throbber
View and review
`Arq Neuropsiquiatr 2011;69(3):536-543
`
`Classical immunomodulatory
`therapy in multiple sclerosis
`How it acts, how it works
`
`Amélia Mendes1, Maria José Sá1,2
`
`ABSTRACT
`Interferon beta (IFNβ) and glatiramer acetate (GA) were the first immunomodulators
`approved to the treatment of relapsing-remitting multiple sclerosis (MS) and clinically
`isolated syndromes. Despite the enlargement of the therapeutic armamentarium, IFNβ
`and GA remain the most widely drugs and the therapeutic mainstay of MS. Objective:
`To review the mechanisms of action of IFNβ and GA and main clinical results in MS.
`Results: IFNβ modulates T and B-cell activity and has effects on the blood-brain barrier.
`The well proved mechanism of GA is an immune deviation by inducing expression of
`anti-inflammatory cytokines. Some authors favor the neuroprotective role of both
`molecules. Clinical trials showed a 30% reduction on the annualized relapse rate and
`of T2 lesions on magnetic resonance. Conclusion: Although the precise mechanisms
`how IFNβ and GA achieve their therapeutics effects remain unclear, these drugs have
`recognized beneficial effects and possess good safety and tolerability profiles. The large
`clinical experience in treating MS patients with these drugs along almost two decades
`deserves to be emphasized, at a time where the appearance of drugs with more selective
`mechanisms of action, but potentially less safer, pave the way to a better selection of the
`most appropriate individualized treatment.
`Key words: multiple sclerosis, interferon beta, glatiramer acetate, immunomodulatory
`therapy.
`
`Terapêutica imunomoduladora clássica na esclerose múltipla: como atua, como
`funciona
`
`RESUMO
`O interferão beta (IFNβ) e o acetato de glatirâmero (GA) foram os primeiros
`imunomoduladores aprovados para o tratamento da esclerose múltipla (EM) surto-
`remissão e doentes com síndromes clinicamente isoladas. Apesar do alargamento
`do armamentário terapêutico, o IFNβ e o GA continuam a ser os medicamentos mais
`usados na EM. Objetivo: Rever os mecanismos de acção do IFNβ e do GA e os principais
`resultados na clínica. Resultados: O IFNβ modula a actividade das células T e B e tem
`efeitos sobre a barreira hemato-encefálica. O mecanismo melhor comprovado do GA é
`o desvio imune através da indução da expressão de citocinas. Alguns autores favorecem
`ainda um papel neuroprotetor para ambos. Os ensaios clínicos mostraram diminuição
`da taxa anualizada de surtos de 30% e das lesões em T2 na ressonância magnética.
`Conclusão: Embora os mecanismos pelos quais o IFNβ e o GA atingem os seus efeitos
`terapêuticos continuem a ser pouco claros, estes fármacos possuem efeitos benéficos
`reconhecidos e bons perfis de segurança e tolerabilidade. A grande experiência clínica
`no tratamento da EM com estes fármacos ao longo de quase duas décadas merece ser
`destacada, numa altura em que o aparecimento de novos fármacos com mecanismos de
`acção mais seletivos, mas potencialmente menos seguros, possibilitarão melhor seleção
`e individualização do tratamento.
`Palavras-chave: esclerose múltipla, interferão beta, acetato de glatirâmero, terapêutica
`imunomoduladora.
`
`1MD, Department of Neurology, Hospital de São João, Porto, Portugal; 2MD, PhD, Department of Neurology, Hospital de São
`João, Porto, Portugal; Health Sciences Faculty, University Fernando Pessoa, Porto, Portugal.
`
`Correspondence
`Amélia Mendes
`Department of Neurology
`Hospital de São João
`Alameda Prof. Hernâni Monteiro
`4200-319 Porto - Portugal
`E-mail: mendes.amelia@gmail.com
`
`Received 21 December 2010
`Received in final form 13 January 2011
`Accepted 20 January 2011
`
`536
`
`Page 1 of 8
`
`YEDA EXHIBIT NO. 2062
`MYLAN PHARM. v YEDA
`IPR2015-00644
`
`

`
`Arq Neuropsiquiatr 2011;69(3)
`
`Multiple sclerosis (MS), the most frequent primary
`demyelinating pathology of the central nervous system
`(CNS), is a chronic and progressive autoimmune disease
`characterized by inflammation, demyelination and axonal
`injury1. The etiology of MS is ultimately unknown, al-
`though there is evidence that complex multifactorial fac-
`tors are implicated, in which environmental are hypothe-
`sized to interact with genetically susceptible individuals2.
`The clinical hallmarks of MS may be summarized
`as follows1: the disease typically begins in young adults
`and affects females more than males (1.77:1); most
`commonly, MS patients alternate relapses with remis-
`sion phases (relapsing-remitting MS or RRMS), some of
`them developing later on a secondary progressive course
`(SPMS), and in a fewer cases, the disease progresses ab
`initio without (progressive MS or PPMS) or with rare
`superimposed relapses (transitional or progressive re-
`lapsing MS-RPMS); the disease is heterogeneous as re-
`gards neurological manifestations, evolution and dis-
`ability; the diagnosis, based in international consensual
`criteria, depends strictly on clinical features and paraclin-
`ical exams, the most important of which is the magnetic
`resonance imaging (MRI); these criteria turned feasible
`the identification of patients with a clinically isolated de-
`myelinating event or syndrome (CIS) that are at risk of
`conversion to a clinically definite disease (CDMS); finally,
`the progressive course and consequent neurological def-
`icits inflict a significant disabling condition to the patient
`and a major burden to relatives, caregivers and society.
`Although on the grounds of non-curative approaches,
`since the early nineties several pharmacological treat-
`ments with immunomodulatory properties were devel-
`oped to treat MS and modify its natural history, com-
`monly designated “disease modifying drugs” (DMD),
`which recognizably represented a major step in the con-
`trol of the disease.
`In this practical review we will focus on the classical
`immunomodulators specifically approved in MS - inter-
`feron beta (IFNβ) and glatiramer acetate (GA) - high-
`lighting their mechanisms of action (how they act) and
`their main clinical and imaging effects (how they work),
`based on the results of pivotal and comparative clinical
`trials. Despite the fast enlargement of the therapeutic ar-
`mamentarium for MS in the last years, with the approval
`of drugs with better efficacy yet potential limiting ad-
`verse effects, as mitoxantrone and natalizumab (usually
`indicated in more severe non-IFNβ-responder cases),
`and the development of oral drugs, exemplified by the
`recently FDA approved fingolimod, IFNβ and GA remain
`up to now the worldwide therapeutic mainstay of MS.
`
`INTERFERON BETA
`Interferons (IFNs) are proteins secreted by cells and
`
`MS: immunomodulatory therapy
`Mendes and Sá
`
`are involved in self defense to viral infections, in the reg-
`ulation of cell growth and in the modulation of immune
`responses. Human IFNβ is a glycoprotein primarily pro-
`duced by fibroblasts with 166 amino acids and 22.5 kDa,
`which is encoded on chromosome 9 without introns3.
`IFNβ was the first therapy to have proved beneficial ef-
`fects on the natural course of MS and has two molecules:
`IFNβ-1a and -1b.
`IFNβ-1a is obtained by eukaryote cell lines derived
`from a Chinese hamster ovary and, similarly to native
`human beta interferon, is glycosilated and has the com-
`plete 166 amino acid sequence; yet, the glycosylation pat-
`tern is not necessarily equal to the human3. IFNβ-1b is
`a product of a bacterial (E. coli) cell line and is not gly-
`cosilated because bacteria do not glycosylate proteins;
`additionally the cystein residue has been substituted
`by a serine at position 17, which prevents incorrect di-
`sulphide bond formation and minimizes the risk of im-
`paired folding of the molecules and the consequent re-
`duced activity; also, the methionine at position 1 has
`been deleted, so the final protein has one less amino
`acid than the natural IFNβ3. Glycosylation decreases
`aggregates formation and immunogenicity, which may
`give a lower potency of IFNβ-1a4, but, on the other side,
`IFNβ-1b has a tight binding to human serum albumin,
`which may contribute to about 10% of IFNβ-1a potency3.
`
`How it acts
`IFNβ binds to a high-affinity type-1 IFN transmem-
`brane receptors and induces a cascade of signaling path-
`ways. After binding to the receptor, phosphorylation and
`activation of two cytoplasmic tyrosine kinases occur.
`This leads to activation of latent transcription factors in
`cell cytoplasm that translocate to the nucleus5. IFNβ has
`a role in the immune system by producing effects on T
`and B cells, and, additionally has influence in blood brain
`barrier (BBB) permeability6.
`
`EFFECTS ON T CELLS
`T cell activation – IFNβ is believed to reduce T cells
`activation, including myelin reactive T cells, because in-
`terferes with antigen processing and presentation by
`downregulating expression of major histocompatibility
`complex (MHC) class II, and reduces the levels of co-
`stimulatory molecules7 and other accessory molecules
`like intercellular cell adhesion molecule-1 (ICAM-1),
`vascular cell adhesion molecule-1 (VCAM-1), and very
`late activation antigen-4 (VLA-4)8.
`T cell differentiation and proliferation – IFNβ
`inhibits the expansion of T cell clones, acting as an
`anti-proliferative agent. The exact mechanism for this
`anti-proliferative effect is unclear. Recently, it was dem-
`onstrated that type I IFNs, in which IFNβ is included,
`
` 537
`
`Page 2 of 8
`
`YEDA EXHIBIT NO. 2062
`MYLAN PHARM. v YEDA
`IPR2015-00644
`
`

`
`MS: immunomodulatory therapy
`Mendes and Sá
`
`could activate the Mnk/eIF4E kinase pathway that plays
`important roles in mRNA translation for IFN-stimu-
`lated genes and generation of IFN-inducible anti-prolif-
`erative responses9. Previous studies have indicated that
`Th17 cells have a critical role in the development of the
`autoimmune response in MS10. IFNβ-1a could induce an
`up-regulation of the TLR (toll-like receptors)-7 signaling
`pathway and inhibit multiple cytokines involved in Th17
`cell differentiation. The authors propose that the exog-
`enously administered high-dose IFNβ-1a augments this
`naturally occurring regulatory mechanism and provides
`a therapeutic effect in patients with RRMS11. Further-
`more, IFNβ inhibits the expression of FLIP, an anti-apop-
`totic protein, leading to an increased incidence of T cells
`death12 and restores T-regulatory cell activity6.
`
`EFFECTS ON CYTOKINES AND CHEMOKINES
`It has been postulated that the modulation of the im-
`mune response by IFNβ may involve an immune devia-
`tion, consisting in a reduction of the expression of Th1
`induced cytokines while enhancing Th2 responses6. Ad-
`ditionally IFNβ has effects on chemokines: it could me-
`diate activity of the chemokine receptor CCR7 which is
`important to direct the entry of T lymphocytes to the
`peripheral lymph nodes rather than to the CNS6. An-
`other chemokine, Regulated on Activation, Normal T
`Expressed and Secreted (RANTES), appears to play a
`role in the pathogenesis of RRMS and was observed a de-
`crease of its sera and peripheral blood adherent mono-
`nuclear cell levels triggered by IFNβ-1b13. A recent study
`suggested that peripheral upregulation of the chemo-
`kines by IFNβ may reduce the chemoattraction of im-
`mune cells to the CNS14.
`
`ANTIGEN PRESENTATION
`Furthermore, IFNβ is postulated to inhibit antigen
`presentation to T cells in conjunction with MHC and
`co-stimulatory molecules as CD80 and CD86, which is a
`crucial event in the ensuing immune response8. Another
`mechanism by which IFNβ can affect antigen presenta-
`tion is by counteracting the effect of IFNγ, because the
`latter cytokine is a potent promoter of MHC class II ex-
`pression on many cell types8.
`
`EFFECTS ON B CELLS
`IFNβ upregulates a B-cell survival factor (BAAF) and
`for those patients in whom B cells play a major impor-
`tant role, this would be a quite undesirable consequence
`of IFNβ therapy. This might partially explain inter-indi-
`vidual differences in the therapeutic response. Other-
`wise, the systemic induction of BAFF by IFNβ therapy
`might facilitate the occurrence of various autoantibodies
`and IFN neutralizing antibodies (NAbs). The authors
`
`538
`
`Arq Neuropsiquiatr 2011;69(3)
`
`conclude that individual MS patients with evidence for
`a significant role of B cells do not appear to be ideal can-
`didates for IFNβ therapy15. However, B cells may trigger
`neurotrophic cytokines that exert positive effects on MS
`autoimmunity, which could outweigh the negative effects
`of IFNβ-induced BAAF responses6.
`
`EFFECTS ON BBB
`IFNβ is able to inhibit the ability of T cells to get
`into the brain by interfering with the expression of sev-
`eral molecules. It was demonstrated that matrix metal-
`loproteinase type 9 (MMP-9) activity can be decreased
`by IFNβ-1b treatment in vitro16, which could difficult the
`migration of lymphocytes across the fibronectin of ce-
`rebral endothelium. Another study did not find any dif-
`ference in the MMP-9 levels during the treatment with
`IFNβ17. Besides the role of the metalloproteinases, IFNβ
`can modulate the expression and traffic of other mol-
`ecules like cytokines, chemokines, adhesion molecules
`and integrins16-18, improving endothelial barrier function
`and prevent the transmigration of leukocytes and other
`neurotoxic mediators across the BBB to sites of CNS
`inflammation10. One example is the possible induction
`of an increase in CD73 expression. Additionally, Uhm
`and colleagues found that the decrease in cell migration
`seems to wane with time as patients who have been re-
`ceiving IFNβ-1b treatment for more than 3.5 years had
`high levels of T-cell migration that were indistinguishable
`from those of MS patients who have never been treated
`with IFNβ19. IFNβ may interfere with T-cell/endothelial
`cell adhesion by inhibiting MHC class II expression on
`endothelial cells, which can also function as ligands for T
`cells20 and by decreasing the expression of VLA-48. IFNβ
`also increases serum concentrations of soluble VCAM1
`(sVCAM1), which might block leukocyte adhesion to ac-
`tivated cerebral endothelium by binding competitively
`with the VLA-4 receptor18. sVCAM1 had been correlated
`with a reduction in the number of MRI gadolinium-en-
`hancing lesions soon after the initiation of treatment21.
`
`ANTIVIRAL EFFECTS
`Both formulations of IFNβ have antiviral properties,
`although IFNβ-1a seems to be more potent in this field6.
`A group of investigators studied the relation of MS-as-
`sociated retrovirus (MSRV) in MS patients treated with
`IFNβ. They found that the viral load in the blood was
`directly related to MS duration and fell below detec-
`tion limits within 3 months of IFN therapy, suggesting
`that evaluation of plasmatic MSRV could be considered
`a prognostic marker for the individual patient to mon-
`itor disease progression and therapy outcome22. Another
`group aimed to analyze IFNβ antiviral efficiency through
`the measurement of human herpesvirus-6 (HHV-6)
`
`Page 3 of 8
`
`YEDA EXHIBIT NO. 2062
`MYLAN PHARM. v YEDA
`IPR2015-00644
`
`

`
`Arq Neuropsiquiatr 2011;69(3)
`
`prevalence in MS patients and they noted a decreased
`number of reactivations of the virus associated with less
`relapses (42.8% of patients with viral reactivations ex-
`perienced at least one relapse versus 22.5% of patients
`without viral reactivations)23.
`
`NEUROPROTECTIVE EFFECTS
`Some studies issued potential neuroprotective effects
`of IFNβ, inducing release of nerve growth factor from
`astrocytes or stimulate the protection of neurons them-
`selves6.
`Other investigators tried to measure the axonal in-
`jury in vivo using MRI spectroscopy to quantify the neu-
`ronal marker, N-acetylaspartate (NAA) and its relation
`with creatine (Cr) and found an increase in NAA/Cr in
`IFNβ-1b treated MS patients. Their data suggest that the
`axonal injury could be partially reversible with IFNβ-1b
`therapy24.
`
`How it works
`CLINICAL AND MRI OUTCOMES
`The first multicenter, randomized and placebo-con-
`trolled study in RRMS patients with IFNβ was pub-
`lished in 199325. This pivotal study demonstrated that
`IFNβ-1b 250 µg subcutaneous (SC) produced a 34% re-
`duction in the clinical relapse rate and in the confirmed
`1-point EDSS progression rate after 2 years, better than
`a lower dose (50 µg), yet the latter was not statistically
`significant compared with the placebo group25. Further-
`more, the number and frequency of T2 active lesions
`on brain MRI were decreased26. Three years later, the
`results of a phase III trial with a similar design using
`IFNβ-1a 30 µg/week intramuscular (IM) showed a 37%
`reduction in the confirmed 1-point EDSS progres-
`sion rate. The median number of MRI-gadolinium en-
`hancing lesions in MS was 33% inferior comparatively
`to the placebo arm. This pivotal trial also showed that
`IFNβ-1a slowed the accumulation of disability27. Since
`then, several trials confirmed these beneficial effects
`in RR form of MS28,29 and in secondary progressive
`with relapses30. The patients with CIS who are consid-
`ered with a high risk of CDMS have a proven benefit
`from early treatment with IFNβ to decrease clinical and
`MRI disease activity, as shown by specific studies con-
`ducted in CIS, either with IFNβ-1a31 or with IFNβ-1b32.
`As regards the route of administration, it does not
`seem to influence the biological effects of the IFNβ for-
`mulations33. Similarly, a dose-dependent effect remains a
`controversial issue. Although the pivotal trials suggested
`a dose-response curve, i.e., clinical and MRI outcomes
`seem to be better with higher doses, the evidence pro-
`vided by them was considered somewhat equivocal34.
`However, other studies pointed out a trend to the same
`
`MS: immunomodulatory therapy
`Mendes and Sá
`
`result, in which higher dose and more frequently admin-
`istered IFNβ was favored35,36, findings that were not cor-
`roborated by others37.
`
`NEUTRALIZING ANTIBODIES (NABS)
`During treatment with IFNβ, a proportion of MS pa-
`tients develop NAbs. The potential impact of NAbs on
`the efficacy of IFN-β treatment in MS is an area of de-
`bate and controversy, although their presence has been
`associated with a significant hampering of the treatment
`effect on the relapse rate and both active lesions and
`burden of disease in MRI. In Europe it is recommended
`that the patients treated with IFNβ are tested for the
`presence of NAbs at 12 and 24 months of therapy. In pa-
`tients with NAbs, the measurement should be repeated
`at intervals of 3-6 months and if the titers continue el-
`evated, IFNβ might be discontinued38. The American
`Academy of Neurology did not find enough evidence
`to make specific recommendations about when to test,
`which test to use, how many tests are necessary, and
`which cutoff titer to apply39.
`
`Side effects
`Therapy with IFNβ is usually well tolerated. The most
`frequent side effects are flu-like symptoms and injection-
`site reaction, which tend to reduce over time. Depres-
`sion, allergic reaction, haematologic and liver function
`abnormalities might also be observed40. IFNβ is a safe
`treatment, but usually is not recommended during preg-
`nancy because of the higher risk of fetal loss and low
`birth weight41.
`
`IFN formulations and indications
`The actual commercially available formulations of
`IFNβ include IFNβ-1a and IFNβ-1b. IFNβ-1a is dosed
`in 30 µg (Avonex®), 22 or 44 µg (Rebif®). The first is ap-
`plied once a week by IM and the second three times a
`week with a SC injection. IFNβ-1b formulations have 250
`µg (Betaferon® or Betaseron®, and Extavia®) and are ad-
`ministered by SC injection every other day. All formula-
`tions are indicated in RRMS, IFNβ-1a IM and IFNβ-1b
`are also approved in patients with CIS at risk of conver-
`sion to CDMS and IFNβ-1b is furthermore approved in
`Europe to treat patients with SPMS still with relapses.
`
`GLATIRAMER ACETATE
`Glatiramer acetate is a synthetic polypeptide com-
`posed of four amino acids (L-glutamic acid, L- lysine, L-
`alanine and L- tyrosine) with an average molecular mass
`of 4700-11.000 Da. It was discovered in the 1960’s, when
`studies to develop a polymer resembling myelin basic
`protein (MBP), a major component of myelin sheath, to
`the model of autoimmune encephalomyelitis (EAE), were
`
` 539
`
`Page 4 of 8
`
`YEDA EXHIBIT NO. 2062
`MYLAN PHARM. v YEDA
`IPR2015-00644
`
`

`
`MS: immunomodulatory therapy
`Mendes and Sá
`
`performed42. One of them, called copolymer 1, demon-
`strated to decrease or prevent EAE, and was later re-
`named as GA43.
`
`How it acts
`Several mechanisms of action have been proposed,
`yet the precise biological effects of GA are not fully un-
`derstood. We present the main effects on T and B lym-
`phocytes and on antigen presenting cells (APCs).
`
`EFFECTS ON T CELLS
`Inhibition of myelin reactive T cells and immune
`deviation – GA binds directly to MHC class II, but also
`seems to be able to interact with MHC class I44. GA in-
`terferes with the activation of myelin-specific T cells
`based on the observation that it acts as an antagonist to
`MBP/MHC at MBP-specific T cell receptor (TCR), op-
`erating as an altered peptide ligand to the 82-100 epitope
`of MBP in vitro42, displacing MBP from the binding site
`on MHC II molecules. Some authors argued that this
`“TCR antagonism” is controversial and, whether it oc-
`curs, is not probably relevant in vivo because GA is un-
`likely to reach sites where it could compete with MBP.
`However, GA-reactive Th2 cells are able to cross the
`BBB and might be activated not only by MBP, but also
`by other cross-reactive antigens44. Myelin reactive T cells
`exposed to increasing doses of GA manifest dose-de-
`pendent inhibition of proliferation and IFNγ production.
`That proliferative response of T cells to GA decreases
`with time. In addition, the observed decrease in GA-re-
`active T cells could be caused by the induction of T cell
`anergy and clonal elimination45. This mechanism of T cell
`anergy can occur in the periphery at the injections sites
`or in their draining lymph nodes where the MBP specific
`cells might be confronted with GA. The used regimen of
`daily SC administration may favor the induction of an-
`ergy rather than a full immunization that requires longer
`intervals between doses46. However, some clonal popula-
`tions of T cells could be expanded, since GA induced the
`conversion of peripheral CD4+CD25– to CD4+CD25+
`regulatory T cells through the activation of transcrip-
`tion factor Foxp3 and lead to proliferation of these cells.
`However, this fact must be interpreted with caution be-
`cause almost all activated human T cells express Foxp342.
`Therapy with GA may improve the immune regulatory
`function of CD8+ T cells42. These data suggest that the
`immunomodulatory effect of GA is attributed to the in-
`duction of a cytokine secretion pattern deviation from
`Th1 to Th2 cytokines, as happens with IFNβ43, which is
`the mechanism with the strongest experimental support.
`
`BYSTANDER SUPPRESSION
`Another potential mechanism of action is the so
`
`540
`
`Arq Neuropsiquiatr 2011;69(3)
`
`called bystander suppression: a phenomenon of T cells
`specific to one antigen which suppress the immunolog-
`ical response induced by another antigen46. This implies
`that GA-reactive Th2 cells are capable of entering the
`CNS and recognizing cross-reactive antigen(s), probably
`myelin antigen(s)44. It is characterized by the secretion
`of anti-inflammatory cytokines by GA-activated T cells
`after they cross the BBB and accumulate in the CNS43.
`
`EFFECTS ON CELL-PRESENTING ANTIGENS
`Although the vast majority of evidence suggests that
`GA acts primarily at the level of T cells, additional ef-
`fects on other immune cells cannot be excluded. For ex-
`ample, GA was reported to inhibit a human monocytic
`cell line, THP-1. In THP-1 cells stimulated with lipopoly-
`saccharide or IFN-γ, GA reduced the percentage of cells
`expressing MHC-DR and DQ antigen and inhibited the
`production of TNF-α and cathepsin-B. In contrast, the
`production of interleukin(IL)-1β was increased47. This
`could also indicate antigen-unspecific modes of action. A
`further study also demonstrated that GA affects mono-
`cytes/macrophages by inducing the production of an
`anti-inflammatory cytokine, the IL-1 receptor antago-
`nist (IL-1Ra), but diminishing the production of IL-1β in
`monocytes, activated by direct contact with stimulated T
`cells in MS patients and in the EAE model48. IL-1Ra can
`be transported through the BBB and exert its immuno-
`modulatory effects in both systemic and CNS compart-
`ments. In addition to the modulation of the adaptive im-
`mune system, GA seems to affect significantly the innate
`immune system48.
`GA may also affect the immune response through
`modifying APCs into anti-inflammatory type II cells. The
`process begins with the presentation of GA to CD8+ and
`CD4+ T cells by APCs. The final step is an alteration of
`cytokine environment that subsequently affect T-cell dif-
`ferentiation as far as concerned to further cytokine se-
`cretion. The T cell CD8+ response becomes oligoclonal
`with expansion and maintenance of CD8+ clone popu-
`lation over long periods of time, in contrast to what hap-
`pens to T cells CD4+ which may increase in number42.
`
`NEUROPROTECTIVE EFFECTS
`Futhermore, GA specific T cells secrete neurotrophic
`factors as brain-derived neurotrophic factor and neuro-
`trophic growth factor, which might favor remyelination
`and axonal protection42,43,49. A study with MRI spectros-
`copy showed a significant increase in NAA/Cr in a group
`of treatment naïve patients with RRMS, who received
`GA compared with untreated patients, suggesting the
`potential role of GA in axonal metabolic recovery and
`protection from sublethal injury50. Another potential ef-
`fect of GA is the delivery of neuroprotective cytokines
`
`Page 5 of 8
`
`YEDA EXHIBIT NO. 2062
`MYLAN PHARM. v YEDA
`IPR2015-00644
`
`

`
`Arq Neuropsiquiatr 2011;69(3)
`
`to the site of inflammation in patients with MS. So, the
`role of GA seems to be the creation of an anti-inflamm-
`matory and neuroprotective environment instead of sup-
`pression the immune activity42.
`
`How it works
`CLINICAL AND MRI OUTCOMES
`The first studies on MS focusing treatment with co-
`polymer 1 were carried out in late 1970s and early 1980s.
`Ten years later, a phase III multicentre, double blind and
`placebo-controlled trial, performed in patients with
`RRMS, showed that 20 mg GA SC daily was effective in
`reducing the annualized relapse rate (ARR) by 29% over
`a 2-year period compared with the placebo51. It also re-
`duced the disability progression in 12%, although this
`change was not statistically significant51. After 10 years
`of open label extension of this pivotal trial, patients orig-
`inally randomized to GA were shown to maintain better
`outcomes than patients who were originally on placebo52,
`although the high dropout rate raised some concerns
`about the power of the study.
`As the initial phase III trial did not include MRI end-
`points, a European/Canadian study was undertaken to
`address this specific issue in MS patients treated with GA
`versus placebo during 9 months53. It was demonstrated a
`reduction in the frequency and volume of new enhancing
`lesions, such as a 35% and 8.3% decrease in the number
`of enhancing lesions and in the median change in T2
`burden of disease, respectively, for the treatment arm,
`an effect that was delayed until 6 months after initia-
`tion of treatment53. Later on, in various studies, ARR re-
`ductions with use of GA in RRMS patients were found
`to be much higher than those seen in its pivotal trial51.
`Recently, the effect of GA on delaying conversion of pa-
`tients presenting with CIS to CDMS was evaluated in
`the PreCISe study, which showed that GA has a benefi-
`cial effect for the treatment of patients with this condi-
`tion54. On the contrary, a large controlled trial with GA
`in PPMS failed to provide any evidence for benefit in this
`population55.
`
`Side effects
`The results of the studies indicate that GA is gener-
`ally safe. The most common adverse reaction is a local
`reaction in the site of injection with erythema and indu-
`ration. GA is less frequently associated with a transient
`post-injection systemic reaction of flushing, chest tight-
`ness, dyspnea, chest palpitations, and anxiety. This self-
`limited systemic reaction may be experienced in 15% of
`the patients and typically resolve within 15-30 minutes
`without sequelae. No significant laboratory abnormal-
`ities have been found. According to the manufacturer,
`rare cases of non-fatal anaphylaxis have also been re-
`
`MS: immunomodulatory therapy
`Mendes and Sá
`
`ported49. Opportunistic infections, malignancies, and the
`development of autoimmune diseases are not risks asso-
`ciated with GA52. Although its use is not recommended
`in pregnancy, there is no evidence to suggest increased
`risk of adverse fetal or pregnancy outcome49,56.
`
`GA formulation and indications
`Glatiramer acetate (Copaxone®) is approved in a SC
`formulation of 20 mg to be administered once a day, to
`treat patients with RRMS and with CIS at risk of con-
`version to CDMS.
`
`Comparative studies
`Recently, the results from three head-to-head trials
`(IFNβ and GA) were published and they did not find
`significant differences between the two molecules in
`the primary endpoints evaluating reduction in relapse
`rates40,57,58. The REGARD study, a randomized, compar-
`ative, parallel-group, open-label trial, compared 44 µg of
`IFNβ-1a SC 3 times a week with 20 mg of GA SC once
`a day for 96 weeks. There was no significant difference
`between groups in the time to first relapse and ARR. Re-
`garding MRI outcomes, no significant differences were
`found in the number and change in volume of T2 ac-
`tive lesions. Patients treated with IFNβ-1a SC had sig-
`nificantly fewer gadolinium enhancing lesions and pa-
`tients treated with GA experienced significantly less
`brain atrophy57. BEYOND study compared 3 groups for
`treatment-naïve early stages RRMS patients: 250 µg of
`IFNβ-1b, 500 µg IFNβ-1b, both SC dosed every other
`day and GA 20 mg SC daily over 2 years. No significant
`differences were found in time to first relapse, overall re-
`lapse rates and proportion of patients who remained re-
`lapse free during the study period. No differences were
`found in T1-hypointense lesion volume change among
`the groups when compared the baseline with the last
`MRI available or annual time points. Change in total
`MRI burden and T2 lesion volume was significantly
`lower in the patients in both IFNβ-1b compared with
`the patients who received GA. However, the differences
`in T2 lesion volume were noted during the first year but
`not in years 2 and 3. The overall median change in brain
`volume was similar in each group. MRI parameters did
`not differ between patients in either IFNβ-1b doses40.
`The BECOME study was conducted to determine the ef-
`ficacy of treatment with IFNβ-1b 250 µg SC every other
`day versus GA 20 mg SC daily in RRMS or CIS patients,
`evaluating MRI outcomes (total number of contrast-en-
`hancing lesions plus new non-enhancing lesions on long
`repetition time scans). The results were similar, as there
`were no significant differences in the effects of the med-
`ications on relapse rates58.
`Therefore, IFNβ and GA are both good options to
`
` 541
`
`Page 6 of 8
`
`YEDA EXHIBIT NO. 2062
`MYLAN PHARM. v YEDA
`IPR2015-00644
`
`

`
`MS: immunomodulatory therapy
`Mendes and Sá
`
`modify the natural course of MS. The choice between
`them is usually a challenging issue in MS Clinics, which in
`our view must be centered on the patient informed deci-
`sion, after a thorough education about the disease and the
`real therapeutic expectations. However, the administra-
`tion routes are rather bothersome to the patients, which
`could contribute to a reduced therapeutic adherence59.
`Pivotal studies of IFNβ and GA in MS demonstrated
`that they are efficacious, lowering the ARR in approxi-
`mately 30%, the lesion burden and their activity, as well
`as the brain atrophy as measured by MRI.
`Even though the mechanisms of action of these clas-
`sical immunomodulatory drugs are not completely un-
`derstood, there is sound evidence that they act on impor-
`tant steps of the inflammatory processes underpinning
`MS. The appearance of drugs with more specific targets,
`as monoclonals and orals, increasing therapeutic efficacy,
`albeit raising new safety and tolerability problems, as well
`as a better understanding of the immunogenetic profiles
`of MS patients, are altogether expected to permit a more
`advanced therapeutic choice in the future. Actually, IFNβ
`and GA are the better known DMD in MS, with proofs
`of their safety and tolerability, so the large clinical expe-
`rience in treating MS patients with them along almost
`two decades, deserves to be emphasized.
`
`1.
`
`2.
`
`5.
`
`7.
`
`8.
`
`9.
`
`REFERENC

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