throbber

`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`The New England Journal of Medicine
`
`Review Article
`
`Medical Progress
`
`M
`
`ULTIPLE
`
` S
`
`CLEROSIS
`
`J
`OHN
`
` L
`, M.D., C
` H. N
`UCCHINETTI
`LAUDIA
`OSEWORTHY
`M
` R
`, M.D.,
`OSES
`ODRIGUEZ
` B
` G. W
`, M.D.
`RIAN
`EINSHENKER
`
`AND
`
`, M.D.,
`
`M
`
`ORE than 100 years has passed since
`Charcot, Carswell, Cruveilhier, and others
`described the clinical and pathological
`characteristics of multiple sclerosis.
` This enigmatic,
`1
`relapsing, and often eventually progressive disorder
`of the white matter of the central nervous system
`continues to challenge investigators trying to under-
`stand the pathogenesis of the disease and prevent its
`progression.
` There are 250,000 to 350,000 patients
`2
`with multiple sclerosis in the United States.
` Multiple
`3
`sclerosis typically begins in early adulthood and has
`a variable prognosis. Fifty percent of patients will need
`help walking within 15 years after the onset of dis-
`ease.
` Advanced magnetic resonance imaging (MRI)
`4
`and spectroscopy may allow clinicians to follow the
`pathological progression of the disease and monitor
`the response to treatment. Recent progress has oc-
`curred in understanding the cause, the genetic com-
`ponents, and the pathologic process of multiple scle-
`rosis. The short-term clinical and MRI manifestations
`of disease activity have been reduced by new therapies,
`although the degree of presumed long-term benefit
`from these treatments will require further study.
`CLINICAL COURSE AND DIAGNOSIS
`A patient’s presenting symptoms and the temporal
`evolution of the clinical findings may suggest the cor-
`rect diagnosis. In relapsing–remitting multiple scle-
`rosis — the type present in 80 percent of patients —
`symptoms and signs typically evolve over a period of
`several days, stabilize, and then often improve, spon-
`taneously or in response to corticosteroids, within
`weeks. Relapsing–remitting multiple sclerosis typi-
`
`From the Department of Neurology, Mayo Clinic and Mayo Founda-
`tion, Rochester, Minn. Address reprint requests to Dr. Noseworthy at the
`Department of Neurology, Mayo Clinic and Mayo Foundation, 200 First
`St., SW, Rochester, MN 55905.
`©2000, Massachusetts Medical Society.
`
`cally begins in the second or third decade of life and
`has a female predominance of approximately 2:1. The
`tendency for corticosteroids to speed recovery from
`relapses often diminishes with time. Persistent signs of
`central nervous system dysfunction may develop after
`a relapse, and the disease may progress between relaps-
`es (secondary progressive multiple sclerosis). Twenty
`percent of affected patients have primary progressive
`multiple sclerosis, which is characterized by a gradu-
`ally progressive clinical course and a similar incidence
`among men and women.
`Relapsing–remitting multiple sclerosis typically
`starts with sensory disturbances, unilateral optic neu-
`ritis, diplopia (internuclear ophthalmoplegia), Lher-
`mitte’s sign (trunk and limb paresthesias evoked by
`neck flexion), limb weakness, clumsiness, gait ataxia,
`and neurogenic bladder and bowel symptoms. Many
`patients describe fatigue that is worse in the afternoon
`and is accompanied by physiologic increases in body
`temperature. The onset of symptoms post partum and
`symptomatic worsening with increases in body tem-
`perature (Uhthoff ’s symptom) and pseudoexacerba-
`tions with fever suggest the diagnosis. Some patients
`have recurring, brief, stereotypical phenomena (par-
`oxysmal pain or paresthesias, trigeminal neuralgia, ep-
`isodic clumsiness or dysarthria, and tonic limb postur-
`ing) that are highly suggestive of multiple sclerosis.
`Prominent cortical signs (aphasia, apraxia, recurrent
`seizures, visual-field loss, and early dementia) and ex-
`trapyramidal phenomena (chorea and rigidity) only
`rarely dominate the clinical picture. Eventually, cog-
`nitive impairment, depression, emotional lability, dys-
`arthria, dysphagia, vertigo, progressive quadriparesis
`and sensory loss, ataxic tremors, pain, sexual dysfunc-
`tion, spasticity, and other manifestations of central
`nervous system dysfunction may become troublesome.
`Patients who have primary progressive multiple scle-
`rosis often present with a slowly evolving upper-
`motor-neuron syndrome of the legs (“chronic pro-
`gressive myelopathy”). Typically, this variant worsens
`gradually, and quadriparesis, cognitive decline, visual
`loss, brain-stem syndromes, and cerebellar, bowel,
`bladder, and sexual dysfunction may develop.
`The diagnosis is based on established clinical and,
`when necessary, laboratory criteria.
` Advances in cer-
`5
`ebrospinal fluid analysis and MRI, in particular, have
`simplified the diagnostic process (Fig. 1).
` The relaps-
`6
`ing forms are considered clinically definite when neu-
`rologic dysfunction becomes “disseminated in space
`and time.” Primary progressive multiple sclerosis may
`be suggested clinically by a progressive course that
`lasts longer than six months, but laboratory studies
`to obtain supportive evidence and efforts to exclude
`
`938
`

`
`September 28, 2000
`
`1
`
`Hopewell EX1006
`
`

`

`
`
`
`
`
`
`
`
`
`
`
`
`
`MEDICAL PROGRESS
`
`A
`
`C
`
`B
`
`D
`
` MRI Scans of the Brain of a 25-Year-Old Woman with Relapsing–Remitting Multiple Sclerosis.
`Figure 1.
`An axial FLAIR (fluid-attenuated inversion recovery) image shows multiple ovoid and confluent hyperintense lesions in the periven-
`tricular white matter (Panel A). Nine months later, the number and size of the lesions have substantially increased (Panel B). After
`the administration of gadolinium, many of the lesions demonstrate ring or peripheral enhancement, indicating the breakdown of
`the blood–brain barrier (Panel C). In Panel D, a parasagittal T
`-weighted MRI scan shows multiple regions in which the signal is
`1
`diminished (referred to as “black holes”) in the periventricular white matter and corpus callosum. These regions correspond to the
`chronic lesions of multiple sclerosis.
`
`2
`
`Volume 343 Number 13
`

`
`939
`
`

`

`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`The New England Journal of Medicine
`
`other, potentially treatable illnesses are advised; for
`example, structural or metabolic myelopathy can be
`identified by appropriate laboratory studies, including
`spinal MRI (Table 1). On MRI, findings of multifo-
`cal lesions of various ages, especially those involving
`the periventricular white matter, brain stem, cerebel-
`lum, and spinal cord white matter, support the clin-
`ical impression. The presence of gadolinium-enhanc-
`ing lesions on MRI indicates current sites of presumed
`inflammatory demyelination (active lesions).
`When there is diagnostic uncertainty, repeated
`MRI after several months may provide evidence that
`the lesions are “disseminated in time.” Cerebrospi-
`nal fluid analysis often shows increased intrathecal
`synthesis of immunoglobulins of restricted specifici-
`ty (oligoclonal bands may be present, or the synthesis
`of IgG may be increased), with moderate lympho-
`cytic pleocytosis (almost invariably there are fewer
`than 50 mononuclear cells). Physiologic evidence of
`subclinical dysfunction of the optic nerves and spinal
`cord (changes in visual evoked responses and soma-
`tosensory evoked potentials) may provide support
`for the conclusion that there is “dissemination in
`space.”
` Therefore, spinal MRI and evoked-potential
`7
`testing may provide evidence of a second lesion that
`can confirm the diagnosis. Abnormalities detected
`by testing of somatosensory evoked potentials and
`spinal MRI may clarify the diagnosis in patients with
`optic neuritis alone or isolated brain-stem abnormal-
`ities and in those suspected of having unifocal cere-
`bral multiple sclerosis on the basis of MRI. If posi-
`tive, abnormalities detected by tests of visual evoked
`responses may support the diagnosis of multiple scle-
`rosis in patients with isolated brain-stem or spinal
`cord lesions.
`The course of multiple sclerosis in an individual
`patient is largely unpredictable. Patients who have a
`so-called clinically isolated syndrome (e.g., optic neu-
`ritis, brain-stem dysfunction, or incomplete transverse
`myelitis) as their first event have a greater risk of both
`recurrent events (thereby confirming the diagnosis
`of clinically definite multiple sclerosis) and disability
`within a decade if changes are seen in clinically asymp-
`tomatic regions on MRI of the brain.
` The presence
`8
`of oligoclonal bands in cerebrospinal fluid slightly in-
`creases the risk of recurrent disease.
`9
`Studies of the natural history of the disease have
`provided important prognostic information that is
`useful for counseling patients and planning clinical
` Ten percent of patients do well for more
`trials.
`4,10,11
`than 20 years and are thus considered to have benign
`multiple sclerosis. Approximately 70 percent will have
`secondary progression.
` Frequent relapses in the first
`4
`two years, a progressive course from the onset, male
`sex, and early, permanent motor or cerebellar find-
`ings are independently, but imperfectly, predictive of
`a more severe clinical course. Women and patients
`with predominantly sensory symptoms and optic neu-
`
`940
`

`
`September 28, 2000
`
`3
`
`T
`
`ABLE
`
` 1.
`
` D
`
`IFFERENTIAL
`
` D
`
`IAGNOSIS
`
`
`
`OF
`
` M
`
`ULTIPLE
`
` S
`
`CLEROSIS
`
`.
`
`Metabolic disorders
`Disorders of B
` metabolism*
`12
`Leukodystrophies
`Autoimmune diseases
`Sjögren’s syndrome, systemic lupus erythematosus, Behçet’s disease, sar-
`coidosis, chronic inflammatory demyelinating polyradiculopathy associat-
`ed with central nervous system demyelination, antiphospholipid-anti-
`body syndrome
`Infections
`†
`HIV-associated myelopathy* and HTLV-1–associated myelopathy,* Lyme
`disease, meningovascular syphilis, Eales’ disease
`Vascular disorders
`Spinal dural arteriovenous fistula*
`Cavernous hemangiomata
`Central nervous system vasculitis, including retinocochlear cerebral
`vasculitis
`Cerebral autosomal dominant arteriopathy with subcortical infarcts and
`leukoencephalopathy
`Genetic syndromes
`Hereditary ataxias and hereditary paraplegias*
`Leber’s optic atrophy and other mitochondrial cytopathies
`Lesions of the posterior fossa and spinal cord
`Arnold–Chiari malformation, nonhereditary ataxias
`Spondylotic and other myelopathies*
`Psychiatric disorders
`Conversion reaction, malingering
`Neoplastic diseases
`Spinal cord tumors,* central nervous system lymphoma
`Paraneoplastic disorders
`Variants of multiple sclerosis
`‡
`Optic neuritis; isolated brain-stem syndromes; transverse myelitis; acute
`disseminated encephalomyelitis, Marburg disease; neuromyelitis optica
`
`*This disorder or group of disorders is of particular relevance in the dif-
`ferential diagnosis of progressive myelopathy and primary progressive mul-
`tiple sclerosis.
`†HIV denotes human immunodeficiency virus, and HTLV-1 human
`T-cell lymphotropic virus type 1.
`‡In many patients with these variants, clinically definite multiple sclerosis
`develops or the course is indistinguishable from that of multiple sclerosis.
`
`ritis have a more favorable prognosis. Life expectan-
`cy may be shortened slightly; in rare cases, patients
`with fulminant disease die within months after the on-
`set of multiple sclerosis. Suicide remains a risk, even
`for young patients with mild symptoms.
`12
`EPIDEMIOLOGIC FEATURES
`The prevalence of multiple sclerosis varies consid-
`erably around the world.
` Kurtzke classified regions
`13
`of the world according to prevalence: a low preva-
`lence was considered less than 5 cases per 100,000
`persons, an intermediate prevalence was 5 to 30 per
`100,000 persons, and a high prevalence was more
`than 30 per 100,000 persons.
` The prevalence is
`14
`highest in northern Europe, southern Australia, and
`the middle part of North America. There has been
`
`

`

`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`MEDICAL PROGRESS
`
`a trend toward an increasing prevalence and incidence,
`particularly in southern Europe.
` Even in areas with
`15,16
`uniform methods of ascertainment and high preva-
`lence, such as Olmsted County, Minnesota, the in-
`cidence has increased from 2 to 6 per 100,000 dur-
`ing the past century.
` However, the incidence has
`17
`actually declined in some,
` but not all,
` areas of
`18,19
`20
`northern Europe. Stable or declining rates have been
`reported most often in regions with high prevalence
`and incidence. The extent to which the observed in-
`creases in incidence are explained by an enhanced
`awareness of the disease and improved diagnostic
`techniques is uncertain. There is a large reservoir of
`mild cases, the recognition of which may depend
`heavily on the zeal and resources of the investigator.
`The reasons for the variation in the prevalence and
`incidence of multiple sclerosis worldwide are not un-
`derstood. Environmental and genetic explanations
`have been offered, and both factors probably have a
`role. The occurrence of rapid shifts in the incidence
`of multiple sclerosis, if not artifactual, is an argu-
`ment for an environmental influence, as is the equiv-
`ocal, but suggestive, evidence of the clustering of cases
`in terms of both geography and time and of epidem-
`ics, especially on the Faroe Islands.
` The apparent
`21
`change in the frequency of multiple sclerosis among
`people
` and their offspring
` who migrate to and
`22,23
`24
`from high-prevalence areas is another factor that has
`been presented to support the existence of an envi-
`ronmental factor. However, each of these relations
`has potential confounders that preclude the drawing
`of a definite conclusion regarding the importance of
`environmental factors.
` The nature of putative envi-
`25
`ronmental factors remains unclear in numerous case–
`control studies. Studies that show that the incidence
`of multiple sclerosis among the adopted children of
`patients with multiple sclerosis is not higher than ex-
`pected seem to argue against the possibility that a
`transmissible factor is primarily responsible for the
`increased risk of the disease among relatives and in-
`stead suggest that genetic factors may be responsible.
`26
`GENETIC FACTORS
`Evidence that genetic factors have a substantial ef-
`fect on susceptibility to multiple sclerosis is unequiv-
`ocal. The concordance rate of 31 percent among
`monozygotic twins is approximately six times the rate
`among dizygotic twins (5 percent).
` The absolute risk
`27
`of the disease in a first-degree relative of a patient with
`multiple sclerosis is less than 5 percent; however, the
`risk in such relatives is 20 to 40 times the risk in the
`general population.
` Since 1973, it has been recog-
`28
`nized that the presence of the HLA-DR2 allele sub-
`stantially increases the risk of multiple sclerosis.
`29
`This effect has been found in all populations, with the
`exception of that in Sardinia.
` The magnitude of the
`30
`relative risk depends on the frequency of the HLA-
`DR2 allele in the general population. Given the high
`
`frequency of this allele in the population, the risk at-
`tributable to the HLA-DR2 allele is considerable. Pop-
`ulations with a high frequency of the allele (e.g., those
`in Scotland) have the highest risk of multiple sclerosis.
`The mode of transmission of genetic susceptibility
`to multiple sclerosis is complex. Most cases are sporad-
`ic, despite the clear excess risk among the relatives of
`patients. Investigators have used the usual genetic
`approaches to identify genes associated with an in-
`creased risk of multiple sclerosis.
`Studies of candidate genes have targeted individu-
`al genes with microsatellite markers with use of asso-
`ciation and linkage strategies. For some genetic re-
`gions, such as the HLA region on chromosome 6, it
`has been difficult to identify the specific polymor-
`phism that predisposes persons to the disease, given
`the high degree of linkage disequilibrium at that locus.
`Candidate-gene studies were followed by four stud-
`ies in which the entire genome was scanned.
` Re-
`31-34
`gions of interest have been identified, although none
`have been linked to the disease with certainty. Con-
`sidering the rather large number of patients evaluated
`in such studies, one might conclude tentatively that no
`single gene, except possibly those for HLA antigens,
`35
`exerts a strong effect.
`Further refinement of the linkage map is in prog-
`ress.
` Whether this approach will prove powerful
`36
`enough to identify genes with a relatively weak effect
`is difficult to predict. To enhance the detection of
`genes with a weak effect, investigators have begun to
`use strategies involving linkage-disequilibrium map-
`ping and transmission-disequilibrium testing. In these
`approaches, putative causative alleles or marker al-
`leles and haplotypes are assessed to determine wheth-
`er they are associated with the disease at a popula-
`tion level or whether they are associated with a
`higher-than-expected rate of transmission of disease
`from heterozygous parents to their children. This ef-
`fort will involve a major expenditure of resources to
`achieve genome-wide coverage. The development of
`novel analytic techniques for these types of genetic
`data sets makes such an undertaking feasible.
`37
`The severity and course of multiple sclerosis may
`also be influenced by genetic factors. Epidemiologic
`evidence to support this premise comes from studies
`examining the rate of concordance for measures that
`describe and quantitate variations in the course of
`disease, including the age at onset, the proportion
`of patients in whom the disease progresses, and the
` HLA-DR and DQ
`extent of disability over time.
`38
`polymorphisms are not associated with the course and
`severity of multiple sclerosis, despite their substantial
`contribution to disease susceptibility.
` Recently, vari-
`39
`ants of the interleukin-1
`–receptor and interleukin-1–
`b
`receptor antagonist genes,
` immunoglobulin Fc re-
`40
`ceptor genes,
` and apolipoprotein E gene
` have been
`41
`42
`associated with the course of the disease, but these
`findings await confirmation.
`
`4
`
`Volume 343 Number 13
`

`
`941
`
`

`

`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`The New England Journal of Medicine
`
`PATHOLOGICAL FEATURES
`AND PATHOGENESIS
`Multiple sclerosis is generally believed to be an im-
`mune-mediated disorder that occurs in genetically sus-
`ceptible people (Fig. 2).
` However, the sequence of
`43
`events that initiates the disease remains largely un-
`known. Given the considerable clinical, genetic, MRI,
`and pathological heterogeneity of multiple sclerosis,
`perhaps more than one pathogenetic mechanism con-
`tributes to tissue injury. This possibility has therapeutic
`implications, because more than one approach to treat-
`ment may be required to treat this disease effectively.
`The pathological hallmark of chronic multiple scle-
`rosis is the demyelinated plaque, which consists of a
`well-demarcated hypocellular area characterized by
`the loss of myelin, relative preservation of axons, and
`the formation of astrocytic scars (Fig. 3). Lesions have
`a predilection for the optic nerves, periventricular
`white matter, brain stem, cerebellum, and spinal cord
`white matter, and they often surround one or several
`medium-sized vessels. Although the lesions are usu-
`ally round or oval, they often have finger-like exten-
`sions along the path of small or medium-sized blood
`vessels (Dawson’s fingers). Inflammatory cells are typ-
`ically perivascular in location, but they may diffusely
`infiltrate the parenchyma. The composition of the
`inflammatory infiltrate varies depending on the stage
`of demyelinating activity. In general, it is composed
`
`of lymphocytes and macrophages; the latter predomi-
`nate in active lesions.
`For meaningful conclusions to be drawn regarding
`the earliest immunologic and molecular events con-
`tributing to the formation of lesions, only actively de-
`myelinating plaques should be considered. Identifying
`myelin-degradation products in macrophages is the
`most reliable method of identifying active lesions (Fig.
`4).
` When stringent criteria are used to define lesion-
`44
`al activity, the frequency of active plaques in patients
`with chronic multiple sclerosis is extremely low. Al-
`though remyelination is minimal in lesions associated
`with chronic multiple sclerosis, plaques in acute and
`early multiple sclerosis may have extensive remyeli-
`nation (referred to as shadow plaques) (Fig. 5). Fur-
`thermore, the lesions of chronic multiple sclerosis
`reportedly contain substantial numbers of oligoden-
`drocyte precursor cells.
` Thus, central nervous sys-
`45
`tem myelin can be repaired, and mechanisms that
`promote endogenous remyelination may represent a
`feasible therapeutic strategy.
`Early symptoms of multiple sclerosis are widely
`believed to result from axonal demyelination, which
`leads to the slowing or blockade of conduction. The
`regression of symptoms has been attributed to the
`resolution of inflammatory edema and to partial re-
`myelination. However, inflammatory cytokines may
`inhibit axonal function, and the recovery of function
`
` Possible Mechanisms of Injury and Repair in Multiple Sclerosis.
`Figure 2 (facing page).
`Genetic and environmental factors (including viral infection, bacterial lipopolysaccharides, superantigens, reactive metabolites, and
`metabolic stress) may facilitate the movement of autoreactive T cells and demyelinating antibodies from the systemic circulation
`into the central nervous system through disruption of the blood–brain barrier. In the central nervous system, local factors (including
`viral infection and metabolic stress) may up-regulate the expression of endothelial adhesion molecules, such as intercellular adhe-
`sion molecule 1 (ICAM-1), vascular-cell adhesion molecule 1 (VCAM-1), and E-selectin, further facilitating the entry of T cells into
`the central nervous system. Proteases, including matrix metalloproteinases, may further enhance the migration of autoreactive im-
`mune cells by degrading extracellular-matrix macromolecules. Proinflammatory cytokines released by activated T cells, such as
`interferon-
` and tumor necrosis factor
` (TNF-
`), may up-regulate the expression of cell-surface molecules on neighboring lympho-
`g
`b
`b
`cytes and antigen-presenting cells. Binding of putative multiple sclerosis (MS) antigens, such as myelin basic protein, myelin-asso-
`ciated glycoprotein, myelin oligodendrocyte glycoprotein (MOG), proteolipid protein,
`B-crystallin, phosphodiesterases, and S-100
`a
`protein, by the trimolecular complex — the T-cell receptor (TCR) and class II major-histocompatibility-complex (MHC) molecules on
`antigen-presenting cells — may trigger either an enhanced immune response against the bound antigen or anergy, depending on
`the type of signaling that results from interactions with surface costimulatory molecules (e.g., CD28 and CTLA-4) and their ligands
`(e.g., B7-1 and B7-2). Down-regulation of the immune response (anergy) may result in the release of antiinflammatory cytokines
`(interleukin-1, interleukin-4, and interleukin-10) from CD4+ T cells, leading to the proliferation of antiinflammatory CD4+ type 2 help-
`er T (Th2) cells. Th2 cells may send antiinflammatory signals to the activated antigen-presenting cells and stimulate pathologic or
`repair-enhancing antibody-producing B cells. Alternatively, if antigen processing results in an enhanced immune response, proin-
`flammatory cytokines (e.g., interleukin-12 and interferon-
`) may trigger a cascade of events, resulting in the proliferation of proin-
`g
`flammatory CD4+ type 1 helper T (Th1) cells and ultimately in immune-mediated injury to myelin and oligodendrocytes. Multiple
`mechanisms of immune-mediated injury of myelin have been postulated: cytokine-mediated injury of oligodendrocytes and myelin;
`digestion of surface myelin antigens by macrophages, including binding of antibodies against myelin and oligodendrocytes (i.e.,
`antibody-dependent cytotoxicity); complement-mediated injury; and direct injury of oligodendrocytes by CD4+ and CD8+ T cells.
`This injury to the myelin membrane results in denuded axons that are no longer able to transmit action potentials efficiently within
`the central nervous system (loss of saltatory conduction). This slowing or blocking of the action potential results in the production
`of neurologic symptoms. The exposed axon segments may be susceptible to further injury from soluble mediators of injury (in-
`cluding cytokines, chemokines, complement, and proteases), resulting in irreversible axonal injury (such as axonal transection and
`terminal axon ovoids). There are several possible mechanisms of repair of the myelin membrane, including resolution of the in-
`flammatory response followed by spontaneous remyelination, spread of sodium channels from the nodes of Ranvier to cover de-
`nuded axon segments and restore conduction, antibody-mediated remyelination, and remyelination resulting from the proliferation,
`migration, and differentiation of resident oligodendrocyte precursor cells. Adapted from a drawing by the Mayo Foundation.
`
`942
`

`
`September 28, 2000
`
`5
`
`

`

`
`
`
`
`
`
`
`MEDICAL PROGRESS
`
`Autoreactive T cell
`
`Endothelial cells
`
`Adhesion
`
`Demyelinating antibodies(cid:2)
`Anti-MOG?
`ICAM-1, VCAM-1,(cid:2)
`E-selectins up-regulated
`
`Systemic circulation
`
`Blood–brain barrier
`
`Activated antigen-(cid:2)
`presenting cell(cid:2)
`(astrocytes, microglia,(cid:2)
`macrophages)
`
`Penetration(cid:2)
`(matrix metalloproteinases)
`
`(cid:2)
`Interferon-g
`TNF-b
`
`(cid:2)
`B7-1, B7-2
`
`CD28 Immune response
`CTLA-4 Anergy
`
`Basement(cid:2)
`membrane
`
`Central(cid:2)
`nervous system
`
`Oligodendrocyte(cid:2)
`precursor cell
`
`TCR
`
`CD4+ T cell
`
`Antiinflammatory signaling(cid:2)
`Interleukin-4, 10, 13
`
`Activated(cid:2)
`B cell
`
`Class II MHC(cid:2)
`molecule
`Putative(cid:2)
`MS antigen
`
`(cid:2)
`
`CD4+(cid:2)
`Th1 cell
`(cid:2)
`TNF-a,
`Interferon-g
`
`
`
`Interleukin-12(cid:2)
`Interferon-g
`
`
`Interleukin-1, 4, 10(cid:2)
`(cid:2)
`
`CD4+(cid:2)
`Th2 cell
`
`(cid:2)
`Interleukin-(cid:2)
`4, 5, 6, 10, 13
`
`Antibodies
`
`Cytokine-mediated(cid:2)
`injury
`
`Macrophage
`
`Complement
`
`Neuronal(cid:2)
`cell body
`
`Glial growth(cid:2)
`factors?
`
`Surface Ig
`
`Proliferation(cid:2)
`Migration(cid:2)
`Differentiation(cid:2)
`Remyelination
`
`Normal(cid:2)
`myelin sheath
`
`Remyelinated(cid:2)
`areas
`
`Immune-(cid:2)
`mediated injury
`
`Class I MHC(cid:2)
`molecule
`
`Class I–restricted(cid:2)
`CD8+ T cell(cid:2)
`up-regulated by(cid:2)
`interferon-g
`
`Antibody-mediated injury(cid:2)
`(cid:2)
`(cid:2)
`(cid:2)
`
`Antibody-mediated(cid:2)
`remyelination?
`
`Primary(cid:2)
`oligodendrogliopathy
`
`Oligodendrocyte
`
`Terminal(cid:2)
`axon ovoid
`
`Increased(cid:2)
`sodium-channel(cid:2)
`density
`
`Degeneration(cid:2)
`of inner glial loop
`
`Postsynaptic neuron
`
`6
`
`Volume 343 Number 13
`

`
`943
`
`

`

`The New England Journal of Medicine
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`A
`
`B
`
` Photomicrographs of a Chronic Multiple Sclerosis
`Figure 3.
`Plaque.
`In Panel A, a well-demarcated hypocellular region of myelin
`loss is evident in the periventricular white matter (luxol fast
`blue and periodic acid–Schiff myelin stain, ¬15). In Panel B,
`neurofilament staining for axons in the same lesion demon-
`strates a reduction in axonal density (¬15).
`
`may result from the redistribution of sodium chan-
`nels across segments of demyelinated axons.
` Irre-
`46,47
`versible axonal injury, gliotic scarring, and exhaustion
`of the oligodendrocyte progenitor pool may result
`from repeated episodes of disease activity and lead to
`progressive loss of neurologic function. Axonal inju-
`ry may occur not only in the late phases of multiple
`sclerosis but also after early episodes of inflammatory
`demyelination.
` The pathogenesis of this early ax-
`48-50
`onal injury is still unclear.
`Experimental in vitro and in vivo models of in-
`flammatory demyelination suggest that diverse disease
`processes, including autoimmunity and viral infection,
`
`944
`

`
`September 28, 2000
`
`7
`
`A
`
`B
`
` Photomicrographs of an Actively Demyelinating Mul-
`Figure 4.
`tiple Sclerosis Lesion (Immunocytochemical Staining of Myelin
`Oligodendrocyte Glycoprotein [Brown] with Hematoxylin Coun-
`terstaining of Nuclei [Blue]).
`In Panel A, at the active edge of a multiple sclerosis lesion (in-
`dicated by the asterisk), the products of myelin degradation are
`present in numerous macrophages (arrowheads) (¬100). In Pan-
`el B (¬100), macrophages containing myelin debris (arrowheads)
`are interdigitated with degenerating myelin sheaths.
`
`

`

`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`MEDICAL PROGRESS
`
`protein is present on the outer lamellae of the myelin
`sheath). Antibodies against both myelin oligodendro-
`cyte glycoprotein and myelin basic protein can be
`found in the brains of patients with multiple sclero-
`sis.
` Deposits of immunoglobulin and activated com-
`57
`plement may be present in multiple sclerosis lesions
`in which myelin is being degraded.
` Taken together,
`58
`these observations suggest that an antibody-mediated
`process may have an important role in the pathogen-
`esis of multiple sclerosis.
`Other factors may also help degrade myelin and
`damage oligodendrocytes. Activated macrophages and
`microglial cells may mediate such activity by produc-
`ing proinflammatory cytokines (such as tumor necro-
`sis factor
` and interferon-
`), generating reactive ox-
`a
`g
`ygen or nitrogen species, producing excitatory amino
`acids, activating complement components, or releas-
`ing proteolytic and lipolytic enzymes. Other factors
`potentially toxic to oligodendroglial cells include sol-
`uble T-cell products (such as perforin), the interaction
`of Fas antigen with Fas ligand, cytotoxicity mediated
`by the interaction of CD8+ T cells with class I ma-
`jor-histocompatibility-complex (MHC) antigens on
`antigen-presenting cells, and persistent viral infec-
`tion.
` Human herpesvirus type 6 can cause a con-
`54
`dition that mimics multiple sclerosis
` and appears in
`59
`oligodendrocytes within multiple sclerosis tissue in
`some patients, but not in control tissue.
` A direct
`60
`causal link, however, remains to be confirmed. In
`one study,
`
`Chlamydia pneumoniae was isolated from
`64 percent of patients with multiple sclerosis, as
`compared with 11 percent of control patients with
`other neurologic diseases, and it was detected in cer-
`ebrospinal fluid by a polymerase-chain-reaction assay
`in 97 percent of patients with multiple sclerosis, as
`compared with 18 percent of control patients.
`61
`These results have yet to be confirmed in other lab-
`oratories.
`62
`Various pathogenic mechanisms may be involved
`in multiple sclerosis. There is an important degree of
`variability among patients in the structural and im-
`munologic features of the lesions of multiple sclero-
`sis.
` The extent of survival of oligodendrocytes varies
`63
`from patient to patient but is uniform within a given
`patient, suggesting that the focus of injury (myelin,
`mature oligodendrocyte, or progenitor cell) varies
`among patients.
` Although most lesions are charac-
`49
`terized by an inflammatory reaction, composed main-
`ly of T lymphocytes and macrophages, diverse pat-
`
`terns of myelin destruction have been described.
`64
`In some lesions, the presence of immunoglobulins
`and activated terminal complement components sug-
`gests that demyelinating antibodies have a pathogenic
`role. In others, a primary oligodendrocyte dystrophy
`manifested by the selective loss of myelin-associated
`glycoprotein and apoptosis of oligodendrocytes has
`been seen. Finally, in other cases, a small rim of ne-
`crotic oligodendrocytes has been found in the nor-
`
`8
`
`Volume 343 Number 13
`

`
`945
`
` Remyelination in a Lesion Associated with Chronic
`Figure 5.
`Multiple Sclerosis.
`The area stained pale blue (indicated by the asterisk) repre-
`sents a region of partial remyelination (a shadow plaque) along
`the periventricular edge of a lesion in a patient with chronic
`multiple sclerosis (luxol fast blue and periodic acid–Schiff my-
`elin stain, ¬15). NAWM denotes normal-appearing white matter.
`
`may induce multiple sclerosis–like inflammatory de-
`myelinated plaques. Activated CD4+ T cells specific
`for one or more self antigens are believed to adhere
`to the luminal surface of endothelial cells in central
`nervous system venules and migrate into the central
`nervous system at the time of disruption of the
`blood–brain barrier. This process is followed by an
`amplification of the immune response after the recog-
`nition of target antigens on antigen-presenting cells.
`The existence of T cells that are reactive to several
`putative self myelin and non-myelin “multiple sclerosis
`antigens,” including myelin basic protein, myelin-asso-
`ciated glycoprotein, myelin oligodendrocyte glycopro-
`tein, proteolipid protein,
`B-crystallin, phosphodi-
`a
`esterases, and S-100 protein, has been proposed.
`51-53
`Additional amplification

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