throbber
Journal of Rehabilitation Research and Development
`Vol. 39 No. 2, March/April 2002
`Pages 273–286
`
`Immunotherapy of multiple sclerosis—Current practice and
`future directions
`
`Mark J. Tullman, MD; Fred D. Lublin, MD; Aaron E. Miller, MD
`The Corinne Goldsmith Dickinson Center for Multiple Sclerosis, Mount Sinai Medical Center, 5 East 98th Street, Box
`1138, New York, NY 10029
`
`Abstract—Over the past decade, multiple sclerosis (MS) has
`become a treatable neurological disease. This paper reviews
`the therapies that have been studied to treat MS and discusses
`various treatment approaches on the horizon. Immunosuppres-
`sive and immunomodulatory therapies have been shown to
`alter the long-term course of MS. Therapies are currently avail-
`able for relapsing-remitting, secondary progressive, and pro-
`gressive relapsing disease. Although effective, these therapies
`have a modest impact on reduction in relapse rate and slowing
`of disease progression. Much work is needed to improve upon
`this modest effect and hopefully obtain a cure.
`
`Key words: disease-modifying agents, immunomodulatory,
`immunosuppressive, immunotherapy, multiple sclerosis, treatment.
`
`INTRODUCTION
`
`In 1993, the first disease-modifying therapy was
`approved in the United States for treatment of multiple
`sclerosis (MS). Since then four additional agents have
`been approved, solidifying MS as a treatable neurological
`disease. Therapy is now available for relapsing-remitting
`(RR), secondary progressive (SP), and progressive relaps-
`ing (PR) MS (see Figure). There are no proven therapies
`
`for primary progressive MS (PPMS). As detailed in the
`following sections, the available therapies for MS are
`modest in their effect on reduction in relapse rate and
`slowing of progression of disease, but nevertheless,
`clearly effective. During the past decade, there have been
`many pilot and pivotal trials in MS, allowing for incre-
`mental improvement in clinical trial design and imple-
`mentation. In addition to refining the clinical scales used
`to assess efficacy, employing many magnetic resonance
`imaging (MRI) metrics has allowed for more rapid
`assessment of treatment effect and a better understanding
`of the underlying immunopathologic process of MS.
`Therapy for MS generally involves either disease-modi-
`fying agents (DMA) (i.e., those that alter the long-term
`course of MS), treatments of acute exacerbations, or symp-
`tomatic therapies. The latter are beyond the scope of this
`paper. The DMA segregate into immunosuppressive thera-
`pies, characterized by relatively nonspecific down-regula-
`tionof immune function and immunomodulatory therapy,
`where the effects on the immune system are more targeted
`and usually less toxic. Currently, the best-studied agents
`have been immunomodulatory.
`
`TREATMENT OF ACUTE ATTACKS
`
`Address all correspondence and requests for reprints to Mark J. Tull-
`man, The Corinne Goldsmith Dickinson Center for Multiple Sclerosis,
`Mount Sinai Medical Center, 5 East 98th Street, Box 1138, New York,
`NY 10029; email: gmtullman@aol.com.
`
`Corticosteroids have anti-inflammatory and immuno-
`suppressive effects. Although they likely do not alter the
`natural course of the disease, corticosteroids shorten the
`duration of an attack and hasten the time to recovery.
`
`273
`
`Merck 2025
`TWi v Merck
`IPR2023-00049
`
`

`

`274
`
`Journal of Rehabilitation Research and Development Vol. 39 No. 2 2002
`
`rent optic neuritis than were the patients in the other two
`groups. Patients treated with IV steroids had a statistically
`significant lower rate of the subsequent development of
`clinically definite MS over a 2-year period than did the
`patients in the other two groups (8). However, this effect
`was temporary and by the end of 3 years, no difference
`existed among the three groups (9).
`Corticosteroids are well tolerated by most patients
`with MS (10). The side effects of short-term pulse doses
`include behavioral disturbances, aseptic necrosis of bone,
`promotion of osteoporosis, risk of infection, and possibly
`peptic ulceration. The long-term risks of pulse doses of
`corticosteroids are unknown. Thus, the risks and benefits
`of corticosteroids need to be considered individually. It is
`perhaps best to treat patients with mild symptoms and
`signs that are not bothersome (e.g., mild sensory symp-
`toms) with a conservative approach.
`The only other treatment shown to be effective for
`acute relapses in MS is plasma exchange (PE). In a small,
`double-blind, placebo-controlled crossover trial by Wein-
`shenker et al., patients with severe attacks of inflamma-
`tory demyelinating disease (all patients had hemiplegia,
`paraplegia, or tetraplegia) who failed to improve after at
`least a 5-day course of high-dose parenteral corticoster-
`oid therapy were randomized to receive either seven
`treatments of PE or sham exchanges over 14 days (11).
`More than 40 percent of patients treated with PE had con-
`siderable improvement of their neurological deficits
`compared with approximately 6 percent of those patients
`who
`received sham exchange. This
`improvement
`occurred early during treatment and was sustained during
`6 months of follow-up. Those that responded were
`treated within an average of 40 days of onset of symp-
`toms. One patient improved after treatment was begun
`61 days after the onset of symptoms. The time from onset
`of symptoms to initiation of treatment was similar in
`those patients that responded to therapy and those that
`failed therapy.
`A reasonable approach to the treatment of acute
`relapses of MS would seem to be a 3- to 7- day course of
`high-dose intravenous corticosteroid therapy. If a patient
`has significant residual disability after 7 days, a trial of
`PE may be warranted.
`
`RR Interferon ß-1a,* Interferon ß-1b, Glatiramer
`Acetate, Mitoxantrone†
`SP Mitoxantrone, Interferon ß-1b‡
`PR Mitoxantrone
`PP None§
`Both 30 μg IM once weekly and 44-μg SC thrice weekly
`have been shown to be effective in RRMS.
`Indicated in patients with worsening RRMS, despite treat-
`ment with interferon ß or glatiramer acetate.
`Two studies of interferon ß–1b in patients with SPMS have
`yielded conflicting results in disease progression.
`Glatiramer acetate and mitoxantrone are currently being
`tested in PPMS.
`RR= relapsing-remitting
`SP = secondary progressive
`PR = progressive relapsing
`PP = primary progressive
`
`* † ‡ §
`
`Figure.
`Successful therapeutic trials in MS by disease type.
`
`They have been the mainstay of treatment for acute exac-
`erbations in MS for more than 30 years (1–4). Whereas
`periodic pulse doses of intravenous (IV) methylpredniso-
`lone are not effective in preventing disability in patients
`with progressive MS, a recent phase II trial concluded
`that they do have an effect on disability in patients with
`RRMS (5,6).
`Although no consensus exists on optimal dose, route,
`or length of therapy, currently, patients with acute attacks
`are often treated with high-dose IV methylprednisolone
`(500–1,000 mg/day) for 3 to 5 days, followed by an oral
`prednisone taper. While some neurologists treat acute
`relapses with oral prednisone, the results of the Optic
`Neuritis Treatment Trial question this practice. In this
`multicenter, blinded, randomized, placebo-controlled trial
`of acute unilateral optic neuritis, 457 patients were ran-
`domized to receive either oral placebo, oral prednisone
`(1 mg/kg) for 14 days or high-dose IV methylpredniso-
`lone for 3 days, followed by 11 days of an oral prednisone
`taper within 8 days of onset of visual symptoms (7).
`Although the vision in patients in the IV steroid- treated
`group improved more rapidly than those of the oral pred-
`nisone- or placebo-treated group, the visual acuity was
`similar in all three groups at 6 months. There was no dif-
`ference in the rate of recovery between the placebo- and
`oral prednisone-treated groups. However, patients treated
`with oral prednisone were more likely to develop recur-
`
`

`

`DISEASE-MODIFYING AGENTS
`
`Immunomodulatory Therapy
`There are four immunomodulatory agents, interferon
`ß-1a (Avonex and Rebif), interferon ß-1b (Betaseron),
`and glatiramer acetate (Copaxone), approved by the Food
`and Drug Administration (FDA) for the treatment of
`RRMS (12–18).
`
`Interferon Beta
`The mechanism of action of interferon ß (IFN ß) in
`MS is unknown. It has a wide range of effects on the
`immune cascade. Potential mechanisms include reduction
`in transport of T cells from the peripheral circulation into
`the central nervous system (CNS) through effects on
`adhesion molecules, chemokines, and matrix metallopro-
`teinases; inhibition of type 1 T-helper cell activation and
`augmentation of suppressor T cell function; and alter-
`ation of cytokine production to favor an anti-inflamma-
`tory state (19).
`Interferon ß-1b (IFN ß-1b) was the first treatment
`shown to have a favorable effect on the natural history of
`MS (12,13). It is produced in Escheria coli and differs
`from human IFN ß by two amino acids. Unlike human
`IFN ß, IFN ß-1b is not glycosylated. In 1993, the FDA
`approved IFN ß-1b for the treatment of RRMS. This
`approval was based on results of a multicenter, double-
`blind, randomized, placebo-controlled trial of 372 patients
`with a baseline Expanded Disability Status Scale (EDSS)
`of 0.0 to 5.5 who had at least two relapses in the previous
`2 years (12,13,20). In this study, patients were randomized
`to receive either placebo, IFN ß-1b 1.6 million interna-
`tional units (MIU) (50 μg), or IFN ß-1b 8 MIU (250 μg)
`subcutaneously (SC) every other day for 2 years. The pri-
`mary end points of the study were a reduction in exacerba-
`tion rate and the percentage of patients remaining
`exacerbation free. At the end of 2 years, the group of
`patients treated with IFN ß-1b 8 MIU had an exacerbation
`rate of 0.84 attacks a year compared with 1.27 attacks a
`year in the group treated with placebo, a statistically sig-
`nificant reduction of approximately 34 percent. At the end
`of 2 years, there was a significantly greater number of
`patients in the IFN-treated group who remained relapse
`free than in the placebo-treated group. However, when the
`study was extended, the difference was no longer signifi-
`cant after 3 years. Patients treated with the IFN ß-1b had a
`longer time to first and second relapse, fewer moderate to
`severe attacks, and required fewer hospitalizations. Con-
`
`TULLMAN et al. Immunotherapy of multiple sclerosis
`
`275
`
`firmed progression of disability had no significant differ-
`ence, as measured by an increase of one or more EDSS
`points that persisted for at least 3 months, between the
`IFN ß-1b and placebo-treated groups. Patients treated with
`IFN ß-1b had a significant reduction in MRI activity and
`burden of disease. The results of various outcome mea-
`sures including exacerbation rate and MRI disease burden
`favored the higher dose treatment group and provided evi-
`dence for a possible dose effect.
`Neutralizing antibodies (NAb) developed in approxi-
`mately 38 percent of the patients treated with IFN ß-1b
`after 2 years (with most developing during the first year
`of treatment) and were associated with a decrease in clin-
`ical and MRI efficacy (12,20). After 18 months, the
`relapse rate of the group of patients treated with IFN ß-1b
`8 MIU who were NAb positive was similar to the group
`of patients receiving placebo (21). However, over time,
`NAb disappeared in the majority of patients, and Rice et
`al. have recently suggested that the measurement of NAb
`may have little clinical use and the treatment decisions
`should be made exclusively on clinical grounds (22).
`In
`the Independent Comparison of Interferon
`(INCOMIN) trial, IFN ß-1b 8 MIU given SC every other
`day and IFN ß-1a (Avonex) 6 MIU once weekly were
`compared in patients with RRMS (23). After 6 months
`of treatment with IFN, the two had similar clinical
`effects. However, after 1 year, IFN ß-1b was superior in
`various unblinded clinical outcomes, including relapse
`rate and sustained EDSS progression. Similarly, MRI
`results (evaluated in a single-blind manner) favored the
`IFN ß-1b-treated group. The recently presented 2-year
`results showed a similar outcome.
`IFN ß-1b has also been shown effective in SPMS
`(24). In a multicenter, double-blind, randomized, placebo-
`controlled European trial, 718 patients with SPMS (EDSS
`3.0 to 6.5) were randomized to receive either IFN ß-1b 8
`MIU or placebo SC every other day for up to 3 years. The
`primary outcome was time to confirmed progression of
`disability measured by a sustained increase of at least 1.0
`point on the EDSS (0.5 point if the EDSS was ≥ 6.0). The
`mean follow-up for both groups was approximately 900
`days. In this study, the group treated with IFN ß-1b had an
`approximate 22-percent reduction in the time to progres-
`sion with an average delay of 9 to 12 months. During the
`study period, nearly one-third fewer patients in the IFN ß-
`1b-treated group became wheelchair-bound compared
`with the placebo-treated group. In addition, patients in the
`IFN ß-1b-treated group had a significant reduction in
`
`

`

`276
`
`Journal of Rehabilitation Research and Development Vol. 39 No. 2 2002
`
`relapse rate, had longer time to first relapse, required
`fewer hospitalizations, and had less steroid use. Positive
`results were also seen for MRI analysis (25). A second
`study of IFN ß-1b in SPMS was performed in North
`America and was recently reported but not yet published
`(26). In this study, SPMS patients were treated with either
`placebo, IFN ß-1b 8 MIU SC every other day, or IFN ß-
`1b 5 MIU/m² (this group received an average dose of 9.6
`MIU) subcutaneously every other day. As opposed to the
`European study, this study showed no effect on the pri-
`mary outcome assessment of time to confirmed progres-
`sion of disability. Similar to the European study, other
`outcome measures, such as reduction in relapse rate and
`MRI activity and lesion load, favored the treatment
`group.
`IFN ß-1b is currently approved by the FDA in ambu-
`latory patients with RRMS to reduce the frequency of
`relapses. The usual dose is 8 MIU (250 μg) SC every
`other day.
`IFN ß-1a (Avonex) is produced in mammalian cell
`culture and is structurally identical to human IFN ß. The
`pivotal trial of Avonex was a multicenter, double-blind,
`randomized, placebo-controlled trial of 311 patients who
`had RRMS with a baseline EDSS of 1.0 to 3.5 and had at
`least two exacerbations in the previous 3 years (14). These
`patients were randomized to receive Avonex 6 MIU
`(30 μg) or placebo via intramuscular (IM) injection once
`weekly. As opposed to the trials of the other interferons
`and glatiramer acetate, the primary outcome measure was
`time to sustained progression of at least 1.0 point on the
`EDSS. After 2 years, there was a significant treatment
`effect. Approximately 35 percent of patients in the pla-
`cebo-treated group had sustained progression of disability
`compared with approximately 22 percent of patients in the
`Avonex-treated group. Several clinical and MRI measures
`served as secondary outcome events. For patients who
`received Avonex for 2 years, the annual exacerbation rate
`was reduced by 32 percent from 0.9 to 0.61. However,
`intent to treat analysis only demonstrated an 18-percent
`reduction in relapse rate. There was a significant reduction
`in the mean number and volume of gadolinium-enhancing
`lesions in the Avonex-treated group. After 1 year, the MRI
`total volume of T2-weighted lesions was significantly
`reduced in the IFN-treated group. However, by the end of
`2 years, no significant difference existed between the two
`groups. Neutralizing antibodies developed in 22 percent of
`patients after 2 years.
`
`There is ongoing debate as to when to initiate pro-
`phylactic therapy in patients with MS. The current prac-
`tice guidelines from the National MS Society state that
`initiation of therapy with an immunomodulatory agent
`should begin as soon as possible following a definite
`diagnosis of MS and determination of a relapsing course
`(27). In the recent CHAMPS study, a multicenter, dou-
`ble-blind, randomized, placebo-controlled trial involving
`383 patients, it was shown that initiating treatment with
`Avonex at the time of a first clinical demyelinating event
`(optic neuritis, incomplete transverse myelitis, brain
`stem, or cerebellar syndrome) in patients with MRI evi-
`dence of prior subclinical demyelination (defined as two
`or more clinically silent lesions greater than 3 mm in
`diameter, one of which had to be periventricular or
`ovoid) is beneficial in reducing the probability of devel-
`oping clinically definite MS (28). Over 3 years, the rate
`of developing clinically definite MS was 44 percent
`lower in the group of patients treated with Avonex than
`the group of patients treated with placebo. However, at
`the end of 3 years, 50 percent of the placebo-treated
`patients did not have a second acute demyelinating event.
`Thus, although the course of MS is unpredictable, treat-
`ment may be unnecessary in a significant proportion of
`patients after a single clinical demyelinating event (i.e.,
`before a diagnosis of clinically definite MS).
`Currently, the FDA has approved the Avonex form of
`IFN ß-1a for relapsing forms of MS to slow the accumu-
`lation of disability and to reduce the rate of relapses. The
`approved dose is 6 MIU (30 μg) IM weekly.
`IFN ß-1a (Rebif) is produced in Chinese hamster
`ovary cells and is structurally identical to human IFN ß
`and Avonex. It was tested in Europe and Canada in a
`multicenter, double-blind, randomized, placebo-con-
`trolled study involving 560 patients with RRMS (EDSS
`0.0 to 5.0) with at least two relapses in the previous
`2 years (15,16). Patients were randomized to receive pla-
`cebo, Rebif 6 MIU (22 μg), or Rebif 12 MIU (44 μg) SC
`three times a week for 2 years. The primary outcome
`measure of the number of relapses over 2 years was
`obtained with a reduction of 33 percent in the Rebif
`12 MIU-treated group and a 27 percent reduction in the
`Rebif 6 MIU-treated group. Patients in both treatment
`groups performed significantly better than the placebo
`group with respect to a number of moderate to severe
`relapses, time to first relapse, percentage of patients
`relapse free, percentage of patients requiring steroid use,
`and time to sustained progression of disability. MRI
`
`

`

`analysis revealed a significant decrease in disease burden
`and fewer gadolinium-enhancing lesions in both treat-
`ment groups compared to the placebo group. Similar to
`what was seen in the IFN ß-1b RRMS trial, a dosing
`effect was suggested with the lower-dose treatment group
`having intermediate results in clinical and MRI out-
`comes. After 2 years, the study was extended an addi-
`tional 2 years with the placebo group crossed over and
`randomized to receive either IFN ß-1a 6 MIU or 12 MIU
`three times a week (29). The treatment effect on clinical
`and MRI measures was maintained during the study for
`both doses of Rebif, with the higher dose group receiving
`the most benefit. Results were superior for patients
`treated with Rebif for all 4 years than for patients in the
`crossover groups, suggesting that early treatment may be
`more beneficial. After 2 years, NAb developed in
`roughly 24 percent of the low-dose treatment group and
`12.5 percent of the high-dose treatment group, and their
`presence did not affect the mean number of relapses (15).
`After 4 years, the proportion of patients that developed
`persistent NAb was similar, but their presence was asso-
`ciated with reduced clinical and MRI efficacy (29).
`In the Evidence for Interferon Dose-Response: Euro-
`pean-North American Comparative Efficacy (EVIDENCE)
`trial, the two forms of IFN ß-1a (Avonex 6 MIU IM weekly
`versus Rebif 12 MIU three times per week) were compared
`in patients with RRMS in a multicenter, evaluator-blinded
`study (30). The 24-week data (the only results published
`thus far) demonstrated a superior effect of Rebif on relapse
`rate, time to first exacerbation, proportion of patients
`relapse-free, and steroid use. MRI outcomes also favored
`the Rebif-treated group.
`Rebif was also tested in SPMS in a multicenter, dou-
`ble-blind, randomized, placebo-controlled trial of 618
`patients (31,32). Although MRI outcomes and relapse
`rate in the Rebif-treated groups had positive results, the
`primary outcome measure of time to confirmed progres-
`sion of disability was not significantly prolonged.
`Based on the data from the EVIDENCE trial, the
`FDA approved Rebif for use in RRMS in March 2002.
`The usual dose is 12 MIU (44 μg) three times per week.
`The main side effects of the interferons are flu-like
`symptoms, injection site reactions, and laboratory abnor-
`malities. The flu-like symptoms (e.g., fever, chills, and
`myalgia) usually occur several hours after injection and
`often resolve within 24 hours. They can be especially
`problematic in the first few weeks after the introduction
`of treatment and may lead to noncompliance and even
`
`TULLMAN et al. Immunotherapy of multiple sclerosis
`
`277
`
`discontinuation of the drug. However, they generally
`resolve within a few months of initiation of therapy, and
`by the end of 1 year, such symptoms are reduced mark-
`edly (20). The flu-like symptoms can often be managed
`with nonsteroidal anti-inflammatory medications or ace-
`taminophen and/or a gradual increasing dose escalation.
`Interferon is frequently given at night to limit side effects
`during wakefulness. All the interferons have been associ-
`ated with injection site reactions that may be as mild as
`local redness to rarely as severe as skin necrosis requiring
`surgical debridement. Skin necrosis is much less com-
`mon with Rebif than Betaseron (12,15). There have been
`no reports of skin necrosis associated with the use of
`Avonex. Injection site reactions can be reduced by proper
`injection preparation and technique.
`A mild increase in liver enzymes, thrombocytopenia,
`anemia, or leukopenia may be seen shortly after initiation
`of therapy with the various interferons and usually
`returns to near baseline by 4 months. However, labora-
`tory abnormalities may develop at any time during ther-
`apy. A complete blood count and liver function tests
`should be obtained before starting therapy and monitored
`during the course of treatment (4 to 6 weeks after initiat-
`ing treatment, at 3 months, and every 3 to 6 months there-
`after). Patients taking other medications that can cause
`hepatotoxicity or myelosuppression require more careful
`laboratory monitoring.
`The interferons may possibly cause or worsen
`depressive symptoms (12,20,29). MS patients have a
`high prevalence of depression and suicidal ideation
`(33–35). Patients on interferon should be evaluated rou-
`tinely for symptoms of depression. IFN ß-1b causes
`menstrual irregularities in some women. No drugs are
`known to interact with the interferons. They are con-
`traindicated in patients with hypersensitivity to the
`drug. Women who are pregnant, trying to become preg-
`nant, or are lactating should not use interferons.
`
`Glatiramer Acetate (Copaxone)
`Glatiramer Acetate (GA) is a synthetic polypeptide
`composed of four amino acids, L-alanine, L-glutamic
`acid, L-lysine, and L-tyrosine. Its mechanism of action is
`unknown but may be related to its capability to enhance
`suppressor T cells or to act as an altered peptide ligand
`(36).
`In a pivotal multicenter, double-blind, randomized,
`placebo-controlled trial, 251 patients with RRMS with an
`EDSS of 0.0 to 5.0 and who had at least two relapses in
`
`

`

`278
`
`Journal of Rehabilitation Research and Development Vol. 39 No. 2 2002
`
`the previous 2 years were randomized to receive either
`placebo or GA 20 mg SC daily for 2 years (17). The pri-
`mary outcome measure of a reduction in relapse rate was
`achieved with a 29-percent reduction from 0.84 in the
`placebo-treated group to 0.59 in the GA-treated group.
`The sustained progression of disability had no significant
`difference (as defined by an increase of at least 1.0 point
`on the EDSS maintained for more than 90 days) between
`the two groups. A subsequent MRI study demonstrated
`that when compared with placebo, GA significantly
`reduced the number of new T2-weighted and gadolin-
`ium-enhancing
`lesions
`(18). However,
`this effect
`occurred more slowly and less intensively than seen with
`IFN ß, suggesting that GA exerts its beneficial effect by a
`different mechanism of action than IFN ß. A large trial of
`GA in PPMS is currently underway (37).
`GA is generally well tolerated, and the most common
`side effects noted during the trial were injection site reac-
`tions and a systemic reaction. The injection site reaction
`consisted of mild redness and induration at the injection
`site. There have been no reports of skin necrosis associ-
`ated with its use. An unpredictable, sporadic transient
`systemic reaction occurred in 15 percent of patients
`receiving GA in the trial. This reaction consisted of a
`combination of chest tightness, flushing, shortness of
`breath, palpitations, and anxiety. It occurred within min-
`utes of injection and was self-limited, lasting for seconds
`to minutes and resolving spontaneously without sequelae.
`In most patients, it occurred only once. GA does not
`cause anemia, leukopenia, or liver enzyme abnormalities,
`and routine laboratory studies are not necessary. GA does
`not cause depressive symptoms. No drugs are known to
`interact with GA. It is contraindicated in patients with
`hypersensitivity to the drug or to mannitol. Women who
`are pregnant or who are lactating should not use GA.
`Neutralizing antibodies do not develop against GA. GA
`is currently FDA approved in patients with RRMS for the
`reduction of relapses. The standard dose is 20 mg SC
`every day.
`Although they certainly are not a cure with a relapse
`reduction of approximately one-third, the DMA that have
`become available in the past decade have provided MS
`patients with hope for a brighter future. According to the
`National MS Society Disease Management Consensus
`Statement, therapy should be continued indefinitely,
`unless a benefit is clearly lacking, side effects are intoler-
`able, new data that reveal other reasons for cessation, or
`better therapy becomes available (27).
`
`IMMUNOSUPPRESSIVE THERAPIES
`
`Mitoxantrone (Novantrone)
`Mitoxantrone is a synthetic anthracendione that inter-
`calates into deoxyribonucleic acid (DNA), producing
`cross- links and strand breaks. It also interferes with ribo-
`nucleic acid (RNA) synthesis. It is indicated to treat pain
`related to advanced hormone refractory prostate cancer
`and as initial therapy of acute nonlymphocytic leukemia.
`Several small randomized, controlled trials suggested
`that mitoxantrone may have a beneficial effect on clinical
`and MRI measures in patients with MS (38–40). These
`results were confirmed in a larger multicenter, observer-
`blind, randomized, placebo-controlled trial in patients
`with severe RRMS, SPMS, or PRMS (41,42). In this
`trial, 194 patients were randomized to receive placebo,
`mitoxantrone 5 mg/m², or mitoxantrone 12 mg/m² intra-
`venously every 3 months for 2 years. A cohort of 110
`patients received annual MRI scans. Mitoxantrone signif-
`icantly reduced the mean change in EDSS; EDSS
`decreased by 0.12 and 0.23 in mitoxantrone 12 mg/m²
`and 5 mg/m² treated groups, respectively, and increased
`by 0.23 in patients in the placebo group. There were also
`significant differences favoring mitoxantrone on various
`clinical measures, including the number of relapses,
`ambulatory impairment, and proportion of patients with
`confirmed EDSS progression. Patients in the higher dose
`mitoxantrone group had superior results in most out-
`comes. On MRI analysis, mitoxantrone significantly
`reduced the number of gadolinium-enhancing lesions and
`the average number of new T2-weighted lesions.
`The major toxicities of mitoxantrone are bone-mar-
`row suppression and dose-related cardiotoxicity (reduced
`left ventricular ejection fraction (LVEF) and irreversible
`congestive heart failure (CHF)). Treatment with mitox-
`antrone is limited to a cumulative lifetime dose of
`140 mg/m² (2 to 3 years in most people). Evaluation of
`LVEF is recommended before therapy is initiated.
`According to the mitoxantrone product insert, MS
`patients with LVEF that is less than 50 percent ordinarily
`should not receive mitoxantrone (43). Further assessment
`of LVEF is recommended in patients who develop symp-
`toms of CHF and before all doses in patients who have
`received a cumulative dose greater than 100 mg/m². Other
`side effects include alopecia, nausea, menstrual irregular-
`ities, and elevated liver enzymes. A complete blood count
`and liver function tests should be monitored before each
`dose. Mitoxantrone is contraindicated in patients with
`
`

`

`hypersensitivity to the drug. It should not be used during
`pregnancy or lactation.
`In 2000, the FDA approved mitoxantrone for the
`treatment of SPMS, PRMS, and worsening RRMS. The
`recommended dose is 12 mg/m² intravenously every
`3 months. This agent is currently being tested in PPMS
`(44).
`
`Azathioprine (Imuran)
`Azathioprine is an imidazolyl derivative of 6-mer-
`captopurine that impairs DNA and RNA synthesis. It is
`used in a variety of autoimmune disorders, including
`myasthenia gravis. In a meta-analysis of seven random-
`ized, blind, controlled trials with 793 patients with all
`forms of MS, azathioprine was shown to significantly
`reduce relapse rate at 1, 2, and 3 years (45). At 1 year, the
`change in EDSS was similar in the treated and control
`groups. After 2 years, there was a trend toward slowing
`disease progression favoring the azathioprine-treated
`group. A small retrospective study analyzed MRI lesion
`load in patients with RRMS (46). This study compared
`two serial MRI scans (mean interval between scans was
`2.5 years) of patients treated with azathioprine and ste-
`roids after acute relapses with the scans of patients
`treated with steroids alone. A significant reduction in
`MRI lesion load was seen in the group treated with aza-
`thioprine and steroids.
`Toxicities of azathioprine include bone marrow sup-
`pression, nausea, vomiting, and liver enzyme elevations.
`An increased risk of cancer may possibly exist with long-
`term use of azathioprine (47). It should not be used dur-
`ing pregnancy.
`Overall, azathioprine has been shown to have a mod-
`est effect on relapses but no convincing effect on progres-
`sive disease. Despite this, azathioprine is probably the
`most common cytostatic agent used in progressive MS,
`based primarily on desperation and anecdotal reports.
`
`Cladribine (Leustatin)
`Cladribine
`is an adenosine deaminase-resistant
`purine-nucleoside analogue. It is an immunosuppressant
`that preferentially targets lymphocytes. It is used in the
`treatment of hairy cell leukemia.
`Several small, randomized, double-blind, placebo-
`controlled studies suggested that cladribine may have a
`favorable effect on clinical and MRI outcomes in patients
`with RRMS and progressive MS (48–50). Subsequently,
`cladribine was studied in a larger, randomized, double-
`
`TULLMAN et al. Immunotherapy of multiple sclerosis
`
`279
`
`blind, placebo-controlled trial in patients with progres-
`sive MS (51). This poorly designed study did not demon-
`strate any clinical benefit after 1 year but did show a
`marked reduction in gadolinium-enhancing lesions in the
`cladribine-treated group. Side effects of cladribine
`include
`long-term bone-marrow suppression, fever,
`fatigue, nausea, and diarrhea. It should not be used during
`pregnancy.
`
`Cyclophosphamide (Cytoxan)
`Cyclophosphamide is an alkylating agent with cyto-
`toxic and anti-inflammatory effects that is used to treat
`neoplastic and autoimmune disorders. Several studies of
`cyclophosphamide in patients with MS have yielded con-
`flicting results (52–54). Comparison between trials is
`always hazardous, and various induction protocols have
`been used, some with the addition of steroids and/or plas-
`mapheresis. The Canadian Cooperative Study provides
`the strongest evidence for its lack of efficacy (55). In this
`multicenter, single-blind, placebo-controlled trial, 168
`patients with progressive MS were randomized to receive
`intravenous cyclophosphamide and oral prednisone; oral
`cyclophosphamide, oral prednisone, and weekly PE; or
`placebo medications and sham exchange. Patients were
`followed for an average of 2.5 years. No significant dif-
`ferences existed between the three groups in the primary
`outcome of progression of disease. Nevertheless, there
`are many anecdotes of success leading to use of this
`agent in desperate cases. Toxicities of cyclophosphamide
`include alopecia, nausea, vomiting, hemorrhagic cystitis,
`sterility, and malignancy. The drug is teratogenic and
`should not be used during pregnancy.
`
`Cyclosporine (Sandimmune)
`Cyclosporine is a cyclic polypeptide with potent
`immunosuppressive properties. It appears to suppress T-
`helper cells. Its main use is in transplant recipients. In a
`large multicenter, double-blind, placebo-controlled trial,
`557 patients with progressive MS were randomi

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