throbber

`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`The New England Journal of Medicine
`
`Review Article
`
`Drug Therapy
`
`A
`
`L A S T A I R
`
` J.J. W
`
`O O D
`
`, M.D.,
`
` Editor
`
`M
`
` M
`
`OF
`ANAGEMENT
`S
`CLEROSIS
`
`ULTIPLE
`
`
`
` A. C
`, M.D., J
` A. R
`R
`OHEN
`EFFREY
`UDICK
`ICHARD
` W
`-G
`, M.D.,
`B
`IANCA
`EINSTOCK
`UTTMAN
`R
` P. K
`, M.D.,
`EVERE
`INKEL
` R
` M. R
`, M.D.
`ICHARD
`ANSOHOFF
`
`AND
`
`, M.D.,
`
`M
`
`ULTIPLE SCLEROSIS is a common dis-
`ease of the central nervous system affect-
`ing approximately 1 million young adults,
`mostly women, worldwide.
` It is characterized by
`1
`episodic neurologic symptoms that are often fol-
`lowed by fixed neurologic deficits, increasing disabil-
`ity, and medical, socioeconomic, and physical decline
`over a period of 30 to 40 years.
`For most of the 20th century, multiple sclerosis
`was considered untreatable. In 1982, the Multiple
`Sclerosis Society of Canada and the National Multiple
`Sclerosis Society of the United States sponsored the
`first international workshop on therapeutic trials.
`2
`This workshop served to usher in an era of activism
`and optimism that has substantially replaced wide-
`spread therapeutic nihilism and skepticism about the
`feasibility of clinical trials in multiple sclerosis.
`There have been a number of important advances
`since the international workshop.
` The Expanded
`3,4
`Disability Status Scale achieved widespread use as a
`single measure of the severity of multiple sclerosis.
`5
`Magnetic resonance imaging (MRI) was invented,
` and quickly established
`applied to multiple sclerosis,
`6
`as a sensitive marker of the pathologic process. Large
`multicenter clinical trials were completed,
` and
`7-14
` The
`monographs on clinical trials were published.
`15,16
`Food and Drug Administration (FDA) approved in-
`terferon beta-1a (Avonex, Biogen, Cambridge, Mass.),
`interferon beta-1b (Betaseron, Berlex Laboratories,
`Richmond, Calif.), and glatiramer acetate (Copax-
`one, Teva Marion Partners, Kansas City, Mo.) for pa-
`
`From the Mellen Center for Multiple Sclerosis Treatment and Research,
`Department of Neurology, Cleveland Clinic Foundation, Cleveland, OH
`44106, where reprint requests should be addressed to Dr. Rudick.
`©1997, Massachusetts Medical Society.
`
`1604
`
`ⴢ
`
`November 27, 1997
`
`tients with relapsing–remitting multiple sclerosis. As
`a result of these advances, effective therapies are now
`available, and clinical trials of other promising thera-
`pies are under way.
`
`DISEASE CHARACTERISTICS
`RELATED TO TREATMENT DECISIONS
`The goal of therapy in patients with multiple scle-
`rosis is to prevent relapses and progressive worsening
`of the disease. Spontaneous recovery is rare when
`neurologic deficits have persisted for longer than six
`months, and there are no known therapies that pro-
`mote regeneration and reverse fixed neurologic def-
`icits. Therefore, disease-modifying therapy should
`be considered before neurologic deficits have per-
`sisted longer than six months. Decisions in individ-
`ual patients should be based both on the course of
`the patient’s disease and on the probability of severe
`disabling disease.
`A standardized nomenclature to describe the
`course of multiple sclerosis (Table 1) was developed
`by consensus.
` The most common pattern at onset
`17
`is relapsing–remitting disease, but it becomes sec-
`ondary progressive disease over time in more than 50
`percent of patients. Approximately 10 percent of pa-
`tients have primary progressive multiple sclerosis.
`They tend to be older at onset (40 to 60 years of age)
`and commonly have a progressive myelopathy. Pa-
`tients with primary progressive multiple sclerosis
`have fewer gadolinium-enhanced lesions on cranial
`MRI scans and fewer inflammatory changes in cere-
`brospinal fluid than patients with secondary progres-
`sive multiple sclerosis.
` Progressive relapsing multi-
`18
`ple sclerosis is a very uncommon pattern of disease.
`The vast majority of patients have relapsing–remit-
`ting multiple sclerosis during the early years and sec-
`ondary progressive multiple sclerosis later. Patients
`with relapsing–remitting multiple sclerosis have the
`best responses to treatment, whereas patients with
`progressive disease are less responsive to treatment.
`Disease-modifying therapy should be considered
`early in the course for patients with an unfavorable
`prognosis. The unfavorable prognostic markers relat-
`ed to more rapid worsening of disease that are listed
`in Table 2 can be used to select patients for treat-
`ment.
` Patients who have multiple cranial MRI le-
`19-22
`sions at the time of their first symptoms are much
`more likely to have major disability later on.
` There-
`23
`fore, in addition to the clinical features, the findings
`on cranial MRI are useful in selecting patients for ear-
`ly treatment. Approximately 10 percent of patients
`have relatively benign disease, however, so not every
`patient should receive disease-modifying therapy.
`
`Hopewell EX1017
`
`1
`
`

`

`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`DRUG THERAPY
`
`DESIGN OF CLINICAL TRIALS
`
`Relation between Treatment Strategies and Pathogenesis
`Various lines of research support the hypothesis
`that multiple sclerosis is an autoimmune process oc-
`curring in genetically susceptible persons after an en-
`vironmental exposure. The geographic heterogeneity
`of the disease and the widely varying prevalence rates
`in different ethnic populations suggest interplay be-
`tween environmental and genetic factors. The obser-
`vation that common viral infections can precipitate
`relapses led to the concept that viruses can trigger
`autoimmune demyelination in susceptible persons.
`24
`No specific pathogen has been reliably linked to mul-
`tiple sclerosis, however, so none of the current treat-
`ment approaches are targeted at microbial patho-
`gens. The children of patients with multiple sclerosis
`have an increased risk (30-fold to 50-fold) of multi-
` Studies of twins and adopted children
`ple sclerosis.
`25
`suggest that the increased risk is largely genetic.
`26
`Candidate-gene and whole-genome screening sug-
`gests that multiple weakly acting genes interact to
`determine the risk of multiple sclerosis.
`27-29
`Most current therapeutic approaches are based on
`the hypothesis that multiple sclerosis is an organ-
`specific autoimmune disease. Inoculation of suscep-
`tible animals with myelin proteins results in a relaps-
`ing–remitting, inflammatory, demyelinating central
`nervous system disease called experimental autoim-
`mune encephalomyelitis. Experimental autoimmune
`encephalomyelitis can be transferred to unimmu-
`nized animals through activated T cells that recog-
`nize small fragments of myelin proteins.
` These
`30
`pathogenic T cells use a restricted array of genes for
`T-cell–antigen receptors. Molecular strategies that in-
`terrupt the interaction between myelin protein pep-
`tides and pathogenic T-cell receptors are effective in
`acute experimental autoimmune encephalomyelitis.
`31
`Human T cells that recognize myelin antigens also
`have restricted use of T-cell receptors, prompting at-
`tempts to eliminate pathogenic T cells with antibod-
` To date, however, evidence of a
`ies or vaccination.
`32
`unique immunologic abnormality in patients with
`multiple sclerosis is lacking. In particular, T cells that
`recognize myelin can be isolated with similar fre-
`quencies from patients with multiple sclerosis and
`normal subjects. Furthermore, as autoimmune dis-
`eases progress, self-peptides are released from the
`target organ, increasing the diversity of the T-cell re-
`sponse. This phenomenon, termed “epitope spread-
`ing,” occurs in animals with chronic experimental
`autoimmune encephalomyelitis
` and in patients
`33,34
`with multiple sclerosis
` and may limit simple treat-
`35
`ment strategies based on blocking the recognition
`of autoantigen.
`The lesions of multiple sclerosis resemble those in-
`duced by delayed hypersensitivity, containing inflam-
`matory cytokines, activated T cells, and mononuclear
`
` C
`T
` 1.
`ABLE
`
` C
`
`LINICAL
`ATEGORIES
`
`OF
`
` M
` S
`.
`ULTIPLE
`CLEROSIS
`
`D
`ISEASE
`
` C
`ATEGORY
`
`Relapsing–remitting
`
`Secondary progressive
`
`Primary progressive
`
`Progressive relapsing
`
`D
`EFINITION
`
`Episodes of acute worsening with recovery and
`a stable course between relapses
`Gradual neurologic deterioration with or with-
`out superimposed acute relapses in a patient
`who previously had relapsing–remitting mul-
`tiple sclerosis
`Gradual, nearly continuous neurologic deterio-
`ration from the onset of symptoms
`Gradual neurologic deterioration from the on-
`set of symptoms but with subsequent super-
`imposed relapses
`
`T
`
` P
` T
` P
` M
`
` 2.
`REDICT
`HAT
`ROGNOSTIC
`ARKERS
`ABLE
`M
` S
` M
` S
`.
`CLEROSIS
`ORE
`EVERE
`ULTIPLE
`
`Progressive disease from the onset of symptoms
`Motor and cerebellar signs at presentation to neurologist
`Short interval between the first two relapses
`Poor recovery from relapse
`-weighted MRI at presentation
`Multiple cranial lesions on T
`2
`
`phagocytes.
` These elements, shown in Figure 1,
`36,37
`are all potential targets for intervention. Function-
`related T-cell surface molecules can be down-regulat-
`ed with antibodies. Cytokine-based therapies, such as
`those involving soluble receptors for tumor necrosis
`factor
` or immunosuppressive cytokines such as
`a
`transforming growth factor
` or interleukin-10, may
`b
`potentially be effective. The inflammation of the cen-
`tral nervous system may also be sensitive to interven-
`tion directed against leukocyte and cerebrovascular
`endothelial adhesion molecules or chemokines, which
`mediate the migration of leukocytes into the central
`nervous system. In both multiple sclerosis and exper-
`imental autoimmune encephalomyelitis, myelin anti-
`bodies are concentrated in the central nervous sys-
`tem, and demyelinating antibodies in experimental
`autoimmune encephalomyelitis synergize with T-cell–
` Pathogenic antibodies are also
`effector mechanisms.
`47
`potential therapeutic targets.
`
`Controlled Clinical Trials
`Therapeutic advances in multiple sclerosis are de-
`pendent on clinical trials because of the highly vari-
`able and unpredictable course of the disease and the
`difficulty in precisely measuring neurologic disability.
`The Expanded Disability Status Scale,
` the most wide-
`5
`ly used outcome measure in clinical trials of multiple
`sclerosis, is an ordinal rating scale ranging from 0 to
`10, in increments of 0.5, with higher scores reflecting
`increasing severity. The fact that there is a single
`score for each patient at each time point makes study
`design and statistical analysis relatively simple, but
`
`Volume 337 Number 22
`
`ⴢ
`
`1605
`
`2
`
`

`

`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`The New England Journal of Medicine
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`Systemic
`Circulation
`
`Blood–Brain
`Barrier
`
`Central Nervous
`System
`
`Recruitment of cells
`
`Myelin injury
`
`Chemokines
`
`T cell
`
`Activation by
`superantigens, molecular
`mimicry, or unknown
`mechanisms
`
`Astrocyte
`
`Microglia
`
`Activation of
`endothelium
`
`Cytokines
`
`4
`
`1
`
`2
`
`1
`
`Conduction block
`
`3
`
`Activated
`autoreactive T cell
`
`Autoreactive
`T cell
`
`Antigen-presenting cell
`
`Figure 1.
` Pathogenesis of Multiple Sclerosis.
` or unknown mechanisms.
` molecular mimicry,
`Circulating autoreactive T cells are activated by stimulation with superantigens,
`39
`38
`Once activated, these autoreactive cells traverse the blood–brain barrier to enter the central nervous system. Perivascular antigen-
`presenting cells provide the signals necessary to result in the activation and clonal expansion of these autoreactive T cells and the
`secretion of proinflammatory cytokines by them. The cytokines, including tumor necrosis factor and interferon-
`, induce astrocytes
`g
`and leukocytes to secrete chemokines
` and stimulate the expression of adhesion molecules by endothelial cells. Activated micro-
`40
`glia and macrophages damage myelin internodes.
` Proinflammatory cytokines may directly inhibit nerve conduction, leading to
`41
`neurologic dysfunction. Immunosuppressive cytokines (not shown) inhibit the inflammatory process, leading to neurologic recov-
`ery. The putative mechanisms of action of the therapeutic effects of interferon beta, as indicated by the numbers, include inhibition
`of the proliferation of autoreactive T cells (1)
`; inhibition of the expression of major-histocompatibility-complex class II molecules,
`42
`43
`leading to reduced antigen presentation within the central nervous system (2); inhibition of metalloproteases,
` leading to re-
`44,45
`duced migration of T cells into and through the central nervous system (3); and induction of immunosuppressive cytokines,
` lead-
`46
`ing to resolution of the inflammatory process (4).
`
`the minimal changes in scores for some patients over
`long intervals and the subjectivity in making the clin-
`ical ratings limit the value of the scale. The usefulness
`of the Expanded Disability Status Scale has been im-
`proved by the addition of a definition of treatment
`failure as sustained worsening of a clinically impor-
` An effort is under way to develop im-
`tant amount.
`48
`proved clinical outcome measures,
` which could de-
`49
`crease the required sample sizes or shorten the
`duration of multiple sclerosis trials.
`Serial MRI studies have shown that new gadolin-
`ium-enhanced lesions are 5 to 10 times as common
`
`as clinical relapses.
` Preliminary evidence of the ef-
`50
`ficacy of treatments on the basis of MRI findings
`will probably serve as the basis for future trials, and
`all will include serial MRI as an important secondary
`outcome measure.
`
`RELAPSING MULTIPLE SCLEROSIS
`
`Corticosteroids
`Corticosteroids are the mainstay of treatment for
`acute relapses of multiple sclerosis. Corticosteroids
`have immunomodulatory and antiinflammatory ef-
`
`1606
`
`ⴢ
`
`November 27, 1997
`
`3
`
`

`

`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`DRUG THERAPY
`
`fects that restore the blood–brain barrier, reduce
`edema, and may possibly improve axonal conduc-
`tion. Corticosteroid therapy shortens the duration
`of the relapse and accelerates recovery, but whether
`the overall degree of recovery is improved or the
`long-term course is altered is not known.
` Corti-
`51-53
`cotropin was demonstrated to help recovery from re-
` but it has been largely replaced by high-dose
`lapse,
`52
`intravenous methylprednisolone, because the latter
`has a more rapid onset of action, produces more
`consistent benefits, and has fewer side effects.
`52,54
`For moderate-to-severe relapses, 1000 mg of meth-
`ylprednisolone per day by intravenous infusion for
`3 to 5 days followed by 60 mg of oral prednisone
`per day, with tapering of the dose over a period of
`12 days, accelerates neurologic recovery.
`In the Optic Neuritis Treatment Trial, 457 patients
`with acute optic neuritis were randomly assigned to
`receive 1000 mg of intravenous methylprednisolone
`per day for 3 days followed by 1 mg of oral predni-
`sone per kilogram of body weight per day for 11
`days; 1 mg of oral prednisone per kilogram per day
`for 14 days; or oral placebo. The rate of recovery of
`vision was significantly faster in the intravenous-
`methylprednisolone group, with the greatest bene-
`fits in patients with visual acuity of 20/50 or worse
` but there were no significant differences
`at entry,
`9
`between groups in visual outcome at six months.
`Prednisone therapy increased the risk of new epi-
`sodes of optic neuritis in either eye, and intravenous
`methylprednisolone reduced by approximately 50
`percent the risk of an attack leading to the diagnosis
`of multiple sclerosis during the two-year follow-up.
`55
`This effect was most evident in patients at highest
`risk for subsequent relapse — those with multicen-
`tric brain lesions on MRI at entry into the study. Af-
`ter three years, differences between the treatment
`groups were no longer significant,
` suggesting that
`56
`intravenous methylprednisolone delayed but did not
`stop the development of multiple sclerosis after op-
`tic neuritis. These results have led to the widespread
`use of intravenous methylprednisolone for patients
`with optic neuritis and abnormal findings on MRI
`of the brain. The results also renewed debate over
`whether intravenous methylprednisolone has long-
`term benefits for patients with multiple sclerosis. A
`clinical trial is under way to determine whether
`pulsed doses of intravenous methylprednisolone giv-
`en every other month slow disease progression in
`patients with moderate disability and secondary pro-
`gressive multiple sclerosis.
`
`Interferon Beta
`Interferon beta is the treatment of choice for pa-
`tients with relapsing–remitting multiple sclerosis.
`Two forms of recombinant interferon beta — 1a and
`1b — have been approved by the FDA and Europe-
`an regulatory agencies. Interferon beta-1a is a glyco-
`
`sylated, recombinant mammalian-cell product, with
`an amino-acid sequence identical to that of natural
`interferon beta. Interferon beta-1b is a nonglycosy-
`lated recombinant bacterial-cell product in which
`serine is substituted for cysteine at position 17.
`Interferon beta-1b was tested in a multicenter trial
`involving 372 patients with relapsing–remitting mul-
`tiple sclerosis and mild-to-moderate disability. Treat-
`ment consisted of either 8 million units (250
`g) or
`m
`1.6 million units (50
`g) of interferon beta-1b or pla-
`m
`cebo given by subcutaneous injection every other day
`for up to five years. As compared with treatment with
`placebo, treatment with the higher dose reduced the
`relapse rate by 31 percent, increased the proportion
`of patients who were relapse-free (27 percent vs. 17
`percent), and reduced by a factor of 2 the number of
`patients who had moderate and severe relapses.
`8
`There was no difference in the proportion of patients
`in whom disability increased or in changes in the dis-
`ability scores between treatment groups. The patients
`in the placebo group had a mean increase of 17 per-
`-weighted MRI at
`cent in the area of the lesions on T
`2
`three years, as compared with a mean decrease of
`6 percent in the patients given high-dose interferon
`beta-1b. There was also a significant reduction in dis-
`ease activity, defined as the finding of new or enlarg-
`ing lesions in serial MRIs.
` The MRI findings in this
`57
`study were pivotal in obtaining FDA approval for in-
`terferon beta-1b and initiated the era in which MRI
`has a key role in assessing therapeutic responses in pa-
`tients with multiple sclerosis.
`Interferon beta-1a was tested in a multicenter trial
`involving 301 patients with relapsing–remitting mul-
`tiple sclerosis and mild-to-moderate disability. Treat-
`ment consisted of weekly intramuscular injections
`(6 million units [30
`g]) or placebo for up to two
`m
`years.
` The principal outcome was the length of
`7,58
`time to the progression of disability, defined as a de-
`crease from base line of at least 1.0 point on the Ex-
`panded Disability Status Scale that persisted for at
`least six months. Treatment with interferon beta-1a,
`as compared with placebo, significantly lowered the
`probability of progression of disability
` and of severe
`7
` In addition, patients treated with inter-
`disability.
`59
`feron beta-1a for two years had a reduction of 32
`percent in the annual rate of relapse, and had fewer
`gadolinium-enhanced lesions on MRI. The favor-
`able effect of interferon beta-1a on gadolinium-
`enhanced lesions, confirmed in a separate study with
`interferon beta-1b,
` suggests that interferon beta
`60
`inhibits new lesion formation.
`Both types of interferon beta are usually well tol-
`erated. The most common side effects are influenza-
`like symptoms for 24 to 48 hours after each injec-
`tion, and these usually subside after two to three
`months of treatment. Injection of interferon beta-1b
`causes redness, tenderness, swelling, and occasional-
`ly, necrosis at the injection site. Interferon beta-1b
`
`Volume 337 Number 22
`
`ⴢ
`
`1607
`
`4
`
`

`

`The New England Journal of Medicine
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`TABLE 3. IMPORTANT UNRESOLVED QUESTIONS
`RELATED TO INTERFERON BETA THERAPY IN PATIENTS
`WITH RELAPSING–REMITTING MULTIPLE SCLEROSIS.
`
`When should therapy be started?
`How long should therapy be continued?
`Can the dose be individualized to achieve maximal therapeu-
`tic benefit?
`What are the therapeutic mechanisms of action of the drug?
`What are the long-term benefits?
`Which preparation of interferon beta is clinically superior?
`
`can also cause slight elevations in serum aminotrans-
`ferase concentrations, leukopenia, or anemia, and a
`few patients have become depressed or have had
`worsening of preexisting depression. Interferon beta-
`1b–neutralizing activity was detected in serum sam-
`ples from 38 percent of patients by the third year of
`treatment
` and correlated with decreased efficacy of
`61
`therapy.
` Serum interferon beta-1a–neutralizing ac-
`62
`tivity was found less often — in 14 percent of pa-
`tients after one year and 22 percent after two years.
`7
`Although interferon beta therapy is effective, im-
`portant questions remain (Table 3). A risk–benefit
`analysis must be done in each patient. The cost of
`therapy, currently approximately $8,000 to $10,000
`per year, and the uncertain long-term risks may out-
`weigh the benefits in patients with mild multiple scle-
`rosis and a favorable prognosis. Whether long-term
`therapy should be started at the time of the first at-
`tack and what constitutes the optimal duration of
`therapy are not known. In a study of recombinant
`interferon alfa-2a (Roferon-A, Hoffmann–LaRoche,
`Nutley, N.J.) in patients with relapsing–remitting
`multiple sclerosis, relapse occurred when therapy was
` suggesting the need for
`stopped after six months,
`63
`more prolonged therapy. The final report from the in-
`terferon beta-1b study
` suggested that patients con-
`61
`tinued to respond to treatment for five years, a finding
`that supports the value of long-term therapy, but the
`high dropout rate (greater than 50 percent) may
`have biased the results in favor of long-term therapy.
`Specific indications to stop therapy were steady pro-
`gression of disability over a period of six months or
`treatment with three courses of corticotropin or cor-
`ticosteroids for acute relapses during a one-year pe-
` The appearance of serum interferon beta–neu-
`riod.
`64
`tralizing antibodies should prompt alternative therapy,
`particularly in patients with disease progression.
`65
`The variable biologic response to interferon beta
`suggests that the dose could be individualized. Side
`effects of the interferon beta-1b correlate with body-
` but there are no established methods to
`surface area,
`61
`individualize the dose for maximal efficacy. Figure 1
`shows the putative sites of action of interferon beta in
`patients with multiple sclerosis. Clarifying the mecha-
`nisms most closely linked to efficacy might lead to
`
`1608
`
`ⴢ
`
`November 27, 1997
`
`better methods to individualize treatment, particularly
`if the therapeutic effect could be monitored easily.
`The best preparation of interferon beta and the
`long-term benefits of such therapy remain controver-
`sial. Both interferon beta-1a and interferon beta-1b
`reduce the relapse rate and disease activity on MRI,
`but interferon beta-1a appears to be better tolerated.
`In addition, interferon beta-1a results in less progres-
`sion of disability,
` suggesting that long-term therapy
`7
`will lessen the eventual impact of the disease.
`
`Glatiramer Acetate
`Glatiramer acetate is a mixture of random syn-
`thetic polypeptides composed of
`-alanine,
`-glutam-
`L
`L
`ic acid, L-lysine, and L-tyrosine in a molar ratio of
`6.0:1.9:4.7:1.0. It was synthesized as an immuno-
`chemical mimic of myelin basic protein, a putative
`autoantigen in multiple sclerosis. After glatiramer
`acetate was found to inhibit experimental autoim-
`mune encephalomyelitis, a small trial suggested effi-
`cacy in patients with relapsing–remitting multiple
`sclerosis.66 It was subsequently tested in a trial involv-
`ing 251 patients with relapsing–remitting multiple
`sclerosis and mild-to-moderate disability. Treatment
`consisted of daily subcutaneous injections of 20 mg
`of glatiramer acetate or placebo for two years.14 The
`annualized relapse rate, the primary end point, was
`29 percent lower in the glatiramer acetate group,
`and the proportion of patients who did not have a
`relapse was higher (34 percent vs. 27 percent). A
`greater proportion of patients in the glatiramer ace-
`tate group had an improvement of 1.0 point or more
`in their score on the Expanded Disability Status Scale
`(25 percent vs. 15 percent), and fewer had worsening
`of disability (21 percent vs. 29 percent). The most
`common side effect was mild reactions at the injec-
`tion site, which occurred in 90 percent of patients
`given glatiramer acetate; 15 percent had brief epi-
`sodes of flushing, chest tightness, shortness of breath,
`palpitations, and anxiety after one or more injec-
`tions. Serum antibodies to glatiramer acetate also
`developed, but the presence of these antibodies had
`no effect on the clinical benefit. MRI scans, which
`were obtained at only one of the study sites, showed
`little change over the course of the study.67
`Glatiramer acetate was approved by the FDA in
`1996. It represents an alternative to interferon beta
`therapy for patients with relapsing–remitting multi-
`ple sclerosis and may be most useful for patients who
`become resistant to interferon beta treatment owing
`to serum interferon beta–neutralizing activity.
`
`Azathioprine
`Azathioprine, a purine analogue, depresses both
`cell-mediated and humoral immunity. A meta-analy-
`sis of five randomized, double-blind, placebo-con-
`trolled trials supported the conclusion that oral aza-
`thioprine (2 to 3 mg per kilogram per day) reduces
`
`5
`
`

`

`DRUG THERAPY
`
`the rate of relapse in multiple sclerosis 68 but has no
`effect on the progression of disability. The concern
`that prolonged azathioprine therapy may increase the
`risk of non-Hodgkin’s lymphoma or skin cancer 69
`was not confirmed in a case–control study.70 Azathi-
`oprine should be considered in patients with relaps-
`ing–remitting multiple sclerosis who do not respond
`to therapy with interferon beta or glatiramer acetate.
`Azathioprine may be useful in patients with Devic’s
`disease,71 a variant of multiple sclerosis affecting the
`optic nerves and cervical spinal cord, or in those with
`recurrent inflammatory myelitis.72
`
`Intravenous Immune Globulin
`Intravenous immune globulin has been used suc-
`cessfully in neuroimmunologic disorders, including
`acute and chronic inflammatory demyelinating poly-
`radiculopathy and myasthenia gravis, but its role in
`patients with multiple sclerosis is not yet clear. In a
`study of 150 patients with relapsing–remitting mul-
`tiple sclerosis who were treated with intravenous im-
`mune globulin (150 to 200 mg per kilogram per
`month) or placebo for two years, there was less wor-
`sening in the scores on the Expanded Disability Sta-
`tus Scale in the group given immune globulin, but
`the differences between groups were small. The
`number of relapses and the annual relapse rate were
`also lower in the immune globulin group. However,
`the treating physicians were aware of the treatment
`assignment, which could have biased the results.
`
`PROGRESSIVE MULTIPLE SCLEROSIS
`Investigators have only recently made a clinical
`distinction between secondary progressive multiple
`sclerosis and primary progressive multiple sclerosis,
`and most studies have not distinguished between
`these two forms of chronic progressive multiple scle-
`rosis. Treatment for chronic progressive multiple
`sclerosis has usually consisted of nonspecific immune
`suppression and has been of only moderate benefit.
`
`Methotrexate
`Methotrexate inhibits dihydrofolate reductase. Low-
`dose oral methotrexate is relatively nontoxic and ef-
`fective in rheumatoid arthritis and psoriasis, presum-
`ably by inhibiting both cell-mediated and humoral
`immunity or as a result of its antiinflammatory effects.
`Sixty ambulatory patients with chronic progressive
`multiple sclerosis and moderate-to-severe disability
`were treated with methotrexate (7.5 mg weekly) or
`placebo for two years.73 As compared with placebo,
`methotrexate significantly reduced sustained worsen-
`ing, according to a composite measure of outcome
`that included the Expanded Disability Status Scale,
`the Ambulation Index,74 and two tests of arm func-
`tion. The methotrexate-treated patients had less dis-
`ease progression, and patients with secondary pro-
`gressive multiple sclerosis benefited most.73,75 The
`
`clinical benefits were considered moderate, but
`toxicity was minimal. Because there are no nontoxic
`alternative treatments for patients with chronic
`progressive multiple sclerosis, low-dose oral metho-
`trexate should be considered for those with progres-
`sive deterioration.
`
`Cyclophosphamide
`Cyclophosphamide is an alkylating agent with po-
`tent cytotoxic and immunosuppressive effects. In
`several controlled but unblinded trials, high-dose in-
`travenous cyclophosphamide with or without subse-
`quent booster injections was effective in patients
`with chronic progressive multiple sclerosis.74,76,77 How-
`ever, one trial failed to demonstrate benefit.10 In this
`trial, the examining neurologist was unaware of the
`treatment assignments, and the patients wore bath-
`ing caps to mask the alopecia induced by cyclophos-
`phamide. Cyclophosphamide has many side effects
`in addition to alopecia, including nausea and vomit-
`ing, hemorrhagic cystitis, leukopenia, myocarditis,
`infertility, and pulmonary interstitial fibrosis. Treat-
`ment with cyclophosphamide may be most appro-
`priate for patients with rapidly progressive disease
`who do not respond to less toxic alternatives such as
`methotrexate.78
`
`Cyclosporine
`Cyclosporine is a potent immunosuppressive drug
`that inhibits several steps in the activation of T cells.
`In a multicenter U.S. study, 550 patients with chronic
`progressive multiple sclerosis were given oral cyclo-
`sporine (initial dose, 7 mg per kilogram per day) or
`placebo for two years.11 The length of time before
`patients became dependent on a wheelchair was
`slightly longer in the cyclosporine group, but neph-
`rotoxicity occurred in 84 percent of cyclosporine-
`treated patients, and hypertension was also frequent.
`The moderate clinical benefits of cyclosporine ap-
`pear to be outweighed by its toxicity.
`
`FUTURE DIRECTIONS
`Long-term placebo-controlled trials may no long-
`er be ethically justifiable in patients with relapsing–
`remitting multiple sclerosis, given the evidence of
`the efficacy of interferon beta and glatiramer acetate.
`Comparisons between treatments are needed but
`will require very large samples, lengthy studies, and
`more sensitive outcome measures.49,79 Analyses of
`cost effectiveness and the quality of life80,81 will be
`increasingly important as more therapeutic options
`become available.
`There are many potential new therapies, and it is
`not clear how best to screen them. Effectiveness in
`experimental autoimmune encephalomyelitis has been
`considered a prerequisite, but many therapies that
`are effective in experimental autoimmune encepha-
`lomyelitis are not useful in patients with multiple
`
`Volume 337 Number 22
`
`ⴢ 1609
`
`6
`
`

`

`The New England Journal of Medicine
`
`sclerosis. Using serial gadolinium-enhanced MRI
`scans to screen therapies for efficacy has been rec-
`ommended,79 but such an approach has the poten-
`tial of screening out treatments that might alter a
`different stage of the pathologic process.
`Specific forms of immune suppression are based
`on deleting or inhibiting autoreactive pathogenic
`T cells. This approach may be limited by the heter-
`ogeneity and complexity of myelin determinant rec-
`ognition in patients with established multiple scle-
`rosis.35 Nevertheless, ongoing trials are testing the
`efficacy of a T-cell–receptor vaccine,32,82 myelin pep-
`tides conjugated to major-histocompatibility-com-
`plex molecules,83 and oral myelin,84 all of which are
`thought to inhibit myelin-reactive T cells. Studies
`of nonspecific immunomodulation with linomide,
`transforming growth factor b, and soluble tumor ne-
`crosis factor–receptor are under way and are planned
`for interleukin-10. Remyelination strategies are large-
`ly at a preclinical stage, although the efficacy of intra-
`venous immune globulin is being tested in patients
`who have fixed neurologic deficits.85 In the future,
`glial-cell transplantation86 or treatment with recom-
`binant growth factors87 could be used to stimulate re-
`generation and functional recovery.
`
`We are indebted to Drs. Henry F. McFarland and W. Ian Mc-
`Donald for their thoughtful review of the manuscript and many
`helpful suggestions.
`
`REFERENCES
`
`1. Williams R, R

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