throbber
“m Check for updates
`
`Review
`Therapeutic Advances in Neurologicai Disorders
`
`Current and emerging therapies in
`multiple sclerosis: a systematic review
`
`Wanda Castro-Barrera. Donna Graves. Teresa C. Frohman. Angela Bates Flores, Paula
`Herdsman. Diana Logan, Megan Orchard. Benjamin Greenberg and Elliot M. Frohman
`
`Thar Adv Naomi Disord
`{2012] Silt} 205—220
`DOI:'|D.11'17I
`l756285612lz50936
`© The Authoris], 2012.
`Reprints and permissions:
`hillewwwsagepubiCduk/
`journalsPermissionsnav
`
`Abstract: Multiple sclerosis [MS] is a potentially disabling chronic autoimmune neurological
`disease that mainly affects young adults. Our understanding of the pathophysiology of MS has
`significantly advanced in the past quarter of a century. This has led to the development of many
`disease-modifying therapies iDMTs] that prevent exacerbations and new lesions in patients
`with relapsing remitting MS iRRMS]. So far there is no drug available that can completely halt
`the neurodegenerative changes associated with the disease. it is the purpose of this review to
`provide concise information regarding mechanism of action. indications. side effects and safety
`of Food and Drug Administration and European Medicines Agency approved agents for MS.
`emerging therapies. and drugs that can be considered for off-label use in MS.
`
`Keywords: disease-modifying therapies. emerging therapies. fingolimod. glatiramer acetate.
`interferon B, multipte scterosis. natalizumab
`
`Introduction
`
`is a chronic autoim—
`Multiple sclerosis (MS)
`mune inflammatory disease of the central nerv-
`ous system (CNS) that mainly affects young
`adults and may lead to significant disability over
`time. Since the first documented case of MS in the
`nineteenth century the knowledge regarding the
`pathophysiology of the disease has significantly
`advanced. The inflammatory cells in MS have
`been well described and include CD4 and CD8
`
`T lymphocytes, microglia and macrophages
`{Govermam 2011]. Also humoral immunity has
`been described as an important component in
`the pathophysiology of MS [Boater et al. 2010].
`
`Within the past 30 years new and effective
`therapies have been developed that decreased
`clinical relapses, reduced newT2 and gadolinium—
`enhancing (Gad+) lesions and aim to halt the
`progression of disease. Since the US Food and
`Drug Administration (FDA) approval of the first
`disease-modifying therapy (DMT)
`in 1993,
`interferon (IFN)—i31b (Betaseron), which was
`also approved in Europe in 1995 under the name
`of Betaferon, we now have a total of eight FDA—
`approved therapies for MS, including an oral
`agent and a single agent approved for secondary
`progressive MS (SPMS) (Table 1). Of note, there
`
`are two agents approved by the European
`Medicines Agency (EMA) for the treatment of
`SPMS, mitoxantrone and iFN—Blb (Betaferon/
`Extavia). All first-line iniectable agents have
`been studied in clinically isolated syndrome
`(CIS) and have demonstrated decreased risk of
`conversion into clinically definite MS (CDMS)
`(Table 2) [Kappos er a]. 2006; Jacobs er al. 20003
`Comi et al. 2001, 2009, 2012a]. So far there is
`no effective therapy to halt progression of disease
`and reduce disability in primary progressive MS
`(PPMS).
`
`There are many new agents in the pipeline which
`will bring great choices into the MS pharmaco—
`logical armamentarium (Table 3).
`
`Correspondence to;
`Wanda Castro-Borrero, MD
`University of Connecticut
`Health Center.
`Neurology Associates,
`263 Farmington Ave.
`Farmington. CT 0603!]-
`5357. USA
`weastrolauchcmdu
`Donna Graves. MD.
`Teresa C.Frohrnal1. PAC,
`Angeta Bales Flores, MD,
`Paula Herdsman, PAC.
`Diana Logan. RN,
`FNP-C. BC, MSCN,
`Hagan Orchard. PAC.
`Ianjamtn Greenherg.
`MD. HHS
`Elliot M. Frohman.
`MD. PhD
`University of Texas
`Southwestern Medical
`Center, Mutlipte Scterosis
`Program. Dallas. TX. USA
`
`FDA- and EMA-approved therapies
`
`Interferon ,6
`IFNs are a family of proteins that play a role in
`the body’s natural defense against microbial, neo-
`plastic and viral insults and have a rote in regu—
`lating the immune response. IFN—B impacts the
`immune system in several ways, such as decreas—
`ing major histocompatibility complex (MHC)
`class II expression, upregulation of interleukin 10
`
`
`
`http:/llansagepubcom
`
`Page 1 of 16
`
`
`.Ireomgenqfig
` ~22};
`
`a
`
` 205
`
`Biogen Exhibit 2225
`
`Mylan v. Biogen
`IPR 2018-01403
`
`Page 1 of 16
`
`Biogen Exhibit 2225
`Mylan v. Biogen
`IPR 2018-01403
`
`

`

`Therapeutic Advances in Neurological Disorders 5 (4]
`
`Table 1. Current Food and Drug Administration/European Medicines Agency approved therapies for multiple
`sclerosis [MS]
`Wanton
`y
`
`. (
`
`IL—10) production, and decreasedT helper (Tm-1
`and Thl'? production, which leads to an overail
`anti—inflammatory effect [Kieseien 2011; Kappos
`at al. 2007].
`
`Subcutaneous interferon [31b fBetaseron, Bayer
`Scherr'ng Pharma AG/Betaferon. Bayer Schering
`Pharma AG/Extavia, Novartis Pharmaceuticals
`Corp). The pivotal phase III trial using IFN—Blb
`was a randomized, double—blind, placebo—con—
`trolled, multicenter trial of 372 patients with
`RRMS over 2 years. This trial demonstrated a
`34% reduction in overall relapses compared with
`placebo. More specifically, there was a 50% reduc—
`tion in annuah'zed relapses classified as moderate
`to severe in the treatment group. Patients receiv-
`ing IFN—Blb were also found to have a lower T2
`
`lesion volume and decreased accumulation of new
`
`lesions [IFNB Multiple Sclerosis Study Group.
`1993]. Each of the IFN-B therapies, as well as
`glatiramer acetate, has been shown to delay conn
`version to CDMS in patients with C18 (Table 2).
`In the 5-year active treatment extension of the
`BENEFIT trial, the effects of early versus delayed
`treatment with IFN Blb were investigated. This
`study showed the risk of conversion to CDMS
`remained lower in the group receiving early treat—
`ment; 46% compared with 57% of patients con-
`verting from CIS to CDMS [hazard ratio (HR)
`0.63; 95% confidence interval (CI) 0.48—0.83; log
`rank test p = 0.003) [Kappos at (212009}.
`
`intramuscular interferon flia lAvonex, Biogen ldec,
`Inc).
`In the pivotal trial including 30} patients
`
`W 2
`
`httpzl/lan.sagepub.com
`
`06
`
`Page 2 of 16
`
`Page 2 of 16
`
`

`

`W Castro-Borrero, D Graves et at.
`
`
`Table 2. Pivotal tria
`
`
`
`
`
`yaw /.
`
`v .
`
`'
`
`a.
`
`
`
`with RRMS, IFN-Bla intramuscularly was shown
`to delay time to progression of disability with
`fewer treated subjects experiencing disability pro—
`gression (21.9% versus 34.9%; p = 0.02) com—
`pared with placebo. Annualized relapse rates
`(ARRs) over a 2-year period were also lower com—
`pared with placebo (ARR 0.61 versus 0.90; p =
`0.03).The accumulation of Gad+ lesions was also
`reduced; however, T2 lesion volume was not sig-
`nificantiy different between the two groups at 2
`years Uacobs at al. 1996].
`
`Subcutaneous interferon 131a {Rebifi EMD Serena,
`Inc). The Prevention of Relapses and Disabil-
`ity by Interferon B—la Subcutaneously in Mul—
`tiple Sclerosis
`(PRISMS)
`trial was a 2—year
`randomized, double-blind, placebo-controlled,
`multimcentered trial of 560 patients with RRMS.
`Subjects treated with either the 22 or 44 ug dose
`of IFN—Bla subcutaneously showed a significant
`reduction in ARRs compared with placebo, 27%
`and 33% respectively. Both treatment groups
`showed a significant reduction in the number of
`new or enlarging T2 lesions; 67% reduction in
`the 22 ug group and 78% reduction in the 44 ug
`group [PRISMS Study Group, 1998]. An exten-
`sion study utilizing a crossover design in which
`placebo-treated patients were randomized to
`
`either 22 or 44 ug of IFN~f31a subcutaneously
`after 2 years revealed patients in both active
`
`treatment groups for the entire 4 years contin—
`ued to show significantly lower number ofrelapses
`per year EPRISMS Study Group, 2001]. IFNs
`have immunogenic properties and treated indi-
`viduals may develop binding and neutralizing
`antibodies (NAbs) to these products. NAbs may
`develop with the use of all formulations of IFN—
`{3; however, they are found more commonly with
`the high-dose, high-frequency IFNs (IFN—Blb
`and IFN—Bla subcutaneously). The issue of
`NAbs is controversial; however, a panel of
`experts met at the Neutralizing Antibodies on
`Interferon Beta in Multiple Sclerosis (NABI-
`NMS) consortium in 2009 in attempts to for-
`mulate a practical approach to the evaluation
`and incorporation of information regarding
`NAbs in the treatment of MS. The group pro—
`posed that both the NAb titer and clinical status
`of the patient should be considered in the deci-
`sion regarding the impact of the presence of
`NAbs on changing DMTs. They also suggested
`reevaluation of the NAbs status prior to making
`a change in therapy unless patients were clearly
`performing poorly clinicaliy {Poiman er a1. 2010].
`
`Glatiramer acetate
`
`Glatiramer acetate (GA) (Copaxone,Teva Neuro-
`science North America / Teva Pharmaceuticals)
`is a first—line therapy for relapsing forms of MS
`
`W h
`
`ttp:/Jtan.sagepub.com
`
`207
`
`Page 3 0f 16
`
`Page 3 of 16
`
`

`

`.WcomGmklD.Smfo.U::..m.m502flwommqmrfimem,mums“
`.Deg.Lmaqummm
`
`
`
`
`.mfimhd.mknmC313“,epncwmmufwdhmm
`omA
`
`rAnuf.deG.mn0mmcmaeehyge.gsCdn5$6.10
`etfi
`5aoouaTCP
`
`Therapeutic Advances in Neurological Disorders 5 {4]
`
`Iu.MamwT
`
`a
`
`Mr
`
`.r.|\
`
`aG2
`
`Page 4 of 16
`
`WWW.L_fxawmmoneM.bsnne,NmoToSamba
`
`bSPBff
`.mdPakacuAsaa.UCOflr)3nnttfi
`
`
`
`.lflnS(Cum
`OduGekcfiomp
`
`upcmhu.lnum
`cashno
`nmesAaP
`3.16n00g
`ECLMUMop
`
`GmummaRbr
`neubmarnmsra10Ce.m.mqdAmeemwmm
`
`cgégfiamummfimAmmm*
`
`flmgmmmemmm.ma.nmsotmaa.
`
`
`
` mswmlamalI.VawmmgmmkaUnmpnmoswfi—e
`
`.mfoomvom
`mmoeombem
`tanauaLrmmicshgnbhgnfl
`
`
`t0.nh.nat.fl
`WuwclmflWMMmCe.uAha0hWmmmprmepup
`eE80.6
`gaflmnaemu
`p.160n«mhPt.1
`ecmtfiCCV...5
`senera6WaraiPmta
`.1V.
`cfrf.met
`
`
`pOOdfibmrplrwtmshkacthSssuh.mfl66W1850Eco.
`
`ID.aid
`
`Page 4 of 16
`
`

`

`W Castro-Borrero, D Graves et at.W
`
`
`
`blocking direct immunologic attack. It was in the
`late 1980s that the immunologic concept ofTh1
`(proinflammatory) and T112 (anti—inflammatory)
`lymphocytes gained momentum.These two types
`of lymphocytes can be identified by the chemicals
`that they manufacture and then secrete. These
`chemicals are known as cytokines, and can be
`divided into inflammatory and proinflammatory.
`In 1997, Aharoni and colleagues published a
`paper that described how GA could stimulate the
`production ofThZ (anti~inflammatory) cells that
`inhibited the inflammatory response by secret—
`ing anti—inflammatory cytokines [Aharoni er a1.
`1997]. The GAs’ effect begins in the peripheral
`tissues in a population of specific lymphocytes
`which circulate in the blood and are capable of
`migrating into the CNS tissue by crossing the
`blood—brain barrier (BBB). These cells then
`encounter fragments of several myelin proteins
`that stimulate the glatiramer cells to multiply and
`begin to produce anti—inflammatory cytokines.
`Since the glatirameruactivated lymphocytes can
`suppress inflammation under way in the diseased
`area of CNS tissue, this process has been given
`the name bystander suppression Uohnson, 2010}.
`To date, data suggest that GA treatment is associ—
`ated with a broader immunomodulatory effect on
`cells of not only the innate but also the adaptive
`immune system. Recent investigations indicate
`that GA treatment may also promote regulatory
`Bncell properties [Lalive er al. 2011].
`
`GA has a relatively narrow adverse effect profile.
`Most frequently patients complain of mild pain
`and pruritis at the injection site. Lipoatrophy and
`skin sire reactions are also seen and may lead to
`discontinuation of therapy. A transient reaction
`called immediate postinjection reaction consists
`of chest tightness, flushing and dyspnea begin—
`ning soon after the injection and lasting no longer
`than 20 min. If no history or evidence of coronary
`artery disease, the patient can be reassured that
`such a reaction is benign [DiPiro er al. 2005].
`
`Multicenter trials with GA have demonstrated
`statistically significant reducu'ons in mean ARR
`that are comparable to those of the IFNs {DiPiro
`er al. 2005]. In two recent studies the efficacy of
`GA was compared with high—dose/highnfi'equency
`IFN—B. In the Rebif versus Glatiratner Acetate in
`Relapsing MS Disease (REGARD) study [Mikol
`er al. 2008}, subcutaneous IFN—B 1 a was compared
`with GA, and in the Betaseron/Betaferon Efficacy
`Yielding Outcomes of a New Dose (BEYOND)
`study [O’Connor
`er al. 2009], subcutaneous
`
`IFN—Blb was compared with GA. In both trials,
`there was no significant difference between IFN
`and GA in the primary endpoints or in any clini—
`cal endpoints, although some differences in mag—
`netic resonance imaging (MRI) measures of
`disease activity have been claimed.
`
`The results from a 15—year analysis of the US
`prospective open-label study of GA indicate that
`long-term continuous use is safe. It also indicates
`that the majority of patients continuing on GA
`therapy in the study have had few relapses and
`minimal disease progression. Of the initial 232
`patients that received at ieast one GA dose since
`study initiation in 1991, only we (43%, ongoing
`cohort) patients continued. Of the 100 patients
`receiving continuous GA as sole immunornodula—
`tory therapy for 15 years (mean disease duration
`of 22 years and mean patient age of 50 years) have
`not transitioned to SPMS, 57% have retained sta-
`ble or improved the Expanded Disability Status
`Scale (EDSS) scores over the course of the study
`and 82% remain ambulatory without mobility aids.
`There was no occurrence of any unforeseen adverse
`events in patients receiving GA therapy.The study
`will continue for 20 years of prospective follow up
`[Ford er a1. 2010] .
`
`Mitoxantrone
`
`Mitoxantrone is an anthracenedione initially
`developed as an anti-neoplastic agent that reduces
`lymphocyte proliferation. Mitoxantrone interca—
`lates into DNA strands, inducing strand breakage
`and inhibition of the DNA repair enzyme topoi-
`somerase II. It is an immunosuppressive agent
`used as a second—line treatment for SPMS, pri—
`mary relapsing multiple sclerosis and worsening
`RRMS. Mitoxantrone was approved for the treat—
`ment of SPMS based on the study by Hartung
`and colleagues {Harmng er al. 2002].
`
`Several studies have shown it to be efficacious in
`reducing exacerbations and number of Gad-I-
`lesions on MRI, and it seems to have elfects on
`disease course up to 5 years after discontinuing
`therapy [Martinelli at al. 2009; Goodin er a1. 2003].
`Nlitoxantrone is given as an intravenous infusion
`over 30 min every 3 months at 12 mg/rn2 for a 2—
`to 3—year period with a maximum cumulative dose
`of 140 mymz. Common side effects include alo-
`pecia, nausea and vomiting, an increased risk of
`infection (particularly urinary and respiratory
`tracts infections) and amenorrhea. Mitoxantrone,
`though eiifective, remains second line due to its
`
`http://lan.sagepub.com
`
`Page 5 0f 16
`
`Page 5 of 16
`
`

`

`
`
`Therapeutic Advances in Neurological Disorders 5 l4]
`
`risk of two serious adverse efiects that can occur
`
`at any time after the first dose is given. The first,
`acute leukemia has an incidence of approximately
`0.81% [Marriott er a1. 2010}. Regular monitor—
`ing of complete blood counts is recommended.
`Mitoxantrone can also cause decreased left ven~
`
`tricular ejection fraction (LVEF) and congestive
`heart failure at a rate of approximately 12% and
`0.4%, respectively [Marriott at at. 2010]. To
`monitor cardiotoxicity, a baseline LVEF must be
`obtained and any patient with an ejection fraction
`less than 50% should not receive mitoxantrone.
`
`It was previously believed that cardiotoxicity could
`only occur with cumulative doses over 96-140
`m m3; however, several reports of cardiotoxicity
`below this threshold have caused the FDA to rec—
`
`ommend monitoring cardiac function before every
`infusion.The therapy must be discontinued if the
`LVEF ever falls below 50% or decreases by 10%
`[Martinelli er al. 2009].
`
`Natalizumab
`
`Migration of leukocytes from the vasculature
`into the parenchyma involves the interaction
`between leukocyte adhesion molecules and their
`complementary ligands on vascular endothelial
`cells. Leukocyte integrins are heterodimeric gly—
`coproteins
`that contain an or and {3 chain
`[Ransohoffi 2007].Vascular cell adhesion mole-
`cule 1 CVCAM—l) is expressed on the surface of
`vascular endothelial cells in the blood vessels
`
`within the CNS and interacts with 004431 integrin
`on lymphocytes to allow for extravasation across
`the BBB. Also, the interaction of ot4l31 integrin
`with fibronectin and osteopontin may modulate
`the survival, priming and activation of leukocytes
`that have entered into the parenchyma of the
`brain and spinal cord. Natalizumab (Tysabri,
`Biogen Idec, Inc.) contains humanized immuno—
`globulin G4K monoclonal antibodies against leu—
`kocyte d4 integrins, including Q40] and 0.407
`integrins, and blocks binding to their endothelial
`receptors (VCAM—1 and mucosal addressin cell
`adhesion molecule l, respectively) [Polman er al.
`2006]. By blocking d4 integrins, natalizumab
`inhibits the migration of leukocytes into the
`brain, which results in reduced inflammation.
`
`Natalizurnab was evaluated for the treatment
`of RRMS in two phase III clinical trials. The
`Natalizumab Safety and Efficacy in relapsing
`remitting multiple sclerosis (AFFIRM) study
`evaluated 942 patients who were randomly
`assigned to receive natalizumab versus placebo
`
`every 4 weeks for 2 years. The primary endpoints
`were the rate of clinical relapse at 1 year and
`the rate of sustained progression of disability,
`measured by the EDSS, at 2 years. Natalizumab
`reduced the risk of sustained disability by 42%
`over 2 years (HR 0.58; 95% CI 0.43—0.77; 13 <
`0.001). It reduced the rate of clinical relapse at
`1 year by 68% (p < 0.001). MRI scans were
`obtained at baseline, 1 year and 2 years.Treatment
`with natalizumab resulted in an 83% reduction of
`
`new or enlarging hyperintense T2 lesions over 2
`years (mean number of lesions 1.9 with natali—
`zumab and 11 with placebo; 13 < 0.001). There
`were 92% fewer Gad+ lesions in the natalizumab
`
`group than in the placebo group at 1 and 2 years
`(p < 0.001).There was also a significant effect on
`Gad+ lesions seen after 6 weeks of natalizumab
`
`treatment [Polman er al. 2006].
`
`The Safety and Eflicacy of Natalizumab in com—
`bination with IFN—fila in patients with RRMS
`(SENTINEL) trial was a 2—year phase III trial
`evaluating treatment with natalizumab or placebo
`in combination with IFN—Bla. The primary end"
`points were the rate of clinical relapse at 1 year
`and accumulative probability of disability pro«
`gression, measured by the EDSS, at 2 years. The
`study showed that treatment with both drugs was
`more effective than treatment with IFN—Bla
`alone. Patients on combination treatment were
`
`less likely to have sustained disability progression
`(23% oersus 29%) and were more likely to remain
`relapse free (61% versus 37%). Combination
`treatment resulted in fewer new or enlarging T2
`lesions (0.9 versus 5.45 p < 0.001) [Rudick er a1.
`2006].The study ended a month early due to the
`occurrence of progressive multifocal
`leukoen—
`cephalopathy (PML) in two patients who received
`natalizumab with IFN—Bla.
`
`The most notable potential adverse effect of natal—
`izumab treatment is the development of PML.
`Following the observation that
`three patients
`treated with natalizumab developed PML, it was
`withdrawn from the market in February 2005
`and reintroduced in July 2006 as monotherapy
`treatment for RRMS. The original risk of PML
`was estimated to be approximately one per 1000
`patients receiving nataliznmab [Berger, 2010]. As
`of 4 January 2012, approximately 96,582 patients
`have received natalizumab since it was marketed
`and there have been 201 confirmed cases of PML
`
`worldwide. Approximately 20% of patients who
`have developed PML have died. Those that sur-
`vived have varying levels of disability, ranging
`
`M 2
`
`http:/lzansagepubtom
`
`10
`
`Page 6 0f 16
`
`Page 6 of 16
`
`

`

`W Castro~Borrero, D Graves et at.M
`
`
`
`from mild to severe. Fewer patients treated and
`wide confidence intervals result in questionable
`estimates beyond 30 months of treatment.
`
`PML is a rare demyelinating disease of the
`brain due to the John Cunningham (JC) virus. It
`is almost always seen in association with an
`underlying immunosuppressive condition. The
`precise explanation for the increased risk ofPML
`with natalizumab therapy remains unknown.
`
`In the natalizumab clinical trials, there was a
`small increase in the rate of infections, including
`herpes infections, pneumonia and urinary tract
`infections. There were no other opportunistic
`infections or increase cases of cancer reported
`[Ransohoff, 2007]. Post—release monitoring dis—
`closed one case of fatal herpes encephalitis,
`one nonfatal case of herpes meningitis, crypto—
`sporidium gastroenteritis, pneumocystis carinii
`pneumonia, varicella pneumonia and mycobac—
`terium avium intracellular complex pneumonia
`[Ransohoff, 20075 Gorelik at 111.2010].
`
`Natalizumab infusions were complicated by seri-
`ous hypersensitivity reactions,
`including fever,
`rash and anaphylaxis, in less than 1% of patients
`and less serious infusion reactions in about 4% of
`patients [Ransohoff, 2007; Polman er al. 20065
`Rudick et al. 2006]. Patients with infusion reac—
`tions were more likely to have persistent NAbs.
`The presence of antibodies lessoned natalizum-
`ab’s clinical efficacy and resulted in clinical and
`radiographic disease activity equivalent to patients
`in the placebo group [Ransohofifi 2007].
`
`Natalizumab is an extremely effective therapy
`for RRMS and is licensed for highly active naive
`patients. Due to the potential risk of PML and
`other opportunistic infections,
`it
`is
`typically
`reserved for patients with clinically or radio-
`graphically extremely active disease either as
`initial therapy or when initial therapy has been
`ineffective or poorly tolerated. Treatment with
`natalizumab requires rigorous ongoing clinical
`surveillance. To minimize the risk of PML,
`patients beginning treatment should have no
`history of immunosuppressive medications in
`the preceding 3 months and should not have
`other conditions that may compromise cell—
`mediated immunity. The FDA and EMA recw
`cmmend the use of the IC virus antibody for
`risk stratification on all patients onTysabri.The
`risk of PML increases after 24 months on ther-
`apy, if there has been prior immunosuppressant
`
`use and the presence of JC virus antibody.
`Patients with positive IC virus antibody, prior
`treatment with an immunosuppressant and who
`have received more than 24 doses of Tysabri
`have an estimated risk of PML of 9—11/1000.
`However, patients without any of those risk fac-
`tors for PML have a risk of PML of less than
`0.1 per 1000 [Sorensen er al. 2012].
`
`Fingolimod
`Fingolimod is an oral sphingosine—l phosphate
`(SIP) receptor modulator. It was approved by the
`FDA in September 2010 as first-line therapy for
`RRMS. However, the EMA has recommended
`that its use be limited to those whose condition
`fails to respond to first—line therapy or only in
`cases of severe, rapidly developing cases of MS. It
`acts as a sphingosine analogue, binding to the
`SIP] receptor on lymphocytes leading to inter~
`nalization and downregulation of their expression
`and thereby preventing the egression of lympho-
`cytes from the lymph nodesAdditionally, through
`interactions with SIP receptors on neural cells,
`fingolimod has been shown to have potentially
`neuroprotective effects in the animal experimen—
`tal autoimmune encephalomyelitis model [Foster
`er al. 2007; Coelho er al. 2007; Miron et at. 2008].
`
`In the 24—month phase III FTY720 Research
`Evaluating Effects of Daily Oral
`therapy in
`Multiple Sclerosis (FREEDOMS) trial compar—
`ing placebo with oral fingolimod at doses of 1.25
`mg and the now FDA—approved 0.5 mg daily
`dose, there was a significant reduction in ARR
`with both doses of fingolimod (0.16 at 1.25 mg
`and 0.18 at 0.5 mg) compared with placebo
`(0.40) which represented a relative reduction of
`60% and 54%, respectively. Furthermore, fingoli—
`mod also reduced the risk of disability progres—
`sion with a probability of disability progression
`(confirmed after 3 months) of 17.7% at the 0.5
`mg dose and 16.6% at the 1.25 mg dose com—
`pared with 24.1% with placebo. Almost 90% of
`patients receiving fingolimod, at either dose, were
`free of enhancing lesions over the course of 2
`years and approximately 50% were free of new or
`enlargingTZ lesions [Kappos et a1. 2010].
`
`The Trial Assessing Injectable Interferon "versus
`FTY720 Oral in RRMS (TRANSFORMS) com—
`paring fingolimod with intramuscular INF-131a
`showed a 52% relative reduction in ARR in the
`patients treated with fingolimod 0.5 mg versus
`IFN. This study showed a similar beneficial
`
`M.
`http:fltan.sagepub.com
`
`Page 7 0f 16
`
`Page 7 of 16
`
`

`

`Therapeutic Advances in Neurological Disorders 5 {4!
`
`effect on MRI markers compared with IFN-Bla;
`however,
`there was no statistically significant
`difference in the disability progression between
`the fingolirnod and IFN—Bla groups [Cohen et al.
`2010].
`
`Despite its eficacy, there are additional safety
`concerns compared with the injectable therapies.
`Data from the two pivotal
`trials showed an
`increased risk of infections, cardiovascular effects,
`including bradycardia and atrioventricular (AV)
`block (first and second degree) with initial dosing
`and macular edema. Each of these was more
`
`common with the higher 1.25 mg dose. Of note,
`there were two deaths related to infections in sub-
`
`jects receiving fingolimod at the 1.25 mg dose in
`TRANSFORMS. One death was secondary to a
`dissemination varicella zoster infection and the
`
`second was related to herpes simplex encephalitis.
`While herpes virus infection has been seen at
`the 0.5 mg dose, cases tended to be mild and
`were not found to occur at a higher rate than the
`control arm [Cohen er a]. 2010].
`
`The EMA recently recommended increased
`patient monitoring during the first dose of fin-
`golimod, including electrocardiogram monitor-
`ing before treatment and then continuously for
`the first 6 h after the first dose is administered,
`and measurement of blood pressure and heart
`rate every hour over the same 6 h.
`
`Off—label therapies
`Immunosuppressive agents, chemotherapies and
`various mAbs have been used off label for many
`
`years as DMTs in MS but the potential benefits
`of these therapies are limited by systemic adverse
`events, such as increased risk of malignancy and
`opportunistic infections. These agents have been
`used in patients who are refractory to or cannot
`tolerate the side efi‘ects of IFN—[i and GA, cannot
`afford FDA-approved therapies, or need intensifi-
`cation of therapy (is. used in combination with
`IFN-B or GA). Also limiting the use of these med»
`ications is the lack of large-scale, controlled trials,
`validating their eficacy.
`
`which is the active metabolite. MA is a potent,
`selective, noncompetitive and reversible inhibitor
`of inosine 5' monophosphate dehydrogenase type
`II. MA inhibits the de novo synthesis pathway of
`guanosine nucleotides without being incorporated
`into DNA. Because '1‘ and B lymphocytes are
`critically dependent for their proliferation on de
`now synthesis of purines, while other cell types
`can utilize salvage pathways, MA has potent cyto—
`static effects on lymphocytes. MA inhibits prolif-
`erative responses ofT and B lymphocytes to both
`mitogenic and allospecific stimulation. MA also
`suppresses antibody formation by B lymphocytes
`[Product information: Cellcept, 2009].
`
`Potential side effects include hypertension, back”
`ache, abdominal pain, diarrhea, nausea, elevated
`transaminases, vomiting, anxiety and tremor.
`Serious side effects include gastrointestinal bleed—
`ing, thrombocytopenia, skin cancer, opportunistic
`infection and PML. Increased susceptibility to
`infection and the possible development of lym—
`phoma may result from immunosuppression.
`
`A retrospective review of experience in treating
`79 patients with MS with MMF showed that
`this agent was well tolerated by the majority of
`patients. Patients were initiated on 500 mg twice
`a day, which was titrated up by 500 mg weekly
`to a maximum of 1000 mg twice a day.While the
`observations were uncontrolled, some of the
`
`patients demonstrated either stabilization or
`improvements in their activities of daily living,
`ambulation and relapse rate [Frohman er al.
`2004]. In a randomized, MRI-blinded, parallel
`group, pilot trial of MMF compared with IFN—
`[513, both drugs appeared safe and well toler—
`ated in the majority of patients. The trial also
`showed a trend toward a lower accumulation of
`combined active MRI lesions. MMF showed a
`
`nonstatistically significant increase in infections
`[Frohman er al. 2010].The dose generally used
`in patients with RRIVIS is 1000 mg twice daily.
`Large, randomized clinical trials are needed to
`better evaluate the safety and efficacy of this
`agent in patients with MS.
`
`Mycophenolate mofetil
`Mycophenolate mofetil (MMF; Cellcept, Roche
`Laboratories, Nutley, N], USA) is FDA and EMA
`approved for preventing rejection of cardiac, liver
`and renal transplants. MMF undergoes rapid and
`complete metabolism to mycophenolic acid (MA),
`
`Azathioprine
`Azathioprine is FDA approved for rejection proph—
`ylaxis (as monotherapy or adjunct) of renal trans—
`plant and rheumatoid arthritis (RA). Although
`not FDA approved, it has been used in the USA
`to treat MS since 1971 [La Mantia er al. 2007].
`Azathioprine is
`licensed for MS therapy in
`
`W 2
`
`httpzfltansagepubtom
`
`12
`
`Page 8 0f 16
`
`Page 8 of 16
`
`

`

`W Castro—Borrero. D Graves at at.W
`
`
`
`Germany. Azathioprine is an imidazole derivative
`of 6—mercaptopurine and acts as an immunosup—
`pressive antimetabolite. It is a purine antagonist
`and afl‘ects DNA replication. It impairs T~cell
`lymphocyte function and is more selective for
`T lymphocytes than for B lymphocytes [Casetta
`at al. 2009].The Cochrane MS Group concluded
`that azathioprine is an appropriate maintenance
`treatment for patients with MS and could be a fair
`alternative to IFN. It is recommended that cumu-
`
`fatigue, headache, muscle
`fever,
`leucopenia,
`spasms and diarrhea. Cases of PML, severe
`mucocutaneous reactions, tumor lysis syndrome
`and fatal infusion reactions have been docu-
`mented. Other severe adverse reactions include
`
`fulminant hepatitis, hepatic failure, bacterial,
`fungal or viral infections, cardiac arrhythmias,
`renal toxicity and bowel obstruction or perfora—
`tion [Prescribing information, 2010].
`
`lative doses do not exceed 600 g due to possibly
`increasing the risk of malignancies [Casetta et at.
`2009].
`
`In a 72—week, open-label phase I trial the safety
`and tolerability of rituxirnab were evaluated in 26
`patients with RRMS. The authors indicated that
`
`Methotrexate
`
`Methotrexate (MTX) is a chemotherapeutic agent
`used for the treannent of severe psoriasis, juvenile
`RA ORA), severe RA, acute lymphoid leukemia
`and other malignancies. MTX reversibly inhibits
`dihydrofolate reductase.Via this mechanism, MTX
`sodium interferes with DNA synthesis, repair
`and cellular
`replication [Product
`information:
`methotrexate, 2000, 2005].
`
`On a systematic review of oral MTX for M8, for
`the Cochrane Multiple Sclerosis Group,
`the
`authors do not recommend the use of MTX for
`
`progressive MS or RRMS due to a lack of high—
`quality evidence. Future trials need to be performed
`using standard outcome measures and objective
`measures, such as MRI [Gray et al. 2004].
`
`Rituximab
`
`Ritnximab is FDA approved for the treatment of
`nonuHodgkin’s lymphoma, chronic lymphocytic
`leukemia, refractory moderate to severe RA,
`Wegener’s granulomatosis and microscopic pol—
`yangiitis [Prescribing information, 2010]. It is
`EMA approved for diffuse large B—cell
`lym—
`phoma and autoimmune arthritis. Rituximab is
`a chimeric murine/human mAb that targets and
`selectively binds CD20, an antigen present on
`pre-B cells and B cells, but not on antibody-
`producing plasma cells or stem cells in the bone
`marrow. By binding CD20, rituximab depletes
`circulating Bucell populations (but not stem cells
`or plasma cells) through a combination of cell—
`mediated and complementmdependent cytotox—
`icity and possibly promoting apoptosis [Bar—Or
`er al. 2008].
`
`Common side effects ofrituximab include naso—
`
`pharyngits, urinary tract
`
`infections, nausea,
`
`no serious adverse events were reported in this
`small cohort with active RRMS and all
`the
`adverse events including infections were mild to
`moderate and did not lead to medication with—
`drawal. No efficacy conclusions were noted due
`to the absence of a control group but they noticed
`a reduction in relapses, Gad+ lesions, new T2
`lesion number and T2 lesion volumes through
`72 weeks [Bar—Or er of. 2008]. In a phase II
`randomized, placebo-controlled trial with 104
`patients there was a reduction in Gad+ lesions
`
`and relapses in patients on rituximab oersus pla—
`cebo [Hauser er a1. 2008]. Rituximab has shown
`efficacy in the treatment of patients with RRMS.
`A recently completed randomized clinical trial
`using a standard dose of rituximab in patients with
`RRMS demonstrated a 91% reduction in the
`
`number of Gad+ lesions on MRI, as we

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