throbber
Expert Opinion on Pharmacotherapy
`
`ISSN: 1465-6566 (Print) 1744-7666 (Online) Journal homepage: http://www.tandfonline.com/loi/ieop20
`
`Glatiramer acetate for the treatment of multiple
`sclerosis
`
`Jerry S Wolinsky
`
`To cite this article: Jerry S Wolinsky (2004) Glatiramer acetate for the treatment of multiple
`sclerosis, Expert Opinion on Pharmacotherapy, 5:4, 875-891, DOI: 10.1517/14656566.5.4.875
`
`To link to this article: http://dx.doi.org/10.1517/14656566.5.4.875
`
`Published online: 02 Mar 2005.
`
`Submit your article to this journal
`
`Article views: 99
`
`View related articles
`
`Citing articles: 1 View citing articles
`
`Full Terms & Conditions of access and use can be found at
`http://www.tandfonline.com/action/journalInformation?journalCode=ieop20
`
`Download by: [UCSF Library]
`
`Date: 15 September 2016, At: 08:33
`
`Page 1 of 18
`
`Coalition Exhibit 1060
`Coalition v. Biogen
`IPR2015-01993
`
`

`
`Drug Evaluation
`
`Glatiramer acetate for the
`treatment of multiple sclerosis
`
`Jerry S Wolinsky
`University of Texas Health Science Center at Houston, 6431 Fannin Street, Houston, TX 77030, USA
`
`Glatiramer acetate (Copaxone®, Teva Pharmaceuticals Ltd) is a collection of
`immunomodulatory, synthetic polypeptides indicated for the treatment of
`relapsing-remitting multiple sclerosis (RR MS). Preclinical and clinical studies
`provide an evolving understanding of the mechanisms by which glatiramer
`acetate exerts both immunological and potential neuroprotective effects
`that account for its clinical efficacy. The results of pivotal controlled clinical
`trials and long-term data, derived from organised extension studies, are eval-
`uated in detail and supportive data from open-label comparison, combina-
`tion treatment and therapeutic switch studies are considered in order to
`determine the place of glatiramer acetate among other approved therapies
`for RR MS. The efficacy of glatiramer acetate is stable or may increase over
`time and the drug has a favourable side effect profile. Glatiramer acetate is
`an appropriate first-line immunomodulatory therapy for RR MS.
`
`Keywords: Copaxone, glatiramer acetate, immunomodulatory therapy, magnetic resonance
`imaging, multiple sclerosis
`
`Expert Opin. Pharmacother. (2004) 5(4):875-891
`
`1. Introduction
`
`Multiple sclerosis (MS) is an inflammatory disease of the CNS that leads to myelin
`destruction and axonal loss. It is the most common, non-traumatic, disabling neuro-
`logical disorder in young adults [1]. Although the aetiology of MS remains unknown,
`its pathogenesis is believed to include autoimmune reactivity to myelin components,
`placing it among the organ-specific autoimmune diseases. In relapsing-remitting
`(RR) MS, the clinical course is punctuated by exacerbations or episodic neurological
`worsening. These attacks or relapses, are followed within weeks to a few months by
`remissions, often with full recovery from clinical symptoms. However, recovery from
`up to 40% of all relapses is incomplete, leaving measurable neurological deficits or
`disability [2]. Progressive forms of MS are characterised by a gradual downhill course
`over many months to years without remissions, so that the patient acquires increas-
`ing clinical deficits, either beginning at presentation (primary-progressive [PP] MS)
`or after a period of RR disease (secondary-progressive [SP] MS).
`Currently approved immunomodulator therapies for RR MS include glatiramer
`
`
`acetate (GA) and the recombinant IFNs, (IFN-β1a, Avonex®, Biogen, Inc.; IFN-β1a,
`Rebif®, Serono, Inc.; IFN-β
`1b, Betaseron®, Berlex Laboratories), all of which modify
`the course of this progressively disabling neurological disease. Immunomodulatory
`treatments reduce disease activity and the accumulation of disability in RR MS. The
`National Multiple Sclerosis Society recommends initiation of therapy with an immu-
`nomodulator as soon as possible following diagnosis of RR MS [201]. Results of mag-
`netic resonance spectroscopy (MRS) and pathology studies show that inflammatory
`activity can cause irreversible axonal damage in the early phases of the disease, rein-
`forcing the need for early and aggressive treatment [3,4]. Mitoxantrone (Novantrone™),
`an antineoplastic agent, is also approved for the treatment of relapsing MS, but is gen-
`erally reserved for secondary progressive and severe RR forms of the disease [5].
`GA (formerly known as copolymer 1 or Cop 1) is indicated for the reduction of
`the frequency of relapses in RR MS. The drug is approved in 42 countries
`
`2004 © Ashley Publications Ltd ISSN 1465-6566
`
`875
`
`1. Introduction
`
`2. Immunopharmacology
`
`3. Clinical efficacy
`
`4. Tolerability
`
`5. Conclusion
`
`6. Expert opinion
`
`For reprint orders, please
`contact:
`reprints@ashley-pub.com
`
`Ashley Publications
`www.ashley-pub.com
`
`Page 2 of 18
`
`

`
`Glatiramer acetate
`
`Systemic
`
`Blood–brain
`barrier
`
`CNS
`
`Pre-Rx
`
`On-Rx
`
`Macrophage
`
`MHC
`
`'Myelin' Ag
`
`TCR
`
`GA
`
`'Myelin'
`Ag
`
`Microglia
`
`Neuron
`
`CD4+ Th1
`IL-2
`IFN-γ
`TNF-α
`LT
`
`CD4+ Th2
`IL-4
`IL-5
`IL-13
`IL-10
`BDGF
`
`BDNF
`
`Myelin internode
`
`Figure 1. A simplified diagrammatic representation of the immunopharmacology of GA in MS therapeutics. The Pre-Rx portion
`of the panel emphasises the baseline state in MS with CD4+ Th1 myelin antigen-reactive cells being activated by systemic antigen
`processing cells, including macrophages, that present foreign antigens that are myelin-like (‘myelin’ Ag) in the context of surface MHC to
`the TCR; invoking the concept of molecular mimicry. Stimulated CD4+ Th1 ‘myelin’ Ag-reactive cells secrete a number of pro-inflammatory
`cytokines (IL-2, IFN-γ, TNF-α and LT). WIth GA therapy, GA may displace some ‘myelin’ Ag. More importantly, on presentation and
`stimulation of GA and ‘myelin’ Ag-reactive CD4+ Th1 cells, GA silences crossreacting CD4+ Th1 ‘myelin’ Ag-reactive cells through anergy,
`apoptosis or antigen-specific mechanisms. Concomitantly, GA stimulates and expands a population of GA-reactive CD4+ Th2 cells that
`secrete anti-inflammatory cytokines (IL-4, -5, -13 and -10) to systemically inhibit ‘myelin’ Ag-reactive CD4+ Th1 cells (red arrow). With
`continued therapy the net result is a reduced proportion of CD4+ Th1 and an increased proportion of GA and ‘myelin’ crossreactive CD4+
`Th2 cells. When these GA and ‘myelin’ crossreactive CD4+ Th2 cells gain access to the CNS by trafficking across the blood–brain barrier,
`they are restimulated by true myelin Ags processed and presented by microglia, a brain-resident macrophage. On restimulation, the GA-
`reactive CD4+ Th2 cells secrete anti-inflammatory cytokines to inhibit ‘myelin’ Ag reactive CD4+ Th1 cells within the CNS and also secrete
`tropic factors, such as BDNF that may facilitate neuronal survival (green arrow). Modified from Neuhaus [14] and other sources.
`Ag: Antibody; BDNF: Brain-derived neurotrophic factor; BDGF: Brain-derived growth factor; GA: Glatiramer acetate; LT: Leukotriene; MHC: Major histocompatibility
`complex; MS: Multiple sclerosis; Rx: Treatment; TCR: T cell receptor.
`
`worldwide, including the US, Canada, Australia, Europe and
`Israel. A comprehensive review of GA was published in this
`journal in 2001 [6]. New data on the long-term clinical experi-
`ence with GA, comparative studies, therapy switching studies
`and magnetic resonance imaging (MRI) findings have since
`become available.
`GA is the acetate salt of a synthetic mixture of polypeptides
`that consists of random sequences of four naturally-occurring
`amino acids: L-glutamic acid, L-lysine, L-alanine and L-tyrosine
`in racemeric form at a defined molar ratio of 1.4:3.4:4.2:1.0,
`respectively. The copolymer was first synthesised in 1967 by
`Arnon et al. at the Weizmann Institute of Science [7] in an
`attempt to simulate some of the then known physicochemical
`
`properties of myelin basic protein (MBP) to induce and then
`dissect experimental allergic encephalomyelitis (EAE). EAE is a
`laboratory animal model of organ-specific autoimmune CNS
`inflammatory disease with some similarities to MS. Even
`though GA proved incapable of inducing EAE, it did demon-
`strate a marked effect in suppressing EAE when animals were
`subsequently challenged with MBP [8,9].
`
`2. Immunopharmacology
`
`The mechanisms of action of GA in humans remain uncertain
`but substantial preclinical data support both immunomodula-
`tory and neuroprotective effects of the drug. At least five
`
`876
`
`Expert Opin. Pharmacother. (2004) 5(4)
`
`Page 3 of 18
`
`

`
`interdependent processes are thought to contribute to the
`effects of GA (Figure 1):
`
`(cid:127) High affinity binding to the major histocompatibility
`complex (MHC) within the antigen binding pocket.
`(cid:127) Competition with MBP at the antigen-presenting cell
`(APC) level for binding to MHC and subsequent inhibi-
`tion of MBP-specific T cell activation through competition
`with MBP–MHC complexes for the T cell receptor.
`(cid:127) Induction of a shift in GA-reactive T cells from a T helper
`Type 1 (Th1) to a T helper Type 2 (Th2) phenotype.
`(cid:127) Migration of GA-specific T cells into the CNS.
`(cid:127) Neuroprotection induced via promotion of neurotrophic
`factors.
`
`As would be expected of repeated injection of any foreign pro-
`tein, immunisation with GA consistently induces GA anti-
`bodies; whether these may contribute or detract from the
`clinical effects of the drug are also considered in this section.
`
`2.1 High-affinity binding to major histocompatibility
`complex
`Fundamental to the development of an antigen-specific,
`T-cell-dependent immune response is the processing and
`presentation of a fragment of the antigen by an APC to a
`T cell precursor. This occurs when an appropriately processed
`antigen is bound by physicochemical interactions within the
`antigen-binding cleft of the MHC of an APC. The resulting
`unique structure is presented on the cell surface of the APC
`where it can interact with the complementary hypervariable
`portions of the T-cell receptors of appropriate T cells. Forma-
`tion of this trimolecular complex is a critical, although not
`necessarily sufficient, prerequisite to stimulating signals that
`activate and condition the behaviour of the T cell.
`Intact GA binds directly to MHC displayed on fixed APCs
`[10]. This binding can be blocked by anti-DR, but not anti-
`DQ, or anti-Class I antibodies, and GA binding to Class II
`must occur at, or very near to, the peptide-binding cleft. Iso-
`lated DR molecules exposed to GA form covalently linked
`complexes [10] and this interaction is not easily blocked by the
`staphylococcal B antigen. Staphylococcal B antigen has a
`known binding site to the Class II antigen that resides out-
`side of the antigen-binding cleft. The binding of GA to a
`Class II antigen is of high avidity and has been demonstrated
`for all common MS-associated DR haplotypes. Based on the
`crystallographic structure of the immunodominant peptide
`of MBP and DR2 [11], it appears that the repeated alanines
`and tyrosines in GA may facilitate the anchoring of GA
`within binding pockets of the Class II binding cleft. Further,
`variation in amino acid sequence inherent to GA could
`account for its ability to efficiently bind to a wide array of
`different Class II haplotypes. However, the high-affinity
`interaction between GA and Class II antigen alone is not suf-
`ficient to explain the mechanism of action of the drug, as the
`immunobiologically inert dextrorotatory form of GA binds
`with similar avidity to DR.
`
`Wolinsky
`
`2.2 Competition with myelin basic protein
`In vitro studies have shown that GA competes with MBP at
`the level of APC for binding to the MHC [12]. GA appears to
`have greater MHC-binding affinity than MBP and other
`myelin-associated proteins (e.g., proteolipid protein [PLP]
`and myelin oligodendrocyte glycoprotein [MOG]). Thus, GA
`efficiently displaces MBP-, PLP- and MOG-derived peptides
`from the MHC binding site, but is not displaced by these
`antigens once it is bound to the MHC [6]. GA isomers have
`the same effect on MHC but do not suppress EAE [13]. After
`binding to MHC, the GA–MHC complex competes with
`available MBP/MHC molecules for binding to T-cell recep-
`tors. As a consequence, some of the myelin-specific, patho-
`genic T cells may become anergic or otherwise altered [14].
`
`2.3 Induction of shift from T helper Type 1 to T helper
`Type 2 lymphocytes
`Data from animal models and ex vivo studies of human lym-
`phocytes show that exposure to GA induces a relative anti-
`inflammatory state by causing a shift in the GA-reactive lym-
`phocyte population from a dominant Th1 state to a Th2 dom-
`inant state [14,15]. Th1 cells produce IL-2, IL-12, IFN-γ and
`TNF-α, which generally behave as pro-inflammatory
`cytokines, whereas Th2 cells produce IL-4, -5, -6, -10 and -13,
`which generally exert anti-inflammatory effects. GA-reactive
`peripheral blood lymphocytes from untreated MS patients
`mostly express TNF-α mRNA, whereas those harvested from
`GA-treated patients mainly express IL-10, transforming
`growth factor (TGF)-β and IL-4 mRNA [16]. The shift towards
`Th2 bias is also demonstrated by the diminished ratio of
`IFN-γ/IL-5 secretion of GA-reactive T cell lines isolated from
`MS patients both before and during GA therapy [15,17]. The
`GA-reactive Th2 cells are believed to act as regulatory cells to
`modulate the pathogenic immune reaction. Many T cell lines
`reactive to a number of potentially encephalitogenic myelin
`proteins, when stimulated in vitro with GA, do not proliferate
`but do secrete cytokines with a predominant IL-5 pattern [18].
`It is important to realise that two phenomena are occurring
`in concert as patients begin therapy with GA. Both naive and
`memory GA-reactive CD4+ T cells are part of the resident
`T cell repertory of mice and men [19]. With initiation of ther-
`apy, the numbers of GA-reactive T cells that can be found using
`proliferation assays progressively falls after a transient increase
`within the first month of therapy. The reduced response is evi-
`dent within 3 – 6 months, substantial at 12 months and is
`decreased by 75% from baseline at 24 months after initiating
`treatment [20]. The proportion of patients with a negative
`in vitro proliferative response to GA increased from 5% at base-
`line to 40% at 2 years. Moreover, treatment with GA results in
`increased apoptosis of a substantial percentage of activated
`(CD69+) CD4+ T cells [21]. Thus, as the shift from a Th1 to a
`Th2 state is being established, the numbers of Th1 GA-reactive
`cells is also falling. Many of the above effects are likely to occur
`systemically, resulting in reduced availability of autoaggressive
`cells for entry into the CNS over time.
`
`Expert Opin. Pharmacother. (2004) 5(4)
`
`877
`
`Page 4 of 18
`
`

`
`Glatiramer acetate
`
`The Th2-biased immunological response seen with GA is
`sustained over long-term treatment. Chen et al. [22] isolated
`48 GA-reactive T cell lines from 10 RR MS patients who had
`taken GA for 6 – 9 years. Proliferative responses, cytokine pro-
`duction and crossreactivity with whole MBP and the MBP
`immunodominant peptide 83-99 were compared with
`responses obtained from 10 MS patients tested before GA
`treatment and after shorter treatment periods (1 – 10 months).
`Long-term treatment with GA resulted in a 2.9-fold decrease
`in the estimated precursor frequency of GA-reactive T cells.
`Nevertheless, the sustained response to GA remained Th2-
`biased and, in part, crossreactive with MBP and MBP (83-99),
`as measured by proliferation and cytokine-release assays [22].
`
`2.4 Migration of activated T cells into the central
`nervous system
`Areas of active CNS demyelination and axonal loss in MS
`white matter lesions contain substantial numbers of inflamma-
`tory cells. GA does not appear to directly inhibit the transmi-
`gration of these inflammatory cells into the CNS. Rather,
`repeated systemic activation by daily GA treatment stimulates
`GA-reactive T cells, which increasingly become Th2-like [23].
`In vitro models demonstrate that Th2 cells can penetrate the
`CNS [24]. Adoptively transferred GA-reactive T cells adminis-
`tered systemically to recipient mice are detectable in the ani-
`mals’ CNS [23,25]. GA-reactive Th2 cells in the CNS are
`postulated to decrease local inflammation through ‘bystander
`suppression’. It is thought that GA-specific Th2 cells within the
`CNS are restimulated by-products of myelin turnover pre-
`sented by local APCs. The antigen presented within the CNS
`cannot be GA, as the drug is rapidly metabolised in subcutane-
`ous tissue at the administration site. Local reactivation of GA-
`specific T cells stimulates the release of anti-inflammatory
`cytokines such as IL-4, -6, -10, TGF-β and brain-derived neu-
`rotrophic factor (BDNF), but not IFN-γ [25,26]. The produc-
`tion of pro-inflammatory cytokines, including IL-2 and IFN-γ,
`is inhibited through this bystander effect. The mechanism of
`bystander suppression may make GA useful in other autoim-
`mune diseases of the CNS where Th1 cells predominate [27].
`
`2.5 Neuroprotection
`A recently identified mechanism of action of GA is related to
`a potential neuroprotective effect of some autoreactive Th1
`and Th2 cells. This possibility was first raised after unex-
`pected findings were reported in a rodent optic nerve crush
`injury model, which typically results in a predictable loss of
`retinal-ganglion neurons. In early experiments, animals
`injected with MBP-reactive T cells immediately after the
`crush injury exhibited attenuated subsequent loss of retinal
`ganglion neurons, but also suffered adoptive transfer EAE [28].
`In subsequent experiments, rats subjected to optic nerve crush
`injury and then injected with GA-specific T cells showed
`increased retinal ganglion neuron survival, compared with
`injured controls, and did not develop EAE [29]. Moreover, in a
`murine model in which intraocular injection of glutamate
`
`destroys retinal ganglion neurons, glutamate toxicity was
`reduced in mice immunised with GA, but not in those immu-
`nised with either MBP or MOG [30]. The neuroprotective
`effects of GA have since been demonstrated in several animal
`models. Compared with untreated controls, GA treatment
`reduced axonal damage in C57/bl mice with chronic EAE [31],
`and increased survival time and improved motor function in a
`murine model of amyotrophic lateral sclerosis (ALS) [32].
`A variety of mechanisms underpinning the neuroprotective
`effects of GA are currently under investigation. Kayhan et al.
`[33] demonstrated that in mice, induction of EAE leads to
`fourfold elevation in nitric oxide (NO) secretion. NO is an
`inflammatory mediator thought to affect regulation of the
`immune response, permeability of the blood–brain barrier,
`trafficking of cells to the CNS and immunosuppression. NO
`has been implicated in primary demyelination via nonspecific
`damage to the myelin sheath of axons, as well as promoting
`direct oligodendrocyte death [34]. Treatment of EAE mice
`with GA leads to a significant decrease in NO secretion by the
`splenocytes in response to the encephalitogen [33].
`Another neuroprotective mechanism may involve GA-stim-
`ulated secretion of BDNF, a neurotrophic factor that plays an
`important role in plasticity and development of the nervous
`system [27,35]. Ziemssen et al. [36] determined that GA-specific
`Th2 and Th1 peripheral blood mononuclear cells produce
`BDNF. Using three GA-specific long-term T cell lines with
`phenotypes Th1, Th1/0 and Th0 derived from a healthy sub-
`ject and one T cell line with a Th2 phenotype derived from a
`GA-treated MS patient, they demonstrated that all four T cell
`lines could be stimulated to produce BDNF [36]. Similarly,
`Chen et al. [26] studied BDNF production in 73 GA-reactive,
`13 MBP-reactive and two tetanus toxoid (TT)-reactive T cell
`lines isolated from 12 MS patients treated with GA. BDNF
`levels produced by GA-specific T cells generated during treat-
`ment were higher than those generated pretreatment. Among
`the 73 GA T cell lines generated, 14% secreted levels of
`BDNF two standard deviations above the GA T cell line
`mean. All GA-reactive T cells that secreted high levels of
`BDNF were Th2 biased. A total of 26, 14 and 13 GA-, MBP-
`and TT T cell lines, respectively, originated from the same
`4 MS patients and could be compared directly. The mean
`BDNF level for the GA-reactive T cell lines was significantly
`higher than that for the MBP- and TT-reactive T cell lines.
`The signal transducing receptor for BDNF, the full-length
`145 tyrosine kinase receptor (trk) B, is expressed in neurons
`and astrocytes in MS lesions [37]. Therefore, BDNF secreted
`by GA-reactive Th1 and Th2 cells in the CNS could exert
`neurotrophic effects directly in the MS target tissue. GA-
`reactive T cells also appear to have a reduced ability to trans-
`form bipolar microglia into a morphologically activated ame-
`boid form [38].
`
`2.6 Glatiramer acetate antibodies
`Immunisation of mice and other laboratory animals with GA
`results in the development of polyclonal GA antibodies.
`
`878
`
`Expert Opin. Pharmacother. (2004) 5(4)
`
`Page 5 of 18
`
`

`
`There is little evidence of crossreaction of the polyclonal GA
`antibodies with MBP or other myelin proteins. However,
`some IgM monoclonal antibodies (mAb) generated against
`either GA or murine MBP do show substantial crossreactivity
`in binding and competition assays [39]. Of interest, polyclonal
`murine GA IgG antibodies stimulate remyelination in a
`murine viral model of inflammatory demyelination [40].
`Patients receiving GA in controlled trials develop demon-
`strable levels of GA-binding antibodies within 1 month of
`starting treatment that greatly exceed background levels found
`in placebo-treated patients. These antibodies are restricted to
`the IgG class, with IgG1 levels several-fold higher than IgG2
`levels and low levels of IgG4 [41] – patterns consistent with a
`Th2-driven response. The GA antibodies peak within
`3 months of treatment initiation, reaching levels of 8- to
`20-fold higher than baseline. GA antibody titres decrease by
`month 6 of therapy, but persist at detectable levels for at least
`2 years of continued treatment [20]. In patients treated for
`2 years, those who were relapse-free at 18 and 24 months of
`therapy had statistically higher GA antibody titres than
`treated patients who had one or more on-trial relapses. No
`correlation between antibody titre and either disability as
`measured by categorical change from baseline EDSS or side
`effect profile, was observed [20].
`Extensive attempts to show that IgG class GA mAbs
`inhibit cellular responses to GA either in vitro and in vivo
`have failed to do so [42]. These have included attempts to
`block the binding of GA to isolated MHC molecules, to
`inhibit the responses of GA-specific T cell lines to proliferate
`or secrete cytokines upon stimulation by GA and to inhibit
`the effect of GA in EAE, either by premixing the mAbs with
`the GA inocula or with passive transfer of the immunised
`mice. Similar negative results were found with high-titre
`human GA-antibody sera from treated MS patients. Moreo-
`ver, 53 serum samples from 34 GA-treated patients failed to
`reduce the proliferative response of a murine GA-specific
`T cell clone to GA or reduce the competitive inhibition of
`proliferation of an MBP-specific human T cell clone by GA.
`Discrepant results were reported by Zhang et al. [43]. Using
`an enzyme-linked
`immunosorbent assay (ELISA)-based
`binding assay, GA antibodies were found in 48% of
`42 RR MS patients treated with GA for 1 – 5 years, with
`most antibody-positive patients seen after 1 year on therapy.
`Of the 14 high-titred sera that were found to inhibit the pro-
`liferation of normal donor peripheral blood cells to GA,
`6 were selected for further study. At low dilution, these 6 sera
`inhibited the proliferate responses of a panel of GA-specific
`T cell lines to GA to a varying extent.
`In summary, the predominance of available data does not
`suggest GA antibodies influence the therapeutic effect of the
`drug. However, given the considerable importance of binding
`and neutralising antibodies to IFN-β that develop during ther-
`apy on longer-term clinical and MRI outcomes [44], additional
`studies of long-term GA-treated patients are appropriate.
`
`Wolinsky
`
`3. Clinical efficacy
`
`3.1 Controlled clinical trials
`3.1.1 Bornstein single centre study
`The first double-blind, randomised, placebo-controlled trial
`of GA in patients with RR MS was conducted in the US in
`the 1980s [45]. A total of 50 patients, 20 – 35 years of age
`(mean: 30.5 years) with ≥ 2 relapses during the 2 years before
`study enrollment (average number: 3.9 relapses) with a
`Kurtzke Disability Status Scale (DSS) score of ≤ 6 (mean
`score: 3) were studied. Patients received GA or placebo
`20 mg/day s.c. for 2 years. The treatment groups were
`matched for gender, number of prior relapses and extent of
`entry disability (DSS 0 – 2 or 3 – 6). The primary end point
`was the proportion of relapse-free patients. Secondary end
`points included the frequency of relapses, change in DSS
`score from baseline and time to progression (defined as
`≥ 1 DSS unit increase maintained for ≥ 3 months).
`The proportion of relapse-free patients was significantly
`lower in the GA-treated cohort (14 of 25 [56%] GA versus
`6 of 23 [26%] placebo; p = 0.045). The overall 2-year relapse
`frequencies were 0.6 and 2.7 in the GA and placebo groups,
`respectively. Multiple regression analysis showed baseline DSS
`score significantly influenced relapse rate; a lower disability
`score at baseline increased the likelihood that a patient would
`remain free of exacerbations (p = 0.003). At the end of the
`2-year study, 84.6% of patients in the lower DSS stratum
`(0 – 2) taking GA were stable or improved versus 30% in the
`placebo group. Mean improvement was 0.5 DSS units with
`GA, whereas those taking placebo worsened by an average of
`1.2 DSS units (p = 0.012). In the higher DSS stratum (3 – 6),
`similar proportions of patients in each treatment group were
`stable, improved or worsened; on average these subjects wors-
`ened by 0.3 (GA) and 0.4 (placebo) DSS units.
`
`3.1.2 The US multi-centre study
`This study had three phases: a placebo-controlled, 24-month,
`double-blind treatment phase (core study); a blinded exten-
`sion phase (up to 36 months) that preserved the original treat-
`ment assignments; and an open-label extension phase in
`which patients who received placebo crossed-over to active
`treatment with GA whereas patients that received GA during
`the double-blind period continued to receive GA. The open-
`label extension phase is ongoing and now in its 12th year; data
`are available from the 6-, 8- and 10-year time points. The pri-
`mary end point (for all study phases) was the number of
`relapses. Secondary end points included the proportion of
`relapse-free patients, time to first relapse, change in Expanded
`Disability Status Scale (EDSS) score relative to baseline and
`proportion of patients with sustained disease progression
`(defined as ≥ 1 point increase in EDSS persisting for
`≥ 3 months). EDSS evaluations were made every 3 months
`during the blinded phase (up to 35 months) and every
`6 months thereafter. Patients were seen within 7 days of each
`
`Expert Opin. Pharmacother. (2004) 5(4)
`
`879
`
`Page 6 of 18
`
`

`
`Glatiramer acetate
`
`suspected relapse. When possible, the same neurologist and
`nurse coordinator completed the assessments of each patient.
`3.1.2.1 Core study phase
`The original study, conducted between 1991 and 1994
`enrolled 251 RR MS patients; 18 – 45 years of age (mean age:
`34 years) [46]. In order to be eligible, patients had to have
`≥ 2 relapses in the 2 years preceding study enrollment (mean
`2.9) and an EDSS score of ≤ 5 (mean 2.5). Patients were ran-
`domised to GA or placebo. Results at 24 months showed a
`relapse rate of 1.19 ± 0.13 for patients receiving GA and
`1.68 ± 0.13 for placebo patients, a 29% reduction in favour of
`GA (p = 0.007); annualised rates were 0.59 and 0.84, respec-
`tively. The proportions of patients who improved, were
`unchanged or worsened by ≥ 1 EDSS unit between baseline
`and the end of 2 years of treatment favoured GA (p = 0.037).
`At 24 months, 33.6 and 27.0% of GA- and placebo-treated
`patients, respectively, were relapse-free (p = 0.098). A post hoc
`analysis suggested that the therapeutic effect of GA may have
`been more pronounced in patients with lower EDSS scores
`(≤ 2 units) at baseline.
`3.1.2.2 Double-blind extension phase
`A total of 203 patients entered the blinded extension. By the
`end of this phase (∼ 35 months of treatment), there was a 32%
`reduction in mean relapse rate with GA (1.34 and 1.98 GA
`versus placebo, respectively; p = 0.002); annualised relapse
`rates were 0.58 and 0.81, respectively [47]. During the entire
`extended trial, 33.6 and 24.6% of GA- versus placebo-treated
`patients remained relapse-free, respectively (p = 0.035). No
`GA-treated patients who were relapse-free during the core
`24-month phase experienced a relapse during the extension
`period. The median time to first relapse was 287 days with GA
`and 198 days with placebo (p = 0.057). Subjects who received
`placebo were significantly more likely to experience multiple
`relapses (p = 0.008). The proportion of patients who improved
`by ≥ 1 EDSS units from entry favoured GA (27.2 and 12.0%
`GA versus placebo, respectively; p = 0.001). Worsening by
`≥ 1.5 EDSS units occurred in 21.6 and 41.6% of GA and pla-
`cebo-treated patients, respectively (p = 0.001). The mean
`EDSS score improved by -0.11 in the GA group and worsened
`by +0.34 in the placebo group (p = 0.006).
`3.1.2.3 Open-label extension phase
`Of the original 251 patients, 208 patients chose to continue
`on the open-label phase of the study. The annualised relapse
`rate of patients treated from the beginning of the study
`dropped each year. At ≥ 6 years of continuous treatment with
`GA since trial entry, 26 of 101 (25.7%) remained relapse free.
`The mean annualised relapse rate for those who received GA
`from randomisation was 0.42 (95% CI = 0.34 – 0.51), a 72%
`decrease compared with their prestudy rate, with a relapse rate
`of 0.23 during year 6 of treatment [48,49]. Of those treated with
`GA from study inception, 69.3% were either neurologically
`unchanged (within 0.5 EDSS units of baseline) or had
`improved by at least 1 unit on the EDSS [49].
`Data beyond 6 years has thus far been presented only in
`abstract form. At 8 years, 142 (56.6%) of the original patients
`
`remained in the study [50]. The annual relapse rate declined to
`0.2. The mean EDSS for the entire cohort was 3.14, which
`represents an increase of 0.5 units from randomisation. It is
`difficult to draw firm conclusions from this long-term cohort
`in the absence of a true placebo group and given the patient
`attrition that occurred over time. However, natural history
`studies of comparable patients would predict a higher level of
`neurological disability, with 50% of patients reaching an
`EDSS of 6 by year 12 following the onset of MS [51]. When
`the 72 patients always on GA were compared to those origi-
`nally randomised to placebo, there were still discernible disa-
`bility differences.
`Of the 208 patients who entered the open-label phase of
`the study, 133 began year 10 of treatment and 122 completed
`that year [52]; 64 were originally randomised to GA and 69 to
`placebo. Before randomisation, their annualised relapse rates
`were 1.52 and 1.46, respectively. By year 10, annual relapse
`rates were 0.22 and 0.23. The mean EDSS score for the group
`always on GA increased from randomisation by 0.9 EDSS
`units to 3.67. However, the majority (64.4%) remained stable
`or improved from randomisation. When patients who were
`switched from placebo to active treatment were compared to
`those continuously on GA, the proportion of patients having
`confirmed progression during the entire 10-year study period
`differed significantly (50 GA from entry versus 72 originally
`randomised to placebo; p = 0.015). These observations are
`consistent with the growing conviction that early and
`extended treatment offers the best outcomes in RR MS.
`
`3.1.3 Meta-analysis of the double-blind,
`placebo-controlled clinical trials
`A meta-analysis using pooled data from all 540 patients in
`three randomised, double-blind, placebo-controlled trials was
`recently published [53]. It was designed to investigate whether
`the treatment effect varied according to disease-related varia-
`bles at baseline. In all trials, patients had RR MS for at least
`1 year, at least 1 or 2 relapses during the 2 years prior to study
`entry and a baseline DSS or EDSS score ranging from 0 to 6.
`Inclusion criteria for one study included at least one gadolin-
`ium (Gd)-enhancing lesion on a MRI screening of the brain
`[54]. Durations of the placebo-controlled phases of the trials
`were 24 [45], 35 [46] and 9 [54] months. Regression models were
`developed to estimate the annualised relapse rate, total number
`of on-trial relapses and time to first relapse. Also explored and
`recently reported in poster form were the effect of GA on

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