throbber
Journal of Autoimmunity and Cell Responses
`ISSN 2054-989X | Volume 1 | Article 3
` Review
`
`
`
` Open Access
`Glatiramer acetate and therapeutic peptide vaccines
`for multiple sclerosis
`
`Jill Conner
`
`Correspondence: Jill.Conner@tevapharm.com
`
`CrossMark
`← Click for updates
`
`Global Specialty Medicines, Teva Pharmaceutical Industries, Ltd., Overland Park, KS, USA.
`
`Abstract
`Therapeutic vaccines are antigen-specific agents that inhibit an unwanted immune response to prevent
`progression of, or eliminate, existing disease. Unlike prophylactic vaccines, developing therapeutic
`vaccines poses a significant challenge because they can require multimodal immunomodulatory activity
`to modulate complex pathogenic disease pathways. This is particularly true in autoimmune disorders,
`including multiple sclerosis, which has complex pathogenesis that, although not fully understood, appears
`to involve dysfunction of both innate and adaptive immune processes. The glatiramoid, glatiramer acetate
`(Copaxone®), a nonbiologic, complex, heterogenous mixture of synthetic polypeptides, is currently
`the only approved therapeutic peptide for treatment of multiple sclerosis. Glatiramer acetate has an
`enormous number of potentially active epitopes (estimated to be ~1030) in the polypeptides mixture. The
`epitopes in glatiramer acetate have not been identified, but they appear to act as altered peptide ligands
`of encephalitogenic epitopes within myelin basic protein, a suspected autoantigen implicated in multiple
`sclerosis. Peptide epitopes in glatiramer acetate compete with autoantigens for binding with major
`histocompatability complex molecules on antigen-presenting cells, thereby altering the functional outcome
`of T cell signaling from inflammatory to anti-inflammatory responses. Other therapeutic vaccines designed
`to more selectively compete with myelin antigens for receptor binding have been shown effective in animal
`models of multiple sclerosis but toxic in human patients in clinical trials. The partially random structure
`of glatiramer acetate and potentially huge number of antigenic sequences may be integral to safety and
`efficacy by influencing multifactorial immune processes and surmounting challenges related to inter- and
`intra-individual heterogeneity of T cell responses and the phenomenon of epitope spreading. Follow-on
`generic versions of glatiramer acetate have been made available to multiple sclerosis patients outside the
`United States. Analyses of these glatiramoids indicate they have different biological and immunological
`activity from that of Copaxone®, illustrating the difficulty of replicating complex glatiramoids and of
`developing safe, effective peptide vaccines in general. Reviewed here are some of the major mechanisms of
`glatiramer acetate activity on innate and adaptive immune pathology and considerations for development
`of future therapeutic peptide vaccines for multiple sclerosis.
`keywords: Autoimmune disease, glatiramer acetate, glatiramoid, multiple sclerosis, peptide, copolymer,
`therapeutic, vaccine
`
`Introduction
`Most vaccines are used for prophylaxis of disease; however,
`therapeutic vaccination involves the use of an antigen-specific
`intervention to inhibit an unwanted immune response,
`with the goal of preventing progression of, or eliminating,
`existing disease [1]. Effective therapeutic vaccines are less
`common than preventive vaccines and are more challenging
`to develop because preventive vaccines typically have
`immunologic specificity for an individual infective agent (e.g.,
`
`viral poliomyelitis, smallpox). In contrast, therapeutic vaccines
`may require multimodal immunomodulatory activity to modu-
`late complex pathogenic disease mechanisms already underway.
`Suppression of harmful inflammatory immune responses by
`antigenic peptide vaccination requires induction of tolerance,
`which can be attained by repeated dosing [2].
`There is great interest in developing therapeutic peptide vac-
`cines for treatment of autoimmune diseases such as multiple
`sclerosis (MS) [3-8]. Synthetic peptides, in principle, could be
`
`© 2014 Jill Conner; licensee Herbert Publications Ltd. This is an Open Access article distributed under the terms of Creative Commons Attribution License
`(http://creativecommons.org/licenses/by/3.0). This permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.
`
`Page 1 of 11
`
`YEDA EXHIBIT NO. 2007
`MYLAN PHARM. v YEDA
`IPR2014-00644
`
`

`
`designed to present selected epitopes to immune cells to
`elicit a desired response. MS is a chronic, progressive,
`neurodegenerative autoimmune disease of the central nervous
`system (CNS) associated with demyelination and axonal
`damage. MS is a disease with no identified specific pathogenic
`target for prophylaxis. The etiology of MS is unknown, and
`pathogenesis is complex and not fully understood, but appears
`to include pathologic mechanisms of innate and adaptive
`immunity working together to exacerbate disease (Figure 1)
`[9-11]. Therefore, an effective vaccine will optimally modulate
`both innate and adaptive immune responses. Currently, the
`glatiramoid, glatiramer acetate (GA, Copaxone®), a non-biologic
`complex drug (NBCD), is the first and only approved therapeutic
`peptide for treatment of MS [1,2,12]. Copaxone® and the
`interferon beta (IFNβ) drugs (protein products produced by
`recombinant DNA techniques, including Avonex®, Rebif®, and
`Betaseron®) were the first disease-modifying therapies (DMTs)
`approved for treatment of MS. The IFNβ drugs have profound
`activities on several components of the process required for
`the migration of inflammatory cells into the CNS. These agents
`have been shown to reduce relapse rate, delay progression
`of neurologic disability, and decrease brain lesions on MRI in
`patients with relapsing-remitting MS (RRMS) [13-16]. Unlike
`GA activity (described below), a primary mechanism of the
`IFNβ drugs is reduction of inflammatory cell trafficking into
`the CNS [10,15,16]. More recently, several oral MS treatments
`have become available, including Aubagio® (teriflunomide),
`Tecfidera® (dimethyl fumarate), and Gilenya® (fingolimod).
`That GA is the only therapeutic peptide approved for MS
`reflects the complexity of the disease and of GA, and the
`challenges of preparing safe and effective therapeutic peptide
`vaccines. Reviewed here are some of the key mechanisms of
`GA activity on innate and adaptive immunity that contribute
`to its efficacy and safety, and considerations for development
`of follow-on versions of GA and new peptide vaccines for MS.
`
`Review
`Glatiramer acetate
`Discovery
`The immunomodulatory benefits of GA were discovered
`serendipitously. In the 1970s, researchers at the Weizmann
`Institute in Israel synthesized a series of amino acid copolymers
`in hopes of developing synthetic antigens that mimic
`myelin basic protein (MBP), the main protein component
`of the lipid-rich myelin sheath and a major autoantigen in
`MS. The copolymers were designed to reproducibly induce
`experimental autoimmune encephalomyelitis (EAE), an
`animal model of MS. However, rather than induce EAE, GA
`immunization proved to be protective against EAE induction
`and reduced symptoms of established EAE in several animal
`species [17-20].
`
`Chemical nature
`GA is a complex, heterogenous, partially random mixture of
`
`Innate
`
`Immature
`dendritic
`cell
`
`TGFβ
`Adaptive
`
`Antigen processing and
`uptake onto MHC class II
`IL-4
`IL-10
`
`IL-4
`IL-10
`
`Mature
`dendritic
`cell
`TGFβ
`IL-6
`
`IL-12
`
`IL-1β
`
`Naive T cell
`
`Naive T cell
`
`TReg cell
`
`Macrophage
`
`TH1 cell IFN-γ
`TNF
`
`B cell
`
`CTL/CD8+ T cell
`
`Innate
`
`Astrocyte
`
`Microglia macrophage
`Reactivation
`
`TH17 cell
`
`IL-17
`
`Blood–brain
`barrier
`
`Central
`nervous
`system
`
`Activation
`TH17 cell
`
`→
`Glutamate
`
`NO
`Neuron
`
`IL-17
`IFN-γ
`TNF
`
`IFN-γ
`TNF
`IFN-γ
`
`TH1 cell
`MMPs
`NO/
`O2•
`
`B cell
`
`CTL/CD8+ T cell
`Plasma
`cell
`
`Autoantibodies
`
`Figure 1. Multiple sclerosis pathogenesis involves innate
`and adaptive immune dysfunction. Reprinted by permission
`from Macmillan Publishers Ltd: Nat Rev Drug Discov.
`Nov;9(11):883-97, copyright 2010 [115].
`
`synthetic polypeptides containing four naturally occurring
`amino acids: L-glutamic acid, L-alanine, L-lysine, and L-tyrosine
`[21]. The polypeptides chains in GA vary in length from 20
`to 200 amino acid residues, with an average of approximately
`60 residues, and molecules in the colloidal GA sus-pension
`range in size from 1.5 nm to 550 nm, making GA one of the
`first true nanomedicines [22,23]. The average molecular
`weight of the polypeptides in GA is 7000-9000 daltons. GA
`has an enormous number of potentially active epitopes
`(estimated to be ~1030 [24]) in the polypeptides mixture, which
`may be integral to drug efficacy and safety [25]. However, the
`complexity of GA makes isolation and identification of the
`active epitopes in the GA polypeptides mixture impossible.
`Nevertheless, it is possible to differentiate among glatiramoids
`(i.e., synthetic copolymer mixtures comprising the four amino
`acids in GA in a defined molar ratio [22]) in analytical and bio-
`logical tests (as described below).
`
`Clinical usage
`Copaxone® is indicated for the treatment of patients with
`relapsing forms of MS [21]. Copaxone® is currently available
`as a daily 20 mg SC injection (approved in the US in 1996)
`and recently approved in the US in January 2014 as a 40 mg
`SC injection administered three times per week.
`
`GA mechanisms of action
`Numerous effects of GA on cellular and humoral immune
`cells have been identified over the last 4 decades, though
`the complete activity of GA remains unknown and new
`mechanisms continue to be discovered (Figure 2) [22,25,26].
`Perhaps the best-known therapeutic mechanisms of GA
`activity involve its effect on adaptive immune cells (T and
`
`2
`
`Jill Conner, Journal of Autoimmunity and Cell Responses 2014,
`http://www.hoajonline.com/journals/pdf/2054-989X-1-3.pdf
`
`doi: 10.7243/2054-989X-1-3
`
`Page 2 of 11
`
`YEDA EXHIBIT NO. 2007
`MYLAN PHARM. v YEDA
`IPR2014-00644
`
`

`
`B cell
`
`Induction of GA-specific antibodies,
`especially GA-specific lgG4 antibodies
`
`T cell
`
`Induction of GA-specific Th2 CD4+ cells
`(Th1→Th2shift of CD4+ T cells)
`Decrease of GA-specific CD4+ T cell proliferation
`Induction of GA-specific CD8+ suppressor T cell
`Induction of GA-specific CD4+CD25+ regulatory
`T cells
`
`APC
`
`More Th2-like differentiation of APC
`Modulation of monocytic cell function
`(e.g., decrease of TNF-α-producing cells)
`Modulation of dendritic cell function
`e.g., restoration of pDC phenotype and function)
`Figure 2. Glatiramer acetate modulates innate and adaptive
`immune cells. Reprinted from Int Rev Neurobiol Vol. 79,
`Ziemssen and Schrempf, Glatiramer acetate: mechanisms
`of action in multiple sclerosis, pp. 537-570 (2007), with
`permission from Elsevier [20].
`
`B lymphocytes). More recently, GA effects on cells of the
`innate immune system (e.g., antigen-presenting cells
`[APCs], natural killer [NK] cells) have been discovered.
`
`GA effects on innate immune cells
`Antigen-presenting cells: The main functions of APCs are
`phagocytosis, antigen presentation, and cytokine production.
`In the peripheral immune compartment, APCs provide three
`sequential signals to activate antigen-specific T cells:
`1) Toll-like receptors (TLRs) on activated APCs recognize
` bacteria, viruses, and other antigens [27,28]. Upon
` phagocytosis, antigenic peptide fragments bound to
` major histocompatibility complex (MHC) molecules on
` the surface of APCs facilitate T cell recognition of the
` cognate antigen by T cell receptors (TCR) on CD4+ cells
` (antigen presentation on MHC class II molecules) or
` CD8+ cytotoxic T cells (antigen presentation by MHC
` class I molecules);
`2) Costimulatory molecules on APCs activate the T cells to
` initiate proliferation and differentiation; and
`3) Cytokine secretion by APCs polarizes differentiation of
` activated CD4+ T cells into different effector (T helper
` Th; e.g., Th1, Th2, Th3, Th17) and regulatory (e.g.,
` CD4+CD25+FoxP3+) T cell subtypes; influences
` cytotoxic CD8+ T cell phenotypes [29]; and instructs
` plasma cell differentiation.
`In MS, peptide epitopes from putative MS autoantigens, in-
`cluding MBP, myelin oligodendrocyte glycoprotein (MOG),
`and proteolipid protein (PLP) molecules, bind to MHC class II
`molecules on APCs. Upon T cell recognition, pro-inflammatory
`(Type I) cytokines are expressed and stimulate differentiation
`and proliferation of pathogenic autoreactive CD4+ Th1 cells
`[10,30]. Reciprocally, APCs exposed to pro-inflammatory T
`cells in vitro tend to express a range of adhesion molecules,
`costimulatory molecules, and cytokines favoring further T
`
`cell differentiation of pro-inflammatory Th1 cells. Activated
`myelin-reactive Th1 cells in the periphery move from the
`systemic circulation across the blood-brain barrier (BBB) and
`enter the CNS [12]. Once Th1 cells establish an inflammatory
`milieu there, pro-inflammatory Th17 cells can enter the CNS
`[31]. Th17 cells play an important role in the pathogenesis of
`inflammatory and autoimmune diseases [32]. Once in the CNS,
`antigenic peptide fragments bound to dendritic cells (DCs),
`microglia, and astrocytes reactivate autoreactive T cells, leading
`to pro-inflammatory cytokine release and myelin damage.
`Circulating APCs of the myelomonocytic lineage, i.e., mono-
`cytes/macrophages, and DCs, may be the primary targets
`of GA immunomodulation. GA activity on monocytes, DCs,
`and microglia is antigen-nonspecific, which may help explain
`why GA has shown efficacy in animal models of several in-
`flammatory and neurodegenerative conditions [33-39].
`GA polypeptides are promiscuous binders to MHC class II
`molecules on APCs, with or without antigen processing [40].
`GA peptides compete with myelin peptides for binding to
`MHC class II molecules on APCs and can preferentially displace
`peptides from MBP [41], MOG [42], and PLP [43] from the MHC
`binding site, but cannot be displaced by them [44]. As a result,
`differentiation of autoaggressive Th1 cells is reduced and GA
`mediates a shift in T cell phenotypes from Th1 to Th2/3 cells.
`Monocytes cultured in anti-inflammatory supernatants push
`T cells toward Th2 differentiation in vitro, which is associated
`with secretion of anti-inflammatory (Type II) cytokines [45].
`GA induces Type II monocyte and microglia differentiation
`in EAE models and in MS patients [45-48]. Adoptive transfer
`of monocytes from GA-treated mice to GA-naïve mice
`with EAE directs T cell differentiation toward Th2 cells and
`CD4+CD25+FoxP3+ Tregs, an important subclass of regulatory
`cells that attenuate autoreactive T cell responses [47].
`Continued GA treatment reduces the ability of APCs to
`present antigen or to respond to various stimuli [45-49]. In
`mice with EAE, GA reduced expression of TLRs on DCs [50],
`and down-regulated osteopontin, a protein implicated in
`chronic inflammatory diseases that induces DC maturation
`toward the Type I inflammatory phenotype. In another study,
`GA inhibited monocyte activation and production of pro-
`inflammatory tumor necrosis factor alpha (TNFα) by human
`DCs in vitro [48], and compared with untreated patients,
`DCs and monocytes from MS patients who received GA for
`1 year showed less activation, accompanied by reduced risk
`of relapse [51].
`The ability of APCs to penetrate the BBB is essential to the
`establishment of autoimmune inflammatory CNS diseases.
`Suppressed expression of chemokines with CNS chemotactic
`properties may also contribute to GA therapeutic activity
`[50]. Macrophage inflammatory protein (MIP)-1 increases
`DC transmigration across endothelial cells and is elevated
`in mice with EAE compared with control mice. Daily GA trea-
`tment suppressed MIP-1α and MIP-1β expression in mice with
`EAE, and also decreased expression of RANTES [50,52], a poly-
`
`3
`
`Jill Conner, Journal of Autoimmunity and Cell Responses 2014,
`http://www.hoajonline.com/journals/pdf/2054-989X-1-3.pdf
`
`doi: 10.7243/2054-989X-1-3
`
`Page 3 of 11
`
`YEDA EXHIBIT NO. 2007
`MYLAN PHARM. v YEDA
`IPR2014-00644
`
`

`
`doi: 10.7243/2054-989X-1-3
`
`peptide with strong chemoattractant activity for T cells, mon-
`ocytes and macrophages associated with pathogenesis of MS
`lesions in the CNS [52].
`Attesting to the primacy of GA effects on APCs, researchers
`showed that GA did not influence T cell polarization when
`added to naïve Th cells activated in an APC-free system. More-
`over, induction of Type II monocytes by GA does not require
`reciprocal signalling by T cells, [53]. In STAT6-deficient mice,
`which cannot generate interleukin (IL)-4-producing Th2 cells,
`monocytes from GA-treated mice exhibited increased Type II
`anti-inflammatory cytokines, IL-10, and transforming growth
`factor beta (TGFβ) [47]. Similarly, monocytes from GA-treated
`RAG-1 mice, which lack mature T and B cells, also showed an
`anti-inflammatory cytokine pattern [47].
`Natural killer cells: NK cells are lymphocytes of the innate
`immune system responsible for immunosurveillance and
`regulating immune responses [54]. NK cells limit damage in
`MS by inhibiting autoreactive T cell responses, and increased
`frequency or functional competence of circulating NK cells
`is associated with decreased disease activity in MS patients
`[55]. NK cells exposed to GA kill both immature and mature
`DCs, impairing the ability of mature DCs to present antigens
`to autoreactive T cells [54,56,57]. This mechanism may be
`involved in GA effects on inhibiting graft-vs-host (GvH) disease
`in laboratory animals [36,58].
`
`GA effect on adaptive immune cells
`CD4+ T cells: Therapeutic effects of GA have been attributed
`in part to the ability of antigenic sequences in hydrolyzed
`GA peptides to act as altered peptide ligands (APLs) of
`encephalitogenic epitopes in MBP [1,12,59,60]. An APL serves
`as a receptor ligand that contains a substitute of one or more
`amino acids of the native ligand to change the functional
`outcome of TCR signaling [61]. An APL must be close enough in
`chemical composition to the native peptide antigen to trigger
`an immune response that is cross-reactive with the infectious
`agent or autoantigen, but must itself be biologically harmless.
`Accordingly, GA is cross-reactive with myelin antigens at both
`the humoral and cellular levels, without being encephalitogenic
`[62-67]. Repeated GA immunization modifies the GA-reactive
`T cell repertoire by skewing them from the Th1 phenotype
`toward the Th2 phenotype [59,66,68,69]. GA-reactive Th2
`cells release anti-inflammatory cytokines and neurotrophic
`factors [69-71] and suppress neighboring auto-aggressive Th1
`cells by means of “bystander suppression” in the CNS [68,69].
`GA treatment also increases the number and suppressive
`capacity of CD4+CD25+FoxP3+ Tregs in MS patients [72,73].
`Tregs dampen autoimmune responses, and are functionally
`impaired in MS patients [74].
`CD8+ T cells: Untreated patients with MS initially show
`low GA-specific CD8+ cell responses compared with healthy
`controls [75], but GA therapy produces proliferative responses
`in CD8+ T cells [75,76]. Adoptive transfer of GA-induced CD8+
`T cells results in amelioration of EAE [77]. GA-reactive CD8+
`
`T cells appear to regulate proliferation of myelin-reactive
`CD4+ cells [75,78]. Over time, continued treatment with GA
`results in decreasing numbers of GA-reactive CD4+ T cells
`while the number of GA-reactive CD8+ T cells increases [79].
`It has been proposed that GA effects on CD8+ T cells may be
`an indispensable component of its therapeutic activity [77].
`B cells: B cells can serve as efficient APCs for T cells and can
`activate autoreactive T cells; likewise, activated T cells can
`trigger B cell activation and formation of autoreactive anti-
`bodies [12,80]. Demyelinating antibodies can travel from the
`systemic circulation across the BBB and into the CNS. Antibody-
`mediated patterns of demyelination are detected in more
`than 50% of MS patients [81].
`All patients treated with GA develop GA-reactive antibodies
`[67,82]. As an antigen-based therapy, by definition anti-GA
`antibodies are not neutralizing and do not appear to interfere
`with MHC class II binding or induction and proliferation of
`suppressor T cell lines and clones [82]. GA treatment produces B
`cell secretion of the Type II cytokine, IL-10, thereby augmenting
`suppression of autoreactive T cells, and promoting induction
`of GA-reactive Th2 cells [83]. GA effects on B cells may also
`be essential to therapeutic activity. Adoptive transfer of B
`cells from GA-treated mice with active EAE inhibited the
`proliferation of autoreactive T cells, whereas GA treatment
`had no effect on EAE in B cell-deficient mice [84]. In a clinical
`study, relapse-free patients tended to develop higher anti-GA
`antibody titers than patients who relapsed [67], suggesting
`beneficial antibody activity. Additionally, remyelination of
`spinal cord axons was promoted by antibodies to GA in mice
`with EAE [85].
`Anti-GA antibody levels peak between 3 and 6 months of
`treatment initiation, and then gradually decline, but remain
`higher than baseline levels [67,82]. Anti-GA antibodies are
`mainly immunoglobulin G1 (IgG1), IgG2, IgG4, and IgA isotypes
`[67,82,86,87]. IgG1 antibody levels remain relatively consistent
`over time. IgG2 antibodies decline with chronic treatment
`[86] and a gradual shift to IgG1 and IgG4 is observed [87]. GA
`was not cross-reactive with MBP when exposed to polyclonal
`antibodies, but cross-reactivity with MBP was evident with
`monoclonal antibodies, in vitro [63]. RRMS patients in clinical
`trials developed GA-reactive antibodies with very low reactivity
`to MBP [67,82].
`
`Selective peptide vaccine attempts
`Given the APL activity of the random GA mixture, selective
`APLs were developed with the intent of creating more potent
`inhibition of autoreactivity. Two APLs of the immunodominant
`peptide region (83–99) of MBP (alanine was substituted for
`lysine at position 91 in both cases) were developed for
`use as therapeutic vaccination for MS. While effective for
`preventing and treating EAE in laboratory animals, they were
`less successful for MS patients, primarily due to toxicity
`problems [88,89]. The selective APL, NBI 5788, was evaluated in
`MS patients randomized to receive once-weekly SC injections
`
`4
`
`Jill Conner, Journal of Autoimmunity and Cell Responses 2014,
`http://www.hoajonline.com/journals/pdf/2054-989X-1-3.pdf
`
`Page 4 of 11
`
`YEDA EXHIBIT NO. 2007
`MYLAN PHARM. v YEDA
`IPR2014-00644
`
`

`
`doi: 10.7243/2054-989X-1-3
`
`of doses ranging from 5 to 50 mg for up to 4 months in a phase
`II clinical trial [88]. The study was stopped prematurely when
`9% of patients developed immediate-type hypersensitivity
`reactions. There was a robust Th2-like immune response in
`NBI 5788-treated patients. Th2 cell responses are associated
`with the promotion of humoral responses, and anti-NBI
`5788 antibody titers were relatively high in patients with
`hypersensitivity reactions, particularly in patients who received
`the highest tested dose. Thus, the strength of the Th2 response
`is an essential consideration in peptide vaccine design; i.e.,
`optimization of the suppression of Th1-related autoimmunity
`must be balanced against the risk of hypersensitivity reactions
`driven by Th2 responses [88].
`Weekly administration of the APL, CGP77116, was assessedin
`another small phase II study [89]. Of 8 patients treated with
`CGP77116, 2 showed improved disease activity, 3 had stable
`disease, and 3 showed worsened disease activity during
`and immediately after treatment. CGP77116 was poorly to-
`lerated and the trial was discontinued before completion. An
`investigation showed activated CGP77116-reactive T cells
`skewed toward the inflammatory Th1 phenotype rather than
`Th2, and on-study relapses were ascribed to CGP77116-induced
`expansion of encephalitogenic T cells specific for MBP(83–99) [89].
`Based on these studies, it was suggested that when relying on
`TCR- or MHC-mediated therapeutic effects (i.e., MHC blockade,
`TCR antagonism or partial agonism, clonal deletion or anergy),
`a single selected APL will be ineffective across individuals with
`different genetic backgrounds, and even intra-individually
`because TCR diversity may preclude a consistent therapeutic
`response [61]. Humans may express up to 8 different class II
`alleles; consequently, there is considerable diversity in the
`number of different antigenic peptides that can be presented
`to T cells among different individuals [61]. Varying autoantigens
`may be important in different individuals and phenotypic T
`cell response to an antigen may be heterogeneous. Further,
`by the time MS becomes clinically evident in humans, it is
`likely that the immune response has spread to more than
`one antigen due to “epitope spreading” [61]. Thus, adverse
`effects seen with NBI 5788 and CGP77116 were likely related
`to the many clonotypes that exist within and among different
`patients against the targeted epitope, MBP(83-99) [90].
`Immune dysregulation at multiple levels appears to cause
`MS, and this may explain an observed resistance to therapeutic
`modalities with high immune selectivity. GA is a pool of
`antigenic peptides of different lengths and composition with
`complex partially random structure and potentially millions
`of antigenic peptides [24,25]. An antigenic epitope in GA for
`a distinct TCR would be present at much lower concentration
`than if the GA structure were defined and not random [61].
`GA administration is thought to lead to anergy of T cells for
`which TCR-binding motifs in GA occur with higher frequency
`and to persistence or expansion of T cells with TCR-binding
`motifs in GA that occur with low frequency [61]. With GA,
`this implies the induction of anergy for Th1/Th0 GA-specific
`
`T cells and persistence/expansion of Th2 GA-specific T cells.
`
`Follow-on glatiramoids-special considerations
`The challenge of creating peptide vaccines is exemplified by
`attempts to create follow-on versions of Copaxone®. GA and
`other NBCDs are heterogenous mixtures of closely related,
`macromolecular, nanoparticulate components that cannot
`be entirely characterized physicochemically using available
`analytical technology [91]. The consistent activity and the
`quality of NBCDs rely on strictly controlled, proprietary
`manufacturing procedures [91-93]. Even small variations to
`the manufacturing method or of ingredient quality used to
`synthesize GA can increase the risk of safety problems or of
`reduced therapeutic efficacy [94-96]. While Copaxone® is not
`encephalitogenic and does not induce autoreactive antibodies,
`the same cannot be assumed for a follow-on GA-like product.
`Several purported “generic” GA products have been mar-
`keted in countries outside of the United States (US), and
`generic GA products await US FDA approval at this writing
`[97,98]. While it is not possible to show that a generic product
`is the same as Copaxone®, demonstration of “similarity” may
`be adequate for generic approval. Thus far, comparisons of
`physicochemical and biological activity have shown differences
`between purported generic GA products and Copaxone®
`[99,100]. A purported generic GA product is currently marketed
`in India (Glatimer®, Natco Pharma, Ltd., Hyderabad, India) [99].
`No information about the safety, efficacy, or immunogenicity
`of this product in RRMS patients has been published at this
`time. However, in analytical tests, Glatimer® has demonstrated
`physicochemical differences from GA [95,96], and gene
`expression studies, which provide a “snapshot” of biological
`processes stimulated by glatiramoid treatment, show Glatimer®
`also has different biologic activities from those of GA [99,100].
`In 2 separate studies, activated splenocytes from GA-treated
`mice showed distinctly different gene transcription profiles
`when reactivated ex vivo by Glatimer® or GA [99,100]. As
`previously described, among GA effects on APCs is down-
`regulation of macrophage and monocyte activation, and
`among GA effects on T cells is skewing T cell differentiation
`toward a Treg phenotype that limits autoimmune activity
`[73,101]. Figure 3 shows the relative expression of Treg-specific,
`macrophage-specific, and monocyte-specific genes in
`splenocyte samples reactivated by GA compared with samples
`activated by Glatimer® [100]. A cell-type enrichment algorithm
`showed the gene expression profile produced by splenocytes
`activated with Glatimer® was significantly enriched in genes
`associated with macrophages and monocytes compared with
`the gene expression profile of splenocytes activated by GA.
`Similarly, compared with GA, the list of genes down-regulated
`by splenocyte activation with Glatimer® was significantly enr-
`iched in genes associated with Tregs. Thus, Glatimer® could have
`very different immunomodulatory effects than GA. Another
`important finding of gene expression studies was poor batch-to-
`batch reproducibility among Glatimer® batches (Figure 4) [99,100].
`
`5
`
`Jill Conner, Journal of Autoimmunity and Cell Responses 2014,
`http://www.hoajonline.com/journals/pdf/2054-989X-1-3.pdf
`
`Page 5 of 11
`
`YEDA EXHIBIT NO. 2007
`MYLAN PHARM. v YEDA
`IPR2014-00644
`
`

`
`GA
`
`Generic
`
`doi: 10.7243/2054-989X-1-3
`
`GA
`N=24
`
`Generic
`N=105
`
`100
`
`80
`
`60
`
`40
`
`20
`
`0
`
`Highly Variable Probes By F Test
`
`Figure 4. Biological impact of glatiramer acetate is
`significantly more consistent than that of the purported
`generic product.
`Among probes with variability induced by activation, 4-fold
`more probes had significant variation by F-test across 11
`generic-activated samples from 5 batches, when compared
`with the number of probes with significant variation by F-test
`across 34 GA-activated samples from 30 batches [100].
`
`Under current regulatory statutes in the US, the first clinical
`exposure to generic GA products in MS patients could occur
`after regulatory approval [108]. Adequate testing of the clinical
`safety, efficacy, and immunogenicity of uncharacterized
`follow-on glatiramoid products in MS patients should
`precede regulatory approval and marketing of these products,
`given their complexity, uncharacterized epitopes, unknown
`mechanism of action, and strong immunogenicity. Moreover,
`the inter- and intra-patient variability of antibody responses to
`these immunogenic drugs should be assessed [109]. To ensure
`that the immunologic and immunogenic safety of a generic
`productare comparable to those of Copaxone® will likely
`require clinical trials in MS patients. Antibody responses to a
`generic product should establish that anti-drug antibodies do
`not neutralize drug efficacy or bind to endogenous proteins.
`Risks of free substitution of a follow-on product for Copaxone®
`should be established in a clinical cross-over study that
`provides direct evidence that repeated switching between
`Copaxone® and a generic drug has no negative impact.
`
`Conclusion and future therapeutic peptide vaccines
`Currently, novel synthetic copolymers designed on the basis
`of the immunological properties of GA are under study for
`use in MS [110]. IP-2301, a peptide copolymer currently in
`pharmaceutical development, has 3 of the same amino acid
`constituents as GA (tyrosine, alanine, and lysine) but replaces
`glutamic acid with phenylalanine. This vaccine has been shown
`
`6
`
`FoxP3+ T Cells
`
`Macrophages
`
`Monocytes
`
`1.00
`0.67
`0.33
`0.00
`-0.33
`-0.67
`-1.00
`
`Figure 3. Cell-type specific differences in the biological
`impact of glatiramer acetate and a purported generic
`glatiramer acetate-like product. Differential gene expression
`in mouse splenocytes activated ex vivo with GA or a generic
`product show GA induces higher expression of Treg-associated
`genes than the generic product, and the generic product
`induces higher expression of macrophage and monocyte-
`associated genes [100].
`
`Differences in gene expression profiles have also been detected
`among other purported generic GA products, including Pro-
`bioglat® (Probiomed S.A. de C.V.; Mexico City, Mexico [102]),
`Escadra® (MR PHARMA S.A., Buenos Aires, Argentina), and
`a generic product marketed by several companies in China
`(“Hangzhou”) [96]. Progesterone receptor membrane com-
`ponent 1 (PGRMC1) is significantly down-regulated by Pro-
`bioglat® relative to medium (i.e., the gene expression profile
`from non-reactivated murine splenocytes), but not by Escadra®
`or Hangzhou (Figure 5). Progesterone is thought to play
`a role in myelin repair [103,104] and to affect the balance
`between Tregs and Th cells [105]. Another relevant gene

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