Treatment of Neuromyelitis Optica Spectrum Disorders
Treatment of Neuromyelitis Optica Spectrum Disorders
Treatment of Neuromyelitis Optica Spectrum Disorders
CURRENT
OPINION Treatment of neuromyelitis optica spectrum
disorders
Andrew R. Romeo and Benjamin M. Segal
Purpose of review
This review discusses concepts for diagnosing neuromyelitis optica spectrum disorders (NMOSD),
distinguishing NMOSD from other inflammatory diseases of the central nervous system, and highlights
recent and forthcoming data on acute and maintenance therapy of NMOSD.
Recent findings
The neurologic manifestations of NMOSD are heterogenous, extending beyond classic presentations of
optic neuritis and longitudinally extensive transverse myelitis. NMOSD may be comorbid with
rheumatologic diseases, such as systemic lupus erythematosus, but is recognized as a distinct entity. Recent
studies of acute treatment of NMOSD support early use of plasmapheresis. Relapse prevention is essential,
as relapses can be disabling and patients may have only partial recovery. Current practice generally
recommends at least 5 years of maintenance treatment. Recent randomized data demonstrates superiority
of rituximab over azathioprine. Phase 3 trials have recently been completed or are underway studying
novel therapies employing B-cell depletion, complement inhibition, and cell-based mechanisms (among
other mechanisms) for maintenance therapy of NMOSD.
Summary
NMOSD is a heterogeneous but well-defined clinical entity, distinct from other neurologic and systemic
inflammatory diseases, and treatment is poised for expansion.
Keywords
immunosuppressive agents, lupus erythematosus, myelitis, neuromyelitis optica, plasmapheresis, systemic
1040-8711 Copyright ß 2019 Wolters Kluwer Health, Inc. All rights reserved. www.co-rheumatology.com 251
more in length, involve central grey as well as white Antiphospholipid syndrome has been associ-
matter, and may be associated with swelling. In ated with both ischemic myelopathy and inflamma-
contrast, MS cord lesions generally span one vertebral tory myelitis. A Mayo Clinic retrospective study of
segment or less, are confined to white matter tracts 24 cases of LETM with antiphospholipid or b2-gly-
(particularly the dorsal columns), and are not associ- coprotein antibodies detected AQP4-IgG in nearly
ated with significant swelling. These distinctions are half of the study participants (and did not find
not absolute, as up to 14% of spinal cord lesions in AQP4-IgG in control cases with antiphospholipid
&&
individuals with seropositive NMOSD are not longi- antibodies but without LETM) [20 ].
tudinally extensive, and initially distinct MS lesions The pathogenesis of CNS involvement in
can become confluent and span multiple segments as patients with Sjögren’s syndrome is somewhat con-
they accumulate [1,14]. Features of orbital MRI that troversial, and likely heterogeneous. Although
are suggestive of NMOSD include bilateral optic patients with Sjögren’s syndrome may have anti-
nerve involvement, posterior nerve predominance, bodies against other aquaporin isoforms, it appears
and involvement of the optic chiasm [1]. only those patients with clinical diagnoses of
NMOSD have AQP4-IgG [21,22]. Some Sjögren’s
syndrome patients with CNS demyelinating syn-
NMOSD AND RHEUMATOLOGICAL dromes meet diagnostic criteria for MS.
CONDITIONS
NMOSD may be comorbid with other autoimmune
syndromes, such as systemic lupus erythematosus ACUTE TREATMENT
(SLE) or Sjögren’s syndrome [1,15]. Optic neuritis NMOSD relapses have significant potential to result
has also been associated with SLE, rheumatoid arthri- in permanent disability, distinct from MS relapses or
tis, and Sjögren’s syndrome [16]. In a multicenter idiopathic acute transverse myelitis [5]. Whereas
retrospective study of patients with NMO (2006 con- relapse severity may govern the need for acute treat-
sensus diagnostic criteria) or seropositive NMOSD, ment in MS, all NMOSD relapses should be treated
approximately 60% of patients had a positive anti- acutely. A common approach to treatment of
nuclear antibody (ANA) (titer greater than 1:40), with NMOSD relapse begins with corticosteroid pulses
approximately 28% of patients exhibiting ANA titer (e.g., methylprednisolone 1000 mg intravenous
at least 1:320 [7]. Approximately 26 and 11% of the daily for 3–5 days). If the initial response to steroids
patients tested positive for Sjögren’s-syndrome- is inadequate, then plasmapheresis should be con-
related antigen A (SS-A) and Sjögren’s-syndrome- sidered. A recent multicenter retrospective found
related antigen B (SS-B), respectively [7]. A recent that using apheresis therapy as a first-line treatment
Brazilian cohort of NMO patients found lower rates of relapse, or initiating apheresis within days of
&
of ANA, antiSS-A, and antiSS-B seropositivity [17]. relapse onset, predicted full recovery [23 ]. There
Birnbaum et al. [18] described two clinical syn- was no difference in efficacy when comparing
dromes of lupus myelitis: ‘gray matter myelitis’ therapeutic plasma exchange (PLEX) with immu-
(‘flaccidity and hyporeflexia’) and ‘white matter noadsorption. An independent, single-center retro-
myelitis’ (‘spasticity and hyperreflexia’). Patients spective analysis of patients with syndromes
with gray myelitis had significantly higher mean reminiscent of NMOSD found that the probability
Systemic Lupus Erythematosus Disease Activity of complete recovery from relapse decreased as the
Index and erythrocyte sedimentation rate. Relapses time interval between relapse onset and PLEX
&
were more common with white matter myelitis. increased [24 ]. The authors concluded that the first
Over half of the patients with white matter myelitis, five days after relapse onset are a critical period to
but none with grey matter myelitis, had a history of initiate PLEX, and that waiting to initiate PLEX after
optic neuritis. Most of the white matter myelitis failure of response to corticosteroids ‘could be dele-
patients (80%) satisfied 2006 diagnostic criteria terious.’ A smaller retrospective from a Chinese
for neuromyelitis optica; four of seven patients with center found an association between earlier initia-
white matter myelitis were seropositive for AQP4- tion of PLEX and favorable recovery, in patients
IgG. Myelitis was most often longitudinally exten- deemed refractory to intravenous corticosteroids
&
sive in both groups. A recent systematic review and/or intravenous immunoglobulin [25 ]. Though
(which included the Birnbaum cohort), corrobo- there are inherent limitations in the designs of each
rated the overlap between NMOSD and white matter of these retrospective studies, collectively they
&&
lupus myelitis [19 ]. In our opinion, cases of myeli- endorse a low threshold to initiate PLEX.
tis and/or optic neuritis that occur in the setting of A number of novel biological agents are being
SLE, which meet criteria for NMOSD, should be investigated for the treatment of acute NMOSD
treated as NMOSD. episodes. In a Phase 1b trial, a C1-esterase inhibitor
(which prevents activation of the complement sys- P ¼ 0.028). There was no significant difference in
tem) was found to be a well-tolerated adjunctive the rate of side-effects, though one patient in the
therapy to intravenous corticosteroids for AQP4-IgG rituximab group had a severe allergic reaction.
seropositive NMOSD patients presenting with acute A large prospective open-label observational
myelitis or optic neuritis [26]. Bevacizumab, a study of azathioprine and mycophenolate mofetil
monoclonal IgG against vascular endothelial in NMOSD patients in China found no significant
growth factor that ostensibly stabilizes the blood– difference in relapse rates. However, more patients
brain barrier, has also been shown to be well toler- on azathioprine discontinued or switched therapy
ated as an add-on to intravenous corticosteroids in because of adverse effects, including leukopenia,
relapses of NMOSD or presumed NMOSD, in a phase hepatotoxicity, hair loss, and nausea/vomiting
&
1b trial [27]. Ublituximab, a B-cell depleting mono- [37 ].
clonal antibody that targets the cell surface mole- Granulocytes are thought to contribute to the
cule CD20, is currently under study as an add-on to inflammatory cascade in NMOSD lesions. Cetirizine
steroids for acute treatment of NMOSD relapse is a second-generation antihistamine, purported to
(NCT02276963). Each of these novel acute treat- stabilize eosinophils. Cetirizine was well tolerated as
ments will need to be studied in randomized trials an add on to typical NMOSD maintenance therapy
to assess efficacy. in a recent open-label pilot study, but a controlled
study will be necessary to demonstrate efficacy [38].
Anecdotally, NMOSD patients have been treated
MAINTENANCE TREATMENT with MS disease modifying therapies, sometimes
Relapse prevention is essential, as relapses can be inadvertently when the spectrum was still being
disabling and patients may have only partial recov- defined. Extrapolating from uncontrolled case
ery. Current practice generally recommends at least reports and series, fingolimod, alemtuzumab, and
5 years of maintenance treatment [28]. To date there dimethyl fumarate are suspected to be (at least)
&
are no published randomized placebo-controlled ineffective in NMOSD [39 ,40]. Interferon-b has
trials for maintenance treatment of NMOSD, been reported to actually exacerbate NMO [8].
though multiple trials are currently underway. Natalizumab is a monoclonal antibody against the
Empiric maintenance regimens have included adhesion molecule, a4-integrin, effective for treat-
mycophenolate mofetil, azathioprine, rituximab, ment of relapsing MS and Crohn’s disease [41,42]. It
& &
and chronic corticosteroids [29,30 ,31 ,32,33,34]. curtails the migration of lymphocytes from the blood
Our practice has favored rituximab for maintenance stream into solid tissues. A recent study identified
treatment. So far no distinctions have been made four natalizumab-treated patients (initially misdiag-
between seronegative and seropositive NMOSD nosed with MS) who met diagnostic criteria for sero-
&
patients in regards to treatment. A multinational, positive NMOSD [43 ]. The same research group
multicenter retrospective analysis found no differ- previously reported five cases of NMOSD patients
ence between seropositive and seronegative initially misdiagnosed with MS and treated with
NMOSD patients in their responsiveness to rituxi- natalizumab [44]. Taken together, all nine patients
&
mab or mycophenolate mofetil [35 ]. Comparative continued to have relapses while on natalizumab.
studies to help guide initial treatment choice or A number of novel therapies have been sug-
sequencing are limited. Our discussion will focus gested or are currently under study for treatment
on recent note-worthy studies of maintenance of NMOSD. Eculizumab is a monoclonal antibody
therapies as well as on-going inquiries and future against terminal complement component 5 (C5); it
directions. has been approved for treatment of paroxysmal
Rituximab and azathioprine were compared nocturnal hemoglobinuria and atypical hemolytic
head-to-head in an open-label randomized trial, uremic syndrome. An open-label pilot study of
which included seropositive and seronegative eculizumab in NMOSD demonstrated safety, and
&&
NMOSD patients [36 ]. The primary outcome was 12 of 14 patients remained relapse-free after
annualized relapse rate (ARR) at 12 months. Despite 12 months [45]. Most of these patients had failed
randomization, the rituximab group had a signifi- standard NMOSD maintenance therapies prior to
cantly higher mean Expanded Disability Status Scale enrollment. A double-blind, placebo-controlled trial
and mean ARR at baseline compared with the aza- of eculizumab has completed and results have not
thioprine group. The rituximab group had a signifi- yet been reported (NCT01892345). A double-blind,
cantly greater decline in ARR from baseline to placebo-controlled trial of MEDI-551, a monoclonal
12 months (1.09 vs. 0.49, P < 0.001), and a signifi- antibody against the B-cell/plasmablast marker
cantly greater proportion of relapse-free patients CD19 is currently underway (NCT02200770) [46].
(65 vs. 41.3% with intention to treat analysis, Tocilizumab is a monoclonal antibody against
1040-8711 Copyright ß 2019 Wolters Kluwer Health, Inc. All rights reserved. www.co-rheumatology.com 253
21. Tzartos JS, Stergiou C, Daoussis D, et al. Antibodies to aquaporins are 37. Chen H, Qiu W, Zhang Q, et al. Comparisons of the efficacy and tolerability of
frequent in patients with primary Sjogren’s syndrome. Rheumatology (Oxford) & mycophenolate mofetil and azathioprine as treatments for neuromyelitis optica
2017; 56:2114–2122. and neuromyelitis optica spectrum disorder. Eur J Neurol 2017; 24:
22. Birnbaum J, Atri NM, Baer AN, et al. Relationship between neuromyelitis 219–226.
optica spectrum disorder and Sjogren’s syndrome: central nervous system This is a prospective open-label study comparing mycophenolate mofetil and
extraglandular disease or unrelated, co-occurring autoimmunity? Arthritis azathioprine for maintenance therapy of NMOSD.
Care Res (Hoboken) 2017; 69:1069–1075. 38. Katz Sand I, Fabian MT, Telford R, et al. Open-label, add-on trial of cetirizine for
23. Kleiter I, Gahlen A, Borisow N, et al. Apheresis therapies for NMOSD attacks: neuromyelitis optica. Neurol Neuroimmunol Neuroinflamm 2018; 5:e441.
& a retrospective study of 207 therapeutic interventions. Neurol Neuroimmunol 39. Yamout BI, Beaini S, Zeineddine MM, Akkawi N. Catastrophic relapses
Neuroinflamm 2018; 5:e504. & following initiation of dimethyl fumarate in two patients with neuromyelitis
This retrospective study demonstrates benefit with early use of plasma exchange optica spectrum disorder. Mult Scler 2017; 23:1297–1300.
for treatment of NMOSD relapse. This report adds to the body of evidence that MS disease modifying therapies may
24. Bonnan M, Valentino R, Debeugny S, et al. Short delay to initiate plasma be ineffective or in some cases detrimental in NMOSD.
& exchange is the strongest predictor of outcome in severe attacks of NMO 40. Azzopardi L, Cox AL, McCarthy CL, et al. Alemtuzumab use in neuromyelitis
spectrum disorders. J Neurol Neurosurg Psychiatry 2018; 89:346–351. optica spectrum disorders: a brief case series. J Neurol 2016; 263:25–29.
This retrospective study demonstrates benefit with early use of plasma exchange 41. Polman CH, O’Connor PW, Havrdova E, et al. A randomized, placebo-
for treatment of NMOSD relapse. controlled trial of natalizumab for relapsing multiple sclerosis. N Engl J
25. Jiao Y, Cui L, Zhang W, et al. Plasma exchange for neuromyelitis optica Med 2006; 354:899–910.
& spectrum disorders in Chinese patients and factors predictive of short-term 42. Sandborn WJ, Colombel JF, Enns R, et al. Natalizumab induction and main-
outcome. Clin Ther 2018; 40:603–612. tenance therapy for Crohn’s disease. N Engl J Med 2005; 353:1912–1925.
This retrospective study demonstrates benefit with early use of plasma exchange 43. Gahlen A, Trampe AK, Haupeltshofer S, et al. Aquaporin-4 antibodies in
for treatment of NMOSD relapse. & patients treated with natalizumab for suspected MS. Neurol Neuroimmunol
26. Levy M, Mealy MA. Purified human C1-esterase inhibitor is safe in acute Neuroinflamm 2017; 4:e363.
relapses of neuromyelitis optica. Neurol Neuroimmunol Neuroinflamm 2014; This retrospective analysis adds to the body of evidence that MS disease
1:e5. modifying therapies may be ineffective for maintenance therapy in NMOSD.
27. Mealy MA, Shin K, John G, Levy M. Bevacizumab is safe in acute relapses of 44. Kleiter I, Hellwig K, Berthele A, et al. Failure of natalizumab to prevent relapses
neuromyelitis optica. Clin Exp Neuroimmunol 2015; 6:413–418. in neuromyelitis optica. Arch Neurol 2012; 69:239–245.
28. Montcuquet A, Collongues N, Papeix C, et al. Effectiveness of mycophenolate 45. Pittock SJ, Lennon VA, McKeon A, et al. Eculizumab in AQP4-IgG-positive
mofetil as first-line therapy in AQP4-IgG, MOG-IgG, and seronegative neu- relapsing neuromyelitis optica spectrum disorders: an open-label pilot study.
romyelitis optica spectrum disorders. Mult Scler 2017; 23:1377–1384. Lancet Neurol 2013; 12:554–562.
29. Wingerchuk DM, Weinshenker BG. Neuromyelitis optica. Curr Treat Options 46. Cree BA, Bennett JL, Sheehan M, et al. Placebo-controlled study in neuromye-
Neurol 2008; 10:55–66. litis optica-ethical and design considerations. Mult Scler 2016; 22:862–872.
30. Bichuetti DB, Perin MM, Souza NA, Oliveira EM. Treating neuromyelitis 47. Ringelstein M, Ayzenberg I, Harmel J, et al. Long-term therapy with interleukin
& optica with azathioprine: 20-year clinical practice. Mult Scler 2018; 6 receptor blockade in highly active neuromyelitis optica spectrum disorder.
1352458518776584. JAMA Neurol 2015; 72:756–763.
This retrospective study analyzes the efficacy of azathioprine for maintenance 48. Tradtrantip L, Zhang H, Saadoun S, et al. Antiaquaporin-4 monoclonal antibody
treatment of NMOSD. blocker therapy for neuromyelitis optica. Ann Neurol 2012; 71:314–322.
31. Huang Q, Wang J, Zhou Y, et al. Low-dose mycophenolate mofetil for 49. Scott K, Hayden PJ, Will A, et al. Bortezomib for the treatment of multiple
& treatment of neuromyelitis optica spectrum disorders: a prospective multi- myeloma. Cochrane Database Syst Rev 2016; 4:CD010816.
center study in South China. Front Immunol 2018; 9:2066. 50. Robak T, Huang H, Jin J, et al. Bortezomib-based therapy for newly diagnosed
This article presents an open-label prosepctive study of mycophenolate mofetil mantle-cell lymphoma. N Engl J Med 2015; 372:944–953.
efficacy and safety as maintenance therapy for NMOSD. 51. Zhang C, Tian DC, Yang CS, et al. Safety and efficacy of bortezomib in
32. Torres J, Pruitt A, Balcer L, et al. Analysis of the treatment of neuromyelitis && patients with highly relapsing neuromyelitis optica spectrum disorder. JAMA
optica. J Neurol Sci 2015; 351:31–35. Neurol 2017; 74:1010–1012.
33. Radaelli M, Moiola L, Sangalli F, et al. Neuromyelitis optica spectrum dis- This is a pilot study of a 26S proteasome inhibitor which may prove to be a novel
orders: long-term safety and efficacy of rituximab in Caucasian patients. Mult maintenance therapy for NMOSD.
Scler 2016; 22:511–519. 52. Burt RK, Balabanov R, Han X, et al. Association of nonmyeloablative hema-
34. Damato V, Evoli A, Iorio R. Efficacy and safety of rituximab therapy in topoietic stem cell transplantation with neurological disability in patients with
neuromyelitis optica spectrum disorders: a systematic review and meta- relapsing-remitting multiple sclerosis. JAMA 2015; 313:275–284.
analysis. JAMA Neurol 2016; 73:1342–1348. 53. Nash RA, Hutton GJ, Racke MK, et al. High-dose immunosuppressive therapy
35. Mealy MA, Kim SH, Schmidt F, et al. Aquaporin-4 serostatus does not predict and autologous hematopoietic cell transplantation for relapsing-remitting
& response to immunotherapy in neuromyelitis optica spectrum disorders. Mult multiple sclerosis (HALT-MS): a 3-year interim report. JAMA Neurol 2015;
Scler 2018; 24:1737–1742. 72:159–169.
This retrospective study of seronegative and seropositive NMOSD patients found 54. Muraro PA, Pasquini M, Atkins HL, et al. Long-term outcomes after autologous
no significant difference in treatment efficacy. hematopoietic stem cell transplantation for multiple sclerosis. JAMA Neurol
36. Nikoo Z, Badihian S, Shaygannejad V, et al. Comparison of the efficacy of 2017; 74:459–469.
&& azathioprine and rituximab in neuromyelitis optica spectrum disorder: a 55. Fu Y, Yan Y, Qi Y, et al. Impact of autologous mesenchymal stem cell infusion
randomized clinical trial. J Neurol 2017; 264:2003–2009. on neuromyelitis optica spectrum disorder: a pilot, 2-year observational study.
This is the first published randomized trial of maintenance therapy in NMOSD. CNS Neurosci Ther 2016; 22:677–685.
Rituximab is shown to have superior efficacy compared to azathioprine, despite 56. Gill S, Maus MV, Porter DL. Chimeric antigen receptor T cell therapy: 25 years
unbalanced treatment assignment potentially favoring azathioprine. in the making. Blood Rev 2016; 30:157–167.
1040-8711 Copyright ß 2019 Wolters Kluwer Health, Inc. All rights reserved. www.co-rheumatology.com 255