Neuromyelitis Optica Spectrum Disorders10
Neuromyelitis Optica Spectrum Disorders10
Neuromyelitis Optica Spectrum Disorders10
Spectrum Disorders C O N T I N U UM A U D I O
I NT E R V I E W A V A I L AB L E
ONLINE
By Fiona Costello, MD, FRCPC
ABSTRACT
PURPOSE OF REVIEW: This article reviews the cardinal clinical features, distinct
immunopathology, current diagnostic criteria, relapse-related risk factors,
emerging biomarkers, and evolving treatment strategies pertaining to
neuromyelitis optica spectrum disorders (NMOSD).
N
euromyelitis optica spectrum disorders (NMOSD), previously agents including azathioprine,
cyclophosphamide, IV
referred to as neuromyelitis optica (NMO), has been an enigmatic
immunoglobulin G,
condition since it was first described more than a century ago by methotrexate, mitoxantrone,
Eugene Devic, for whom it was originally named.1 For many years, mycophenolate mofetil,
rituximab, and tocilizumab for
NMO was considered an overlap syndrome with multiple sclerosis the treatment of neuromyelitis
(MS), distinguished from the latter only by its relative severity and specific optica spectrum disorders.
predilection for optic nerve and spinal cord involvement.2 The recognition of
NMOSD as an autoimmune astrocytopathy has been relatively recent. In 2004, © 2022 American Academy
Lennon and colleagues3 discovered serum antibodies that target the water of Neurology.
CONTINUUMJOURNAL.COM 1131
FIGURE 7-1
Schematic of the immune pathophysiology of neuromyelitis optica spectrum disorder
(NMOSD), myelin oligodendrocyte glycoprotein (MOG), glial fibrillary acidic protein (GFAP),
and multiple sclerosis (MS) optic neuritis. NMOSD: Aquaporin-4 (AQP4)-immunoglobulin
G (IgG) enters through defects in the blood-brain barrier (BBB), binds to astrocytes (ASTs),
and initiates complement dependent cytotoxicity (CDC) by assembling complexes for
complement C1q (C1q) binding. AQP4-IgG also activates antibody-dependent cell-mediated
cytotoxicity (ADCC) by natural killer (NK) cells, and complement products stimulate
degranulation of polymorphonuclear cells (PMNs). Membrane attack complex (MAC) may
transit to adjacent oligodendrocytes, resulting in damage represented histopathologically
by myelin vesiculation. Degenerating ASTs alter oligodendrocyte physiology, resulting in
axonal swelling. Myelin debris is removed by infiltrating macrophages (MΦ). MOG:
Perivenous and confluent demyelination are mediated by combined humoral and cellular
mechanisms. CD4-lymphocyte and granulocytic infiltrates emerging from venous and
meningeal sources result in focal and confluent regions of demyelination highlighted by
nascently demyelinated axons with split myelin sheaths and vesiculation, myelin-laden
macrophages within active demyelinating regions, and activated microglia (MG) in the
periplaque area. Peripherally generated MOG-IgG may contribute to myelin destruction
through CDC and ADCC, as well as activated T-cell infiltration by facilitating phagocytosis
and antigen presentation. Perivenous MAC deposition and diffuse myelin protein loss are
histologic features supporting diffuse antibody-mediated myelin destruction. GFAP: GFAP
papillitis results from secondary axonal swelling. GFAP-IgG is predominantly generated
intrathecally; however, its role in driving disease pathology is unclear. Animal models
demonstrate predominantly perivascular, meningeal, and vascular CD8 T-cell infiltrates.
MS: Active MS lesions are characterized by the deposition of complement and immunoglobulin.
The perivenous inflammatory infiltrates are mainly composed of CD8+ T cells and B cells
producing intrathecal IgG in association with activated microglia and macrophages. MAC
complexes are observed along myelin sheaths and within myelin-laden MΦ, suggestive of
active CDC.
Reprinted with permission from DeLott L, et al, J Neurol.13 © 2022 Springer-Verlag GmbH.
AN AUTOIMMUNE ASTROCYTOPATHY
A detailed review of NMOSD immunopathology is beyond the scope of this
review, yet it is important to highlight immune pathophysiologic aspects that
distinguish this condition from other inflammatory disorders of the central
nervous system (CNS) such as MS, myelin oligodendrocyte glycoprotein
(MOG)-IgG–associated disorders, and glial fibrillary acidic protein (GFAP)
astrocytopathy (FIGURE 7-1).13 As a point of clarification, NMOSD is no longer
considered an especially severe inflammatory demyelinating disease of the CNS
but is recognized instead as an autoimmune astrocytopathy.2,4,6,14 Specifically,
the pathologic lesions in NMOSD are heterogeneous and different from MS,14
showing more extensive loss of immunostaining for AQP4 and GFAP than
myelin damage.2 Owing to severe astrocyte injury, GFAP levels are higher in the
CSF of seropositive patients with NMOSD during acute exacerbations compared
with levels seen with MS relapses.2 The binding of APQ4-IgG antibody to
AQP4 on astrocytic foot processes initiates the complement pathway, with
immunologic consequences that ultimately disrupt the blood-brain barrier,
CONTINUUMJOURNAL.COM 1133
FIGURE 7-2
Two cases of transverse myelitis associated with neuromyelitis optica spectrum disorder
(NMOSD). A 21-year-old woman presented with a 1-week history of hiccups, a burning
sensation in her neck, trunk, and legs, decreased rectal tone, and lower limb weakness.
A, Sagittal T2-weighted MRI shows poorly delineated hyperintense signal changes within an
enlarged spinal cord extending over eight to nine spinal segments (longitudinally extensive
transverse myelitis). A hyperintense dorsal medulla/area postrema lesion is also seen (arrow).
B, Sagittal postcontrast T1-weighted MRI shows patchy enhancement of the spinal cord
lesion. As a second example, a 37-year-old man presented with left monocular vision loss
and progressive upper extremity weakness. C, Axial fluid-attenuated inversion recovery
(FLAIR) MRI shows hyperintense signal change in the left retrobulbar optic nerve (arrow).
D, Sagittal T2-weighted MRI shows hyperintense signal changes in the cervical cord
extending over five to six spinal segments. The lesion occupied more than two-thirds of the
cross-sectional diameter of the cord and did not enhance (not shown).
Reprinted with permission from Costello F and Scott JN, Continuum (Minneap Minn).21
© 2019 American Academy of Neurology.
The patient’s clinical presentation and course can provide important clues.
Symptoms of NMOSD are not hyperacute in onset (nadir reached within
4 hours) nor are they protracted (worsening over 4 or more weeks).9
Furthermore, neurologic deficits in patients with NMOSD are tied to relapses as
opposed to interevent progression, as is seen in individuals with MS. Therefore,
neurologic deficits that progress over weeks to months would be atypical for
CONTINUUMJOURNAL.COM 1135
patients with NMOSD and suggest an alternative etiology.9 Ancillary tests should
be evaluated with due diligence because virtually every radiologic feature of
NMOSD can be caused by something else. Specifically, clinical and radiologic
features of myelopathy may be caused by sarcoidosis and other systemic
inflammatory disorders, MOG-associated disorders, GFAP astrocytopathy,
spondylotic myelopathy, ischemia, metabolic disorders (deficiency in vitamin
B12, copper, folate, or vitamin E), neoplasms, paraneoplastic myelopathy, spinal
dural arteriovenous fistulas, and Leber hereditary optic neuropathy plus
syndromes.32-34 MRI findings indicating possible chiasmatic/optic neuritis have
Optic neuritis Vision loss often characterized by severe, painless decline in visual acuity to Snellen equivalent of
20/200 or worse, with altitudinal visual field defects at presentation9; acutely, the optic nerve
may be swollen or normal in appearance; prognosis for visual recovery is guarded13
Transverse myelitis Sensory and motor dysfunction is often severe, with associated painful tonic spasms at times and
pruritis; neurologic recovery may be limited, and worse outcomes are predicted by more
extensive longitudinally extensive transverse myelitis lesions with subsequent cord atrophy9,32
Area postrema syndrome Area postrema syndrome is characterized by persistent nausea, vomiting, and hiccups9 and may
be initially misdiagnosed as a primary gastrointestinal syndrome, causing treatment delay;
approximately 58% of patients with neuromyelitis optic spectrum disorders (NMOSD) experience
area postrema syndrome during the course of their disease, and this syndrome often heralds
other NMOSD-related relapses over the disease course10
Brainstem syndromes Variable presentations of cranial nerve palsies, including hearing loss, gaze palsies, opsoclonus,
myoclonus, ataxia, and limb weakness may occur9,30
Diencephalic syndromes Patients may manifest narcolepsy, hypothermia, syndrome of inappropriate secretion of
antidiuretic hormone (SIADH), anorexia, daytime somnolence, and obesity9,30
Acute myopathy with Recurrent hyperCKemia with muscle pathology compatible with complement-activating IgG
hyperCKemia targeting sarcolemmal aquaporin-4 (AQP4) confirms organ involvement beyond the central
nervous system (CNS) as a component of NMOSD30,31
Neuropsychiatric Psychiatric manifestations may be more common in NMOSD than multiple sclerosis (MS), with
recurrent major depressive disorder being the most commonly reported psychiatric diagnosis;
patients with NMOSD may also have mania, dysthymic disorder, anxiety, psychosis, and eating
disorders; suicide risk is reported to be higher in NMOSD than MS25-27
Uveitis Vision loss, blurred vision, floaters, and photosensitivity due to inflammation of the uveal tract
may occur in 1% of NMOSD cases, with bilateral anterior uveitis being the most common subtype;
uveitis attacks may precede relapses among 67% of patients29
Diagnostic criteria for neuromyelitis optica spectrum disorder (NMOSD) with positive
aquaporin-4 (AQP4)-IgG
◆ At least one core clinical characteristic
◆ Positive test for AQP4-IgG using best available detection method (cell-based assay
strongly recommended)
◆ Exclusion of alternative diagnoses
Diagnostic criteria for NMOSD without AQP4-IgG or NMOSD with unknown AQP4-IgG status
◆ At least two core clinical characteristics occurring as a result of one or more clinical
attacks and meeting all of the following requirements:
◇ At least one core clinical characteristic must be optic neuritis, acute transverse myelitis
with longitudinally extensive transverse myelitis (LETM), or area postrema syndrome
◇ Dissemination in space (two or more different core clinical characteristics)
◇ Fulfillment of MRI requirements below as applicable
◆ Negative tests for AQP4-IgG using best available detection method or testing unavailable
◆ Exclusion of alternative diagnoses
Core clinical characteristics
◆ Optic neuritis
◆ Acute transverse myelitis
◆ Area postrema syndrome: episode of otherwise unexplained hiccups or nausea and vomiting
◆ Acute brainstem syndrome
◆ Symptomatic narcolepsy or acute diencephalic clinical syndrome with NMOSD-typical
diencephalic MRI lesions
◆ Symptomatic cerebral syndrome with NMOSD-typical brain lesions
Additional MRI requirements for seronegative NMOSD
◆ Acute optic neuritis: requires brain MRI showing (1) normal findings or only nonspecific
white matter lesions, OR (2) optic nerve MRI with a T2-hyperintense lesion or T1-weighted
gadolinium-enhancing lesion extending over more than half the optic nerve length or
involving optic chiasm
◆ Acute transverse myelitis: requires associated intramedullary MRI lesion extending over
three contiguous segments (LETM) OR three contiguous segments of focal spinal cord
atrophy in a patient who has a history that is compatible with acute transverse myelitis
◆ Area postrema syndrome: requires associated dorsal medulla/area postrema lesions
◆ Acute brainstem syndrome: requires associated periependymal brainstem lesions
CONTINUUMJOURNAL.COM 1137
CONTINUUMJOURNAL.COM 1139
CASE 7-1 A 34-year-old man who was previously well presented with a 3-day
history of pain and vision loss in his right eye. His examination showed a
best-corrected Snellen visual acuity of 20/200 in the right eye and 20/20
in the left eye. His pupils were 6 mm in darkness constricting to 3 mm in
bright light with a right relative afferent pupillary defect. The fundus
examination of the right eye showed optic disc edema with hemorrhage
superior to the optic disc (FIGURE 7-3A). Fundus findings were normal in the
left eye (FIGURE 7-3B). Ocular motility was normal, and the remaining
findings on the neurologic examination were unremarkable.
MRI of the brain was normal. Yet, fat-suppressed T1-weighted,
gadolinium-enhanced views of the orbits (FIGURE 7-3C) showed
enhancement of the right optic nerve at the level of the optic canal. The
patient underwent serologic testing for aquaporin-4 (AQP4)-IgG
antibodies, which proved negative. Serum testing for myelin
oligodendrocyte glycoprotein (MOG)-IgG antibodies (cell-based assay)
was positive. The patient was treated with high-dose IV
methylprednisolone for 5 days and had good visual recovery within
2 weeks of symptom onset.
COMMENT Several features of this case are typical for MOG-associated disorders. The
patient is a young man; although neuromyelitis optica spectrum disorder
(NMOSD) is more likely to affect middle-aged women, MOG-associated
disorders may be just as likely to affect men as women. The manifestations
of prominent optic disc edema with hemorrhage are more typical findings
of MOG-associated optic neuritis than NMOSD optic neuritis or optic
neuritis associated with multiple sclerosis. The patient has prominent optic
nerve enhancement extending through the optic canal. Accordingly,
patients with MOG-associated optic neuritis may be more likely to
demonstrate longitudinal lesions affecting the intraorbital optic nerve
segments, with prominent perineural enhancement as compared with
patients who have optic neuritis with MS or NMOSD. Finally, the patient
experienced very good visual recovery with high-dose IV
methylprednisolone, which is more likely to occur in cases of
MOG-associated optic neuritis than NMOSD optic neuritis.
FIGURE 7-3
Imaging findings from the patient in CASE 7-1. A, The
fundus examination of the right eye shows optic
disc edema with hemorrhage superior to the optic
disc. B, Fundus findings are normal in the left eye.
C, A Fat-suppressed axial postcontrast
T1-weighted view of the orbits shows
enhancement of the right optic nerve at the level
of the optic canal and orbital apex (arrow).
CONTINUUMJOURNAL.COM 1141
CONTINUUMJOURNAL.COM 1143
FIGURE 7-4
Imaging findings from the patient in CASE 7-2. The fundus examination shows a normal-
appearing optic nerve in the right eye (A) and temporal optic disc pallor in the left eye (B).
MRI of the orbits shows a longitudinal fat-suppressed fluid-attenuated inversion recovery
(FLAIR) hyperintensity within the left optic nerve (white arrow), affecting the intraorbital
segment of the nerve and extending along its length to the optic chiasm (C). Axial FLAIR
imaging of the brain reveals several subcortical white matter signal changes (D, open arrow).
This case illustrates several important points that ultimately helped render COMMENT
the diagnosis of NMOSD but not before the patient was left with
permanent vision loss. The older age of the patient and absence of any
visual recovery after 8 weeks were atypical features for sporadic optic
neuritis or optic neuritis heralding a diagnosis of MS. In “typical” cases of
optic neuritis, patients regain a line or two of Snellen visual acuity function
within 3 weeks of symptom onset. The orbital images in this case
demonstrated a longitudinal lesion affecting the left intraorbital optic
nerve, extending to involve the chiasm, which can be seen in NMOSD.
Importantly, the patient experienced symptoms consistent with area
postrema syndrome 3 months before her left optic neuritis event, which
were mistakenly attributed to a gastrointestinal illness. Regrettably, a
cranial MRI was not performed at that time, which may have shown
classical features for area postrema syndrome in the dorsal medulla.
Furthermore, had high-dose corticosteroid therapy and
immunosuppressive treatment been administered, the patient may or may
not have experienced an optic nerve relapse shortly thereafter. Given the
patient’s core clinical characteristics (optic neuritis and area postrema
syndrome), positive serum APQ4-IgG result, and lack of another diagnosis
that could otherwise explain her clinical presentation, she fulfilled
diagnostic criteria for NMOSD as outlined in TABLE 7-2.
CONTINUUMJOURNAL.COM 1145
Serum Testing
In addition to the known specificity and sensitivity of the AQP4-IgG antibody as
a diagnostic biomarker, serum neurofilament light chain has also gained recent
interest in the evaluation of patients with NMOSD. Neurofilament light chain is a
scaffolding protein in the neuronal cytoskeleton that is released in the
extracellular space following axonal damage, eventually making its way into CSF
and serum.72-74 Neurofilament light chain levels are increased in the CSF of
patients with neurodegenerative and traumatic disorders, including MS and
NMOSD. Initially, neurofilament light chain was measured in the CSF, which
posed obvious challenges in the clinical arena because testing was likely
considered too invasive to be practical. The debut of single-molecule array
testing enabled highly accurate and reproducible serum neurofilament light
chain measurements, which consistently parallel neurofilament light chain levels
in CSF.72-74 Studies have shown that higher serum neurofilament light chain
levels are observed in patients with MS compared with healthy controls.
Moreover, serum neurofilament light chain levels correlate with an increased
burden of T2-weighted and gadolinium-enhancing MRI lesions and worsening
CSF
CSF analysis is supportive for the diagnosis of NMOSD if oligoclonal bands
are absent, in the appropriate clinical context.9 Yet, oligoclonal bands may be
found in approximately 20% to 30% of patients with NMOSD, most often
during an attack. In contrast, oligoclonal bands are present in more than 85% of
patients with MS. Analysis of CSF from patients with NMOSD often shows
normal to highly inflammatory profiles, and this is typically more prominent for
episodes of transverse myelitis than optic neuritis. Pleocytosis of the CSF,
frequently with neutrophilic or eosinophilic predominance, is useful for
distinguishing NMOSD from MS.6,9 In addition to oligoclonal bands, CSF GFAP
levels have gained interest as a biomarker in NMOSD. GFAP is a principal
intermediate filament that forms the astrocyte cytoskeleton. Accordingly,
patients with NMOSD have elevated GFAP levels in the CSF and serum, owing to
astrocyte injury. Higher serum and CSF GFAP levels have been reported during
NMOSD attacks and predict the presence of relapses.10 Akin to neurofilament
light chain, CSF and serum GFAP levels show strong correlations, meaning
serum GFAP has the potential role as a biomarker for disease severity and
predictor of future disease activity in patients with AQP4-IgG–positive NMOSD,
even in phases of clinical remission. Finally, a higher serum GFAP/serum
neurofilament light chain quotient at relapse may help differentiate NMOSD
from MS,75 but this has been more practically calculated based on serum levels.
CONTINUUMJOURNAL.COM 1147
Serology
Other autoantibodies Positive systemic lupus Support diagnosis of Higher autoantibody levels are
erythematosus, Sjögren syndrome, NMOSD, particularly in seen with cases of clinically
myasthenia gravis antibodies AQP4-IgG– active connective tissue
seronegative cases diseases that co-associate with
NMOSD
Serum neurofilament Serum neurofilament light chain May have a role in Single-molecule array allows for
light chain correlates with clinical and MRI facilitating diagnosis accurate, reproducible levels
measures of disease activity, and detecting relapses that closely correlate with CSF
reflects drug effects, and captures neurofilament light chain;
NMOSD relapse effects notably, serum neurofilament
light chain will increase with
other pathologic states of the
central nervous system
Serum glial fibrillary GFAP is a principal intermediate Relapse detection Serum GFAP levels correlate
acidic protein (GFAP) filament that forms the astrocyte well with CSF GFAP levels
cytoskeleton; patients with NMOSD
have elevated GFAP levels in the CSF
and serum owing to astrocyte injury;
higher CSF GFAP levels have been
reported during NMOSD attacks
CSF
Oligoclonal bands Oligoclonal bands are commonly May have a role in Oligoclonal bands may be found
found in patients with multiple facilitating diagnosis in a minority of patients with
sclerosis (MS) and rare in NMOSD NMOSD (15%) and do not
exclude the diagnosis
GFAP GFAP is a principal intermediate Relapse detection: Serum GFAP levels correlate
filament that forms the astrocyte higher CSF GFAP levels well with CSF GFAP levels
cytoskeleton; patients with NMOSD have been reported
have elevated GFAP levels in the CSF during NMOSD attacks
and serum owing to astrocyte injury and predict the
presence of relapses
Peripapillary retinal Peripapillary retinal nerve fiber layer Relapse (optic Peripapillary retinal nerve fiber
nerve fiber layer changes represent axonal injury in the neuritis) detection and layer changes are not specific to
thickness afferent visual pathway; owing to the follow-up NMOSD and should be
severity of optic neuritis in patients interpreted in the context of a
with NMOSD, more peripapillary thorough ophthalmologic
retinal nerve fiber layer thinning examination
occurs over weeks to months after
NMOSD optic neuritis relative to MS
optic neuritis
Macular ganglion cell Macular ganglion cell inner plexiform Optic neuritis relapse Macular ganglion cell inner
layer/ganglion cell inner layer thinning represents retinal detection and follow-up plexiform layer changes are not
plexiform layer thickness ganglion cell (neuronal) injury in specific to NMOSD and should
NMOSD be interpreted in the context of
a thorough ophthalmologic
examination
Microcystic macular Microcystic macular edema may be Relapse (optic neuritis) Microcystic macular edema is a
edema a marker of more severe optic detection and follow-up nonspecific finding and may be
neuritis and is more often seen in observed across a spectrum of
NMOSD optic neuritis than MS optic optic neuropathies
neuritis
MRI
Optic nerve lesions Longitudinal intracranial optic nerve Diagnosis and follow-up Longitudinal optic nerve and
lesions extending to the chiasm are chiasmal lesions can be found in
characteristic of NMOSD other disorders (eg, neoplasms,
sarcoidosis, lymphoma)
Optic nerve lesion length Optic nerve lesion length, Prognostic Worse optical coherence
particularly affecting the intraorbital tomography–measured retinal
and canalicular segments, correlates ganglion layer and ganglion layer
with long-term visual acuity in thinning can be paired with MRI
NMOSD measures to predict
vision-related quality of life
outcomes
Longitudinally extensive Spinal cord relapses in NMOSD Diagnosis, follow-up, Spinal lesions may evolve over
transverse myelitis usually cause an LETM, with the and predicting future time, so longitudinal follow-up
(LETM) spinal cord lesion spanning three or relapses may be needed
more consecutive vertebral
segments; up to 20% of patients may
present with a short-segment
transverse myelitis
Dorsal medulla lesions Lesions in the area postrema are May have a role in Area postrema lesions may be
common in NMOSD and may herald facilitating diagnosis in transient; therefore, it is
disease onset; patients present with the right clinical important to image soon after
hiccups, nausea, and emesis context symptom onset because
gadolinium-enhanced
sequences may improve
detection
Periependymal lesions Periependymal lesions are located May have a role in Periaqueductal lesions are seen
around the cerebral aqueduct and facilitating diagnosis in in NMOSD and MOG-associated
the third and fourth ventricles in the right clinical setting disorders but are rare in MS
circumventricular regions that are
highly vascularized and lack a
blood-brain barrier; these include
lesions in the diencephalon
(hypothalamus and thalamus) and
brainstem, which represent areas of
high AQP4-IgG expression
CONTINUUMJOURNAL.COM 1149
Bridge-arch lesions NMOSD corpus callosal lesions are May have a role in Callosal lesions in MS
often extensive and oriented along facilitating diagnosis in are characteristically
the long axis in a “bridge-arch” the right clinical setting well-circumscribed and ovoid
pattern; these lesions are shaped, oriented perpendicular
sometimes acutely edematous with to the lateral ventricles (Dawson
a marbled pattern fingers)
Brain lesion The enhancement pattern of brain May have a role in MS lesions are often
enhancement patterns lesions in NMOSD often shows facilitating diagnosis, open-ring or nodular with
pencil-thin linear periependymal or detecting disease well-demarcated enhancement
cloudlike configuration with poorly activity, and
marginated enhancement; prognostication
leptomeningeal enhancement may
also occur; enhancing lesions may
predict future relapses and later
disability
Cervical cord Mean upper cervical cross-sectional Detecting disease The association between
(cross-sectional area) area as a measure of spinal cord activity and cervical cord cross-sectional
atrophy is associated with an prognostication area and disability has been
increased number of myelitis found even in patients with
relapses and has been predictive of NMOSD with and without a
subsequent clinical disability clinical history of myelitis
(evidence is conflicting)
Bright spotty lesions Bright spotty lesions appear as May have a role in Bright spotty lesions may
discrete hyperintense lesions on facilitating diagnosis in facilitate the discrimination of
axial T2-weighted imaging that are the right clinical setting NMOSD from other LETM
more hyperintense or of equivalent causes; rarely bright spotty
intensity to the surrounding CSF lesions can occur with other
causes of transverse myelitis
Spinal cord lesion length Longer spinal cord lesion length at Prognostic Studies comparing patients with
time of attack is associated with NMOSD with long and short
increased disability, both at attack spinal cord lesions have
nadir and after recovery demonstrated that shorter
lesions are associated with
better prognosis for recovery
MRI
MRI represents an important diagnostic biomarker, particularly for
seronegative individuals suspected of having NMOSD (TABLE 7-2). In most
cases of NMOSD, the cranial MRI scan is viewed as being unremarkable
relative to imaging in patients with MS. Yet, lesions are detected 50% to 85% of
the time in patients fulfilling the revised 2006 NMO diagnostic criteria.32
Nonspecific punctate and hyperintense subcortical changes (best seen with
fluid-attenuated inversion recovery [FLAIR] and T2-weighted sequences) have
been reported in 43% to 70% of patients with NMOSD at initial presentation.32
For patients presenting with specific clinical syndromes such as optic neuritis,
transverse myelitis, and area postrema syndrome, characteristic radiologic
lesions can help render the diagnosis (TABLE 7-2).9 In 2019, Cacciaguerra and
colleagues79 proposed criteria to distinguish MS from NMOSD. These criteria
included (1) the absence of combined juxtacortical/cortical lesions, (2) the
absence of ovoid periventricular lesions, (3) the absence of so-called Dawson
fingers, (4) the presence of an LETM (FIGURE 7-2), and (5) the observation of
periependymal lesions along the lateral ventricles. It was noted that fulfillment of
at least two of the five aforementioned criteria distinguished NMOSD from MS
with 92% sensitivity and 91% specificity in training samples (82% sensitivity and
91% specificity in validation samples).32,79 Unfortunately, the Cacciaguerra
criteria are less reliable in differentiating NMOSD and MOG-associated disorder
cases from one another.79
As stated, additional MRI findings may be relatively specific for NMOSD
diagnosis in the right clinical context. For example, a 2021 study found bilateral
involvement in almost 50% of NMOSD cases with optic nerve lesions.55 These
lesions tend to extend over 50% of the posterior aspects of the optic nerve as it
extends toward the chiasm.55 In cases of area postrema syndrome, a characteristic
high-signal lesion in the posterior medulla at the floor of the fourth ventricle
(best seen with T2-weighted and FLAIR MRI sequences) can help render the
diagnosis (FIGURE 7-5).55,80 Other brain MRI abnormalities observed in NMOSD
include large (greater than 3 cm) and confluent cerebral hemispheric lesions
located in the subcortical white matter.55 These lesions may resemble lesions of
ADEM or have a tumefactive appearance (FIGURE 7-5).55,80 MRI frequently
reveals corpus callosal lesions in both NMOSD and MS, but these are more likely
to be oriented in a “bridge arch” and show a marbled pattern in NMOSD
(FIGURE 7-5).32,55,80 Rounded lesions with a ground glass–like heterogeneous
appearance within the body of the corpus callosum are also considered specific
for NMOSD.55 In contrast, corpus callosal lesions in MS tend to be well
circumscribed, ovoid shaped, and oriented perpendicular to the lateral
ventricles.32 The central vein sign often seen in MS (in 80% of patients with MS)
is found in only one-third of patients with NMOSD.32,81 Diffuse leptomeningeal
CONTINUUMJOURNAL.COM 1151
FIGURE 7-5
MRI lesions characteristic of
neuromyelitis optica spectrum
disorder. Axial brain MRI images
showing diencephalic lesions
surrounding (A) the third ventricles and
cerebral aqueduct, which include the
thalamus, hypothalamus (B), and
anterior border of the midbrain (C).
Axial brain MRI images reveal a dorsal
brainstem lesion adjacent to the fourth
ventricle (D), whereas a sagittal cervical
cord MRI image depicts a linear
medullary lesion that is contiguous with
cervical cord lesion (E); an edematous
and extensive dorsal brainstem lesion
involving the cerebellar peduncle is
shown in an axial brain MRI (F). The
sagittal (G, left) and coronal (G, right)
brain MRI images capture a callosal
lesion immediately next to the lateral
ventricle, following the ependymal
lining, a “marbled-pattern” callosal
lesion is shown on an axial MRI image
(H) whereas an axial image (I) shows the
classic “bridge-arch pattern” callosal
lesion. The series of axial brain MRI
images show tumefactive hemispheric
white matter lesions (J), a long
spindlelike or radial-shaped lesion
following white matter tracts (K), and
extensive and confluent hemispheric
lesions with increased diffusivity on
apparent diffusion coefficient maps
suggesting vasogenic edema (L), and
hemispheric lesions in the chronic phase with cystlike cavitary changes (M). Corticospinal
tract lesions are seen involving the posterior limb of the internal capsule (N) and on axial
lesions of the cerebral peduncle of the midbrain (O), whereas coronal images show a
longitudinally extensive lesion following the pyramidal tract ( P). Axial MRI images
demonstrate cloudlike enhancement (Q), and linear enhancement of the ependymal surface
of the lateral ventricles (R). The axial MRI image (S, left) and sagittal image (S, right) show
meningeal enhancement.
Reprinted with permission from Kim HJ, et al, Neurology.80 © 2015 American Academy of Neurology.
CONTINUUMJOURNAL.COM 1153
FIGURE 7-6
Imaging characteristics of spinal cord lesions in aquaporin-4 (AQP4)-positive neuromyelitis
optica spectrum disorders (NMOSD) and myelin oligodendrocyte glycoprotein (MOG)-
positive myelitis. AQP4-immunoglobulin G (IgG)–positive disease, sagittal T2 sequences:
longitudinally extensive lesions, extension of cervical cord lesions rostrally into the dorsal
medulla, and subsequent cord atrophy and discontinuity of lesions in chronic longitudinally
extensive transverse myelitis (LETM). MOG-IgG–positive disease, sagittal and axial T2
sequences: short-segment spinal cord lesions are demonstrated, although LETM may also
occur. MOG-IgG–positive disease, sagittal T2 and sagittal and axial postcontrast T1-weighted
sequences: although bright spotty lesions are more frequently associated with AQP4-IgG–
positive NMOSD, they may rarely occur with other causes of transverse myelitis, as seen here.
The short length and peripheral location of this lesion, in addition to serologic testing, helped
confirm the diagnosis of MOG-IgG–positive disease.
Gd = gadolinium.
Reprinted from Solomon JM, et al, Ther Adv Neurol Disord.32 © 2021 The Authors.
CONTINUUMJOURNAL.COM 1155
TABLE 7-4 Therapeutic Agents Used in the Treatment of Patients With Neuromyeltis
Optica Spectrum Disorders
Dosing options
(regimens may
Drug Mechanism vary) Adverse effects Indication
Off-label immunosuppressive (induction and maintenance) therapies
Azathioprine A purine analogue that Suggested Neutropenia, leukopenia, Has been shown to be
disrupts synthesis of dosing: 2 mg/kg/ pancytopenia, and severe inferior to rituximab and
DNA and RNA and d to 3 mg/kg/d myelosuppression; tocilizumab on relapses
induces T-cell apoptosis combined with opportunistic viral, and disability
oral prednisone bacterial, and fungal progression in
initially (30 mg/d infections neuromyelitis optica
which is tapered spectrum disorders
after 6-9 months) (NMOSD)
Dosing options
(regimens may
Drug Mechanism vary) Adverse effects Indication
Mitoxantrone A type II topoisomerase Suggested Because of the side Has been associated
inhibitor that disrupts dosing: 12 mg/ effects (cardiotoxicity, with improved relapse
cellular DNA synthesis m2/mo for therapy-related acute rates and disability
and repair by 6 months, leukemia) and the limited
intercalation between followed by duration of the therapy,
DNA bases; inhibits monthly mitoxantrone use is
proliferation of maintenance generally not
macrophages, B and T dose of 6 mg/m2; recommended; the
cells; induces apoptosis total cumulative maximum cumulative dose
of B cells, monocytes, dose <120 mg/m2 should not exceed
and dendritic cells; and 100 mg/m2 body
decreases secretion of surface area
proinflammatory
cytokines
Cyclophosphamide An alkylating agent that The treatment Bladder cancer, gonadal Has been associated
forms DNA crosslinks, may be applied in toxicity, hemorrhagic with improved relapse
which is irreversible, and immune-ablative cystitis; alopecia, nausea/ rates and disability, but
causes cell apoptosis, doses (2000 mg/ vomiting, transient results are mixed82; it is
eliminates T regulatory d for 4 days) or at myelosuppression, and only recommended
cells, and induces T-cell a dose of 500 mg/ transient azoospermia when other
growth factors including m2 to 600 mg/m2 immunosuppressive
type I interferons body surface therapies fail or are not
area per available
administration;
the dose should
be adjusted
according to
changes in the
total leukocyte
count
CONTINUUMJOURNAL.COM 1157
CONTINUED
CONTINUED FROM FROM PAGE 1157
PAGE 1157
Dosing options
(regimens may
Drug Mechanism vary) Adverse effects Indication
IV immunoglobulin Consists of polyclonal Suggested Fever and headache; Data are lacking; IVIg
(IVIg) IgG pooled from donors; dosing regimens aseptic meningitis, therapy is suggested as
the mechanism by which vary but include thrombosis, and an alternative for
it exerts its anti- IVIg treatment anaphylaxis patients with
inflammatory effect in 0.7 g/kg body contraindication to one
specific diseases is weight/d for of the other treatments
unknown but may stem 3 days, repeated or for children
from both functional at various
domains of the IgG intervals (monthly
molecule, the or otherwise)
antigen-binding
fragment (F[ab]2), and
the fragment
crystallizable region
Eculizumab Monoclonal antibody 900 mg/wk IV for Headaches, leukopenia, The PREVENT trial84
that binds to the 4 weeks, then and lymphopenia; showed that eculizumab
complement component 1200 mg IV on complement inhibition reduced the risk of
C5, inhibiting its cleavage week 5 and every increases the risk of relapse by 94%
and preventing the 2 weeks infection with compared with placebo,
generation of the thereafter encapsulated bacteria, and the risk reduction
terminal complement especially Neisseria persisted at 48 and
complex; inhibits AQP4 meningitidis 144 weeks
antibody formation of (meningococcal
the membrane attack vaccination is mandatory
complex before starting
treatment); upper/lower
respiratory tract
infections,
nasopharyngitis, influenza,
pharyngitis, and bronchitis
Dosing options
(regimens may
Drug Mechanism vary) Adverse effects Indication
Satralizumab A monoclonal antibody Subcutaneous Leukopenia, In the SAkuraSky study
against the IL-6 satralizumab nasopharyngitis and (satralizumab plus
receptor; blocks the 120 mg at weeks 0 upper/lower respiratory background
IL-6R and B-cell antibody and 2 and every tract infections immunotherapy) 8 (20%) of
production 4 weeks 41 participants in the
thereafter, has treatment arm relapsed
been tested as over the course of the trial
monotherapy and compared with 18 (43%) of
as add-on 42 controls, resulting in a
therapy in two 62% reduction in
double- adjudicated relapse risk
blind placebo- over time; post hoc
controlled phase analysis by AQP4-Ig
3 trials serostatus revealed a
(SAkuraStar and greater effect in the
SAkuraSky)86,87 seropositive subgroup,
with a 79% reduction in
relapse risk compared
with seropositive
participants in the control
group; serum AQP4-IgG–
negative participants (n =
28; 14 in each arm) showed
no reduction in risk of
relapse with satralizumab
compared with placebo11
CONTINUUMJOURNAL.COM 1159
Dosing options
(regimens may
Drug Mechanism vary) Adverse effects Indication
Dosing options
(regimens may
Drug Mechanism vary) Adverse effects Indication
Ublituximab A third-generation Suggested Leukopenia with other Compared with
chimeric IgG1 dosing: single serious side effects TBD rituximab, ublituximab
monoclonal antibody dose of 450 mg IV permits less dosing and
with high affinity to the may have fewer adverse
Fcy receptor, an epitope events; a phase 1 clinical
on CD20+ B cells, which trial of ublituximab as an
is not targeted by add-on to steroids for
rituximab; causes the treatment of acute
depletion of CD20+ B NMOSD has shown that
cells 4 of 5 subjects achieved
an acute B-cell depletion
with at least 2 months of
durability89,100,101;
although not designed
for efficacy, this trial
demonstrated that
neurologic disability
improved in the three
subjects who achieved
immunologic remission
CONTINUUMJOURNAL.COM 1161
CONTINUUMJOURNAL.COM 1163
SPECIAL CIRCUMSTANCES
NMOSD co-associates with numerous other pathologic states including
infections, systemic inflammatory conditions, cancers, and immune-mediated
connective tissue diseases.10 With respect to the association between connective
tissue diseases and NMOSD, the aforementioned international panel concluded
that the presence of SLE and Sjögren syndrome in NMOSD should be viewed as a
coexistence, rather than a complication.9,15 Indeed, the presence of SLE and
Sjögren syndrome in the context of an NMOSD core clinical syndrome such as
optic neuritis or transverse myelitis lends support for the diagnosis of NMOSD.9,15
This co-association has clinical implications that require consideration. Positive
AQP4-IgG antibodies can occur in asymptomatic patients with connective tissue
diseases; moreover, positive antinuclear antibodies, anti–double-stranded DNA
antibodies, anti–Sjögren syndrome A (SSA)/Ro antibodies, and anti-SSB/La
antibodies are also found in patients with NMOSD without clinically apparent
systemic autoimmune diseases.15 For this reason, patients with NMOSD should
be asked about systemic symptoms and undergo a rheumatologic workup if an
underlying connective tissue disease is suspected. Similarly, neurologic
manifestations such as area postrema syndrome, optic neuritis, or transverse
myelitis in a patient with a known connective tissue disease should prompt
evaluation for NMOSD, including an MRI of the brain with and without contrast,
as well as serum antibody testing for AQP4-IgG and MOG-IgG.15 Acutely, these
patients may be treated with high-dose corticosteroids; yet, deciding on the
appropriate long-term immunosuppressive therapy may be challenging and
require multidisciplinary collaboration. Rituximab, and to a lesser extent
mycophenolate mofetil, have been used to treat patients with both connective
tissue diseases and NMOSD.15 Importantly, therapies that interfere with tumor
necrosis factor-alpha such as infliximab, adalimumab, or etanercept would best
be avoided because these agents have been associated with causing and
exacerbating CNS inflammatory disorders.15
CONCLUSION
Over the past decade, scientific understanding regarding the clinical
manifestations, relapse-related risk factors, and therapeutic approach to NMOSD
has rapidly evolved. In many respects, the path of scientific discovery has been
an affirmation of a “bench-to-beside” model that started with the identification
of key immunologic therapeutic targets and culminated in the approval of three
new therapies for NMOSD in 2019. Currently, NMOSD poses not only a
diagnostic conundrum but also treatment challenges. The role of MOG-IgG
antibodies in the diagnostic criteria for NMOSD needs to be fully elucidated.
Better biomarkers are needed to predict relapses, and better treatments are
needed to prevent them. Areas of clinical priority in the management of patients
with NMOSD include the need for regenerative therapies to ameliorate the
effects of attack-dependent disability. In the modern treatment era, scientific
exploration, clinical rigor, and patient-oriented advocacy will be needed to
ensure safe and effective therapies are accessible to all patients with NMOSD.
CONTINUUMJOURNAL.COM 1165
REFERENCES
1 Devic E. Myelite aigue compliquee de nevrite 14 Misu T, Hoftberger R, Fujihara K, et al. Presence
optique. Bull Med (Paris) 1894;8:1033-1034. of six different lesion types suggests diverse
mechanisms of tissue injury in neuromyelitis
2 Fujihara K. Neuromyelitis optica spectrum
optica. Acta Neuropathol 2013;125(6):815-827.
disorders: still evolving and broadening. Curr
doi:10.1007/s00401-013-1116-7
Opin Neurol 2019;32(3):385-394. doi:10.1097/
WCO.0000000000000694 15 Cruz RA, Chaudhary S, Guevara M, Meltzer E.
Neuromyelitis optica spectrum disorders
3 Lennon VA, Wingerchuk DM, Kryzer TJ, et al. A
(NMOSD) and connective tissue disease (CTD):
serum autoantibody marker of neuromyelitis
an update for the rheumatologist. Curr
optica: distinction from multiple sclerosis.
Rheumatol Rep 2021;23(6):33. doi:10.1007/
Lancet 2004;364:(9451):2106-2112. doi:10.1016/
s11926-021-01000-2
S0140-6736(04)17551-X
16 Mealy MA, Wingerchuk DM, Greenberg BM, Levy
4 Lucchinetti CF, Guo Y, Popescu B, et al. The
M. Epidemiology of neuromyelitis optica in the
pathology of an autoimmune astrocytopathy:
United States: a multicenter analysis. Arch Neurol
lessons learned from neuromyelitis optica. Brain
2012;69(9):1176-1180. doi:10.1001/
Pathol 2014;24(1):83-97. doi:10.1111/bpa.12099
archneurol.2012.314
5 Winkler A, Wrzos C, Haberl M, et al. Blood-brain
17 Weinshenker BG, Wingerchuk DM. Neuromyelitis
barrier resealing in neuromyelitis optica occurs
spectrum disorders. Mayo Clin Proc 2017;92(4):
independently of astrocyte regeneration. J Clin
663-679. doi:10.1016/j.mayocp.2016.12.014
Invest 2021;131(5):e141694. doi:10.1172/JCI141694
18 Jiao Y, Fryer JP, Lennon VA, et al. Updated
6 Fujihara K, Misu T, Nakashima I, et al.
estimate of AQP4-IgG serostatus and disability
Neuromyelitis optica should be classified as an
outcome in neuromyelitis optica. Neurology 2013;
astrocytopathic disease rather than a
81(14):1197-1204. doi:10.1212/WNL.
demyelinating disease. Clin Exp Neuroimmunol
0b013e3182a6cb5c
2012;3:58-73. doi: 10.1111/j.1759-1961.2012.00030.x
19 Jarius S, Ruprecht K, Wildemann B, et al.
7 Hor JY, Asgari N, Nakashima I, et al. Epidemiology
Contrasting disease patterns in seropositive and
of neuromyelitis optica spectrum disorder and its
seronegative neuromyelitis optica: a multicentre
prevalence and incidence worldwide. Front
study of 175 patients. J Neuroinflammation 2012;
Neurol 2020;11:501. doi:10.3389/fneur.2020.00501
9:14. doi:10.1186/1742-2094-9-14
8 Papp V, Magyari M, Aktas O, et al. Worldwide
20 Kitley J, Leite MI, Nakashima I, et al. Prognostic
incidence and prevalence of neuromyelitis
factors and disease course in aquaporin-4
optica. Neurology 2021;96(2):59-77. doi:10.1212/
antibody-positive patients with neuromyelitis
WNL.0000000000011153
optica spectrum disorder from the United
9 Wingerchuk DM, Banwell B, Bennett JL, et al. Kingdom and Japan. Brain 2012;135(Pt 6):
International consensus diagnostic criteria for 1834-1849. doi:10.1093/brain/aws109
neuromyelitis optica spectrum disorders.
21 Costello F, Scott J. Imaging in neuro-
Neurology 2015;85(2):177-189. doi:10.1212/
ophthalmology. Continuum (Minneap Minn)
WNL.0000000000001729
2019;25(5, Neuro-ophthalmology):1438-1490.
10 Ma X, Kermode AG, Hu X, Qui W. Risk of relapse in doi:10.1212/CON.0000000000000783
patients with neuromyelitis optica spectrum
22 Mealy MA, Mossburg SE, Kim SH, et al. Long-term
disorder: recognition and preventive strategy.
disability in neuromyelitis optica spectrum
Mult Scler Relat Disord 2020;46:102522.
disorder with a history of myelitis is associated
doi:10.1016/j.msard.2020.102522
with age at onset, delay in diagnosis/preventive
11 Levy M, Fujihara K, Palace J, et al. New therapies treatment, MRI lesion length and presence of
for neuromyelitis optica spectrum disorder. symptomatic brain lesions. Mult Scler Relat
Lancet Neurol 2021;20(1):60-67. doi:10.1016/ Disord 2019;28:64-68. doi:10.1016/j.msard.
S1474-4422(20)30392-6 2018.12.011
12 Brod SA. Review of approved NMO therapies 23 Hinson SR, Lennon VA, Pittock SJ. Autoimmune
based on mechanism of action, efficacy and AQP4 channelopathies and neuromyelitis optica
long-term effects. Mult Scler Relat Disord 2020; spectrum disorders. Handb Clin Neurol 2016;133:
46:102538. doi:10.1016/j.msard.2020.102538 377-403. doi:10.1016/B978-0-444-63432-0.
00021-9
13 DeLott L, Bennett JL, Costello F, et al. The
changing landscape of optic neuritis: a narrative 24 Wingerchuk DM, Lennon VA, Lucchinetti CF, et al.
review. J Neurol 2022;269(1):111-124. doi:10.1007/ The spectrum of neuromyelitis optica. Lancet
s00415-020-10352-1 Neurol 2007;6(9):805-815. doi:10.1016/S1474-
4422(07)70216-8
CONTINUUMJOURNAL.COM 1167
50 Kessler RA, Mealy MA, Levy M, et al. Early 62 Akaishi T, Takeshita T, Himori T, et al. Rapid
indicators of relapses vs pseudorelapses in administration of high-dose intravenous
neuromyelitis optica spectrum disorder. Neurol methylprednisolone improves visual outcomes
Neuroimmunol Neuroinflamm 2016;3(5):e269. doi: after optic neuritis in patients with AQP4-IgG-
10.1212/NXI.0000000000000269 positive NMOSD. Front Neurol 2020;11:932.
doi:10.3389/fneur.2020.00932
51 Tackley G, O’Brien F, Rocha J, et al. Neuromyelitis
optica relapses: race and rate, 63 Siritho S, Nopsopon T, Pongpirul K, et al.
immunosuppression and impairment. Mult Scler Therapeutic plasma exchange vs conventional
Relat Disord 2016;7:21-25. doi:10.1016/j. treatment with intravenous high dose steroid for
msard.2016.02.014 neuromyelitis optica spectrum disorders
(NMOSD): a systematic review and meta-
52 Orman G, Wang KY, Pekcevik Y, et al. Enhancing
analysis. J Neurol 2021;268(12):4549-4562. doi:10.
brain lesions during acute optic neuritis and/or
1007/s00415-020-10257-z
longitudinally extensive transverse myelitis may
portend a higher relapse rate in neuromyelitis 64 Takai Y, Kuroda H, Misu T, et al. Optimal
optica spectrum disorders. AJNR Am J management of neuromyelitis optica spectrum
Neuroradiol 2017;38(5):949-953. doi:10. disorder with aquaporin-4 antibody by oral
3174/ajnr.A5141 prednisolone maintenance therapy. Mult Scler
Relat Disord 2021;49:102750. doi:10.1016/j.msard.
53 Wang Y, Zhang L, Zhang B, et al. Comparative
2021.102750
clinical characteristics of neuromyelitis optica
spectrum disorders with and without medulla 65 Banerjee A, Ng J, Coleman J, et al. Outcomes
oblongata lesions. J Neurol 2014;261(5):954-962. from acute attacks of neuromyelitis optica
doi:10.1007/s00415-014-7298-7 spectrum disorder correlate with severity of
attack, age and delay to treatment. Mult Scler
54 Mariano R, Messina S, Roca-Fernandez A, et al.
Relat Disord 2019;28:60-63. doi:10.1016/j.
Quantitative spinal cord MRI in MOG-antibody
msard.2018.12.010
disease, neuromyelitis optica and multiple
sclerosis. Brain 2021;144(1):198-212. doi:10.1093/ 66 Kim SH, Kim W, Huh SY, et al. Clinical efficacy of
brain/awaa347 plasmapheresis in patients with neuromyelitis
optica spectrum disorder and effects on
55 Clarke L, Arnett S, Lilley K, et al. Magnetic
circulating anti-aquaporin-4 antibody levels.
resonance imaging in neuromyelitis optica
J Clin Neurol 2013;9(1):36-42. doi:10.3988/jcn.
spectrum disorder. Clin Exp Immunol 2021;206(3):
2013.9.1.36
251-265. doi:10.1111/cei.13630
67 Merle H, Olindo S, Jeannin S, et al. Treatment of
56 Kessler RA, Mealy MA, Levy M, et al. Treatment
optic neuritis by plasma exchange (add-on) in
of neuromyelitis optica spectrum disorder:
neuromyelitis optica. Arch Ophthalmol 2012;
acute, preventive, and symptomatic. Curr Treat
130(7):858-862. doi:10.1001/archophthalmol.2012.
Options Neurol 2016;18(1):2. doi:10.1007/s11940-
1126
015-0387-9
68 Biomarkers Definitions Working Group.
57 Horton L, Bennett JL. Acute management of
Biomarkers and surrogate endpoints: preferred
optic neuritis: an evolving paradigm.
definitions and conceptual framework. Clin
J Neuroophthalmol 2018;38(3):358-367.
Pharmacol Ther 2001;69(3):89-95. doi:10.1067/
doi:10.1097/WNO.0000000000000700
mcp.2001.113989
58 Bonnan M, Valentino R, Debeugny S, et al. Short
69 Aronson JK. Biomarkers and surrogate endpoints.
delay to initiate plasma exchange is the strongest
Br J Clin Pharmacol 2005;59(5):491-494. doi:10.1111/
predictor of outcome in severe attacks of NMO
j.1365-2125.2005.02435.x
spectrum disorders. J Neurol Neurosurg
Psychiatry 2018;89(4):346-351. doi:10.1136/jnnp- 70 Hill AB. The environment and disease:
2017-316286 association or causation? Proc R Soc Med 1965;
58(5):295-300.
59 Levy M. Plasmapheresis for acute attacks in
neuromyelitis optica spectrum disorders. Neurol 71 Costello F, Burton JM, et al. Retinal imaging with
Neuroimmunol Neuroinflamm 2018;5:e510. doi: optical coherence tomography: a biomarker in
10.1212/NXI.0000000000000510 multiple sclerosis? Eye Brain 2018;10:47-63.
doi:10.2147/EB.S139417
60 Petzold A, Braithwaite T, Oosten BW, et al. Case
for a new corticosteroid treatment trial in optic 72 Disanto G, Barro C, Benkert P, et al. Serum
neuritis: review of updated evidence. J Neurol neurofilament light: a biomarker of neuronal
Neurosurg Psychiatry 2020;91(1):9-14. doi:10.1136/ damage in multiple sclerosis. Ann Neurol 2017;
jnnp-2019-321653 81(6):857-870. doi:10.1002/ana.24954
61 Kleiter I, Gahlen A, Borisow N, et al. Apheresis 73 Barro C, Chitnis T, Weiner HL, et al. Blood
therapies for NMOSD attacks. A retrospective neurofilament light: a critical review of its
study of 207 therapeutic interventions. Neurol application to neurologic disease. Ann Clin Transl
Neuroimmunol Neuroinflamm 2018;5(6): Neurol 2020;7(12):2508-2523. doi:10.1002/
e0170847. doi:10.1212/NXI.0000000000000504 acn3.51234
84 Pittock SJ, Berthele A, Fujihara K, et al. 94 Brod SA. Immune reconstitution therapy in
Eculizumab in aquaporin-4-positive NMOSD. Mult Scler Relat Disord 2021;52:102971.
neuromyelitis optica spectrum disorder. N Engl J doi:10.1016/j.msard.2021.102971
Med 2019;381(7):614-625. doi:10.1056/
95 Greco R, Bondanza A, Oliveria MC, et al.
NEJMoa1900866
Autologous hematopoietic stem cell
85 Cree BAC, Bennett JL, Kim HJ, et al. Inebilizumab transplantation in neuromyelitis optica: a registry
for the treatment of neuromyelitis optica study of the EBMT Autoimmune Diseases
spectrum disorder (N-MOmentum): a double- Working Party. Mult Scler 2015;21(2):189-197. doi:
blind, randomised placebo-controlled phase 2/3 10.1177/1352458514541978
trial. Lancet 2019;394(10206):1352-1363. doi:10.
96 Burton JM, Duggan P, Costello F, et al. A pilot trial
1016/S0140-6736(19)31817-3
of autologous hematopoietic stem cell
transplant in neuromyelitis optic spectrum
disorder. Mult Scler Relat Disord 2021;53:102990.
doi:10.1016/j.msard.2021.102990
CONTINUUMJOURNAL.COM 1169
97 Costello F, Burton JM. Contemporary 101 Tugizova M, Vlahovic L, Tomczak A, et al. New
management challenges in seropositive NMOSD therapeutic landscape in neuromyelitis optica.
[published online July 11, 2022]. J Neurol. Curr Treat Options Neurol 2021;23(4):13. doi:
doi: 10.1007/s00415-022-11241-5 10.1007/s11940-021-00667-3
98 Burt RK, Balabanov R, Han X, et al. Autologous 102 Holmøy T, Høglund RA, Illes Z, et al. Recent
nonmyeloablative hematopoietic stem cell progress in maintenance treatment of
transplantation for neuromyelitis optica. neuromyelitis optica spectrum disorder.
Neurology 2019;93(18):e1732-e1741. doi:10.1212/ J Neurol 2020;268(12):4522-4536. doi:10.1007/
WNL.0000000000008394 s00415-020-10235-5
99 Tappenden P, Saccardi R, Confavreux C, et al. 103 Tenembaum S, Yeh EA, et al. The Guthy-Jackson
Autologous haematopoietic stem cell Foundation International Clinical Consortium
transplantation for secondary progressive (GJCF-ICC). Pediatric NMOSD: a review and
multiple sclerosis: an exploratory position statement on approach to work-up and
cost-effectiveness analysis. Bone Marrow diagnosis. Front Pediatr 2020;8:339. doi:10.
Transplant 2010;45(6):1014-1021. doi:10.1038/bmt. 3389/fped.2020.00339
2009.305
104 Mao-Draayer Y, Thiel S, Mills EA, et al.
100 Held F, Klein AK, Berthele A, et al. Drug Neuromyelitis optica spectrum disorders and
treatment of neuromyelitis optica spectrum pregnancy: therapeutic considerations. Nat Rev
disorders: out with the old, in with the new? Neurol 2020;16(3):154-170. doi:10.1038/s41582-
Immunotargets Ther 2021;10:87-101. doi:10.2147/ 020-0313-y
ITT.S287652