throbber
GLATIRAMER ACETATE: MECHANISMS OF ACTION IN
`MULTIPLE SCLEROSIS
`
`Tjalf Ziemssen and Wiebke Schrempf
`
`Multiple Sclerosis Center Dresden, Neurological University Clinic
`Dresden University of Technology, Dresden 01307, Germany
`
`I. Introduction
`II. Pathology and Immunology
`III. MS as Neurodegenerative Disease
`IV. The Janus Face of CNS-Directed Autoimmune Inflammation
`V. Neurotrophic Factors Are Released by DiVerent Immune Cells
`VI. Glatiramer Acetate: Historical Remarks
`VII. GA: Overview of Clinical Studies
`VIII. GA: Imaging Studies
`IX. GA: Animal Models
`X. GA in MS: Mechanisms of Action
`A. Effects of GA by Binding to MHC Class II Molecules
`B. Effects of GA on the APC Level
`XI. EVects of GA on the B-Cell Level
`XII. EVects of GA on the T-Cell Level
`A. Th1–Th2 Shift
`B. GA as Altered Peptide Ligand
`C. Secretion of Neurotrophic Factors
`
`D. Induction of GA-Specific CD8þ Suppressor T Cells
`E. Induction of Regulatory CD4þCD25þ T Cells
`
`XIII. Conclusions
`References
`
`Glatiramer acetate (GA), formerly known as copolymer 1, is a mixture of
`synthetic polypeptides composed of four amino acids resembling the myelin basic
`protein (MSP). GA has been shown to be highly eVective in preventing and
`suppressing experimental autoimmune encephalomyelitis (EAE),
`the animal
`model of multiple sclerosis (MS). Therefore, it was tested in several clinical studies
`and so approved for the immunomodulatory treatment of relapsing-type MS.
`In contrast to other immunomodulatory MS therapies, GA has a distinct
`mechanism of action: GA demonstrates an initial strong promiscuous binding to major
`histocompatibility complex molecules and consequent competition with various
`(myelin) antigens for their presentation to T cells. In addition, antigen-based therapy
`generating a GA-specific immune response seems to be the prerequisite for GA
`
`INTERNATIONAL REVIEW OF
`NEUROBIOLOGY, VOL. 79
`DOI: 10.1016/S0074-7742(07)79024-4
`
`537
`
`Copyright 2007, Elsevier Inc.
`All rights reserved.
`0074-7742/07 $35.00
`
`Page 1 of 34
`
`YEDA EXHIBIT NO. 2083
`MYLAN PHARM. v YEDA
`IPR2015-00644
`
`

`
`538
`
`ZIEMSSEN AND SCHREMPF
`
`therapy. GA treatment induces an in vivo change of the frequency, cytokine secretion
`
`pattern and the eVector function of GA-specific CD4þ and CD8þ T cells, probably
`by aVecting the properties of antigen-presenting cells such as monocytes and dendritic
`cells. As demonstrated extensively in animal experiments, GA-specific, mostly,
`T helper 2 cells migrate to the brain and lead to in situ bystander suppression of the
`inflammatory process in the brain. Furthermore, GA-specific cells in the brain express
`neurotrophic factors like the brain-derived neurotrophic factor (BDNF) in addition
`to anti-inflammatory T helper 2-like cytokines. This might help tip the balance in favor
`of more beneficial influences because there is a complex interplay between detrimental
`and beneficial factors and mediators in the inflammatory milieu of MS lesions.
`
`I. Introduction
`
`Multiple sclerosis (MS) is the most common inflammatory demyelinating disease
`of the central nervous system (CNS). It is believed to be a multifocal immune-
`mediated disorder in which the myelin sheath or the oligodendrocyte is targeted
`by the immune system in genetically susceptible people. There is a considerable
`heterogeneity in terms of clinical, radiological, and pathological changes, mirrored
`by a high intrapatient and interpatient variability in the clinical course and its
`manifestations. The disease aVects approximately 0.1% of the population in tem-
`perate climates. It is a disease of young people with a lifelong and often disabling
`course. MS is manifested in physical symptoms (relapses and disability progression),
`CNS inflammation, brain atrophy and cognitive dysfunction.
`About 85% of the patients begin with a relapsing-remitting disease, where
`neurological symptoms and signs develop over several days, plateau, and then
`usually improve over days to weeks. Approximately two-thirds of patients with
`relapsing-remitting MS (RRMS) undergo a conversion to a secondary progressive
`disease course (SPMS), where relapse frequency lessens over time and progressive
`neurological dysfunction emerges. The remaining 15% of patients begin the
`disease course with a gradually progressive neurological dysfunction, typically a
`slowly worsening myelopathy [primary progressive MS (PPMS)].
`
`II. Pathology and Immunology
`
`The pathological hallmark of MS is the demyelinating plaque which consists
`of infiltrating lymphocytes and macrophages, damage to the blood–brain barrier,
`and loss of myelin.
`
`Page 2 of 34
`
`YEDA EXHIBIT NO. 2083
`MYLAN PHARM. v YEDA
`IPR2015-00644
`
`

`
`GLATIRAMER ACETATE: MECHANISMS OF ACTION IN MULTIPLE SCLEROSIS 539
`
`Periphery
`
`CNS
`
`APC
`
`Blood−brain barrier
`
`6
`
`1
`
`3
`
`4
`
`5
`
`TMyelin
`
`2
`
`BMyelin
`
`BMyelin
`
`APC
`
`TMyelin
`
`FIG. 1. Hypothetical pathophysiological cascade in MS. Details (inclusive numbers) in the text.
`
`Oligodendrocytes synthesize and maintain the axonal myelin sheath of up to
`40 neighboring nerve axons in the CNS. Compact myelin consists of a condensed
`membrane, spiraled around axons to form the insulating segmented sheath
`needed for saltatory axonal conduction; voltage-gated sodium channels cluster
`at the unmyelinated nodes of Ranvier between myelin segments, from where the
`action potential is propagated and spreads down the myelinated nerve segment to
`trigger another action potential at the next node.
`In MS, the composition of the inflammatory infiltrate varies depending on the
`stage of demyelinating activity. Early symptoms of MS are widely believed to
`result from this inflammatory demyelination which leads to slowing or blockade
`of axonal conduction. The regression of symptoms has been attributed to the
`resolution of inflammatory edema and to partial remyelination.
`Current concepts assume that MS occurs as a consequence of immune toler-
`ance breakdown in genetically susceptible individuals (Hafler, 2004). The major
`contributing factors thus include genetics, environment, and immune dysregula-
`tion. T helper (Th) cells are considered to play a pivotal role in the whole self-
`reactive immune response of the CNS, primarily characterized by inflammatory
`demyelination. Putting experimental data from human studies and animal experi-
`ments together, the following pathophysiological cascade seems quite attractive
`which is shown and numbered in Fig. 1:
`
`Page 3 of 34
`
`YEDA EXHIBIT NO. 2083
`MYLAN PHARM. v YEDA
`IPR2015-00644
`
`

`
`540
`
`ZIEMSSEN AND SCHREMPF
`
`1. Autoreactive T cells, known to exist in any individual, are activated in the
`periphery, probably by molecular mimicry (i.e., recognition of epitopes
`that are common to autoantigens and microbial structures as exogenous
`triggers) or by self-antigens.
`2. T cells may trigger B-cell activation and antibody formation, the latter
`potentially exerting detrimental eVector functions at the myelin sheath
`(Ziemssen and Ziemssen, 2005).
`3. T-cell activation enables transmigration through the blood–brain barrier
`to the sites of inflammation, a cascade of events influenced by adhesion
`molecules, chemotactic factors and migration promoters.
`4. Recognizing their antigens presented by microglia, the local antigen-
`presenting cells (APCs), the autoreactive T cells are reactivated.
`
`5. T cells of both CD4þ and CD8þ cytotoxic phenotypes release proinflam-
`
`matory cytokines. An inflammatory cascade is initiated followed by the
`further recruitment of inflammatory cells like monocytes.
`6. Underlying immunoregulatory defects, such as decrease of regulatory
`T cells in the circulation of patients with MS, allow the further pathological
`activation of autoreactive T cells.
`
`III. MS as Neurodegenerative Disease
`
`Although MS seems to be primarily an inflammatory autoimmune disease, it has
`become evident that axonal loss plays an important role in the pathogenesis of
`disability in patients with MS. While axonal pathology was elegantly and precisely
`described in classic MS neuropathologic studies more than a century ago (Charcot,
`1877), it has reemerged as a major focus of research (Trapp et al., 1998). The
`important question to be addressed is not whether there is axonal loss in MS but
`when and to what extent does the axonal loss occur. The timing and degree of axonal
`loss is of importance not only in its relationship to the etiology of the disease but may
`be central to the appearance of clinical symptoms and the progressive deterioration
`associated with the disease. The fact that axonal loss is irreversible has important
`implications for when and which therapeutic intervention should be used.
`It is likely that various mechanisms contribute to axonal damage during diVerent
`stages of disease. In active lesions, the extent of axonal transection correlates with
`inflammatory activity while even there seems to be an inflammation-independent
`axonal loss (Perry and Anthony, 1999). Hence, axonal loss may be caused by
`inflammatory products of activated immune and glial cells, including proteolytic
`enzymes, cytokines, oxidative products, and free radicals, although the precise
`molecular mechanisms of axonal damage are poorly understood. In addition, the
`magnitude of axonal loss in chronic MS lesions without pronounced inflammatory
`
`Page 4 of 34
`
`YEDA EXHIBIT NO. 2083
`MYLAN PHARM. v YEDA
`IPR2015-00644
`
`

`
`GLATIRAMER ACETATE: MECHANISMS OF ACTION IN MULTIPLE SCLEROSIS 541
`
`infiltrates suggests that mechanisms other than inflammatory demyelination con-
`tribute to the degeneration of axons. Several conditions interfere with attempts of
`axonal regrowth after lesions develop. These include the lack of neurotrophic factors
`that support growth, the presence of a glial scar (depending on the site of lesion) or
`the presence of inhibitory molecules that impede axonal growth. Evidence shows
`that axon degeneration following injury has similarities with the cellular mechanisms
`underlying programmed cell death (Perry and Anthony, 1999).
`The biochemical events underlying the inflammatory and neurodegenerative
`phases of the disease are not known in detail and may be quite diVerent (Steinman,
`2001). Inflammation involves activation of T and B cells in the periphery, crossing
`the blood–brain barrier and homing to the lesion site. In the lesion, T cells are
`reactivated by myelin antigens, release cytokines that attract macrophages and
`activate microglia which start to destroy the myelin sheath. Anti-myelin antibodies
`bind complement, attract macrophages, and stimulate opsonization of myelin.
`Demyelination leads to reversible and to some extent irreversible impairment of
`functionality of the axon whose conduction properties are deteriorated, thus
`accounting for the clinical symptoms associated with relapses.
`Neurodegeneration is likely to be a complex process (Chitnis et al., 2005;
`Grigoriadis et al., 2004), especially when it takes place in inflammatory disorders
`like MS. To a significant extent, axonal loss seems to be a major consequence of
`
`demyelination and inflammation, for example, by binding of CD8þ T cells to
`
`exposed axons and secretion of toxic factors. However, other mechanisms not
`directly related to demyelination and inflammation are also likely to be important.
`An example is excitotoxicity: glutamic acid can bind to excitatory amino acid
`receptors on the cell bodies, dendrites, or axon terminals of neurons and initiate a
`process of neuronal cell death (Steinman, 2001).
`Theoretically, there are several possible relationships between inflammation
`and neurodegeneration in MS. Three exclusive hypotheses can be envisaged:
`(a) that neurodegeneration is entirely secondary to inflammation, (b) that inflam-
`mation is entirely secondary to neurodegeneration, or (c) that inflammation and
`neurodegeneration are entirely independent. On the other hand, nonexclusive
`hypotheses stating that neurodegeneration is partially dependent and partially
`independent of inflammation or vice versa can also be put forward, and these
`seem intuitively more likely (Ziemssen, 2005).
`There are a number of clinical arguments in favor of some independence
`between the inflammatory and neurodegenerative processes. For example, in a
`clinical trial of alemtuzumab (Campath-1H), a monoclonal antibody directed
`against CD52 which leads to T-cell depletion, in secondary progressive MS (SPMS),
`a gradual extinction of exacerbations and lesion activity visible on magnetic reso-
`nance imaging (MRI) was demonstrated (Coles et al., 1999). However, disability
`continued to progress in about half the patients in whom progressive brain atrophy
`and axonal degeneration could be observed using MRI and magnetic resonance
`
`Page 5 of 34
`
`YEDA EXHIBIT NO. 2083
`MYLAN PHARM. v YEDA
`IPR2015-00644
`
`

`
`542
`
`ZIEMSSEN AND SCHREMPF
`
`spectroscopy (MRS). The investigators concluded firstly that inflammation and
`demyelination were responsible for relapses of MS and could be prevented
`by alemtuzumab treatment and secondly that ongoing axonal degeneration
`accounted for the progressive phase of disability. Even though axonal injury may
`have been conditioned by prior inflammation, this process can continue despite
`complete suppression of inflammatory activity.
`There is also neuropathologic evidence for a dissociation between inflamma-
`tory demyelination and axonal injury from a series of 42 biopsy samples obtained
`from patients with MS (Bitsch et al., 2000). Acute axonal injury was visualized by
`amyloid precursor protein (APP) staining. There was no relationship between the
`expression of APP or axonal density and the extent of demyelination, with axonal
`injury being observed even in lesions that were successfully remyelinating. Simi-
`larly, there was no association between the extent of axonal injury and markers of
`acute inflammation such as tumor necrosis factor (TNF)- or inducible nitric
`oxide synthase. However, axonal injury was correlated to some extent with the
`extent of infiltration by CD8þ T lymphocytes and macrophages. A dissociation
`
`between neurodegeneration and inflammatory demyelination is also observed in
`lesions within the cortex. These lesions are characterized by a significant degree of
`axonal transection and apoptosis of neuronal cell bodies. However, the extent of
`infiltration by T lymphocytes and macrophages and the expression of inflamma-
`tory markers are low (Bo et al., 2003; Peterson et al., 2001).
`MRI studies in very early disease also suggest that inflammation and neuronal
`injury are not strictly related. A study of 31 subjects presenting with a clinically
`isolated syndrome evaluated inflammatory lesion activity with classical T2- and
`T1-weighted images after gadolinium enhancement and measured a surrogate
`marker of axonal injury, the size of the N-acetylaspartate peak (NAA) determined
`in the whole brain (Filippi et al., 2003). In these patients, the mean size of the NAA
`peak was some 20% lower than that observed in matched controls. No correlation
`was observed between the size of the NAA peak and lesion volume on either T1 or
`T2 images. The investigators concluded that significant axonal injury occurs early
`in the disease and that this is only indirectly linked to inflammatory activity.
`Studies such as these suggest that treatment strategies for MS need to address
`both the inflammatory and neurodegenerative components of the disease, and that
`anti-inflammatory therapies may only be able to control the inflammation-related
`neurodegenerative process adequately (Chitnis et al., 2005).
`
`IV. The Janus Face of CNS-Directed Autoimmune Inflammation
`
`Inflammation is considered to be a key feature in MS pathogenesis. The
`neurotoxic eVects of inflammation are well established and thought to be at least
`partially responsible for the observed axonal damage. Recently, an increasing
`
`Page 6 of 34
`
`YEDA EXHIBIT NO. 2083
`MYLAN PHARM. v YEDA
`IPR2015-00644
`
`

`
`GLATIRAMER ACETATE: MECHANISMS OF ACTION IN MULTIPLE SCLEROSIS 543
`
`body of experimental evidence supports the view of a dual role of the immune
`system in CNS-directed autoimmune inflammation. A number of studies have
`proposed that autoimmune inflammation may have neuroprotective eVects in the
`CNS (Ziemssen, 2005).
`On the one hand, MS and its animal models, representing neuroimmunolo-
`gical diseases which arise when immune cells attack the nervous system, provide
`the paradigm for the deleterious interaction between cells of the immune and
`nervous systems. EAE, the MS animal model, can be induced by active immuni-
`zation with CNS autoantigens MBP or by the transfer of autoantigen-specific
`T cells into naive syngenic recipients (Gold et al., 2006). On the other hand, it was
`demonstrated that MBP-specific, encephalitogenic T cells may have seemingly
`neuroprotective (side) eVects. The neuroprotective and regenerative potential of
`immune cells was first coined by Schwartz et al. (1999).
`They demonstrated that autoimmune T cells could protect neurons in an
`animal model of secondary degeneration after a partial crush injury of the optic
`nerve (Moalem et al., 1999). In several experiments, T cells with diVerent specifi-
`cities [specific for MBP, control antigen ovalbumin (OVA), or a heat-shock
`protein (hsp) peptide] were activated by restimulation with their respective anti-
`gens in vitro, and then injected into rats immediately after a unilateral optic nerve
`injury. Seven days after injury, the optic nerves were analyzed by immunohisto-
`chemistry for the presence of T cells. Small numbers of T cells could be found in
`the intact (uninjured) optic nerves of rats injected with anti-MBP T cells, which is
`consistent with previous observations that activated MBP-specific T cells home to
`intact CNS white matter.
`A much more pronounced accumulation of T cells, however, was observed in
`the crushed optic nerves of the rats injected with T cells specific for MBP, hsp peptide,
`or OVA. The degree of primary and secondary damage to the optic nerve axons
`and their attached retinal ganglion cells was measured by injecting a neurotracer
`distal to the site of the optic nerve lesion immediately after the injury, and again
`after 2 weeks. The number of labeled retinal ganglion cells as marker for viable
`axons was significantly greater in the retinae of the rats injected with anti-MBP
`T cells than with anti-OVA or anti-hsp peptide T cells. Thus, although all three
`T-cell lines (TCLs) accumulated at the site of injury, only the MBP-specific,
`autoimmune T cells had a substantial eVect in limiting the extent of secondary
`degeneration. This neuroprotective eVect was confirmed by electrophysiological
`studies.
`The results demonstrate that T-cell autoimmunity can mediate significant
`neuroprotection after CNS injury. The authors speculate that after injury, ‘‘cryptic’’
`epitopes might become available and might be recognized by endogenous non-
`encephalitogenic (benign) T cells. After local stimulation, these protective autoreac-
`tive T cells could exert their neuroprotective eVect. The findings further substantiate
`the idea that ‘‘natural autoimmunity’’ can be benign and may even function as a
`protective mechanism (Cohen, 1992).
`
`Page 7 of 34
`
`YEDA EXHIBIT NO. 2083
`MYLAN PHARM. v YEDA
`IPR2015-00644
`
`

`
`544
`
`ZIEMSSEN AND SCHREMPF
`
`Macrophages seem to represent another type of immune cell which is capable
`of mediating neuroprotection and/or stimulating recovery of CNS lesions. The
`injection of activated macrophages into transected rat spinal cord stimulated
`tissue repair and partial recovery of motor function (Rapalino et al., 1998). The
`neuroprotective activity of immune cells is not restricted to the CNS. After
`experimental axotomy of the facial nerve of immunodeficient SCID mice, the
`survival of facial motor neurons was severely impaired compared to immunocom-
`petent wild-type mice. Reconstitution of SCID mice with wild-type splenocytes
`containing T and B cells restored the survival of facial motor neurons in these
`mice to the level of the wild-type controls (Serpe et al., 1999).
`It is equally evident that a large number of neurotoxic and proinflammatory
`mediators are produced and released by immune cells (Kerschensteiner et al.,
`2003). The neutralization of toxic inflammatory mediators may improve the
`outcome of MS and experimental models of CNS damage. On the other hand,
`we know that immune cells can supply a number of neuroprotective mediators,
`including neurotrophic factors, anti-inflammatory cytokines, and prostaglandins
`which may reduce tissue damage (Hohlfeld et al., 2006).
`The idea that inflammatory reactions may not always be harmful under
`certain conditions even confers neuroprotection and repair, has important con-
`sequences for the design of immunomodulatory therapies for MS (Hohlfeld et al.,
`2006). Undebatably, there is convincing rationale for immunosuppressive treat-
`ment when the noxious eVects of the inflammatory reaction prevail. Because
`nonselective immunosuppressive treatments will suppress both destructive and
`beneficial components of inflammation, therapy is likely to fail when the benefi-
`cial eVects of CNS inflammation outweigh its negative consequences. It seems, for
`example, possible that the lack of beneficial (side) eVects of inflammation during
`the late phase of MS with little inflammation but ongoing axonal loss contributes
`to the pathogenesis of neurodegeneration. In MS, it is unfortunately unclear
`whether there is a stage of the disease when the inflammatory reaction is more
`beneficial than harmful.
`The concept of the neuroprotective role of inflammation can be extended to
`neurodegenerative, ischemic, and traumatic lesions of the CNS, considering that
`inflammation is a universal tissue reaction crucial for defense and repair (Ziemssen
`and Ziemssen, 2005).
`
`V. Neurotrophic Factors Are Released by Different Immune Cells
`
`The precise mechanisms involved in immune-mediated neuroprotection re-
`main to be clarified. A number of recent studies have shown that several types of
`immune cells and hematogenic progenitor cells express one or more neurotrophic
`
`Page 8 of 34
`
`YEDA EXHIBIT NO. 2083
`MYLAN PHARM. v YEDA
`IPR2015-00644
`
`

`
`GLATIRAMER ACETATE: MECHANISMS OF ACTION IN MULTIPLE SCLEROSIS 545
`
`factors. For example, nerve growth factor (NGF) is produced by B cells, which
`also express the trkA receptor and p75 NGF receptor (Torcia et al., 1996). Because
`neutralization of endogenous NGF caused apoptosis of memory B cells, it was
`concluded that NGF is an autocrine growth factor for memory B cells.
`More recently, another neurotrophin, brain-derived neurotrophic factor
`(BDNF), was found to be expressed in immune cells. BDNF was originally cloned
`in 1989 as the second member of the neurotrophin family which includes NGF
`and neurotrophins (NT)-3, -4/5, -6, and -7 (Lewin and Barde, 1996). Since then,
`the important role of BDNF in regulating the survival and diVerentiation of
`various neuronal populations, including sensory neurons, cerebellar neurons,
`and spinal motor neurons, has been firmly established. Neurons are the major
`source of BDNF in the nervous system. BDNF binds to diVerent types of recep-
`tors: the tyrosine kinase receptor B (trkB) which exists in two isoforms—the full-
`length receptor (gp145trkB) and the truncated receptor (gp95trkB) lacking the
`tyrosine kinase domain—and the p75 neurotrophin receptor (Klein et al., 1991). It
`is thought that BDNF and NT-4/5 exert their biological function via the full-
`length form of trkB receptor which expression seems to be restricted to neuronal
`cell populations.
`Immune cells can be a potent source of the neuroprotective factor BDNF
`in neuroinflammatory disease (Hohlfeld et al., 2006). Activated human T cells,
`B cells, and monocytes are able to secrete bioactive BDNF after in vitro activation
`(Kerschensteiner et al., 1999). The BDNF secreted by immune cells is bioactive as
`it supports neuronal survival in vitro. In histology, BDNF immunoreactivity was
`found in T cells and macrophages in active and inactive MS lesions.
`Similar observations were recently reported by several other groups of in-
`vestigators. After experimental injury of the striatum, activated macrophages and
`microglia cells transcribe mRNA for glial cell line-derived neurotrophic factor
`(GDNF) and BDNF (Batchelor et al., 1999). This could help to explain the
`sprouting of dopaminergic neurons observed after experimental injury. Tran-
`scripts for BDNF and NT-3 and their receptors trkB and trkC were found in
`subpopulations of human peripheral blood cells (Besser and Wank, 1999). BDNF
`protein was secreted by cultured T-cell clones.
`Besides BDNF, there is a robust expression of the full-length BDNF receptor
`gp145trkB in neurons in the vicinity of MS plaques (Stadelmann et al., 2002).
`Single neurons with clearly pronounced trkB immunoreactivity close to MS
`lesions can be observed, suggesting an upregulation of trkB in a proportion of
`damaged neurons. Additionally, full-length trkB immunoreactivity is present in
`reactive astrocytes within the lesions. The restriction of trkB expression to neural
`cell types in MS underscores the possibility that there may be BDNF signaling
`from infiltrating cells to neurons in neuroinflammatory lesions, as would be
`necessary for immune cells to support neuronal survival or provide axonal
`protection.
`
`Page 9 of 34
`
`YEDA EXHIBIT NO. 2083
`MYLAN PHARM. v YEDA
`IPR2015-00644
`
`

`
`546
`
`ZIEMSSEN AND SCHREMPF
`
`Within the MS lesion, immune cells seem to be the major source of BDNF
`(Stadelmann et al., 2002). They are likely to release this substance in the immedi-
`ate vicinity of nerve cell processes, which—according to the observed trkB
`expression—are likely to be responsive to the neuroprotective eVects of BDNF.
`This neurotrophin-mediated neuroimmune signaling network could be a major
`factor that helps to preserve axons in a microenvironment that is clearly capable
`of exerting significant neurotoxicity. Thus, it should be considered as a beneficial
`aspect of neuroinflammation that could be worth preserving therapeutically, or
`even reinforcing using tailored immunomodulatory treatment strategies.
`
`VI. Glatiramer Acetate: Historical Remarks
`
`w
`
`), formerly known as copolymer 1, is the
`Glatiramer acetate (GA, Copaxone
`acetate salt of a standardized mixture of synthetic polypeptides containing the
`four amino acids L-alanine, L-glutamic acid, L-lysine, and L-tyrosine with a
`defined molar ratio of 0.14:0.34:0.43:0.09 and an average molecular mass of
`4.7–11.0 kDa, that is, an average length of 45–100 amino acids (Arnon et al.,
`1996; Teitelbaum et al., 1997a,b).
`In the 1960s, Drs. Sela, Arnon, and their colleagues at the Weizmann
`Institute, Israel were involved in studies on the immunologic properties of a series
`of polymers and copolymers which were developed to resemble MBP, a myelin
`protein. MBP in Freund’s complete adjuvant induces EAE, the best animal model
`of MS. They were interested in evaluating whether these polypeptides could
`simulate the ability of MBP and fragments and regions of the MBP molecule to
`induce EAE (Teitelbaum et al., 1971, 1973, 1974a,b). None of these series was
`capable of inducing EAE, but several polypeptides were able to suppress EAE in
`guinea pigs. Copolymer 1, later known as GA, was shown to be the most eVective
`polymer in preventing or decreasing the severity of EAE. The suppressive eVect is
`a general phenomenon and not restricted to a particular species, disease type, or
`encephalitogen used for EAE induction.
`Abramsky et al. (1977) were the first who treated a group of severe RRMS
`patients with intramuscular GA 2–3 mg every 2–3 days for 3 weeks, then weekly for
`2–5 months. No conclusions could be drawn regarding drug eYcacy but there were
`no significant undesirable side eVects. Three clinical trials in the 1980s were
`performed showing some evidence of eYcacy that was adequate for support of the
`Food and Drug Administration (FDA) approval and a good safety profile (Bornstein
`et al., 1982, 1984, 1987). However, the results of these studies must be interpreted
`with caution because before 1991 the production of the drug was not standardized
`( Johnson, 1996). DiVerent batches had variable suppressive eVects on EAE which
`could also imply variable eVects in MS patients.
`
`Page 10 of 34
`
`YEDA EXHIBIT NO. 2083
`MYLAN PHARM. v YEDA
`IPR2015-00644
`
`

`
`GLATIRAMER ACETATE: MECHANISMS OF ACTION IN MULTIPLE SCLEROSIS 547
`
`In 1991, a phase III multicenter trial with a daily 20-mg dose of an s.c.
`administered, highly standardized GA preparation was started in the United
`States. This double-blind, placebo-controlled study demonstrated that GA signif-
`icantly reduced the relapse rate without significant side eVects ( Johnson et al.,
`1995).
`In 1996, GA was approved by the US FDA as a treatment of ambulatory
`patients with active RRMS. Since then GA has been licensed for approval in
`many other countries (Ziemssen et al., 2002b).
`
`VII. GA: Overview of Clinical Studies
`
`Four early exploratory open studies were performed in the late 1970s and
`early 1980s in order to obtain indications of dosing and safety (Abramsky et al.,
`1977; Bornstein et al., 1982, 1984, 1987). In total, 41 patients with relapsing-
`remitting or SPMS were enrolled in these studies. The treatment schedules, doses
`of GA, and treatment duration were quite variable from study to study. The
`maximum dose of 20 mg/day was well tolerated and no severe adverse eVects
`were detected in these studies.
`To assess more comprehensively the eYcacy of GA in patients with RRMS, a
`definitive phase III trial was conducted at 11 US medical centers with a total of
`251 RRMS patients who received GA at a dosage of 20 mg or placebo by daily
`s.c. injection for 2 years ( Johnson et al., 1995). As the primary end point, the mean
`annualized relapse rates were 0.59 for the GA-treated group and 0.84 for the
`
`placebo group, a 29% reduction was statistically significant ( p ¼ 0.007). Trends in
`
`the proportion of relapse-free patients and median time to first relapse favored
`GA treatment. Patients in both groups with higher disability at entry, measured by
`the Expanded Disability Status Scale (EDSS), had a higher relapse rate, while the
`largest reduction in relapse rate between groups occurred in patients with a
`baseline EDSS of 0–2 (33% vs a reduction of 22% in patients with an EDSS
`score at entry >2). When the proportion of patients who improved, were un-
`changed, or worsened by 1 EDSS step from baseline to end of study (2 years)
`worsened ( p ¼ 0.037). The eVect of treatment was constant throughout the entire
`
`was evaluated, significantly more patients on GA improved and more on placebo
`
`study duration. Patient withdrawals were 19 (15.2%) from the copolymer 1 group
`and 17 (13.5%) from the placebo group at approximately the same intervals. The
`treatment was well tolerated. The most common adverse experience was the local
`injection-site reaction. Rarely, the transient self-limiting immediate post-injection
`reaction followed the injection in 15.2% of those treated with GA and 3.2% of
`those treated with placebo.
`
`Page 11 of 34
`
`YEDA EXHIBIT NO. 2083
`MYLAN PHARM. v YEDA
`IPR2015-00644
`
`

`
`548
`
`ZIEMSSEN AND SCHREMPF
`
`In an extension of this study up to 35 months with unchanged blinding and
`study conditions, the clinical benefit of GA for both the relapse rate and for
`neurological disability was sustained ( Johnson et al., 1998). Thus, the reduction of
`
`annual relapse rate was 32% in favor of GA ( p ¼ 0.002). The results of this
`
`extension study confirmed the excellent tolerability and safety profile of GA. This
`study was further on extended as open-label study with all patients receiving
`active drug. The published data from approximately 10 years of ongoing trial
`showed a persistent reduced relapse rate of about 1 relapse per 5 years (Ford et al.,
`2006). After more than 10 years of observation, 108 out of 232 patients (47%)
`were still participating. Sixty-two percent of the group who stayed on GA without
`interruption were neurologically unchanged or improved from baseline by at least
`one step of the EDSS scale in contrast to 28% of the withdrawn patients. This is
`the first time that the benefits of an immunomodulatory treatment over this
`clinically highly relevant long-term time period are demonstrated in a planned
`neurological follow-up study.
`Studies using an oral formulation of GA (CORAL) (Filippi et al., 2006) and
`applying GA to PPMS patients (PROMISE) (Wolinsky and PROMiSe Trial
`Study Group, 2004) failed to demonstrate significant treatment eVects. There
`are promising new data on using a higher GA dose (40 mg) per injection which
`are studied in a multicenter, controlled trial (FORTE).
`
`VIII. GA: Imaging Studies
`
`Several early MRI studies indicated a trend towards benefit with GA, but
`were limited by the small number of evaluated patients. Wolinsky et al. were able
`to demonstrate a definite, but modest eVect of GA on MRI enhancements of MS
`patients in the US open-label GA extension MRI trial for relap

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