throbber

`
`Multiple Sclerosis Therapeutics
`
`Fourth Edition
`
`Edited by
`Jeffrey A. Cohen, MD
`Jmlo: Lx|::rmv:n'.J|
`'t'traL»;-uII:sf'3-gran .‘.-1.P|:':'Le"re't3r‘.’..lIp|e\;’!e'3ss |'.=>.1rvrmr:1'::.'.-:‘<.rn‘ ".:*.n.l-:1«'_;m VNH hyni
`
`-rm" :,f r~.a°~,r.-
`
`:7-Iv:|<»‘.
`
`Riehard A. Rudick, MD
`‘W.“('fl(v"l1lI?3f\"SfP|T'VIC‘???hCw][I‘C"lTi1CHfS(‘:]f:'1."l:J'3|C1QLdl"1LIi1.i.“(‘(‘\t’.llt§:\'|i(',3-PIPfl1I,‘}H ll".-‘~
`
`.. 3'5 CAMBRIDGE
`UNIVERSITY PRESS
`
`
`
`Page 1 0f 16
`
`Biogen Exhibit 2066
`Coalition v. Biogen
`IPR2015—01993
`
`

`

`CAMBRIDGE UNIVERSITY PRESS
`Cambridge, New York, Melbourne, Madrid, Cape Town,
`Singapore, Sao Paulo, Delhi, Tokyo, Mexico City
`
`Cambridge University Press
`The Edinburg Building, Cambridge CB2 8RU, UK
`
`Published in the United States of America by Cambridge University
`Press, New York
`
`www.cambridge.org
`Information on this title: www.cambridge.org/
`978052 I 7662 72
`
`© Cambridge University Press 2011
`
`This publication is in copyright. Subject to statutory exception
`and to the provisions of relevant collective licensing agreements,
`no reproduction of any part may take place without the written
`permission of Cambridge University Press.
`
`First published 2011
`
`Printed in the United Kingdom at the University Press, Cambridge
`
`A catalog recordfor this publication is availablefrom the British Library
`
`Library of Congress Cataloging in Publication data
`Multiple sclerosis therapeutics / edited by Jeffrey A. Cohen,
`Richard A. Rudick. — 4th ed.
`p.
`;
`cm.
`
`Includes bibliographical references and index.
`ISBN 978-0-521-76627-2 (hardback)
`I. Cohen, Jeffrey A. (Jeffrey Alan). 1954-
`III. Title.
`
`II. Rudick, Richard A.
`
`2. Clinical Trials as Topic —
`[DNLM: 1. Multiple Sclerosis - therapy.
`methods.
`3. Magnetic Resonance Imaging.
`4. Multiple Sclerosis ~
`pathology.
`5. Outcome Assessment (Health Care) WL 360]
`LC classification not assigned
`6l6.8'34 — dc23
`2011030483
`
`ISBN 978-0-521-76627-2 Hardback
`
`Cambridge University Press has no responsibility for the persistence
`or accuracy of URLs for external or third-party internet websites
`referred to in this publication, and does not guarantee that any
`content on such websites is, or will remain, accurate or appropriate.
`
`Every effort has been made in preparing this book to provide accurate
`and up-to«date information which is in accord with accepted
`standards and practice at the time ofpublication. Although case
`histories are drawn from actual cases, every effort has been made to
`disguise the identities of the individuals involved. Nevertheless, the
`authors, editors and publishers can make no warranties that the
`information contained herein is totally free from error, not least
`because clinical standards are constantly changing through research
`and regulation. The authors, editors and publishers therefore disclaim
`all liability for direct or consequential damages resulting from the use
`ofmaterial contained in this book. Readers are stronglyadvised to pay
`careful attention to information provided by the manufacturer ofany
`drugs or equipment that they plan to use.
`
`Page 2 of 16
`
`

`

`
`
`l l
`
`Measurement of CNS atrophy
`Elizabeth Fisher
`
`128
`
`Axonal pathology in patients with multiple
`sclerosis: Evidence from in vivo proton
`magnetic resonance spectroscopy
`150
`
`Sridar Narayanan, Zografos Caramanos, Paul M.
`Matthews, and Douglas L. Arnold
`
`Imaging of gray matter lesions in multiple
`sclerosis
`165
`
`Mike P. Wattjcs, Jerocn I.G. Geurts, and Frederik Barkhof
`
`Functional imaging in multiple sclerosis
`Kyle C. Kern and Nancy L. Sicotte
`
`175
`
`Diffusion imaging in multiple sclerosis
`
`186
`
`Stephen
`
`lones and Michael D. Phillips
`
`The use of MRI in multiple sclerosis clinical
`trials
`198
`
`Robert A. Bermel, Elizabeth Fisher, Peter B. lmrey,
`and Jeffrey A. Cohen
`
`Optical coherence tomography to monitor
`axonal and neuronal integrity in multiple
`sclerosis
`213
`
`Kristin M. Galetta and Laura]. Balcer
`
`The process of drug development and
`approval in the United States, the European
`Union, and Asia
`225
`
`Nadine Cohen, Ann Dodds-Frerichs, Tammy
`Phinncy, and Paula Sandler
`
`Selection, interpretation, and development of
`end-points in multiple sclerosis clinical trials
`Marc K. Walton
`
`232
`
`The challenge of demonstrating long-term
`benefit of disease-modifying therapies in
`multiple sclerosis
`244
`Maria Trojano
`
`l6
`
`l8
`
`I9
`
`20
`
`Contents
`
`List ofcontribulors
`Abbreviationslist
`Foreword
`xv
`
`page viii
`xiii
`
`Stephen C. Reingold
`Preface
`xvii
`
`Section I — Introduction
`
`Aspects of multiple sclerosis that relate to
`experimental therapeutics
`1
`Richard A. Rudick and Jeffrey A. Cohen
`
`The pathology of multiple sclerosis
`Ranjan Dutta and Bruce D. Trapp
`
`12
`
`The immunology of multiple sclerosis
`Amit Bar-Or and Peter]. Darlington
`
`20
`
`35
`The genetics of multiple sclerosis
`Iorge R. Oksenberg, Sergio E. Baranzini, and
`Stephen L. Hauser
`
`The epidemiology of multiple sclerosis
`Ruth Ann Marric and Helen Tremlett
`
`46
`
`Section II — Clinical trial methodology
`
`Measures of neurological impairment and
`disability in multiple sclerosis
`56
`
`Gary R. Cutter, Charity J. Morgan, Amber R.
`Salter, Stacey S. Coiield, and Laura J. Balcer
`
`Assessment of neuropsychological function in
`multiple sclerosis
`65
`Stephen M. Rao
`
`Health-related quality of life assessment in
`multiple sclerosis
`79
`Deborah M. Miller, i\/Iichael W. Kattan, and Alex 7.. F11
`
`Ki‘!
`
`6)
`
`Measures of acute and chronic lesions
`
`visualized by conventional magnetic
`resonance imaging
`91
`lack H. Simon and Jerry S. Wolinsky
`
`10
`
`Measures of magnetization transfer
`Massimo Filippi, Ioscph C. McGowan. and
`Maria A. Rocca
`
`112
`
`
`
`Page 3 of 16
`
`

`

`"l
`
`I Contents
`
`
`
`_i
`
`37
`
`38
`
`39
`
`40
`
`Use of immunosuppressants to treat multiple
`sclerosis
`436
`
`James M. Stankiewicz and Howard L. Wciner
`
`intravenous immunoglobulin to treat multiple
`sclerosis
`444
`
`Franz Fazekas, Siegrid Fuchs, Per Soelberg
`Srzirensen, and Ralf Gold
`
`Plasma exchange treatment for CNS
`454
`inflammatory demyelinating disease
`Brian G. Weinshenker, B. Mark Keegan, leffrey L.
`Winters, Ichiro Nakashima, and Kazuo Fujihara
`
`465
`Statins in multiple sclerosis
`Martin S. Weber, Emmanuelle Waubant, and Scott
`S. Zamvil
`
`/ll
`
`42
`
`43
`
`44
`
`45
`
`46
`
`47
`
`48
`
`49
`
`50
`
`T-cell-based therapies for multiple
`sclerosis
`472
`
`Tanuja Chitnis and Samia I. Khoury
`
`B-cell-based therapies for multiple
`sclerosis
`483
`
`Emmanuelle Waubant and Amit Bar-Or
`
`Sex hormones and other pregnancy-related
`factors with therapeutic potential in multiple
`sclerosis
`498
`Rhonda R. Voskuhl
`
`Hematopoietic stem cell transplantation to
`treat multiple sclerosis
`508
`
`Richard K. Burt and Francesca Milanetti
`
`Mesenchymal stem cell transplantation to
`treat multiple sclerosis
`520
`Don Mahad, Sarah M. Planchon, and Ieffrey A. Cohen
`
`Neuroprotection in multiple sclerosis
`Avindra Nath and Peter A. Calabresi
`
`535
`
`Combination therapy in multiple
`sclerosis
`547
`
`Michelle Fabian and Fred D. Lublin
`
`Dalfampridine in multiple sclerosis
`Andrew D. Goodman
`
`557
`
`Complementary and alternative treatments in
`multiple sclerosis
`562
`
`Vijayshree Yadav, Lynne Shinto, and Dennis N.
`Bourdette
`
`The role of chronic cerebrospinal venous
`insufficiency in multiple sclerosis
`574
`Devon Conway, Soo Hyun Kim, and Alexander
`Rae—Grant
`
`21
`
`22
`
`23
`
`24
`
`The growing need for alternative clinical trial
`designs for multiple sclerosis
`253
`Stephen C. Reingold, Henry F. McFarland, and
`A. John Petkau
`
`Ethical considerations in multiple sclerosis
`clinical trials
`261
`
`Aaron E. Miller, Nada Gligorov, and Stephen C. Krieger
`Pharmacogenomics and related
`discovery-driven approaches in multiple
`sclerosis
`272
`
`Paulo Fontoura and David Leppert
`
`Neutralizing antibodies directed against
`biologic agents to treat multiple sclerosis
`Per Soelberg Sorensen
`
`287
`
`25
`
`26
`
`27
`
`28
`
`29
`
`30
`
`31
`
`32
`
`33
`
`34
`
`35
`
`36
`
`Section III — Clinical trials of multiple
`sclerosis therapies
`
`Interferon beta to treat multiple sclerosis
`Richard A. Rudick
`
`300
`
`Glatiramer acetate to treat multiple
`sclerosis
`315
`
`Jenny Guerre and Corey C. Ford
`
`330
`Natalizumab to treat multiple sclerosis
`Chris H. Polman, Ioep Killestein, and Richard A. Rudick
`
`Mitoxantrone to treat multiple sclerosis
`Gilles Edan and Emmanuelle Le Page
`
`344
`
`Cladribine to treat multiple sclerosis
`Gavin Giovannoni and Stuart D. Cook
`
`358
`
`Fingolimod to treat multiple sclerosis
`Ieifrey A. Cohen
`
`370
`
`Dimethyl fumarate to treat multiple sclerosis
`Robert J. Fox and Ralf Gold
`
`387
`
`Alemtuzumab to treat multiple sclerosis
`Orla Tuohy and Alasdair I. Coles
`
`393
`
`Dacllzumab to treat multiple sclerosis
`Iaume Sastre-Garriga and Xavier Montalban
`
`399
`
`Laqulnimod to treat multiple sclerosis
`Douglas R. Ielfery
`
`405
`
`Teriflunomide to treat multiple sclerosis
`Paul W. O'Connor
`
`410
`
`High-dose methylprednisolone to treat
`multiple sclerosis
`418
`Robert]. Fox and R. Philip Kinkel
`
`vi
`
`Page 4 of 16
`
`

`

`51
`
`52
`
`53
`
`54
`
`55
`
`56
`
`Section IV — Therapy in clinical practice
`
`Disease-modifying therapy for multiple
`sclerosis in clinical practice
`583
`Jeffrey A. Cohen and Andrew D. Goodman
`
`Treatment for patients with primary
`progressive multiple sclerosis
`604
`Zhaleh Khaleeli and Alan J. Thompson
`
`Diagnosis, pathogenesis, and treatment of
`neuromyelitls optica (NMO) spectrum
`disorders
`614
`
`Sean J. Pittock
`
`Management of pediatric multiple
`sclerosis
`632
`
`F.. Ann Yeh and Bianca Weinstock-Guttman
`
`Use of MRI in the clinical management of
`multiple sclerosis
`645
`
`I. Theodore Phillips and Lacl A. Stone
`
`Multiple sclerosis-associated fatigue
`Lauren B. Krupp and Dana J. Serafin
`
`654
`
`Contents /I
`
`57
`
`58
`
`60
`
`61
`
`62
`
`666
`Management of spastlcity
`Francois A. Bethoux and Matthew Sutliff
`
`Management of bladder and sexual
`dysfunction in multiple sclerosis
`676
`Natasha Frost, Jessica Szpak, Scott Litwiller, and
`Alexander Rae-Grant
`
`Depression in multiple sclerosis
`Adam I. Kaplin and Ryan E. Stagg
`
`696
`
`Assessment and treatment of pain disorders
`in multiple sclerosis
`707
`Jahangir Maleki and Amy Sullivan
`
`Management of medical comorbidities in
`patients with multiple sclerosis
`714
`John R. Scagnelli and Myla D. Goldman
`
`Rehabilitation in multiple sclerosis
`Francois A. Bethoux and Matthew Sutliff
`
`724
`
`Index
`
`731
`
`Color plates appear between p. 398 and 399
`
`Page 5 of 16
`
` ~
`
`vii
`
`

`

`
`
`w Introduction
`
`Chapter
`
`Aspects of multiple sclerosis that relate to
`experimental therapeutics
`
`Richard A. Rudick and Jeffrey A. Cohen
`
`The purpose of this chapter is to discuss key features of multi-
`ple sclerosis (MS) that relate to clinical trial design or treatment.
`The emphasis will be on aspects of the disease that create chal-
`lenges for developing effective therapies, and for using them in
`practice. These include the subclinical nature of early-stage MS,
`phenotypic and disease heterogeneity, and complexities related
`to measuring disease severity. Many of these topics are covered
`in greater detail throughout the book. This chapter will end with
`a brief discussion about current controversies in the MS exper-
`imental therapeutics field.
`
`Disease features relevant to clinical trials
`
`MS pathology is largely subclinical in early MS
`Relapsing remitting MS (RRMS) patients have periodic relapses
`occurring at variable rates, but generally less than one per year.
`Serial MRI demonstrates many more new lesions than clin-
`ical relapses, with most studies demonstrating a rate of new
`MRI lesions about 10-20-fold higher than clinical relapses.” In
`some patients, MRI shows active disease for years with no clin-
`ical relapses, indicating that MS disease activity may be entirely
`subclinical in some patients during the early stage ofthe disease.
`New inflammatory lesions in white matter begin with gadolini-
`ium (Gd) enhancement, marking sites of inllammatory lesions
`(Chapter 9).5’5 Approximately 50% of untreated RRMS patients
`have Gd—enhancing lesions on a cranial MRI scan obtained
`when the disease is inactive clinically.“ Even the number of
`Gd-positive lesions drastically underestimates disease activity,
`however. First, gray matter pathology is present in MS patients,
`even early in the disease (Chapters 2, II, and I3), and Gd-
`enhancement rarely occurs in gray matter lesions. Second, dis-
`ease in normal appearing white matter is well recognized, and
`correlates with progressive atrophy. Therefore, Gd-enhancing
`lesions, themselves mostly asymptomatic, are just the “tip of the
`iceberg" with respect to MS pathology.
`There are several implications of this for clinical trials. First,
`relapse counts are meaningful clinical outcomes in RRMS, but
`inherently insensitive, and it is difficult to measure clinical dis-
`ability in RRMS patient groups, because RRMS patients don’t
`generally get disabled during the time-frame of a clinical trial.
`Second, insensitivity of clinical outcomes in RRMS drives up
`
`sample sizes, which become prohibitive in active arm designs
`(Chapter 21). 'Ihird, the subclinical nature of disease activity in
`RRMS forms the basis for screening putative therapies using
`MRI outcomes, including MRI parameters as secondary out-
`comes, and potentially using MRI as a primary outcome mea-
`sure in RRMS trials. In that regard, Sormani and colleagues
`conducted a pooled analysis of 23 clinical trials that included
`MRI lesion measurements, to test the effect of interventions
`on lesions and relapse rate.” The effect of the intervention on
`MRI lesions was strongly correlated with the effect of the inter-
`vention on relapses, accounting for over 80% of the total vari-
`ance. This indicates that formation of new lesions in RRMS is
`clinically relevant, and supports the argument that MRI disease
`activity could be used as a primary outcome for RRMS trials.
`While many have advocated for this, use of MRI as a primary
`outcome measure has not achieved regulatory agency accep-
`tance (Chapter 18).
`
`Progressive destructive pathology starts early
`in the disease
`
`The rationale for early intervention in MS is the presence of
`widespread tissue damage at
`the earliest stages of the dis-
`ease."‘ " Once RRMS is established, residual clinical disability
`is usually minimal or absent early in the disease, yet there is
`ongoing tissue damage, as evidenced by accumulation of T2-
`bright MRI lesions,‘ T1-hypointense lesions,” and brain atro-
`phy (Chapter II).”‘"'’ Gray matter lesions are frequent in MS
`autopsy material.” Although these lesions are not visualized
`with standard MRI methods, gray matter atrophy has been doc-
`umented early in the disease.”“9 It is widely believed that the
`ongoing destructive pathology sets the stage for later conver-
`sion to secondary progressive MS (SPMS), in which disability
`accumulates. According to this model, tissue destruction pro-
`ceeds without frank neurological disability progression until a
`threshold is surpassed. Beyond that stage, progressive neurolog-
`ical disability ensues. 'lhe threshold hypothesis was supported
`by studies correlating retinal nerve fiber layer (RNFI.) thickness
`with visual acuity?" Visual acuity was maintained until RNFL
`thickness declined to about 75 microns, and then fell off rapidly
`with further loss of RN FL thickness. The implication of this for
`
`Multiple Sclerosis Therapeutics, Fourth Edition, ed. Jeffrey A. Cohen and Richard A. Rudick. Published by Cambridge University Press.
`© Cambridge University Press 2011.
`
`Page 6 of 16
`
` r
`
`

`

`Section I: Introduction
`
`clinical trials is that disease-modifying drug therapy should be
`viewed as providing secondary neuroprotection, i.e. treatment
`prevents neuroclegeneration by inhibiting inflammation, and
`thereby decreases the amount of brain injury and delays or pre-
`vents SPMS.
`
`Disease manifestations are heterogeneous
`Another factor complicating MS clinical
`trials is disease
`heterogeneity, which is one of the hallmarks of MS. Patients
`manifest varying patterns of clinical features, variable clinical
`course, and variable disease severity. This creates hurdles
`for clinical
`trials,
`as heterogeneity complicates outcomes
`assessment, and increases required sample sizes. The myriad
`clinical manifestations include neuropsychological impairment
`(itself multifaceted), visual loss, eye movement abnormalities,
`weakness, spasticity, incoordination, imbalance, sensory loss,
`paresthesias, gait impairment, bowel and bladder dysfunction,
`sexual dysfunction,
`fatigue, and paroxysmal phenomena.
`Individuals manifest these features in varying combinations,
`and the symptoms change over time. Even within multi-
`ply affected families, there is striking clinical heterogeneity
`between affected family members. Disease heterogeneity is
`poorly understood in MS, as genome—wide association studies
`have mostly focused on disease susceptibility genes, rather
`than disease modifying genes (Chapter 4).
`Managing the wide variety of MS symptoms is crucially
`important for patient well-being, but is increasingly challenging
`with increasing complexity and emphasis on disease modifying
`drug therapy. Heterogeneity in clinical manifestations also
`presents significant challenges for the design of clinical trials.
`Subjects in separate trials and treatment arms within a given
`trial exhibit variable admixtures of clinical manifestations that
`
`are not necessarily evenly matched between study groups. Mul-
`tidimensional clinical outcome measures are needed to capture
`the range of ways in which MS affects patients (Chapter 6). The
`traditional clinical outcome measure — Expanded Disability
`Status Scale (BDSS) - is heavily weighted to motor impairment,
`particularly gait dysfunction. Common symptoms such as cog-
`nitive dysfunction, sphincter disturbances, pain, and fatigue
`have significant effects on functional status and quality of life
`(QOL), but may not correlate well with measures of physical
`impairment and disability. This forms a strong rationale for
`including patient-reported outcomes in clinical
`trials as a
`measure of the impact of intervention on disease-related
`symptoms (Chapter 8). It is possible that therapies may have
`different effects on specific disease manifestations, i.e. benefit
`for some with no effect or deleterious effects on others. This is
`
`an under-explored area.
`
`Evolution of the MS disease process —
`the ”MS categories”
`Because the clinical course of MS evolves over decades, there
`has been interest in subcategorizing MS into discrete groupings.
`
`The current classification system was based on clinical phe-
`nomenology of the clinical disease course.“ not on the under-
`lying biological mechanisms. According to this classification,
`MS begins with a clinically isolated syndrome (CIS), defined
`as an initial clinical episode with features typical for inflam-
`matory demyelination (e.g. optic neuritis, partial transverse
`myelitis). With additional clinical episodes, CIS evolves to clin-
`ically definite RRMS. Even in the absence of a second relapse. a
`patient meets criteria for clinically definite MS when new MRI
`lesions are observed during follow-up.” RRMS then evolves to
`SPMS in many but not all patients. About 15% of patients have
`primary progressive MS (PPMS), meaning that progressive dis-
`ability ensues without prior relapses. In RRMS patients, peri-
`odic relapses occur at irregular and unpredictable intervals,
`averaging approximately one per year, but declining with dis-
`ease duration. Episodic attacks of neurological dysfunction are
`followed by partial or complete recovery, separated by clini-
`cally stable intervals. Relapses become less conspicuous over the
`years, and over 60% of RRMS patients transition to SPMS. Dur-
`ing this stage physical and cognitive disability gradually wors-
`ens, and disease worsening is refractory to known treatment.
`RRMS and SPMS present different challenges in study
`design. In RRMS, relapses are infrequent, occur at irregular
`intervals, and pose significant measurement challenges, and
`disability progression tends to be minimal during the course
`of a clinical trial. There seems to be some “drift” in MS sever-
`
`ity in the direction of more benign disease. This may be driven
`by increased awareness of MS and widespread use of MRI scan-
`ning for patients with non-specific symptoms such as fatigue,
`paresthesias, or headache.” The SPMS stage of the disease is
`also difficult to study, but for different reasons. Deterioration
`occurs slowly over the course of years, and there is significant
`within-patient and between-patient variability. Further, while
`trials tend to restrict patients by disease category, transition
`from RRMS to SPMS does not occur at a precise point in time.
`Clinical relapses become less distinct, recovery becomes less
`complete, and gradual worsening in the absence of relapses
`eventually becomes apparent. Transition to the SPMS stage,
`which commonly occurs during the fourth and fifth decade of
`life, can be estimated only in retrospect, once it is clear that the
`patient has gradually worsened in the absence of acute relapses.
`Because of the indistinct boundary between RR and SPMS,
`many patients could be entered into either a RRMS or a SPMS
`clinical trial, depending on how the clinician chooses to classify
`the individual patient.
`A consensus has emerged that PPMS (Chapter 52) should
`be considered separately for clinical trials. This is based on the
`uncertainty about the etiological relationship between PPMS
`and SPMS. Prototypical PPMS patients have symptom onset at
`a later age, typically between ages 40 and 60, and the female
`preponderance seen with relapsing forms of MS is not evi-
`dent. These patients commonly present with insidiously pro-
`gressive spastic weakness, imbalance, and sphincter dysfunc-
`tion; diffuse and less nodular T2—hyperintense lesions on cranial
`MRI; few if any Gd-enhancing lesions; and less indication of
`
`Page 7 of 16
`
`
`
`

`

`?*
`
`L__
`
`inflammation in cerebrospinal fluid (CSF).“ PPMS may be less
`dependent on inflammation, and neurodegenerative mecha-
`nisms may underlie the disease. Some PPMS patients have clini-
`cal, MRI, and CSF findings similar to SPMS. These patients may
`be similar to SPMS. but without clinically distinct relapses dur-
`ing the early disease stage. This is probably also true of another
`clinical category — progressive relapsing MS (PRMS) - in which
`there is gradual neurological progression from onset but with
`subsequent superimposed relapses. Thus, studies in PPMS are
`problematic for two reasons. These cases are relatively uncom-
`mon, and clinical trial groups contain admixtures of disease eti-
`ologies. It is unknown whether “SPMS—lil<e PPMS” and “pure
`PPMS” patients have similar pathogenic mechanisms driving
`disease progression, or whether they would respond similarly
`to treatment intervention.
`
`Common practice has been to select relatively homoge-
`neous patient groups for inclusion in clinical trials by enter-
`ing patients with a specified disease category, and creating dis-
`ability limits based on the EDSS.” As a result of widespread
`acceptance of the disease categories, separate trials have been
`conducted for patients with C18, RRMS, SPMS, and PPMS.
`This strategy aims to reduce between—patient variability and to
`increase the power to show therapeutic effects with a given sam-
`ple size. However, there are some drawbacks. Narrow entry cri-
`teria impede recruitment; it may not be clear whether the results
`of a trial enrolling a highly selected cohort of patients can be
`extrapolated to other groups of MS patients; and the distinc-
`tion between clinical disease categories is imprecise and based
`on clinical features that are disconnected from underlying dis-
`ease mechanisms. Conversely, different clinical trials that nom-
`inally studied the same patient population almost certainly con-
`tain different mixes of patients. This point is well illustrated by
`the European and North American trials of interferon beta-lb
`(IFNB—lb) in SPMS. These two trials used similar entry crite-
`ria, but enrolled distinct patient populations that yielded differ-
`ent results with the same therapeutic agent.“ The problem of
`classifying patients is most problematic at the interface between
`RRMS and SPMS. as discussed above. Biological markers for the
`different MS categories would be valuable, but are not currently
`available.
`
`Disease severity can not be accurately predicted in
`individuals or groups
`Because of the highly variable future course for newly diag-
`nosed MS patients, there is a compelling need for prognos-
`tic markers for treatment decision—making at the individual
`patient level. Prognostic markers would not only serve the
`need for better clinical decision-making, but also would help
`with informative enrollment into clinical trials. Data from the
`
`pre—therapeutic era suggested that 50% of MS patients were
`unable to carry out household and employment responsibilities
`10 years after disease onset, 50% required an assistive device to
`walk after 15-20 years, and 50% were unable to walk at all after
`
`Chapter 1:Chal|enges in experimental therapies
`
`l
`
`25 years.” About 10% of patients have an unusually severe dis-
`ease course, deteriorating to severe disability in only a few years,
`while 10%—20% exhibit mild disease with minimal disability
`decades after symptom onset. Distinguishing these severe and
`mild cases early after symptom onset has proved difficult.
`Selective enrollment has been attempted in clinical trials.
`The approach has been to enroll patients at risk for disease activ-
`ity, excluding patients not likely to change during the trial. In
`groups of patients, milder disease has been associated with sen-
`sory symptoms or optic neuritis at onset, good recovery from
`relapses and infrequent relapses early in the disease course.”' 15
`Conversely, symptom onset at an older age, progressive disease
`from onset, and poor relapse recovery mark a relatively worse
`prognosis. Clinical features have not been useful for informa-
`tive enrollment, however. The presence of multiple white matter
`lesions at the time of first MS symptom has proven very useful,
`as it is associated with much higher risk of disease activity in
`the next 5 years." Also, the amount of T2 lesion accrual during
`the initial 5 years after onset is a modest predictor of EDSS 20
`years later.” Despite this, T2 lesion load has not been used for
`informative enrollment strategies in clinical trials.
`Most trials employ relapses or progression during a speci-
`fied time period prior to the trial, or Gd-enhancing lesions on
`screening MRIs to identify patients with increased likelihood
`of disease activity during the trial. This is supported by a study
`showing that relapse rate prior to the trial and disease duration
`were the best predictors of on-study relapse rate.” In that study,
`disease course and Gd-enhancement status did not provide
`additional information. That study used a pooled data set from
`natural history studies and the placebo groups of randomized
`clinical trials, with a substantially larger sample size compared
`with previous analyses. A second study examined factors
`that predicted on-study Gd—enhancement, a common efficacy
`end-point in Phase 2 studies.“ A combination of younger
`age at onset, shorter disease duration, recent relapses, and T2
`lesion volume predicted Gd-enhancement. In other studies,
`the presence of Gd—enhancing lesions at baseline predicted
`frequency of clinical relapses, as well as increased T2 lesion
`volume and brain atrophy progression over the subsequent two
`years.”'32 However, all of the identified predictors, alone or in
`combination, are only modestly predictive of disease activity
`during a trial. The advantages of informative enrollment need
`to be balanced against the difficulty of finding eligible patients,
`and the problem of generalizing results when the entry criteria
`are restrictive.
`
`Heterogeneity in pathological mechanisms
`Studies of a large number of biopsy and autopsy specimens
`suggested that the mechanisms leading to tissue damage differ
`from patient to patient.”‘3“ Four distinct patterns of pathol-
`ogy were proposed. Analogous to experimental autoimmune
`encephalomyelitis,
`in patterns I and II
`the myclin sheath
`appears to be the target of the destructive process, mediated
`by macrophages in pattern I and antibody and complement
`
`,..—n-mm.-..
`
`Page 8 of 16
`
`
`
`

`

`I
`
`Section I: Introduction
`
`deposition in pattern 11. Pattern III is characterized by an
`ill-defined lesion border with early loss of adaxonal myelin-
`associated glycoprotein. This pattern is similar to that seen
`in some viral encephalitides and in cerebral ischemia. In pat-
`tern IV, there is a sparse inflammatory reaction, with pron1i-
`nent non-apoptotic degeneration of oligodendrocytes in the
`periplaque white matter. At present, pathologically distinct MS
`subgroups cannot be defined on the basis of biomarkers, or
`functional assays. However, most now recognize neuromyeli-
`tis optica (NMO) as a distinct disorder (Chapter 53). It has
`long been known that NMO differs from typical MS clinically,
`by imaging features, pathology,” and response to MS disease-
`modifying drugs. But the watershed event was the observa-
`tion that NMO is associated with antibodies to aquaporin—4,
`an astrocyte water channel.”‘'” Presumably, better understand-
`ing of MS pathological heterogeneity will lead to more rationale
`approaches to personalized use of disease—modifying drugs.
`
`(omplexities related to measurement tools
`that impact clinical trials
`Clinical measures: relapses, physical function,
`neuropsychological performance (Chapters 6-8)
`The annualized relapse rate or the number of relapses are the
`most common primary outcome measure for RRMS clinical
`trials. Relapse frequency was the primary outcome measure
`in pivotal trials of two of the three IFNB products,‘l""” the
`glatiramer acetate trial,“ and the natalizumab trials.43““ These
`studies led to world-wide approval by regulatory agencies, and
`marketing of the products. Relapses are considered clinically
`relevant by regulatory agencies, because they are defined by new
`neurological symptoms and signs and are therefore assumed to
`have clinical impact. The relationship between relapse number
`and future disability is weak, however.“ Relapses may be sub-
`jective, and influenced by bias, over- or under-reporting, and
`treatment unmasking. There are no accepted methods to quan-
`tify relapse severity or recovery from relapse. Lastly, the amount
`of relapse rate reduction considered “clinically important” has
`never been defined. The rate of relapse in MS clinical trial pop-
`ulations has fallen over time. This indicates that n1ore recent tri-
`
`als have enrolled patients with less active disease, lowering the
`power of recent trials to show treatment arm differences, and
`making comparison across trials completely impractical.
`The EDSS is an ordinal scale ranging from O to 10 that classi-
`fies disability severity according to 19 steps.” A score of0 means
`a normal neurological examination; a score of 3.5 is computed
`when there is moderate disability in more than one functional
`system (e.g. visual, motor, cerebellar, sensory, bowel, bladder,
`etc.), but the patient is able to walk an unlimited distance with-
`out assistance. A score between 4.0 and 6.0 indicates limited dis-
`
`tance walking. Level 6.0 indicates the need for unilateral assis-
`tance to walk, 6.5 bilateral assistance, and 37.0 measures sever-
`
`ity in non-ambulatory patients. There is debate whether the
`EDSS measures disability accurately at the low end, because it
`
`has been very difficult to standardize the scoring for the func-
`tional system scales a11d small changes within the functional
`systems have unclear clinical relevance; and the middle and
`high ranges are insensitive to change, and so lower the power
`of clinical trials. Despite criticism, the EDSS has been the stan-
`dard measure of neurologic disability in nearly all MS clinical
`trials.
`
`Since the mid-1990s, the EDSS has been used to deter-
`
`mine “disability progression,” by identifying patients with con-
`firmed worsening from the baseline score. The proportion of
`patients in different treatment arms are compared directly, or
`using survival curves. The most common definition of “disabil-
`ity progression” in RRMS trials is worsening from baseline by at
`least 1.0 EDSS point, confirmed at the next three-month study
`visit. A minority of trials have required six-month confirma-
`tion. The EDSS may revert to baseline more commonly if the
`three-month definition is used,“ probably because of residual
`effects of relapses still present at three-months. The relevance
`of confirmed EDSS worsening in the early stages of MS remains
`controversial. One study showed a strong observed correlation
`between six-month confirmed EDSS worsening and clinical
`outcome eight years later.” There are no similar studies using
`three-month confirmation. A pooled analysis of multiple clini-
`cal trials demonstrated a strong association between treatment
`effect on relapse rate, and treatment effect on confirmed EDSS
`worsening, suggesting these two measures are i11ter-related in
`RRMS patients.” Despi

This document is available on Docket Alarm but you must sign up to view it.


Or .

Accessing this document will incur an additional charge of $.

After purchase, you can access this document again without charge.

Accept $ Charge
throbber

Still Working On It

This document is taking longer than usual to download. This can happen if we need to contact the court directly to obtain the document and their servers are running slowly.

Give it another minute or two to complete, and then try the refresh button.

throbber

A few More Minutes ... Still Working

It can take up to 5 minutes for us to download a document if the court servers are running slowly.

Thank you for your continued patience.

This document could not be displayed.

We could not find this document within its docket. Please go back to the docket page and check the link. If that does not work, go back to the docket and refresh it to pull the newest information.

Your account does not support viewing this document.

You need a Paid Account to view this document. Click here to change your account type.

Your account does not support viewing this document.

Set your membership status to view this document.

With a Docket Alarm membership, you'll get a whole lot more, including:

  • Up-to-date information for this case.
  • Email alerts whenever there is an update.
  • Full text search for other cases.
  • Get email alerts whenever a new case matches your search.

Become a Member

One Moment Please

The filing “” is large (MB) and is being downloaded.

Please refresh this page in a few minutes to see if the filing has been downloaded. The filing will also be emailed to you when the download completes.

Your document is on its way!

If you do not receive the document in five minutes, contact support at support@docketalarm.com.

Sealed Document

We are unable to display this document, it may be under a court ordered seal.

If you have proper credentials to access the file, you may proceed directly to the court's system using your government issued username and password.


Access Government Site

We are redirecting you
to a mobile optimized page.





Document Unreadable or Corrupt

Refresh this Document
Go to the Docket

We are unable to display this document.

Refresh this Document
Go to the Docket