throbber
Neurotherapeutics: The Journal of the American Society for Experimental NeuroTherapeutics
`
`Mechanism of Action of Glatiramer Acetate in Treatment of
`Multiple Sclerosis
`
`Martin S. Weber,* Reinhard Hohlfeld,†‡ and Scott S. Zamvil*
`
`*Department of Neurology and Program in Immunology, University of California, San Francisco, CA 94143; †Institute for
`Clinical Neuroimmunology, Klinikum Grosshadern, Ludwig Maximilians University, Munich, 81377, Germany; and ‡Department
`of Neuroimmunology, Max Planck Institute of Neurobiology, Martinsried, 82152, Germany
`
`Summary: Glatiramer acetate (GA) (Copolymer-1, Copaxone,
`Teva, Israel, YEAK) is a polypeptide-based therapy approved for
`the treatment of relapsing-remitting multiple sclerosis. Most in-
`vestigations have attributed the immunomodulatory effect of GAs
`to its capability to alter T-cell differentiation. Specifically, GA
`treatment is believed to promote development of Th2-polarized
`GA-reactive CD4⫹ T-cells, which may dampen neighboring in-
`flammation within the central nervous system. Recent reports in-
`dicate that the deficiency in CD4⫹CD25⫹FoxP3⫹ regulatory T-
`
`cells in multiple sclerosis is restored by GA treatment. GA also
`exerts immunomodulatory activity on antigen presenting cells,
`which participate in innate immune responses. These new findings
`represent a plausible explanation for GA-mediated T-cell immune
`modulation and may provide useful insight for the development of
`new and more effective treatment options for multiple sclerosis.
`Key Words: Multiple sclerosis, glatiramer acetate, immuno-
`modulatory agents, mechanism of action, antigen presenting
`cells.
`
`INTRODUCTION
`
`Glatiramer acetate (GA) (Copolymer-1, Copaxone,
`Teva, Israel, YEAK) is a pool of synthetic peptides ran-
`domly composed of L-tyrosine (Y), L-glutamic acid (E),
`L-alanine (A), and L-lysine (K) with an average length of
`40 to 100 residues. GA was synthesized in this manner
`more than 30 years ago to most closely resemble the
`encephalitogenic properties of myelin basic protein
`(MBP), one suspected auto-antigen in multiple sclerosis
`(MS). Surprisingly, instead of inducing experimental au-
`toimmune encephalomyelitis (EAE), the murine model
`of MS, immunizations with GA protected mice from
`subsequent attempts to induce EAE.1 This seminal ob-
`servation was
`followed by various clinical
`trials.
`Whereas early open-label studies already suggested clin-
`ical benefit in the 1980s,2,3 these findings had to be
`interpreted with caution as drug production was not yet
`standardized. In 1991, a phase III multicenter, double-
`blind, placebo-controlled trial with standardized GA
`preparation was initiated in 11 medical centers in the
`
`Address correspondence and reprint requests to: Scott S. Zamvil,
`M.D., Ph.D., Department of Neurology, Program in Immunology, Uni-
`versity of California, San Francisco, 513 Parnassus Avenue, S-268, San
`Francisco, CA 94143. E-mail: zamvil@ucsf.neuroimmnunol.org.
`
`United States, with 251 relapsing–remitting MS pa-
`tients.4 Within two years of treatment, the relapse rate
`decreased approximately 30% in GA-treated patients
`leading to approval of GA treatment of MS in many
`countries worldwide in 1995. A later double-blind, pla-
`cebo-controlled study demonstrated a reduction in the
`number of gadolinium-enhancing lesions in patients re-
`ceiving GA compared to a placebo during a nine-month
`study period.5 Additional data showed that GA may also
`have a favorable effect in preventing tissue loss at a later
`diseased stage.6,7 Based on these favorable clinical and
`imaging data, subcutaneously administered GA is one of
`the most widely prescribed drugs used today for the
`treatment of relapsing–remitting MS.
`Many investigators have attempted to address the im-
`munologic basis for the clinical effects of GA in MS and
`MS models.8,9 Although different potential mechanisms
`have been considered, most investigations have attrib-
`uted the immunomodulatory activity of GA to alterations
`in T-cell antigen reactivity, focusing on its influence on
`the adaptive immune response. Early in vitro studies
`established that GA can bind to major histocompatibility
`complex (MHC) class II molecules and suggested that
`GA might preferentially alter presentation of myelin an-
`tigens to auto-reactive T-cells.10,11 Studies in EAE and
`MS have extensively demonstrated that GA treatment
`
`Vol. 4, 647– 653, October 2007 © The American Society for Experimental NeuroTherapeutics, Inc.
`
`647
`
`MYLAN PHARMS. INC. EXHIBIT 1090 PAGE 1
`
`

`
`648
`
`WEBER ET AL.
`
`promotes development of Th2-polarized GA-reactive
`CD4⫹ T cells.1,12–14 These Th2 cells can potentially
`accumulate in the CNS15 where they might release anti-
`inflammatory cytokines12,15–17 and neurotrophic fac-
`tors,18 dampening the activity of nearby auto-aggressive
`T cells, a process known as “bystander suppression.”15
`Recent reports indicate that GA treatment also exerts
`immunomodulatory activity on antigen presenting cells
`(APCs),19 –23 a part of the innate immune system. These
`newer findings may provide a plausible explanation for
`the observed Th2 deviation under GA treatment and raise
`the question as to whether GA is solely an antigen-
`specific T cell-directed immunotherapeutic agent as cur-
`rently assumed. In this article, we review the various
`effects of GA on the adaptive and innate immune system
`and describe how these two arms of the immune system
`interact with one another during GA therapy.
`
`Effects on the adaptive immune system
`As with conventional peptide antigens, GA can bind to
`MHC class II molecules on the surface of APCs.10,24 In
`association with MHC class II molecules, GA is recog-
`nized by T cells via their antigen-specific T-cell receptor.
`Early in vitro studies indicated that GA may compete
`with myelin antigens for the binding to MHC class II.
`Specifically, it was observed that GA binding to MHC
`class II could inhibit the activation of T cell lines specific
`for MBP.25 However, a later study demonstrated that the
`stereoisomer of GA, D-GA, which contains solely D-
`amino acids, could effectively bind to MHC class II,26
`but failed to suppress EAE.27 These findings indicate that
`GA may not primarily act as an MHC class II antagonist.
`It is well established that in most MS patients, GA
`treatment induces a population of CD4⫹ GA-reactive
`Th2 cells,12–14,28 which is associated with clinical bene-
`fit.29 It seems very unlikely that sufficient amounts of
`GA can reach the CNS to locally activate GA-reactive T
`cells. It is believed that GA-reactive Th2 cells are gen-
`erated in the periphery, accumulate (along with patho-
`genic non-GA-specific elements) in the CNS of patients
`with MS and release anti-inflammatory cytokines in a
`process termed “bystander suppression” (see FIG. 1).
`Many studies in EAE and MS have generated the con-
`cept
`that GA-reactive Th2 cells may be reactivated
`within the CNS through cross-recognition of myelin an-
`tigen.12,16 This assumption was supported by two obser-
`vations. First, GA-reactive Th2 cells could be identified
`in the CNS of GA-treated mice protected from EAE.15
`Second, in some but not all studies,30 several GA-spe-
`cific Th2 cell lines generated from MS patients or mice
`could cross react with MBP at the level of cytokine
`secretion.12,15–17,31
`CD4⫹CD25⫹ regulatory T cells (Treg) are an important
`subclass of regulatory cells that engage in the maintenance
`of immunologic tolerance by actively suppressing self-
`
`Neurotherapeutics, Vol. 4, No. 4, 2007
`
`reactive lymphocytes.32,33 Forkhead transcription factor
`Foxp3 is the key transcription factor in the physiological
`development of Treg.34 Its genetic defect results in im-
`paired function of Treg, which is associated with in an
`autoimmune and inflammatory syndrome in humans as
`well as in mice.35 Similarly, the experimental deletion of
`Treg in mice causes various spontaneous organ-specific
`autoimmune diseases.36 Viglietta et al.37 reported that in
`patients with MS, similar to other autoimmune condi-
`tions,38 effector function and frequency of Treg is sig-
`nificantly decreased in the peripheral blood. Several
`studies provided evidence for a role and mechanism of
`action of GA in the induction of CD4⫹CD25⫹ Treg. In
`vitro exposure to GA resulted in an elevated production
`of interleukin-10 (IL)-10 by Treg.39 In another study, GA
`promoted the conversion of CD4⫹CD25- to CD4⫹CD25⫹
`Treg through the activation of Foxp3.40 GA treatment led to
`a significant increase in Foxp3 expression in CD4⫹ T cells
`in MS patients whose Foxp3 expression was reduced at
`baseline. GA-reactive CD4⫹CD25⫹ T-cell lines generated
`from GA-treated MS patients expressed high levels of
`Foxp3 that correlated with increased T-cell regulation.40
`Thus, besides the well-known preferential Th2 differentia-
`tion of T cells, GA appears to normalize frequency and
`function of Treg in MS, which represents an additional
`immunomodulatory effect of GA.
`More recently, it was reported that GA treatment also
`induces a population of CD8⫹ GA-reactive T cells. In
`untreated MS patients, GA-reactive CD8⫹ T-cell re-
`sponses were found to be significantly lower compared
`with healthy individuals. Treatment with GA restored
`these CD8⫹ responses41 and enhanced release of IFN-␥
`by these cells,42 which appears to be associated with a
`positive clinical response.42 Although the in vivo func-
`tion of these cells is still not entirely understood, a recent
`report indicated that GA-reactive CD8⫹ T cells may
`suppress pro-inflammatory effector T-cell function in a
`manner similar to CD4⫹CD25⫹ Treg.43,44
`Besides activation and alteration of T cells, GA treat-
`ment also induces a humoral response to itself in most
`patients, which peaks approximately 3 months after treat-
`ment initiation.45 Just as individuals who are naive to GA
`treatment sometimes have pre-existing (naive) GA reac-
`tive T cells,28 some untreated MS patients reveal an
`unprimed humoral response against GA, mainly of an
`IgM, IgG1, and IgG2 isotype.46 GA-treated MS patients
`also produce IgG1 and IgG2 anti-GA antibodies, but in
`contrast to unexposed individuals, GA-treated MS pa-
`tients frequently develop high titers of IgG4 antibodies
`against GA.46 Preferential secretion of IgG4 antibodies
`might occur secondary to the induction of GA-reactive
`Th2 cells, as isotype switching to IgG4 is regulated by
`the Th2 cytokine IL-4. To date, it is considered contro-
`versial whether antibodies against GA are of clinical
`relevance. In general, IgG4 antibodies have strong neu-
`
`MYLAN PHARMS. INC. EXHIBIT 1090 PAGE 2
`
`

`
`GLATIRAMER ACETATE TO TREAT MS
`
`649
`
`Periphery
`
`CNS
`
`?
`
`CD4+CD25+
`FoxP3+ Treg
`
`Y
`GA/Ag
`
`MHC
`
`Type II
`APC
`
`GA/Ag
`
`Y
`
`CD4+
`Th-2
`
`CD4+
`Th-2
`
`YAg
`
`Type II
`APC
`
`local
`or
`recruited
`
`Neurotrophic
`factors
`
`Anti-inflammatory
`cytokines
`
`Type II
`microglia
`
`GA
`
`CD8+
`T cell
`
`?
`
`?
`
`Plasma cell
`
`Y Y
`Y
`GA-reactive IgG4
`Y
`Y
`FIG. 1. Cross-talk between type II antigen presenting cells (APCs) and regulatory T-cell populations in glatiramer acetate (GA)-mediated
`immune modulation. GA treatment exerts effects on APC and T cells that result in the induction of a specific population of Th2 cells and
`CD4⫹CD25⫹FoxP3⫹ regulatory T cells (Treg) in the periphery. Type II APC and Th2 cells may facilitate the development of each in a
`positive feedback mechanism, as type-2 monocytes tend to induce Th2 cells, and Th2 cell-derived anti-inflammatory cytokines may
`promote development of type II APC. GA-reactive Th2 cells are believed to cross the blood-brain barrier and to be locally reactivated
`within the CNS through cross recognition of myelin antigen. In response, these cells may secrete anti-inflammatory cytokines and
`neurothrophic factors dampening neighboring inflammation (“bystander suppression”). Another feedback loop between APC and T cells
`may develop within the CNS, as Th2-cytokines might promote type II differentiation of resident APC, such as microglia. GA treatment
`is also associated with induction of GA-reactive CD8⫹ T cells, although their in vivo function remains to be determined. Finally,
`consistent with the Th2 shift, GA-reactive plasma cells secrete anti-GA antibodies ( Ɱ), preferentially of an IgG4 isotype. Whether these
`antibodies enter the CNS or may neutralize some of the immunomodulatory effects of GA is not yet known. (GA-Ag ⫽ glatiramer acetate
`antigen; ? ⫽ Presumed transmigration of immune cells across the blood-brain barrier.)
`
`tralizing activity, although they do not bind to fragment
`crystallizable (Fc) receptors or activate the complement
`system. However, serum from GA-treated patients con-
`taining antibodies against GA did not inhibit the ability
`of GA to stimulate GA-reactive T cells, indicating that in
`vitro anti-GA Ig had no neutralizing effect.45 Interest-
`ingly, Brenner et al.47 reported that relapse-free patients
`displayed higher titers against GA than patients with an
`active disease course under GA treatment, indicating a
`beneficial rather than effect-neutralizing role of antibod-
`ies against GA. In fact, in an animal model of CNS
`demyelinating disease, GA-specific antibodies were
`shown to promote myelin repair,48 an effect which might
`contribute to the proposed neuroprotective properties of
`GA in MS.
`
`Effects on the innate immune system
`Although past investigations of GA primarily focused
`on its effects on the adaptive immune system, especially
`on T cells, emerging evidence supports the concept that
`GA may also act on APCs. The interaction between
`APCs and T cells is fundamental for any adaptive T-cell
`immune response. Several groups have reported that in
`vitro GA treatment leads to a broad antigen-nonspecific
`alteration of APC function.19-21,49 –52 Possibly the first
`report regarding the effect of GA on the innate immune
`system was derived from an in vitro study in which GA
`altered the activation of a human monocytic cell line.49
`Specifically, GA inhibited the induction of HLA proteins
`as well as the release of tumor necrosis factor (TNF) and
`cathepsin B by THP-1 cells. In vitro GA treatment was
`
`Neurotherapeutics, Vol. 4, No. 4, 2007
`
`MYLAN PHARMS. INC. EXHIBIT 1090 PAGE 3
`
`

`
`650
`
`WEBER ET AL.
`
`also shown to alter the activation status of freshly iso-
`lated human monocytes.19,20 Weber et al.19 reported that
`GA inhibited lipopolysaccharide (LPS)-mediated expres-
`sion of APC activation markers,
`including CD150/
`SLAM, CD25, and CD69. Furthermore, GA-treated
`monocytes released significantly lower levels of TNF-
`and IL-12, two inflammatory Th1-polarizing cytokines.
`Another study demonstrated that GA treatment not only
`reduced the release of inflammatory cytokines, but also
`enhanced production of Th2 polarizing IL-10 by mono-
`cytes.20 A similar cytokine shift was observed in micro-
`glial cells, an APC population that is believed to have a
`key role in the reactivation of T cells within the CNS. In
`vitro-generated human dendritic cells also released less
`TNF21 and IL-1250 on in vitro GA exposure. Most no-
`tably, GA-treatment of dendritic cells promoted Th2 dif-
`ferentiation of naive T cells without affecting APC ca-
`pability for inducing T-cell proliferation.21
`Two independent studies investigated how GA af-
`fects monocytes in MS patients.
`In both studies,
`monocytes were freshly isolated from GA-treated pa-
`tients without any additional in vitro exposure to GA.
`Compared to untreated MS patients and healthy sub-
`jects, monocytes from GA-treated MS patients ex-
`pressed significantly lower levels of the activation
`marker CD150/SLAM and released less TNF on stim-
`ulation with low concentration of LPS.19 In the second
`study, Kim and colleagues20 reported that the basal
`and induced release of IL-10 was significantly en-
`hanced in monocytes
`from GA-treated patients,
`whereas the production of IL-12 was reduced, defining
`an anti-inflammatory “type II” monocyte phenotype.
`These studies clearly indicate a systemic effect of GA
`treatment on monocytes that may promote Th2 differ-
`entiation of T cells in vivo. Theoretically, these find-
`ings raise the possibility that GA treatment may com-
`promise innate immune responses in GA-treated MS
`patients. However, GA-treatment does not appear to
`be predisposed to infections. In this regard, one in
`vitro finding might be of relevance (i.e., GA only
`inhibited activation of monocytes that were challenged
`with suboptimal concentrations of toll-like receptor
`ligands, such as LPS).19 Higher concentrations of LPS
`could override the inhibitory effect of GA, which
`could explain why the capability of monocytes to ef-
`ficiently clear infections is not diminished in GA-
`treated MS patients. Future longitudinal studies are
`necessary to define whether initiation of GA treatment
`truly leads to a reduction of APC reactivity in the
`individual MS patient. This type of study will also
`allow correlation between altered APC reactivity and a
`drug-related benefit
`to determine the extent of the
`clinical relevance of these GA-mediated effects on the
`APCs.
`
`Neurotherapeutics, Vol. 4, No. 4, 2007
`
`Cross-talk between type II APC and regulatory T-
`cell populations
`It has become established that the phenotype of APC
`influences differentiation of T cells and that reciprocally
`differentiated T cells modify APC function. In this re-
`gard, monocytes cultured with Th2 supernatants devel-
`oped a phenotype similar to GA-treated monocytes. This
`finding indicates that GA-reactive Th2 cells can exert a
`positive feedback on the development of type II mono-
`cytes. In fact, type II monocyte development may even
`occur secondary to the induction of GA-reactive Th2
`cells. However, other evidence suggests the opposite
`scenario (i.e., that APCs may be the primary target of GA
`and that GA-induced type II APCs mediate T-cell devi-
`ation). First, in vitro, GA exerted a direct effect on var-
`ious APC populations resembling its effect in vivo, in the
`absence of T cells.19,20 These GA-treated APCs were
`capable of promoting development of Th2 cells when
`co-cultured with naive (untreated) Th0 cells in the ab-
`sence of GA.21 Second, in vivo GA treatment exerted a
`systemic effect on monocytes and possibly on monocyte-
`derived APCs. However, the frequency of GA reactive
`Th2 cells in the peripheral blood of GA-treated MS
`patients is only approximately 1 in 20,000, raising the
`question as to whether Th2 cytokines derived from these
`cells could be sufficient to mediate type II APC devel-
`opment. Most strikingly, studies in genetically altered
`mice indicate that in vivo GA treatment can induce type
`II monocytes in the absence of T cells.53 Further studies
`are necessary to determine the pathway by which GA
`treatment may alter APC and T-cell function in MS
`patients.
`Assuming that APCs are the primary target through
`which GA mediates T-cell immune deviation, one would
`anticipate that Th2 deviation and/or induction of Treg
`should not be restricted to GA-reactive T cells. A recent
`study by Allie et al.54 investigated the phenotype of
`T-cell lines specific for GA, MBP, or tetanus toxoid
`generated from MS patients before and after GA treat-
`ment. T-cell differentiation was assessed by the ratio
`between IFN-␥ and IL-5 release. In this longitudinal
`study, in vivo GA treatment biased differentiation of all
`T cell lines toward a Th2 phenotype, indicating that Th2
`differentiation occurred independent of T-cell antigen
`specificity.54 However, another study did not describe an
`antigen-independent Th2 deviation of established T-cell
`responses on GA treatment, and supported the concept
`that Th2 deviation may primarily occur in GA-reactive T
`cells.42 Although apparently conflicting, both findings
`might be valid. First, a cross-sectional study comparing
`untreated patients to GA-treated patients may be less
`sensitive to detect minor changes in T-cell differentiation
`compared to a longitudinal study investigating the same
`patients before and after treatment. Second, it is plausible
`that an APC-driven Th2 deviation may be pronounced in
`
`MYLAN PHARMS. INC. EXHIBIT 1090 PAGE 4
`
`

`
`GLATIRAMER ACETATE TO TREAT MS
`
`651
`
`GA-reactive T cells, as every APC that presents GA
`should have been in contact with GA and undergone type
`II differentiation prior to T-cell activation. The concept
`of an antigen-nonspecific effect of GA is further sup-
`ported by the fact that GA treatment has been shown to
`be clinically beneficial in other models of autoimmune or
`inflammatory conditions, such as arthritis, uveoretini-
`tis,55 inflammatory bowel disease,56 and graft rejec-
`tion.57
`Although T cells might not be the primary target of
`GA, they are most likely the effector cells of GA-medi-
`ated immune modulation. Deficiencies in regulatory T
`cells have been associated with MS pathogenesis38 and
`GA-mediated restoration of T-cell regulation correlates
`with clinical benefit.40 In EAE, adoptive transfer of GA-
`reactive T cells alone can inhibit EAE induction by var-
`ious encephalitogens,16,31,58 similar to GA treatment it-
`self, and GA-reactive T cells accumulate in the CNS of
`protected animals. Thus, whereas GA may mediate a
`primary effect on APC independent of T cells, the type II
`APC-induced regulatory T cells may be the effector cells
`of GA-mediated immune modulation.
`
`Possible neurotrophic effects
`Some experimental data indicate that GA may have
`direct neuroprotective properties. In vitro, GA-reactive T
`cells can produce neurotrophic factors such as brain-
`derived neurotrophic factor (BDNF).17,18 BDNF is an
`important factor for differentiation and survival of neu-
`rons and is required for maintenance of various glial cell
`functions.59 Although the ability to produce BDNF is
`unlikely to be restricted to GA-reactive T cells, it may
`relate to the activation status of immune cells (e.g., T
`cells).60 In this regard, the continuous activation by daily
`GA application may promote BDNF production of GA-
`reactive T cells in vivo. As activation of immune cells
`also facilitates their transmigration across the blood-
`brain barrier,61 accumulation of BDNF-producing GA-
`reactive T cells within the CNS of patients with MS may
`occur, in proportion to the population frequency of these
`cells. This concept is supported by findings derived from
`EAE studies. Adoptively transferred GA-reactive T cells
`are detected within the CNS of mice with EAE15 and
`produce BDNF in situ.62 These putative neurotrophic
`effects of GA may not be restricted to CNS autoimmune
`disease. In an optic-nerve injury model, GA-specific T
`cells prevented the secondary degeneration of axons and
`similarly accumulated at the site of injury producing
`neurotrophic factors.63 In an animal model of glaucoma,
`GA reduced loss of retinal ganglion cells without affect-
`ing intraocular pressure.64 GA administration protected
`motor neurons from acute and chronic degeneration65
`and adoptive transfer of GA-reactive T cells enhanced
`survival of dopaminergic neurons in a mouse model of
`Parkinson’s disease.66,67 Thus, GA may exert neurotro-
`
`phic and/or protective properties in addition to immuno-
`modulatory effects. Their relevance in human neurode-
`generative diseases,
`including MS,
`remains
`to be
`determined.
`
`CONCLUSIONS
`
`Although GA is one of the most widely prescribed
`drugs for treatment of relapsing–remitting MS, its mech-
`anism of action is still not entirely understood. GA treat-
`ment induces a preferential Th2 deviation of T cells and
`promotes restoration of frequency and function of Treg
`in MS. Recent reports demonstrated that GA also exerts
`immunomodulatory effects on APCs, such as monocytes.
`These new findings may provide a plausible explanation
`for GA-mediated T-cell immune modulation. Whereas it
`remains to be determined whether APCs, T cells, or both
`are the primary pharmacological target for GA, immune
`modulation of APC and T cells appears to engage a
`positive feedback mechanism. These novel observations
`should contribute to a better understanding of the mech-
`anism of action of GA and may provide useful insight for
`the development of new and more efficient agents.
`
`REFERENCES
`
`1. Teitelbaum D, Meshorer A, Hirshfeld T, Arnon R, Sela M. Sup-
`pression of experimental allergic encephalomyelitis by a synthetic
`polypeptide. Eur J Immunol 1971;1:242–248.
`2. Bornstein MB, Miller AI, Teitelbaum D, Arnon R, Sela M. Mul-
`tiple sclerosis: trial of a synthetic polypeptide. Ann Neurol 1982;
`11:317–319.
`3. 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.
`4. 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.
`5. Comi G, Filippi M, Wolinsky JS. European/Canadian multicenter,
`double-blind, randomized, placebo-controlled study of the effects
`of glatiramer acetate on magnetic resonance imaging—measured
`disease activity and burden in patients with relapsing multiple
`sclerosis. European/Canadian Glatiramer Acetate Study Group.
`Ann Neurol 2001;49:290 –297.
`6. Filippi M, Rovaris M, Rocca MA, Sormani MP, Wolinsky JS,
`Comi G. Glatiramer acetate reduces the proportion of new MS
`lesions evolving into ”black holes.” Neurology 2001;57:731–733.
`7. Sormani MP, Bruzzi P, Comi G, Filippi M. The distribution of the
`magnetic resonance imaging response to glatiramer acetate in mul-
`tiple sclerosis. Mult Scler 2005;11:447– 449.
`8. Neuhaus O, Farina C, Wekerle H, Hohlfeld R. Mechanisms of
`action of glatiramer acetate in multiple sclerosis. Neurology 2001;
`56:702–708.
`9. Farina C, Weber MS, Meinl E, Wekerle H, Hohlfeld R. Glatiramer
`acetate in multiple sclerosis: update on potential mechanisms of
`action. Lancet Neurol 2005;4:567–575.
`10. 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 U S A
`1994;91:4872– 4876.
`11. Teitelbaum D, Fridkis-Hareli M, Arnon R, Sela M. Copolymer 1
`inhibits chronic relapsing experimental allergic encephalomyelitis
`induced by proteolipid protein (PLP) peptides in mice and inter-
`
`Neurotherapeutics, Vol. 4, No. 4, 2007
`
`MYLAN PHARMS. INC. EXHIBIT 1090 PAGE 5
`
`

`
`652
`
`WEBER ET AL.
`
`responses. J Neuroimmunol
`
`feres with PLP-specific T cell
`1996;64:209 –217.
`12. 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 U S A2000;97:7452–7457.
`13. Dhib-Jalbut S. Mechanisms of action of interferons and glatiramer
`acetate in multiple sclerosis. Neurology 2002;58(8 Suppl 4):S3–9.
`14. Duda PW, Schmied MC, Cook SL, Krieger JI, Hafler DA. Glati-
`ramer acetate (Copaxone) induces degenerate, Th2-polarized im-
`mune responses in patients with multiple sclerosis. J Clin Invest
`2000;105:967–976.
`15. Aharoni R, Teitelbaum D, Leitner O, Meshorer A, Sela M, Arnon
`R. Specific Th2 cells accumulate in the central nervous system of
`mice protected against experimental autoimmune encephalomyeli-
`tis by copolymer 1. Proc Natl Acad Sci U S A2000;97:11472–
`11477.
`16. 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 U S A1997;94:10821–10826.
`17. Chen M, Gran B, Costello K, Johnson K, Martin R, Dhib-Jalbut S.
`Glatiramer acetate induces a Th2-biased response and cross reac-
`tivity with myelin basic protein in patients with MS. Mult Scler
`2001;7:209 –219.
`18. Ziemssen T, Kumpfel T, Klinkert WE, Neuhaus O, Hohlfeld R.
`Glatiramer acetate-specific T-helper 1- and 2-type cell lines pro-
`duce BDNF: implications for multiple sclerosis therapy. Brain-
`derived neurotrophic factor. Brain 2002;125(Pt 11):2381–2391.
`19. Weber MS, Starck M, Wagenpfeil S, Meinl E, Hohlfeld R, Farina
`C. Multiple sclerosis: glatiramer acetate inhibits monocyte reac-
`tivity in vitro and in vivo. Brain 2004;127(Pt 6):1370 –1378.
`20. Kim HJ, Ifergan I, Antel JP, et al. Type 2 monocyte and microglia
`differentiation mediated by glatiramer acetate therapy in patients
`with multiple sclerosis. J Immunol 2004;172:7144 –7153.
`21. Vieira PL, Heystek HC, Wormmeester J, Wierenga EA, Kapsen-
`berg ML. Glatiramer acetate (copolymer-1, copaxone) promotes
`Th2 cell development and increased IL-10 production through
`modulation of dendritic cells. J Immunol 2003;170:4483– 4488.
`22. Stasiolek M, Bayas A, Kruse N, et al. Impaired maturation and
`altered regulatory function of plasmacytoid dendritic cells in mul-
`tiple sclerosis. Brain 2006;129(Pt 5):1293–1305.
`23. Stuve O, Youssef S, Weber MS, et al. Immunomodulatory synergy
`by combination of atorvastatin and glatiramer acetate in treatment
`of CNS autoimmunity. J Clin Invest 2006;116:1037–1044.
`24. Fridkis-Hareli M, Strominger JL. Promiscuous binding of syn-
`thetic copolymer 1 to purified HLA-DR molecules. J Immunol
`1998;160:4386 – 4397.
`25. 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 U S A1992;89:137–141.
`26. Aharoni R, Schlegel PG, Teitelbaum D, et al. Studies on the
`mechanism and specificity of the effect of the synthetic random
`copolymer GLAT on graft-versus-host disease. Immunol Lett
`1997;58:79 – 87.
`27. 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.
`28. Wiesemann E, Klatt J, Sonmez D, Blasczyk R, Heidenreich F,
`Windhagen A. Glatiramer acetate (GA) induces IL-13/IL-5 secre-
`tion in naive T cells. J Neuroimmunol 2001;119:137–144.
`29. Farina C, Wagenpfeil S, Hohlfeld R. Immunological assay for
`assessing the efficacy of glatiramer acetate (Copaxone) in multiple
`sclerosis: a pilot study. J Neurol 2002;249:1587–1592.
`30. Lisak RP, Zweiman B, Blanchard N, Rorke LB. Effect of treatment
`with Copolymer 1 (Cop-1) on the in vivo and in vitro manifesta-
`tions of experimental allergic encephalomyelitis (EAE). J Neurol
`Sci 1983;62:281–293.
`31. Aharoni R, Teitelbaum D, Sela M, Arnon R. Bystander suppres-
`sion of experimental autoimmune encephalomyelitis by T cell lines
`and clones of the Th2 type induced by copolymer 1. J Neuroim-
`munol 1998;91:135–146.
`32. Hori S, Nomura T, Sakaguchi S. Control of regulatory T cell
`
`Neurotherapeutics, Vol. 4, No. 4, 2007
`
`transcription
`
`factor Foxp3. Science
`
`the
`by
`development
`2003;299:1057–1061.
`33. Brunkow ME, Jeffery EW, Hjerrild KA, et al. Disruption of a new
`forkhead/winged-helix protein, scurfin, results in the fatal lympho-
`proliferative disorder of the scurfy mouse. Nat Genet 2001;27:68 –
`73.
`34. Fontenot JD, Gavin MA, Rudensky AY. Foxp3 programs the de-
`velopment and function of CD4⫹CD25⫹ regulatory T cells. Nat
`Immunol 2003;4:330 –336.
`35. Bacchetta R, Passerini L, Gambineri E, et al. Defective regulatory
`and effector T cell functions in patients with FOXP3 mutations.
`J Clin Invest 2006;116:1713–1722.
`36. Sakaguchi S, Fukuma K, Kuribayashi K, Masuda T. Organ-specific
`autoimmune diseases induced in mice by elimination of T cell
`subset. I. Evidence for the active participation of T cells in natural
`self-tolerance; deficit of a T cell subset as a possible cause of
`autoimmune disease. J Exp Med 1985;161:72– 87.
`37. Kukreja A, Cost G, Marker J, et al. Multiple immuno-regulatory
`defects in type-1 diabetes. J Clin Invest 2002;109:131–140.
`38. Viglietta V, Baecher-Allan C, Weiner HL, Hafler DA. Loss of
`functional suppression by CD4⫹CD25⫹ regulatory T cells in
`patients with multiple sclerosis. J Exp Med 2004;199:971–979.
`39. Putheti P, Soderstrom M, Link H, Huang YM. Effect of glatiramer
`acetate (Copaxone) on CD4⫹CD25high T regulatory cells and
`their IL-10 production in multiple sclerosis. J Neuroimmunol
`2003;144:125–131.
`40. Hong J, Li N, Zhang X, Zheng B, Zhang JZ. Induction of
`CD4⫹CD25⫹ regulatory T cells by copolymer-I through activa-
`tion of transcription factor Foxp3. Proc Natl Acad Sci U S A
`2005;102:6449 – 6454.
`41. Karandikar NJ, Crawford MP, Yan X, et al. Glatiramer acetate
`(Copaxone) therapy induces CD8(⫹) T cell responses in patients
`with multiple sclerosis. J Clin Invest 2002;109:641– 649.
`42. Farina C, Then Bergh F, Albrecht H, et al. Treatment of multiple
`sclerosis with Copaxone (COP): Elispot assay detects COP-in-
`duced interleukin-4 and interferon-gamma response in blood cells.
`Brain 2001;124(Pt 4):705–719.
`43. Tennakoon DK, Mehta RS, Ortega SB, Bhoj V, Racke MK, Karan-
`dikar NJ. Therapeutic induction of regulatory, cytotoxic CD8⫹ T
`cells in multiple sclerosis. J Immunol 2006;176:7119 –7129.
`44. Biegler BW, Yan SX, Ortega SB, Tennakoon DK, Racke MK,
`Karandikar NJ. Glatiramer acetate (GA) therapy induces a focused,
`oligoclonal

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