throbber
Aois DRUG EVALUATION
`
`.$;';,*:+.;7;,;°,?.?;;:.g;§>;g.m
`© Adis International Urnlled. All rlgfls reserved.
`
`Glatiramer Acetate
`
`A Review of its Use in Relapsing-Remitting Multiple Sclerosis
`
`Dene Simpson, Stuart Noble and Caroline Perry
`
`Adis International Limited, Auckland, New Zealand
`
`Various sections of the manuscript reviewed by:
`R. Arnon, Department of Immunology, Weizmann Institute of Science, Rehovot, Israel; M. Pilippi,
`Department of Neuroscience, Scientific Institute Ospendale San Raffaele, Milan, Italy; M. S. Freedman,
`Multiple Sclerosis Research Clinic, The Ottawa Hospital, Ottawa, Canada; G. Giovannoni, Department of
`Neurochemistry, University of London, London, UK; K. P. Johnson, Department of Neurology, University
`of Maryland, Baltimore, Maryland, USA; D. Teitelbaum, Department of Immunology, Weizmann Institute
`of Science, Rehovot, Israel; B. Weinstock-Guttman, Baird Center for Multiple Sclerosis, The State University
`of New York, University at Buffalo, Buffalo, New York, USA.
`
`
`
`Contents
`
`.
`.
`Summary
`1.
`Introduction .
`
`.
`.
`
`.
`.
`
`.
`.
`
`.
`.
`
`.
`.
`
`.
`.
`
`.
`.
`
`.
`.
`
`.
`.
`
`.
`.
`
`.
`.
`
`.
`.
`
`.
`.
`
`.
`.
`
`.
`.
`
`.
`
`.
`.
`
`.
`.
`
`.
`.
`
`.
`.
`
`.
`.
`
`.
`.
`
`.
`.
`
`.
`.
`
`.
`.
`
`.
`.
`
`.
`.
`
`.
`.
`
`.
`.
`
`.
`.
`
`.
`.
`
`.
`.
`
`.
`.
`
`.
`.
`
`.
`.
`
`.
`.
`
`.
`.
`
`.
`.
`
`.
`.
`
`.
`.
`
`.
`.
`
`.
`.
`
`.
`.
`
`.
`.
`
`.
`.
`
`.
`.
`
`.
`.
`.
`
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`. 826
`. 829
`
`. 830
`. 830
`. 830
`. 831
`. 833
`. 833
`. 834
`. 834
`. 834
`. 834
`. 835
`. 835
`. 837
`. 839
`. 841
`. 841
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`2. Pharmacological Properties
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`2.1 Pathogenesis of Multiple Sclerosis
`.
`.
`.
`.
`.
`2.2 Proposed Mechanisms of Action of Glatiramer Acetate .
`2.2.1 Induction of G|atiramerAcetate-Specific SuppressorTCe||s .
`2.2.2 Inhibition of Myelin-Reactive T-Cell Responses .
`.
`.
`.
`.
`.
`.
`.
`.
`2.2.3 Potential for Neuroprotecfion .
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`2.3 Immunological Effects .
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`2.3.1 Non-Neutralisin Antibodies .
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`2.3.2 Selectivity .
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`2.4 Pharmacokinetic Properties .
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`3. Therapeutic Efficacy .
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`3.1 Effects on Relapse Rate .
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`3.2 Magnetic Resonance Imaging Assessments
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`3.3 Effects on Disability .
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`3.4 Other Effects
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`4. Pharmacoeconomics .
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`5. Tolerability .
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`
`.
`
`.
`
`.
`
`. 842
`
`AMNEAL
`
`EXHIBIT NO. 1029 Page 1
`
` AMNEAL
`
`

`
`826
`
`Simpson et a1.
`
`.
`.
`.
`.
`.
`Injection-Site Reactions
`5.1
`5.2 Post—|njection Systemic Reactions
`5.3 Other Effects
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`
`.
`
`.
`.
`.
`
`.
`.
`.
`
`.
`.
`.
`
`.
`.
`.
`
`.
`.
`.
`
`.
`.
`.
`
`.
`.
`.
`
`.
`.
`.
`
`.
`.
`.
`
`.
`.
`.
`
`.
`.
`.
`
`.
`.
`.
`
`.
`.
`.
`
`.
`.
`.
`
`.
`.
`.
`
`.
`.
`.
`
`.
`.
`.
`
`.
`.
`.
`
`.
`.
`.
`
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`6. Dosage and Administration .
`7. Place of Glatiramer Acetate in the Management of Relapsing-Remitting
`Multiple Sclerosis
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`
`.
`
`.
`
`.
`.
`.
`
`.
`
`.
`
`.
`.
`.
`
`.
`
`.
`
`.
`.
`.
`
`.
`
`.
`
`.
`.
`.
`
`.
`
`.
`
`.
`.
`.
`
`.
`
`.
`
`.
`.
`.
`
`.
`
`.
`
`.
`.
`.
`
`.
`
`.
`
`.
`.
`.
`
`.
`
`.
`
`.
`.
`.
`
`.
`
`.
`
`.
`.
`.
`
`.
`
`.
`
`.
`.
`.
`
`.
`
`.
`
`.
`.
`.
`
`.
`
`.
`
`. 843
`. 843
`. 844
`
`. 844
`
`. 844
`
`Summary
`
`Abstract
`
`Glatiramer acetate is a synthetic copolymer composed of a random mixture of
`four amino acids that modifies the immune response that results in the CNS
`inflammation, demyelination and axonal loss characteristic of relapsing-remitting
`multiple sclerosis (RRMS).
`In three randomised, double-blind trials in patients with RRMS, subcutaneous
`glatiramer acetate 20 mg/day was significantly more effective than placebo for
`the primary outcome measure of each trial (mean relapse rate, proportion of
`relapse-free patients and number of gadolinium-enhancing lesions on magnetic
`resonance imaging [MRI] scans). The mean relapse rate was significantly reduced
`at endpoint (approximately one-third less) in the two larger trials (the US pivotal
`trial [primary endpoint] and the European/Canadian study [tertiary endpoint]) in
`patients receiving glatiramer acetate compared with those receiving placebo. The
`rate was 78% less for glatiramer acetate than placebo patients in the pilot trial
`that investigated a slightly different patient population. Glatiramer acetate sig-
`nificantly decreased disease activity and burden of disease, as assessed in the
`European/Canadian study using a range of MRI measures. Patients with RRMS
`treated with glatiramer acetate in the US trial were significantly more likely to
`experience improved disability (whereas placebo recipients were more likely to
`experience worsening disability) and their overall disability status was signifi-
`cantly improved compared with placebo recipients. Data from the active—treatment
`extension of the US trial suggest that glatiramer acetate has sustained clinical
`benefits up to 8 years.
`
`Glatiramer acetate was generally well tolerated; the most commonly reported
`treatment-related adverse events were localised injection-site reactions and tran-
`sient post-injection systemic reactions. Both reactions were generally mild and
`self limiting but were responsible for the majority of withdrawals from treatment
`(up to 6.5 and 3.5%, respectively). Glatiramer acetate is not associated with the
`influenza-like syndrome or neutralising antibodies that are reported in patients
`treated with interferon-I3 for RRMS.
`
`The cost effectiveness of glatiramer acetate has yet to be definitively deter-
`mined as assessment of available data is confounded by very different models,
`data sources and assumptions.
`Conclusion. Glatiramer acetate has shown efficacy in well controlled clinical
`trials in patients with RRMS; it reduces relapse rate and decreases MRI-assessed
`disease activity and burden. It is generally well tolerated and is not associated
`with the influenza-like symptoms and formation of neutralising antibodies seen
`with the interferons—B. Based on available data and current management guide-
`lines, glatiramer acetate is a valuable first—line treatment option for patients with
`RRMS.
`
`© Adls International Umlred. All fights reserved.
`
`CNS Drugs 2[XD; lo (12)
`
`AMNEAL
`
`EXHIBIT NO. 1029 Page 2
`
` AMNEAL
`
`

`
`Glatiramer Acetate in Relapsing-Rernitting MS
`
`827
`
`Pharmacologlcal
`Propelfles
`
`Therapeuflc Efllcacy
`
`The proposed mechanism of action of glatiramer acetate in modulating the auto-
`immune response in relapsing-remitting multiple sclerosis (RRMS) consists of
`two components. The first is the induction of glatiramer acetate-specific suppres-
`sor T cells (i.e. type 2 helper T lymphocytes [Th2]) that are capable of directly
`and indirectly downregulating the inflammation in the CNS associated with mul-
`tiple sclerosis (MS). Human studies have shown that these glatiramer acetate-
`reactive T cells are initially and predominantly T111 type (pro-inflarrnnatory), but
`with exposure to glatiramer acetate there is a shift to a T112/T1,3-type response
`(anti-inflamrnatory). It is these glatiramer acetate-specific suppressor T cells, not
`glatiramer acetate itself, that may migrate into the CNS and downregulate the
`inflammation that is triggered by the antigenic products of demyelination (myelin
`basic protein [MBP] and other myelin antigens). In a small study involving pa-
`tients with RRMS, this shift from Th1- to Th2-type T cells induced by glatiramer
`acetate was accompanied by clinical benefits.
`The second feature of glatiramer acetate’s mechanism of action is the inhibi-
`tion of the autoreactive MBP- and other myelin antigen-specific T cells that would
`otherwise be stimulated to proliferate and release inflammatory cytokines. Cur-
`rent hypotheses include glatiramer acetate acting as an altered peptide ligand and
`engaging various T-cell receptor (TCR)s, and glatiramer acetate engaging the
`TCR and downregulating the MBP-specific T-cell response possibly by deliver-
`ing a non-activating signal (anergy).
`
`Recent research suggests that neuroprotection may be another mechanism of
`action accounting for the beneficial clinical effects of glatiramer acetate in RRMS.
`
`Antibodies stimulated by glatiramer acetate treatment are non—neutra1ising
`and do not affect the clinical efficacy of the drug.
`Few pharmacokinetic data are available for glatiramer acetate; following sub-
`cutaneous administration, the drug is rapidly degraded in the periphery, resulting
`in very low or undetectable serum concentrations. Glatiramer acetate is not re-
`quired to be present in the serum to exert its anti—inflammatory action but absorp-
`tion in proportion to the dose administered was rapid.
`
`Glatiramer acetate has shown efficacy in treating patients with RRMS. In three
`randomised, double-blind trials (including a 2-year pilot trial and the larger US
`pivotal [2-year] and European/Canadian [9-month] studies) glatiramer acetate
`20mg once daily, administered subcutaneously in patients with RRMS, was sig-
`nificantly more effective than placebo for the respective primary endpoint of each
`trial (proportion of relapse—free patients, relapse rate and number of enhancing
`lesions on magnetic resonance imaging [MRI] scans).
`
`For patients receiving glatiramer acetate compared with those receiving pla-
`cebo in the two larger comparative studies, the mean relapse rate (covariate ad-
`justed) at study endpoint was 29% lower in the large US trial (where relapse rate
`was the primary endpoint) and 33% lower in the European/Canadian study (where
`relapse rate was the tertiary endpoint). In the pilot trial, glatiramer acetate recip-
`ients had a mean relapse rate 78% lower, and they were more than twice as likely
`to be relapse free, than placebo recipients. Re1apse—related results in this pilot
`trial have not been reproduced in larger trials, possibly due to the patient popu-
`lation’s having a shorter duration of disease and a higher baseline relapse rate
`than those in subsequent studies.
`Glatiramer acetate decreased disease activity and burden of disease, as assessed
`by analysis of MRI scans, in patients enrolled in the European/Canadian study
`
`© Adls International Umlted. All fights reserved.
`
`CNS Drugs 2[XD; "IO (12)
`
`AMNEAL
`
`EXHIBIT NO. 1029 Page 3
`
` AMNEAL
`
`

`
`828
`
`Simpson et al.
`
`Pharmacoeconomlcs
`
`where certain MRI measures were the primary and secondary endpoints. For the
`primary outcome measure, patients in the glatiramer acetate—treated group dem-
`onstrated 29% fewer gadolinium-enhancing CNS lesions (areas of acute inflam-
`mation representing disruption of the blood-brain barrier) than patients in the
`placebo group. For secondary MRI outcomes, glatiramer acetate showed signif-
`icantly greater lesion reductions (ranging from 30 to 82.6%) than placebo. Al-
`though this 9-month trial period was considered too short to demonstrate a
`significant reduction in the volume of hypointense T1 lesions (representing areas
`of demyelination and axonal loss), further analysis of these scans has shown that,
`after 8 months, the proportion of new T2 lesions evolving into these hypointense
`T1 lesions (‘black holes’) in patients receiving glatiramer acetate was half that
`shown in patients receiving placebo.
`Progression to sustained disability, as measured by the Kurtzke Expanded
`Disability Status Scale (EDSS), was a secondary endpoint in the two long—term
`trials. Patients with RRMS treated with glatiramer acetate in the pivotal US trial
`were significantly more likely to experience improved disability, and placebo
`recipients were more likely to experience worsening disability. The overall dis-
`ability status was also significantly improved in this trial, although the change
`was modest. The pilot trial showed positive trends in delaying the onset or wors-
`ening of disability, although it did not have adequate statistical power to evaluate
`this outcome.
`
`Preliminary results data from the active—treatment extension of the US trial
`suggest that glatiramer acetate has sustained clinical benefits up to 8 years.
`
`Two studies conducted in 2000 and 2001 investigating the cost effectiveness of
`glatiramer acetate in the treatment of RRMS are difficult to compare as they used
`different models and data sources and led to different conclusions.
`
`According to a cost-utility analysis based on the clinical outcomes of a large
`placebo—controlled trial (the US pivotal trial) and its extensions, glatiramer ace-
`tate is cost effective compared with best supportive care alone for RRMS, from
`the perspective of the UK National Health Service. Cost-utility ratios improved
`with a longer duration of treatment for all three cost variables. At 8 years, cost
`per relapse avoided was £11 000, cost per disability unit avoided was £8 862 and
`cost per quality-adjusted life-year (QALY) gained was £22 586 (year 2000 costs).
`An analysis conducted by the National Institute of Clinical Excellence used
`longer term modelling and concluded that neither glatiramer acetate nor the
`interferons—[3 were cost effective in the treatment of RRMS. The best mean cost
`per QALY gained (i.e. at 20 years of treatment and including MS Research Trust
`data on quality of life), expressed as a range covering all the agents under inves-
`tigation, was between £35 000 and £104 000, which was more than the value
`considered favourable in the UK (£30 000).
`
`Given the complexities of cost-effectiveness assessments in RRMS, a lifelong,
`disabling disease (for which clinical benefits of long—term treatment have only
`recently been published) and the limited amount of information available at pres-
`ent, it is impossible to draw a single definitive conclusion regarding the cost
`effectiveness of glatiramer acetate, and further data and evaluations in this field
`would be useful.
`
`Tolerablllty
`
`Subcutaneously administered glatiramer acetate 20mg is generally well tolerated.
`The most commonly reported treatment-related adverse events (data from three
`placebo—controlled clinical trials and pooled results from two of these and other
`
`© Adls International Umlted. All fights reserved.
`
`CNS Drugs 2[XD; 1o (12)
`
`AMNEAL
`
`EXHIBIT NO. 1029 Page 4
`
` AMNEAL
`
`

`
`Glatiramer Acetate i.n Relapsing-Remitting MS
`
`829
`
`trials) are localised injection-site reactions and transient post-injection systemic
`reactions. The incidence of injection-site reactions (manifesting mainly as pain
`and erythema) was 64 and 73% with glatiramer acetate versus 37 and 38% with
`
`placebo (no p value reported).
`Post-injection systemic reactions occurred with an incidence of 10-38% with
`glatiramer acetate versus <1—13% with placebo (no p value reported) and mani-
`fested as one or more symptoms (facial flushing, chest tightness, dyspnoea, pal-
`pitations, tachycardia, anxiety and/or sweating) occurring within minutes of an
`injection and lasting for 30 seconds to 30 minutes.
`Both reactions were generally mild and self limiting but accounted for the
`majority of withdrawals from treatment. Overall withdrawal rates ranged from
`
`6—8% with localised injection—site reactions accounting for up to 6.5% and post-
`injection systemic reactions for up to 3.5% (vs 0.8% with placebo, no p value
`stated).
`
`Serious treatment-related events occurred in 52% of patients enrolled in clin-
`ical trials and, although no anaphylaxis was reported during the trials, three non-
`fatal cases of allergic reaction have since been recorded.
`Glatiramer acetate is not associated with the influenza-like syndrome reported
`in patients treated with interferon-B.
`
`Glatiramer acetate is indicated for the long-term management of RRMS and is
`currently approved in numerous countries worldwide including the US A, Canada,
`
`the UK and many other European countries. Glatiramer acetate is administered
`once daily by subcutaneous injection at a standard dose of 20mg. Data on the use
`of glatiramer acetate in pregnant and nursing women, the elderly, patients youn-
`ger than 18 years and those with impaired renal function are limited. Contraindi-
`cations include intravenous administration and hypersensitivity to glatiramer
`acetate or mannitol (which is included in the injection formulation).
`
`Dosage and
`Admlnlsttatlon
`
`I. Introduction
`
`Multiple sclerosis (MS) is a chronic, inflamma-
`
`tory disease of the CNS usually diagnosed in young
`adults (aged 20-40 years)“] and affecting an esti-
`mated 2.5 million people worldwide.[2] It is an auto-
`immune condition,[3'5] possibly triggered in genet-
`
`ically susceptible individuals by one or more agents
`in the environment,[5=7] that results in unchecked
`
`inflammation causing demyelination of areas in
`
`the brain and spinal cord. Relapsing-remitting MS
`
`fatigue and dizziness.[1°s”1 These relapses are in-
`terspersed with periods of complete or partial re-
`
`mission. Although some patients continue on this
`course without becoming seriously disabled,[5:7"2=131
`the majority (about 80%)[14] enter a phase within
`
`5-15 years in which they experience an increase in
`overall disability with or without relapses (second-
`ary progressive MS).[7=3-13-14] Within 10-15 years
`of a diagnosis of MS, 50% of patients are unable
`to walk unassisted,[6’151 and after 25 years 50% are
`wheelchair bound.[‘5]
`
`(RRMS) is the most common type of MS; approx-
`
`Management of RRMS includes multidiscipli-
`
`imately 85% of patients present with this type of
`MS.“'7~3] It manifests as self-limited attacks of neu-
`
`rological dysfunction (relapses)[91 during which
`
`the patient experiences a sudden worsening of neu-
`
`rological symptoms such as numbness, tingling,
`
`muscle weakness, spasticity, visual disturbances,
`
`nary rehabilitation, pharmacotherapy for symp-
`toms and treatment of relapses and, more recently,
`
`pharmacotherapy for modifying the underlying
`disease in an attempt to prevent relapses and delay
`the progression to disability.[5»3»12]
`Limited success (from an efficacy, tolerability
`
`© Adls International Umlted. All fights reserved.
`
`CNS Drugs 2[XD; 16 (12)
`
`AMNEAL
`
`EXHIBIT NO. 1029 Page 5
`
` AMNEAL
`
`

`
`Simpson et al.
`
`or cost perspective) with general immunosuppres-
`sant agents (azathioprine, mitoxantrone and oth-
`ers)[1~5"51 and intravenous immunoglobu1ins[3""”]
`(not approved in the USA) and progress in under-
`standing the pathogenesis of RMS have led to the
`introduction and use of more specific immuno-
`modulatory agents, such as glatiramer acetate (pre-
`viously copolymer-1) and two forms of recombi-
`nant interferon-|3,[3"51 for altering the natural
`course of the disease (see section 7).
`
`Glatiramer acetate is a synthetic copolymer
`
`composed of a random mixture of four amino acids
`that was initially developed to mimic myelin basic
`protein (MBP), one of the antigens thought to be
`involved in the pathogenesis of MSJ13] However,
`in a variety of animal species,[19’2°] it unexpectedly
`inhibited experimental autoimmune encephalitis
`(EAE), the primary animal model for MS. The sup-
`pressive and protective effects of the drug, in EAE
`induced by MBP and other myelin antigens (e.g.
`proteolipid protein [PLP] and myelin oligodendro-
`cyte glycoprotein [MOG]), are now well estab-
`lishedl“-19=2” In patients with RRMS, glatiramer
`acetate downregulates the immune response to these
`myelin antigens. It is considered to be the first
`agent to do this by engaging the T-cell receptor
`(TCR).[3’22] This article reviews pharmacological
`and clinical data on the use of glatiramer acetate in
`patients with RRMS.
`
`2. Pharmacological Properlies
`
`Glatiramer acetate consists of a mixture of four
`
`naturally occurring amino acids ( L-glutamic acid,
`L-alanine, L-tyrosine and L-lysine) produced by
`random polymerisation.[23’24] This agent simulates
`MBP, an antigen thought to be involved in MS,
`although both the pathogenesis of this disease and
`
`the mode of action of glatiramer acetate have yet
`to be fully elucidated. Nevertheless, the beneficial
`effects of glatiramer acetate in RRMS are thought
`to stem from its modification of immune processes
`implicated in the pathogenesis of the disease. This
`section focuses on the human studies that have at-
`
`tempted to characterise the pharmacological ef-
`fects and exact mechanism of action of glatiramer
`
`acetate and includes studies in patients with all
`types of MS. Animal data are provided where hu-
`man data are limited.
`
`2.1 Palhogenesis of Multiple Sclerosis
`
`The pathogenesis of MS involves a cascade of
`immunological events beginning with the activa-
`tion (by viral, environmental or other triggers)[6=7]
`of autoreactive myelin-reactive T cells in the pe-
`riphery. The subsequent release of inflammatory
`mediators and the upregulation of adhesion mole-
`cules[7~13] facilitate the passage of these T cells
`through an ‘altered’ blood-brain barrier into the
`CNS,[7’25v251 where they are reactivated by the anti-
`genic products of demyelination, predominantly
`MBP[3’7’27] but possibly PLP and MOG as well.
`These activated myelin-reactive T cells release
`
`cytokines and mediators, which recruit and stimu-
`late other inflammatory cells (microglia, astro-
`
`cytes and plasma cells) that precipitate the demy-
`elination, oligodendrocyte damage[9] and axonal loss
`characteristic of MS.[3’4=7-1236331
`
`2.2 Proposed Mechanisms of Action of
`Glaiira mer Acetate
`
`The disease-specific mechanism of action of
`glatiramer acetate in modulating the immune re-
`sponse in MS appears to be a complex process in-
`volving:
`
`0 induction of glatiramer acetate-specific sup-
`pressor T cells that are capable of directly and
`indirectly downregulating the inflammation in
`the CNS[21’22’26’27’29] (see section 2.2.1);
`
`0 inhibition of the autoreactive MBP-specific T
`cells that would otherwise be stimulated to pro-
`liferate and release inflammatory cytokines that
`lead to CNS inflammation and damage[27’3°]
`(see section 2.2.2).
`
`Although its eventual site of action is the
`CNSJ3” glatiramer acetate itself is unlikely to
`cross the blood-brain barrierlz‘-“Z1 as it is degraded
`into smaller fragments in the periphery after sub-
`cutaneous administration (see section 2.4).[33] It is
`
`the glatiramer acetate-induced T cells that migrate
`
`© Adls International Umlted. All I1gh1’s reserved.
`
`CNS Drugs 2[XD; 1o (12)
`
`AMNEAL
`
`EXHIBIT NO. 1029 Page 6
`
` AMNEAL
`
`

`
`Glatiramer Acetate i.n Relapsing-Remitting MS
`
`831
`
`into the CNS and downregulate the inflammation
`associated with MS (see sections 2.2.1 and 2.2.2).
`
`the proliferation of glatiramer acetate-specific T-
`
`cell lines. The glatiramer acetate-reactive T cells
`
`A summary of the key pharmacological effects
`of glatiramer acetate of potential relevance to its
`use in the treatment of RRMS is presented in table I.
`
`2.2. l Induction of Glatiramer Acetate-specific
`Suppressor T Cells
`
`Glatiramer acetate binds to the major histo-
`compatibility complex (MHC) class II molecule
`on the antigen-presenting cells[35’36] in the periph-
`ery at the injection sites or in the lymph nodes
`draining the injection sites.[291 The subsequent
`binding of this glatiramer acetate/MHC complex
`to the glatiramer acetate-specific TCRW91 induces
`
`secrete predominantly T-helper (T1,) 1-type pro-
`inflammatory cytokines,[41] but with exposure to
`
`glatiramer acetate the T cells are shifted more to-
`wards the T112 type (anti-inflammatory).[34~4°=‘”=53]
`
`These suppressor T cells may migrate through the
`blood-brain barrier into the CNS [251 Where they en-
`
`counter MBP and other pathogenic myelin antigens
`
`(products of CNS inflammation and demyelin-
`
`ation) presented by the MHC. Because glatiramer
`acetate is cross-reactive with these antigcns,[“°"”’43=47]
`
`the glatiramer acetate-activated T cells are reacti-
`
`vated to produce Th2 anti-inflammatory cytokines,
`
`Table I. Pharmacological effects of glatiramer acetate (GA) of relevance to its use in the treatment of relapsing-remitting mulfple sclerosis
`MS)"
`
`MHC binding in the periphery
`Binds to MHC with high alfinityl35l
`Promiscuous binding to multiple HLA-DR alleles,l3°l including DBFl1*1501 and DR4 haplotypes (linked to known MS susceptibility in
`cenain population groups)l37l
`Successfully corrpetes with MBP and other myelin antigens tor MHC binding [5'37‘393’
`
`Induction of suppressor T cells
`GA-reactive T cells shift from Ti-.1 type (pro-inflammatory) to Th2 and Th3 type (anti-inflamn1atory)[“”'i“*“"
`Shitt evident as early as 1 monthl“3' aftertreatment and sustained for up to 9 years,“ despite reduction in GA-reactive T-cell
`proliferation[‘‘*‘‘'‘’'‘51
`
`Inhibition of MBP-specific T-cell response
`Dose dependently inhbits proliferation of MBP-specific T cell<J27-3°]
`Downregulates the MBP-specific T-cell response by engaging the TCR and inducing anergy (acts as an altered peptide ligand and
`delivers a non-activating signaI)l2°-27'
`TCR antagonism demonstratedl“°] (inconsistent results in another studyl27])°
`
`T-cell migration through blood-brain barrier
`GA-induced Th2-type T cells accumulate in the CNS'‘’‘'’'
`
`Cross-reactivity
`Induces T cells cross—reactive with MBP,l“°-‘”“‘3v“71 MOGI5-4°‘ and PLPl2°~‘"1
`Cross—reactivity shown at the level of cytokine expression!”-4‘-“I
`More T-cell lines show cross-reactivity with continued GA treatment"‘°l
`
`Bystander suppression
`GA-induced suppressor T cells. reactivated by MBP. inhbit T-cell responses to other myelin antigenslz’-'2°'23-“i“-“5°] and downregulate
`turther inflammatory processes
`
`Neuroprotection
`Increased production of brain-derived neurotrophic factor detected in GA-specific T cells“ -521
`a All findings are from in vitro human studies except two studies by Aharoni et a|.l2°'3"
`b Unlikely to be relevant in vivo as GA and MBP remain in separate sites.
`c Unlikely to be relevant in vivo as MBP-specific T cells and GA remain in separate sites.
`HLA = human leucocyte antigen; MBP = myelin basic protein; MHC = major histocompatibility complex: MOG = myelin oligodendrocyte
`glyooprotein; PLP = proteolipid protein: TCFI = T-cell receptor: Tr. = T helper.
`
`© Adls Internctlonal Umlted. All rights reserved.
`
`CNS Drugs 2[XD; 1o (12)
`
`AMNEAL
`
`EXHIBIT NO. 1029 Page 7
`
` AMNEAL
`
`

`
`832
`
`Simpson et al.
`
`
`
`.GA Binding of GA to the MHC
`
`
`
`,,,h5bm°,, ofMBPT_ce"
`response lane-_r9v 0'
`TOR a"'a9°"'sm?)
`
`_
`_
`22:33:: f.-fceus
`
`S""”'°""Th‘ ‘°
`secretion
`
`CNS
`
`Migration of GA
`T °°"‘ ""°“9"
`blood-brain barrier
`
`GA T cells secrete
`
`Th2fT,.,3 cytokines in
`response to stimulation
`
`W MBP (°'°55"°3°“V“Y)
`
`
`
`
`
`inflammation in the CNS
`
`
`
`_Fi9-1-P'°P°5ed '"e°"a"lf5'“5°‘a_°f‘°"°‘9'?“'a"'°'a°‘%'3t°(GA)
`IN the treatmentofrelapslng-remitting multiple sclerosus.APc=
`antigen-presenting oell; MBP = myelin basic protein; MHC = major
`histocompatibility complex; TcFl =T-cell receptor; Tr. = T helper.
`
`which directly and indirectly downregulate the
`CNS-based inflammation associated with MS [2239]
`
`(figure 1). The ability of glatiramer acetate in the
`
`periphery to induce this remote suppression of in-
`
`flammation in the CNS is due to the cross-reactivity
`of glatiramer acetate-specific T cells with not only
`MBP,[26=27=29’4"431 but also other myclin antigens
`implicated in the pathogenesis of MS (possibly
`MOG[5~43] and PLP[2°"”1). This phenomenon, where-
`by the anti-inflammatory cytokines secreted are in-
`dependent of the primary autoantigen or antigen
`
`specificity of the T cells, is known as ‘bystander
`Suppression» .[22,26,28,4l ,44,so,s4]
`Evidence of a shift of T-cell generation from
`predominantly Thl-type (pro-inflammatory) to Th2-
`
`and Th3-type T cells (both anti-inflammatory) was
`
`accompanied by clinical benefits in a nonblind
`study that enrolled ten patients with RRMS treated
`with subcutaneous glatiramer acetate 20mg once
`daily for 12 months.[‘'‘‘1 There was a 2- to 6-fold
`increase in serum and peripheral blood lymphocyte
`(PBL) levels of indicators for the anti-inflamma-
`
`tory Th2- and Th3-type T cell at 3 or 6 months of
`treatment compared with pretreatment levels. At
`12 months the levels were higher than baseline val-
`ues.l44] Patients with active MS have been shown
`
`to have defective production and low levels of
`interleukin (IL)-10, IL-4 and transforming growth
`factor [3 (TGFB) [anti-inflarrnnatory cytokines].[“"']
`The mean serum level of IL-10 in patients treated
`with glatiramer acetate increased nearly 2-fold at
`
`3 months compared with baseline (from approxi-
`mately 1.4 to 2.6 ng/L, p = 0.04 [estimated from a
`graph]), although the increase was not significant
`
`at 6 and 12 months. Messenger RNA (mRNA) ex-
`pression of IL-4 and TGFB in the PBLs (estimated
`from graph) showed a 4- and 6-fold enhancement
`
`at 6 months (p = 0.03 vs baseline), and transform-
`
`ing growth factor-B remained significantly higher
`than baseline at 12 months (p = 0.02).[44]
`mRNA expression of the pro-inflammatory
`
`Thl-type cytokine tumour necrosis factor-oi in
`PBLs had decreased 78% from the pretreatment
`level at 12 months (estimated from a graph) [P <
`_
`0.004] .[44] A decrease from baseline was also 0b-
`
`© Adls International Umlted. All fights reserved.
`
`CNS Drugs 2[XD; 1o (12)
`
`AMNEAL
`
`EXHIBIT NO. 1029 Page 8
`
` AMNEAL
`
`

`
`Glatiramer Acetate i.n Relapsing-Remitting MS
`
`served at 12 months in the mean level of soluble
`
`IL-2 receptor; there was substantial interpatient
`variability in this reduction at all timepoints. The
`12-month decrease, which was not significant, was
`preceded by an increase at 3 months (p = 0.04 vs
`baseline).[‘‘‘‘]
`
`The shift from Th1- to Th2- and Th3-type T cells
`
`was accompanied by improvements in clinical
`markers of disease activity. There was a significant
`reduction in mean annual relapse rate (1.4 to 0.6,
`
`p = 0.001), and nine of ten patients had improved
`or stable disability, according to absolute changes
`in the Kurtzke Expanded Disability Status Scale
`(EDSS) score.[441
`There is recent evidence of a sustained shift in
`
`cytokine profile of the glatiramer acetate-induced
`T cellsm] in ten patients treated with glatiramer
`acetate for 6-9 years.[55] This shift, coupled with
`the cross-reactivity between glatiramer acetate and
`
`MBP and other myelin antigens, is consistent with
`the anti-inflammatory effects of glatiramer acetate
`and the phenomenon of ‘bystander suppression’
`(see above).
`
`The induction of glatiramer acetate-specific,
`protective T cells is detected early and the time
`required for the development of an adequate and
`effective T-cell population is reflected in the tim-
`ing of the onset of significant therapeutic action of
`glatiramer acetate (see section 3).[23v31'32’54]
`
`2.2.2 Inhibition of Myelin -Reactive
`T-Cell Responses
`
`The beneficial effects of glatiramer acetate are
`further explained by the inhibition of the autoreac-
`tive MBP- and other myelin antigen-specific T-cell
`
`responses implicated in the pathogenesis of MS
`(figure 1). Studies have shown that after initially
`inducing proliferation of the T cells, glatiramer ac-
`etate inhibits both their proliferation 97-301 and their
`secretion of IL-2 (a Th1 cytokine).[3°] It has been
`suggested that glatiramer acetate can alter the T
`cells in some way. Indeed, possibly acting as an
`altered peptide ligand, it engages various TCRs,
`altering the T-cell cytokine secretion to a Th2 pro-
`file. It may also downregulate the MBP-specific
`T-cell response by delivering a non-activating sig-
`
`nal (anergy)[2‘5=27] [figure 1]. TCR antagonism
`(competition for the TCR by the glatiramer acetate!
`MHC and MBP/MHC complexes), which is another
`mechanism proposed for this T-cell inhibition, is
`likely to remain an in vitro observation as glatira-
`
`mer acetate remains in the periphery and MBP in
`the CNS.[29~46]
`
`Glatiramer acetate might also interfere with the
`interaction between activated T cells and micro-
`
`glia in the CNS (which leads to the production of
`inflammatory cytokines), thereby accounting for
`the reduced inflammation in the CNS observed
`
`with glatiramer acetate treatment of RRMSJ56]
`
`2.2.3 Potential for Neuroprotecfion
`
`induced by treatment with
`Neuroprotection,
`glatiramer acetate, has been demonstrated in ani-
`malls" and in vitro human[51] studies. In experi-
`mental crush lesions of the optic nerve in a rat
`model, adoptive transfer of the no

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