throbber
bs_bs_banner
`
`doi:10.1111/cei.12206
`
`Clinical and Experimental Immunology
`
`TRANSLATIONAL NEUROIMMUNOLOGY REVIEW SERIES
`Series originators and editors: Olaf Stu¨ve and Uwe Zettl
`
`Requirement for safety monitoring for approved multiple sclerosis
`therapies: an overview
`
`OTHER ARTICLES PUBLISHED IN THIS SERIES
`Paraneoplastic neurological syndromes. Clinical and Experimental Immunology 2014, 175: 336–48.
`Diagnosis, pathogenesis and treatment of myositis: recent advances. Clinical and Experimental Immunology 2014, 175: 349–58.
`Monoclonal antibodies in treatment of multiple sclerosis. Clinical and Experimental Immunology 2014, 175: 373–84.
`CLIPPERS: chronic lymphocytic inflammation with pontine perivascular enhancement responsive to steroids. Review of an increasingly recognized entity within
`the spectrum of inflammatory central nervous system disorders. Clinical and Experimental Immunology 2014, 175: 385–96.
`Disease-modifying therapy in multiple sclerosis and chronic inflammatory demyelinating polyradiculoneuropathy: common and divergent current and future
`strategies. Clinical and Experimental Immunology 2014, 175: 359–72.
`Myasthenia gravis: an update for the clinician. Clinical and Experimental Immunology 2014, 175: 408–18.
`Cerebral vasculitis in adults: what are the steps in order to establish the diagnosis? Red flags and pitfalls. Clinical and Experimental Immunology 2014,
`175: 419–24.
`Multiple sclerosis treatment and infectious issues: update 2013. Clinical and Experimental Immunology 2014, 175: 425–38.
`
`P. S. Rommer,* U. K. Zettl,†
`B. Kieseier,‡ H.-P. Hartung,‡
`T. Menge,‡ E. Frohman,¶
`B. M. Greenberg,¶ B. Hemmer§ and
`O. Stüve‡¶§**
`*Department of Neurology, Medical University
`of Vienna, Vienna, Austria, †Department of
`Neurology, University of Rostock, Rostock,
`‡Department of Neurology, Medical Faculty,
`Heinrich Heine University, Düsseldorf,
`§Department of Neurology, Klinikum rechts der
`Isar, Technische Universität München, Munich,
`Germany, ¶Department of Neurology, University
`of Texas Southwestern Medical Center, and
`**Neurology Section, VA North Texas Health
`Care System, Dallas VA Medical Center, Dallas,
`TX, USA
`
`Accepted for publication 11 September 2013
`Correspondence: O. Stüve, Neurology Section,
`VA North Texas Health Care System, Medical
`Service, 4500 South Lancaster Road, Dallas, TX
`75216, USA.
`E-mail: olaf.stuve@utsouthwestern.edu
`
`Summary
`
`During the last two decades, treatment options for patients with multiple
`sclerosis (MS) have broadened tremendously. All agents that are currently
`approved for clinical use have potential side effects, and a careful risk–benefit
`evaluation is part of a decision algorithm to identify the optimal treatment
`choice for an individual patient. Whereas glatiramer acetate and interferon
`beta preparations have been used in MS for decades and have a proven safety
`record, more recently approved drugs appear to be more effective, but poten-
`tial risks might be more severe. The potential complications of some novel
`therapies might not even have been identified to their full extent. This review
`is aimed at the clinical neurologist in that it offers insights into potential
`adverse events of each of the approved MS therapeutics: interferon beta,
`glatiramer acetate, mitoxantrone, natalizumab, fingolimod and terifluno-
`mide, as well as recently approved therapeutics such as dimethyl fumarate
`and alemtuzumab. It also provides recommendations for monitoring the dif-
`ferent drugs during therapy in order to avoid common side effects.
`
`Keywords: activation, acute respiratory distress syndrome, multiple sclerosis
`(MS)
`
`Introduction
`
`Multiple sclerosis (MS) is an inflammatory disease affecting
`young adults and is a major cause of disability [1]. MS phe-
`notypes have been differentiated into relapsing–remitting
`MS (RRMS), primary progressive MS (PPMS) and second-
`ary progressive MS (SPMS). In the majority of patients,
`RRMS will proceed eventually to SPMS [2]. While relapsing
`
`forms of MS appear to be driven primarily by central
`nervous system (CNS) inflammation, progressive forms of
`MS are also characterized by extensive neurodegeneration
`[3]. Two decades ago, the first therapeutic agents were
`approved for treatment of relapsing forms of this disorder
`[4]. Since then, the therapeutic options have broadened
`tremendously. While it is now possible to lower the rate
`of clinical attacks and the lesion burden on magnetic
`
`© 2013 British Society for Immunology, Clinical and Experimental Immunology, 175: 397–407
`
`397
`
`Merck 2034
`TWi v Merck
`IPR2023-00049
`
`

`

`TRANSLATIONAL NEUROIMMUNOLOGY REVIEW SERIES
`
`resonance images (MRI), questions remain regarding the
`long-term benefits derived from any of the approved agents.
`In addition, all the drugs that are currently available for use
`in patients with MS have potential side effects [5], and a
`careful risk–benefit evaluation often helps the neurologist
`to identify the best agent for an individual patient.
`Currently, glatiramer acetate (GA) (Copaxone®), inter-
`feron beta (IFN-β) preparations (Betaseron®, Extavia®,
`Rebif®, Avonex®), mitoxantrone (Novantrone®), natalizu-
`mab (Tysabri®), fingolimod (Gilenya®) and teriflunomide
`(Aubagio®) are approved for therapy of relapsing forms of
`MS in the United States and other countries. Laboratory
`screening tests for specific complications of these agents are
`becoming increasingly complex and a daily routine for MS
`neurologists. The requirement for such tests will be dis-
`cussed in this review.
`
`GA (Copaxone®)
`
`GA is indicated for therapy in RRMS and in first clinical
`relapses in patients with MRI compatible with MS. It was
`approved for RRMS in 1996 by the Food and Drug Admin-
`istration (FDA) [4]. GA reduces the risk of relapses in
`RRMS [6] and may decrease disease progression compared
`to patients who terminated treatment with it
`[7].
`Twenty mg GA are administered subcutaneously (s.c.)
`daily [8].
`GA is a polymer of amino acids (glutamic acid, lysine,
`alanine and tyrosine). This co-polymer was originally gen-
`erated to mimic myelin basic protein, and to allow the
`induction of the MS animal model experimental autoim-
`mune
`encephalomyelitis
`(EAE). Unexpectedly, disease
`resistance was observed [9], which led to clinical trials in
`MS patients. The complex mode of action of GA is still not
`understood fully. However, cellular immune responses are
`shifted from inflammatory to anti-inflammatory cytokines.
`This T helper type 1 (Th1) to Th2 shift seems to be respon-
`sible for some of the effects. Other mechanisms are inhibi-
`tion of activation and proliferation of encephalitogenic T
`cells and a modulation of antigen-presenting cells [10–12].
`Brain-derived neurotrophic factor (BDNF) production is
`increased in response to GA treatment. BDNF may possess
`neuroprotective capacity [13], as it may play an important
`role in the protection of axons [14].
`Potential side effects of GA include immediate post-
`injection reactions such as flushing, chest pain, palpitations,
`anxiety, dyspnoea, urticaria and constriction of the throat.
`These side effects are usually self-limited and occur unpre-
`dictably several months after initiation. Chest pain may be
`associated with post-injection status, but it also may occur
`without a temporal relation to injections. Lipoatrophy has
`been reported [15,16]. To avoid skin necrosis the patients
`should follow injection techniques as stated in the prescrib-
`ing information. Antibodies
`targeting GA have been
`reported, but it is not plausible that they antagonize its
`
`P. S. Rommer et al.
`
`actions in vivo. Usually, many of these side effects are self-
`limited or can be avoided by proper injection [8]. Whereas
`flu-like symptoms are less frequent in patients on GA
`therapy compared to patients on IFN-β treatment, injection
`side reactions are more common in patients under GA
`treatment [17]. Recent reports have shown hepatic toxicity
`under treatment with GA [18,19]. These reports need to be
`validated.
`No laboratory monitoring is required during GA therapy
`(see Table 1).
`GA is a pregnancy category B (see Table 2), meaning that
`no adverse effects on embryonal development were observed
`in animal reproduction studies. Well-controlled clinical
`trials
`in pregnant women are lacking. Consequently,
`GA should be used during pregnancy only if clearly
`needed [8].
`
`IFN-β
`IFN-β was first approved for MS in the in 1993. There are
`three different products available – IFN-β-1b preparations
`(Betaseron®, Extavia®) that are administered s.c. every
`other day, and IFN-β-1a preparations that are either admin-
`istered s.c. (Rebif®) three times weekly or intramuscularly
`(i.m.) (Avonex®) once a week [4].
`IFN-β is a purified, lyophilized protein product generated
`by recombinant DNA techniques. In response to viruses,
`IFN-β is produced by the innate immune systems. A reduc-
`tion of T cell activation, a cytokine shift in favour of
`anti-inflammatory effects, induction of regulatory T cells
`and prevention of leucocytes from crossing the blood–brain
`barrier have been shown. In addition, IFN-β leads to higher
`neutrotrophic factor expression, promotes anti-viral effects
`and apoptosis of autoreactive T cells. The exact mechanisms
`by which IFN-β benefits patients with MS are currently not
`known [20].
`The most common side effects are flu-like symptoms.
`Symptoms may be minimized by the intake of analgesics
`or anti-pyretics prior to injection. As with GA, injection
`site necrosis and reactions have been reported. Again,
`proper injection technique and the change of injection site
`are of importance in reducing the occurrence of these side
`effects. Injection side effects are more common in patients
`receiving IFN-β-1a three times weekly s.c. when compared
`with patients under IFN-β-1a weekly i.m. [21]. Allergic
`reactions and anaphylaxis are rare complications that have
`to be considered severe. In the case of anaphylaxis, treat-
`ment with interferon has
`to be discontinued. Severe
`hepatic injury has been reported under treatment with
`IFN-β preparations, mainly when therapy occurs in com-
`bination with other hepatotoxic agents. Depression has
`been reported in patients treated with IFN-β. Therefore,
`symptoms of depression have to be monitored, and treat-
`ment discontinued as indicated. In addition, haematolo-
`gical abnormalities including lymphopenia, neutropenia,
`
`398
`
`© 2013 British Society for Immunology, Clinical and Experimental Immunology, 175: 397–407
`
`

`

`Safety monitoring in MS therapies
`
`Table 1. Therapy monitoring in approved therapeutics. Possible side effects.
`
`TRANSLATIONAL NEUROIMMUNOLOGY REVIEW SERIES
`
`Potential side effects
`
`Recommended monitoring
`
`Glatiramer
`acetate
`Interferon beta
`
`Mitoxantrone
`
`Natalizumab
`
`Fingolimod
`
`Teriflunomide
`
`Alemtuzumab
`
`Dimethyl fumarate
`
`Flushing, chest pain, dyspnoea
`Palpitations, urticaria, skin necrosis
`Flu-like symptoms, injection-site necrosis
`Depression, allergic reactions
`Hepatic injury, neutropenia
`Lipoatrophy
`Congestion heart failure
`Urine colour blue-green
`Birth deficiency, sterility
`Hair loss, nausea
`PML, fever, joint pain
`Liver disease, melanoma
`Allergic reactions
`Bradycardia, heart failure
`Fever, diarrhoea, liver disease
`Macular oedema, skin cancers
`Enzephalities
`Hepatic injury, elevated liver enzymes,
`infections, polyneuropathy
`Autoimmune disorders (thyroid disorders, immune
`thrombocytic purpura), infusion-related side effects
`Lymphopenia, gastrointestinal side effects
`
`None
`
`Liver enzymes
`Blood count
`Thryroid testing
`Neutralizing antibodies
`Left ventricular ejection fraction
`ECG, differential blood count,
`Liver enzymes, pregnancy testing
`
`JC-virus
`Neutralizing antibodies
`
`ECG, cardiological evaluation
`Ophtalmological evaluation
`VZV-antibodies, liver enzymes
`
`Liver enzymes, pregnancy testing,
`white blood count
`Complete monthly blood counts,
`testing for autoimmunity
`White blood cell count
`
`PML: progressive multi-focal leucoencephalopathy; ECG: electrocardiography; VZV: varicella zoster virus.
`
`anaemia and leukopenia have been reported [22–27]. Two
`other relatively common side effects of IFN-β therapy in
`patients with MS are thyroid autoimmunity and hypothy-
`roidism [28], although other reports could not show a sig-
`nificant increase in thyroid dysfunction or anti-thyroid
`autoantibody positivity [29,30].
`In conclusion, treatment with IFN-β is considered safe
`and well tolerated. However, after the approval of IFN-β
`cases with autoimmune diseases,
`including idiopathic
`thrombocytopenia, hypo- and hyperthyroidism and auto-
`immune hepatitis, have been reported. Thus, liver enzymes
`should be monitored in regular intervals in the absence of
`signs of liver injury (1 month, 3 months and 6 months after
`
`initiation and each 6 months afterwards). Known liver
`disease is a contraindication to therapy with IFN-β. Liver
`transaminase levels of greater than five times of normal
`should lead to a dose reduction. If enzyme levels do not
`convert to normal, treatment has to be discontinued. If
`enzyme levels normalize after a dose reduction, a return to
`the full dose can be initiated with ongoing hepatic monitor-
`ing. In addition, complete blood counts should be obtained
`after 1 month, 3 months, 6 months and each 6 months
`thereafter. Thyroid testing should be performed initially and
`afterwards only in the case of abnormalities every 6 months
`and when clinical signs of hypo- or hyperthyroidism are
`obvious (see Table 1).
`
`Table 2. Potential risk of MS therapeutics in pregnancy.
`
`FDA pregnancy
`category
`
`A
`B
`
`C
`
`D
`
`X
`
`Interpretation
`
`Therapeutic agent
`
`Well-controlled trials in pregnant women revealed no increased risk for fetus
`No well-controlled trials, but animal trials revealed no increased risk or
`well-controlled trials revealed no risk, whereas animal trials have shown
`adverse effects
`Animal studies have shown increased risk for the fetus or have not been
`conducted; no well-controlled trials in pregnant women
`Studies have shown harm to the fetus; however, the benefit may outweigh risk
`under certain circumstances
`Studies in animals or humans have demonstrated fetal abnormalities and/or there
`is positive evidence of human fetal risk based on adverse reaction data from
`investigational or marketing experience, and the risks involved in use of the
`drug in pregnant women clearly outweigh potential benefits
`
`Glatiramer acetate
`
`Interferon beta, natalizumab, fingolimod,
`alemtuzumab, dimethyl fumarate
`Mitoxantrone
`
`Teriflunomide
`
`© 2013 British Society for Immunology, Clinical and Experimental Immunology, 175: 397–407
`
`399
`
`

`

`TRANSLATIONAL NEUROIMMUNOLOGY REVIEW SERIES
`
`Like all therapeutic proteins, IFN-β is immunogenic and
`can induce the production of binding and neutralizing anti-
`bodies [31]. Neutralizing antibodies are up to seven times
`more prevalent in patients receiving IFN-β-1b every other
`day or IFN-β-1a s.c. three times weekly, when compared
`with IFN-β-1a i.m. once a week [21,32]. Routine testing for
`IFN-β neutralizing antibodies is currently not universally
`recommended. Testing for neutralizing antibodies might be
`recommended in the setting of clinical disease progression
`under IFN-β treatment. If testing is performed, the presence
`of high titres against IFN-β on recurrent testing or the
`failure to induce interferon inducible protein (MxA) should
`perhaps lead to the discontinuation of therapy and a switch
`to a different class of drug [33].
`One lethal case of capillary leak syndrome was reported
`in a patient with monoclonal gammopathy of unknown sig-
`nificance (MGUS) after one administration of IFN-β-1b.
`Post-mortem measurements showed a deficiency of C1
`inhibitor (C1-INH) that controls the complement system.
`The release of proinflammatory cytokines appears to have
`resulted in an uncontrolled activation of complement
`factor. There are reports of associations of MGUS and
`C1-INH. An autopsy did not confirm the diagnosis of clini-
`cal definite MS [34]. In patients with MGUS and MS who
`are candidates for IFN-β therapy, the level of C1-INH
`should be determined.
`IFN-β-1b has been assigned pregnancy category C. In
`animals,
`significant
`increases
`in embryolethal
`and
`arbotifacient effects could be shown under doses approxi-
`mately two to three times higher than the doses used in
`patients with MS. Well-controlled trials in humans are
`lacking and not feasible. However, spontaneous abortions
`have been reported in clinical trials. Women should be
`informed about this risk and treatment should be discon-
`tinued when women intend to become pregnant or during
`pregnancy [35–37].
`A polyethylene glycol (PEGylated) formulation of IFN-β
`with longer injection intervals is currently under investiga-
`tion, showing promising preliminary data according to a
`recent press release [Action in Diabetes and Vascular
`Disease: Preterax and Diamicron MR Controlled Evaluation
`(ADVANCE) study] [38].
`
`Mitoxantrone
`
`Mitoxantrone (Novantrone®) was approved in 2000 by the
`FDA for rapidly worsening RRMS or secondary progressive
`MS [39]. It has proved its efficacy in several trials [40,41].
`Mitoxantrone is administered at doses of 12 mg/m2 every
`3 months intravenously (i.v.) as short infusions. It is an
`anti-neoplastic
`cytotoxic agent
`that
`inhibits
`type
`II
`topoisomerase and disrupts DNA synthesis. Furthermore,
`mitoxantrone showed effects on the proliferation of T and
`B cells and induces natural killer (NK) cell maturation
`[42,43]. It was first used in cancer therapy. From cancer
`
`P. S. Rommer et al.
`
`patients receiving mitoxantrone it is known that there is a
`dose-dependent risk of developing cardiomyopathy [44].
`Because of
`reports of congestive heart
`failure and
`decreases in the left cardiac ejection fraction, cardiac moni-
`toring has been recommended. Heart failure may occur
`during or after termination of therapy with mitoxantrone
`[45,46]. The risk correlates with accumulating doses of
`mitoxantrone, and a cumulative dose of 140 mg/m2 should
`not be exceeded. The incidence of secondary lymphoid
`cancer is estimated to be between 0·25 and 6%. There seems
`to be no correlation between the applied dose and the likeli-
`hood for lymphoma. These complications have substan-
`tially limited the use of mitoxantrone despite its proven
`efficacy [47].
`Prior to initiation of therapy, left ventricular ejection
`fraction (LVEF) should be obtained by echocardiogram,
`multi-gated radionucleotide angiography (MUGA) or
`MRI. Prior to each infusion with mitoxantrone an electro-
`cardiogram (ECG) should be performed. In addition, a
`quantitative re-evaluation of LVEF should be performed
`before initiation of mitoxantrone, during therapy with
`mitoxantrone and yearly after termination of mitoxantrone
`using the same method utilized at baseline [48]. A signifi-
`cant reduction of LVEF (below 50%) is a contraindication
`for initiation of therapy with mitoxantrone and a reason for
`terminating therapy.
`Because mitoxantrone leads to a reduction in the number
`of leucocytes, administration of mitoxantrone is not recom-
`mended when neutrophil numbers fall below 1500 mm3.
`Complete blood count and differential blood count, as well
`as thrombocytes and liver enzymes, should be tested prior
`to each administration. Patients with hepatic insufficiency
`with threefold elevated liver enzymes should not be admin-
`istered mitoxantrone, as it is metabolized in the liver (see
`Table 1) [48,49]. Liver toxicity has been reported in as many
`as 15% of treated patients [50].
`During therapy with mitoxantrone, vaccinations with live
`virus vaccines should be avoided. The application of other
`anti-neoplastic agents should be avoided. The patient
`should be aware that the urine may be blue–green in colour
`for some days after infusion. Other side effects include tran-
`sient hair loss or thinning and nausea, and menstrual disor-
`ders in females. If there are signs of extravasation, the
`infusion has to be stopped immediately to avoid tissue
`necrosis [49].
`Patients who have not completed their family planning
`should be informed that mitoxantrone may cause sterility.
`As mitoxantrone may cause birth defects, contraception is
`required during therapy. A pregnancy test should be con-
`ducted prior to each administration [49]. Well-controlled
`trials are currently lacking in pregnant women [51].
`Mitoxantrone has been assigned to pregnancy category D
`by the FDA. Animal data suggest fetotoxicity (low fetal birth
`weight and retarded development of the fetal kidney) and
`premature delivery [48] (see Table 2).
`
`400
`
`© 2013 British Society for Immunology, Clinical and Experimental Immunology, 175: 397–407
`
`

`

`TRANSLATIONAL NEUROIMMUNOLOGY REVIEW SERIES
`
`Safety monitoring in MS therapies
`
`Natalizumab
`
`Natalizumab is a humanized recombinant monoclonal anti-
`body against the α4-chain of integrins that was designed to
`diminish leucocyte migration from the peripheral blood
`into the CNS. Specifically, very late activating antigen-4
`identical with α4-chain of α4β1-integrin) is
`(VLA-4;
`decreased in its ability to bind its ligand, vascular cell adhe-
`sion molecule (VCAM)-1 [52,53]. Natalizumab received
`accelerated approval by the FDA in 2004 based on the
`results after 1 year of treatment in two placebo-controlled
`trials. The trials were ongoing for another year [54,55]. The
`agent was withdrawn voluntarily by its manufacturers in
`2005,
`after
`three
`cases
`of
`progressive multi-focal
`leucoencephalopathy (PML) in patients with MS and
`Crohn’s disease were reported [5,56,57]. PML is an infec-
`tion of cells in the CNS with the human polyoma virus JC
`(JCV).
`In 2006, natalizumab was
`reintroduced.
`It
`is
`approved for relapsing forms of MS in the United States
`[58] and for highly active forms of RRMS (defined as failed
`response to other therapeutics such as IFN-β or GA, or if
`disease is evolving rapidly) [58,59].
`Potential side effects are common and can be observed in
`about 10% of all patients. Side effects include fever, joint
`pains, headache, dizziness, depression, vaginitis, gastroen-
`teritis, feeling or being sick and sore throat [60]. Herpes
`infections have also been reported in MS patients under
`natalizumab therapy, but it is unclear that there is an
`increased incidence compared to the general population
`[61].
`Severe side effects and complications include PML, aller-
`gic reactions and liver disorders. Recently, risk stratification
`for patients with MS on natalizumab became possible.
`Specifically, a positive anti-JCV antibody status reflecting
`infection with JCV [62], previous treatment with immuno-
`suppressants such as mitoxantrone or cyclophosphamide
`and treatment duration with natalizumab (more than 24
`monthly infusions) were determined to be correlated with a
`higher risk of PML. The risk differs from fewer than one in
`10 000 in patients with no risk factors to up to 11 in 1000 in
`patients with positive JCV status, previous treatment with
`immunosuppressants and treatment duration longer than
`24 months [63]. Recently, a trial investigating the accuracy
`of JCV seropositivity revealed that there is a false negative
`rate of JCV in the serum of 37% when compared with the
`virus load in the urine. Thus, a negative JCV test may
`underestimate the rate of JCV latency in a given individual
`[64]. Regular monitoring warrants for clinical signs for
`PML and JCV testing should be repeated in negative
`patients every 6 months. Upon suspicion of PML, treatment
`with natalizumab should be terminated immediately. The
`clinical and imaging diagnosis of PML should be confirmed
`by MRI scan, cerebrospinal fluid (CSF) and polymerase
`chain reaction (PCR) testing for JCV. In cases with sus-
`pected PML and absence of JCV copies by PCR in CSF, a
`
`brain biopsy could be considered. Plasma exchange is often
`performed to accelerate the elimination of natalizumab.
`However, there is no evidence that the use of plasma
`exchange favourably alters clinical outcomes. In addition,
`nearly all PML patients develop paradoxical deterioration
`after termination of natalizumab. Responsible for this dete-
`rioration is the immune reconstitution inflammatory syn-
`drome (IRIS), which is known from cases of PML in AIDS
`patients. In this situation, the use of glucocorticosteroids is
`recommended [65,66].
`Further side effects include liver dysfunction with an
`increase of liver enzymes and an increase of bilirubin. These
`effects can be observed typically within days of treatment
`initiation [60], although delayed reactions have also been
`described [67]. Even in the absence of relevant clinical
`signs, liver enzymes should be tested prior to treatment,
`after 1 month and after 3 months of therapy initiation.
`Complete blood counts with cell differential, as well as
`platelet count, should be determined 1, 3 and 6 months
`after initiation, and every 6 months thereafter [60] (see
`Table 1). Skin cancers have been reported under treatment
`with natalizumab [68,69].
`Persistent anti-idiotypic antibodies against natalizumab
`(detected at two time-points) will prevent the drug from
`being efficient. Thus, therapy has to be terminated. In the
`case of anaphylaxis or allergic reaction, neutralizing anti-
`bodies are typically detectable [54]. The prevalence of neu-
`tralizing antibodies appears higher in patients in whom
`natalizumab therapy was stopped within 6 months of
`initiation and then restarted later. Because of the risk of
`allergic reaction, post-infusion observation for 1 h is
`recommended.
`Well-controlled trials in pregnant women are currently
`lacking. Natalizumab has been assigned a pregnancy cat-
`egory C. In animal studies, a higher rate of abortion was
`observed at doses seven times the human dose (see Table 2).
`Natalizumab therapy should be reserved for those patients
`in whom potential benefits outweigh potential risks [60]. As
`there is currently no exit strategy for natalizumab that pre-
`vents disease reactivation, discontinuation during preg-
`nancy presents its own challenges.
`
`Fingolimod (Gilenya®)
`
`Fingolimod is approved by the FDA for RRMS [70] and in
`Europe by the European Medicines Agency (EMA) for
`patients with RRMS and disease activity, despite first-line
`treatment, or in patients with evolving severe RRMS. It is
`administered orally
`as 0·5-mg
`capsules daily
`[71].
`Fingolimod binds to sphingosine-1-phospate (S1P) recep-
`tors on immune cells. Consequently, these immune cells are
`unable to egress from lymphatic tissue, and subsequently
`into the CNS [72]. Only lymphocytes that reside within sec-
`ondary lymphoid organs are affected, which account for
`approximately 2% of all circulating lymphocytes.
`In
`
`© 2013 British Society for Immunology, Clinical and Experimental Immunology, 175: 397–407
`
`401
`
`

`

`TRANSLATIONAL NEUROIMMUNOLOGY REVIEW SERIES
`
`addition to the effects on immune cells, there is emerging
`evidence that fingolimod may modulate S1P receptors in
`the CNS and may reduce neurodegenerative processes
`[73,74].
`The most common side effects of fingolimod are head-
`ache, flu-like symptoms, diarrhoea, back pain, liver enzyme
`elevations and cough. More severe side effects such as
`cardiac complications are common, with an incidence
`between 1 and 10% of patients treated with fingolimod. A
`first-degree atrioventricular block was reported in about
`4·7% in patients treated with 0·5 mg fingolimod. Other
`risks include a minor increase in blood pressure, decrease in
`lung function, macular oedema and an increased frequency
`of viral infections, in particular varizella zoster [75–79].
`Macular oedema may lead to progressive visual loss. The
`pathogenic role of fingolimod in macular oedema is cur-
`rently not understood fully. In two Phase III trials, 13
`patients developed macular oedema, 10 of them within the
`first 4 months of treatment. Eleven of the 13 patients with
`macular oedema were administered a dose that is more than
`twice as high as the currently approved dose [77,78].
`Cutaneous neoplasias were reported more often in the
`fingolimod group than in the IFN or placebo control
`groups. Basaliomas and melanomas (in situ) were reported
`[75,76]. A dermatology screening examination could be
`suggested before the initiation of fingolimod therapy.
`In the context of one Phase II clinical trial, one MS
`patient died from varicella zoster infection and consecutive
`hepatic failure, and another patient died from herpes sim-
`plex virus 1 encephalitis. A third patient was diagnosed with
`a life-threatening HSV encephalitis. All these patients were
`treated with the higher, non-approved 1·25 mg dose [78].
`FDA and EMA currently recommend 6-h heart monitor-
`ing with continuous ECG monitoring during the first
`administration of fingolimod. If bradycardia occurs within
`the first 6 h, cardiac monitoring should be extended for
`another 2 h. The occurrence of severe bradycardia, QTC
`interval prolongation, AV block II Wenckeback or AV
`block III
`requires overnight observation.
`In patients
`who become clinically symptomatic during fingolimod-
`related bradyarrhythmias or who take other bradycardia-
`promoting agents should be assessed by a cardiologists to
`determine the feasibility of fingolimod therapy. In patients
`with atrioventricular block II, significant QT-prolongation,
`symptomatic known bradycardia or history of syncope,
`ischaemic heart disease or history of myocardial infarctions
`or cerebrovascular infarction, uncontrollable arterial hyper-
`tension or congestive heart disease, fingolimod cannot be
`recommended. Because of the first-dose cardiac side effects
`of fingolimod, cardiac monitoring has to be repeated in all
`patients who experience a treatment hiatus of 14 days or
`longer [78,79]. Additionally, the EMA requires for repeated
`monitoring when the treatment is interrupted for 1 day
`during the first 2 weeks of treatment, or 7 days during week
`3 and 4 of treatment [80].
`
`P. S. Rommer et al.
`
`Moreover, the majority of cases with macular oedema
`occurred within the first 3–4 months after onset of therapy.
`Patients may report blurred vision or decreased vision or
`may be asymptomatic. The incidence is about 0·4% in the
`0·5 mg group, with a higher incidence with patients with a
`history of uveitis. After termination of therapy, macular
`oedema usually resolves spontaneously; therefore, evalua-
`tion of the fundus has to be performed prior to initiation of
`therapy, within 4 months after onset of therapy and at any
`time of decreased visual acuity [77,78].
`Vaccination during therapy with fingolimod may be less
`effective. Vaccination with live attenuated virus vaccines
`should be avoided during, and 2 months after, fingolimod
`therapy as it may carry the risk of infections. In addition,
`patients without a history of chickenpox or vaccination
`against varicella zoster virus (VZV) should be tested for
`VZV antibodies. In those without antibodies vaccination
`should be considered before initiation [81]. Vaccination is
`recommended 1 month prior to initiation with fingolimod
`therapy in order to ensure immunization. Monitoring of
`therapy should also include complete cell counts at initia-
`tion of therapy, months 1, 3 and 6 and in periodic intervals
`thereafter [82] (see Table 1).
`Fingolimod has been assigned to pregnancy category C
`by the FDA. Animal studies demonstrated evidence of fetal
`outcomes,
`including teratogenicity and embryolethality.
`Well-controlled trials in women are currently lacking. Prior
`to initiation a pregnancy test has to be conducted. If a
`patient becomes pregnant during treatment, application of
`fingolimod should be terminated [82,83] (see Table 2).
`
`Teriflunomide (Aubagio®)
`
`Teriflunomide was approved in 2012 by the FDA for treat-
`ment of relapsing forms of MS [84] after it had docu-
`mented efficacy in several trials [85–87]. Teriflunomide is a
`pyrimidine synthesis inhibitor and has anti-inflammatory
`properties by inhibiting dihydroorotate dehydrogenase
`which, in turn, is necessary for pyrimidine synthesis. Prolif-
`eration of autoreactive B and T cells is reduced. Addition-
`ally,
`immunomodulatory effects on the expression of
`cytokines by lymphocytes have been shown [88,89]. The
`exact mechanisms by which teriflunomide mediates its ben-
`efits in MS are not understood fully. Teriflunomide is avail-
`able in two doses: 7 or 14 mg once daily in the United States
`[90] and 14 mg in Europe [91].
`Hepatotoxicity was reported with leflunomide, the parent
`drug of teriflunomide, in patients with rheumatoid arthri-
`tis. Adverse effects of teriflunomide include decreased white
`blood count (WBC), and infections. Specifically, cases of
`tuberculosis were reported. Polyneuropathy, renal failure,
`skin reactions, hair thinning and an increase in blood pres-
`sure have also been observed [86,92].
`Teriflunomide is contraindicated in patients with severe
`hepatic injury. In other patients, liver enzymes have to be
`
`402
`
`© 2013 British Society for Immunology, Clinical and Experimental Immunology, 175: 397–407
`
`

`

`TRANSLATIONAL NEUROIMMUNOLOGY REVIEW SERIES
`
`Safety monitoring in MS therapies
`
`monitored prior to initiation and for at least 6 months after
`initiation. A complete blood count has to be conducted. In
`addi

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