throbber
Views & Reviews
`
`CME Mechanisms of action of glatiramer
`acetate in multiple sclerosis
`
`Oliver Neuhaus, MD; Cinthia Farina, PhD; Hartmut Wekerle, MD; and Reinhard Hohlfeld, MD
`
`Article abstract—Glatiramer acetate (GA, Copaxone [Teva Pharmaceuticals, Kansas City, MO], formerly known as
`copolymer-1) and interferon- (IFN)-b are both used for the immunomodulatory treatment of multiple sclerosis, but they
`act in different ways. Four major mechanisms of GA have been identified: 1) competition with myelin-basic protein (MBP)
`for binding to major histocompatibility complex (MHC) molecules; 2) competition of GA/MHC with MBP/MHC for binding
`to the T-cell receptor; 3) partial activation and tolerance induction of MBP-specific T cells (action as an altered peptide
`ligand); and 4) induction of GA-reactive T-helper 2- (TH2)-like regulatory cells. Of these four mechanisms, 1 and 2
`presumably occur only in vitro and are therefore irrelevant for the in vivo effects of GA. In contrast, mechanisms 3 and 4
`could occur in vivo and both could contribute to the clinical effects of GA.
`NEUROLOGY 2001;56:702–708
`
`Glatiramer acetate (GA, Copaxone [Teva Pharma-
`ceuticals, Kansas City, MO], formerly known as
`copolymer-1) and interferon- (IFN)-b are now widely
`used for the immunomodulatory treatment of MS.1-4
`The mechanisms of action of these agents, although
`not completely understood, seem to be fundamen-
`tally different. Whereas IFN-b exerts its multiple
`immunomodulatory effects in an antigen-nonspecific
`way, GA seems to preferentially affect immune cells
`specific for myelin basic protein (MBP) and perhaps
`other myelin antigens.5-7 This view rests mainly on
`evidence obtained in experimental autoimmune en-
`cephalomyelitis (EAE), an animal model of MS. Until
`recently the effects of GA on the human immune
`system were largely unknown. However, several new
`articles have shed light on the mechanisms of action
`of GA in MS. In this article, we briefly review these
`novel findings and put them into perspective with
`the previous observations in animal models.
`
`Clinical effects of GA in EAE and MS. GA is a
`standardized, randomized mixture of synthetic
`polypeptides consisting of L-glutamic acid, L-lysine,
`L-alanine, and L-tyrosine with a defined molar resi-
`due ratio of 0.14 : 0.34 : 0.43 : 0.09 and an average
`molecular mass of 4.7 to 11.0 kDa, i.e., an average
`length of 45 to 100 amino acids. It has been known
`for a long time that GA has both suppressive and
`protective effects in EAE induced by various enceph-
`alitogenic antigens in different species.5,8-13 With
`some exceptions (murine graft-versus-host disease
`
`[in doses higher than required to suppress EAE],14
`experimental uveoretinitis,15 and inhibition of type II
`collagen-reactive T cells in vitro16), GA seems to be
`ineffective in other autoimmune models.6 In addition
`to the subcutaneous (s.c.) route of administration,
`the oral form of GA has also been shown to be effec-
`tive in EAE.17,18 Daily s.c. administration of GA has
`beneficial effects on the clinical and MRI-defined
`course of patients with MS.19-24
`
`Overview of the immunologic effects of GA in
`EAE. During the last three decades, the pioneering
`work of Michael Sela, Ruth Arnon, and their col-
`leagues has laid the foundations for the approval
`of GA for use in the treatment of MS.6,25 A large
`body of experimental evidence in numerous EAE
`models suggests that GA acts by several different
`mechanisms7:
`Results of in vitro studies suggest that GA com-
`petes in some way with MBP.26-28 Specifically, GA
`competes with MBP at the antigen-presenting cell
`(APC) level for binding to the major histocompatibil-
`ity complex (MHC), and GA/MHC competes with
`MBP/MHC for binding to the T-cell receptor (TCR).
`GA binds to many different alleles of MHC class II
`molecules (“promiscuous binding”).29 Interestingly,
`the stereoisomer of GA, D-GA, which is composed of
`D-amino acids, binds as effectively to MHC class II30
`but fails to suppress EAE.31 This suggests that com-
`petition for MHC binding alone is insufficient to ex-
`plain the beneficial effects of GA.5 At the TCR level,
`
`From the Department of Neuroimmunology (Drs. Neuhaus, Farina, Wekerle, and Hohlfeld), Max-Planck Institute of Neurobiology, Martinsried; and the
`Institute for Clinical Neuroimmunology and the Department of Neurology (Dr. Hohlfeld), Ludwig Maximilians University, Munich, Germany.
`Supported by a grant from TEVA. O.N. is a postdoctoral fellow supported by Deutsche Forschungsgemeinschaft and Max-Planck-Gesellschaft. The Institute
`for Clinical Neuroimmunology is supported by the Hermann and Lilly Schilling Foundation. R.H. is a consultant to TEVA.
`Received September 29, 2000. Accepted in final form December 10, 2000.
`Address correspondence and reprint requests to Dr. Reinhard Hohlfeld, Institute for Clinical Neuroimmunology, Klinikum Grosshadern, Ludwig Maximil-
`ians University, Marchioninistrasse 15, 81366 Munich, Germany; e-mail: hohlfeld@neuro.mpg.de
`
`702 Copyright © 2001 by AAN Enterprises, Inc.
`
`Page 1 of 7
`
`YEDA EXHIBIT NO. 2065
`MYLAN PHARM. v YEDA
`IPR2015-00643
`
`

`
`GA has been reported to act as an antagonist of the
`antigenic peptide MBP 82-100, but not MBP 1-11
`and proteolipid protein (PLP) 139-151.32
`Results of in vivo studies indicate that GA induces
`regulatory T cells of the T-helper 2- (TH2)-type in
`the peripheral immune system outside the CNS.
`When spleen cells from GA-treated mice were adop-
`tively transferred into syngeneic animals, these cells
`protected from EAE induced by different CNS
`antigens.33-35 Earlier researchers demonstrated a
`similar inhibitory effect with a “soluble factor” ex-
`tracted from these cells,36 which were later identified
`as anti-inflammatory TH2 cytokines.34,35 Further
`support for the proposed protective role of GA-
`reactive regulatory T cells comes from the recent
`demonstration that GA-specific TH2 cells are
`present in the CNS of GA-treated mice.37 GA-specific
`T cells also have a neuroprotective effect after adoptive
`transfer into rats with experimental crush lesions of
`the optic nerve.38 This latter finding suggests the possi-
`bility that GA-specific TH2-like regulatory T cells not
`only provide protective cytokines such as interleukin-
`(IL)-4, IL-5, IL-13, and transforming growth factor-
`(TGF)-b, but also neurotrophic factors such as brain-
`derived neurotrophic factor (BDNF).38,39
`
`Immunologic effects of GA in human subjects.
`The different effects of GA are listed and compared
`with the effects of IFN-b in the table.
`High frequency of GA-reactive proliferating T cells
`In contrast to the lack of ef-
`in untreated subjects.
`fect of GA on immune cells isolated from untreated
`animals, GA induces vigorous proliferation of periph-
`eral blood lymphocytes (PBL)
`from untreated
`(unprimed) human subjects.40-42 Phenotypic analyses
`revealed that the GA-responsive T-cell population in
`untreated subjects is polyclonal43,44 and predomi-
`nantly originates from the memory T-cell pool.44 Re-
`cent findings indicate that the proliferative response
`to GA depends on both MHC class I- and MHC class
`II-restricted T cells.45
`Reduced proliferative response in GA-treated pa-
`tients. The proliferative response to GA decreases
`with time in GA-treated patients.6,42,46-48 Recent re-
`sults from our own group indicate that this decrease
`is specific to GA because it is not observed with re-
`call antigens like tetanus toxoid and tuberculin.49
`Using limiting dilution assays, Schmied et al. ob-
`served that the constitutively high frequency of GA-
`reactive T cells in untreated patients initially tends
`to increase during the first months of GA therapy
`and only later decreases below baseline.50 Theoreti-
`cally, the observed decrease in GA-reactive T cells
`could be caused by anergy induction or activation-
`induced cell death of GA-specific T cells.50
`Deviation from TH1 to TH2. TH cells can be di-
`vided into several types based on their characteristic
`cytokine secretion patterns and effector functions.51-55
`TH1 cells produce proinflammatory cytokines such as
`
`IL-2, IL-12, IFN-g, and tumor necrosis factor- (TNF)-a.
`In contrast, TH2 cells produce downregulatory cyto-
`kines such as IL-4, IL-5, IL-6, IL-10, and IL-13. TH1
`cells mediate proliferative and delayed hypersensitivity
`responses, whereas TH2 cells are involved in allergic
`pathways and support antibody production by B cells.52
`Different lines of evidence suggest that GA
`treatment induces a shift from TH1 to TH2. GA
`treatment was shown to increase serum IL-10 lev-
`els and TGF-b and IL-4 mRNA in PBL, whereas it
`suppressed TNF-a mRNA.55 Using intracellular
`double-immunofluorescence flow cytometry, we dem-
`onstrated that long-term GA-reactive T-cell lines
`(TCL) from patients with untreated MS and healthy
`controls predominantly produce IFN-g and are to be
`classified as TH1 cells, whereas GA-reactive TCL
`from patients with GA-treated MS predominantly
`produce IL-4, i.e., behave like TH2-cells.43 Recent ob-
`servations on short-term44,48 and long-term56 GA-
`reactive TCL by other groups are consistent with
`these findings. In contrast to MBP-reactive TCL,
`GA-reactive TCL secrete IL-6, a TH2-related cyto-
`kine.56 In addition, the IFN-g : IL-5 ratio was biased
`toward IFN-g in MBP-reactive TCL and toward IL-5
`in GA-reactive TCL, in both treated and untreated
`patients.42
`In contrast to the limiting dilution assay (which
`detects proliferation), results obtained with an auto-
`mated ELISPOT assay (which detects cytokine pro-
`duction of individual cells) indicate that during GA
`therapy, there is an increase of GA-reactive T cells
`producing IL-4 or IFN-g.49 Specifically, the study by
`Farina et al.49 demonstrated that patients with GA-
`treated MS show 1) a significant reduction of
`GA-induced proliferation of peripheral blood mono-
`nuclear cells; 2) a positive IL-4 ELISPOT response
`mediated predominantly by CD41 T cells after in
`vitro stimulation with a wide range of GA concentra-
`tions; and 3) an elevated IFN-g response partially
`mediated by CD81 T cells after stimulation with
`high GA concentrations. All three effects were GA-
`specific because they were not observed with control
`antigens.49 The GA-induced changes were stable over
`time and allowed the correct identification of GA-
`treated and untreated donors in most cases.49 It
`therefore appears that during therapy, GA-reactive
`T cells are not physically deleted, but rather they are
`modified in such a way that they respond to in vitro
`challenge with GA by secreting cytokines but not by
`proliferating.
`Effects on migratory potential of T cells. Using
`an in vitro model of lymphocyte migration, Prat et al.
`demonstrated that the migratory potential of lym-
`phocytes freshly isolated from GA-treated patients
`was reduced compared with untreated patients.57 In
`vitro treatment with IFN-b, but not GA, reduced
`lymphocyte migration rates, indicating that IFN-b
`acts directly on cell migration, whereas GA acts in-
`directly.57 GA did not change the expression of ad-
`hesion molecules on human brain microvascular
`endothelial cells.58 It is currently not known whether
`March (2 of 2) 2001 NEUROLOGY 56 703
`
`Page 2 of 7
`
`YEDA EXHIBIT NO. 2065
`MYLAN PHARM. v YEDA
`IPR2015-00643
`
`

`
`Figure. Schematic view of the putative
`mechanism of action of glatiramer ace-
`tate (GA). In the periphery, outside the
`CNS, GA initially stimulates a popula-
`tion of TH1-like T cells. During treat-
`ment, the properties of the GA-stimulated
`T cells change, and they become more
`TH2-like (dotted arrow). The activated
`GA-specific T cells enter the CNS, where
`they encounter CNS antigens like MBP
`bound to MHC class II and presented on
`the surface of microglia cells. The GA-
`reactive T cells are stimulated to secrete
`downmodulatory cytokines like IL-4,
`which exert a bystander suppressive ef-
`fect on other T cells. TCR 5 T-cell recep-
`tor; MHC 5 major histocompatibility
`complex; Ag 5 antigen.
`
`the reduced migratory potential is related to the
`TH1-to-TH2
`shift
`observed with GA-specific
`TCL.42-44,48,56
`Cross-stimulation with MBP and other antigens.
`GA was originally developed to mimic MBP for EAE
`induction.6,8 Surprisingly, however, it turned out
`that GA inhibits MBP-induced EAE.6 Ever since
`then, some form of partial cross-reaction or competi-
`tion between MBP and GA has been considered cru-
`cial to the mechanism of GA. Most investigators of
`the human immune response to GA found that GA is
`not cross-reactive with MBP at the level of prolifera-
`tion.41,56 An exception was reported by Gran et al.,
`who observed that a small number of GA-reactive
`TCL isolated from a patient treated with GA for
`more than 6 years proliferated in response to whole
`MBP and peptide MBP 83-99.44
`Despite the lack of cross-stimulation at the prolif-
`eration level, there is clear evidence that GA and
`MBP may cross-stimulate T cells at the level of cyto-
`kine production. In our own study, about 10% of the
`tested GA-specific T cell
`lines could be cross-
`stimulated with MBP to produce low levels of cyto-
`In these experiments, TH1-type TCL
`kines.43
`preferentially produced IFN-g, whereas TH2-type
`TCL produced IL-4.43 Similar results were reported
`by others, using either MBP-specific TCL42 for cross-
`stimulation with GA, or GA-reactive TCL44 for cross-
`stimulation with MBP.
`Interestingly, two of our GA-specific T-cell lines
`could be stimulated to produce IFN-g with another
`myelin autoantigen, myelin-oligodendrocyte glyco-
`protein (MOG).43 This indicates that the cross-
`stimulatory effect on cytokine production is not
`entirely restricted to MBP. It may also occur with
`other autoantigens. Indeed, one group of investiga-
`tors found evidence that the immune response to GA
`in GA-treated patients becomes more “degenerate.”48
`The authors reported that with increasing duration
`of treatment, the surviving GA-reactive T cells re-
`sponded to an increasing number of components from
`a combinatorial peptide library.48 These observations
`704 NEUROLOGY 56 March (2 of 2) 2001
`
`would add an interesting facet to the mechanism of GA
`action, implying that GA-reactive, TH2-like (protective)
`T cells might be activated not only by MBP, but also by
`other cross-reactive antigens. This might help to ex-
`plain why GA had beneficial effects not only in EAE
`induced by MBP but also by MOG or PLP, and also in
`a few other experimental autoimmune diseases.6,14,15
`Inhibition of MBP-specific T cells by GA. GA was
`found to inhibit the MBP-induced proliferation and
`IL-2 secretion of human MBP-specific TCL.28 A triv-
`ial toxic effect of GA could be excluded because the
`inhibition was overcome by increasing the concen-
`tration of MBP.28 A control antigen, tuberculin pu-
`rified protein derivative (PPD), did not have any
`inhibitory effect.28 GA also inhibited influenza vi-
`rus hemagglutinin- and Borrelia burgdorferi-
`specific T-cell clones, although to a lesser extent
`than MBP-specific T cells.44 In addition, GA was
`shown to inhibit the cytolytic ability of human MBP-
`specific TCL restricted to MS-associated HLA-DR
`types.59 In a recent study, Gran et al. showed that
`GA inhibited IFN-g production in MBP-reactive T
`cells in a dose-dependent manner and had a less
`pronounced effect on the secretion of IL-4 and IL-5.44
`In a subset of the analyzed T-cell clones, GA had a
`differential effect, i.e., it inhibited proliferation and
`IFN-g production and induced IL-4 and IL-5 secre-
`tion. This indicates a differential influence of GA on
`the T-cell activation parameters.44 Furthermore, GA
`was shown to induce a state of nonresponsiveness
`(anergy) in MBP-specific T-cell clones.44 In principle,
`these inhibitory effects could occur at the level of
`binding of GA to the MHC or to the TCR.
`Interaction with MHC. GA binds to purified
`HLA-DR molecules without antigen processing29 and
`without any obvious preference for particular al-
`leles.60 Although distinct binding motifs of GA to MS-
`associated HLA-DR molecules could be defined,
`virtually all of the GA polypeptides seem to have a
`binding capacity to MHC class II, owing perhaps to
`their random composition.61 In addition to MHC
`class II molecules, GA also seems to be able to inter-
`
`Page 3 of 7
`
`YEDA EXHIBIT NO. 2065
`MYLAN PHARM. v YEDA
`IPR2015-00643
`
`

`
`Table Comparison of major immunologic effects of glatiramer acetate (GA) and interferon (IFN)-b
`
`Effects
`
`T-cell proliferation in vitro
`
`Proliferation during
`treatment
`Regulation of MHC
`expression
`MHC binding
`
`T-cell migration
`
`TH2 shift in PBL
`
`TH2 shift in GA-specific T
`cells
`Cross-stimulation with
`MBP
`Cross-inhibition of MBP-
`specific TCL
`TCR antagonism
`
`APL effect on MBP-
`specific T cells
`Effects on antigen-
`presenting cells
`
`GA
`
`IFN-b71-73
`
`Suppression of proliferation of MBP-reactive T
`cells in vitro28
`Decreased proliferation of PBL to GA during
`treatment6,42,47-49
`No known effect
`
`Direct and promiscuous binding of GA to different
`HLA-DR alleles60,61
`Reduced migration of PBL from GA-treated
`patients (unknown mechanism)57; no effect on
`adhesion molecule expression on human brain
`microvascular endothelial cells58
`
`Increased levels of IL-10 in serum and of mRNA
`for TGF-b and IL-455; reduction of mRNA for
`TNF-a in PBL55
`Shift of GA-reactive T cells from TH1 towards TH2
`during GA treatment43,48
`Induction by GA of cytokine production in MBP-
`specific T cells and vice versa42-44
`Inhibition of proliferation of T cells specific for
`MBP and some other antigens28,44
`TCR antagonism with MBP 82–100 (controversial
`findings)32,44
`Induction of anergy in MBP-specific T-cell clones44
`
`Antigen-nonspecific suppression of T-cell
`proliferation74,75
`No known effect
`
`Inhibition of IFN-g-induced upregulation of MHC
`class II expression76,77
`No known effect
`
`Reduced T-cell migration caused by inhibition of
`matrix metalloproteinases (MMP),78-80 increase of
`soluble adhesion molecules (sICAM-1, sVCAM-
`1),80,81 and decrease of surface-expressed
`adhesion molecules (VLA-4)82
`Induction of TH2 cytokines and reduction of TH1
`cytokines in PBL83-85
`
`No known effect
`
`No known effect
`
`No known effect
`
`No known effect
`
`No known effect
`
`Inhibition of TNF-a and cathepsin-B production in
`a monocytic cell line62
`
`Several effects, e.g., inhibition of IFN-g induction
`of FcgRI expression in monocytes86
`
`Due to space limitations, only representative articles are cited for each mechanism.
`
`APL 5 altered peptide ligand; HLA 5 human histocompatibility leukocyte antigen; IL 5 interleukin; MBP 5 myelin-basic protein;
`MHC 5 major histocompatibility complex; mRNA 5 messenger ribonucleic acid; PBL 5 peripheral blood lymphocytes; sICAM 5 soluble
`intercellular adhesion molecule; sVCAM 5 soluble vascular cell adhesion molecule; TCL 5 T-cell lines; TCR 5 T-cell receptor; TGF 5
`transforming growth factor; TH 5 T-helper; TNF 5 tumor necrosis factor; VLA 5 very late antigen.
`
`act with MHC class I.45,49 It is therefore likely that
`part of the inhibitory effects described in the previ-
`ous section occur by competition between GA and
`other antigens for MHC binding. Clearly, this type of
`competition could occur with any antigen and is
`therefore antigen-nonspecific. However, for reasons
`explained below, this mechanism is probably irrele-
`vant for the in vivo effects of GA.
`In addition to competition
`Interaction with TCR.
`at the MHC class II level, and consistent with ani-
`mal data, GA was reported to act as a “TCR antago-
`nist” against the MBP 82-100 peptide.32 In contrast,
`in another study TCR-antagonistic effects were not
`observed.44 However, when MBP-specific TCL were
`stimulated with different concentrations of GA, their
`subsequent response to the nominal antigen MBP
`was turned off, i.e., the T cells had been “anergized”
`by GA.44 This suggests that a specific TCR engage-
`ment by GA does occur. This mechanism may be
`relevant to the in vivo effects of GA because it does
`
`not require the simultaneous presence of the nomi-
`nal antigen (e.g., MBP).
`Effect on monocytes. Although the vast majority
`of evidence suggests that GA acts primarily at the
`level of T cells, additional effects on other immune
`cells cannot be excluded. For example, GA was
`reported to inhibit a human monocytic cell line,
`THP-1.62 In THP-1 cells stimulated with lipopoly-
`saccharide or IFN-g, GA reduced the percentage of
`cells expressing HLA-DR and DQ antigen and inhib-
`ited the production of TNF-a and cathepsin-B. In
`contrast, the production of IL-1b was increased.62
`The mechanism of these effects and their relevance
`to the overall mechanism of action of GA currently
`are unknown.
`
`Conclusion: proposed mechanism of action of
`GA in multiple sclerosis. The results discussed
`in the previous sections suggest four major effects of
`GA on human T cells:
`March (2 of 2) 2001 NEUROLOGY 56 705
`
`Page 4 of 7
`
`YEDA EXHIBIT NO. 2065
`MYLAN PHARM. v YEDA
`IPR2015-00643
`
`

`
`1. GA binds “promiscuously” to MHC class II and
`perhaps MHC class I molecules, thereby competing
`with the MHC binding of other antigens. This effect,
`which by its nature is antigen-nonspecific, is un-
`likely to play a role in vivo because after s.c. admin-
`istration GA is quickly degraded to free amino acids
`and small oligopeptides.63 Therefore it is not likely to
`reach the CNS where it could compete with the rele-
`vant auto-antigens for MHC binding.
`2. GA/MHC competes with MBP for binding to the
`antigen-specific surface receptor of MBP-specific T cells
`(“TCR antagonism”). The experimental evidence sup-
`porting this effect is controversial. If it occurs, it is
`unlikely to be relevant in vivo because GA is unlikely
`to reach sites where it could compete with MBP.
`3. GA/MHC binds to the TCR of T cells specific for
`MBP and, perhaps, other myelin antigens. In this
`view, GA acts like an “altered peptide ligand” (APL)
`relative to MBP. As a consequence, some of the
`myelin-specific, pathogenic T cells might become “an-
`ergic” or be otherwise changed in their properties,
`e.g., in their migratory potential. This effect would
`be relatively antigen-specific and presumably occur
`in the periphery at the injection sites or in their
`draining lymph nodes where the MBP-specific T cells
`might be confronted with GA. Although some in vitro
`findings support this mechanism, it is not yet known
`whether the functional properties of MBP-specific T
`cells are altered in GA-treated patients. It may be of
`relevance in this connection that we were unable to
`isolate MBP-specific TCL from GA-treated patients.43
`4. GA treatment induces a TH1-to-TH2 shift in GA-
`reactive T cells in vivo. The GA-reactive T cells act
`as regulatory cells and have beneficial effects on the
`pathogenic autoimmune reaction. Compared with
`the other putative mechanisms, this currently has
`the strongest experimental support. We would like to
`propose the following scenario (figure): GA-reactive
`TH2-like T cells are able to cross the blood-brain
`barrier because they are activated by daily immuni-
`zation.64 During treatment, the properties of the GA-
`reactive T cells are changed in such a way that they
`increasingly become TH2-like.43,48 Inside the CNS,
`the GA-reactive T cells are confronted with products
`of myelin turnover presented by local APC.65 Some of
`the GA-reactive cells cross-react with MBP or MOG
`and are therefore stimulated to release anti-
`inflammatory cytokines such as IL-4, IL-6, IL-10,
`and even neurotrophic factors.38,39,66 Subsequently,
`the production of pro-inflammatory cytokines such
`as IL-2 and IFN-g by other inflammatory cells is
`reduced via a suppressive bystander effect.34,35,67,68
`Hypothetically, a similar process might occur in the
`periphery: Any viral or bacterial antigens that stim-
`ulate peripheral MBP-specific T cells by molecular
`mimicry69,70 might also activate peripheral GA-
`specific, cross-reactive downregulatory T cells.
`
`Acknowledgment
`The authors thank Ms. Judy Benson for her helpful comments and
`careful reading of the manuscript.
`
`706 NEUROLOGY 56 March (2 of 2) 2001
`
`References
`1. Wolinsky JS. Copolymer 1: a most reasonable alternative
`therapy for early relapsing-remitting multiple sclerosis with
`mild disability. Neurology 1995;45:1245–1247.
`2. Noseworthy JH. Progress in determining the causes and treat-
`ment of multiple sclerosis. Nature 1999;399(suppl):A40–A47.
`3. Milo R, Panitch H. Glatiramer acetate or interferon-b for mul-
`tiple sclerosis? A guide to drug choice. CNS Drugs 1999;11:
`289–306.
`4. Wolinsky JS. Glatiramer acetate. In: Hawkins CP, Wolinsky
`JS, eds. Principles of treatments in multiple sclerosis. Oxford,
`Woburn: Butterworth-Heinemann, 2000:71–94.
`5. Arnon R, Sela M, Teitelbaum D. New insights into the mech-
`anism of action of copolymer 1 in experimental allergic en-
`cephalomyelitis and multiple sclerosis. J Neurol 1996;
`243(suppl 1):S8–S13.
`6. Arnon R. The development of Cop-1 (Copaxone®), an innova-
`tive drug for the treatment of multiple sclerosis: personal re-
`flections. Immunol Lett 1996;50:1–15.
`7. Teitelbaum D, Arnon R, Sela M. Cop 1 as a candidate drug for
`multiple sclerosis. J Neural Transm 1997;49(suppl):85–91.
`8. Teitelbaum D, Meshorer A, Hirshfeld T, Arnon R, Sela M.
`Suppression of experimental allergic encephalomyelitis by a
`synthetic polypeptide. Eur J Immunol 1971;1:242–248.
`9. Teitelbaum D, Webb C, Meshorer A, Arnon R, Sela M. Sup-
`pression by several synthetic polypeptides of experimental al-
`lergic encephalomyelitis induced in guinea pigs and rabbits
`with bovine and human basic encephalitogen. Eur J Immunol
`1973;3:273–279.
`10. Teitelbaum D, Webb C, Bree M, Meshorer A, Arnon R, Sela M.
`Suppression of experimental allergic encephalomyelitis in rhe-
`sus monkeys by a synthetic basic copolymer. Clin Immunol
`Immunopathol 1974;3:256–262.
`11. Lisak RP, Zweiman B, Blanchard N, Borke LB. Effect of treat-
`ment with copolymer 1 (Cop-1) on the in vivo and in vitro
`manifestations of experimental allergic encephalomyelitis
`(EAE). J Neurol Sci 1983;62:281–293.
`12. Teitelbaum D, Fridkis-Hareli M, Arnon R, Sela M. Copolymer
`1 inhibits chronic relapsing experimental allergic encephalo-
`myelitis induced by proteolipid protein (PLP) peptides in mice
`and interferes with PLP-specific T cell responses. J Neuroim-
`munol 1996;64:209–217.
`13. Ben-Nun A, Mendel I, Bakimer R, et al. The autoimmune
`reactivity to myelin oligodendrocyte glycoprotein (MOG) in
`multiple sclerosis is potentially pathogenic: effect of copolymer
`1 on MOG-induced disease. J Neurol 1996;243(suppl 1):S14–
`S22.
`14. Schlegel PG, Aharoni R, Chen Y, et al. A synthetic random
`basic copolymer with promiscuous binding to class II major
`histocompatibility complex molecules inhibits T-cell prolifera-
`tive responses to major histocompatibility antigens in vitro
`and confers the capacity to prevent murine graft-versus-host
`disease in vivo. Proc Natl Acad Sci USA 1996;93:5061–5066.
`15. Zhang M, Chan CC, Vistica B, Hung V, Wiggert B, Gery I.
`Copolymer 1 inhibits experimental autoimmune uveoretinitis.
`J Neuroimmunol 2000;103:189–194.
`16. Fridkis-Hareli M, Rosloniec EF, Fugger L, Strominger JL.
`Synthetic amino acid copolymers that bind to HLA-DR pro-
`teins and inhibit type II collagen-reactive T cell clones. Proc
`Natl Acad Sci USA 1998;95:12528–12531.
`17. Teitelbaum D, Arnon R, Sela M. Immunomodulation of ex-
`perimental autoimmune encephalomyelitis by oral adminis-
`tration of copolymer 1. Proc Natl Acad Sci USA 1999;96:
`3842–3847.
`18. Weiner HL. Oral tolerance with copolymer 1 for the treatment
`of multiple sclerosis. Proc Natl Acad Sci USA 1999;96:3333–
`3335.
`19. Bornstein MB, Miller A, Slagle S, et al. A pilot trial of Cop 1
`in exacerbating–remitting multiple sclerosis. N Engl J Med
`1987;317:408–414.
`20. Johnson KP, Brooks BR, Cohen JA, et al. Copolymer 1 reduces
`relapse rate and improves disability in relapsing-remitting
`multiple sclerosis. Results of a phase III multicenter, double-
`blind, placebo-controlled trial. Neurology 1995;45:1268–1276.
`21. Johnson KP, Brooks BR, Cohen JA, et al. Extended use of
`glatiramer acetate (Copaxone) is well tolerated and maintains
`
`Page 5 of 7
`
`YEDA EXHIBIT NO. 2065
`MYLAN PHARM. v YEDA
`IPR2015-00643
`
`

`
`its clinical effect on multiple sclerosis relapse rate and degree
`of disability. Neurology 1998;50:701–708.
`22. Comi G, Filippi M, for the Copaxone MRI Study Group. The
`effect of glatiramer acetate (Copaxone®) on disease activity as
`measured by cerebral MRI in patients with relapsing-remitting
`multiple sclerosis (RRMS): a multi-center, randomized, double
`blind, placebo-controlled study extended by open-label treat-
`ment. Neurology 1999;52(suppl 2):A289. Abstract.
`23. Mancardi GL, Sardanelli F, Parodi RC, et al. Effect of
`copolymer-1 on serial gadolinium enhanced MRI in relapsing
`remitting multiple sclerosis. Neurology 1998;50:1127–1133.
`24. Ge Y, Grossman RI, Udupa JK, et al. Glatiramer acetate
`(Copaxone) treatment in relapsing-remitting MS. Quantita-
`tive MR assessment. Neurology 2000;54:813–817.
`25. Sela M. Probing into the realm of proteins and immunity.
`Protein Sci 1998;7:1653–1659.
`26. Webb C, Teitelbaum D, Arnon R, Sela M. In vivo and in vitro
`immunological cross-reactions between basic encephalitogen
`and synthetic basic polypeptides capable of suppressing exper-
`imental allergic encephalomyelitis. Eur J Immunol 1973;3:
`279–286.
`27. Teitelbaum D, Aharoni R, Arnon R, Sela M. Specific inhibition
`of the T cell response to myelin basic protein by the synthetic
`copolymer Cop-1. Proc Natl Acad Sci USA 1988;85:9724–9728.
`28. Teitelbaum D, Milo R, Arnon R, Sela M. Synthetic copolymer
`1 inhibits human T-cell lines specific for myelin basic protein.
`Proc Natl Acad Sci USA 1992;89:137–141.
`29. Fridkis-Hareli M, Teitelbaum D, Gurevich E, et al. Direct
`binding of myelin basic protein and synthetic copolymer 1 to
`class II major histocompatibility complex molecules on living
`antigen-presenting cells—specificity and promiscuity. Proc
`Natl Acad Sci USA 1994;91:4872–4876.
`30. Aharoni R, Schlegel PG, Teitelbaum D, et al. Studies on the
`mechanism and specificity of the synthetic copolymer GLAT
`on graft-versus-host disease. Immunol Lett 1997;58:79–87.
`31. Webb C, Teitelbaum D, Herz A, Arnon R, Sela M. Molecular
`requirements involved in suppression of EAE by synthetic
`basic copolymers of amino acids. Immunochemistry 1976;13:
`333–337.
`32. Aharoni R, Teitelbaum D, Arnon R, Sela M. Copolymer 1 acts
`against the immunodominant epitope 82-100 of myelin basic
`protein by T cell receptor antagonism in addition to major
`histocompatibility complex blocking. Proc Natl Acad Sci USA
`1999;96:634–639.
`33. Aharoni R, Teitelbaum D, Arnon R. T suppressor hybridomas
`and interleukin-2 dependent lines induced by copolymer 1 or
`by spinal cord homogenate down-regulate experimental aller-
`gic encephalomyelitis. Eur J Immunol 1993;23:17–25.
`34. Aharoni R, Teitelbaum D, Sela M, Arnon R. Copolymer 1
`induces T cells of the T helper type 2 that crossreact with
`myelin basic protein and suppress experimental autoimmune
`encephalomyelitis. Proc Natl Acad Sci USA 1997;94:10821–
`10826.
`35. Aharoni R, Teitelbaum D, Sela M, Arnon R. Bystander sup-
`pression of experimental autoimmune encephalomyelitis by T
`cell lines and clones of the Th2 type induced by copolymer 1.
`J Neuroimmunol 1998;91:135–146.
`36. Lando Z, Dori Y, Teitelbaum D, Arnon R. Unresponsiveness to
`experimental allergic encephalomyelitis in mice: replacement
`of suppressor cells by a soluble factor. J Immunol 1981;127:
`1915–1919.
`37. Aharoni R, Teitelbaum D, Leitner O, Meshorer A, Sela M,
`Arnon R. Specific TH2 cells are present in the central nervous
`system of mice protected against EAE by copolymer 1. Proc
`Natl Acad Sci USA 2000;97:11472–11477.
`38. Kipnis J, Yoles E, Porat Z, et al. T cell immunity to copolymer
`1 confers neuroprotection on the damaged optic nerve: possi-
`ble therapy for optic neuropathies. Proc Natl Acad Sci USA
`2000;97:7446–7451.
`39. Kerschensteiner M, Gallmeier E, Behrens L, et al. Activated
`human T cells, B cells, and monocytes produce brain-derived
`neurotrophic factor (BDNF) in vitro and in brain lesions: a
`neuroprotective role of inflammation? J Exp Med 1999;189:
`865–870.
`40. Brosnan CF, Litwak M, Neighbour PA, et al. Immunogenic
`potentials of copolymer 1 in normal human lymphocytes. Neu-
`rology 1985;35:1754–1759.
`
`41. Burns J, Krasner LJ, Guerrero F. Human cellular immune
`response to copolymer I and myelin basic protein. Neurology
`1986;36:92–94.
`42. Qin Y, Zhang DQ, Prat A, Pouly S, Antel J. Characterization
`of T cell lines derived from glatiramer-acetate treated multi-
`ple sclerosis patients. J Neuroimmunol 2000;108:201–206.
`43. Neuhaus O, Farina C, Yassouridis A, et al. Multiple sclerosis:
`comparison of copolymer-1 reactive T cell lines from treated
`and untreated subjects reveals cytokine shift from T helper 1
`to T helper 2 cells. Proc Natl Acad Sci USA 2000;97:7452–
`7457.
`44. Gran B, Tranquill LR, Chen M, et al. Mechanisms of immuno-
`modulation by glatiramer acetate. Neurology 2000;55:1704–
`1714.
`45. Ragheb S, Lisak RP. The lym

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