toxins 989–990 Post-transplant lymphoproliferative disorder (PTLD) 1233, 1241 classification 1233 intestinal transplantation (ITx) 1270 multivisceral and intestinal (MVI) transplantations 1313, 1314 pancreas transplantation 1282 small bowel transplantation 1287–1288 Post-traumatic bacterial meningitis 1365 Postural tachycardia syndrome (PoTS) 172, 173 Posture see Orthostatic hypotension (OH) Potassium chloride 721 Potassium perchlorate 813 Pott’s disease 446, 1530, 1531 Powassan virus 1378, 1380 Praeder–Labhart–Willi syndrome 796 Pramipexole 522 Pramlintide 817, 820–821 Prasugrel 1129, 1639 Pravastatin 1638 Praziquantel 1421, 1422, 1426, 1429, 1545 neurocysticercosis 1455, 1537 Prazosin 994, 1636 Precocious presentation, brain metastases 1144 Prednisolone 1513–1514, 1608, 1613, 1718 Prednisone 459, 481, 506, 1333, 1470, 1545 lung cancer 354 pregnancy 1608, 1613 sarcoidosis 321–322, 323, 325 Pre-eclampsia humoral factors 1597 hypertension/stroke risk mitigation 1598 long-term effects 1597–1598 posterior reversible encephalopathy syndrome (PRES) 1693–1694 role of placenta 1597 stroke 1596–1598 ischemic 1598–1602 treatment 1604 Pregabalin 521, 780, 1332–1333 elimination 418, 425, 426, 427 Pregnancy 1595–1622 brain tumors 1614–1616, 1615 epilepsy 1606–1607 future directions 1616–1617

Pregnancy (Continued ) headache 1613–1614 primary 1614 secondary 1614 multiple sclerosis (MS) 1607–1608 disease-modifying therapies (DMTs) 1606, 1607–1608 predictions 1607 relapse rates 1607 neuromuscular disease 1608–1613 acquired inflammatory demyelinating polyneuropathy (AIDP) 1611 chronic inflammatory demyelinating polyneuropathy (CIDP) 1611 hereditary neuropathy/dystrophy/ myopathy 1612 immunosuppressant treatment 1613 inflammatory myopathy 1612 lower extremity mononeuropathy/ radiculopathy 1609 upper extremity mononeuropathy/ radiculopathy 1608–1609 multifocal motor neuropathy (MMN) 1611–1612 myasthenia gravis (MG) 1610–1611 outcomes 1612–1613 plexopathy 1610 spinal cord disorders 1616 stroke 1595–1606 cerebral venous thrombosis (CVT) 1605–1606 epidemiology 1595 hemorrhagic 1602–1605 maternal physiologic changes 1596 underlying mechanisms 1595–1596 vascular endothelial dysfunction 1595, 1596–1598 Pregnancy in Multiple Sclerosis Group (PRIMS) 1607 Premature aging, vitamin D 881 Premature ventricular contractions (PVCs) 130 Prenatal stress 517 Pressure control ventilation (PCV) 279 Pressure palsy 1681 Presynaptic toxins 990 Pre-syncope 172 Prevention of Venous Thromboembolism After Acute Ischemic Stroke with Low Molecular Weight Heparin (PREVAIL) study 296–297, 300 Preventive Services Task Force (USA) 1654 Prickly heat 948 Primary angiitis of central nervous system (PACNS) 486–487, 1725–1726, 1736 clinical features 486–487, 486 diagnosis 487 differential diagnosis 487 Primary brain gliomas 1149 Primary brain tumors 726 Primary care, developing world 1774 Primary central nervous system lymphoma (PCNSL) 1028–1030 brain metastases 1149

I35 Primary central nervous system lymphoma (PCNSL) (Continued ) clinical presentation 1036 radiotherapy (RT) 1185–1186 Primary central nervous system lymphoma (PCNSL), HIV-associated 1321, 1326–1327 brain biopsy 1326 clinical presentation 1326 management 1326–1327 neuroimaging 1326 pathogenesis 1326 Primary central sleep apnea (CSA) 263, 263 Primary cerebral angiitis 1736 Primary dural lymphoma, clinical presentation 1036 Primary graft dysfunction (PGD) 1238, 1239 Primary headache disorder 1450 thunderclap 1726 Primary hyperparathyroidism (PHPT) 738–742, 866–870 clinical/biochemical features 739–741, 867 cardiovascular manifestations 867 gastrointestinal manifestations 867 neurologic/muscular manifestations 739–741 neuromuscular/neuropsychiatric manifestations 867–869 renal manifestations 867 skeletal manifestations 867 diagnosis 866 etiology 866 management 869–870 pathophysiology 738–739 Third International Workshop 869 treatment 741–742 Primary myelofibrosis (PMF) 1077–1078 clinical characteristics 1077 defined 1073 diagnostic criteria/risk assessment 1077–1078 JAK2 gene 1073–1074 neurologic manifestations 1078 treatment 1078 Primary ovarian failure (POF) 793–796, 794 Primary plasma cell leukemia 1032 Primary sleep apnea of infancy 264–265 Primidone 418, 421, 425, 427 PRISM (Randomised Trial of Intensive versus Symptomatic Management of Paget’s Disease) trial 536 PRKAR1A gene 1566 Probenecid 1534 Procainamide 132, 135 Procarbazine 1184–1185, 1201, 1201, 1210 Prochlorperazine 634, 639 Procyclidine 636–637 PRODIGE study, low molecular weight heparin (LMWH) 299 Progressive craniovertebral subluxation 437–438

I36 Progressive multifocal leukoencephalopathy (PML) 1149, 1378, 1379 intestinal transplantation (ITx) 1272 JC polyomavirus 1232, 1263 liver transplantation 1263 rituximab 1278–1279 treatment 1136 Progressive multifocal leukoencephalopathy (PML), HIVassociated 1327–1328 clinical presentation 1327 diagnosis 1327–1328, 1327 epidemiology 1327 pathogenesis 1327 treatment 1328 Progressive polyradiculopathy 1334, 1335 Progressive systemic sclerosis (PSS) 468 Prolactin 686, 690 Prolactin blocking agents 1655 Prolactinomas 687, 691, 697 Prolonged mechanical ventilation (PMV) 283 Promethazine 634, 639, 1642 Pro-opiomelanocortin (POMC) 750 Propafenone 132, 135 Propionibacterium acnes 318 Propofol 846–847, 1260–1261, 1262, 1624, 1625, 1630 Propofol infusion syndrome 1677, 1758 Propranolol 187, 721, 1274, 1571 adverse effects 132, 135, 1636 Propylthiouracil (PTU) 813–814, 813 Prospective Investigation of Pulmonary Embolism Diagnosis (PIOPED) 293 Prostacyclin 1738 Prostaglandin E 56 Prosthetic heart valves 63–65 Protamine 1132 Protease inhibitors (PIs) 1324–1325, 1340, 1494 Proteasome inhibitors 1136–1137, 1203 PROTECT AF study, warfarin treatment 143 Protein 622 Protein kinase C (PKC) pathway 778 Protein S100B 34 Prothrombin complex concentrates (PCCs) 1128, 1131–1132 Proton magnetic resonance spectroscopy (H-MRS) 516 Proton pump inhibitors 638–639 Proton radiobiology 1183 Protozoa 1404–1414, 1407 PRPS1 gene 830 PSE-Syndrom-Test 663–664, 664, 665 Pseudechis spp 992 Pseudohyponatremia 365 Pseudohypoparathyroidism see Hypoparathyroidism/ pseudohypoparathyroidism Pseudomonas aeruginosa 1369, 1532 Pseudonaja spp 992

INDEX Pseudopseudohyperparathyroidism 745 Pseudotumors, hemophilic 1053–1054 Pseudoxanthoma elasticum (PXE) (Gr€ onblad–Strandberg syndrome) 571–572, 1568 clinical features 571, 572, 1568 cutaneous manifestations 1568 eye involvement 1568 vascular manifestations 1568 diagnosis/management 1568–1569, 1569 neurologic complications 571–572 Psychiatric manifestations hyperthyroidism 722–723 hypothyroidism 706–707 inflammatory bowel diseases (IBD) 601 neurocysticercosis (NCC) 1450 porphyria 842 see also Mental disturbances Psychiatric transient loss of consciousness (TLOC) 179, 180–181, 184 clinical features 180–181, 180 etiology/pathophysiology 181 falls 184 Psychogenic pseudosyncope see Psychiatric transient loss of consciousness (TLOC) Psychosis 465 Psychostimulants 1644, 1645 Puerperium see Pregnancy Pufferfish 964, 988 Pulmonary cement embolization (PCE) 1747–1748 Pulmonary disorders angiography 293 embolism (PE) see Venous thromboembolism (VTE) hypertension 258 obstructive sleep apnea syndrome (OSAS) 258 parenchymal/vascular pathology 266 vein isolation 143–144 Pure red cell aplasia (PRCA) 1010 Purified protein derivative (PPD) 1247–1248 Purine analogs 1200 Purine metabolism, disorders of 827–833 inborn errors of metabolism, indicators 835 nucleoside phosphorylase (PNP) deficiency 828, 832 overview 827, 828 PURSUIT (Platelet glycoprotein IIb/IIIa in Unstable angina. Receptor Suppression using Integrin Therapy) trial 101–103 Purulent meningitis 1411 Pyrazinamide 1490, 1491, 1495 Pyridostigmine 185, 354, 781, 1628 Pyridoxine (pyridoxal) see Vitamin B6 deficiency Pyrimethamine 1325 Pyrimidine analogs 1134, 1395, 1649

Pyrimidine metabolism, disorders of 833–835, 834 catabolic disorders 835 inborn errors of metabolism, indicators 835 nucleotide depletion and overactive cytosolic 5’-nucleotidase 834, 834 overview 827

Q Quality of life (QOL) 868–869, 870 Quatember and Maly Clocktest 261 Quinagolide 810, 811 Quinidine 132, 136, 1637 Quinine 1517

R R173W gene 841 Rabeprazole 638–639 Rabies 1501–1506 clinical features 1501–1502 diagnosis 1502–1504, 1505 differential diagnosis 1502 encephalitis 1378, 1379 diagnosis 1380 management 1504 natural history 1504 pathogenesis/pathology 1502, 1503 prevention 1504–1506 post-exposure treatment 1506, 1506 vaccines 1504–1506, 1506, 1550, 1553–1554 renal transplantation 1250 Radial artery catheterization 5 Radiation see Radiotherapy (RT) Radiation Therapy Oncology Group (RTOG) 1191, 1326 study 9802 1184, 1185–1186 Radiculomyelitis 1495 Radiculoneuritis 1474–1475 Radiculopathy 456, 1322, 1334, 1335 lower extremity 1609 upper extremity 1608–1609 Radiofrequency ablation (RFA) 262 Radioimmunotherapy 1136 Radiosensitizers 1200, 1204, 1212 Radiosurgery 1186–1187, 1188, 1189, 1190 stereotactic see Stereotactic radiosurgery (SRS) Radiotherapy (RT) 947, 1181–1198 biology 1183, 1183 -induced conditions 1150 irradiation process 1181–1183, 1182 neurosarcoidosis 325 normal tissue, effects on 1189–1193 early 1190 delayed 1190–1191 late 1191–1193 sarcoidosis 327 treatment, neurologic disease 1184–1189 benign tumors 1186–1189 malignant tumors 1184–1186 nontumor indications 1189 Ramosetron 636

INDEX Ranirestat 779 Ranitidine 638–639 Rapamune/rapamycin see Sirolimus Rattlesnakes (Crotalus) 992–993 Reactive arthritis 457 Recluse (violin) spiders 994 Recombinant factor VIIa (NovoSeven®) 1128 Recombinant vaccines 1550 Recurrent laryngeal nerve palsy 726 Red blood cells (RBC) 1005–1006 distribution width (RDW) 1006, 1008 Refsum disease 1562, 1579–1580 clinical manifestations 1579 diagnosis/treatment 1579 Regional wall motion abnormality (RWMA) 6–7 186Re-HEDP, adverse effects 1655 Relapsing polychondritis (RP) 457–459 clinical findings 457–458 diagnosis 458 natural history 458 pathogenesis/treatment 458–459 RE-LY (Randomized Evaluation of Long-Term Anticoagulation Therapy) trial 119–120, 121–122 Remethylation defects 935, 935 Remifentanil 1626–1628 Renal disease, acute/chronic 383–394 central nervous system (CNS) complications 383–385 renal replacement therapy 385–388 peripheral nervous system (PNS) disorders 388–391 see also Antiepileptic drugs (AEDs), hepatic/renal disease; Dialysis patients Renal manifestations Fabry disease (FD) 1564 primary hyperparathyroidism (PHPT) 867 Renal replacement therapy 385–388 Renal transplantation 1245–1255 central nervous system (CNS) 1247–1250 bacterial infection 1247–1248 fungi 1248–1249 parasites 1248 viral infection 1249–1250 cerebrovascular disease 1251 diagnostic approach 1252–1253, 1252 drug-related neurotoxicity 1245–1247 calcineurin inhibitors (CNI) 1245–1246, 1246 immunosuppression 1246 steroids 1246 metabolic causes 1251–1252 neoplastic diseases 1250–1251 peripheral neuropathy 1251 treatments, neurologic 1253 Renzapride 638 Repaglinide 817, 820 Repeatable Battery for the Assessment of Neuropsychological Status (RBANS) 663–664 Reperfusion injury 20, 25–26

Replacement therapies 385–388, 1125–1128 Rescue ICP trial 1759–1760 Reserpine 20 Respiratory disorders achondroplasia 556–557 acid-base 375 acidosis 375–376 acute respiratory distress syndrome (ARDS) 280 alkalosis 375–376 muscle weakness 1357 see also Breathing Respiratory failure (RF), acute/chronic 273–288 classification 273 diagnosis 273–275, 274 management principles 279–283 acute respiratory distress syndrome (ARDS) 280 mechanical ventilation 279–283, 280, 281, 282 noninvasive ventilation (NIV) 281 neuromuscular disease (NMD) 273–275, 284–285 home mechanical ventilation (HMV) 285 muscle function devices 284 noninvasive ventilation (NIV) 284–285 oropharyngeal/tracheobronchial secretions 283 specific diseases 275–279 types 273 Respiratory tract drugs 1655 Restless legs syndrome (RLS) 257 dialysis patients 398–399 iron disorders 861 renal disease 390 RET gene 804, 806 Reteplase 104 Retigabine 418, 425, 427, 429 Retinal degeneration 354 Retinal injury, radiotherapy (RT) 1192 Retinoids 1201, 1204, 1212–1213, 1652 Retinopathy, carcinoma-associated 351 Retroviral agents 1648 Retroviridae family 1527 Return of spontaneous circulation (ROSC) 28–30 Reversible cerebral vasoconstriction syndrome (RCVS) 1599–1600, 1599, 1650, 1725–1741 causes/associated conditions 1726–1727, 1727 posterior reversible encephalopathy syndrome (PRES), overlap 1727 clinical findings 1727–1729 demographics 1727–1728 diagnostic criteria 1729, 1729, 1730 focal deficits/seizures 1728 general examination 1729 thunderclap headache 1726, 1728 differential diagnosis 1734–1737 exertion/sexual activity 1737

I37 Reversible cerebral vasoconstriction syndrome (RCVS) (Continued ) migraine 1736–1737 primary cerebral angiitis 1736 thunderclap headache 1730, 1734–1736, 1737 etiology/pathophysiology 1727, 1737 future directions 1738 historical perspective 1725–1726 laboratory investigations 1731 management 1737–1738 natural history 1729–1731, 1731 neuroimaging 1731–1734 abnormalities, timing 1734 brain computed tomography (CT)/ magnetic resonance imaging (MRI) 1731–1732, 1732, 1733, 1734, 1735 cerebral angiography 1732–1733, 1735, 1736 pathology 1733 ultrasound 1733 nomenclature 1726 posterior reversible encephalopathy syndrome (PRES) 1692 postpartum 1726–1727 terminology 1725–1726, 1726 Reversible posterior leukoencephalopathy syndrome (RPLS) see Posterior reversible encephalopathy (leukoencephalopathy) syndrome (PRES/PRLS) Reversible splenial lesion syndrome 369 Reye’s syndrome 582, 645–646 Rhabdomyolysis intensive care unit (ICU) 1676–1677 renal transplantation 1252 Rhabdomyoma 216 Rhabdoviridae family 1501 Rhesus macaque monkey, Lyme disease 1475 Rheumatic mitral valve disease 61–62 Rheumatic valvular heart disease (RVHD) 61–62 Rheumatoid arthritis (RA) 449–454 atlantoaxial subluxation 440, 453 autonomic nervous system (ANS) dysfunction 452 cerebral vasculitis 451–452, 452 classification criteria 450 craniocervical dislocation 435, 440, 441–442, 442 leptomeningeal (LM) involvement 451 management 452–453 nerve entrapment 450–451, 450 prognosis 454 subclinical neurologic disease 449, 450 treatment, complications 453–454 vasculitic neuropathy 451 Rhinocerebral syndromes, fungal infection 1386 Rhinocerebral zygomycosis 1385, 1396–1397, 1397 Rhizomucor 1383, 1396 Rhizopus 1383, 1396

I38 Ri-Abs (antibodies) 1164 Ribavirin (Rebetol®) 676, 679, 1348–1349, 1381, 1504 mechanism of action 679 neurotoxicity 679 Riboflavin see Vitamin B2 deficiency Rickettsia 1403 Ridley–Jopling classification, leprosy 1509–1510, 1510 Rifabutin 1247–1248, 1338, 1494, 1495 Rifampicin (rifamycin/rifampin) 1357, 1512–1513, 1575–1576, 1646 tuberculosis (TB) 1247–1248, 1338, 1494 tuberculous meningitis(TBM) 1490, 1491, 1492 Rikettsiae (Rhizobiales/Rickettsiales) 1403–1404 clinical manifestations 1403, 1405 epidemiology 1404 pathogens 1403 therapy 1403–1404 Riley–Day syndrome 172 Risedronate 878 Risperidone 1642 Rituximab (Rituxan®) 485–486, 718–719, 1120, 1136, 1170, 1173 Riva-Rocci, Scipione 161 Rivaroxaban 139, 295, 1639 Rivastigmine 1642 Rochester (Minnesota) Epidemiology Project 1462 Rocuronium 1509 Rofecoxib 580, 1649 Ropinirole 522 Rosiglitazone 819 Rotavirus vaccine 1651 Roundworms 1414, 1429–1436 Royal College of Physicians, Gulstonian Lectures (1885) 75–76 Royal Medico-Chirurgical Society 529 Rubella virus 1349–1350 clinical manifestations 1349–1350 acquired rubella infection 1349–1350 congenital rubella syndrome (CRS) 1350 diagnosis 1350 epidemiology 1349 measles-mumps-rubella (MMR) vaccine 1349, 1350, 1550–1551, 1550, 1651 treatment/prognosis/prevention 1350 Rubitecan 1212 Ruboxistaurin 779 Rufinamide 418, 425, 427, 428–429 Russell’s viper (Daboia russelii) 988, 990, 992–993

S Saddle embolism 296 S-adenosylmethionine (SAM) 928, 928, 929, 1206, 1541 St Louis encephalitis virus 1378, 1379, 1380 Salicylates 1648 Salmonella spp 1532 SAR1B gene 624

INDEX Sarcoidosis 305–334 brain metastases 1149–1150 clinical course 313 clinical findings 305–313, 306 CNS parenchymal disease 309–312 hydrocephalus 308–309, 309 meningeal disease 308, 308, 309 myopathy 313 neurosarcoidosis see Neurosarcoidosis peripheral neuropathy 312–313, 323 epidemiology/pathophysiology/genetics 317–320, 319 historical perspective 305 investigations 313–317 differential diagnosis 313–314, 315 laboratory 306, 314–316 neuroimaging 308, 309, 312, 316–317, 317 management 321–328 alternative treatments 321, 324–327, 325 radiation therapy 327 supportive care 327–328 surgical considerations 324 treatment 321–323, 321 pathology 320–321, 320 Sarcoma, Paget’s 531–532, 536 sarcomatous transformation 531 SAVE (Survival and Ventricular Enlargement) trial 113, 114 Saw-scaled (carpet) vipers (Echis) 992–993 Saxagliptin 817, 820 Scandinavian Glioblastoma Study Group 1184 SCD-HeFT (Sudden Cardiac Death in Heart Failure Trial) 114 Schilder–Addison disease 760 Schilling test 921 Schistosoma 1419–1421, 1419, 1535–1536 Schistosoma haematobium 1419–1420, 1535 Schistosoma japonicum 1420, 1535, 1777 Schistosoma mansoni 1420, 1535 Schistosomiasis see Bilharziasis Schmidt’s syndrome see Autoimmune polyglandular syndrome, type II (APSII) Scleroderma (systemic sclerosis) (SSc) 468–470 autonomic nervous system (ANS) 469 central nervous system (CNS) 468, 469 musculoskeletal involvement 468 neurologic complications 464, 468 peripheral nervous system (PNS) 468–469 treatment 469–470 SCN1A gene 1358 Scoliosis 559 Scopolamine 187 Scorpionida 993 Scorpions see under Venomous bites Scurvy 891, 908–910 Sea kraits (Laticauda) 992

Sea snakes 989, 992 Second International Study of Infarct Survival (ISIS-2 Collaborative Group) 105 Secondary adrenal insufficiency 689 Secondary care, developing world 1774 Secondary hyperparathyroidism 742–743, 744 Secondary hypogonadism 689–690 Secondary hypothyroidism 689 Secondary intracranial hypertension 365 Sedatives 519, 522, 1240, 1644 Seizures bacterial meningitis 1372 brain metastases 1144–1145 cardiac arrest 31 cardiac catheterization 197 cardiac surgery 55 chronic 1149 coronary artery bypass grafting (CABG) 198 decompression illness (DCI) 964 dialysis, causes 386, 386 end of life (EOL) phase 1219–1220 heart transplantation 1230, 1233–1234, 1235 Henoch–Sch€ onlein purpura (HSP) 1105 hyperthyroidism 722 hypothyroidism 706 infective endocarditis 69 intestinal transplantation (ITx) 1272, 1273, 1274 liver transplantation 1261 neurocysticercosis (NCC) 1448–1449 neuropsychiatric systemic lupus erythematosus (NPSLE) 465 neurosarcoidosis 311–312, 323 obstructive sleep apnea syndrome (OSAS) 259 pancreas/small bowel transplantation 1278 posterior reversible encephalopathy syndrome (PRES) 1696 psychogenic nonepileptic 180–181 reversible cerebral vasoconstriction syndrome (RCVS) 1728 small bowel transplantation 1287–1288 treatment 846–847, 1221 Seldinger technique 41 Selective COX-2 inhibitors 1649 Selective nerve root block (SNRB) 1642 Selective serotonin norepinephrine reuptake inhibitors (SSNRIs) 776, 781 Selective serotonin reuptake inhibitors (SSRIs) 519, 520, 678–679, 781, 1642, 1651 Selegiline 1641 Selenium 31 deficiency 591 Selenocosmiinae 994 Semple vaccine 1554 Semustine (methyl-CCNU) 1208–1209 Sensorineural hearing loss (SNHL) 201, 1544

INDEX Sensory/sensory motor polyneuropathy 353, 774 Sepsis 1694 Sepsis syndrome 281 Sequential compression devices (SCDs) 291, 294–295, 296, 297, 298–299 Serial Dotting Test (SDT) 663–664, 664 Serotonin 3 receptor antagonists 636–637 Serotonin 5-HT3 receptor antagonists 519, 522 Serotonin reuptake inhibitors 187 Serotonin syndrome (SS) 639, 949–951, 950, 1651 treatment 951 Serotonin-norepinephrine reuptake inhibitors (SNRIs) 519, 520–521, 1642 Sertindole 1642 Sertraline 520 Serum angiotensin-converting enzyme (SACE) 314 Serum creatine kinase (sCK) activity 501 Setrons 636–637, 639 Severe combined immunodeficiency (SCID) 832 Severe hypermagnesemia 871 Sevoflurane 1626, 1627, 1630–1631 SF-36 questionnaire (health-related quality of life) 507, 514, 536, 868–869 Sheehan syndrome 756–757, 1604–1605 Shellfish 988 Shiga toxin (Stx) 1115, 1120 Shiga toxin-producing Escherichia coli (STEC) 1113–1114, 1114, 1115, 1118 Shigella spp, hemolytic uremic syndrome (HUS) 1113–1114, 1114, 1120 Shingles see Herpes zoster Shock 1694 Shock liver 645–646 Short bowel syndrome (SBS) 1286, 1305–1307 Shunt-associated meningitis 1369 Sick sinus syndrome (SSS) 130, 141 Sickle cell disease (SCD) 1015–1027 clinical findings 1015–1016 genetics 1018–1019 historical perspective 1015 laboratory investigations 1016–1017 management 1020–1022 natural history 1016 neuroimaging 1017–1018 pathology 1019–1020 Sildenafil 782, 1654 Silent cerebral infarct 1016 Silent lesions 77 Silent myocardial infarction 94 Simvastatin 1638 ‘Single child family’ (China) 1778–1779 Single-nucleotide polymorphisms (SMPs) 1362 Single-photon emission computed tomography (SPECT) 515, 1715 Sinus node dysfunction 140–141 Sipple syndrome see Multiple endocrine neoplasia (MEN), type 2A

Sirolimus (rapamune/rapamycin) 1246, 1275 adverse effects 676, 677–678 transplantation 1231, 1260, 1281–1282, 1289–1290 Sitagliptin 817, 820 Situational syncope 174 Sj€ ogren–Larsson syndrome (SLS) 1562, 1589–1590 clinical symptoms 1589 cutaneous 1589 neurologic 1589 ophthalmologic 1589 diagnosis/testing 1589 radiology 1589–1590 treatment 1590 Sj€ ogren’s syndrome (SS) autonomic nervous system (ANS) 471 central nervous system (CNS) 470 muscle involvement 471 neurologic complications 464, 470 peripheral nervous system (PNS) 470–471 treatment 471 Skeletal manifestations hyperthyroid myopathy 713–714 primary hyperparathyroidism (PHPT) 867 Skin biopsy 1367 Skin lesions, Behc¸et’s syndrome (BS) 1704 Skull base, metastases 345 Paget’s disease of bone (PDB) 530 Skull fractures depressed 1760 repair of depressed 1760 traumatic brain injury (TBI) 1758 ‘Slapped cheek’ rash 1351 Sleep-related disorders 257 dialysis patients 398 hypothyroidism 708 nonobstructive idiopathic alveolar hypoventilation 265 sleep apnea syndrome (SAS) 398, 556 ‘sleep attacks’/sleepiness 177, 180–181, 184 sleeping sickness, African 1412, 1413, 1777 see also Central sleep apnea (CSA); Obstructive sleep apnea (OSA)/ syndrome (OSAS) Sleep-related hypoventilation /hypoxemia, lower airway obstruction 265–266 neuromuscular/chest wall disorders 265, 266 pulmonary parenchymal/vascular pathology 266 Small bowel transplantation 1285–1290 diagnostic evaluation 1289 neurologic complications 1277–1278 perioperative 1286–1287 postoperative 1278–1279, 1287–1288, 1287 short bowel syndrome 1286

I39 Small bowel transplantation (Continued ) total parenteral nutrition (TPN) 1286 neuropathology 1288, 1289 period preceding 1277 treatment 1289–1290 Small fiber polyneuropathy 774 Small tyrosine kinase inhibitors 1205, 1213 Small-cell lung cancers (SCLC) antibodies 1162–1163 brain metastases 1143–1144, 1146, 1150, 1151 Lambert–Eaton myasthenic syndrome (LEMS) 1171 limbic encephalitis (LE) 1168, 1169 subacute cerebellar degeneration (SCD) 1164 subacute sensory neuropathy (SSN) 1170 see also Lung cancer Smallpox vaccine 1552 Small-vessel encephalitis 1577–1578 Smoking 257 Smoldering myeloma (SM) 1031–1032, 1083, 1086, 1088 Snakes see under Venomous bites Sneddon’s syndrome (SS) 1562, 1566–1567 Snoring 253–254 Society of Critical Care Medicine (SCCM) 951 Sodium nitroprusside 165, 165, 1636, 1697 Sodium oxybate 522 Solitary plasmacytoma 1083, 1084, 1089, 1092 extramedullary 1032 SOLVD (Studies of Left Ventricular Dysfunction) trial 111–112, 114, 119 Somatosensory evoked potentials (SSEPs) 1630–1631 cardiac arrest 32–33 Somatostatin 810–811 Somatostatin analogs (SSA) 697, 810–811 Somatropin 810, 811–812 Somatropin agonists 811–812 Somatropin antagonists 812 Sones technique 41 Sorafenib 1213 SOSTM1 gene 532–533 Sotalol 132, 136 South American trypanosomiasis (Chagas disease) 1413–1414 SPARCL (Stroke Prevention by Aggressive Reduction in Cholesterol Levels) trial 105 Sparganosis 1427–1428 clinical manifestations 1428 diagnosis 1428 epidemiology 1427 pathogen 1427–1428 treatment 1428 Speech apraxia 1272 Spiders see under Venomous bites Spinal arachnoiditis 1450 Spinal cord disorders 247–248, 1331–1332, 1616 compression 964, 1186, 1186

I40 Spinal cord disorders (Continued ) ischemia see Cerebral/spinal cord ischemia leukemias 1040 lymphomas 1035–1036 myelopathy 964 neurocysticercosis 1451–1453 neurosyphilis 1465, 1465 sarcoidosis 312 tropics see Tropical myelopathy tuberculosis (TB) 1495–1496 Spinal cord injury (SCI) 794, 796, 1761–1767 clinical evaluation 1762–1763, 1764 epidemiology 1761 etiology 1521, 1522 imaging 1763–1764, 1765 management 1764–1767 acute 1764–1766 surgical 1766–1767 pathophysiology 1761–1762 without radiographic abnormality (SCIWORA) 1761 Spinal epidural abscess 1532, 1532 Spinal epidural metastases 340–343 incidence/clinical features 340–341, 341 treatment 341–343 Spinal fractures, acute 455 Spinal headache 1747 Spinal intramedullary metastases 343 Spinal muscular atrophy (SMA) 277 Spinal neurocysticercosis 1538 Spinal stenosis 541–550 causes 542 classification 541 definition 541 diagnosis 543–546 differential 545–546 neurophysiology 545 physical examination 543–544 symptoms 543 epidemiology 541–542 historical perspective 541 imaging 544–545 computed tomography (CT) 544–545, 544 magnetic resonance (MRI) 545, 545 radiographs 544 management 546–547 operative 546–547, 547, 548 nonoperative 546 outcome 547–548 pathophysiology 542–543 claudication 542–543 myelopathy 543 narrowing/stenosis 542, 542 spondyloarthropathies 457 Spinal syndromes, fungal infection 1386 Spinal vascular syphilis 1534 Spine Injury Study Group 1761–1762 Spine, Paget’s disease of bone (PDB) 531, 535 Spinocerebellar ataxia, Machado Joseph’s 794

INDEX Spirochaeta pallida 1461 Spirochetal myelopathy 1532–1535 Spirometra spp 1422, 1427–1428 Spirometra mansoni 1431 Spironolactone 764–765 Spondylitis, tuberculous 1495–1496 Spondyloarthritis, acute myelopathy 1524–1525 Spondylodiscitis 79 Spondyloepiphyseal dysplasia 444 Spontaneous carotid-cavernous fistula 567, 574 Spontaneous meningitis, adults 1369 Sporadic inclusion body myositis (sIBM) see Idiopathic inflammatory myopathy (IIM) Sri Lankan hump-nosed pit viper (Hypnale) 992–993 ST elevation MI (STEMI) 93, 101 ‘Staggers’ 962 Stanford Sleepiness Scale 260 Staphylococcus spp 68 Staphylococcus aureus 1369, 1532, 1532 see also Infective endocarditis Staphylococcus epidermidis 67 STARFlex closure device 113 Starr–Edwards valves 65 ‘Starry sky’ appearance 1537 Statins 1636–1637, 1638 Status cataplecticus 180–181 Status epilepticus (SE) 8 Stem cell transplantation (SCT) 1009, 1035 Stereotactic biopsy 1149 Stereotactic radiosurgery (SRS) 1189, 1191–1192 brain metastases 338–339, 1151–1152 Steroidogenesis, impaired 756–757 Steroids 1173, 1513–1514, 1628–1629, 1676 adverse effects 601, 1201, 1246, 1647 bacterial meningitis 1369–1370, 1371 cerebral vasculitis 479, 483 giant cell arteritis (GCA) 229, 481 neuro-Behc¸et syndrome (NBS) 1719–1720 transplantation 1231, 1268, 1274 tuberculosis (TB) 1490–1493 Stiff man syndrome (SMS) 352, 410–411, 614 ‘Stocking and glove ’ pattern 774 STOP (Stroke Prevention Trial in Sickle Cell Anemia) study 1018, 1020, 1021–1022 STOP II 1020 Streptococcus spp 1532 Streptococcus agalactiae 1361–1362, 1367 Streptococcus bovis 77, 81 Streptococcus pneumoniae 1367, 1369, 1777 bacterial meningitis 1361–1362, 1369, 1372, 1373 hemolytic uremic syndrome (HUS) 1114, 1114, 1115, 1120 Streptococcus suis 1369 Streptococcus viridans 79, 81 Streptokinase 104

Streptomycin (SM) 1490, 1491, 1492 Streptozotocin 1208–1209 Stress cardiomyopathy (Takotsubo cardiomyopathy/apical ballooning syndrome ) 6, 20, 22, 22, 26, 115–117 ‘String of beads’ appearance 1650 Stroke aortic surgery 234 arrhythmia 129–130 arterial ischemic 405–407, 408–409 cardiac surgery 54–55 coronary artery bypass grafting (CABG) 93, 198 developing world 1777–1778 dialysis patients 400 electrophysiologic procedures 142 essential thrombocythemia (ET) 1076 external cardioversion 139–140 heart transplantation 1233, 1235 hypertension 165 hyperthyroidism 723–724 obstructive sleep apnea syndrome (OSAS) 258 pregnancy see under Pregnancy primary myelofibrosis (PMF) 1078 respiratory failure (RF) 275, 275 sickle cell disease (SCD) 1015–1016 sudden death after 23 syndromes, fungal infection 1386 valvular surgery 199–200 venous thromboembolism (VTE) 291–292 see also Acute ischemic stroke; Acute myocardial infarction (AMI); Arterial ischemic stroke (AIS), risks; Cardiomyopathy Stroke Data Bank 129 Strongylida hookworms 1432–1433 Strongyloides 1287–1288 Strongyloides stercoralis 1435–1436 clinical manifestations 1435 diagnosis 1435–1436 epidemiology 1435 hyperinfection syndrome 1429 pathogen 1435 therapy 1436 Sturge–Weber syndrome (SWS) 1562, 1570–1571 clinical manifestations 1570–1571, 1570 ophthalmologic 1571 radiologic features 1570, 1571 treatment 1571 Subacute cerebellar degeneration (SCD) 1160, 1160, 1163–1165 autoantibodies 1164–1165 clinical/biological characteristics 1163 onconeuronal antibodies (ON-Abs) 1163 CV2/CRMP5-Abs 1164 Hu-Abs 1164 Ri-Abs 1164 Tr-Abs 1161, 1164 VGCC-Abs 1164 Yo-Abs 1163–1164 seronegative 1165

INDEX Subacute combined degeneration (SCD) 590, 919, 929, 934–935, 1542 spinal cord 927 Subacute measles encephalitis 1346, 1348 Subacute sclerosing panencephalitis (SSPE) 1345, 1346, 1346 treatment 1348–1349 Subacute sensory neuronopathy (SSN) 352, 876, 1160, 1160 antibodies 1162–1163 Subarachnoid hemorrhage (SAH) 116, 298, 1629 pregnancy 1602–1604 reversible cerebral vasoconstriction syndrome (RCVS) 1730, 1734 vascular/intensive care neurology 5–6, 6 Subarachnoid neurocysticercosis 1451, 1453 Subclavian steal syndrome (SSS) 175, 198 Subcortical structures breathing 245–247, 247 disease 245–247, 247 Subcutaneous neurofibromas 1581–1582 Subdural hematomas (SDH) 387, 554, 1752, 1753, 1760, 1761 Succinylcholine 1628 Sudden cardiac death (SCD) 19–24 classification 19 clinical manifestations 21–22 electrical conduction abnormalities 21–22, 21 stress cardiomyography 22, 22 definition 19–20 historical perspective 19 pathophysiology/histopathology 20 precipitants 20–21, 20 preventive strategies 23 specific scenarios 22–23 Sudden death in adults 556 in children 556 unexplained, in epilepsy (SUDEP) 8, 22–23 Sufentanil 1626 Suffocation 254 Sulfadiazine 1324–1325, 1325 Sulfamethoxazole see Trimethoprim/ sulfamethoxazole (TMP-SMX, TMP-SMZ) Sulfonamides 818, 1646 Sulfonylureas (SU) 816, 820, 1654 Sunitinib 1213 Superficial siderosis (SS) 861 Superficial venous thrombosis (SVT) see Venous thromboembolism (VTE) Supraventricular arrhythmias 153 Supraventricular tachycardia 130 Suramin 1200, 1204, 1652 Surgery, cardiac see Cardiac surgery/ interventions Susac syndrome 1717

Sustained Treatment of Paroxysmal-AF trial 157 Sweet disease (SD) 1717 SWITCH (Stroke with Transfusions Changing to Hydroxyurea) trial 1021–1022 Sydenham’s chorea 1780 SYDNEY 2 (Symptomatic Diabetic Neuropathy) trial 779 Sympathetic outflow tracts, central nervous system (CNS) 4 Symptomatic hydrocephalus 345 Synacthen test 759 Synchronized intermittent mandatory ventilation (SIMV) 279 Synchronous presentation 1144 Syncope see Transient loss of consciousness (TLOC)/syncope Syndrome of inappropriate secretion of antidiuretic hormone (SIADH) 842, 843, 1486 hypernatremia 372 hyponatremia 365, 367–369, 368 Syphilis aortitis 232–233 clinical manifestations 1464 meningovascular 1464 see also Neurosyphilis Syphilitic meningomyelitis 1464–1465, 1534 Syringobulbia-myelia 557, 557 Systemic amyloidosis, clinical presentation 1037 Systemic disorders, nephrotic syndrome 407 Systemic inflammatory response syndrome (SIRS) 650 Systemic lupus erythematosus (SLE) 442, 463–468 antiphospholipid syndrome (APLS) 466 incidence 464 management 467–468 neurologic manifestations 464 neuropsychiatric syndromes see Neuropsychiatric systemic lupus erythematosus (NPSLE) Systemic Lupus International Collaborating Clinics (SLICC) 464 Systemic sclerosis (SSc) see Scleroderma Systemic vasculitis 458

T Tabes dorsalis 1465, 1533–1534, 1534 Tacrolimus 675, 676, 1275 neurotoxicity 1245, 1269–1270, 1271, 1278–1279 central nervous system (CNS) 1281–1282 small bowel transplantation 1287–1288 transplantation 1231, 1260, 1268, 1274, 1280 Tadalafil 782, 1654 Taenia spp 1428 Taenia multiceps 1428

I41 Taenia solium (pig tapeworm) 1422, 1536–1537, 1777 see also Neurocysticercosis (NCC) Taeniasis 1445 Taipan (Oxyuranus) 992 Takatsuki syndrome see POEMS syndrome Takayasu disease 477–479 classification 476, 478 clinical features 478–479, 478 differential diagnosis 479 prognosis 479 treatment 479 Takotsubo cardiomyopathy see Stress cardiomyopathy Tamoxifen 1152–1153, 1201, 1212, 1655 Tapeworms 1414, 1422–1428 fish see Diphyllobothrium latum pig see Taenia solium Tardive dyskinesias (TD) 635, 638 Targeted therapies 1136–1137, 1204–1205, 1212–1213 molecular 339–340, 1152–1153 Taxanes 1135, 1152 adverse effects 1135–1136, 1202, 1210 Tegaserod 638 Telangiectasias, mucocutaneous 1571–1572, 1571 Telbivudine (Tyzeka®) 676, 680 Temozolomide 339, 1184–1185, 1209 Temperature see Body temperature Temporal arteritis see Giant cell arteritis (GCA) Tenatoprazole 638–639 Tenecteplase 31, 104 Teniposide 1203, 1210–1211 Tenoxicam 965 10/66 group, dementia 1778–1779 Teriparatide 813, 814, 879 Terminal arbor degeneration 1202 Tertiary care, developing world 1774 Tertiary hyperparathyroidism 742–743, 744 Testicular germ cell tumors 787 Testis see Ovaries/testis, disease of Tetanus 1506–1509 clinical findings 1506–1508, 1507 complications 1508 differential diagnosis 1508 immunization 1509 diphtheria-tetanus-pertussis (DTP) vaccine 1358, 1550, 1555–1556, 1651 management 1508–1509 autonomic dysfunction 1509 intensive care unit (ICU) 1509 resource poor settings 1508–1509 natural history 1508 prophylaxis 1509 Tetany 377, 377 Tetracycline 625, 1337, 1468–1469, 1646 Tetrahydrofolate (THF) 917, 928 Tetratiomolybdate 856 Thalidomide 1136, 1513–1514 adverse effects 1200, 1204, 1212, 1652 multiple myeloma (MM) 1032, 1089, 1090

I42 Thalidomide-induced peripheral neuropathy (TiPN) 1090 Theophylline 1655 Thermal injuries 981–986 clinical findings 981–982 future directions 985 historical perspective 981 laboratory investigations 982–983 electrocardiography (ECG) monitoring 982, 983 myoglobinuria 982–983 management 983–985 excision/wound care 984–985 fluid resuscitation 983–984 myoglobinuria clearance 984 rehabilitation 985 trauma, associated 984 natural history 982 neuroimaging 983 pathology 983, 983 Thermogard VP® system 954 ThermoSuit® system 954 Thiamazole 813 Thiamin see Vitamin B1 deficiency Thiazide diuretics 745 Thiazolidinediones 819 Thienopyridine therapy 101 Thimerosal (thiomersal) 1556 Thiobarbiturates 1624 Thionamides 813–814 6-Thiopurines 1134 Thiopental 31, 1624, 1625 Thiotepa 1201, 1209 Thor inhibitors, heart transplantation 1231 Thoracic aortic aneurysms 225–226 Thoracic-lumbar-sacral orthotic (TLSO) 1767 Thoracoabdominal aortic aneurysm (TAAA) 226, 227, 233–234, 233 Thrombin inhibitors 139, 295 Thromboembolic stroke 964 Thromboembolism, catheter ablation atrial fibrillation (AF) 151–153, 152 during 154, 155, 156 supraventricular/ventricular arrhythmias 153 Thromboembolism, venous see Venous thromboembolism (VTE) Thrombolysis acute coronary syndrome (ACS) 95, 103–104, 104 acute ischemic stroke 86 acute myocardial infarction 44–45, 45 Thrombolysis In Myocardial Infarction (TIMI) study 101–103 Thrombolytic drugs, adverse effects 1637–1638 Thrombophilic states 1061–1072 arterial ischemic stroke (AIS), risks 1066–1068 acquired thrombophilia 1067 children 1067–1068 inherited thrombophilia 1066–1067 mixed thrombophilia 1067 paradoxical embolism 1068

INDEX Thrombophilic states (Continued ) cerebral venous sinus thrombosis (CVST), risks 1064–1066 acquired thrombophilia 1065 children 1065–1066 epidemiology/symptoms 1064 inherited thrombophilia 1064–1065, 1064 mixed thrombophilia 1065 definition 1061 venous thromboembolism (VTE), risks 1061–1064, 1062 acquired thrombophilia 1063 inherited thrombophilia 1061–1063 lipoprotein(a) 1064 mixed thrombophilia 1063–1064 Thromboprophylaxis 294–296, 295, 299–300 Thrombosis, polycythemia vera (PV) 1075 Thrombotic microangiopathy 1113 Thrombotic thrombocytopenic purpura (TTP) 1005, 1113, 1119 epidemiology/etiology 1114–1115 hematologic disorders 1129 treatment 1120 Thromboxane synthase inhibitors 1129–1130 Thrombus 111–112 Thunderclap headache 1650 see also under Reversible cerebral vasoconstriction syndrome (RCVS) Thyroid disease 703–735 miscellaneous disorders 726 ophthalmopathy see Graves’ ophthalmopathy see also Hyperthyroidism; Hypothyroidism Thyroid hormones 812–813, 813 Thyroid storm 721–722 Thyroid-stimulating hormone (TSH) 686 acromegaly 691–692 central hypothyroidism (CH) 689 excess 693 -producing adenomas 693 Thyrotoxic hypokalemic periodic paralysis (TPP) 719–721 clinical features 719–720 neurologic features 719 non-neurologic features 719–720 diagnosis 720 management 720–721, 720 emergency treatment 721 prevention 721 underlying hyperthyroidism 721 pathophysiology 720 Thyrotropin (TSH) see Thyroid stimulating hormone (TSH) Thyroxine 711 Tiabendazole 1433, 1435 Tiagabine 846 adverse effects 1640, 1640 elimination 418, 425, 426, 427 Ticagrelor 1129 Tickborne encephalitis virus 1378 diagnosis 1380

Ticlopidine (Ticlid®) 1129, 1639 Tiger snake (Notechis) 992 Tilt-table testing 517 Tinel’s sign 1510 Tinzaparin 299 Tipifarnib 1205 TIPSS (transjugular intrahepatic portosystemic shunt) 662 Tirofiban 1639 Tityus 988, 993–994 Tityus serrulatus 993–994 Tizanidine 1332, 1640 TNM (tumor-nodes-metastasis) system 335 TO-ACT (Thrombolysis Or Anticoagulation for Cerebral Venous Thrombosis) trial 1605–1606 Tocainide 132, 136 Tocilizumab 459, 479, 1719 Tolvaptan 810, 812 Topiramate 1640, 1640 elimination 418, 424, 425, 427 Topoisomerase inhibitors 1135, 1204, 1212 Topotecan 1212 Tositumomab (Bexxar®) 1136 Total body irradiation (TBI) 1295–1296, 1299, 1300 Total iron binding capacity (TIBC) 1008 Total parenteral nutrition (TPN) 1286, 1305–1307 Tourette’s syndrome, management 1644 Toxic myeloneuropathy 1544–1545 Toxocara canis/cati 1431, 1434–1435, 1538, 1539 clinical manifestations 1434–1435 diagnosis 1435 epidemiology 1434 pathogen 1434 therapy 1435 Toxocariasis 1539 Toxoplasma gondii 1232, 1248 see also Toxoplasmosis entries Toxoplasmosis, HIV-associated 1323–1326 clinical manifestations 1323 human infection 1323 laboratory diagnosis 1324 neuroimaging 1324, 1324 prophylaxis 1325–1326, 1325 Toxoplasma encephalitis 1324 treatment 1324–1325, 1325 response 1325 Toxoplasmosis, renal transplantation 1248, 1248, 1252 Tr-Abs (antibodies), subacute cerebellar degeneration (SCD) 1161, 1164 Tracheobronchial secretions 283 Training, developing world 1774–1775 Train-of-four muscle twitch assessment 30–31 Tramadol 519, 522, 780–781 Tranexamic acid (AMCA) 1128 Trans-4-hydroxy-2-nonenal (HNE) 831

INDEX Transcranial Doppler (TCD) monitoring 155 Transesophageal echocardiography (TEE) 7–8, 52, 139–140 Transfusion, reactions/risks 1127–1128 Transfusion-related acute lung injury (TRALI) 649–650, 1127–1128 Transient cortical blindness 44, 44, 197 Transient ischemic attacks (TIAs) cardiomyopathy 111, 113–114, 118–119 dialysis patients 400 essential thrombocythemia (ET) 1075–1076 heart transplantation 1233 infective endocarditis 67 neurosarcoidosis 311 polycythemia vera (PV) 1074–1075 primary myelofibrosis (PMF) 1078 syncope 175 vertebrobasilar 181 Transient loss of consciousness (TLOC)/ syncope 169–193 causes 170–181, 170 medical 180 neurologic 176, 177, 179–180 syncopal, cerebral hypoperfusion 170–176 nonsyncopal 176–181, 179 definitions 169 diagnostic workup 184–186 clinical history 179, 185 investigations 185–186 physical examination 185 differential diagnosis 181–184, 181 ‘absences’/‘absent-mindedness’ 184 ‘drop attacks’ 181–184 epidemiology 169–170 recognition 179 treatment 186–188 syncopal 186–187 nonsyncopal 170, 187 driving restrictions 187–188 Transient unresponsiveness in elderly 183 Transplantation acute liver failure (ALF) 649 hemolytic uremic syndrome (HUS) 1120 see also specific organs Trastuzumab 1152–1153, 1205 Trauma acute myelopathy 1524 see also Neurotraumatology Traumatic brain injury (TBI) 6–7, 298, 688, 1751–1761 bacterial meningitis 1372 clinical evaluation 1754–1755, 1754 developing world 1780 epidemiology 1751–1752, 1752 imaging 1755–1757, 1755, 1756, 1764 Lund concept of pathophysiology-based management 1753 management 1757–1760 acute medical 1757–1758 intracranial hemorrhage (ICH) 1759

Traumatic brain injury (TBI) (Continued ) intracranial pressure (ICP), increased 1758–1760 surgical guidelines 1760 neuroanesthesia 1628 outcomes 1760–1761 pathophysiology 1752–1754 Traumatic craniovertebral subluxation 438, 438, 439 Traumatic transient loss of consciousness (TLOC) 181 Trazodone 1642 Trematodes (flukes) 1414, 1419–1422 Tremor 715 Treponema 1532 Treponema pallidum see Neurosyphilis Tretinoin 1212–1213 Triazoles 1649 Trichinella spp 1433–1434 Trichinellosis 1433–1434 clinical manifestations 1434 diagnosis 1434 pathogen 1434 therapy 1434 Trichoderma polysporum 1299–1300 Triclabendazole 1422 Tricyclic antidepressants (TCA) adverse effects 1642 diabetic neuropathy 780, 781, 781 fibromyalgia 519–520, 519 Trientine 856, 857 Triethylenetetramine 856–857 Trigeminal nerve (cranial nerve V) 307 Trigeminal neuralgia, radiotherapy (RT) 1189, 1190 Trimeresurus complex 992–993 Trimethoprim 1646 Trimethoprim-sulfamethoxazole (TMPSMX, TMP-SMZ) 628, 1246, 1247, 1248, 1532 toxoplasmosis 1323, 1325, 1325 Triple A (Allgrove) syndrome 756 Triple-symptom complex 1703 Triptans 8, 1274, 1571, 1651 TRITON-TIMI 38 (Trial to Assess Improvement in Therapeutic Outcomes by Optimizing Platelet Inhibition with PrasugrelThrombolysis in Myocardial Infarction) trial 101 Trochlear nerve (cranial nerve IV) 307 Troglitazone 819 TROICA (Thrombolysis in Cardiac Arrest) trial 31 Tropheryma whipplei 80–81, 627, 628 Tropical myeloneuropathy 1521–1522 ataxic neuropathy (TAN) 1542–1543 historical perspective 1523 nutritional 1540–1544 toxic 1544–1545 Tropical myelopathy 1521–1548 acute myelopathy, causes 1522, 1524–1525 disc herniation/spondyloarthritis 1524–1525

I43 Tropical myelopathy (Continued ) trauma 1524 vascular 1525 clinical aspects 1524 etiologies 1522 historical perspective 1522–1524, 1523 immune-inflammatory disorders 1540 acute transverse myelitis 1540 multiple sclerosis (MS) 1540 infectious myelopathy 1522, 1525–1540 bacterial 1530–1532 chronic retroviral 1529–1530 fungal 1535 parasitic 1535–1540 spirochetal 1532–1535 vacuolar see under HIV-infected patients viral 1527 nutritional myeloneuropathy 1540–1544 toxic- 1542–1544 Vitamin B12 see vitamin B12 (cobalamin (Cbl)) deficiency toxic myeloneuropathy 1544–1545 cyanide 1544–1545 fluorosis 1547–1546 lathyrism 1547–1546 Tropical (nutritional) amblyopia 1543 Tropical sprue (TS) 625 Tropisetron 522, 636 Trousseau’s sign 377, 743 Trypanosoma 1777 Trypanosoma brucei 1412–1413 Trypanosoma brucei gambiense 1412–1413 Trypanosoma brucei rhodesiense 1412–1413 Trypanosoma cruzi 1407, 1413–1414 TSC1 gene 1584 TSC2 gene 1584 Tsetse flies 1412 Tuberculoid tuberculoid leprosy 1573–1575 Tuberculomas (tuberculous granulomas) 1149, 1492–1494, 1493 Tuberculosis (TB), central nervous system (CNS) 1485–1499 clinical findings 1486–1496 developing world 1777 etiologic agent 1485 HIV see under HIV-infected patients pathogenesis 1485–1486, 1486 renal transplantation 1247–1248, 1252 spinal cord 1530, 1530, 1531 Tuberculosis-associated immune reconstitution inflammatory syndrome (TB-IRIS) 1495 Tuberculous abscess 1494, 1494 Tuberculous meningitis (TBM) 1486–1492 associated features 1487 cerebrospinal fluid (CSF) findings 1488, 1488 complications 1489–1490 diagnosis 1488–1489 management 1490–1492, 1491 multidrug-resistant (MDR) meningitis 1491, 1492 neuroimaging 1489, 1489

I44 Tuberculous meningitis (TBM) (Continued ) prognosis 1492 signs/symptoms 1487 Tuberculous myelitis 1495 Tuberculous spondylitis 1495–1496 Tuberous sclerosis complex (TSC) 1562, 1584–1586 clinical symptoms 1584–1585 cutaneous 1584, 1585 neurologic 1584–1585 diagnosis/genetic testing 1586, 1586 Tumor necrosis factor (TNF) blockade 453 Tumor necrosis factor-a (TNF-a) 507 inhibitors 481 Tumors brain see Brain tumors cardiac see Cardiac tumors central nervous system (CNS) 1250–1251, 1263 flare syndrome 1209 identification 1172 neurologic 1582–1583, 1583, 1584 ovarian 789, 791–792 pituitary see Pituitary tumors primary 1143–1144 radiotherapy (RT) 1184–1189 resection 1149 spinal cord 1616 TNM (tumor-nodes-metastasis) system 335 Turner syndrome, genetics 53 Tyrosine kinase inhibitors 1205, 1213, 1652

U Ulcerative colitis (UC) see Inflammatory bowel diseases (IBD) Ulcers genital 1704 oral 1704 Ultrasound 1733 venous duplex (VDU) 291, 293, 294, 300 UMOD gene 827–829 Undersea and Hyperbaric Medical Society 156 Unfractionated heparin (UFH) 137, 137, 1077, 1130, 1131, 1132 adverse effects 137, 137, 1638 hematologic disorders 1077, 1130, 1131, 1132 intracranial hemorrhage (ICH) 297, 298–299 venous thromboembolism (VTE) 295, 296–297, 300 Unilateral lower compartment syndrome 592 United Kingdom Prospective Diabetes Study (UKPDS) 773–774, 778–779 United Network for Organ Sharing (UNOS) 1238 Unstable angina (UA) 93, 99–100 Unsteady gait 1145 UPB1 gene 835 Upper extremity mononeuropathy/ radiculopathy 1608–1609 Urapidil 1697

INDEX Urea derivatives 818 Urea transporters (UTs) 396–397 Ureidopropionase deficiency 834, 835 Uremic encephalopathy 383–385, 384 acute vs chronic 383–384 clinical symptoms 384, 385 diagnosis 384–385 dialysis patients 395–396 pathogenesis 384 therapy 385, 385 Uremic myopathy, renal disease 391 Uremic polyneuropathy (PNP) 388–390 clinical picture 389 diagnosis 389 pathogenesis 389 therapy 389–390 Uridine-5-monophosphate (UMP) 827, 833–834 synthase 833 Urtoxazumab 1120 US Navy Diving Manual 962, 965 US Navy Treatment Table 6 protocol 156 Uveo-parotid fever 305 Uvulopalatopharyngoplasty (UPPP) 262

V Vaccinations 1549–1557 bacterial vaccines 1554–1556 viral vaccines 1550–1554 mechanisms of action 1550, 1550 Vaccine Adverse Events Reporting System (VAERS) 1549, 1551, 1553, 1554, 1556 Vaccine Safety Datalink (VSD) 1549, 1553 VACTERL (vertebral anomalies, imperforate anus, cardiac anomalies, tracheoesophageal fistula, renal anomalies, limb anomalies) 53 Vacuolar myelopathy (VM) see under HIV-infected patients Vagus nerve (cranial nerve X) 308 Valaciclovir 1380–1381 Valdecoxib 580 Valganciclovir 1232, 1331 VALIANT (VALsartan In Acute myocardial iNfarcTion) trial 94, 100 Valproic acid (valproate) 846, 847, 1324–1325 adverse effects 1640, 1640 brain metastases 337, 1153 elimination 418, 420–421, 425, 427 Valsalva maneuver 51–52, 1738 Valvular heart disease 61–74 anticoagulation, complications 65–66 calcific 62–63 infective endocarditis see Infective endocarditis mitral valve prolapse (MVP) 63 noninfective endocarditis 69–70 prosthetic heart valves 63–65 bioprosthetic 64–65 mechanical heart valves 65 rheumatic (RVHD) 61–62 Valvular surgery 194, 195, 199–200

Vancomycin 1369, 1371, 1379–1380 Vapreotide 810 Vardenafil 782, 1654 Varicella zoster virus (VZV) encephalitis 1377, 1378, 1379 diagnosis 1380 treatment 1381 infection heart transplantation 1232 mononeuropathy 1333–1334 renal transplantation 1249 treatment 1380 vaccine 1550, 1551–1552, 1651 Vascular compression theory 543 Vascular conditions acute myelopathy 1525 dementia (VaD) 9, 931–932, 1778 endothelial dysfunction 1595, 1596–1598 hemostasis 1596 leukemias 1040–1041 lymphomas 1037–1038 neurology see Cardiology, vascular/ intensive care neurology neuropathy 310–311, 311, 451 pattern reaction 1719 pseudoxanthoma elasticum (PXE) 1568 ‘steal’ syndrome 877–878 zoster 1569 Vascular Ehlers–Danlos syndrome (vEDS) 565–568 description 565–567, 566, 567 diagnostic criteria 567 neurologic complications 567–568 Vasculature in Uppsala Seniors, perspective investigation (PIVUS) 115 Vasculitis 598–599 see also Cerebral vasculitis Vasopressin 812 Vasopressin analogs 812 Vasopressin antagonists 812 Vasospasm, neurocardiogenic injury 20 Vasovagal syncope 174, 175 Velocardiofacial syndrome, genetics 52, 53 Venereal Disease Research Laboratory (VDRL) 1334–1336, 1533–1534 Venezuelan equine encephalitis virus 1378, 1380 Venlafaxine 520, 780, 1090 Venomous bites 987–1003 historical perspective 987 insect stings 995 clinical manifestations/treatment 995 natural toxins 996 pathophysiology 988–990 nervous system, effects 988–989 neurotoxins 989–990, 990 scorpions 988, 993–994 clinical manifestations 994 management 994 snakes 988, 990–993 elapidae 987, 991–992 epidemiology 990–991 management 996–997

INDEX Venomous bites (Continued ) viperidae 987, 992–993 spiders (Araneae) 988, 994–995 clinical effects 994–995 treatment 995 tick paralysis 988, 995–996 venom 987–988, 988 apparatus 987 Venous duplex ultrasonography (VDU) 291, 293, 294, 300 Venous emboli 51 Venous sinus thrombosis (VST) 1041 Venous thromboembolism (VTE) 289–304 epidemiology 289–292, 290 general principles 292–300 presentation/diagnosis 292–294, 294 risks 1061–1064, 1062 treatment issues 294–296 acute conditions 296 neurologic/neurosurgical disease 296–300 screening 300 thromboprophylaxis 294–296, 295, 299–300 Ventilators, weaning from 1240 Ventricula neurocysticercosis 1451 Ventricular arrhythmias, catheter ablation 153 Ventriculitis 1577–1578 Verapamil 132, 136, 1636, 1737–1738 Verotoxin 1115, 1117–1118 Vertebral artery dissection, decompression illness (DCI) 964 Vertebral canal stenosis 557, 558–559, 558 Vertebral column malalignment 559, 559, 560 Vertebral hemangiomas 1616 Vertebrobasilar transient ischemic attacks (TIAs) 181 Vertebroplasty, complications 1747–1748, 1748 Very long chain fatty acids (VLCFA) 759–761, 762 Very low density lipoproteins (VLDLs) 623–624 Vespa 995 Vespula vulgaris 995 Vestibular syncope 181–182 Vestibulocochlear nerve (cranial nerve VIII) 308 Veterans Affairs Cooperative Study Group on Antihypertensive Agents 161 Cooperative Study on Valvular Heart Disease 64 Diabetes Trial (VADT) 778–779 Normative Aging Study (USA) 931 VGCC-Abs (antibodies) 1161, 1164 VGKC-Abs (antibodies) 1161 limbic encephalitis (LE) 1166, 1167 Vigabatrin 846, 1584–1585, 1640 elimination 418, 425, 426–427, 427 Vildagliptin 817, 820 Vinblastine 1201–1202

Vinca alkaloids 1089, 1135, 1152 adverse effects 1200, 1201–1202, 1210 Vincristine 1089, 1090, 1184–1185, 1201–1202 Vindesine 1201–1202 Vinorelbine 1201–1202 Violin spiders 994 Viperidae 987, 992–993 Viperinae 992–993 Viral encephalitis developing world 1777 serology/CSF studies 1380 Viral infection, central nervous system (CNS) 1249–1250 Viral myelopathy 1527 Viral vaccines 1550–1554 Virchow, Rudolf 93 Visual evoked potentials (VEP) 315, 316 VITACOG trial 934 Vitamins absorption 622 adverse effects 1654–1655 see also Hydrosoluble vitamins Vitamin A deficiency, short bowel syndrome 1286 Vitamin B1 (thiamin) deficiency 891, 896–901 bariatric surgery 589 biochemical function 896 clinical findings 896–898 genetics 898 laboratory investigations 894, 898 management 900–901 natural sources 896 neuroimaging 898, 899 overview 892 pathology 898–900 toxicity 898 Vitamin B1 (thiamine) deficiency, short bowel syndrome 1286 Vitamin B2 (riboflavin) deficiency 901–902 chemical function 901 clinical findings 901–902 genetics 901, 902 headache 1274 laboratory investigations 894, 902 management 902 natural sources 901 neuroimaging 902 overview 892 pathology 902 toxicity 902 Vitamin B3 (niacin/nicotinic acid) deficiency 891, 902–904 bariatric surgery 591 biochemical function 902 clinical findings 903 genetics 903–904 laboratory investigations 894, 903 management 904 natural sources 902–903 neuroimaging 903 overview 892 pathology 904

I45 Vitamin B5 (pantothenic acid) deficiency 904–905 clinical findings 905 laboratory investigations 894, 905 management 905 natural sources 904 overview 892 short bowel syndrome 1286 Vitamin B6 (pyridoxine/pyridoxal) deficiency 779, 905–908 biochemical function 905 clinical findings 906–907 genetics 907 homocysteine concentration 931, 934 laboratory investigations 894, 907 management 907–908 natural sources 906 neuroimaging 907 overview 892 pathology 907 Vitamin B7 (biotin) deficiency, short bowel syndrome 1286 Vitamin B8 (biotin) deficiency laboratory investigations 894 overview 892 Vitamin B9 (folate) deficiency see Folic acid deficiency Vitamin B12 (cobalamin (Cbl)) deficiency 915–926, 1541–1542 absorption 1541 bariatric surgery 591 causes 588, 591–592 clinical manifestations 592 coenzyme form 1541 diagnosis 1542 epidemiology 1541 functions/kinetics 916–917, 916 hematologic disorders 1125–1126 hemolytic uremic syndrome (HUS) 1120 historical perspective 1541 investigations 592 megaloblastic anemia 1007 neurologic manifestations 1541–1542 neuropathology 1542 pathogenesis 1541 requirements/sources 915–916 short bowel syndrome 1286 terminology 915 treatment 1542 see also Folic acid deficiency Vitamin C (ascorbic acid) deficiency 891, 908–910 biochemical function 908 clinical findings 908–909 genetics 909 laboratory investigations 894, 909 management 909–910 neuroimaging 909 overview 892 pathology 909 Vitamin D deficiency bariatric surgery 591 calcium 865 central nervous system (CNS) 879–882 disorders 881–882 fibromyalgia 523

I46 Vitamin D deficiency (Continued ) homeostasis 737–738 hypercalcemia 741–742, 865–866 hypocalcemia 377, 742–743, 744, 745, 870 hypoparathyroidism 871–872 neurogenesis 880 production/presence 879–880 short bowel syndrome 1286 Vitamin E deficiency 623 short bowel syndrome 1286 Vitamin K treatment 1131–1132 Voglibose 818, 819 Vogt–Koyanagi-Harada syndrome (VKH) 1717 Volume control ventilation (VCV) 279 von Gierke disease 833 von Hippel-Lindau disease 765 von Recklinghausen’s neurofibromatosis see Neurofibromatosis type 1 (NF1) von Willebrand disease (VWD) 1045, 1046, 1056–1057 clinical presentation 1047, 1056 diagnosis 1056–1057 treatment 1055, 1057 types 1056 Voodoo’ Death (Cannon) 19 Voriconazole 1248, 1329–1330

W W198X gene 841 Waldenstr€om’s macroglobulinemia (WM) (lymphoplasmacytic lymphoma) 1033, 1083, 1085, 1086, 1093–1094 central nervous system (CNS) 1093–1094 clinical presentation 1037 peripheral nervous system (PNS) 1093 Wall motion abnormalities (WMA) 6 Wallenberg’s syndrome 437–438 WAPS (warfarin in the antiphospholipid syndrome) study 1067 Warfarin 62, 70, 113, 200, 295, 1131 adverse effects 137, 137, 138, 1638–1639 bleeding risk 122–123, 123 cardiomyopathy 118, 120, 121–122 intracranial hemorrhage (ICH) 1131–1132, 1131 valvular heart disease 65, 66 Warfarin-Aspirin Recurrent Stroke Study (WARSS) 112 Wasp stings 995 WATCH (Warfarin and Antiplatelet Therapy in Chronic Heart Failure) trial 120 WATCHMAN® (Left Atrial Appendage Closure Technology) device 143 Water restriction strategy 368–369 Waterhouse–Friederichsen syndrome 1363 Watershed cerebral infarction 131 Weakness muscle 184, 1252, 1357 see also Intensive care unit-acquired weakness (ICUAW), generalized

INDEX Wegener disease (ANCA-associated granulomatous vasculitis) 483–484 clinical features 484 Wells criteria, venous thromboembolism (VTE) 293–294 Wermer’s syndrome see Multiple endocrine neoplasia (MEN), type 1 Wernicke–Korsakoff syndrome 896, 897 Wernicke’s encephalopathy (WE) 386–387, 397 bone marrow transplantation 1299 intestinal transplantation (ITx) 1272 laboratory investigations 898 malabsorption 625–626 management 900 neuroimaging 898, 899 pathology 900 vitamin B1 589, 897, 899 West Haven criteria, encephalopathy 646 West Nile virus encephalitis 1378, 1379 diagnosis 1380 treatment 1381 renal transplantation 1249–1250, 1252 Western equine encephalitis virus 1378 diagnosis 1380 Whipple’s disease (WhD) 627–628 White matter abnormalities, gluten-related diseases (GRD) 612, 612 Whole killed vaccines 1550 Whole-body 18F-FDG PET scan 1149 Whole-brain radiation therapy (WBRT) 337 brain metastases 1150–1151, 1152 prophylactic 340 Widow spiders 994–995 Williams syndrome, genetics 53 Wilson’s disease (WD) 626, 851, 853, 854, 1257, 1780 clinical features 854–855 diagnosis 855–856, 856 physiopathology 854 treatment 856–857 WIN 3 ( Randomized Trial of Interventions to Improve Warfarin Adherence) trial 120 Wohlfahrtia 1436 Wolbachia 1429 Wolff–Parkinson–White (WFW) syndrome 130 Women with epilepsy (WWE) 1606–1607 Worcester Heart Attack Study 94 World Federation of Neurology (WFN) 1774–1775, 1778 World Headache Alliance 1776 World Health Organization (WHO) bilharziasis 1535 endocrine drugs 809 epilepsy 1775–1776 essential thrombocythemia (ET) 1076 leprosy 1512–1514 monoclonal gammopathy of undetermined significance (MGUS) 1031 multidrug-resistant (MDR) meningitis 1492

World Health Organization (WHO) (Continued ) multiple sclerosis (MS) 1779 myeloid and lymphoid neoplasms 1027, 1029 neurological disorders 1773 osteoporosis 875 paragonimiasis 1422 PNM system 1426–1427 polycythemia vera (PV) 1074 primary myelofibrosis (PMF) 1077–1078 rabies 1504–1506 stroke 1777–1778 syphilis 1468, 1469 tetanus 1506 tuberculosis (TB) 1485 vaccines 1556–1557 World Stroke Organization 1775 Wuchereria bancrofti 1429

X Xanthine oxidase deficiency (hereditary xanthinuria) 828, 833 Xeroderma pigmentosum (XP) 1562, 1579 clinical manifestations 1579 diagnosis/treatment 1579 Ximelagatran 139, 1639 X-linked cutis laxa see Occipital horn syndrome (OHS)

Y Yellow fever vaccine 1550, 1554, 1555 Yellow-bellied sea snake (Pelamis platurus) 992 Yersinia spp 1113–1114, 1114 Yo-Abs (antibodies) 1161, 1163 subacute cerebellar degeneration (SCD) 1163–1164 Yohimbine 781–782 Young Adult Myocardial Infarction and Ischemic Stroke (YAMIS) study 1068 Yttrium-90 (Y-90) 1136, 1189

Z Zidovudine 1340, 1340 Zinc salts 856–857 Ziprasidone 1642 Zivkovic study 1287–1288 Zoledronic acid (zoledronate) 536, 878 Zollinger–Ellison syndrome (ZES) 804–805 Zolpidem 522 Zona see Herpes zoster Zonisamide 1640 elimination 418, 425, 427–428, 427 Zopiclone 522 Zoster vaccine, adverse effects 1651 see also Herpes zoster (zona/shingles); Varicella zoster virus (VZV) Zygomycetes spp 1385, 1386, 1397 Zygomycosis 1385