throbber
Autoimmunity Reviews 6 (2007) 469 – 475
`
`www.elsevier.com/locate/autrev
`
`Glatiramer acetate: Mechanisms of action in multiple sclerosis
`Wiebke Schrempf, Tjalf Ziemssen ⁎
`
`Multiple Sclerosis Center, Department of Neurology, Dresden University of Technology, Fetscherstraße 74, 01307 Dresden, Germany
`
`Received 2 January 2007; accepted 6 February 2007
`Available online 6 March 2007
`
`Abstract
`
`Glatiramer acetate (GA) is a mixture of synthetic polypeptides composed of four amino acids resembling myelin basic
`protein (MBP). GA has been shown to be effective in preventing and suppressing experimental autoimmune encephalomyelitis
`(EAE), the animal model of multiple sclerosis. It was tested in several clinical studies and approved for the immunomodulatory
`treatment of
`relapsing-type MS in 1996. Glatiramer acetate demonstrates a strong promiscuous binding to major
`histocompatibility complex molecules and inhibits the T cell response to several myelin antigens. In addition, it was shown
`to act as a T cell receptor antagonist for the 82–100 MBP epitope. Glatiramer acetate treatment causes in vivo changes of the
`frequency, cytokine secretion pattern and effector function of GA-specific T cells. It was shown to induce GA-specific
`regulatory CD4+ and CD8+ T cells and a TH1–TH2 shift with consecutively increased secretion of antiinflammatory cytokines.
`GA-specific TH2 cells are able to migrate across the blood–brain barrier and cause in situ bystander suppression of
`autoaggressive TH1 T cells. In addition glatiramer acetate was demonstrated to influence antigen presenting cells (APC) such as
`monocytes and dendritic cells. Furthermore secretion of neurotrophic factors with potential neuroprotective effects was shown.
`© 2007 Elsevier B.V. All rights reserved.
`
`Keywords: Glatiramer acetate; Multiple sclerosis; Experimental Autoimmune Encephalomyelitis (EAE); Neuroprotection; Myelin Basic Protein
`(MBP)
`
`Contents
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`1. Multiple sclerosis.
`2. Drug development and clinical studies .
`3. GA as antigen-based therapy .
`.
`.
`.
`.
`.
`3.1.
`Effects of GA on APC .
`.
`.
`.
`.
`3.2.
`Effects of GA on T cells .
`.
`.
`.
`3.3.
`Effects of GA on B cells .
`.
`.
`.
`3.4. Neuroprotection .
`.
`.
`.
`.
`.
`.
`.
`.
`3.5.
`Biomarkers .
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`Take-home messages .
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`Take-home messages
`References .
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`. 470
`. 470
`. 470
`. 471
`. 471
`. 473
`. 473
`. 473
`. 473
`. 473
`
`⁎ Corresponding author. Tel.: +49 351 458 3859; fax: +49 351 458 5873.
`E-mail address: Ziemssen@web.de (T. Ziemssen).
`
`1568-9972/$ - see front matter © 2007 Elsevier B.V. All rights reserved.
`doi:10.1016/j.autrev.2007.02.003
`
`Page 1 of 7
`
`YEDA EXHIBIT NO. 2009
`MYLAN PHARM. v YEDA
`IPR2014-00644
`
`

`

`470
`
`W. Schrempf, T. Ziemssen / Autoimmunity Reviews 6 (2007) 469–475
`
`1. Multiple sclerosis
`
`Multiple sclerosis (MS) is a chronic, inflammatory
`demyelinating disease affecting about 0.1% of the
`population in temperate climates. It varies in terms of
`clinical, radiological and pathological features and is
`characterized by physical and neuropsychological
`symptoms. Pathological findings include axonal dam-
`age, brain atrophy and demyelinating plaques consisting
`of inflammatory cells [1]. Different disease entities are
`known leading from relapsing–remitting (RRMS) to
`primary (PPMS) and secondary progressive forms
`(SPMS). Current concepts assume that MS occurs as a
`consequence of immune tolerance breakdown in genet-
`ically susceptible individuals. The major contributing
`factors include environmental factors and immune
`dysregulation.
`The mechanisms of CNS inflammation involve
`activation of autoreactive, myelin specific T helper (TH)
`cells in the periphery possibly by molecular mimicry.
`These activated lymphocytes are able to cross the blood–
`brain barrier and are reactivated by recognising their
`specific target antigen presented by antigen presenting
`cells (APC) e.g. microglia. Reactivation of T cells results
`in increased secretion of proinflammatory cytokines and
`chemokines. An inflammatory cascade is initiated which
`causes activation and recruitment of macrophages and
`other inflammatory cells to the site of inflammation.
`Activation of B cells leads to augmented release of anti-
`myelin antibodies which bind complement and stimulate
`opsonization of myelin peptides. This ongoing destruc-
`tion of the myelin sheath causes reversible and to some
`extent irreversible impairment of functionality of the axon
`by deterioration of its conduction properties. As a major
`consequence of inflammation, axonal loss by binding of
`cytotoxic CD8+ T cells to demyelinated axons, release of
`cytotoxic factors and cytokines is proposed. In addition
`mechanisms not directly related to demyelination and
`inflammation i.e. excitotoxicity caused by glutamic acid is
`shown to initiate a process of neuronal cell death [1].
`
`2. Drug development and clinical studies
`
`Glatiramer acetate is a random mixture of synthetic
`polypeptides composed of four amino acids (L-glutamic
`acid, L-lysine, L-alanine and L-tyrosine) in a residue
`molar ratio 4.2 : 3.4 : 1.4 : 1.0 with an average molecular
`mass of 4700–11,000 Da. Myelin basic protein (MBP)
`is a major protein of the myelin sheath and is used as
`encephalitogenic protein for induction of experimental
`autoimmune encephalomyelitis (EAE)
`in animals.
`Glatiramer acetate was discovered in the 1960s when
`
`studies on the immunological properties of a series of
`polymers and copolymers developed to resemble MBP
`were conducted. Instead of EAE induction several of
`these polypeptides were able to decrease the severity or
`prevent EAE [2]. Copolymer 1,
`later known as
`glatiramer acetate, was shown to be the most effective
`polymer. It took several years until first exploratory
`open studies on patients with either relapsing remitting
`or secondary progressive multiple sclerosis were
`performed in the late 1970s and early 1980s. However
`the results of these studies must be interpreted with
`caution as drug production was not standardized before
`1991. In 1991 a phase III multicentre, double-blind,
`placebo-controlled trial with standardized glatiramer
`acetate preparation was conducted at 11 US medical
`centres with 251 RRMS patients receiving treatment for
`two years. Relapse rate decreased about 30% which was
`statistically significant (P = 0.007) [4]. In 1996 GA was
`approved by the US Food and Drug Administration
`(FDA) as a treatment for patients with active relapsing–
`remitting MS. Since then, studies using an oral formu-
`lation of GA (CORAL) and applying GA to primary
`progressive multiple sclerosis patients (PROMISE)
`failed to demonstrate significant
`treatment effects.
`There are promising new data on using a higher GA
`dose (40 mg) which are currently studied in a multi-
`centre, double-blind, controlled trial (FORTE).
`
`3. GA as antigen-based therapy
`
`In contrast to other immunomodulatory MS therapies
`glatiramer acetate seems to preferentially affect immune
`cells in an antigen-specific way. The peptide mixture is
`applied to patients with a putative autoimmune disease
`over many years by daily injection. From a broader
`perspective GA is one of the few practical examples of
`therapeutic vaccination distinct
`from prophylactic
`vaccination against
`infectious diseases. The precise
`mechanism of action of glatiramer acetate is not fully
`understood yet. The mechanisms discussed include
`competition with myelin peptides especially MBP for
`binding to major histocompatibility molecules (MHC)
`on APC, antagonism at the T cell receptor of myelin
`specific T cells and induction of GA-specific suppres-
`sive/regulatory T cells secreting potentially antiinflam-
`matory cytokines upon activation (Table 1). The vast
`majority of studies performed initially in EAE and
`subsequently in MS have focused on evaluating and
`targeting T cell responses especially the role of CD4+ T
`cells. Nevertheless there is increasing evidence for
`immunomodulating effects of GA on CD8+ T cells but
`also on B cells and APC.
`
`Page 2 of 7
`
`YEDA EXHIBIT NO. 2009
`MYLAN PHARM. v YEDA
`IPR2014-00644
`
`

`

`W. Schrempf, T. Ziemssen / Autoimmunity Reviews 6 (2007) 469–475
`
`471
`
`Table 1
`Immunological effects of glatiramer acetate
`
`MHC II-complex
`
`MBP–MHC-
`complex
`T cell receptor
`
`T cell proliferation in
`vivo after treatment
`T cell proliferation
`in vitro
`
`T cell migration
`T cell features
`
`Effect
`• Promiscuous binding to various
`HLA-DR alleles [5]
`• Displacement of already bound MBP
`from binding site of MHC II-complex
`• T cell receptor antagonism to the
`82–100 MBP epitope [15]
`• Decreased GA specific T cell frequency
`in patients on GA treatment [15,18]
`• Suppression of MBP specific
`T cell lines [6]
`• Reduced proliferation of GA-specific
`CD4+ T cells [23]
`• Reduced, mechanism unclear
`• Shift from TH1 to TH2 cells [6,14,16,17]
`• Bystander suppression [22]
`• Induction of suppressive/regulatory CD4+
`and CD8+ T cells [25–27]
`Effects on monocytes • Reduced activation upon stimulation [7]
`• Increased IL-10, IL-4 secretion [9]
`• Inhibition of TNF-α secretion in vitro [8]
`• Inefficient maturation [12]
`• Reduced ability to stimulate T-cells [10,11]
`• Increased IL-10 secretion [9,10]
`• Controversial findings regarding
`TNF-α [8,9]
`• Decreased IL-12 secretion [10]
`• Increased IL-10 secretion
`• Decreased production of IL-12/TNF-α [11]
`• Enhanced secretion of
`IL-10 and TGF-β [14]
`• Inhibition of transformation to an
`activated microglia form [13]
`• Enhanced secretion of neurotrophic
`factors (BDNF, NT-3/4) [34]
`• Decreasing neuronal damage [35]
`• Increased neuronal proliferation [35]
`
`Effects on dendritic
`cells
`
`Effects on
`macrophages
`Astrocyte/microglia
`
`Neuroprotection
`
`3.1. Effects of GA on APC
`
`In vitro studies have shown that GA competes with
`myelin peptides from binding to MHC molecules and is
`able to displace MBP peptides from APC. GA binds to
`many different alleles of MHC class II molecules without
`antigen processing and without any obvious preference
`(“promiscuous binding”) [5]. Thus GA inhibits MHC II
`assisted activation of myelin specific T cells [6]. GA
`treatment leads to a generalized, antigen-non-specific
`modulation of APC function i.e. monocytes and macro-
`phages resulting in a reduced reactivity towards proin-
`flammatory stimuli [7]. First
`in vitro experiments
`demonstrated that GA blocked the activation of a
`monocytic cell line in the absence of T cells by inhibiting
`the induction of HLA-DR and HLA-DQ proteins leading
`to a reduction of Tumor Necrosis Factor α (TNF-α)
`
`release and decline of cathepsin B activity [8]. GA-treated
`monocytes and macrophages also produced increased
`amounts of Interleukin-10 (IL-10) [9]. GA-treated
`dendritic cells were able to shift the phenotype of naive
`T cells towards TH2 like T cells. In addition GA inhibited
`the secretion of IL-12 and TNF-α by human dendritic
`cells and macrophages [10]. In contrast to these findings
`GA exposed bone-marrow-derived dendritic cells pro-
`duced increased amounts not only of IL-10 but also of
`TNF-α upon stimulation maybe due to a less effective
`autoregulation via IL-10 [9]. Sanna et al. [11] recently
`demonstrated that in MS patients pretreatment with GA
`significantly decreased the proliferative effect of dendritic
`cells on lymphocytes in vitro. Furthermore GA treated
`MS patients showed a significant reduction of GA
`induced proliferation of peripheral blood mononuclear
`cells. Plasmacytoid dendritic cells (pDCs) from multiple
`sclerosis patients were shown to exhibit an inefficient
`maturation process after stimulation compared to controls.
`GA treatment partially restored phenotype and function of
`pDCs in these patients [12]. Chabot et al.
`[13]
`demonstrated a reduced ability of GA-treated T cells to
`interact with microglia. Consecutively the secretion of
`proinflammatory cytokines was reduced in microglia-T
`cell-coculture experiments. Furthermore CNS infiltration
`of GA-specific T cells resulted in increased expression of
`Transforming Growth Factor β (TGF-β) and IL-10 by
`resident astrocytes and microglia [14].
`
`3.2. Effects of GA on T cells
`
`In addition to competition for binding to MHC mole-
`cules and thus inhibiting the T cell response to several
`myelin antigens, GA is proposed to act as an antagonist to
`MBP/MHC at MBP-specific T cell receptors (TCR). It
`was demonstrated to operate as an “altered peptide ligand”
`to the 82–100 epitope of MBP [15]. This effect presum-
`ably occurs in the periphery at the injection sites or in the
`corresponding draining lymph nodes. The immunomod-
`ulatory effect of GA is primarily attributed to its ability to
`induce in vivo changes of the cytokine secretion pattern
`and the effector function of GA-reactive T helper cells.
`Modulation of the immune response with glatiramer
`acetate leads to deviation of cytokine production in
`response to MBP from so-called TH1 cytokines to TH2
`cytokines [6,16,17]. This so called TH1–TH2 shift is
`characterized by increased secretion of antiinflamma-
`tory cytokines like IL-4 and decreased TNF-α and
`Interferon-γ (IFN-γ) production [18]. After prolonged
`GA treatment T cell response of MS patients remains
`TH2 biased [19]. In EAE experiments GA-reactive TH2
`cells could be detected in the CNS [20]. Furthermore,
`
`Page 3 of 7
`
`YEDA EXHIBIT NO. 2009
`MYLAN PHARM. v YEDA
`IPR2014-00644
`
`

`

`472
`
`W. Schrempf, T. Ziemssen / Autoimmunity Reviews 6 (2007) 469–475
`
`human TH1 and TH2 GA-reactive T cells were shown
`to migrate across an artificial blood–brain barrier in vitro
`[21]. So it can be concluded that activated TH2 cells are
`able to cross the blood–brain barrier (BBB), accumulate in
`the CNS [20] and express in situ antiinflammatory
`cytokines like IL-4, IL-10, TGF-β and IL-5, which not
`only modulate the local milieu but are able to suppress the
`activity of encephalitogenic TH1 cells [16–18]. This so
`called bystander suppression is regarded as essential part
`of the mechanisms of action of GA [22] (see Fig. 1). It
`seems very unlikely that sufficient amounts of GA reach
`the CNS to compete with locally degraded myelin
`antigens. So it might be speculated that GA-specific TH2
`cells are reactivated in the CNS when crossreacting with
`locally presented myelin autoantigens.
`In the setting of autoimmunity, one of the goals of
`successful therapeutic immune modulation is the induc-
`tion of peripheral tolerance, a large part of which is
`mediated by regulatory T cells. GA treatment gradually
`reduces the proliferative reactivity of GA-specific
`CD4+ T cells [23] probably by inducing apoptosis in
`some T cells [24]. Karandikar et al. [25] demonstrated
`
`that during GA treatment proliferative CD4+ T cell
`responses were comparable in healthy individuals and
`MS patients, whereas untreated MS patients showed a
`deficit in CD8+ T cell-mediated proliferation towards
`GA compared with healthy subjects. GA-specific treat-
`ment enhanced the frequency and suppressive ability of
`GA specific CD8+ T cells to those seen in healthy
`individuals. CD8+ T cells from GA treated patients and
`healthy subjects, but not those from untreated patients
`with MS, exhibited potent, HLA class I-restricted, GA-
`specific cytotoxicity [26]. In addition to these regula-
`tory CD8+ T cells Hong et al. [27] demonstrated that
`expression of Foxp3 in CD4+ T cells was significantly
`increased in MS patients treated with GA. This
`induction of regulatory CD4+ T cells was mediated by
`IFN-γ and to a lesser degree TGF-β1. Adoptive transfer
`of GA-specific T cells suppresses induction of EAE by
`different encephalitogens, but cannot cure ongoing
`EAE, which shows that T cells alone have only a limited
`role when disease is established [15,22]. By contrast
`GA injections not only prevent but also suppress
`established EAE [2].
`
`Fig. 1. Mechanisms of action of glatiramer acetate in multiple sclerosis. 1. GA exhibits competitive binding at the MHC-II complex and TCR-
`antagonism. In addition GA is able to displace MBP from the binding site on MHC-II molecules. Treatment with GA leads to the induction of antigen
`specific TH2 T cells in the periphery. 2. In addition CD8+ and CD4+CD25+ regulatory T cells are induced by GA therapy. 3. The constant activation
`seems to have an important impact on the induction and maintenance of the regulatory/suppressive immune cells. 4. Because of the daily activation,
`GA T cells are believed to be able to cross the blood–brain barrier. 5. Inside the CNS, some GA-specific T cells cross-react with products of local
`myelin turnover presented by local APCs. 6. In response anti-inflammatory cytokines are secreted which dampen the local inflammatory process
`(bystander suppression). 7. Furthermore GA specific T cells secrete neurotrophic factors which might favor remyelination and axonal protection.
`
`Page 4 of 7
`
`YEDA EXHIBIT NO. 2009
`MYLAN PHARM. v YEDA
`IPR2014-00644
`
`

`

`W. Schrempf, T. Ziemssen / Autoimmunity Reviews 6 (2007) 469–475
`
`473
`
`3.3. Effects of GA on B cells
`
`A report including data from three different therapeutic
`trials demonstrated that all GA treated patients developed
`anti-GA antibodies which declined 6 months after
`treatment initiation but were still detectable. Anti-MBP-
`antibody titers were low and remained unchanged during
`treatment. Antibodies were of the IgG class with IgG1
`isotype levels two- to threefold higher than those of IgG2
`[3]. Farina et al. showed GA-specific IgG4 antibodies in
`GA-treated MS patients in a cross-sectional study. In
`addition they showed the occurrence of GA antibodies of
`the IgM, IgG1 and IgG2 class in some untreated patients
`[28]. Also in GA-treated primary progressive MS patients
`GA-reactive antibodies of the IgG1 subclass predomi-
`nated [29]. IgG1 subsequently decreased while anti-GA
`antibodies of the IgG4 subclass increased and remained
`high for the 3 years of follow-up. This isotype switch to
`IgG4 seems to reflect the interaction of B cells with TH2
`lymphocytes [30]. The presence of GA-reactive antibodies
`in some untreated control subjects suggests that such
`antibodies, which are mainly of the IgM isotype, are present
`in the common B cell immune repertoire. One might
`speculate that they are T cell independent, polyspecific
`“natural” antibodies produced by so called B1 cells.
`Teitelbaum et al. did not observe any neutralizing
`activity in sera of GA-treated patients. GA antibodies did
`not interfere with GA action neither in terms of MHC
`binding and T cell stimulation nor regarding suppression
`of EAE [31]. However in vitro experiments by Salama
`et al. demonstrated inhibition of GA-specific T cell lines
`by purified GA antibodies [32]. No clear evidence has yet
`been found that GA antibodies inhibit clinical efficacy in
`vivo. Moreover relapse-free patients seem to develop
`higher antibody titers, which would be consistent with a
`beneficial rather than neutralizing activity of the anti-
`bodies [3]. In a murine model of demyelinating disease
`induced by Theiler's virus remyelination of spinal cord
`axons was enhanced by GA antibodies [33]. These results
`support the hypothesis that the antibody response in GA
`treated patients may be beneficial by facilitating repair of
`demyelinated lesions.
`
`3.4. Neuroprotection
`
`Human GA-specific TH1-, TH2-, and TH0 cells all
`showed low levels of basal secretion of Brain Derived
`Neurotrophic Factor (BDNF) and an increase of BDNF
`production after stimulation [34]. This ability of GA-
`specific cell could also be demonstrated in situ in the
`mouse model of MS (EAE) resulting not only in de-
`creased neuronal damage, but also increased neuronal
`
`proliferation [35]. Gilgun-Sherki et al. demonstrated a
`significant reduction of axonal loss and neuronal damage
`in myelin oligodendrocyte glycoprotein (MOG)-induced
`EAE, a model characterized by a chronic disease course
`[36]. Kipnis et al. showed that in a model of traumatic
`injury of the optic nerve the posttraumatic spread of
`degeneration could be attenuated by either active im-
`munization with GA on the day of injury or by adoptive
`transfer of GA-specific T cells [37]. After restimulation
`by cross-reactive myelin antigens in the CNS, GA-
`reactive T cells seem to secrete not only immunomod-
`ulatory cytokines but also neurotrophic factors. There-
`fore these T cells might confer neuroprotection in
`addition to bystander suppression.
`
`3.5. Biomarkers
`
`Similar to effects seen in patients treated with other
`immunomodulatory drugs some GA-treated patients do
`not respond to therapy. Early identification of such non-
`responders would be very useful for adjusting therapy
`regimens. For this reason there has been much interest in
`the development of appropriate biomarkers. We are just
`running a study (COPIMMUNONET) which aims at
`finding a link between clinical outcome and immuno-
`logical parameters specific for GA.
`
`Take-home messages
`
`• Glatiramer acetate is an approved drug for the
`treatment of relapsing–remitting multiple sclerosis, a
`chronic inflammatory disease of the central nervous
`system.
`• Treatment with glatiramer acetate results in reduction
`of clinical exacerbations and progression rate in
`multiple sclerosis patients as well as a decrease of
`burden lesion shown on MRI scans.
`• Its proposed mechanisms of action include competi-
`tion with myelin antigens for binding to APC,
`antagonism at specific T cell receptors, induction of
`GA-specific regulatory CD4+ and CD8+ T cells which
`downregulate inflammation by secretion of antiin-
`flammatory cytokines and cause bystander suppres-
`sion of proinflammatory TH1 cells in the CNS.
`• In addition, neuroprotective effects as well as reg-
`ulatory effects on B cells, monocytes and dendritic
`cells have been shown.
`
`References
`
`[1] Steinman L. Multiple sclerosis: a two-stage disease. Nat
`Immunol 2001;2:762–4.
`
`Page 5 of 7
`
`YEDA EXHIBIT NO. 2009
`MYLAN PHARM. v YEDA
`IPR2014-00644
`
`

`

`474
`
`W. Schrempf, T. Ziemssen / Autoimmunity Reviews 6 (2007) 469–475
`
`[2] Teitelbaum D, Meshorer A, Hirshfeld T, Arnon R, Sela M.
`Suppression of experimental allergic encephalomyelitis by a
`synthetic polypeptide. Eur J Immunol 1971;1:242–8.
`[3] Brenner T, Arnon R, Sela M, Abramsky O, Meiner Z, Riven-
`Krietman R. Humoral and cellular
`immune responses to
`Copolymer 1 in multiple sclerosis patients treated with Copaxone.
`J Neuroimmunol 2001;115:152–60.
`[4] Johnson KP, Brooks BR, Cohen JA, Ford CC, Goldstein J, Lisak
`RP. Copolymer 1 reduces relapse rate and improves disability in
`relapsing–remitting multiple sclerosis: results of a phase III
`multicenter, double-blind placebo-controlled trial. The Copolymer
`1 Multiple Sclerosis Study Group. Neurology 1995;45:1268–76.
`[5] Fridkis-Hareli M, Teitelbaum D, Gurevich E. Direct binding of
`myelin basic protein and synthetic copolymer 1 to class II major
`histocompatibility complex molecules on living antigen-present-
`ing cells-specificity and promiscuity. Proc Natl Acad Sci U S A
`1994;91:4872–6.
`[6] Gran B, Tranquill M, Chen B, Bielekova W, Zhou W, Dhib-
`Jalbut S, et al. Mechanisms of immunmodulation by glatiramer
`acetate. Neurology 2000;55:1704–14.
`[7] Weber MS, Starck M, Wagenpfeil S, Meinl E, Hohlfeld R, Farina
`C. Multiple sclerosis: glatiramer acetate inhibits monocyte
`reactivity in vitro and in vivo. Brain 2004;127:1370–8.
`[8] Li Q, Milo R, Panitch H, Swoveland P, Bever Jr CT. Glatiramer
`acetate blocks the activation of THP-1 cells by interferon-
`gamma. Eur J Pharmacol 1998;342:303–10.
`[9] Jung S, Siglienti I, Grauer O, Magnus T, Scarlato G, Toyka KV.
`Induction of IL-10 in rat peritoneal macrophages and dendritic
`cells by glatiramer acetate. J Neuroimmunol 2004;148:63–73.
`[10] 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;170:4483–8.
`[11] Sanna A, Fois ML, Arru G, Huang YM, Link H, Pugliatti M, et
`al. Glatiramer acetate reduces lymphocyte proliferation and
`enhances IL-5 and IL-13 production through modulation of
`monocyte-derived dendritic cells in multiple sclerosis. Clin Exp
`Immunol 2006;143:357–62.
`[12] Stasiolek M, Bayas A, Kruse N, Wieczarkowiecz A, Toyka KV,
`Gold R, et al. Impaired maturation and altered regulatory
`function of plasmacytoid dendritic cells in multiple sclerosis.
`Brain 2006;129:1293–305.
`[13] Chabot S, Yong FP, Le DM, Metz LM, Myles T, Yong VW.
`Cytokine production in T-lymphocyte–microglia interaction is
`attenuated by glatiramer acetate: a mechanism for therapeutic
`efficacy in multiple sclerosis. Mult Scler 2002;8:299–306.
`[14] Aharoni R, Kayhan B, Eilam R, Sela M, Arnon R. Glatiramer
`acetate-specific T cells in the brain express T helper 2/3 cytokines
`and brain-derived neurotrophic factor in situ. Proc Natl Acad Sci
`U S A 2003;100:14157–62.
`[15] 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 U S A
`1999;96:634–9.
`[16] Duda PW, Schmied MC, Cook SL, Krieger JI, Hafler DA.
`Glatiramer acetate (Copaxone) induces degenerate, Th2-polar-
`ized immune responses in patients with multiple sclerosis. J Clin
`Invest 2000;105:967–76.
`[17] Neuhaus O, Farina C, Yassouridis A, Wiendl H, Then Bergh F,
`Dose T. 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 A
`2000;97:7452–7.
`[18] Miller A, Shapiro S, Gershtein R, Kinarty A, Rawashdeh H,
`Honigman S, et al. Treatment of multiple sclerosis with
`copolymer-1 (Copaxone): implicating mechanisms of Th1 to
`Th2/Th3 immune-deviation. J Neuroimmunol 1998;92:113–21.
`[19] Chen M, Conway K, Johnson KP, Martin R, Dhib-Jalbut S.
`Sustained immunological effects of Glatiramer acetate in patients
`with multiple sclerosis treated for over 6 years. J Neurol Sci
`2002;201:71–7.
`[20] 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
`U S A 2000;97:11472–7.
`[21] Kim HJ, Biernacki K, Prat A, Antel JP, Bar-Or A. Inflammatory
`potential and migratory capacities across human brain endothelial
`cells of distinct glatiramer acetate-reactive T cells generated in
`treated multiple sclerosis patients. Clin Immunol 2004;111:38–46.
`[22] Aharoni R, Teitelbaum D, Sela M, Arnon R. Bystander
`suppression of experimental autoimmune encephalomyelitis by
`T cell lines and clones of the Th2 type induced by copolymer 1.
`J Neuroimmunol 1998;91:135–46.
`[23] Ragheb S, Abramczyk S, Lisak D, Lisak R. Long-term therapy
`with glatiramer acetate in multiple sclerosis: effect on T cells.
`Mult Scler 2001;7:43–7.
`[24] Ruggieri M, Avolio C, Scacco S. Glatiramer acetate induces pro-
`apoptotic mechanisms involving Bcl-2, Bax and Cyt-c in
`peripheral lymphocytes from multiple sclerosis patients. J Neurol
`2006;253:231–6.
`[25] Karandikar NJ, Crawford MP, Yan X, Ratts RB, Brenchley JM,
`Ambrozak DR, et al. Glatiramer acetate (Copaxone) therapy
`induces CD8(+) T cell responses in patients with multiple
`sclerosis. J Clin Invest 2002;109:641–9.
`[26] Tennakoon DK, Mehta RS, Ortega SB, Bhoj V, Racke MK,
`Karandikar NJ. Therapeutic induction of regulatory, CD8+
`T cells in Multiple Sclerosis. J Immunol 2006;176:7119–29.
`[27] Hong J, Li N, Zhang X, Zheng B, Zhang JZ. Induction of
`CD4+CD25+ regulatory T cells by copolymer-I
`through
`activation of transcription factor Foxp3. Proc Natl Acad Sci
`U S A 2005;102:6449–54.
`[28] Farina C, Vargas V, Heydari N, Kümpfel T, Meinl E, Hohlfeld R.
`Treatment with glatiramer acetate induces specific IgG4
`antibodies in multiple sclerosis patients. J Neuroimmunol
`2002;123:188–92.
`[29] Basile E, Gibbs E, Aziz T, Oger J. During 3 years treatment of
`primary progressive multiple sclerosis with glatiramer acetate,
`specific antibodies switch from IgG1 to IgG4. J Neuroimmunol
`2006;177:161–6.
`[30] Ishizaka A, Sakiyama Y, Nakanishi M, Tomizawa K, Oshika E,
`Kojima K, et al. The inductive effect of interleukin-4 on IgG4 and
`IgE synthesis in human peripheral blood lymphocytes. Clin Exp
`Immunol 1990;79:392–6.
`[31] Teitelbaum D, Brenner T, Abramsky O, Aharoni R, Sela M,
`Arnon R. Antibodies to glatiramer acetate do not interfere with its
`biological
`functions and therapeutic efficacy. Mult Scler
`2003;9:592–9.
`[32] Salama HH, Hong J, Zang YCQ, El Mongui A, Zhang JW.
`Blocking effects of serum reactive antibodies induced by glatiramer
`acetate treatment in multiple sclerosis. Brain 2003;126:2638–47.
`[33] Ure DR, Rodriguez M. Polyreactive antibodies to glatiramer
`acetate promote myelin repair in murine model of demyelinating
`disease. FASEB J 2002;16:1260–2.
`
`Page 6 of 7
`
`YEDA EXHIBIT NO. 2009
`MYLAN PHARM. v YEDA
`IPR2014-00644
`
`

`

`W. Schrempf, T. Ziemssen / Autoimmunity Reviews 6 (2007) 469–475
`
`475
`
`[34] Ziemssen T, Kümpfel T, Klinkert WE, Neuhaus O, Hohlfeld R.
`Glatiramer acetate-specific T-helper 1- and 2-type cell
`lines
`produce BDNF:
`implications for multiple sclerosis therapy.
`Brain-derived neurotrophic factor. Brain 2002;125:2381–91.
`[35] Aharoni R, Eilam R, Domev H, Labunskay G, Sela M, Arnon R.
`The immunomodulator glatiramer acetate augments the expres-
`sion of neurotrophic factors in brains of experimental autoim-
`mune encephalomyelitis mice. Proc Natl Acad Sci U S A
`2005;102:19045–50.
`
`[36] Gilgun-Sherki Y, Panet H, Holdengreber V, Mosberg-Galili R,
`Offen D. Axonal damage is reduced following glatiramer ace-
`tate treatment in C57/bl mice with chronic-induced experi-
`mental autoimmune encephalomyelitis. Neurosci Res 2003;47:
`201–7.
`[37] Kipnis J, Yoles E, Porat Z, Cohen A, Mor F, Sela M, et al. T cell
`immunity to copolymer 1 confers neuroprotection on the
`damaged optic nerve: possible therapy for optic neuropathies.
`Proc Natl Acad Sci U S A 2000;97:7446–51.
`
`Prevalence of autoantibodies (ANA, anti ds-DNA, ENA, IMF) and rheumatic syndromes in patients with
`lymphoproliferative diseases.
`
`In the development of rheumatic syndromes as well as of lymphoproliferative disorders it is probable that there are
`common genetic, environmental and immunoregulatory pathogenetic mechanisms. The purpose of this study,
`(Chloraki-Bobota A. et. al. (J BUON 2006; 11: 485-9) was to determine the frequency of simultaneous
`presentation of both lymphoma or other lymphoproliferative diseases, and rheumatic syndromes. All patients with
`lymphoproliferative diseases (1920 patients) followed at Metaxa Cancer Hospital, Piraeus, Greece, during the last
`5 years, were included in this study. 312/1920 (16.2%) patients presented with non-Hodgkin's lymphoma (NHL),
`645/1920 (33.5%) had myeloma, 558/1920 (29%) had leukemia and miscellaneous other hematological
`malignancies. Anti nuclear antibodies (ANA), ribosomal P and intermediate filament antibodies (IMF), Anti-
`dsDNA and extractable nuclear antigens (ENA: Sm, RNP, SSA), were analyzed in all. 388/1920 (4.6%) patients
`were ANA positive. On the other hand, clinical symptoms attributed to autoimmune diseases (arthralgias, morning
`stiffness etc) plus autoantibodies other than ANA were present only in 8/312 (2.5%) patients with NHL, among
`them one with anti-cardiolipin antibodies. It is interesting that from these 8 patients, 3 had MALT lymphoma and 3
`diffuse B-cell large lymphoma. It was concluded that the simultaneous presence of lymphoproliferative diseases
`and rheumatic syndromes are more frequent among lymphoma patients than in other lymphoproliferative diseases.
`Therefore, the screening of antibodies in MHL patients may be useful for the discovery and the treatment of an
`underlying autoimmune disease.
`
`Proinflammatory mediator-induced reversal of CD4+CD25+ regulatory T cell-mediated suppression in rheumatoid
`arthritis.
`
`CD4+CD25+ T regulatory cells are present in increased numbers in the synovial fluid (SF) of rheumatoid arthritis
`(RA) patients and display enhanced suppressive activity as compared with their peripheral blood (PB)
`counterparts. Despite the presence of these immunoregulatory cells in RA, c

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