Skip to main content
Advertisement
  • Neurology.org
  • Journals
    • Neurology
    • Clinical Practice
    • Education
    • Genetics
    • Neuroimmunology & Neuroinflammation
  • Online Sections
    • Neurology Video Journal Club
    • Neurology: Neuroimmunology & Neuroinflammation COVID-19 Article Hub
    • Diversity, Equity, & Inclusion (DEI)
    • Innovations in Care Delivery
    • Practice Buzz
    • Practice Current
    • Residents & Fellows
    • Without Borders
  • Collections
    • COVID-19
    • Disputes & Debates
    • Health Disparities
    • Infographics
    • Null Hypothesis
    • Patient Pages
    • Topics A-Z
    • Translations
  • Podcast
  • CME
  • About
    • About the Journals
    • Contact Us
    • Editorial Board
  • Authors
    • Submit New Manuscript
    • Submit Revised Manuscript
    • Author Center

Advanced Search

Main menu

  • Neurology.org
  • Journals
    • Neurology
    • Clinical Practice
    • Education
    • Genetics
    • Neuroimmunology & Neuroinflammation
  • Online Sections
    • Neurology Video Journal Club
    • Neurology: Neuroimmunology & Neuroinflammation COVID-19 Article Hub
    • Diversity, Equity, & Inclusion (DEI)
    • Innovations in Care Delivery
    • Practice Buzz
    • Practice Current
    • Residents & Fellows
    • Without Borders
  • Collections
    • COVID-19
    • Disputes & Debates
    • Health Disparities
    • Infographics
    • Null Hypothesis
    • Patient Pages
    • Topics A-Z
    • Translations
  • Podcast
  • CME
  • About
    • About the Journals
    • Contact Us
    • Editorial Board
  • Authors
    • Submit New Manuscript
    • Submit Revised Manuscript
    • Author Center
  • Home
  • Articles
  • Issues
  • COVID-19 Article Hub
  • Infographics & Video Summaries

User menu

  • My Alerts
  • Log in

Search

  • Advanced search
Neurology Neuroimmunology & Neuroinflammation
Home
A peer-reviewed clinical and translational neurology open access journal
  • My Alerts
  • Log in
Site Logo
  • Home
  • Articles
  • Issues
  • COVID-19 Article Hub
  • Infographics & Video Summaries

Share

January 2021; 8 (1) Views & ReviewsOpen Access

Targeting B cells to modify MS, NMOSD, and MOGAD

Part 2

View ORCID ProfileJonas Graf, Jan Mares, Michael Barnett, View ORCID ProfileOrhan Aktas, View ORCID ProfilePhilipp Albrecht, Scott S. Zamvil, Hans-Peter Hartung
First published December 16, 2020, DOI: https://doi.org/10.1212/NXI.0000000000000919
Jonas Graf
From the Department of Neurology (J.G., O.A., P.A., H.-P.H.), University Hospital, Medical Faculty Heinrich Heine University, Düsseldorf, Germany; Department of Neurology (J.M.), Palacky University, Olomouc, Czech Republic; Department of Neurology, Brain and Mind Centre (M.B., H.-P.H.), Department of Neurology, University of Sydney, New South Wales, Australia; and Department of Neurology (S.S.Z.), UCSF Weill Institute of Neurosciences, University of California at San Francisco.
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
  • ORCID record for Jonas Graf
Jan Mares
From the Department of Neurology (J.G., O.A., P.A., H.-P.H.), University Hospital, Medical Faculty Heinrich Heine University, Düsseldorf, Germany; Department of Neurology (J.M.), Palacky University, Olomouc, Czech Republic; Department of Neurology, Brain and Mind Centre (M.B., H.-P.H.), Department of Neurology, University of Sydney, New South Wales, Australia; and Department of Neurology (S.S.Z.), UCSF Weill Institute of Neurosciences, University of California at San Francisco.
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Michael Barnett
From the Department of Neurology (J.G., O.A., P.A., H.-P.H.), University Hospital, Medical Faculty Heinrich Heine University, Düsseldorf, Germany; Department of Neurology (J.M.), Palacky University, Olomouc, Czech Republic; Department of Neurology, Brain and Mind Centre (M.B., H.-P.H.), Department of Neurology, University of Sydney, New South Wales, Australia; and Department of Neurology (S.S.Z.), UCSF Weill Institute of Neurosciences, University of California at San Francisco.
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Orhan Aktas
From the Department of Neurology (J.G., O.A., P.A., H.-P.H.), University Hospital, Medical Faculty Heinrich Heine University, Düsseldorf, Germany; Department of Neurology (J.M.), Palacky University, Olomouc, Czech Republic; Department of Neurology, Brain and Mind Centre (M.B., H.-P.H.), Department of Neurology, University of Sydney, New South Wales, Australia; and Department of Neurology (S.S.Z.), UCSF Weill Institute of Neurosciences, University of California at San Francisco.
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
  • ORCID record for Orhan Aktas
Philipp Albrecht
From the Department of Neurology (J.G., O.A., P.A., H.-P.H.), University Hospital, Medical Faculty Heinrich Heine University, Düsseldorf, Germany; Department of Neurology (J.M.), Palacky University, Olomouc, Czech Republic; Department of Neurology, Brain and Mind Centre (M.B., H.-P.H.), Department of Neurology, University of Sydney, New South Wales, Australia; and Department of Neurology (S.S.Z.), UCSF Weill Institute of Neurosciences, University of California at San Francisco.
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
  • ORCID record for Philipp Albrecht
Scott S. Zamvil
From the Department of Neurology (J.G., O.A., P.A., H.-P.H.), University Hospital, Medical Faculty Heinrich Heine University, Düsseldorf, Germany; Department of Neurology (J.M.), Palacky University, Olomouc, Czech Republic; Department of Neurology, Brain and Mind Centre (M.B., H.-P.H.), Department of Neurology, University of Sydney, New South Wales, Australia; and Department of Neurology (S.S.Z.), UCSF Weill Institute of Neurosciences, University of California at San Francisco.
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Hans-Peter Hartung
From the Department of Neurology (J.G., O.A., P.A., H.-P.H.), University Hospital, Medical Faculty Heinrich Heine University, Düsseldorf, Germany; Department of Neurology (J.M.), Palacky University, Olomouc, Czech Republic; Department of Neurology, Brain and Mind Centre (M.B., H.-P.H.), Department of Neurology, University of Sydney, New South Wales, Australia; and Department of Neurology (S.S.Z.), UCSF Weill Institute of Neurosciences, University of California at San Francisco.
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Full PDF
Citation
Targeting B cells to modify MS, NMOSD, and MOGAD
Part 2
Jonas Graf, Jan Mares, Michael Barnett, Orhan Aktas, Philipp Albrecht, Scott S. Zamvil, Hans-Peter Hartung
Neurol Neuroimmunol Neuroinflamm Jan 2021, 8 (1) e919; DOI: 10.1212/NXI.0000000000000919

Citation Manager Formats

  • BibTeX
  • Bookends
  • EasyBib
  • EndNote (tagged)
  • EndNote 8 (xml)
  • Medlars
  • Mendeley
  • Papers
  • RefWorks Tagged
  • Ref Manager
  • RIS
  • Zotero
Permissions

Make Comment

See Comments

Downloads
2634

Share

  • Article
  • Figures & Data
  • Info & Disclosures
Loading

Abstract

Ocrelizumab, rituximab, ofatumumab, ublituximab, inebilizumab, and evobrutinib are immunotherapies that target various B cell–related proteins. Most of these treatments have proven efficacy in relapsing and progressive forms of MS and neuromyelitis optica spectrum disease (NMOSD) or are in advanced stages of clinical development. Currently, ocrelizumab and inebilizumab are licensed for treatment of MS and NMOSD, respectively. This part of the review focuses on monoclonal antibody B cell–depleting strategies in NMOSD and the emerging related myelin oligodendrocyte glycoprotein (MOG) immunoglobulin G–associated disease (MOGAD). Case series and phase 2/3 studies in these inflammatory disorders are assessed. The safety profile of long-term B-cell depletion in MS, NMOSD, and MOGAD will be highlighted. Finally implications of the current coronavirus disease 2019 pandemic on the management of patients with these disorders and the use of B cell–depleting agents will be discussed.

Glossary

COVID-19=
coronavirus disease 2019;
IgG=
immunoglobulin G;
NEMOS=
Neuromyelitis Optica Study Group;
MOG=
myelin oligodendrocyte glycoprotein;
MOGAD=
MOG IgG–associated disease;
NMOSD=
neuromyelitis optica spectrum disease;
PML=
progressive multifocal leukoencephalopathy;
SARS-CoV2=
severe acute respiratory syndrome coronavirus 2

NMOSD

Following identification of immunoglobulin G (IgG) antibodies to aquaporin 4 in some 75%–80% of patients, research in humans and animal models has implicated aberrant B-cell responses in the pathogenesis of neuromyelitis optica spectrum disease (NMOSD)1,–,4 These include defects in central and peripheral tolerance mechanisms allowing emergence of pathogenic antibodies, impairment of B regulatory activity, heightened production of proinflammatory cytokines, and complement activation.

Rituximab

Relevant CD19− and CD20− B lymphocyte–depleting studies in NMOSD are summarized in the table. Most studies were observational and retrospective.5,–,7 Rituximab has long been considered a first-line treatment in NMOSD.8 This recommendation for many years relied on multiple smaller observational studies and expert opinion (Class IV evidence).7,9,10 Two recent meta-analyses analyzed 4385 and 577 patients6 from some 25 interpretable studies. The largest multicenter retrospective study from Italy included 73 patients. Two regimens were mostly applied: either 375 mg/m2 weekly for 3 weeks or 1 g every 2 weeks twice. Rituximab reduces relapse frequency and neurologic disability but does not alter the frequencies of autoreactive B cells and does not reset defective early B-cell tolerance checkpoints.11 B cells remain depleted in the circulation for up to 12 months. Bimonthly assessment of CD19/CD27-positive memory B cells was proposed to determine the time point for reintroduction of rituximab therapy.12,–,16 A multinational 14-center study reported real-world data on 67 children having aquaporin 4 antibody–positive NMOSD.17 They had a median follow-up of 4 years. Their mean age at onset was 10.2 years. Twenty-nine children received rituximab, half of them first line and one-third second line. In the entire group, the annualized relapse rate dropped from 2.5 to 0.14 on treatment. The patients on first-line rituximab treatment experienced no further relapse; 24% had further attacks. Two patients were switched to ofatumumab due to severe infusion-related reactions. One child developed persistent neutropenia.17

View this table:
  • View inline
  • View popup
  • Download powerpoint
Table

Trials of CD20−and CD19-depleting monoclonal antibodies for NMOSD

Recently, a multicenter, randomized, double-blind, placebo-controlled clinical trial has been performed in Japan, proving its capacity to prevent relapses,18 which can be considered a breakthrough.19 Given the small size and some other methodological limitations, a larger, randomized, controlled phase 3 trial would need to be undertaken to validate these observations.

Strategies to individualize rituximab treatment by changing intervals of redosing rely on determining CD19-positive or CD19+ CD27+ memory B cells or switched memory B cells (CD19+/CD27+/IgM−/IgD−) as a percentage of peripheral blood mononuclear cells.13,20

Inebilizumab

Inebilizumab is a glycoengineered, afucosylated anti-CD19 antibody that was specifically designed to increase affinity to FcyRIIA and thereby enhance antibody-dependent cytotoxicity.21,22 CD19 is more broadly expressed on cells of the B-cell lineage. Late-stage memory B cells and plasmablasts carry CD19 on their surface but are CD20 negative. Hence, it was predicted that depleting CD19 carrying cells would affect pathogenic autoantibody production more markedly than targeting CD20 cells. In vitro assays examining antibody-dependent cell-mediated cytotoxicity demonstrated B-cell depletion occurring at lower antibody concentrations than with rituximab.21,23 Studies in preclinical models and phase 1 trails in systemic sclerosis and MS provided evidence for effective CD19 B-cell depletion.23,24

The N-MOmentum trial was the largest ever conducted in NMOSD. This international multicenter double-blind, randomized placebo-controlled phase 2/3 study with an open-label extension period investigated the safety and efficacy of this CD19-depleting monoclonal antibody in aquaporin 4 IgG-positive and -negative patients with NMOSD. Of 231 patients, 175 were randomized to inebilizumab and 56 to placebo. Ninety-two percent and 93% were aquaporin 4 IgG positive. The primary outcome was time to onset of an NMOSD attack determined by an adjudication committee. Secondary end points included worsening of Expanded Disability Status Scale from baseline, low-contrast visual acuity, cumulative number of active MR lesions (new gadolinium-enhancing or new or enlarging T2 lesions), and number of disease-related hospitalizations. Inebilizumab markedly reduced the risk of attacks, the main cause of disability in this crippling disease25 (table). This trial was terminated early by an independent data-monitoring committee because of a clear demonstration of efficacy. Twelve percent of the patients receiving inebilizumab vs 39% of the patients receiving placebo encountered an attack (p < 0.0001; hazard ratio 0–272). Significant and robust B-cell depletion occurred within 4 weeks. Expanded Disability Status Scale worsening was less with inebilizumab; no difference was observed in low-contrast visual acuity, but post hoc analysis showed a lower risk of inebilizumab-treated patients to experience optic neuritis. Cumulative active MRI lesion count was lower in inebilizumab-treated participants as were NMOSD-related hospitalizations.25

Adverse events occurred at similar frequency in both treatment arms. Thus, CD19 depletion is a well-validated treatment option for NMOSD.21,–,23,26 Further safety studies are needed as long-term CD19 depletion may be associated with an elevated risk of opportunistic infections.27

Perspective

A phase 1 study of ublituximab as an add-on therapy to methylprednisolone in an acute relapse of NMOSD suggests that CD20 depletion is safe in this regime and may improve neurologic outcome.28 Further placebo-controlled studies are to confirm these findings. Studies that emphasize the efficacy of anti–interleukin 6 receptor antibodies, e.g., sartralizumab and tocilizumab, underline the importance of interleukin 6 as a B cell–activating factor in NMOSD.29,–,31

MOGAD

MOG, myelin oligodendrocyte glycoprotein, is a minor component of CNS myelin expressed on its outer layer. Conformation-dependent antibodies have been described in a number of CNS inflammatory disease, but recent evidence suggests that MOG IgG–associated disease (MOGAD), affecting children and adults, is a distinct clinical and pathologic entity with optic neuritis, myelitis, isolated brainstem, encephalitis, encephalopathic, or acute disseminated encephalomyelitis-like presentation. It runs a monophasic or more frequently relapsing course.1,32,–,38 Pathologically, inflammation, demyelination, and preservation of astrocytes have been observed. Studies on the pathogenicity of MOG IgG have been largely undertaken in rodent models where antibodies and autoreactive T cells in isolation or in concert induce injury.4,38,–,40 In 60% of 21 MOG IgG–positive patients, MOG-reactive B cells could be isolated. They displayed a heterogeneous pattern of antibody production.41 In a flow cytometric study looking at B- and T-cell populations in 19 MOG IgG–positive patients, regulatory B cells were lower and memory B cells higher in number compared with controls.42 A range of immunosuppressive and immunomodulatory drugs have been used, among them rituximab, mostly to prevent further attacks.

In an EU Pediatric Demyelinating Disease Consortium study, 102 children received rituximab first, second, or third line. The majority continued to relapse despite effective peripheral B-cell depletion.43 An Australian multicenter study of 33 children and 26 adult patients with relapsing MOG IgG–associated demyelination found 1 of 7 patients not responding to rituximab, although B cells were depleted. The German Neuromyelitis Optica Study Group (NEMOS) reported relapses in 6 of 9 patients receiving rituximab.32 In a prospective French study of 16 adult patients with MOG IgG, one-third of patients relapsed in the presence of less than 0.05% memory B cells.44 In the largest international cohort retrospectively analyzing data from 121 patients, relapse rates on rituximab declined by 37%. After 2 years, 33% were predicted to remain relapse free.45 Effect size was largest with first-line administration and higher in adult compared with pediatric patients. In summary, rituximab has shown efficacy in up to two-thirds of patients with MOGAD, but a sizable fraction continued to have attacks with full B-cell depletion.

CD20/CD19 B-cell depletion in MS, NMOSD, and MOGAD: adverse events and safety profiles

The biggest concerns with immune cell depletion therapies are the occurrence of serious infections, opportunistic infections, and malignancies as consequences of lymphopenia and impaired lymphocyte function.46

Three controlled phase 2 and phase 2/3 trials and retrospective series are available for determining the safety of rituximab in MS,46 NMOSD, and MOGAD. Overall, rituximab was well tolerated apart from infusion-related reactions, which tended to diminish in severity with repeat infusion. The largest observational study with a retrospective design comes from Sweden where rituximab has been commonly used as preferred high-efficacy disease-modifying treatment of MS. In 2016, safety results from 822 rituximab-treated patients (557 relapsing-remitting, 198 secondary progressive, and 67 primary progressive MS) were reported.47 Again, infusion-related reactions were the most frequent adverse events occurring during 7.8% of infusions. They were usually mild. Importantly, 76 infections developed in 72 patients. A 2-center retrospective study from the United States recognized infections at a rate of 38.6/1,000 patient-years in 907 patients studied.48 A comparative study of infection risks among patients on various disease-modifying agents again from the Swedish national MS registry cohort associated rituximab use with the highest incidence of infections.49 Most common were upper respiratory and lower urinary tract infections and pneumonia. In this study of 3,260 patients receiving rituximab, 2 cases of carry-over progressive multifocal leukoencephalopathy (PML) were diagnosed. In a study from Finland of 72 patients with MS, rituximab caused severe neutropenia in 2 requiring discontinuation.50 In the US study referred to above, neutropenia of less than 500 cells/mm3 was noted in 1.2% of patients on rituximab.48 The same group of investigators observed low values of IgG evolving over a mean treatment period of 31.1 months and a mean cumulative dose of 4,012 mg in 6%.48

When the FDA Adverse Event Reporting Database was recently interrogated,51 623 and 7,984 reports for rituximab and ocrelizumab, respectively, were identified. Serious adverse events were more commonly reported for rituximab as were adverse events related to blood, lymphatic, and immune system. On the other hand, infections, although mild or moderate such as nasopharyngitis and upper respiratory tract infection, were twice as frequent with ocrelizumab whose safety profile was in line with published data. The authors of this report speculated about a different or more extensive depletion of B cells by ocrelizumab underlying these observations but also admitted a number of methodological limitations51 inherent in interpreting data from such a registry.

Linking data from the Swedish national MS registry and Swedish Cancer registry, Alping et al.52 did not find a higher incidence of invasive cancers in 4,187 first-ever treated rituximab MS patients. These data are in line with the long-term experience of over 11 years in the Rheumatoid Arthritis Global Clinical Trial Program that enrolled 3,595 patients receiving a mean of 4 courses.53

In essence, safety data for rituximab in NMOSD are comparable. A systematic review and meta-analysis of 46 studies published between 2000 and 2015 encompassing 438 patients found infusion-related adverse events in 10.3%, infections in 9.1%, persistent leukopenia in 4.6%, and posterior reversible encephalopathy in 0.5%.5 A single-center prospective observational study from Milan recorded serious infections in 5 of 21 patients who had leukopenia and hypogammaglobulinemia.10

Regarding ocrelizumab, the safety profile has recently been systematically reviewed.54 Data on longer-term side effects, particularly relating to reactivation of infections and induction of malignancies following completion of the pivotal trials, have been presented at conferences and in a recent report of the 5-year open-label extension pooled analysis of the twin OPERA (Study of Ocrelizumab in Comparison With Interferon Beta-1a (Rebif) in Participants With Relapsing Multiple Sclerosis) studies in relapsing MS.55 These analyses did not identify new safety signals among the approximately 150,000 patients who had been treated with ocrelizumab worldwide (180,000 patient-years) (Roche, data on file, March 2, 2020).56 In the pivotal clinical trials, 11 cancer cases occurred in patients with PPMS on ocrelizumab in the placebo-controlled phase. These included 4 cases of breast cancer and 3 cases of basal cell carcinoma, as well as 1 case each of endometrioid adenocarcinoma, anaplastic large cell lymphoma, malignant fibrous histiocytoma, and pancreatic carcinoma.

The open-label extension study reported 1 case of basal cell carcinoma and 1 case of squamous cell carcinoma. The placebo group included 1 basal cell carcinoma and 1 cervical adenocarcinoma in situ.57 Among patients with relapsing MS receiving ocrelizumab, there were 2 cases of breast cancer, 1 renal cell carcinoma, and 1 malignant melanoma compared with the placebo group, which reported 1 mantle cell carcinoma and 1 squamous cell carcinoma case. The ocrelizumab open-label extension study reported 2 cases of breast cancer, 2 cases of basal cell carcinoma, and 1 case of malignant melanoma.58 A more recent analysis of 5,051 patients who received ocrelizumab in controlled trials and open-label extensions for up to 6.5 years presented the latest available data set at the 2020 EAN Meeting. Thirty-six patients with breast cancer (benign, malignant, and nonspecified) were recorded and 4 patients with malignant melanoma. Single cases of other malignancies were observed. However, these rates were in the range of incidence data from the Danish MS Registry and the US National Cancer Institute SEER (Surveillance, Epidemiology, and End Results) general population database and hence do not signify a specific malignancy signal associated with ocrelizumab.58

In the phase 3 trials, patients with relapsing MS treated with ocrelizumab, compared with interferon beta-1a, exhibited a higher incidence of varicella zoster infections (17 vs 8, respectively78), and patients with PPMS receiving ocrelizumab, compared with placebo, had a higher prevalence of oral herpes infections (2.3% vs 0.4%, respectively).58 Case reports highlight the relevance of hepatitis reactivation in the setting of CD20 cell depletion59,60 and emphasize the need for and the benefits of patient risk stratification before the start of therapy.e1

Similarly to its precursor, the chimeric CD20 monoclonal antibody rituximab,e2 the postauthorization use of ocrelizumab increased the risk of developing late-onset neutropenia,e3,e4 tumefactive demyelinating lesions,e5 and PML. As of January 31, 2020, when 150,000 patients were on ocrelizumab worldwide, 9 confirmed PML cases were reported.54,57,58 Eight patients developed PML on ocrelizumab after switching from previous disease-modifying therapies, while to date, only 1 case of non–carry-over PML has been reported in an elderly patient who had low T lymphocyte counts before starting ocrelizumab. He finally passed away.e6 Isolated case reports of herpes simplex type 2 encephalitis,e7 B19 parvovirus infection,e8 and 2 cases of meningitise9 have been published in patients treated with ocrelizumab.e9 Ocrelizumab therapy is unlikely to be associated with an elevated risk of tuberculosis infection.e10 Secondary immunoglobulin deficiency can precipitate serious infections.e11 A review investigating potential associations of B-cell depletion treatment with rituximab and the risk of developing hypogammaglobulinemia or infection failed to identify any significant risk factors.e12 Ongoing data analyses examine the incidence over time of hypogammaglobulinemia in patients treated with ocrelizumab. Over the 5 years of the double-blind pooled OPERA trials in relapsing MS and their open-label extension up to 5 years, IgG levels were lowered below lower limit of normal in 5.4%, IgA in 5.1%, and IgM in 29.5% of patients.55 A drug reaction with eosinophilia and systemic symptoms after ocrelizumab therapy has also been described.e13

There were no specific safety concerns (i.e., spontaneous abortions or fetal malformations) in pregnant women who underwent CD20 depletion with rituximab during the first 6 months of pregnancy.e14 A current study suggests that monoclonal antibodies may be safe in breastfeeding.e15 Although a retrospective analysis of patients with neuroimmunologic diseases who underwent rituximab therapy over a period of 7 yearse16 and a meta-analysis of patients with rheumatoid arthritise17 confirm rituximab's good safety profile, the effects of long-term B-cell depletion still remain unclear.

A retrospective study demonstrated that the incidence of infusion-associated reactions was significantly reduced when histamine antagonists and oral fluid were also administered.e18 There are very limited data available on the intrathecal administration of CD20 B cell–depleting monoclonal antibodies.e19

Given the aggregate evidence, it makes sense to perform regular clinical surveillance including routine blood work of patients with MS undergoing CD20 cell–depleting therapies.e20–e22

Implications of the COVID-19/SARS-CoV2 pandemic

There is concern that B cell–depleting agents may increase the risk of viral diseases, including severe acute respiratory syndrome coronavirus 2 (SARS-CoV2)/coronavirus disease 2019 (COVID-19). Rituximab has in the past not been associated with an elevated risk of developing viral infections.e23 Initially, evidence suggested there was not a more severe course of COVID-19 in ocrelizumab-treated patients with MS.e24 This is based on a pharmacovigilance analysis conducted by Roche. As of 30 April 2020, 26 suspected and 74 confirmed cases of patients with MS on ocrelizumab contracting SARTS-CoV2 were identified. The great majority were asymptomatic, mild, or moderately affected and 64 of 64 patients fully recovered.77

In line with these observations are a case report of a patient with MS receiving ocrelizumab who had a mild COVID-19 infectione25 and a case series of 60 patients under CD20 depletion from Spain.e26 A recently published large case series from France found that rituximab- and ocrelizumab-treated patients with MS who got infected with SARS-CoV2 have a milder course of COVID-19.e27 However, a cross-sectional survey in Iran revealed that B-cell depletion may increase the susceptibility to contracting COVID-19.e28 A recent large scale study of some 200 people with MS and confirmed COVID-19 infection from Italy noted a higher infection frequency among patients on anti-CD20 monoclonal antibody therapy and a more severe course.e29 Of interest, a patient with MS on fingolimod who developed a severe COVID-19 infection benefitted from short-term interleukin 6 receptor blockage with tocilizumab.e30 Practitioners may consider a temporary delay of lymphocyte-depleting therapies in patients with MS.e31 Clearly, management of inflammatory demyelinating CNS diseases with immunomodulatory and immunosuppressive treatments needs to take into account the potential impact of COVID-19, and it will be important to explore responsiveness to SARS-CoV2 vaccines in these patients.e36,e37 The implications of the current pandemic on neurologic diseases have recently been discussed.e32,e33

Open issues and conclusion

Regarding MS, further studies are needed to better understand the mode of action and safety profile of (long-term) CD20 depletion. Publication of the 2 completed phase 3 trials of ofatumumab in MS may provide crucial evidence as it is still unknown whether a complete B-cell depletion is necessary for a therapeutic effect. Oral Bruton's tyrosine kinase inhibitors may be a promising therapy in the future, but phase 3 studies must prove a beneficial effect on clinical parameters. Today, it remains uncertain if CD19 depletion may be an option in patients with MS not responding to CD20 depletion.24,e34 Of interest, 1 report suggested that cladribine preferentially depletes B cells producing only a modest diminution of circulating T cells.e35

Regarding NMOSD, the CD19 cell–depleting antibody inebilizumab is the first B cell–directed treatment licensed. It enlarges the therapeutic arsenal for this disabling disease. Controlled trials are desirable in MOGAD. Clearly, for both conditions, development of evidence-based treatment algorithms is highly desirable.

In summary, much has been achieved in the treatment of MS, NMOSD, and MOGAD in recent years. The approval of ocrelizumab constituted another important step toward the effective treatment of patients with MS. Ofatumumab has just been licensed for the treatment of relapsing forms of MS in the United States and is expected to receive market authorization in the EU early 2021. Importantly, ocrelizumab is the first drug shown to be effective in the treatment of PPMS.

Rigorous pharmacovigilance, analysis of registry data, and the results of phase 3b and 4 trials and real-world data are indispensable. There is also a strong case for further mechanistic studies to clarify the effects of CD19 and CD20 B-cell depletion. Ublituximab and kinase inhibitors are promising new immunotherapies, which may open a path toward more individualized treatment of MS and related inflammatory demyelinating disorders of the CNS.

Study funding

No targeted funding reported.

Disclosure

J. Graf received conference and educational support from Biogen, Merck Serono, Sanofi Genzyme, and Grifols and a research fellowship from the Deutsche Forschungsgemeinschaft (project number 438899010). J. Mares reports no conflicts. M. Barnett has received institutional support for research, speaking, and/or participation in advisory boards for Biogen, Merck, Novartis, Roche, and Sanofi Genzyme; is a consulting neurologist for RxMx; and is Research Director for the Sydney Neuroimaging Analysis Centre. O. Aktas received, with approval of the Rector of Heinrich Heine University, grants from the German Research Foundation (DFG), the German Ministry for Education and Research (BMBF) as part of the German Competence Network Multiple Sclerosis (KKNMS; for NEMOS NationNMO-PAT FKZ 01GI1602B), and the Eugène Devic European Network (EU-FP7) and honoraria and travel/accommodation/meeting expenses from Almirall, Bayer, Biogen, MedImmune, Merck Serono, Novartis, Roche, Sanofi Genzyme, and Teva. P. Albrecht received, with approval of the Rector of Heinrich Heine University and the CEO of University of Düsseldorf Hospital, personal fees, research grants, and nonfinancial support from Allergan, Biogen, Celgene, Ipsen, Merck Serono, Merz Pharmaceuticals, Novartis, and Roche and personal fees and nonfinancial support from Bayer HealthCare and Sanofi-Aventis/Genzyme, outside the submitted work. S.S. Zamvil is Deputy Editor of Neurology: Neuroimmunology and Neuroinflammation and is an Associate Editor for Frontiers in Immunology and Frontiers in Neurology. He serves on the Advisory Committee for the American Congress on Treatment and Research in Multiple Sclerosis (ACTRIMS) and on the grant review committee for the National Multiple Sclerosis Society (NMSS). He has served as a consultant and received honoraria from Biogen Idec, EMD-Serono, Genzyme, Novartis, Roche/Genentech, and Teva Pharmaceuticals, Inc., and has served on Data Safety Monitoring Boards for Lilly, BioMS, Teva, and Opexa Therapeutics. He receives research grant support from the NIH, NMSS, Weill Institute, Race to Erase MS, and the Maisin Foundation. H.-P. Hartung received honoraria for serving on steering and data-monitoring committees, adboards, and speaking at scientific symposia from Bayer HealthCare, Biogen, Celgene BMS, GeNeuro, MedImmune, Merck, Novartis, Roche, Teva, TG Therapeutics, and Viela Bio with the approval of the Rector of Heinrich Heine University. Go to Neurology.org/NN for full disclosures.

Appendix Authors

Table
Table

Footnotes

  • Go to Neurology.org/NN for full disclosures. Funding information is provided at the end of the article.

  • The Article Processing Charge was funded by Heinrich-Heine-Universität.

  • See page e918

  • Received August 3, 2020.
  • Accepted in final form September 11, 2020.
  • Copyright © 2020 The Author(s). Published by Wolters Kluwer Health, Inc. on behalf of the American Academy of Neurology.

This is an open access article distributed under the terms of the Creative Commons Attribution-NonCommercial-NoDerivatives License 4.0 (CC BY-NC-ND), which permits downloading and sharing the work provided it is properly cited. The work cannot be changed in any way or used commercially without permission from the journal.

References

  1. 1.↵
    1. Fujihara K,
    2. Cook LJ
    . Neuromyelitis optica spectrum disorders and myelin oligodendrocyte glycoprotein antibody-associated disease: current topics. Curr Opin Neurol 2020;33:300–308.
    OpenUrl
  2. 2.↵
    1. Bennett JL,
    2. O'Connor KC,
    3. Bar-Or A, et al
    . B lymphocytes in neuromyelitis optica. Neurol Neuroimmunol Neuroinflamm 2015;2:e104. doi: 10.1212/NXI.0000000000000104.
    OpenUrlAbstract/FREE Full Text
  3. 3.↵
    1. Cotzomi E,
    2. Stathopoulos P,
    3. Lee CS, et al
    . Early B cell tolerance defects in neuromyelitis optica favour anti-AQP4 autoantibody production. Brain 2019;142:1598–1615.
    OpenUrl
  4. 4.↵
    1. Mader S,
    2. Kümpfel T,
    3. Meinl E
    . Novel insights into pathophysiology and therapeutic possibilities reveal further differences between AQP4-IgG- and MOG-IgG-associated diseases. Curr Opin Neurol 2020;33:362–371.
    OpenUrlPubMed
  5. 5.↵
    1. Damato V,
    2. Evoli A,
    3. Iorio R
    . Efficacy and safety of rituximab therapy in neuromyelitis optica spectrum disorders: a systematic review and meta-analysis. JAMA Neurol 2016;73:1342–1348.
    OpenUrl
  6. 6.↵
    1. Gao F,
    2. Chai B,
    3. Gu C, et al
    . Effectiveness of rituximab in neuromyelitis optica: a meta-analysis. BMC Neurol 2019;19:36.
    OpenUrl
  7. 7.↵
    1. Annovazzi P,
    2. Capobianco M,
    3. Moiola L, et al
    . Rituximab in the treatment of neuromyelitis optica: a multicentre Italian observational study. J Neurol 2016;263:1727–1735.
    OpenUrlPubMed
  8. 8.↵
    1. Trebst C,
    2. Jarius S,
    3. Berthele A, et al
    . Update on the diagnosis and treatment of neuromyelitis optica: recommendations of the Neuromyelitis Optica Study Group (NEMOS). J Neurol 2014;261:1–16.
    OpenUrlCrossRefPubMed
  9. 9.↵
    1. Cabre P,
    2. Mejdoubi M,
    3. Jeannin S, et al
    . Treatment of neuromyelitis optica with rituximab: a 2-year prospective multicenter study. J Neurol 2018;265:917–925.
    OpenUrl
  10. 10.↵
    1. Radaelli M,
    2. Moiola L,
    3. Sangalli F, et al
    . Neuromyelitis optica spectrum disorders: long-term safety and efficacy of rituximab in Caucasian patients. Mult Scler 2016;22:511–519.
    OpenUrlCrossRefPubMed
  11. 11.↵
    1. Chamberlain N,
    2. Massad C,
    3. Oe T,
    4. Cantaert T,
    5. Herold KC,
    6. Meffre E
    . Rituximab does not reset defective early B cell tolerance checkpoints. J Clin Invest 2016;126:282–287.
    OpenUrlCrossRefPubMed
  12. 12.↵
    1. Kim SH,
    2. Huh SY,
    3. Lee SJ,
    4. Joung A,
    5. Kim HJ
    . A 5-year follow-up of rituximab treatment in patients with neuromyelitis optica spectrum disorder. JAMA Neurol 2013;70:1110–1117.
    OpenUrl
  13. 13.↵
    1. Kim SH,
    2. Hyun JW,
    3. Kim HJ
    . Individualized B cell-targeting therapy for neuromyelitis optica spectrum disorder. Neurochem Int 2019;130:104347.
    OpenUrl
  14. 14.↵
    1. Cohen M,
    2. Romero G,
    3. Bas J, et al
    . Monitoring CD27+ memory B-cells in neuromyelitis optica spectrum disorders patients treated with rituximab: results from a bicentric study. J Neurol Sci 2017;373:335–338.
    OpenUrl
  15. 15.↵
    1. Evangelopoulos ME,
    2. Andreadou E,
    3. Koutsis G, et al
    . Treatment of neuromyelitis optica and neuromyelitis optica spectrum disorders with rituximab using a maintenance treatment regimen and close CD19 B cell monitoring. A six-year follow-up. J Neurol Sci 2017;372:92–96.
    OpenUrl
  16. 16.↵
    1. Novi G,
    2. Bovis F,
    3. Capobianco M, et al
    . Efficacy of different rituximab therapeutic strategies in patients with neuromyelitis optica spectrum disorders. Mult Scler Relat Disord 2019;36:101430.
    OpenUrl
  17. 17.↵
    1. Paolilo RB,
    2. Hacohen Y,
    3. Yazbeck E, et al
    . Treatment and outcome of aquaporin-4 antibody-positive NMOSD: a multinational pediatric study. Neurol Neuroimmunol Neuroinflamm 2020;7:e387. doi: 10.1212/NXI.0000000000000104.
    OpenUrl
  18. 18.↵
    1. Tahara M,
    2. Oeda T,
    3. Okada K, et al
    . Safety and efficacy of rituximab in neuromyelitis optica spectrum disorders (RIN-1 study): a multicentre, randomised, double-blind, placebo-controlled trial. Lancet Neurol 2020;19:298–306.
    OpenUrlPubMed
  19. 19.↵
    1. Hartung HP,
    2. Aktas O
    . Old and new breakthroughs in neuromyelitis optica. Lancet Neurol 2020;19:280–281.
    OpenUrl
  20. 20.↵
    1. Trewin BP,
    2. Adelstein S,
    3. Spies JM, et al
    . Precision therapy for neuromyelitis optica spectrum disorder: a retrospective analysis of the use of class-switched memory B-cells for individualised rituximab dosing schedules. Mult Scler Relat Disord 2020;43:102175.
    OpenUrl
  21. 21.↵
    1. Herbst R,
    2. Wang Y,
    3. Gallagher S, et al
    . B-cell depletion in vitro and in vivo with an afucosylated anti-CD19 antibody. J Pharmacol Exp Ther 2010;335:213–222.
    OpenUrlAbstract/FREE Full Text
  22. 22.↵
    1. Ward E,
    2. Mittereder N,
    3. Kuta E, et al
    . A glycoengineered anti-CD19 antibody with potent antibody-dependent cellular cytotoxicity activity in vitro and lymphoma growth inhibition in vivo. Br J Haematol 2011;155:426–437.
    OpenUrlCrossRefPubMed
  23. 23.↵
    1. Chen D,
    2. Gallagher S,
    3. Monson NL,
    4. Herbst R,
    5. Wang Y
    . Inebilizumab, a B Cell-Depleting anti-CD19 antibody for the treatment of autoimmune neurological diseases: insights from preclinical studies. J Clin Med 2016;5:107.
    OpenUrl
  24. 24.↵
    1. Agius MA,
    2. Klodowska-Duda G,
    3. Maciejowski M, et al
    . Safety and tolerability of inebilizumab (MEDI-551), an anti-CD19 monoclonal antibody, in patients with relapsing forms of multiple sclerosis: results from a phase 1 randomised, placebo-controlled, escalating intravenous and subcutaneous dose study. Mult Scler 2019;25:235–245.
    OpenUrlCrossRef
  25. 25.↵
    1. Cree BAC,
    2. Bennett JL,
    3. Kim HJ, et al
    . Inebilizumab for the treatment of neuromyelitis optica spectrum disorder (N-MOmentum): a double-blind, randomised placebo-controlled phase 2/3 trial. Lancet 2019;394:1352–1363.
    OpenUrlCrossRefPubMed
  26. 26.↵
    1. Gallagher S,
    2. Turman S,
    3. Yusuf I, et al
    . Pharmacological profile of MEDI-551, a novel anti-CD19 antibody, in human CD19 transgenic mice. Int Immunopharmacol 2016;36:205–212.
    OpenUrl
  27. 27.↵
    1. Stüve O,
    2. Warnke C,
    3. Deason K, et al
    . CD19 as a molecular target in CNS autoimmunity. Acta Neuropathol 2014;128:177–190.
    OpenUrlCrossRefPubMed
  28. 28.↵
    1. Mealy MA,
    2. Levy M
    . A pilot safety study of ublituximab, a monoclonal antibody against CD20, in acute relapses of neuromyelitis optica spectrum disorder. Medicine (Baltimore) 2019;98:e15944.
    OpenUrl
  29. 29.↵
    1. Traboulsee A,
    2. Greenberg BM,
    3. Bennett JL, et al
    . Safety and efficacy of satralizumab monotherapy in neuromyelitis optica spectrum disorder: a randomised, double-blind, multicentre, placebo-controlled phase 3 trial. Lancet Neurol 2020;19:402–412.
    OpenUrl
  30. 30.↵
    1. Zhang C,
    2. Zhang M,
    3. Qiu W, et al
    . Safety and efficacy of tocilizumab versus azathioprine in highly relapsing neuromyelitis optica spectrum disorder (TANGO): an open-label, multicentre, randomised, phase 2 trial. Lancet Neurol 2020;19:391–401.
    OpenUrl
  31. 31.↵
    1. Bennett JL,
    2. Greenberg B,
    3. Traboulsee A, et al
    . Efficacy of satralizumab as monotherapy in pre-specified subgroups of sakurastar, a double-blind placebo-controlled phase 3 clinical study in patients with neuromyelitis optica spectrum disorder (NMOSD). Mult Scler Relat Disord 2020;37:101576.
    OpenUrl
  32. 32.↵
    1. Jarius S,
    2. Ruprecht K,
    3. Kleiter I, et al
    . MOG-IgG in NMO and related disorders: a multicenter study of 50 patients. Part 2: Epidemiology, clinical presentation, radiological and laboratory features, treatment responses, and long-term outcome. J Neuroinflammation 2016;13:280.
    OpenUrlCrossRefPubMed
  33. 33.↵
    1. Papathanasiou A,
    2. Tanasescu R,
    3. Davis J, et al
    . MOG-IgG-associated demyelination: focus on atypical features, brain histopathology and concomitant autoimmunity. J Neurol 2020;267:359–368.
    OpenUrl
  34. 34.↵
    1. Höftberger R,
    2. Guo Y,
    3. Flanagan EP, et al
    . The pathology of central nervous system inflammatory demyelinating disease accompanying myelin oligodendrocyte glycoprotein autoantibody. Acta Neuropathol 2020;139:875–892.
    OpenUrlPubMed
  35. 35.↵
    1. Jurynczyk M,
    2. Messina S,
    3. Woodhall MR, et al
    . Clinical presentation and prognosis in MOG-antibody disease: a UK study. Brain 2017;140:3128–3138.
    OpenUrlPubMed
  36. 36.↵
    1. Takai Y,
    2. Misu T,
    3. Kaneko K, et al
    . Myelin oligodendrocyte glycoprotein antibody-associated disease: an immunopathological study. Brain 2020;143:1431–1446.
    OpenUrlPubMed
  37. 37.↵
    1. Ciotti JR,
    2. Eby NS,
    3. Wu GF,
    4. Naismith RT,
    5. Chahin S,
    6. Cross AH
    . Clinical and laboratory features distinguishing MOG antibody disease from multiple sclerosis and AQP4 antibody-positive neuromyelitis optica. Mult Scler Relat Disord 2020;45:102399.
    OpenUrl
  38. 38.↵
    1. Spadaro M,
    2. Winklmeier S,
    3. Beltrán E, et al
    . Pathogenicity of human antibodies against myelin oligodendrocyte glycoprotein. Ann Neurol 2018;84:315–328.
    OpenUrlPubMed
  39. 39.↵
    1. Reindl M,
    2. Waters P
    . Myelin oligodendrocyte glycoprotein antibodies in neurological disease. Nat Rev Neurol 2019;15:89–102.
    OpenUrlPubMed
  40. 40.↵
    1. Peschl P,
    2. Bradl M,
    3. Höftberger R,
    4. Berger T,
    5. Reindl M
    . Myelin oligodendrocyte glycoprotein: deciphering a target in inflammatory demyelinating diseases. Front Immunol 2017;8:529.
    OpenUrlCrossRefPubMed
  41. 41.↵
    1. Winklmeier S,
    2. Schlüter M,
    3. Spadaro M, et al
    . Identification of circulating MOG-specific B cells in patients with MOG antibodies. Neurol Neuroimmunol Neuroinflamm 2019;6:625. doi: 10.1212/NXI.0000000000000104.
    OpenUrlAbstract/FREE Full Text
  42. 42.↵
    1. Li X,
    2. Wang L,
    3. Zhou L, et al
    . The imbalance between regulatory and memory B cells accompanied by an increased number of circulating T-follicular helper cells in MOG-antibody-associated demyelination. Mult Scler Relat Disord 2019;36:101397.
    OpenUrl
  43. 43.↵
    1. Hacohen Y,
    2. Wong YY,
    3. Lechner C, et al
    . Disease course and treatment responses in children with relapsing myelin oligodendrocyte glycoprotein antibody-associated disease. JAMA Neurol 2018;75:478–487.
    OpenUrl
  44. 44.↵
    1. Durozard P,
    2. Rico A,
    3. Boutiere C, et al
    . Comparison of the response to rituximab between myelin oligodendrocyte glycoprotein and aquaporin-4 antibody diseases. Ann Neurol 2020;87:256–266.
    OpenUrl
  45. 45.↵
    1. Whittam DH,
    2. Cobo-Calvo A,
    3. Lopez-Chiriboga AS, et al
    . Treatment of MOG-IgG-associated disorder with rituximab: an international study of 121 patients. Mult Scler Relat Disord 2020;44:102251.
    OpenUrlPubMed
  46. 46.↵
    1. Winkelmann A,
    2. Loebermann M,
    3. Reisinger EC,
    4. Hartung HP,
    5. Zettl UK
    . Disease-modifying therapies and infectious risks in multiple sclerosis. Nat Rev Neurol 2016;12:217–233.
    OpenUrlCrossRefPubMed
  47. 47.↵
    1. Salzer J,
    2. Svenningsson R,
    3. Alping P, et al
    . Rituximab in multiple sclerosis: a retrospective observational study on safety and efficacy. Neurology 2016;87:2074–2081.
    OpenUrl
  48. 48.↵
    1. Vollmer BL,
    2. Wallach AI,
    3. Corboy JR,
    4. Dubovskaya K,
    5. Alvarez E,
    6. Kister I
    . Serious safety events in rituximab-treated multiple sclerosis and related disorders. Ann Clin Transl Neurol 2020;7:1477–1487.
    OpenUrl
  49. 49.↵
    1. Luna G,
    2. Alping P,
    3. Burman J, et al
    . Infection risks among patients with multiple sclerosis treated with fingolimod, natalizumab, rituximab, and injectable therapies. JAMA Neurol 2020;77:184–191.
    OpenUrl
  50. 50.↵
    1. Airas L,
    2. Nylund M,
    3. Mannonen I,
    4. Matilainen M,
    5. Sucksdorff M,
    6. Rissanen E
    . Rituximab in the treatment of multiple sclerosis in the hospital district of southwest Finland. Mult Scler Relat Disord 2020;40:101980.
    OpenUrl
  51. 51.↵
    1. Caldito NG,
    2. Shirani A,
    3. Salter A,
    4. Stuve O
    . Adverse event profile differences between rituximab and ocrelizumab: findings from the FDA adverse event reporting database. Mult Scler 2020. doi: 10.1177/1352458520949986.
  52. 52.↵
    1. Alping P,
    2. Askling J,
    3. Burman J, et al
    . Cancer risk for fingolimod, natalizumab, and rituximab in multiple sclerosis patients. Ann Neurol 2020;87:688–699.
    OpenUrl
  53. 53.↵
    1. van Vollenhoven RF,
    2. Fleischmann RM,
    3. Furst DE,
    4. Lacey S,
    5. Lehane PB
    . Longterm safety of rituximab: final report of the rheumatoid arthritis global clinical trial program over 11 years. J Rheumatol 2015;42:1761–1766.
    OpenUrlAbstract/FREE Full Text
  54. 54.↵
    1. Ng HS,
    2. Rosenbult CL,
    3. Tremlett H
    . Safety profile of ocrelizumab for the treatment of multiple sclerosis: a systematic review. Expert Opin Drug Saf 2020;19:1069–1094.
    OpenUrl
  55. 55.↵
    1. Hauser SL,
    2. Kappos L,
    3. Arnold DL, et al
    . Five-years of ocrelizumab in relapsing multiple sclerosis: OPERA studies open-label extension. Neurology 2020;95:e1854–e1867.
    OpenUrl
  56. 56.↵
    1. Hauser SL,
    2. Kappos L,
    3. Montalban X, et al
    . Safety of ocrelizumab in multiple sclerosis: updated analysis in patients with relapsing and primary progressive multiple sclerosis. Presented at the 70th American Academy of Neurology (AAN) Annual Meeting; April 21–27, 2018; Los Angeles, CA, USA; Platform presentation number S36.001 [online]. Available at: ocreposters.com/uploads/AAN%202018%20General%20Safety_Platform_Global_Hauser%20et%20al_Updated.pdf. Accessed June 15, 2020.
  57. 57.↵
    1. Montalban X,
    2. Hauser SL,
    3. Kappos L, et al
    . Ocrelizumab versus placebo in primary progressive multiple sclerosis. N Engl J Med 2017;376:209–220.
    OpenUrlCrossRefPubMed
  58. 58.↵
    1. Hauser SL,
    2. Kappos L,
    3. Montalban X, et al
    . Safety of ocrelizumab in multiple sclerosis: updated analysis in patients with relapsing and primary progressive multiple sclerosis. Presented at the 6th Congress of the European Academy of Neurology (EAN) [online]. Available at: medically.roche.com/en/search/pdfviewer.ed54f34c-1b9b-461a-82ed-dac932793643.html?cid=slpsxx2005nexxean2020. Accessed June 15, 2020.
  59. 59.↵
    1. Ciardi MR,
    2. Iannetta M,
    3. Zingaropoli MA, et al
    . Reactivation of hepatitis B virus with immune-escape mutations after ocrelizumab treatment for multiple sclerosis. Open Forum Infect Dis 2019;6:ofy356.
    OpenUrl
  60. 60.↵
    1. Nicolini LA,
    2. Canepa P,
    3. Caligiuri P, et al
    . Fulminant hepatitis associated with echovirus 25 during treatment with ocrelizumab for multiple sclerosis. JAMA Neurol 2019;76:866–867.
    OpenUrl
  61. Additional e-references (e1-e37) available at: links.lww.com/NXI/A347.

Letters: Rapid online correspondence

No comments have been published for this article.
Comment

REQUIREMENTS

If you are uploading a letter concerning an article:
You must have updated your disclosures within six months: http://submit.neurology.org

Your co-authors must send a completed Publishing Agreement Form to Neurology Staff (not necessary for the lead/corresponding author as the form below will suffice) before you upload your comment.

If you are responding to a comment that was written about an article you originally authored:
You (and co-authors) do not need to fill out forms or check disclosures as author forms are still valid
and apply to letter.

Submission specifications:

  • Submissions must be < 200 words with < 5 references. Reference 1 must be the article on which you are commenting.
  • Submissions should not have more than 5 authors. (Exception: original author replies can include all original authors of the article)
  • Submit only on articles published within 6 months of issue date.
  • Do not be redundant. Read any comments already posted on the article prior to submission.
  • Submitted comments are subject to editing and editor review prior to posting.

More guidelines and information on Disputes & Debates

Compose Comment

More information about text formats

Plain text

  • No HTML tags allowed.
  • Web page addresses and e-mail addresses turn into links automatically.
  • Lines and paragraphs break automatically.
Author Information
NOTE: The first author must also be the corresponding author of the comment.
First or given name, e.g. 'Peter'.
Your last, or family, name, e.g. 'MacMoody'.
Your email address, e.g. higgs-boson@gmail.com
Your role and/or occupation, e.g. 'Orthopedic Surgeon'.
Your organization or institution (if applicable), e.g. 'Royal Free Hospital'.
Publishing Agreement
NOTE: All authors, besides the first/corresponding author, must complete a separate Publishing Agreement Form and provide via email to the editorial office before comments can be posted.
CAPTCHA
This question is for testing whether or not you are a human visitor and to prevent automated spam submissions.

Vertical Tabs

You May Also be Interested in

Back to top
  • Article
    • Abstract
    • Glossary
    • NMOSD
    • MOGAD
    • CD20/CD19 B-cell depletion in MS, NMOSD, and MOGAD: adverse events and safety profiles
    • Implications of the COVID-19/SARS-CoV2 pandemic
    • Open issues and conclusion
    • Study funding
    • Disclosure
    • Appendix Authors
    • Footnotes
    • References
  • Figures & Data
  • Info & Disclosures
Advertisement

Use of Whole-Genome Sequencing for Mitochondrial Disease Diagnosis

Dr. Robert Pitceathly and Dr. William Macken

► Watch

Related Articles

  • Targeting B Cells to Modify MS, NMOSD, and MOGADPart 1

Topics Discussed

  • HIV

Alert Me

  • Alert me when eletters are published

Recommended articles

  • Views & Reviews
    Targeting B Cells to Modify MS, NMOSD, and MOGAD
    Part 1
    Jonas Graf, Jan Mares, Michael Barnett et al.
    Neurology: Neuroimmunology & Neuroinflammation, December 16, 2020
  • Article
    Ocrelizumab Extended Interval Dosing in Multiple Sclerosis in Times of COVID-19
    Leoni Rolfes, Marc Pawlitzki, Steffen Pfeuffer et al.
    Neurology: Neuroimmunology & Neuroinflammation, July 14, 2021
  • Views & Reviews
    Unraveling B lymphocytes in CNS inflammatory diseases
    Distinct mechanisms and treatment targets
    Bruna Klein da Costa, Renata Brant de Souza Melo, Giordani Rodrigues dos Passos et al.
    Neurology, September 09, 2020
  • Views & Reviews
    B-cell–targeted therapies in relapsing forms of MS
    Divyanshu Dubey, Thomas Forsthuber, Eoin P. Flanagan et al.
    Neurology: Neuroimmunology & Neuroinflammation, October 23, 2017
Neurology - Neuroimmunology Neuroinflammation: 10 (3)

Articles

  • Articles
  • Issues
  • Popular Articles

About

  • About the Journals
  • Ethics Policies
  • Editors & Editorial Board
  • Contact Us
  • Advertise

Submit

  • Author Center
  • Submit a Manuscript
  • Information for Reviewers
  • AAN Guidelines
  • Permissions

Subscribers

  • Subscribe
  • Sign up for eAlerts
  • RSS Feed
Site Logo
  • Visit neurology Template on Facebook
  • Follow neurology Template on Twitter
  • Visit Neurology on YouTube
  • Neurology
  • Neurology: Clinical Practice
  • Neurology: Education
  • Neurology: Genetics
  • Neurology: Neuroimmunology & Neuroinflammation
  • AAN.com
  • AANnews
  • Continuum
  • Brain & Life
  • Neurology Today

Wolters Kluwer Logo

Neurology: Neuroimmunology & Neuroinflammation
Online ISSN: 2332-7812

© 2023 American Academy of Neurology

  • Privacy Policy
  • Feedback
  • Advertise