You are on page 1of 9

The n e w e ng l a n d j o u r na l of m e dic i n e

Review Article

Allan H. Ropper, M.D., Editor

Neuromyelitis Optica Spectrum Disorder


Dean M. Wingerchuk, M.D., and Claudia F. Lucchinetti, M.D.​​

N
euromyelitis optica spectrum disorder (NMOSD) is a rare auto- From the Department of Neurology,
immune disease. It causes multifocal central nervous system (CNS) in- Mayo Clinic, Scottsdale, AZ (D.M.W.);
and the Department of Neurology, Mayo
flammation that primarily affects the optic nerves and spinal cord, and it Clinic, Rochester, MN (C.F.L.). Dr. Wing-
typically results in attacks of visual loss and paralysis. Associations between optic erchuk can be contacted at ­wingerchuk
nerve and spinal cord disease were noted in the 19th century, especially after De- ​.­dean@​­mayo​.­edu or at the Department
of Neurology, Mayo Clinic, 13400 E. Shea
vic’s description of “neuro-myélite optique aiguë” (acute neuromyelitis optica).1 In Blvd., Scottsdale, AZ 85259.
the 20th century, “Devic’s disease” was considered a severe, monophasic optic and
N Engl J Med 2022;387:631-9.
spinal variant of multiple sclerosis that spared the brain.2 However, the discovery DOI: 10.1056/NEJMra1904655
of IgG-class antibodies that bind to the water channel aquaporin-4 (AQP4-IgG) in Copyright © 2022 Massachusetts Medical Society.

serum from patients with Devic’s disease, but not from those with typical multiple
sclerosis, established neuromyelitis optica as a distinct entity with a chronic, re- CME
lapsing course.3-5 Studies of patients who are AQP4-IgG–seropositive have led to the at NEJM.org
recognition that many additional clinical and neuroimaging findings occur, in-
cluding brain involvement, which are captured by the term neuromyelitis optica
spectrum disorder.6,7 The AQP4-IgG autoantibody is detectable in more than 80%
of patients with NMOSD and is pathogenic, initiating the inflammatory CNS lesions
and clinical manifestations of the disease.8,9

Epidemiol o gy
NMOSD accounts for 1 to 2% of all cases of CNS inflammatory demyelinating
disease in the United States and Europe, with multiple sclerosis being much more
common, but NMOSD accounts for one third or more of cases of CNS inflamma-
tion in Asian and other non-White populations. Incidence and prevalence estimates
have ranged from 0.037 to 0.73 cases per 100,000 person-years and 0.7 to 10.0 cases
per 100,000 persons, respectively, but these estimates vary widely among studies,
with the highest rates among Africans and in the Afro-Caribbean region and the
lowest rates among Whites and persons in Australasia, probably reflecting genetic
and environmental influences and differences in ascertainment methods.10 A
population-based study in the United States showed a higher prevalence for Blacks
(13.0 cases per 100,000 persons) than for Whites (4.0 per 100,000).11
The median age at onset of the disorder is 40 years, but the disorder can affect
people at any age, and up to 20% of cases are in children or in adults over the age
of 65 years. Seropositive disease has a female preponderance that approaches 90%,
whereas seronegative cases have an equal sex distribution.10,12 Up to 3% of cases
are familial.13 Whole-genome sequencing has shown that AQP4-IgG is associated
with major-histocompatibility-complex region variants, and there is a causal relation-
ship between known risk variants and systemic lupus erythematosus, which may
coexist with antibody-positive NMOSD.14 It has been suggested that environmental
factors may play a role in the genesis of the disorder, but none have been identified.
In pediatric NMOSD, unlike in multiple sclerosis, exposures to common herpes
viruses, including Epstein–Barr virus, are not implicated.15

n engl j med 387;7  nejm.org  August 18, 2022 631


The New England Journal of Medicine
Downloaded from nejm.org at UNIVERSIDAD PERUANA CAYETANO HEREDIA on August 30, 2022. For personal use only. No other uses without permission.
Copyright © 2022 Massachusetts Medical Society. All rights reserved.
The n e w e ng l a n d j o u r na l of m e dic i n e

Acute attacks of NMOSD, including the initial taken for convulsions. Severe high cervical my-
episode, usually occur in the absence of an iden- elitis can cause neurogenic respiratory failure.2
tified inciting event, but approximately one third Area postrema syndrome is the presenting
of cases are preceded by an infection, which is feature of NMOSD in 10% of patients and occurs
often viral, and in rare cases, an acute attack fol- at some time during the disease in 15 to 40% of
lows vaccination.2 However, no specific infection patients.18,19 Vomiting lasts a median of 2 weeks
or vaccine has been strongly implicated as a trig- and, in up to two thirds of cases, presages an at-
ger of disease activity. The initial attack of NMOSD tack of optic neuritis or myelitis.19 The syndrome
with AQP4-IgG has been associated with the use can accompany myelitis when a destructive cer-
of immune checkpoint inhibitor therapies.16,17 vical lesion ascends into the brain stem, or it
may be isolated and caused by nondestructive
dorsal medullary lesions. When the syndrome is
Cl inic a l Ch a r ac ter is t ic s
isolated, especially as the inaugural presentation
Six core clinical syndromes have been identified of NMOSD, patients are typically evaluated by
in NMOSD. They are classified by their location: internists or gastroenterologists for intractable
optic nerve, spinal cord, area postrema of the vomiting, and the diagnosis may be delayed un-
dorsal medulla, other brain-stem regions, dien- til there are subsequent neurologic symptoms.
cephalon, or cerebrum. These syndromes are The other NMOSD syndromes occur less fre-
manifested as acute attacks and relapses of neu- quently, except that cerebral syndromes are com-
rologic dysfunction, with symptoms that typically mon in children and cause generalized or focal
evolve over a period of days.7 The most common signs, including encephalopathy, hemiparesis,
syndromes are optic neuritis and transverse my- hemianopia, or seizures.7,20 Brain-stem lesions may
elitis, as well as area postrema syndrome, which cause oculomotor dysfunction, hearing loss, ver-
causes intractable or repetitive vomiting and hic- tigo, dysarthria, or other cranial-nerve symptoms;
cups. Multifocal lesions may result in a simulta- as with optic neuritis, these features alone are not
neous or rapidly sequential combination of syn- initially distinguishable from similar syndromes
dromes (e.g., optic neuritis and myelitis). Attacks in multiple sclerosis. Lesions in diencephalic struc-
are identified on the basis of new clinical symp- tures such as the thalamus and hypothalamus
toms and objective neurologic signs (except in the have been reported to cause narcolepsy-like symp-
case of isolated area postrema symptoms) and toms, endocrinopathies, temperature dysregula-
by ruling out other causes, such as infection, that tion, eating disorders, and a syndrome of inap-
can simulate an attack. Detection of new lesions propriate antidiuretic hormone secretion.
on magnetic resonance imaging (MRI) in the rel-
evant neuroanatomical region provides support- Neuroim aging
ive evidence that a neurologic event is due to
NMOSD. Confirmation that an attack is due to NMOSD is
The sentinel attack involves the optic nerve or greatly aided by the detection on MRI of charac-
spinal cord in more than 85% of affected adults. teristic lesions in the brain, optic nerve, optic chi-
Patients with optic neuritis present with unilat- asm, or spinal cord21 (Fig. 1). In the acute phase,
eral or bilateral visual loss or scotoma, dyschro- T2-weighted MRI sequences may show new or
matopsia, and ocular pain exacerbated by eye enlarging inflammatory lesions, and T1-weighted
movement that is not distinguishable from optic sequences obtained with gadolinium may show
neuritis in multiple sclerosis or from an idiopathic enhancement of actively inflamed lesions. Abnor-
form. Acute transverse myelitis causes limb weak- malities on orbital MRI have a predilection for the
ness, numbness, sensory loss or pain below the optic chiasm or the adjacent posterior optic nerve
lesion level, and bladder and bowel dysfunction. but may occupy the full length of the nerve. MRI
Other myelitis symptoms include Lhermitte’s sign of the spinal cord typically shows longitudinally
(trunk or limb paresthesias), elicited by neck flex- extensive transverse myelitis (Fig. 1A), defined as
ion, and episodes of paroxysmal tonic spasms a lesion that is at least three contiguous vertebral
lasting 15 to 60 seconds, with painful agonist segments in length.2,22 Although this pattern is
and antagonist muscle cocontraction, often mis- characteristic of NMOSD, about 15% of sentinel

632 n engl j med 387;7  nejm.org  August 18, 2022

The New England Journal of Medicine


Downloaded from nejm.org at UNIVERSIDAD PERUANA CAYETANO HEREDIA on August 30, 2022. For personal use only. No other uses without permission.
Copyright © 2022 Massachusetts Medical Society. All rights reserved.
Neuromyelitis Optica Spectrum Disorder

myelitis attacks are associated with a shorter le- undergo testing for both antibodies; however,
sion that simulates multiple sclerosis.23 Detection AQP4-IgG and MOG-IgG antibodies rarely coex-
of a dorsal medullary lesion with the use of MRI ist. Patients who test negative for both antibodies
is confirmatory of area postrema syndrome, but may be described as having “double seronegative”
the lesion is small and visible for only a few days.7 NMOSD, a category that could be narrowed with
Seen on MRI scans, focal lesions in the brain stem, the discovery of new autoantibodies.
diencephalon, or cerebrum, even those that are
asymptomatic, may also be indicative of NMOSD.7 Dise a se C our se
Attacks of NMOSD usually reach peak severity in
Di agnosis
several days, plateau, and then spontaneously sub-
International consensus–based criteria for the di- side, frequently leaving moderate-to-severe and
agnosis of NMOSD (Table 1)7 require the manifes- permanent functional deficits.27 In more than
tation of one or more of the six typical syndromes 90% of cases, the disease has a relapsing course,
and emphasize that clinical–radiologic correla- similar to that of multiple sclerosis, with a period
tions and a positive serologic test for AQP4-IgG of months or years between attacks. Neurologic
distinguish NMOSD from multiple sclerosis and impairment is stable or may diminish during
other disorders. About 80% of cases are seroposi- remission, but relapses lead to stepwise accrual
tive, but current criteria allow for the diagnosis of disability. The type, frequency, and severity of
of NMOSD if AQP4-IgG testing is negative or not relapses are influenced by age, sex, and ethnic
available, which may be the case in some low- group.28,29 Data regarding these factors have been
income regions of the world. However, according obtained from retrospective series. Among women
to the international consensus criteria, the diag- with seropositive NMOSD, relapse rates have been
nosis of NMOSD is almost ensured if there is a higher during the first 3 months post partum
characteristic first attack and AQP4-IgG antibodies than in the period before or during pregnancy,
are present.7 and the risk of miscarriage has been increased.30
The standard serum AQP4-IgG reference test Five years after the onset of disease, nearly
is a live cell–based flow-cytometric assay with one quarter of untreated AQP4-IgG–seropositive
more than 80% sensitivity and more than 99% patients require gait assistance, more than 40%
specificity.24,25 False positive low-titer results are blind in at least one eye, and mortality ap-
may occur with the use of enzyme-linked im- proaches 10%.28,31 Hospitalization rates and use
munosorbent assays and, in an atypical case, of health resources are substantial.32 In con-
should prompt reconsideration of the diagnosis trast to relapsing multiple sclerosis, in which
and confirmation with a cell-based assay. False late neurodegeneration and progressive function-
negative AQP4-IgG results can occur if serum is al deterioration may dominate the clinical pic-
sampled during immunosuppressive drug treat- ture, NMOSD rarely has a secondary progressive
ment or after plasma exchange. Retesting at an course.33
interval after discontinuation of treatment or dur-
ing a relapse is recommended in such cases.7 Test- A sso ci at ions w i th O ther
ing for cerebrospinal fluid AQP4-IgG is relatively Disor der s
insensitive.
The categorization of NMOSD with negative Up to half of patients with NMOSD and AQP4-IgG
or unknown AQP4-IgG status requires stringent have other detectable serum autoantibodies
clinical and MRI criteria and a search for other (e.g., thyroperoxidase, antinuclear, and Ro/SS-A
causes of the CNS symptoms.7 The most common antibodies), and one third have an autoimmune
alternative diagnosis is a CNS demyelinating dis- disease, most commonly thyroiditis, systemic
order associated with serum antibodies to myelin lupus erythematosus, or Sjögren’s syndrome.2 It
oligodendrocyte glycoprotein (MOG-IgG). This is advisable to perform AQP4-IgG testing before
disorder overlaps with both AQP4-IgG–seroposi- making a diagnosis of “lupus myelitis” or other
tive NMOSD and multiple sclerosis, both of which CNS complications of rheumatologic disease, be-
also can cause optic neuritis or myelitis.26 Pa- cause AQP4-IgG seropositivity usually indicates
tients with NMOSD clinical syndromes generally the coexistence of one of these diseases and

n engl j med 387;7  nejm.org  August 18, 2022 633


The New England Journal of Medicine
Downloaded from nejm.org at UNIVERSIDAD PERUANA CAYETANO HEREDIA on August 30, 2022. For personal use only. No other uses without permission.
Copyright © 2022 Massachusetts Medical Society. All rights reserved.
The n e w e ng l a n d j o u r na l of m e dic i n e

A B C D

E F G

H I J

NMOSD, with NMOSD usually accounting for Pathol o gic a l Fe at ur e s


the CNS syndrome.34 Up to 5% of antibody-pos- a nd Patho gene sis
itive NMOSD cases are paraneoplastic, and a
search for an occult malignant process may be AQP4 is an integral membrane water channel
considered in patients with risk factors for can- protein expressed by many cell types, including
cer, particularly patients in whom NMOSD de- gastrointestinal, lung, retinal, renal, and muscle
velops at a late age. cells. In the CNS, AQP4 is expressed on astro-

634 n engl j med 387;7 nejm.org August 18, 2022

The New England Journal of Medicine


Downloaded from nejm.org at UNIVERSIDAD PERUANA CAYETANO HEREDIA on August 30, 2022. For personal use only. No other uses without permission.
Copyright © 2022 Massachusetts Medical Society. All rights reserved.
Neuromyelitis Optica Spectrum Disorder

Figure 1 (facing page). MRI Patterns in Neuromyelitis Table 1. Core Clinical Characteristics and Diagnostic Criteria for
Optica Spectrum Disorder (NMOSD). Neuromyelitis Optica Spectrum Disorder (NMOSD).*
A sagittal T2-weighted scan shows a longitudinally ex-
tensive cervical spinal cord lesion that extends into the Core clinical characteristics
medulla and has been considered relatively characteris- The six core clinical characteristics are optic neuritis, acute myelitis, area
tic of NMOSD (Panel A); the lesion is associated with postrema syndrome, acute brain-stem syndrome (with associated
an area of ringlike contrast enhancement on T1-weight- periependymal brain-stem lesions on MRI in AQP4-IgG–negative
ed imaging (Panel B). Axial T1-weighted sequences patients), symptomatic narcolepsy or acute diencephalic clinical
with contrast material show increased signal along the syndrome with diencephalic lesions on MRI that are characteristic
length of the left optic nerve (Panel C) and optic chi- of NMOSD, and symptomatic cerebral syndrome with brain lesions
that are characteristic of NMOSD
asm (Panel D). A sagittal fluid-attenuated inversion re-
covery (FLAIR) sequence shows increased signal in the Criteria for NMOSD with AQP4-IgG
dorsal medulla (Panel E), which is associated with area One or more of the six core clinical characteristics
postrema syndrome. A pair of small foci of contrast en-
hancement on T1-weighted sequences confirms a re- Positive serum test for AQP4-IgG with the use of the best available detection
method (cell-based assay strongly recommended)
lapse of acute area postrema syndrome (Panel F). An
axial FLAIR sequence shows diencephalic lesions in- No alternative diagnoses
volving the hypothalamus (Panel G). Characteristic Criteria for NMOSD without AQP4-IgG or with unknown antibody status
brain lesions include large, confluent white-matter le-
sions, seen on an axial FLAIR sequence (Panel H), with Two or more of the six core clinical characteristics resulting from one or
cloudlike contrast enhancement on an axial T1-weight- more clinical attacks; must include at least one of the following three
core clinical characteristics:
ed, contrast-enhanced sequence (Panel I), and lesions
involving the long axis of the corpus callosum, seen on Acute optic neuritis with either no abnormalities or only nonspecific
a sagittal FLAIR sequence (Panel J). The arrows in the white-matter lesions in the brain on MRI, or with orbital MRI show-
panels point to lesions. Additional images are shown in ing a lesion extending over more than half the optic-nerve length or
involving the optic chiasm
Figure S1 in the Supplementary Appendix, available
with the full text of this article at NEJM.org. Acute myelitis with a longitudinally extensive cord lesion on MRI
Area postrema syndrome with a dorsal medullary lesion on MRI
Negative test (or tests) for AQP4-IgG with use of best available detection
cytic end-feet that abut endothelial cells, which method or testing unavailable
form the glia limitans component of the blood– No alternative diagnoses
brain barrier. When the barrier is breached, or
in regions such as the area postrema where it is * Diagnostic criteria are from Wingerchuk et al.7 AQP4-IgG denotes IgG-class
lacking, circulating AQP4-IgG gains access and antibodies against aquaporin-4.
binds to its target antigen, initiating inflamma-
tory responses that generate NMOSD lesions. The AQP4-IgG can originate.41 Cross-reactivity with
pathological hallmark of NMOSD with AQP4-IgG bacterial antigens resembling AQP4 or, in para-
is a focal inflammatory CNS lesion with marked neoplastic cases, initiation of an antibody-medi-
or complete loss of AQP4.35,36 Human immuno- ated response to tumor-cell expression of AQP4
pathological findings support the concept that may be the originating mechanism. Complement
NMOSD is an autoimmune astrocytopathy9 and activation is an immediate consequence of the
that AQP4-specific autoantibodies initiate lytic selective binding of AQP4-IgG to its antigen. Ac-
and sublytic astrocyte disease with variable po- tivated complement, together with interleukin-6,
tential for reversibility.35,37,38 Animal and tissue- increases the permeability of the blood–brain bar-
based models that use patient-derived monoclo- rier and is a chemoattractant, signaling eosino-
nal antibodies can reproduce some characteristics phils and neutrophils to enter the evolving lesion
of NMOSD, further supporting the pathogenic and degranulate. Astrocyte injury occurs from
role of AQP4-IgG.39 the effects of C5 and membrane attack complex
The current concept of NMOSD immunopatho- components of complement. Demyelination oc-
genesis is outlined in Figure S2 in the Supplemen- curs as a secondary consequence of myelinolysis
tary Appendix, available with the full text of this and oligodendrocyte injury.42
article at NEJM.org.40 The putative steps in the Despite widespread tissue expression of
pathogenesis begin with immune tolerance check- AQP4, extra-CNS clinical disease such as myosi-
point defects that allow the development of a set tis is rare.43 One potential explanation for selec-
of autoreactive B cells from which pathogenic tive CNS vulnerability is that the CNS lacks

n engl j med 387;7  nejm.org  August 18, 2022 635


The New England Journal of Medicine
Downloaded from nejm.org at UNIVERSIDAD PERUANA CAYETANO HEREDIA on August 30, 2022. For personal use only. No other uses without permission.
Copyright © 2022 Massachusetts Medical Society. All rights reserved.
The n e w e ng l a n d j o u r na l of m e dic i n e

Table 2. Treatment for Prevention of NMOSD Relapse.*

Drug† Mechanism of Action Route and Dose Side Effects Key Risks
Prednisone Immunosuppression Oral; initial dose: 40–80 mg Dose-related systemic Gastrointestinal bleeding
daily; maintenance dose: glucocorticoid
5–30 mg daily effects
Azathioprine T-cell and B-cell suppression Oral; 2–3 mg/kg daily Nausea, vomiting, Leukopenia, liver toxicity, infec-
fatigue tions, cancer
Mycophenolate T-cell and B-cell suppression Oral; 1–3 g daily Nausea, diarrhea Leukopenia, hypertension, tera-
mofetil togenicity; REMS program
required for reproductive risks
Rituximab B-cell depletion; anti-CD20 Intravenous; initial dose: 1 g, Infusion reactions Infections, hepatitis B reactiva-
monoclonal antibody repeated after 2 wk; main- tion, immunoglobulin defi-
tenance dose: 0.5–2 g at ciency with prolonged use,
6-mo intervals reduced vaccine response
Eculizumab Complement inhibition; anti- Intravenous; initial dose: Nausea, headache, Meningococcal vaccine and
C5 monoclonal antibody 900 mg weekly for 4 wk and back pain REMS program required for
1200 mg 1 wk later; main- risk of meningococcal infec-
tenance dose: 1200 mg at tion‡
2-wk intervals
Inebilizumab B-cell depletion; anti-CD19 Intravenous; initial dose: Infusion reactions Infections, immunoglobulin
monoclonal antibody 300 mg, repeated after deficiency with prolonged
2 wk; maintenance dose: use, reduced vaccine re-
300 mg at 6-mo intervals sponse‡
Satralizumab Interleukin-6 inhibition; Subcutaneous; initial dose: Injection-site reac- Infections, neutropenia, amino-
anti–interleukin-6 receptor 120 mg at wk 0, 2, and 4; tions, nasophar- transferase and lipid abnor-
monoclonal antibody maintenance dose: 120 mg yngitis, headache, malities‡
at 4-wk intervals arthralgia

* REMS denotes Risk Evaluation and Mitigation Strategy.


† Eculizumab, inebilizumab, and satralizumab are approved by the Food and Drug Administration for relapse prevention in adults who have
NMOSD with AQP4-IgG. There are no approved therapies for NMOSD in the absence of AQP4-IgG.
‡ Eculizumab is contraindicated in patients with unresolved Neisseria meningitidis infection. Inebilizumab and satralizumab are contraindi-
cated in patients with active hepatitis B and those with active or latent tuberculosis.

protective complement regulatory proteins that Preventive Immunotherapy


are coexpressed with AQP4 in peripheral tissues The principal goal of NMOSD management is
such as the kidney.44 prevention of relapse, both for patients who are
AQP4-IgG–seropositive at the initial presentation,
who have more than a 70% risk of relapse in the
T r e atmen t
subsequent year, and for all patients, whether
Acute Relapses and Symptoms seropositive or seronegative for AQP4-IgG, who
Acute relapses are initially treated with intra- have an established relapsing course. Prevention
venous glucocorticoids, but one study has of relapse is expected to preserve long-term neuro-
shown short-term remission of symptoms in logic function because the accumulation of lesions
only 19% of patients treated with these drugs.27 with each attack would be averted; a secondary
Moderate-to-severe relapses, including those progressive course of NMOSD, independent of
that respond incompletely to glucocorticoids, relapses, is rare.
may be ameliorated with early administration Available preventive therapies are listed in
of plasma exchange.27 Management of residual Table 2. Until 2019, there were no approved drugs
effects of relapses such as gait impairment, for NMOSD, but observational data suggested
weakness and spasticity, neuropathic pain, low that immunosuppression reduces the frequency
vision, neurogenic bladder and bowel, and cog- of relapse among both AQP4-IgG–seropositive
nitive and mood disorders can improve the patients and AQP4-IgG–seronegative patients,45,46
quality of life. and one small, randomized, placebo-controlled

636 n engl j med 387;7  nejm.org  August 18, 2022

The New England Journal of Medicine


Downloaded from nejm.org at UNIVERSIDAD PERUANA CAYETANO HEREDIA on August 30, 2022. For personal use only. No other uses without permission.
Copyright © 2022 Massachusetts Medical Society. All rights reserved.
Neuromyelitis Optica Spectrum Disorder

trial showed a benefit of rituximab in seroposi- body-producing plasmablasts and plasma cells,
tive patients.47 The most frequently used drugs than anti-CD20 rituximab. In a phase 2–3 trial
have been rituximab, mycophenolate mofetil, and that included AQP4-IgG–seropositive and AQP4-
azathioprine. Many patients who do not have ac- IgG–seronegative patients and did not permit
cess to recently approved drugs (discussed below) concomitant treatment with immunosuppressive
continue to receive these therapies if they have drugs, the risk of relapse was reduced by 73%
been in prolonged remission with one of these overall and by 77% among seropositive patients.50
treatments or if they have seronegative, relapsing Inebilizumab reduced the rates of accumulating
NMOSD; these drugs are also used in children. disability, accumulating lesions on brain MRI,
Low-dose prednisone (or another glucocorticoid), and hospitalization. Two patients died, one from
methotrexate, mitoxantrone, or cyclophosphamide a relapse of NMOSD and the other from an in-
is used for relapse prevention in various regions determinate CNS cause.
of the world. Satralizumab targets the interleukin-6 recep-
Three approved monoclonal antibody therapies tor and thereby inhibits the downstream effects
for adults with NMOSD and AQP4-IgG autoanti- of interleukin-6; another drug targeting this re-
bodies48 are eculizumab, inebilizumab, and sa- ceptor, tocilizumab, reduced the risk of relapse
tralizumab; each targets steps in the immuno- as compared with azathioprine in an open-label,
pathogenesis of NMOSD and has been tested in randomized trial.51 Two trials of satralizumab for
trials. In randomized, double-blind, placebo- the treatment of patients who have NMOSD with
controlled trials, the time to the first adjudicated or without AQP4-IgG have been completed, one
relapse has been used as the primary outcome. of which allowed concurrent immunosuppres-
Differences in trial design have included enroll- sive therapy.52,53 Efficacy was achieved only in the
ment criteria (i.e., whether enrollment has been seropositive groups in the two studies, with a 74%
restricted to AQP4-IgG–positive patients), attack and a 79% reduction in relapse risk, and no deaths
definition, and the use of placebo alone or pla- or cancers occurred.
cebo added to existing therapy as the comparator. Considerations in selecting one of the recently
In one trial, eculizumab, which inhibits C5 and approved preventive therapies include efficacy,
presumably the formation of the membrane at- problems with current treatment (side effects or
tack complex mediated by C5b, reduced the risk lack of a response), safety, frequency and route
of relapse by 94% as compared with placebo in of administration, potential for adherence, effect
AQP4-IgG–seropositive adults, some of whom on vaccine response, planning for conception,
were receiving concomitant immunosuppressive drug access, and cost. Several drugs appear to be
agents.49 No formal inferences regarding overall relatively safe when taken during pregnancy, but
disability or quality-of-life outcomes could be the data are limited.30 Aside from confirmation
made, because the period of follow-up after a of eligibility on the basis of AQP4-IgG–seroposi-
relapse was limited (6 weeks) and the between- tive status, there are no biomarkers that facilitate
group difference in a measure of disability pro- drug selection.
gression was not significant. Patients receiving Extension studies for approved drugs have
eculizumab as monotherapy accounted for 24% of not identified major additional safety concerns
the trial participants, and the reduction in the and have shown low relapse rates, but inferences
risk of relapse in this group was similar to the about sustained efficacy and long-term risks are
reduction in the group of patients who received limited because these studies were unblinded for
eculizumab in addition to an established immu- the participants and investigators, observations
nosuppressive therapy. Upper respiratory tract have been uncontrolled, and the longest extension
infections and headache were more common in study followed treated patients for a mean of 4 to
the eculizumab group. There were no infections 5 years.54,55 Monitoring of AQP4-IgG titers or le-
by encapsulated organisms such as Neisseria men- sions seen on MRI is not currently recommended
ingitidis; infections are a known risk of comple- for therapeutic decision making, but serum bio-
ment inhibition. One death occurred from pulmo- markers such as glial fibrillary acidic protein are
nary empyema. being studied for confirming or predicting clin-
Inebilizumab targets CD19; it depletes a broad- ical relapse.56 Breakthrough relapses usually
er spectrum of the B-cell lineage, including anti- prompt changes in therapy, possibly a switch to

n engl j med 387;7  nejm.org  August 18, 2022 637


The New England Journal of Medicine
Downloaded from nejm.org at UNIVERSIDAD PERUANA CAYETANO HEREDIA on August 30, 2022. For personal use only. No other uses without permission.
Copyright © 2022 Massachusetts Medical Society. All rights reserved.
The n e w e ng l a n d j o u r na l of m e dic i n e

a drug with a different mechanism of action, but Sum m a r y


a consensus definition of treatment failure and
more data are needed. Combination therapy, other The canonical clinical features of neuromyelitis
immunosuppressive agents, and autologous stem- optica have been supplemented by other charac-
cell transplantation57 have been considered for teristic syndromes and the finding of pathogenic
treatment-refractory disease. Indefinite treatment AQP4-IgG autoantibodies. The expanded disease
is recommended for NMOSD with AQP4-IgG, spectrum, NMOSD, is treated with drugs that tar-
since retrospective data suggest that stopping pre- get the steps in pathogenesis. Promising approach-
ventive therapies leads to high relapse rates.58 es to predicting and preventing relapses, enhanc-
Research on new treatments, guided by an ing recovery from relapse, and achieving immune
understanding of the pathobiologic features of tolerance are under investigation.
NMOSD, is focused on approaches to the resto-
ration of immune tolerance; inhibition of AQP4- Disclosure forms provided by the authors are available with
the full text of this article at NEJM.org.
IgG binding or pathogenicity; targeting of the ef- We thank Mr. Seth Lambert and Mr. Bryce Bergene, Mayo
fects of complement, granulocytes, and microglia; Clinic, for assistance with earlier versions of the figures.
and neuroprotection.59,60

References
1. Devic E. Myélite aiguë compliquée de autoimmunity and neuromyelitis optica optica spectrum disorder: an internation-
névrite optique. Bull Med (Paris) 1894;​8:​ spectrum. Ann Neurol 2016;​79:​775-83. al update. Neurology 2015;​84:​1165-73.
1033-4. 12. Jarius S, Ruprecht K, Wildemann B, et 22. Ciccarelli O, Cohen JA, Reingold SC,
2. Wingerchuk DM, Hogancamp WF, al. Contrasting disease patterns in sero- et al. Spinal cord involvement in multiple
O’Brien PC, Weinshenker BG. The clinical positive and seronegative neuromyelitis sclerosis and neuromyelitis optica spec-
course of neuromyelitis optica (Devic’s optica: a multicentre study of 175 pa- trum disorders. Lancet Neurol 2019;​ 18:​
syndrome). Neurology 1999;​53:​1107-14. tients. J Neuroinflammation 2012;​9:​14. 185-97.
3. Lennon VA, Wingerchuk DM, Kryzer 13. Matiello M, Kim HJ, Kim W, et al. Fa- 23. Flanagan EP, Weinshenker BG,
TJ, et al. A serum autoantibody marker of milial neuromyelitis optica. Neurology Krecke KN, et al. Short myelitis lesions in
neuromyelitis optica: distinction from mul- 2010;​75:​310-5. aquaporin-4-IgG-positive neuromyelitis
tiple sclerosis. Lancet 2004;​364:​2106-12. 14. Estrada K, Whelan CW, Zhao F, et al. optica spectrum disorders. JAMA Neurol
4. Lennon VA, Kryzer TJ, Pittock SJ, A whole-genome sequence study identi- 2015;​72:​81-7.
Verkman AS, Hinson SR. IgG marker of fies genetic risk factors for neuromyelitis 24. Waters P, Reindl M, Saiz A, et al. Mul-
optic-spinal multiple sclerosis binds to optica. Nat Commun 2018;​9:​1929. ticentre comparison of a diagnostic assay:
the aquaporin-4 water channel. J Exp Med 15. Graves J, Grandhe S, Weinfurtner K, aquaporin-4 antibodies in neuromyelitis
2005;​202:​473-7. et al. Protective environmental factors for optica. J Neurol Neurosurg Psychiatry
5. Wingerchuk DM, Lennon VA, Pittock neuromyelitis optica. Neurology 2014;​83:​ 2016;​87:​1005-15.
SJ, Lucchinetti CF, Weinshenker BG. Re- 1923-9. 25. Redenbaugh V, Montalvo M, Sechi E,
vised diagnostic criteria for neuromyelitis 16. Narumi Y, Yoshida R, Minami Y, et al. et al. Diagnostic value of aquaporin-4-IgG
optica. Neurology 2006;​66:​1485-9. Neuromyelitis optica spectrum disorder live cell based assay in neuromyelitis op-
6. Wingerchuk DM, Lennon VA, Lucchi- secondary to treatment with anti-PD-1 an- tica spectrum disorders. Mult Scler J Exp
netti CF, Pittock SJ, Weinshenker BG. The tibody nivolumab: the first report. BMC Transl Clin 2021;​7:​20552173211052656.
spectrum of neuromyelitis optica. Lancet Cancer 2018;​18:​95. 26. Marignier R, Hacohen Y, Cobo-Calvo
Neurol 2007;​6:​805-15. 17. Weiss D, Cantré D, Zettl UK, Storch A, A, et al. Myelin-oligodendrocyte glycopro-
7. Wingerchuk DM, Banwell B, Bennett Prudlo J. Lethal form of a late-onset aqua- tein antibody-associated disease. Lancet
JL, et al. International consensus diag- porin-4 antibody-positive NMOSD related Neurol 2021;​20:​762-72.
nostic criteria for neuromyelitis optica to the immune checkpoint inhibitor 27. Kleiter I, Gahlen A, Borisow N, et al.
spectrum disorders. Neurology 2015;​85:​ nivolumab. J Neurol 2022;​269:​2778-80. Neuromyelitis optica: evaluation of 871
177-89. 18. Misu T, Fujihara K, Nakashima I, attacks and 1,153 treatment courses. Ann
8. Lucchinetti CF, Mandler RN, McGavern Sato S, Itoyama Y. Intractable hiccup and Neurol 2016;​79:​206-16.
D, et al. A role for humoral mechanisms nausea with periaqueductal lesions in 28. Kitley J, Leite MI, Nakashima I, et al.
in the pathogenesis of Devic’s neuromye- neuromyelitis optica. Neurology 2005;​65:​ Prognostic factors and disease course in
litis optica. Brain 2002;​125:​1450-61. 1479-82. aquaporin-4 antibody-positive patients
9. Lucchinetti CF, Guo Y, Popescu BF, 19. Shosha E, Dubey D, Palace J, et al. with neuromyelitis optica spectrum dis-
Fujihara K, Itoyama Y, Misu T. The pathol- Area postrema syndrome: frequency, cri- order from the United Kingdom and Ja-
ogy of an autoimmune astrocytopathy: teria, and severity in AQP4-IgG-positive pan. Brain 2012;​135:​1834-49.
lessons learned from neuromyelitis opti- NMOSD. Neurology 2018;​91(17):​e1642- 29. Kim S-H, Mealy MA, Levy M, et al.
ca. Brain Pathol 2014;​24:​83-97. e1651. Racial differences in neuromyelitis optica
10. Papp V, Magyari M, Aktas O, et al. 20. Chitnis T, Ness J, Krupp L, et al. Clin- spectrum disorder. Neurology 2018;​91(22):​
Worldwide incidence and prevalence of ical features of neuromyelitis optica in e2089-e2099.
neuromyelitis optica: a systematic review. children: US Network of Pediatric MS Cen- 30. Mao-Draayer Y, Thiel S, Mills EA, et
Neurology 2021;​96:​59-77. ters report. Neurology 2016;​86:​245-52. al. Neuromyelitis optica spectrum disor-
11. Flanagan EP, Cabre P, Weinshenker 21. Kim HJ, Paul F, Lana-Peixoto MA, et ders and pregnancy: therapeutic consider-
BG, et al. Epidemiology of aquaporin-4 al. MRI characteristics of neuromyelitis ations. Nat Rev Neurol 2020;​16:​154-70.

638 n engl j med 387;7  nejm.org  August 18, 2022

The New England Journal of Medicine


Downloaded from nejm.org at UNIVERSIDAD PERUANA CAYETANO HEREDIA on August 30, 2022. For personal use only. No other uses without permission.
Copyright © 2022 Massachusetts Medical Society. All rights reserved.
Neuromyelitis Optica Spectrum Disorder

31. Jiao Y, Fryer JP, Lennon VA, et al. Up- 42. Pittock SJ, Lucchinetti CF. Neuromye- 52. Yamamura T, Kleiter I, Fujihara K, et
dated estimate of AQP4-IgG serostatus litis optica and the evolving spectrum of al. Trial of satralizumab in neuromyelitis
and disability outcome in neuromyelitis autoimmune aquaporin-4 channelopa- optica spectrum disorder. N Engl J Med
optica. Neurology 2013;​81:​1197-204. thies: a decade later. Ann N Y Acad Sci 2019;​381:​2114-24.
32. Ajmera MR, Boscoe A, Mauskopf J, 2016;​1366:​20-39. 53. Traboulsee A, Greenberg BM, Bennett
Candrilli SD, Levy M. Evaluation of co- 43. Guo Y, Lennon VA, Popescu BF, et al. JL, et al. Safety and efficacy of satrali-
morbidities and health care resource use Autoimmune aquaporin-4 myopathy in zumab monotherapy in neuromyelitis
among patients with highly active neuro- neuromyelitis optica spectrum. JAMA optica spectrum disorder: a randomised,
myelitis optica. J Neurol Sci 2018;​384:​96- Neurol 2014;​71:​1025-9. double-blind, multicentre, placebo-con-
103. 44. Yao X, Verkman AS. Complement reg- trolled phase 3 trial. Lancet Neurol 2020;​
33. Wingerchuk DM, Pittock SJ, Lucchi- ulator CD59 prevents peripheral organ 19:​402-12.
netti CF, Lennon VA, Weinshenker BG. A injury in rats made seropositive for neuro- 54. Wingerchuk DM, Fujihara K, Palace J,
secondary progressive clinical course is myelitis optica immunoglobulin G. Acta et al. Long-term safety and efficacy of
uncommon in neuromyelitis optica. Neu- Neuropathol Commun 2017;​5:​57. eculizumab in aquaporin-4 IgG-positive
rology 2007;​68:​603-5. 45. Poupart J, Giovannelli J, Deschamps NMOSD. Ann Neurol 2021;​89:​1088-98.
34. Pittock SJ, Lennon VA, de Seze J, et al. R, et al. Evaluation of efficacy and tolera- 55. Rensel M, Zabeti A, Mealy MA, et al.
Neuromyelitis optica and non organ-spe- bility of first-line therapies in NMOSD. Long-term efficacy and safety of inebili-
cific autoimmunity. Arch Neurol 2008;​65:​ Neurology 2020;​94(15):​e1645-e1656. zumab in neuromyelitis optica spectrum
78-83. 46. Mealy MA, Wingerchuk DM, Palace J, disorder: analysis of aquaporin-4-immu-
35. Roemer SF, Parisi JE, Lennon VA, et Greenberg BM, Levy M. Comparison of noglobulin G-seropositive participants
al. Pattern-specific loss of aquaporin-4 relapse and treatment failure rates among taking inebilizumab for ≥4 years in the
immunoreactivity distinguishes neuro- patients with neuromyelitis optica: multi- N-MOmentum trial. Mult Scler 2022;​28:​
myelitis optica from multiple sclerosis. center study of treatment efficacy. JAMA 925-32.
Brain 2007;​130:​1194-205. Neurol 2014;​71:​324-30. 56. Aktas O, Smith MA, Rees WA, et al.
36. Misu T, Fujihara K, Kakita A, et al. 47. Tahara M, Oeda T, Okada K, et al. Serum glial fibrillary acidic protein: a
Loss of aquaporin 4 in lesions of neuro- Safety and efficacy of rituximab in neuro- neuromyelitis optica spectrum disorder
myelitis optica: distinction from multiple myelitis optica spectrum disorders (RIN-1 biomarker. Ann Neurol 2021;​89:​895-910.
sclerosis. Brain 2007;​130:​1224-34. study): a multicentre, randomised, dou- 57. Burt RK, Balabanov R, Han X, et al.
37. Takai Y, Misu T, Suzuki H, et al. Stag- ble-blind, placebo-controlled trial. Lancet Autologous nonmyeloablative hematopoi-
ing of astrocytopathy and complement Neurol 2020;​19:​298-306. etic stem cell transplantation for neuro-
activation in neuromyelitis optica spec- 48. Levy M, Fujihara K, Palace J. New myelitis optica. Neurology 2019;​ 93(18):​
trum disorders. Brain 2021;​144:​2401-15. therapies for neuromyelitis optica spectrum e1732-e1741.
38. Guo Y, Lennon VA, Parisi JE, et al. disorder. Lancet Neurol 2021;​20:​60-7. 58. Kim SH, Jang H, Park NY, et al. Dis-
Spectrum of sublytic astrocytopathy in 49. Pittock SJ, Fujihara K, Palace J, et al. continuation of immunosuppressive ther-
neuromyelitis optica. Brain 2022;​ 145:​ Eculizumab monotherapy for NMOSD: apy in patients with neuromyelitis optica
1379-90. data from PREVENT and its open-label spectrum disorder with aquaporin-4 anti-
39. Duan T, Verkman AS. Experimental extension. Mult Scler 2022;​28:​480-6. bodies. Neurol Neuroimmunol Neuroin-
animal models of aquaporin-4-IgG-sero- 50. Cree BAC, Bennett JL, Kim HJ, et al. flamm 2021;​8(2):​e947.
positive neuromyelitis optica spectrum Inebilizumab for the treatment of neu- 59. Bar-Or A, Steinman L, Behne JM, et
disorders: progress and shortcomings. romyelitis optica spectrum disorder al. Restoring immune tolerance in neuro-
Brain Pathol 2020;​30:​13-25. (N-MOmentum): a double-blind, ran- myelitis optica: part II. Neurol Neuroim-
40. Bennett JL, Owens GP. Neuromyelitis domised placebo-controlled phase 2/3 munol Neuroinflamm 2016;​3(5):​e277.
optica: deciphering a complex immune- trial. Lancet 2019;​394:​1352-63. 60. Tradtrantip L, Asavapanumas N,
mediated astrocytopathy. J Neuroophthal- 51. Zhang C, Zhang M, Qiu W, et al. Safe- Verkman AS. Emerging therapeutic tar-
mol 2017;​37:​291-9. ty and efficacy of tocilizumab versus aza- gets for neuromyelitis optica spectrum
41. Cotzomi E, Stathopoulos P, Lee CS, et thioprine in highly relapsing neuromyeli- disorder. Expert Opin Ther Targets 2020;​
al. Early B cell tolerance defects in neuro- tis optica spectrum disorder (TANGO): an 24:​219-29.
myelitis optica favour anti-AQP4 autoanti- open-label, multicentre, randomised, phase Copyright © 2022 Massachusetts Medical Society.
body production. Brain 2019;​142:​1598-615. 2 trial. Lancet Neurol 2020;​19:​391-401.

n engl j med 387;7  nejm.org  August 18, 2022 639


The New England Journal of Medicine
Downloaded from nejm.org at UNIVERSIDAD PERUANA CAYETANO HEREDIA on August 30, 2022. For personal use only. No other uses without permission.
Copyright © 2022 Massachusetts Medical Society. All rights reserved.

You might also like