throbber
REVIEW ARTICLE
`
`Drugs 2008; 68 (17): 2445-2468
`0012-6667/08/0017-2445/$53.45/0
`
`© 2008 Adis Data Information BV. All rights reserved.
`
`Disease-Modifying Agents for
`Multiple Sclerosis
`Recent Advances and Future Prospects
`
`Til Menge,1 Martin S. Weber,2,3 Bernhard Hemmer,2 Bernd C. Kieseier,1
`Hans-Christian von B ¨udingen,4 Clemens Warnke,1 Scott S. Zamvil,3 Aaron Boster,5
`Omar Khan,6 Hans-Peter Hartung1 and Olaf St ¨uve7,8,9
`1 Department of Neurology, Heinrich Heine-University, D ¨usseldorf, Germany
`2 Department of Neurology, Klinikum rechts der Isar, Technical University,
`M ¨unchen, Germany
`3 Department of Neurology, University of California, San Francisco, California, USA
`4 Department of Neurology, University of Z ¨urich, Z ¨urich, Switzerland
`5 Department of Neurology, The Ohio State University, Columbus, Ohio, USA
`6 Department of Neurology, The Multiple Sclerosis Clinical Research Center, Wayne State
`University School of Medicine, Detroit, Michigan, USA
`7 Neurology Section, VA North Texas Health Care System, Medical Service, Dallas, Texas, USA
`8 Department of Neurology, University of Texas Southwestern Medical Centre at Dallas, Dallas,
`Texas, USA
`9 Department of Immunology, University of Texas Southwestern Medical Centre at Dallas,
`Dallas, Texas, USA
`
`Contents
`Abstract . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2445
`1. Pathogenesis of Multiple Sclerosis (MS) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2447
`1.1 Genetic and Environmental Factors . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2447
`1.2 Activation and Migration of Immune Cells . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2447
`1.3 Lesion Formation . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2448
`1.4 Neurodegeneration . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2448
`2. Testing the Efficacy of Pharmacological Agents in MS . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2449
`3. Current Treatment Regimens . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2449
`4. Overview of Future Compounds . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2452
`4.1 Suppressing the Immune System . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2452
`4.2 Modulating the Immune System . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2454
`4.2.1 Compounds with Broad Modes of Action . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2454
`4.2.2 Compounds with Selective Modes of Action . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2455
`4.2.3 Compounds with Highly Selective and Monospecific Modes of Action . . . . . . . . . . . . . 2456
`5. Protection and Repair of the CNS . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2458
`6. Adverse Effects . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2459
`7. Conclusions . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2459
`
`Abstract
`
`Multiple sclerosis (MS) is a chronic autoimmune disease of the CNS. Current-
`ly, six medications are approved for immunmodulatory and immunosuppressive
`treatment of the relapsing disease course and secondary-progressive MS. In the
`
`Page 1 of 24
`
`YEDA EXHIBIT NO. 2077
`MYLAN PHARM. v YEDA
`IPR2015-00644
`
`

`
`2446
`
`Menge et al.
`
`first part of this review, the pathogenesis of MS and its current treatment options
`are discussed.
`During the last decade, our understanding of autoimmunity and the pathogene-
`sis of MS has advanced substantially. This has led to the development of a number
`of compounds, several of which are currently undergoing clinical testing in phase
`II and III studies. While current treatment options are only available for parenteral
`administration, several oral compounds are now in clinical trials, including the
`immunosuppressive agents cladribine and laquinimod. A novel mode of action
`has been described for fingolimod, another orally available agent, which inhibits
`egress of activated lymphocytes from draining lymph nodes. Dimethylfumarate
`exhibits immunomodulatory as well as immunosuppressive activity when given
`orally. All of these compounds have successfully shown efficacy, at least in
`regards to the surrogate marker contrast-enhancing lesions on magnetic resonance
`imaging.
`Another class of agents that is highlighted in this review are biological agents,
`namely monoclonal antibodies (mAb) and recombinant fusion proteins. The
`humanized mAb daclizumab inhibits T-lymphocyte activation via blockade of the
`interleukin-2 receptor. Alemtuzumab and rituximab deplete leukocytes and
`B cells, respectively; the fusion protein atacicept inhibits specific B-cell growth
`factors resulting in reductions in B-cells and plasma cells. These compounds are
`currently being tested in phase II and III studies in patients with relapsing MS.
`The concept of neuro-protection and -regeneration has not advanced to a level
`where specific compounds have entered clinical testing. However, several agents
`approved for conditions other than MS are highlighted. Finally, with the advent of
`these highly potent novel therapies, rare, but potentially serious adverse effects
`have been noted, namely infections and malignancies. These are critically
`reviewed and put into perspective.
`
`efficacy and good safety profiles. Over the past 10
`Multiple sclerosis (MS) is a chronic disease con-
`years, specific anatomical, cellular and molecular
`fined to the CNS. Its pathological hallmarks are
`targets have become the focus of drug development.
`neuroinflammation, de- and remyelination, neurode-
`A more in-depth understanding of the inflammatory
`generation and astrogliosis. To date, the aetiology of
`cascade underlying MS disease activity will allow
`MS remains unknown; however, growing evidence
`the development of increasingly specific, and hope-
`supports an autoimmune pathogenesis triggered by
`fully safe and effective, pharmacological agents for
`environmental factors in genetically susceptible in-
`all clinical MS phenotypes. As a consequence, fu-
`dividuals. Perhaps not surprisingly, immunomodu-
`ture treatments will have to be designed to tackle the
`latory therapies have been the mainstay of pharma-
`neurodegenerative processes inherent to MS.
`cotherapy for many decades. Currently, clinicians
`On the basis of the current pathogenetic concepts
`have access to two distinct treatment strategies. In
`of MS, we provide an overview of future com-
`the past, our limited knowledge of MS pathogenesis
`pounds, describe the mechanisms by which they
`allowed only general modulation or suppression of
`immune responses. During the last decade, several modulate the immune system in patients with MS,
`agents belonging to the class of immunomodulators
`known adverse effects and their stages of clinical
`were shown to be effective in clinical trials, and
`development. Publications were retrieved by search-
`were approved. All of these agents have modest
`ing PubMed Entrez provided by the National Center
`
`© 2008 Adis Data Information BV. All rights reserved.
`
`Drugs 2008; 68 (17)
`
`Page 2 of 24
`
`YEDA EXHIBIT NO. 2077
`MYLAN PHARM. v YEDA
`IPR2015-00644
`
`

`
`Disease-Modifying Agents for Multiple Sclerosis
`
`2447
`
`for Biotechnology Information. The search strategy
`consisted of a combination of
`the following
`keywords: multiple sclerosis OR experimental
`autoimmune encephalomyelitis AND therapy, clin-
`ical trial(s), drug development, experimental, drug
`efficacy, drug safety. Latest search dates assessed
`were June 2008. The National Institute of Health’s
`website and the website of the National Multiple
`Sclerosis Society (http://www.nationalmssociety.
`org/research/clinical-trials/index.aspx) have been
`utilized to screen for clinical trials of compounds
`related to MS.
`
`1. Pathogenesis of Multiple
`Sclerosis (MS)
`
`In order to highlight current advances in the
`treatment of MS, one must appreciate the current
`pathogenic concepts of the disease. Typically, MS
`becomes clinically apparent during early adulthood.
`The disease is more prevalent in females than males.
`It is very likely that in many patients, CNS inflam-
`mation starts many years before the onset of clinical
`signs and symptoms.[1] Despite tremendous progress
`in our understanding of the disease and in the devel-
`opment of specific therapies, MS remains one of the
`leading causes of neurological disability among
`young individuals, second only to trauma.[2-4] Fol-
`lowing a period of monophasic disease, termed clin-
`ically isolated syndrome (CIS), the disease in the
`majority of patients enters a relapsing-remitting MS
`(RRMS) disease course. After another 10–15 years,
`the disease enters a chronic progressive phase in
`about 30–50% of patients, termed secondary-pro-
`gressive MS (SPMS).[2,5] Approximately 10–20%
`are affected by a primary-progressive MS (PPMS)
`disease course[4] and a rather small group of patients
`(<5%) experience a progressive-relapsing disease
`course that most rapidly progresses, resulting in loss
`of ambulation in a median of 7 years.[6]
`
`1.1 Genetic and Environmental Factors
`
`The aetiology of MS remains unknown. How-
`ever, there is strong evidence emerging that suggests
`a complex interplay of multiple genes and environ-
`mental factors.[7-9] The identification of specific sus-
`
`ceptibility genes has been difficult. Until recently,
`the only strong and consistent linkage identified has
`been at chromosome 6p21, the location of the major
`histocompatibility complex (MHC).[10-13] For pa-
`tients with MS of Northern European ethnicity,
`there is widespread consensus on the role of one
`common HLA allele: DRB1*1501. One allele of
`this gene increases the disease risk by an odds ratio
`of about 3.[14,15] More recently, it was shown by an
`international collaborative efforts that predominant-
`ly single nucleotide polymorphisms in the genes that
`encode parts of the interleukin (IL)-2 and -7 recep-
`tors appear to be associated with an increased risk of
`developing MS.[16,17] Interestingly, these genes play
`a critical role in inflammatory immune responses
`and neurodevelopment. In addition, a number of
`microbial agents, bacterial and viral, have been im-
`plicated in the pathogenesis of MS, of which Ep-
`stein-Barr virus, human herpesvirus (HHV)[6] and
`varicella zoster virus are currently being extensively
`studied.[18-22]
`It is conceivable that in an individual carrying
`such disease susceptibility genes, an infection or
`sequential infections may eventually lead to an aber-
`rant response of the immune system against self-
`antigens.[23,24]
`
`1.2 Activation and Migration of
`Immune Cells
`
`Both the innate and the adaptive immune systems
`play a role in the pathogenesis of MS. Structural
`homology of microbial antigens with CNS epitopes
`may lead to chronic activation of the immune sys-
`tem against self-antigens. This phenomenon is
`termed molecular mimicry. Animal studies have
`demonstrated that acquired immune responses to
`CNS antigens are initiated in the lymph nodes and
`spleen, where antigen-specific T and B cells become
`activated and clonally expand.[25] Activation enables
`these cells to cross biological membranes, including
`the blood-brain barrier (BBB). This is essential to
`physiological immune surveillance, including the
`CNS.[26] Unfortunately, it leads to autoimmunity if
`the antigen recognized in the lymph nodes resem-
`bles an autoantigen in the CNS. During the early
`
`© 2008 Adis Data Information BV. All rights reserved.
`
`Drugs 2008; 68 (17)
`
`Page 3 of 24
`
`YEDA EXHIBIT NO. 2077
`MYLAN PHARM. v YEDA
`IPR2015-00644
`
`

`
`2448
`
`Menge et al.
`
`sion.[40-43] Some of these maturation steps may also
`phase of MS, microglia activation and an amplifica-
`take place in the CNS within lymphoid follicle-like
`tion of immune cell infiltration into the brain and
`structures found in late stages of MS.[44-46] The ulti-
`spinal cord are seen.[27] The physiologically tightly
`sealed BBB likely becomes compromised, and the mate target specificities of these B cells still remain
`enigmatic.[47] Subsets of memory B cells and plasm-
`transmigration of additional immune cells, includ-
`ing macrophages and dendritic cells, is facilitated.
`ablasts, which are attracted to the intrathecal space
`and parenchymal lesions,[48] produce antibodies that
`Cell migration from the periphery into the CNS
`are detectable in MS lesions and are likely to be the
`involves a complex sequential interaction of adhe-
`cause of demyelination via complement activation
`sion molecules, matrix metalloproteinases (MMPs)
`in most MS patients.[49,50]
`and chemokines.[28] Namely, the firm contact be-
`There is emerging evidence that in the progres-
`tween leukocytes destined to enter the CNS and
`endothelial cells of the BBB activated by cytokines
`sive stages of MS, immune responses are further
`is, amongst others, mediated by α4-integrins on
`compartmentalized (see earlier in this section[44-46])
`and locally sustained,[51] thus impeding the access of
`leukocytes and vascular cell adhesion molecule-1
`(VCAM-1) on endothelial cells.[29] Other potentially
`a number of immunoactive therapeutic compounds
`relevant adhesion molecules and chemokine recep-
`to the lesion site, including monoclonal antibodies,
`tors are also expressed by lymphoid and myeloid
`neurotrophic and gliotrophic factors, and cell thera-
`cells.[30]
`pies.
`
`1.3 Lesion Formation
`
`1.4 Neurodegeneration
`
`While demyelination is the hallmark characteris-
`It was demonstrated by several investigators that
`tic of MS and occurs most prominently in areas of
`macrophages and T cells (both CD4+ and CD8+)
`infiltrate the CNS parenchyma in MS.[31-33] In addi-
`acute inflammation within the white matter, the
`neurodegenerative and neuroregenerative features
`tion, clonotypic CD8+ T cells were also detected in
`cerebrospinal fluid (CSF).[31,32] CD4+ T cells and
`of MS have only recently been rediscovered after
`their initial description by Charcot.[52-54] Damage to
`B cells appear to be more prominent in the perivas-
`cular spaces adjacent to lesions.[27] Macrophages
`or even loss of axons occurs in early disease stages
`and appears to correlate with the degree of neuro-
`and T cells within the lesion secrete a wide range of
`logical disability.[55,56] Axonal injury is evident both
`molecules toxic to the myelin sheath, including pro-
`at sites of inflammatory infiltrates and also in their
`teases, reactive oxygen species, nitric oxide deriva-
`conspicuous absence.[53] In the context of inflamma-
`tives and cytokines, that orchestrate the inflamma-
`tory damage.[34] Whereas some studies suggested a
`tion, the underlying mechanisms may be quite simi-
`pro-inflammatory T helper (Th)-1 and Th17 cyto-
`lar to those of demyelination: a direct attack on the
`kine signature in MS lesions,[33,35,36] other investiga-
`axon by CD8+ T cells, complement-mediated anti-
`tors demonstrated that both Th1 and Th2 cytokines,
`body-dependent phagocytosis of axons after binding
`of antibodies to neuronal membrane antigens,[57] T-
`and their receptors are up-regulated in the CNS of
`MS patients.[37] More recently, it has also been dis-
`cell-dependent recruitment and activation of macro-
`phages that express inflammatory mediators and
`cussed whether cytokines may be an essential com-
`toxic molecules all may lead to acute axonal trans-
`ponent of CNS repair mechanisms, for example,
`remyelination after an acute attack.[38] In addition, it
`section. In contrast, the gradual loss of oligodendro-
`cytes and axo-glial disconnection may eventually
`was proposed that the inflammatory milieu itself
`may support neuroprotection.[39] B cells in the CSF
`deprive axons of trophic support and further aug-
`and MS plaques are oligoclonally expanded, expres- ment their insidious damage; once denuded, axons
`sing somatically mutated B-cell receptor genes that
`appear to be particularly vulnerable to noxious med-
`are compatible with an antigen-driven expan-
`iators, including nitric oxide metabolites and ex-
`
`© 2008 Adis Data Information BV. All rights reserved.
`
`Drugs 2008; 68 (17)
`
`Page 4 of 24
`
`YEDA EXHIBIT NO. 2077
`MYLAN PHARM. v YEDA
`IPR2015-00644
`
`

`
`Disease-Modifying Agents for Multiple Sclerosis
`
`2449
`
`ing lesions on T1-weighted images, and the number
`citotoxic molecules, and hence undergo degenera-
`tion.[58] In these denuded axons, the redistribution
`of newly emerging lesions. However, there is a
`general notion that the use of these MRI disease
`and enhanced activities of sodium channels can lead
`to mitochondrial energy failure and calcium over- makers may correlate insufficiently with histopa-
`flow, ultimately activating proteases capable of dis-
`thology and clinical disease progression; hence,
`integrating the axonal cytoskeleton.[57,59]
`MRI may never substitute for clinical outcome mea-
`sures.[68] Biomarkers that unequivocally correlate to
`Up to 50% of all demyelinating lesions remye-
`disease parameters have not yet been established.[69]
`linate spontaneously.[60] Remyelination occurs in all
`The animal model of MS, experimental autoim-
`disease subtypes and takes place even late during the
`mune encephalomyelitis (EAE), has been extensive-
`disease course.[61] It appears to be initiated by
`ly studied to investigate CNS autoimmunity, but
`oligodendrocyte progenitor cells (OPCs), which
`does not faithfully reflect and model the hetero-
`proliferate within the lesion, differentiate into pre-
`geneity and insidious onset of the human disease
`myelinating oligodendrocytes and finally into ma-
`and its pathogenetic hallmarks.[70] EAE represents
`ture oligodendrocytes. This orderly process requires
`some of the characteristic features of MS (CNS
`an appropriately permissive microenvironment,
`damage mediated by CD4+ T cells and macro-
`such as growth factors and support of neurons.[62]
`phages), while others are largely missing or incom-
`Even
`in chronic MS
`lesions, premyelinating
`plete (CNS damage mediated by CD8+ T cells and
`oligodendrocytes are present, but seem to be im-
`B cells, neurodegeneration in the absence of inflam-
`paired in their remyelinating activity.[62,63] As yet, it
`mation).[70-72] This may explain why compounds
`remains enigmatic as to why remyelination occurs in
`shown to be highly efficacious in EAE have failed in
`some patients, but is absent or fails in other pa-
`clinical trails.[73,74] Perhaps even more problematic
`tients.[64,65] Possible mechanisms include ongoing
`is the fact that many pharmaceutical companies do
`destruction of OPCs, or the compromise of other
`not pursue drug development of compounds that fail
`glia cells and neurons that may support remyelina-
`to show a benefit in the EAE model.
`tion.[66,67]
`Novel EAE models, including transgenic human-
`Figure 1 illustrates the current concept of MS
`ized models, have been generated to better replicate
`pathogenesis. Compounds that selectively interfere
`B-cell- and CD8+ T-cell-mediated demyelination
`with certain pathogenic pathways are depicted in
`and axonal damage, and may be applied to preclini-
`figure 1 to highlight their mode of action.
`cal testing of novel compounds in the future.[72,75-80]
`Animal models of virus-induced autoimmunity
`or demyelination may add relevant information
`when evaluating pharmaceuticals for patients with
`Clinically, pathogenetically and histopathologi- MS.[23,70,71,81]
`cally, MS is a complex and highly heterogeneous
`disease with an often unpredictable disease
`course.[4,49,50] Thus, clinical trials would have to
`enrol very large numbers of patients (and controls)
`and would have to last many years to attain mean-
`ingful results if they only relied on clinical out-
`comes. In the past two decades, disease surrogate
`markers on brain magnetic resonance images
`(MRIs) have been used to substitute for clinical
`outcomes. These paraclinical tests aim to capture
`subclinical disease activity. Conventional imaging
`sequences include T2 lesion load, contrast enhanc-
`
`Acute relapses are managed by intravenous corti-
`costeroids: typically 3–5 days of methylpred-
`nisolone 1 g with or without oral tapering. This
`regimen was shown to shorten the duration of the
`relapse, mediated through a number of genomic and
`non-genomic actions.[82-84]
`Two classes of agents are currently approved as
`first-line treatment for the prevention of clinical
`relapses. These drugs are considered first-line thera-
`
`2. Testing the Efficacy of
`Pharmacological Agents in MS
`
`3. Current Treatment Regimens
`
`© 2008 Adis Data Information BV. All rights reserved.
`
`Drugs 2008; 68 (17)
`
`Page 5 of 24
`
`YEDA EXHIBIT NO. 2077
`MYLAN PHARM. v YEDA
`IPR2015-00644
`
`

`
`2450
`
`Periphery
`
`T helper cell
`
`Th2 and
`FoxP3+ Treg
`
`Adhesion
`
`B cells
`
`Proliferation
`
`Migration
`
`Neuron
`
`Menge et al.
`
`CNS
`
`Plasma cell
`
`Secretion of
`myelin-specific
`antibodies
`
`Astrocyte
`
`Ag presentation
`
`Proliferation
`Th1 and Th17
`differentiation
`
`Adhesion
`
`T cells
`
`Peripheral APC
`(e.g. dendritic cell)
`
`BBB
`
`Myelin
`sheath
`
`Migration
`
`Oligodendrocyte
`
`Secretion of
`inflammatory cytokines
`
`Local or
`recruited
`APC
`(e.g. microglia)
`
`Fig. 1. Mechanisms of action of currently approved treatments for multiple sclerosis. Interferon-β (IFNβ, yellow) reduces leukocyte
`proliferation, expression of adhesion molecules and secretion of matrix metalloproteinases required for leukocyte migration. Glatiramer
`acetate (GA, green) alters presentation of antigen (Ag) by antigen-presenting cells (APC) and promotes development of T helper (Th)-2 and
`FoxP3+ regulatory T cells (Treg). Through inhibition of cell replication, mitoxantrone (red) reduces proliferation and activation of leukocytes.
`Natalizumab (purple) blocks the adhesion molecule very late activating antigen (VLA)-4 inhibiting leukocyte adhesion to the blood-brain-
`barrier (BBB).
`
`pies based predominantly on their safety record, not
`their efficacy. Recombinant interferon-β (IFNβ) is
`available in three formulations for subcutaneous or
`intramuscular administration. All three formulations
`were approved on the basis of successful phase III
`trials in CIS, RRMS and at least for IFNβ-1b in
`SPMS.[85-91] There is now overwhelming evidence
`that early disease initiation with IFNβ slows down
`disease progression (recently reviewed in this jour-
`nal by St¨uve et al.[92]). IFNβ exerts a plethora of
`effects at multiple critical checkpoints of MS patho-
`genesis (see figure 1).[93,94] It induces the expression
`
`of a number of gene products, including 2′,5′-
`oligoadenylate synthetase, neopterin, tryptophan,
`β2-microglobulin and human Mx protein, and some
`of these markers are used in assays that assess the
`bioavailability of these drugs in individual pa-
`tients.[82] Treatment with IFNβ results in MHC class
`I gene expression, antiviral and antiproliferative ac-
`tions, and monocyte activation, to name a few. How-
`ever, the way that IFNβ mediates its beneficial
`effects in MS is not completely understood. There is
`convincing evidence that several different mechan-
`isms of action may play a role, although some have
`
`© 2008 Adis Data Information BV. All rights reserved.
`
`Drugs 2008; 68 (17)
`
`Page 6 of 24
`
`YEDA EXHIBIT NO. 2077
`MYLAN PHARM. v YEDA
`IPR2015-00644
`
`

`
`Disease-Modifying Agents for Multiple Sclerosis
`
`2451
`
`only been demonstrated in vitro. These include the
`inhibition of leukocyte proliferation, the decrease of
`antigen presentation by microglia, a modulatory ef-
`fect on the IgG synthesis of plasma cells, the reduc-
`tion of T-cell MMP expression, as well as the
`downregulation of adhesion molecules that allow T-
`cell migration into the brain.[95,96]
`
`decreased expression of MMPs.[107,108] Because of
`its potential cumulative cardiotoxicity, the individu-
`al maximal dosage is limited and the timepoint of
`treatment initiation has to be carefully consid-
`ered.[109] Mitoxantrone may slow down disease pro-
`gression in some patients with SPMS.[91,104,105]
`
`Another second-line treatment option for escalat-
`ing therapy in RRMS patients who do not respond to
`The second first-line compound, glatiramer ace-
`IFNβ or GA has been approved only recently. The
`tate (GA), is a random polypeptide made up of four
`highly specific humanized monoclonal antibody
`amino acids (L-glutamic acid, L-lysine, L-alanine
`(mAb) natalizumab is a novel compound that is the
`and L-tyrosine) in a specific molar ratio that resem-
`first product of rational drug design for this disease
`bles myelin basic protein (MBP). Interestingly, ser-
`in that it targets a molecule that propagates CNS
`endipity has led to its discovery as a MS treatment
`inflammation. Specifically, natalizumab targets the
`option, because initially it was developed to induce
`α4-integrin of the adhesion molecule very late acti-
`rather than ameliorate EAE.[97] While not all mech-
`vating antigen (VLA)-4 on leukocytes, an adhesion
`anisms of action of GA are fully understood, the
`following immunomodulatory effects of GA in MS molecule crucial for the initial contact between leu-
`may be attributed mainly to altered T-cell activation
`kocytes and endothelial cells of the BBB. The adhe-
`and differentiation:[93] (i) induction of GA-reactive
`sion molecule VLA-4 binds to VCAM-1 on brain
`vascular endothelium (figure 1).[29] By specific
`Th2-like regulatory suppressor cells; (ii) restoration
`blockade of the subunit α4-integrin, natalizumab
`of CD4+CD25+FoxP3+ regulatory T cells; and (iii)
`direct
`immunomodulatory activity on antigen- was designed to prevent egress of T cells from the
`presenting cells (APCs), which participate in innate
`blood into the brain. Proof-of-concept was demon-
`immune responses (figure 1).[98-100] GA is approved
`strated in the EAE model.[110] After several mainly
`for the treatment of RRMS.[101,102] A phase III trial in MRI-centred phase II studies,[111-114] some 7 years
`CIS (preCISe) was prematurely terminated in late
`later clinical efficacy was successfully explored in
`2007 as primary endpoints were already met in the
`two of the largest phase III trials conducted at the
`interim analysis.[103]
`time, enrolling over 2000 MS patients.[115,116] This
`led to US FDA fast-track approval of natalizumab in
`the US in 2004 after reviewing the 1-year data of the
`AFFIRM (Natalizumab Safety and Efficacy in Re-
`trial.[115]
`lapsing Remitting Multiple Sclerosis)
`Natalizumab reduced relapse rates by more than
`two-thirds and slowed disease progression during
`the 2-year observation period.[115-118]
`
`Because of its problematic safety profile, the
`alkylating chemotherapeutic agent mitoxantrone is
`only considered a second-line treatment option for
`patients with relapsing forms of MS who do not
`respond to IFNβ or GA.[104,105] Mitoxantrone inter-
`calates into DNA, resulting in cross-links and strand
`breaks.[106] In addition, mitoxantrone interferes with
`the enzyme topoisomerase II,[106] an enzyme that
`Inhibition of immune cell migration to the CNS
`had rare but serious adverse effects;[119] progressive
`transiently forms double-strand breaks that occur
`when DNA is altered during replication or transcrip- multifocal leucencephalopathy (PML) occurred in
`tion (figure 1). Consequently, mitoxantrone affects
`0.1% of patients, and subsequently led to the death
`replication predominantly in rapidly dividing cells.
`in two patients and severe disability in another pa-
`tient.[120] PML is a deadly opportunistic CNS infec-
`Predominantly as a result of this effect, there are a
`number of secondary effects on the immune system,
`tion for which specific treatment is not available. It
`including interference with antigen presentation, re-
`is caused by reactivation of a clinically latent JC
`duction in the expression of pro-inflammatory cyto-
`polyomavirus infection. As of March 2008, approxi-
`kines and attenuation of leukocyte migration via a mately 32 000 patients worldwide have received
`
`© 2008 Adis Data Information BV. All rights reserved.
`
`Drugs 2008; 68 (17)
`
`Page 7 of 24
`
`YEDA EXHIBIT NO. 2077
`MYLAN PHARM. v YEDA
`IPR2015-00644
`
`

`
`2452
`
`Menge et al.
`
`4. Overview of Future Compounds
`
`MS is a chronic inflammatory autoimmune dis-
`ease. Therefore, it is not surprising that the majority
`of current and proposed treatment options are direct-
`ed towards the modulation or suppression of the
`immune system. However, as more and more know-
`ledge is gathered regarding the potential of neurore-
`generation in MS, therapeutic avenues aiming to
`support neuroregeneration are becoming increasing-
`ly more popular. Therapies can be discerned in
`regards to their specificity (i.e. broad-based vs spe-
`cifically interfering with certain pathways). Most of
`the established therapies can be considered broad-
`based, including IFNβ and GA, while newer drugs
`are rather specific, including natalizumab. The spe-
`cific advantages and disadvantages of broad-based
`and highly specific agents that are currently in de-
`velopment or in clinical trials are discussed in this
`section, and table I summarizes the information
`provided for novel and only recently approved bi-
`opharmaceuticals.
`
`natalizumab in the post-marketing setting, 3600 of ment trials at the time were not unequivocal.[131] The
`benefit-risk ratio of azathioprine is considered un-
`whom have been exposed for at least 18 months,
`favourable, given the efficacy of IFNβ and GA, and
`with three further reported cases of PML to date.[121]
`the increased risk for malignancy in long-term treat-
`Further cases of PML have not yet been made pub-
`ment in an adolescent patient population.[130,132]
`lic. Notably, the two MS patients had received com-
`bination therapy with IFNβ, while the third patient
`As yet, treatment options for the progressive
`phases of MS are limited. There is currently no
`with Crohn’s disease had been pretreated with other
`agent approved for PPMS (i.e. none has shown
`immunomodulators and immunosuppressants. It is
`sustained efficacy).[133]
`thus tempting to speculate that effective global sup-
`pression of immune reactions may compromise
`immune surveillance of the CNS towards potentially
`noxious agents such as JC virus.[122,123] In fact, natal-
`izumab treatment of patients was shown to lead to
`the reactivation of HHV-6.[123] Additionally, since
`natalizumab was shown
`to mobilize CD34+
`haematopoietic stem cells from the bone marrow, it
`may be argued that potentially JC virus-bearing cells
`may also be sequestered systemically.[124]
`It remains to be further evaluated whether natal-
`izumab differentially impacts on migration of the
`various immune cell subsets, and whether additional
`effects (e.g. co-stimulation, cell maturation, apopto-
`sis, virus reactivation) play a role in its mode of
`action.[123,125] Sensitive enrolment procedures, in-
`cluding identification of compromised immunocom-
`petence prior to natalizumab initiation and height-
`ened awareness of possible complications during
`treatment, have been recommended.[126,127] Open
`questions remain concerning the long-term and ad-
`verse effects in patients receiving natalizumab, and
`the generation of biomarkers to a priori identify
`patients at risk for PML.
`In addition to the drugs listed in this section, all
`of which are approved for the treatment of patients
`with MS, a number of experimental agents have
`been used extensively in clinical practice. Cyclo-
`phosphamide is an alternative option when mitoxan-
`trone is not indicated or has failed, for instance, in
`patients with rapidly worsening MS.[128,129] There is
`no inherent risk of cardiotoxicity but there is for
`bladder toxicity. Oral immunosuppressants, such as
`azathioprine, methotrexate and ciclosporin, do not
`play a role in current MS treatment.[130] In the pre-
`IFNβ era, azathioprine was used to treat a larger
`percentage of patients with RRMS, although treat-
`
`4.1 Suppressing the Immune System
`
`New immunosuppressive drugs with less serious
`adverse effects than mitoxantrone have been devel-
`oped and are currently being evaluated in clinical
`trials. Laquinimod (ABR-215062) is an orally
`bioavailable quinoline-3-carboxamide. It is an ana-
`logue of linomide (roquinimex), which was chemi-
`cally modified after serious adverse effects occurred
`in patients with MS, including endocarditis and ser-
`ositis.[155] After demonstration o

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