1
|
Wang AA, Luessi F, Neziraj T, Pössnecker E, Zuo M, Engel S, Hanuscheck N, Florescu A, Bugbee E, Ma XI, Rana F, Lee D, Ward LA, Kuhle J, Himbert J, Schraad M, van Puijenbroek E, Klein C, Urich E, Ramaglia V, Pröbstel AK, Zipp F, Gommerman JL. B cell depletion with anti-CD20 promotes neuroprotection in a BAFF-dependent manner in mice and humans. Sci Transl Med 2024; 16:eadi0295. [PMID: 38446903 DOI: 10.1126/scitranslmed.adi0295] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2023] [Accepted: 02/13/2024] [Indexed: 03/08/2024]
Abstract
Anti-CD20 therapy to deplete B cells is highly efficacious in preventing new white matter lesions in patients with relapsing-remitting multiple sclerosis (RRMS), but its protective capacity against gray matter injury and axonal damage is unclear. In a passive experimental autoimmune encephalomyelitis (EAE) model whereby TH17 cells promote brain leptomeningeal immune cell aggregates, we found that anti-CD20 treatment effectively spared myelin content and prevented myeloid cell activation, oxidative damage, and mitochondrial stress in the subpial gray matter. Anti-CD20 treatment increased B cell survival factor (BAFF) in the serum, cerebrospinal fluid, and leptomeninges of mice with EAE. Although anti-CD20 prevented gray matter demyelination, axonal loss, and neuronal atrophy, co-treatment with anti-BAFF abrogated these benefits. Consistent with the murine studies, we observed that elevated BAFF concentrations after anti-CD20 treatment in patients with RRMS were associated with better clinical outcomes. Moreover, BAFF promoted survival of human neurons in vitro. Together, our data demonstrate that BAFF exerts beneficial functions in MS and EAE in the context of anti-CD20 treatment.
Collapse
Affiliation(s)
- Angela A Wang
- Department of Immunology, University of Toronto, Toronto, M5S 1A8, Canada
| | - Felix Luessi
- Department of Neurology, Focus Program Translational Neuroscience (FTN) and Immunotherapy (FZI), Rhine Main Neuroscience Network (rmn2), University Medical Center of the Johannes Gutenberg University Mainz, 55131 Mainz, Germany
| | - Tradite Neziraj
- Department of Neurology, University Hospital of Basel and University of Basel, 4031 Basel, Switzerland
- Departments of Biomedicine and Clinical Research, University Hospital of Basel and University of Basel, 4031 Basel, Switzerland
- Research Center for Clinical Neuroimmunology and Neuroscience Basel (RC2NB), University Hospital of Basel and University of Basel, 4031 Basel, Switzerland
| | - Elisabeth Pössnecker
- Department of Neurology, University Hospital of Basel and University of Basel, 4031 Basel, Switzerland
- Departments of Biomedicine and Clinical Research, University Hospital of Basel and University of Basel, 4031 Basel, Switzerland
- Research Center for Clinical Neuroimmunology and Neuroscience Basel (RC2NB), University Hospital of Basel and University of Basel, 4031 Basel, Switzerland
| | - Michelle Zuo
- Department of Immunology, University of Toronto, Toronto, M5S 1A8, Canada
| | - Sinah Engel
- Department of Neurology, Focus Program Translational Neuroscience (FTN) and Immunotherapy (FZI), Rhine Main Neuroscience Network (rmn2), University Medical Center of the Johannes Gutenberg University Mainz, 55131 Mainz, Germany
| | - Nicholas Hanuscheck
- Department of Neurology, Focus Program Translational Neuroscience (FTN) and Immunotherapy (FZI), Rhine Main Neuroscience Network (rmn2), University Medical Center of the Johannes Gutenberg University Mainz, 55131 Mainz, Germany
| | - Alexandra Florescu
- Department of Immunology, University of Toronto, Toronto, M5S 1A8, Canada
| | - Eryn Bugbee
- Department of Immunology, University of Toronto, Toronto, M5S 1A8, Canada
| | - Xianjie I Ma
- Department of Immunology, University of Toronto, Toronto, M5S 1A8, Canada
| | - Fatima Rana
- Department of Immunology, University of Toronto, Toronto, M5S 1A8, Canada
| | - Dennis Lee
- Department of Immunology, University of Toronto, Toronto, M5S 1A8, Canada
| | - Lesley A Ward
- Department of Immunology, University of Toronto, Toronto, M5S 1A8, Canada
| | - Jens Kuhle
- Department of Neurology, University Hospital of Basel and University of Basel, 4031 Basel, Switzerland
- Departments of Biomedicine and Clinical Research, University Hospital of Basel and University of Basel, 4031 Basel, Switzerland
- Research Center for Clinical Neuroimmunology and Neuroscience Basel (RC2NB), University Hospital of Basel and University of Basel, 4031 Basel, Switzerland
| | - Johannes Himbert
- Department of Neurology, Focus Program Translational Neuroscience (FTN) and Immunotherapy (FZI), Rhine Main Neuroscience Network (rmn2), University Medical Center of the Johannes Gutenberg University Mainz, 55131 Mainz, Germany
| | - Muriel Schraad
- Department of Neurology, Focus Program Translational Neuroscience (FTN) and Immunotherapy (FZI), Rhine Main Neuroscience Network (rmn2), University Medical Center of the Johannes Gutenberg University Mainz, 55131 Mainz, Germany
| | | | - Christian Klein
- Roche Innovation Center Zurich, Roche Glycart AG, 8952 Schlieren, Switzerland
| | - Eduard Urich
- Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., 4052 Basel, Switzerland
| | - Valeria Ramaglia
- Department of Immunology, University of Toronto, Toronto, M5S 1A8, Canada
| | - Anne-Katrin Pröbstel
- Department of Neurology, University Hospital of Basel and University of Basel, 4031 Basel, Switzerland
- Departments of Biomedicine and Clinical Research, University Hospital of Basel and University of Basel, 4031 Basel, Switzerland
- Research Center for Clinical Neuroimmunology and Neuroscience Basel (RC2NB), University Hospital of Basel and University of Basel, 4031 Basel, Switzerland
| | - Frauke Zipp
- Department of Neurology, Focus Program Translational Neuroscience (FTN) and Immunotherapy (FZI), Rhine Main Neuroscience Network (rmn2), University Medical Center of the Johannes Gutenberg University Mainz, 55131 Mainz, Germany
| | | |
Collapse
|
2
|
Kiyat P, Karti O, Gercik Ö, Şak T. Choroidal, retinal, and optic nerve changes in rheumatoid arthritis and primary sjogren's syndrome patients: comparıson with each other and healthy subjects. Int Ophthalmol 2024; 44:24. [PMID: 38324105 DOI: 10.1007/s10792-024-02970-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Accepted: 12/04/2023] [Indexed: 02/08/2024]
Abstract
PURPOSE The present study aims to evaluate the optic nerve, macula, and choroidal changes in both rheumatoid arthritis (RA) and primary Sjögren's syndrome (SjS) patients, and to compare these findings with age-matched healthy volunteers. METHODS This study included 46 RA patients, 33 primary SjS patients, and 37 age-matched healthy volunteers. All of the patients underwent a thorough ophthalmological examination, during which measurements of the retinal nerve fiber layer (RNFL), ganglion cell layer(GCL), and subfoveal choroidal thickness (CT) were taken using OCT (optical coherence tomography). The measurements taken from the right eye of each patient were used to compare among the groups. RESULTS RNFL thickness in superior quadrant was found to be statistically significantly thinner in the eyes with RA when compared to the control group (p = 0.022). In the nasal quadrant, the RNFL thickness was significantly thinner in patients with primary SjS compared to healthy individuals (p = 0.036). Also, the temporal quadrant RNFL was significantly thinner in RA patients than in the primary SjS patients (p = 0.033). GCL thickness was observed to be thinner in all quadrants of both RA and primary SjS groups compared to the control group. However, the difference was not found to be statistically significant. Subfoveal CT was observed to be thicker in RA and SjS groups compared to the control group, but this difference was also not statistically significant. CONCLUSION Systemic autoimmune diseases like RA and primary SjS can lead to a decrease in RNLF and GCL thickness, which can impair visual acuity even in the absence of ocular symptoms. Therefore, monitoring changes in the optic nerve, retina, and choroid layer are crucial in these patients.
Collapse
Affiliation(s)
- Pelin Kiyat
- Department of Ophthalmology, İzmir Democracy University, Buca Seyfi Demirsoy Training and Research Hospital, Kozağaç Mah., Özmen Sok., No:147, Buca, İzmir, Turkey.
| | - Omer Karti
- Department of Ophthalmology, İzmir Democracy University, Buca Seyfi Demirsoy Training and Research Hospital, Kozağaç Mah., Özmen Sok., No:147, Buca, İzmir, Turkey
| | - Önay Gercik
- Department of Rheumatology, İzmir Democracy University, Buca Seyfi Demirsoy Training and Research Hospital, İzmir, Turkey
| | - Tuncer Şak
- Department of Rheumatology, İzmir Democracy University, Buca Seyfi Demirsoy Training and Research Hospital, İzmir, Turkey
| |
Collapse
|
3
|
Kim W, Kim HJ. An update on biologic treatments for neuromyelitis optica spectrum disorder. Expert Rev Clin Immunol 2023; 19:111-121. [PMID: 36414430 DOI: 10.1080/1744666x.2023.2151441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
INTRODUCTION Neuromyelitis optica spectrum disorder (NMOSD) is an autoimmune disease of the central nervous system mediated by antibodies targeting the aquaporin-4 (AQP4) water channel expressed on astrocytes. The binding of specific antibodies to AQP4 causes complement-dependent cytotoxicity, leading to inflammation and demyelination. Several recent phase 2 and 3 randomized placebo-controlled trials showed the efficacy and safety of monoclonal antibody therapies targeting B-cells, interleukin-6 receptor, and complement. AREAS COVERED Current biologic treatments for NMOSD and developments therein, and unresolved issues in NMOSD treatment. EXPERT OPINION New biologic treatments demonstrate high efficacy and good safety for patients with AQP4-IgG-positive NMOSD. The optimal therapeutics for seronegative NMOSD, pediatric patients, and female patients who are pregnant or wish to be are unclear, and further research is needed. Also, real-world studies of new biological agents and the data on the durability of their beneficial effects and their long-term safety are required. Effective rescue therapy for an acute attack is critical given permanent disability in NMOSD is attack-related, and biologic agents that treat acute attack are emerging. If such treatments are to become widely applied, studies on the most cost-effective treatment strategies are needed.
Collapse
Affiliation(s)
- Woojun Kim
- Department of Neurology, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Ho Jin Kim
- Department of Neurology, Research Institute and Hospital of National Cancer Center, Goyang, Korea
| |
Collapse
|
4
|
Pique J, Nicolas P, Marignier R. Neuromielite ottica acuta (malattia di Devic). Neurologia 2022. [DOI: 10.1016/s1634-7072(22)47095-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022] Open
|
5
|
Zhang C, Zhang TX, Liu Y, Jia D, Zeng P, Du C, Yuan M, Liu Q, Wang Y, Shi FD. B-Cell Compartmental Features and Molecular Basis for Therapy in Autoimmune Disease. NEUROLOGY-NEUROIMMUNOLOGY & NEUROINFLAMMATION 2021; 8:8/6/e1070. [PMID: 34465614 PMCID: PMC8409132 DOI: 10.1212/nxi.0000000000001070] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Accepted: 07/21/2021] [Indexed: 11/16/2022]
Abstract
Background and Objectives To assess the molecular landscape of B-cell subpopulations across different compartments in patients with neuromyelitis optica spectrum disorder (NMOSD). Methods We performed B-cell transcriptomic profiles via single-cell RNA sequencing across CSF, blood, and bone marrow in patients with NMOSD. Results Across the tissue types tested, 4 major subpopulations of B cells with distinct signatures were identified: naive B cells, memory B cells, age-associated B cells, and antibody-secreting cells (ASCs). NMOSD B cells show proinflammatory activity and increased expression of chemokine receptor genes (CXCR3 and CXCR4). Circulating B cells display an increase of antigen presentation markers (CD40 and CD83), as well as activation signatures (FOS, CD69, and JUN). In contrast, the bone marrow B-cell population contains a large ASC fraction with increased oxidative and metabolic activity reflected by COX genes and ATP synthase genes. Typically, NMOSD B cells become hyperresponsive to type I interferon, which facilitates B-cell maturation and anti–aquaporin-4 autoantibody production. The pool of ASCs in blood and CSF were significantly elevated in NMOSD. Both CD19− and CD19+ ASCs could be ablated by tocilizumab, but not rituximab treatment in NMOSD. Discussion B cells are compartmentally fine tuned toward autoreactivity in NMOSD and become hyperreactive to type I interferon. Inhibition of type I interferon pathway may provide a new therapeutic avenue for NMOSD.
Collapse
Affiliation(s)
- Chao Zhang
- From the China National Clinical Research Center for Neurological Diseases (C.Z., Y.W., F.-D.S.), Beijing Tiantan Hospital, Capital Medical University; and Department of Neurology (C.Z., T.-X.Z., Y.L., D.J., P.Z., C.D., M.Y., Q.L., F.-D.S.), Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin Medical University, China
| | - Tian-Xiang Zhang
- From the China National Clinical Research Center for Neurological Diseases (C.Z., Y.W., F.-D.S.), Beijing Tiantan Hospital, Capital Medical University; and Department of Neurology (C.Z., T.-X.Z., Y.L., D.J., P.Z., C.D., M.Y., Q.L., F.-D.S.), Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin Medical University, China
| | - Ye Liu
- From the China National Clinical Research Center for Neurological Diseases (C.Z., Y.W., F.-D.S.), Beijing Tiantan Hospital, Capital Medical University; and Department of Neurology (C.Z., T.-X.Z., Y.L., D.J., P.Z., C.D., M.Y., Q.L., F.-D.S.), Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin Medical University, China
| | - Dongmei Jia
- From the China National Clinical Research Center for Neurological Diseases (C.Z., Y.W., F.-D.S.), Beijing Tiantan Hospital, Capital Medical University; and Department of Neurology (C.Z., T.-X.Z., Y.L., D.J., P.Z., C.D., M.Y., Q.L., F.-D.S.), Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin Medical University, China
| | - Pei Zeng
- From the China National Clinical Research Center for Neurological Diseases (C.Z., Y.W., F.-D.S.), Beijing Tiantan Hospital, Capital Medical University; and Department of Neurology (C.Z., T.-X.Z., Y.L., D.J., P.Z., C.D., M.Y., Q.L., F.-D.S.), Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin Medical University, China
| | - Chen Du
- From the China National Clinical Research Center for Neurological Diseases (C.Z., Y.W., F.-D.S.), Beijing Tiantan Hospital, Capital Medical University; and Department of Neurology (C.Z., T.-X.Z., Y.L., D.J., P.Z., C.D., M.Y., Q.L., F.-D.S.), Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin Medical University, China.
| | - Meng Yuan
- From the China National Clinical Research Center for Neurological Diseases (C.Z., Y.W., F.-D.S.), Beijing Tiantan Hospital, Capital Medical University; and Department of Neurology (C.Z., T.-X.Z., Y.L., D.J., P.Z., C.D., M.Y., Q.L., F.-D.S.), Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin Medical University, China
| | - Qiang Liu
- From the China National Clinical Research Center for Neurological Diseases (C.Z., Y.W., F.-D.S.), Beijing Tiantan Hospital, Capital Medical University; and Department of Neurology (C.Z., T.-X.Z., Y.L., D.J., P.Z., C.D., M.Y., Q.L., F.-D.S.), Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin Medical University, China
| | - Yongjun Wang
- From the China National Clinical Research Center for Neurological Diseases (C.Z., Y.W., F.-D.S.), Beijing Tiantan Hospital, Capital Medical University; and Department of Neurology (C.Z., T.-X.Z., Y.L., D.J., P.Z., C.D., M.Y., Q.L., F.-D.S.), Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin Medical University, China
| | - Fu-Dong Shi
- From the China National Clinical Research Center for Neurological Diseases (C.Z., Y.W., F.-D.S.), Beijing Tiantan Hospital, Capital Medical University; and Department of Neurology (C.Z., T.-X.Z., Y.L., D.J., P.Z., C.D., M.Y., Q.L., F.-D.S.), Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin Medical University, China.
| |
Collapse
|
6
|
Shi B, Zhao M, Qiao L, Huang F, Zhou S, Wei Y, Wang J, Wang N. Relapses shortly after rituximab treatment in neuromyelitis optica spectrum disorder. Mult Scler Relat Disord 2021; 54:103143. [PMID: 34273608 DOI: 10.1016/j.msard.2021.103143] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2021] [Revised: 06/12/2021] [Accepted: 07/05/2021] [Indexed: 10/20/2022]
Abstract
OBJECTIVE Rituximab (RTX), an anti-CD20 monoclonal antibody, has been demonstrated to be a useful maintenance therapy for neuromyelitis optica spectrum disorder (NMOSD). However, few patients may suffer from relapses shortly after RTX. In order to investigate the clinical features of RTX-related relapses and guide therapeutic strategy, 3 patients in our department were reported and literatures were reviewed. METHODS We reported three NMOSD patients suffered from relapses shortly after rituximab treatment in our hospital and reviewed 13 patients reported in literatures. Their demographic characteristics, clinical features and therapeutic strategy were retrospectively analyzed. RESULTS Sixteen patients, including three cases reported in this study, experienced 21 attacks within 1 month after RTX infusion. All of them were women with an age at onset of 34.0 ± 15.0 years. Fourteen patients were seropositive for aquaporin-4 antibody, and one was seropositive for myelin oligodendrocyte glycoprotein antibody. 57.1% (12/21) of RTX-related relapses occurred after the first use of RTX. Their clinical manifestations included optic neuritis (8/21), myelitis (11/21), and the other two relapses without detailed descriptions. Also, 62.5% (10/16) of patients had a history of prior relapses within 3 months before RTX infusions, and the location of nine relapses overlapped with previous relapses. RTX was given again after the first RTX-related relapse in eight patients, three of them with low-dosage RTX stayed stable for years, and five patients with full-dosage RTX experienced another RTX-related relapse. CONCLUSIONS Relapses may occur shortly after RTX treatment in NMOSD. RTX-related relapse did not necessarily mean that RTX was ineffective in low-dosage regimen. Timely and sufficient treatment of RTX is crucial to prevent a relapse. It may be more reasonable to monitor B cell repopulation so as to determine a re-treatment regimen. RTX-related relapse following full-dosage RTX may be a predictor for a second time RTX-related relapse and it may be reasonable to switch to other immunosuppressants in early stage.
Collapse
Affiliation(s)
- Bingxin Shi
- Department of Neurology, Yuquan Hospital, School of Clinical Medicine, Tsinghua University, No.5 Shijingshan Road, Beijing 100049, PR China
| | - Mangsuo Zhao
- Department of Neurology, Yuquan Hospital, School of Clinical Medicine, Tsinghua University, No.5 Shijingshan Road, Beijing 100049, PR China.
| | - Liyan Qiao
- Department of Neurology, Yuquan Hospital, School of Clinical Medicine, Tsinghua University, No.5 Shijingshan Road, Beijing 100049, PR China.
| | - Fangjie Huang
- Department of Neurology, Yuquan Hospital, School of Clinical Medicine, Tsinghua University, No.5 Shijingshan Road, Beijing 100049, PR China
| | - Shimei Zhou
- Department of Neurology, Yuquan Hospital, School of Clinical Medicine, Tsinghua University, No.5 Shijingshan Road, Beijing 100049, PR China
| | - Yan Wei
- Department of Neurology, Yuquan Hospital, School of Clinical Medicine, Tsinghua University, No.5 Shijingshan Road, Beijing 100049, PR China
| | - Jing Wang
- Department of Neurology, Yuquan Hospital, School of Clinical Medicine, Tsinghua University, No.5 Shijingshan Road, Beijing 100049, PR China
| | - Ning Wang
- Department of Neurology, Yuquan Hospital, School of Clinical Medicine, Tsinghua University, No.5 Shijingshan Road, Beijing 100049, PR China
| |
Collapse
|
7
|
Cacciaguerra L, Tortorella P, Rocca MA, Filippi M. Targeting Neuromyelitis Optica Pathogenesis: Results from Randomized Controlled Trials of Biologics. Neurotherapeutics 2021; 18:1623-1636. [PMID: 33909234 PMCID: PMC8608970 DOI: 10.1007/s13311-021-01055-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/31/2021] [Indexed: 02/04/2023] Open
Abstract
Advances in neuromyelitis optica spectrum disorder pathogenesis have allowed the development of targeted drugs. These treatments act on core elements of the disease, including the pro-inflammatory IL-6 pathway (tocilizumab and satralizumab), B cells (rituximab and inebilizumab), and complement (eculizumab). According to recent phase II-III trials, biologics significantly reduced the risk of relapses in aquaporin-4-seropositive patients, whereas results were less striking in the small cohorts of aquaporin-4-seronegative patients. Most adverse events were mild to moderate, with systemic symptoms (headache, arthralgia) or infections (upper respiratory and urinary tracts) being most commonly reported.
Collapse
Affiliation(s)
- Laura Cacciaguerra
- Neuroimaging Research Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Neurology Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | | | - Maria A Rocca
- Neuroimaging Research Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Neurology Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | - Massimo Filippi
- Neuroimaging Research Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy.
- Neurology Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy.
- Vita-Salute San Raffaele University, Milan, Italy.
- Neurorehabilitation Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy.
- Neurophysiology Service, IRCCS San Raffaele Scientific Institute, Milan, Italy.
| |
Collapse
|
8
|
Gibril M, Walters R. Neuromyelitis optica spectrum disorder as a paraneoplastic syndrome: a rare and challenging diagnosis. BMJ Case Rep 2021; 14:e239389. [PMID: 33947671 PMCID: PMC8098970 DOI: 10.1136/bcr-2020-239389] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/15/2021] [Indexed: 01/17/2023] Open
Abstract
Neuromyelitis optica spectrum disorder (NMOSD) is an uncommon antibody-mediated disease of the central nervous system, often associated with aquaporin-4 antibodies (AQP4-Ab). NMOSD may present as a subacute myelopathy, progressing over days with MRI revealing a contiguous inflammatory lesion of the spinal cord, ≥3 vertebral segments, a longitudinally extensive transverse myelitis. We describe an unusual paraneoplastic form of AQP4-Ab NMOSD that developed in a patient with an advanced diffuse large B-cell lymphoma. The patient had an unusual hyperacute onset, reaching a clinical nadir within hours.
Collapse
|
9
|
Kang YR, Kim KH, Nam TS, Lee KH, Kang KW, Lee SJ, Choi SY, Chandrasekaran G, Kim MK. Fulminant Course of Neuromyelitis Optica in a Patient With Anti-MDA5 Antibody-Positive Dermatomyositis: A Case Report. Front Med (Lausanne) 2020; 7:576436. [PMID: 33262991 PMCID: PMC7686760 DOI: 10.3389/fmed.2020.576436] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Accepted: 10/07/2020] [Indexed: 11/21/2022] Open
Abstract
Anti-melanoma differentiation-associated gene 5 (anti-MDA5) antibody is a myositis-specific marker detected in clinically amyopathic dermatomyositis (DM). DM with anti-MDA5 antibody can be accompanied by rapidly progressive interstitial lung disease (RP-ILD) and other autoimmune disorders. Until now, only one case of neuromyelitis optica (NMO) with anti-MDA5-positive DM has been reported worldwide, in which the patient achieved a favorable outcome with intensive immunotherapy. We report a case of NMO in a patient with anti-MDA5-positive DM complicated by ILD and rheumatoid arthritis. Our patient experienced a fulminant course of NMO, rather than RP-ILD, in the presence of hyperferritinemia, which resulted in profound neurological sequelae despite immunotherapy including rituximab.
Collapse
Affiliation(s)
- You-Ri Kang
- Department of Neurology, Chonnam National University Medical School, Chonnam National University Hospital, Gwangju, South Korea
| | - Kun-Hee Kim
- Department of Biomedical Sciences, Chonnam National University Medical School, Gwangju, South Korea
| | - Tai-Seung Nam
- Department of Neurology, Chonnam National University Medical School, Chonnam National University Hospital, Gwangju, South Korea
| | - Kyung-Hwa Lee
- Department of Pathology, Chonnam National University Medical School, Gwangju, South Korea
| | - Kyung Wook Kang
- Department of Neurology, Chonnam National University Medical School, Chonnam National University Hospital, Gwangju, South Korea
| | - Seung-Jin Lee
- Department of Radiology, Chonnam National University Medical School, Gwangju, South Korea
| | - Seok-Yong Choi
- Department of Biomedical Sciences, Chonnam National University Medical School, Gwangju, South Korea
| | | | - Myeong-Kyu Kim
- Department of Neurology, Chonnam National University Medical School, Chonnam National University Hospital, Gwangju, South Korea
| |
Collapse
|
10
|
Carnero Contentti E, Rojas JI, Cristiano E, Marques VD, Flores-Rivera J, Lana-Peixoto M, Navas C, Papais-Alvarenga R, Sato DK, Soto de Castillo I, Correale J. Latin American consensus recommendations for management and treatment of neuromyelitis optica spectrum disorders in clinical practice. Mult Scler Relat Disord 2020; 45:102428. [DOI: 10.1016/j.msard.2020.102428] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2020] [Revised: 07/24/2020] [Accepted: 07/27/2020] [Indexed: 02/06/2023]
|
11
|
Klein da Costa B, Brant de Souza Melo R, Passos GRD, Gomes Meneses Sevilha Castro D, Becker J, Bar-Or A, Sato DK. Unraveling B lymphocytes in CNS inflammatory diseases: Distinct mechanisms and treatment targets. Neurology 2020; 95:733-744. [PMID: 32907966 DOI: 10.1212/wnl.0000000000010789] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Accepted: 08/03/2020] [Indexed: 12/15/2022] Open
Abstract
Specific therapies targeting B lymphocytes in multiple sclerosis (MS) have demonstrated reductions in disease activity and disability progression. Several observational studies have also shown the effects of targeting B lymphocytes in other rare CNS inflammatory diseases, such as neuromyelitis optica spectrum disorder (NMOSD) and autoimmune encephalitis (AE). However, some drugs targeting cytokine receptors involved in B-lymphocyte maturation and proliferation resulted in negative outcomes in MS. These apparently conflicting findings have stimulated research on the pathophysiologic mechanisms of B lymphocytes in CNS inflammatory diseases. It has been demonstrated that B lymphocytes participate in the pathogenesis of these conditions as antigen-presenting cells, producing proinflammatory cytokines that induce Th1 and Th17 responses and producing antibodies. However, they are also able to produce anti-inflammatory cytokines, such as interleukin-10, functioning as regulators of autoimmunity. Understanding these diverse effects is essential for the development of focused treatments. In this review, we discuss the possible mechanisms that underlie B-lymphocyte involvement in MS, NMOSD, and AE and the outcomes obtained by treatments targeting B lymphocytes.
Collapse
Affiliation(s)
- Bruna Klein da Costa
- From the Brain Institute of Rio Grande do Sul (BraIns) and School of Medicine (B.K.d.C., R.B.d.S.M., G.R.d.P., J.B., D.K.S.), Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre; Santa Casa de Belo Horizonte (R.B.d.S.M.), Belo Horizonte; Produtos Roche Químicos e Farmacêuticos S.A. (D.G.M.S.C.), São Paulo, Brazil; and Perelman Center for Advanced Medicine (PCAM) (A.B.-O), University of Pennsylvania, Philadelphia.
| | - Renata Brant de Souza Melo
- From the Brain Institute of Rio Grande do Sul (BraIns) and School of Medicine (B.K.d.C., R.B.d.S.M., G.R.d.P., J.B., D.K.S.), Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre; Santa Casa de Belo Horizonte (R.B.d.S.M.), Belo Horizonte; Produtos Roche Químicos e Farmacêuticos S.A. (D.G.M.S.C.), São Paulo, Brazil; and Perelman Center for Advanced Medicine (PCAM) (A.B.-O), University of Pennsylvania, Philadelphia
| | - Giordani Rodrigues Dos Passos
- From the Brain Institute of Rio Grande do Sul (BraIns) and School of Medicine (B.K.d.C., R.B.d.S.M., G.R.d.P., J.B., D.K.S.), Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre; Santa Casa de Belo Horizonte (R.B.d.S.M.), Belo Horizonte; Produtos Roche Químicos e Farmacêuticos S.A. (D.G.M.S.C.), São Paulo, Brazil; and Perelman Center for Advanced Medicine (PCAM) (A.B.-O), University of Pennsylvania, Philadelphia
| | - Douglas Gomes Meneses Sevilha Castro
- From the Brain Institute of Rio Grande do Sul (BraIns) and School of Medicine (B.K.d.C., R.B.d.S.M., G.R.d.P., J.B., D.K.S.), Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre; Santa Casa de Belo Horizonte (R.B.d.S.M.), Belo Horizonte; Produtos Roche Químicos e Farmacêuticos S.A. (D.G.M.S.C.), São Paulo, Brazil; and Perelman Center for Advanced Medicine (PCAM) (A.B.-O), University of Pennsylvania, Philadelphia
| | - Jefferson Becker
- From the Brain Institute of Rio Grande do Sul (BraIns) and School of Medicine (B.K.d.C., R.B.d.S.M., G.R.d.P., J.B., D.K.S.), Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre; Santa Casa de Belo Horizonte (R.B.d.S.M.), Belo Horizonte; Produtos Roche Químicos e Farmacêuticos S.A. (D.G.M.S.C.), São Paulo, Brazil; and Perelman Center for Advanced Medicine (PCAM) (A.B.-O), University of Pennsylvania, Philadelphia
| | - Amit Bar-Or
- From the Brain Institute of Rio Grande do Sul (BraIns) and School of Medicine (B.K.d.C., R.B.d.S.M., G.R.d.P., J.B., D.K.S.), Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre; Santa Casa de Belo Horizonte (R.B.d.S.M.), Belo Horizonte; Produtos Roche Químicos e Farmacêuticos S.A. (D.G.M.S.C.), São Paulo, Brazil; and Perelman Center for Advanced Medicine (PCAM) (A.B.-O), University of Pennsylvania, Philadelphia
| | - Douglas Kazutoshi Sato
- From the Brain Institute of Rio Grande do Sul (BraIns) and School of Medicine (B.K.d.C., R.B.d.S.M., G.R.d.P., J.B., D.K.S.), Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre; Santa Casa de Belo Horizonte (R.B.d.S.M.), Belo Horizonte; Produtos Roche Químicos e Farmacêuticos S.A. (D.G.M.S.C.), São Paulo, Brazil; and Perelman Center for Advanced Medicine (PCAM) (A.B.-O), University of Pennsylvania, Philadelphia
| |
Collapse
|
12
|
Whittam DH, Cobo-Calvo A, Lopez-Chiriboga AS, Pardo S, Gornall M, Cicconi S, Brandt A, Berek K, Berger T, Jelcic I, Gombolay G, Oliveira LM, Callegaro D, Kaneko K, Misu T, Capobianco M, Gibbons E, Karthikeayan V, Brochet B, Audoin B, Mathey G, Laplaud D, Thouvenot E, Cohen M, Tourbah A, Maillart E, Ciron J, Deschamps R, Biotti D, Rostasy K, Neuteboom R, Hemingway C, Forsyth R, Matiello M, Webb S, Hunt D, Murray K, Hacohen Y, Lim M, Leite MI, Palace J, Solomon T, Lutterotti A, Fujihara K, Nakashima I, Bennett JL, Pandit L, Chitnis T, Weinshenker BG, Wildemann B, Sato DK, Kim SH, Huda S, Kim HJ, Reindl M, Levy M, Jarius S, Tenembaum S, Paul F, Pittock S, Marignier R, Jacob A. Treatment of MOG-IgG-associated disorder with rituximab: An international study of 121 patients. Mult Scler Relat Disord 2020; 44:102251. [PMID: 32629363 PMCID: PMC7895306 DOI: 10.1016/j.msard.2020.102251] [Citation(s) in RCA: 98] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Revised: 05/25/2020] [Accepted: 05/29/2020] [Indexed: 11/22/2022]
Abstract
OBJECTIVE To assess the effect of anti-CD20 B-cell depletion with rituximab (RTX) on relapse rates in myelin oligodendrocyte glycoprotein antibody-associated disorder (MOGAD). METHODS Retrospective review of RTX-treated MOGAD patients from 29 centres in 13 countries. The primary outcome measure was change in relapse rate after starting rituximab (Poisson regression model). RESULTS Data on 121 patients were analysed, including 30 (24.8%) children. Twenty/121 (16.5%) were treated after one attack, of whom 14/20 (70.0%) remained relapse-free after median (IQR) 11.2 (6.3-14.1) months. The remainder (101/121, 83.5%) were treated after two or more attacks, of whom 53/101 (52.5%) remained relapse-free after median 12.1 (6.3-24.9) months. In this 'relapsing group', relapse rate declined by 37% (95%CI=19-52%, p<0.001) overall, 63% (95%CI=35-79%, p = 0.001) when RTX was used first line (n = 47), and 26% (95%CI=2-44%, p = 0.038) when used after other steroid-sparing immunotherapies (n = 54). Predicted 1-year and 2-year relapse-free survival was 79% and 55% for first-line RTX therapy, and 38% and 18% for second-/third-line therapy. Circulating CD19+B-cells were suppressed to <1% of total circulating lymphocyte population at the time of 45/57 (78.9%) relapses. CONCLUSION RTX reduced relapse rates in MOGAD. However, many patients continued to relapse despite apparent B-cell depletion. Prospective controlled studies are needed to validate these results.
Collapse
Affiliation(s)
- Daniel H Whittam
- Department of Neurology, The Walton Centre NHS Foundation Trust, Liverpool, United Kingdom; Institute of Infection and Global Health, University of Liverpool, Liverpool, United Kingdom.
| | - Alvaro Cobo-Calvo
- Pathologies de la Myéline et Neuro-inflammation, Hôpital Neurologique Pierre Wertheimer Hospices Civils de Lyon, Centre de Référence des Maladies Inflammatoires Rares du Cerveau et de la Moelle, INSERM U1028, CNRS UMR 5292, Lyon 1 University, Center for Research in Neuroscience of Lyon, Lyon, France
| | | | - Santiago Pardo
- Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, United States
| | - Matthew Gornall
- Liverpool Clinical Trials Centre, University of Liverpool, Liverpool, United Kingdom
| | - Silvia Cicconi
- Liverpool Clinical Trials Centre, University of Liverpool, Liverpool, United Kingdom
| | - Alexander Brandt
- Experimental and Clinical Research Center Max Delbrueck Center for Molecular Medicine, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Klaus Berek
- Medical University of Innsbruck, Innsbruck, Austria
| | - Thomas Berger
- Department of Neurology, Medical University of Vienna, Vienna, Austria
| | | | - Grace Gombolay
- Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, United States; Emory & Children's Pediatric Institute, Atlanta, Georgia, United States
| | | | - Dagoberto Callegaro
- Hospital das Clinicas Faculty of Medicine, University of São Paulo, São Paulo, Brazil
| | - Kimihiko Kaneko
- Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Tatsuro Misu
- Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Marco Capobianco
- Neurology Dept. - Regional MS Centre, S. Luigi University Hospital, Orbassano, Italy
| | - Emily Gibbons
- Department of Neurology, The Walton Centre NHS Foundation Trust, Liverpool, United Kingdom; Institute of Infection and Global Health, University of Liverpool, Liverpool, United Kingdom
| | | | - Bruno Brochet
- CHU de Bordeaux & INSERM U 1215, University of Bordeaux, Bordeaux, France
| | - Bertrand Audoin
- Aix Marseille University, APHM, Hôpital de La Timone, Marseille, France
| | | | | | - Eric Thouvenot
- Hôpital Carémeau, Nimes University Hospital, Nimes, France
| | - Mikaël Cohen
- Université Côte d'Azur, Centre Hospitalier Universitaire de Nice, Nice, France
| | - Ayman Tourbah
- Reims University Hospital, University of Reims Champagne-Ardenne, Reims, LPN EA 2027, University of Paris VIII, Saint-Denis, France
| | | | | | | | | | - Kevin Rostasy
- Department of Pediatric Neurology, Children's Hospital Datteln, University Witten/Herdecke, Witten, Germany
| | - Rinze Neuteboom
- Department of Paediatric Neurology, Erasmus MC-Sophia, Rotterdam, the Netherlands; Department of Neurology, MS Centre ErasMS, Erasmus MC, Rotterdam, the Netherlands
| | - Cheryl Hemingway
- Department of Paediatric Neurology, Great Ormond Street Hospital for Children, London, United Kingdom
| | - Rob Forsyth
- Institute of Neuroscience, Newcastle University, Newcastle, United Kingdom
| | - Marcelo Matiello
- Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, United States
| | - Stewart Webb
- Department of Neurology, Queen Elizabeth University Hospital, Glasgow, United Kingdom
| | - David Hunt
- Anne Rowling Regenerative Neurology Clinic, University of Edinburgh, Edinburgh, United Kingdom
| | - Katy Murray
- Anne Rowling Regenerative Neurology Clinic, University of Edinburgh, Edinburgh, United Kingdom
| | - Yael Hacohen
- Department of Paediatric Neurology, Great Ormond Street Hospital for Children, London, United Kingdom; Department of Neuroinflammation, Queen Square MS Centre, UCL Institute of Neurology, London, United Kingdom
| | - Ming Lim
- Children's Neuroscience, Evelina London Children's Hospital @ Guy's and St Thomas' NHS Foundation Trust, London, United Kingdom; Faculty of Life Sciences and Medicine, King's College London, London, United Kingdom
| | - M Isabel Leite
- Nuffield Department of Clinical Neurosciences, John Radcliffe Hospital, Oxford, United Kingdom
| | - Jacqueline Palace
- Nuffield Department of Clinical Neurosciences, John Radcliffe Hospital, Oxford, United Kingdom
| | - Tom Solomon
- Department of Neurology, The Walton Centre NHS Foundation Trust, Liverpool, United Kingdom; Institute of Infection and Global Health, University of Liverpool, Liverpool, United Kingdom
| | | | - Kazuo Fujihara
- Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Ichiro Nakashima
- Department of Neurology, Tohoku Medical and Pharmaceutical University, Sendai, Japan
| | - Jeffrey L Bennett
- Departments of Neurology and Ophthalmology, Program in Neuroscience, University of Colorado School of Medicine, Aurora, Colorado, United States
| | | | - Tanuja Chitnis
- Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, United States
| | | | - Brigitte Wildemann
- Molecular Neuroimmunology Group, Department of Neurology, University of Heidelberg, Heidelberg, Germany
| | - Douglas Kazutoshi Sato
- Hospital das Clinicas Faculty of Medicine, University of São Paulo, São Paulo, Brazil; Tohoku University Graduate School of Medicine, Sendai, Japan; Pontificia Universidade Catolica Rio Grande Do Sul, Porto Alegre RS, Brazil
| | - Su-Hyun Kim
- Research Institute and Hospital of National Cancer Center, Goyang, South Korea
| | - Saif Huda
- Department of Neurology, The Walton Centre NHS Foundation Trust, Liverpool, United Kingdom; Institute of Infection and Global Health, University of Liverpool, Liverpool, United Kingdom
| | - Ho Jin Kim
- Research Institute and Hospital of National Cancer Center, Goyang, South Korea
| | | | - Michael Levy
- Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, United States
| | - Sven Jarius
- Molecular Neuroimmunology Group, Department of Neurology, University of Heidelberg, Heidelberg, Germany
| | - Silvia Tenembaum
- National Pediatric Hospital Dr. Juan P. Garrahan, Ciudad de Buenos Aires, Argentina
| | - Friedemann Paul
- Experimental and Clinical Research Center Max Delbrueck Center for Molecular Medicine, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Sean Pittock
- Department of Neurology, Mayo Clinic, Rochester, Minnesota, United States
| | - Romain Marignier
- Pathologies de la Myéline et Neuro-inflammation, Hôpital Neurologique Pierre Wertheimer Hospices Civils de Lyon, Centre de Référence des Maladies Inflammatoires Rares du Cerveau et de la Moelle, INSERM U1028, CNRS UMR 5292, Lyon 1 University, Center for Research in Neuroscience of Lyon, Lyon, France
| | - Anu Jacob
- Department of Neurology, The Walton Centre NHS Foundation Trust, Liverpool, United Kingdom; Institute of Infection and Global Health, University of Liverpool, Liverpool, United Kingdom; Department of Neurology, The Cleveland Clinic Abu Dhabi, United Arab Emirates.
| |
Collapse
|
13
|
Differential Effects of MS Therapeutics on B Cells-Implications for Their Use and Failure in AQP4-Positive NMOSD Patients. Int J Mol Sci 2020; 21:ijms21145021. [PMID: 32708663 PMCID: PMC7404039 DOI: 10.3390/ijms21145021] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Revised: 07/11/2020] [Accepted: 07/13/2020] [Indexed: 12/25/2022] Open
Abstract
B cells are considered major contributors to multiple sclerosis (MS) pathophysiology. While lately approved disease-modifying drugs like ocrelizumab deplete B cells directly, most MS medications were not primarily designed to target B cells. Here, we review the current understanding how approved MS medications affect peripheral B lymphocytes in humans. These highly contrasting effects are of substantial importance when considering these drugs as therapy for neuromyelitis optica spectrum disorders (NMOSD), a frequent differential diagnosis to MS, which is considered being a primarily B cell- and antibody-driven diseases. Data indicates that MS medications, which deplete B cells or induce an anti-inflammatory phenotype of the remaining ones, were effective and safe in aquaporin-4 antibody positive NMOSD. In contrast, drugs such as natalizumab and interferon-β, which lead to activation and accumulation of B cells in the peripheral blood, lack efficacy or even induce catastrophic disease activity in NMOSD. Hence, we conclude that the differential effect of MS drugs on B cells is one potential parameter determining the therapeutic efficacy or failure in antibody-dependent diseases like seropositive NMOSD.
Collapse
|
14
|
Vakrakou AG, Tzanetakos D, Argyrakos T, Koutsis G, Evangelopoulos ME, Andreadou E, Anagnostouli M, Breza M, Tzartos JS, Gialafos E, Dimitrakopoulos AN, Velonakis G, Toulas P, Stefanis L, Kilidireas C. Recurrent Fulminant Tumefactive Demyelination With Marburg-Like Features and Atypical Presentation: Therapeutic Dilemmas and Review of Literature. Front Neurol 2020; 11:536. [PMID: 32714265 PMCID: PMC7344179 DOI: 10.3389/fneur.2020.00536] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Accepted: 05/14/2020] [Indexed: 12/31/2022] Open
Abstract
Atypical forms of demyelinating diseases with tumor-like lesions and aggressive course represent a diagnostic and therapeutic challenge for neurologists. Herein, we describe a 50-year-old woman presenting with subacute onset of left hemiparesis, memory difficulties and headache. Brain MRI revealed a tumefactive right frontal-parietal lesion with perilesional edema, mass effect and homogenous post-contrast enhancement, along with other small atypical lesions in the white-matter. Brain biopsy of cerebral lesion ruled out lymphoma or any other neoplastic process and patient placed on corticosteroids with complete clinical/radiological remission. Two years after disease initiation, there was disease exacerbation with reappearance of the tumor-like mass. The patient initially responded to high doses of corticosteroids but soon became resistant. Plasma-exchange sessions were not able to limit disease burden. Resistance to therapeutic efforts led to a second biopsy that showed perivascular demyelination, predominantly consisting of macrophages, with a small number of T and B lymphocytes, and the presence of reactive astrocytes, typical of Creutzfeldt-Peters cells. The patient received high doses of cyclophosphamide with substantial clinical/radiological response but relapsed after 7-intensive cycles. She received 4-weekly doses of rituximab with disease exacerbation and brainstem involvement. She eventually died with complicated pneumonia. We present a very rare case of recurrent tumefactive demyelinating lesions, with atypical tumor-like characteristics, with initial response to corticosteroids and cyclophosphamide, but subsequent development of drug-resistance and unexpected exacerbation upon rituximab administration. Our clinical case raises therapeutic dilemmas and points to the need for immediate and appropriate immunosuppression in difficult to treat tumefactive CNS lesions with Marburg-like features.
Collapse
Affiliation(s)
- Aigli G Vakrakou
- Demyelinating Diseases Unit, 1st Department of Neurology, School of Medicine, Aeginition Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Dimitrios Tzanetakos
- Demyelinating Diseases Unit, 1st Department of Neurology, School of Medicine, Aeginition Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | | | - Georgios Koutsis
- Demyelinating Diseases Unit, 1st Department of Neurology, School of Medicine, Aeginition Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Maria-Eleptheria Evangelopoulos
- Demyelinating Diseases Unit, 1st Department of Neurology, School of Medicine, Aeginition Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Elisabeth Andreadou
- Demyelinating Diseases Unit, 1st Department of Neurology, School of Medicine, Aeginition Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Maria Anagnostouli
- Demyelinating Diseases Unit, 1st Department of Neurology, School of Medicine, Aeginition Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Marianthi Breza
- Demyelinating Diseases Unit, 1st Department of Neurology, School of Medicine, Aeginition Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - John S Tzartos
- Demyelinating Diseases Unit, 1st Department of Neurology, School of Medicine, Aeginition Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Elias Gialafos
- Demyelinating Diseases Unit, 1st Department of Neurology, School of Medicine, Aeginition Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Antonios N Dimitrakopoulos
- Demyelinating Diseases Unit, 1st Department of Neurology, School of Medicine, Aeginition Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Georgios Velonakis
- Research Unit of Radiology, 2nd Department of Radiology, National and Kapodistrian University of Athens, Athens, Greece
| | - Panagiotis Toulas
- Research Unit of Radiology, 2nd Department of Radiology, National and Kapodistrian University of Athens, Athens, Greece
| | - Leonidas Stefanis
- Demyelinating Diseases Unit, 1st Department of Neurology, School of Medicine, Aeginition Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Constantinos Kilidireas
- Demyelinating Diseases Unit, 1st Department of Neurology, School of Medicine, Aeginition Hospital, National and Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
15
|
Tahara M, Oeda T, Okada K, Kiriyama T, Ochi K, Maruyama H, Fukaura H, Nomura K, Shimizu Y, Mori M, Nakashima I, Misu T, Umemura A, Yamamoto K, Sawada H. 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. [PMID: 32199095 DOI: 10.1016/s1474-4422(20)30066-1] [Citation(s) in RCA: 205] [Impact Index Per Article: 51.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Revised: 02/05/2020] [Accepted: 02/19/2020] [Indexed: 02/08/2023]
Abstract
BACKGROUND Pharmacological prevention against relapses in patients with neuromyelitis optica spectrum disorder (NMOSD) is developing rapidly. We aimed to investigate the safety and efficacy of rituximab, an anti-CD20 monoclonal antibody, against relapses in patients with NMOSD. METHODS We did a multicentre, randomised, double-blind, placebo-controlled clinical trial at eight hospitals in Japan. Patients aged 16-80 years with NMOSD who were seropositive for aquaporin 4 (AQP4) antibody, were taking 5-30 mg/day oral steroids, and had an Expanded Disability Status Scale (EDSS) score of 7·0 or less were eligible for the study. Individuals taking any other immunosuppressants were excluded. Participants were randomly allocated (1:1) either rituximab or placebo by a computer-aided dynamic random allocation system. The doses of concomitant steroid (converted to equivalent doses of prednisolone) and relapses in previous 2 years were set as stratification factors. Participants and those assessing outcomes were unaware of group assignments. Rituximab (375 mg/m2) was administered intravenously every week for 4 weeks, then 6-month interval dosing was done (1000 mg every 2 weeks, at 24 weeks and 48 weeks after randomisation). A matching placebo was administered intravenously. Concomitant oral prednisolone was gradually reduced to 2-5 mg/day, according to the protocol. The primary outcome was time to first relapse within 72 weeks. Relapses were defined as patient-reported symptoms or any new signs consistent with CNS lesions and attributable objective changes in MRI or visual evoked potential. The primary analysis was done in the full analysis set (all randomly assigned patients) and safety analyses were done in the safety analysis set (all patients who received at least one infusion of assigned treatment). The primary analysis was by intention-to-treat principles. This trial is registered with the UMIN clinical trial registry, UMIN000013453. FINDINGS Between May 10, 2014, and Aug 15, 2017, 38 participants were recruited and randomly allocated either rituximab (n=19) or placebo (n=19). Three (16%) patients assigned rituximab discontinued the study and were analysed as censored cases. Seven (37%) relapses occurred in patients allocated placebo and none were recorded in patients assigned rituximab (group difference 36·8%, 95% CI 12·3-65·5; log-rank p=0·0058). Eight serious adverse events were recorded, four events in three (16%) patients assigned rituximab (lumbar compression fracture and infection around nail of right foot [n=1], diplopia [n=1], and uterine cancer [n=1]) and four events in two (11%) people allocated to placebo (exacerbation of glaucoma and bleeding in the right eye chamber after surgery [n=1], and visual impairment and asymptomatic white matter brain lesion on MRI [n=1]); all patients recovered. No deaths were reported. INTERPRETATION Rituximab prevented relapses for 72 weeks in patients with NMOSD who were AQP4 antibody-positive. This study is limited by its small sample size and inclusion of participants with mild disease activity. However, our results suggest that rituximab could be useful maintenance therapy for individuals with NMOSD who are AQP4 antibody-positive. FUNDING Japanese Ministry of Health, Labour and Welfare, Japan Agency for Medical Research and Development, and Zenyaku Kogyo.
Collapse
Affiliation(s)
- Masayuki Tahara
- Clinical Research Centre and Department of Neurology, National Hospital Organization Utano National Hospital, Kyoto, Japan.
| | - Tomoko Oeda
- Clinical Research Centre and Department of Neurology, National Hospital Organization Utano National Hospital, Kyoto, Japan
| | - Kazumasa Okada
- Department of Neurology, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Takao Kiriyama
- Department of Neurology, Nara Medical University School of Medicine, Nara, Japan
| | - Kazuhide Ochi
- Department of Clinical Neuroscience and Therapeutics, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Hirofumi Maruyama
- Department of Clinical Neuroscience and Therapeutics, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Hikoaki Fukaura
- Department of Neurology, Saitama Medical University, Kawagoe, Japan
| | - Kyoichi Nomura
- Department of Neurology, Saitama Medical University, Kawagoe, Japan
| | - Yuko Shimizu
- Department of Neurology, Tokyo Women's Medical University School of Medicine, Tokyo, Japan
| | - Masahiro Mori
- Department of Neurology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Ichiro Nakashima
- Department of Neurology, Tohoku Medical and Pharmaceutical University, Sendai, Japan
| | - Tatsuro Misu
- Department of Neurology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Atsushi Umemura
- Clinical Research Centre and Department of Neurology, National Hospital Organization Utano National Hospital, Kyoto, Japan
| | - Kenji Yamamoto
- Clinical Research Centre and Department of Neurology, National Hospital Organization Utano National Hospital, Kyoto, Japan
| | - Hideyuki Sawada
- Clinical Research Centre and Department of Neurology, National Hospital Organization Utano National Hospital, Kyoto, Japan
| |
Collapse
|
16
|
Nicolas P, Ruiz A, Cobo-Calvo A, Fiard G, Giraudon P, Vukusic S, Marignier R. The Balance in T Follicular Helper Cell Subsets Is Altered in Neuromyelitis Optica Spectrum Disorder Patients and Restored by Rituximab. Front Immunol 2019; 10:2686. [PMID: 31803192 PMCID: PMC6877601 DOI: 10.3389/fimmu.2019.02686] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2019] [Accepted: 10/31/2019] [Indexed: 01/18/2023] Open
Abstract
Neuromyelitis optica spectrum disorder (NMOSD) is a rare and severe auto-immune disease of the central nervous system driven by pathogenic antibodies mainly directed against aquaporin-4 (AQP4-Ab). Treatment of NMOSD currently relies on immunosuppressants (mycophenolate mofetil, azathioprine) or B-cell-depleting therapy (rituximab). B-cell differentiation into antibody-producing cells requires T follicular helper cells (Tfh). There are several Tfh subsets that differentially affect B-cell differentiation; Tfh2 and Tfh17 subsets strongly support B-cell differentiation. By contrast, Tfh1 lack this capacity and T follicular regulatory cells (Tfr), inhibit B-cell differentiation into antibody-producing cells. We performed a broad characterization of circulating Tfh subsets in 25 NMOSD patients and analyzed the impact of different treatments on these subsets. Untreated NMOSD patients presented a Tfh polarization toward excessive B-helper Tfh subsets with an increase of Tfh17 and (Tfh2+Tfh17)/Tfh1 ratio and a decrease of Tfr and Tfh1. Rituximab restored the Tfh polarization to that of healthy controls. There was a trend toward a similar result for azathioprine and mycophenolate mofetil. Our results suggest that NMOSD patients present an impaired balance in Tfh subsets favoring B-cell differentiation which may explain the sustained antibody production. These findings provide new insights into the pathophysiology of NMOSD, and further suggest that Tfh and Tfr subsets could be considered as potential therapeutic target in NMOSD because of their upstream role in antibody production.
Collapse
Affiliation(s)
- Philippe Nicolas
- Service de Neurologie, Sclérose en Plaques, Pathologies de la Myéline et Neuro-Inflammation, et Centre de Référence des Maladies Inflammatoires Rares du Cerveau et de la Moelle (MIRCEM), Hôpital Neurologique Pierre Wertheimer, Hospices Civils de Lyon, Bron, France.,Centre de Recherche en Neurosciences de Lyon, INSERM U1028-CNRS UMR5292-UCBL, Bron, France
| | - Anne Ruiz
- Centre de Recherche en Neurosciences de Lyon, INSERM U1028-CNRS UMR5292-UCBL, Bron, France
| | - Alvaro Cobo-Calvo
- Service de Neurologie, Sclérose en Plaques, Pathologies de la Myéline et Neuro-Inflammation, et Centre de Référence des Maladies Inflammatoires Rares du Cerveau et de la Moelle (MIRCEM), Hôpital Neurologique Pierre Wertheimer, Hospices Civils de Lyon, Bron, France.,Centre de Recherche en Neurosciences de Lyon, INSERM U1028-CNRS UMR5292-UCBL, Bron, France
| | - Guillaume Fiard
- NeuroBioTec, Hôpital Neurologique Pierre Wertheimer, Bron, France
| | - Pascale Giraudon
- Centre de Recherche en Neurosciences de Lyon, INSERM U1028-CNRS UMR5292-UCBL, Bron, France
| | - Sandra Vukusic
- Service de Neurologie, Sclérose en Plaques, Pathologies de la Myéline et Neuro-Inflammation, et Centre de Référence des Maladies Inflammatoires Rares du Cerveau et de la Moelle (MIRCEM), Hôpital Neurologique Pierre Wertheimer, Hospices Civils de Lyon, Bron, France.,Centre de Recherche en Neurosciences de Lyon, INSERM U1028-CNRS UMR5292-UCBL, Bron, France
| | - Romain Marignier
- Service de Neurologie, Sclérose en Plaques, Pathologies de la Myéline et Neuro-Inflammation, et Centre de Référence des Maladies Inflammatoires Rares du Cerveau et de la Moelle (MIRCEM), Hôpital Neurologique Pierre Wertheimer, Hospices Civils de Lyon, Bron, France.,Centre de Recherche en Neurosciences de Lyon, INSERM U1028-CNRS UMR5292-UCBL, Bron, France.,NeuroBioTec, Hôpital Neurologique Pierre Wertheimer, Bron, France
| |
Collapse
|
17
|
Novi G, Bovis F, Capobianco M, Frau J, Mataluni G, Curti E, Zuliani L, Cavalla P, Brambilla L, Annovazzi P, Repice AM, Lanzillo R, Esposito S, Benedetti L, Maietta I, Sica F, Buttari F, Malucchi S, Fenu G, Landi D, Bosa C, Realmuto S, Malentacchi M, Granella F, Signori A, Bonavita S, Uccelli A, Sormani MP. Efficacy of different rituximab therapeutic strategies in patients with neuromyelitis optica spectrum disorders. Mult Scler Relat Disord 2019; 36:101430. [DOI: 10.1016/j.msard.2019.101430] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2019] [Revised: 09/17/2019] [Accepted: 10/02/2019] [Indexed: 11/17/2022]
|
18
|
Whittam DH, Tallantyre EC, Jolles S, Huda S, Moots RJ, Kim HJ, Robertson NP, Cree BAC, Jacob A. Rituximab in neurological disease: principles, evidence and practice. Pract Neurol 2019; 19:5-20. [PMID: 30498056 DOI: 10.1136/practneurol-2018-001899] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
Rituximab is a widely used B-cell-depleting monoclonal antibody. It is unlicensed for use in neurological disorders and there are no treatment guidelines. However, as a rapidly acting, targeted therapy with growing evidence of efficacy and tolerability in several neuroinflammatory disorders, it is an attractive alternative to conventional immunomodulatory medications. This practical review aims to explain the basic principles of B-cell depletion with therapeutic monoclonal antibodies. We present the evidence for using rituximab in neurological diseases, and describe the practical aspects of prescribing, including dosing, monitoring, safety, treatment failure and its use in special circumstances such as coexisting viral hepatitis, pregnancy and lactation. We provide an administration guide, checklist and patient information leaflet, which can be adapted for local use. Finally, we review the safety data of rituximab and ocrelizumab (a newer and recently licensed B-cell-depleting therapy for multiple sclerosis) and suggest monitoring and risk reduction strategies.
Collapse
Affiliation(s)
- Daniel H Whittam
- Department of Neurology, The Walton Centre NHS Foundation Trust, Liverpool, UK
| | - Emma C Tallantyre
- Helen Durham Centre for Neuroinflammation, University Hospital or Wales, Cardiff, UK
- Division of Psychological Medicine and Clinical Neurosciences, School of Medicine, Cardiff University, Cardiff, UK
| | - Stephen Jolles
- Immunodeficiency Centre for Wales, University Hospital of Wales, Cardiff, UK
- School of Medicine, Cardiff University, Cardiff, UK
| | - Saif Huda
- Department of Neurology, The Walton Centre NHS Foundation Trust, Liverpool, UK
| | - Robert J Moots
- Department of Musculoskeletal Diseases, Institute of Ageing and Chronic Diseases, University of Liverpool, Liverpool, UK
| | - Ho Jin Kim
- Department of Neurology, Research Institute and Hospital of National Cancer Center, Goyang, South Korea
| | - Neil P Robertson
- Helen Durham Centre for Neuroinflammation, University Hospital or Wales, Cardiff, UK
- Division of Psychological Medicine and Clinical Neurosciences, School of Medicine, Cardiff University, Cardiff, UK
| | - Bruce A C Cree
- Weill Institute for Neurosciences, University of California, San Francisco, California, USA
| | - Anu Jacob
- Department of Neurology, The Walton Centre NHS Foundation Trust, Liverpool, UK
- School of Medicine, University of Liverpool, Liverpool, UK
| |
Collapse
|
19
|
Kim SH, Hyun JW, Kim HJ. Individualized B cell-targeting therapy for neuromyelitis optica spectrum disorder. Neurochem Int 2018; 130:104347. [PMID: 30513364 DOI: 10.1016/j.neuint.2018.11.022] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Revised: 11/12/2018] [Accepted: 11/30/2018] [Indexed: 12/23/2022]
Abstract
Neuromyelitis optica spectrum disorder (NMOSD) is an autoimmune inflammatory disease of the central nervous system characterized by severe attacks of optic neuritis (ON), longitudinally extensive transverse myelitis (LETM), and area postrema syndrome. The majority of patients with NMOSD are seropositive for autoantibodies against the astrocyte water channel aquaporin-4 (AQP4). As convergent clinical and laboratory-based investigations have indicated that B cells play a fundamental role in NMO immunopathology, B cells have become an attractive therapeutic target. Rituximab is a therapeutic monoclonal antibody against CD20 expressed on B cells and increasingly used for the treatment of NMOSD. Although there is robust evidence for the efficacy and safety of rituximab in NMOSD, considerable variability has been noted in biological and clinical responses in patients. Therefore, the focus now is on understanding the mechanisms underlying the variability in response to rituximab and optimizing the use of rituximab for NMOSD. Identification of biomarkers for prediction of clinical response, and effective dosing and timing of treatment may provide useful tools for patient-tailored treatment in NMOSD. Herein, we review current evidence on factors that affect biological and clinical responses to rituximab and highlight the importance of individualized therapies for NMOSD.
Collapse
Affiliation(s)
- Su-Hyun Kim
- Department of Neurology, Research Institute and Hospital of National Cancer Center, Republic of Korea
| | - Jae-Won Hyun
- Department of Neurology, Research Institute and Hospital of National Cancer Center, Republic of Korea
| | - Ho Jin Kim
- Department of Neurology, Research Institute and Hospital of National Cancer Center, Republic of Korea.
| |
Collapse
|
20
|
Araki M. Blockade of IL-6 signaling in neuromyelitis optica. Neurochem Int 2018; 130:104315. [PMID: 30342072 DOI: 10.1016/j.neuint.2018.10.012] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2018] [Revised: 10/04/2018] [Accepted: 10/17/2018] [Indexed: 01/14/2023]
Abstract
Neuromyelitis optica (NMO) and neuromyelitis optica spectrum disorder (NMOSD) are autoimmune diseases associated with a disease-specific autoantibody directed against the water channel protein aquaporin-4. Standard immunotherapy, immunosuppressive agents, and corticosteroids can prevent acute attacks and maintain remission in most patients with NMOSD. However, there is a strong need for additional options for patients who are refractory to standard treatments. Emerging therapies targeting specific molecules related to the pathogenicity of NMOSD are currently being developed. The review focuses on improving preventive treatments for NMOSD, including ongoing randomized clinical trials using biological drugs targeting CD19 and CD20 on B cells, interleukin-6, and complement protein C5. The anti-IL-6 receptor monoclonal antibody tocilizumab (TCZ), which can block IL-6 signaling, was shown to be highly effective for refractory patients with NMOSD. Notably, TCZ has marked effects on chronic neuropathic pain and general fatigue in patients refractory to standard medications. TCZ is a promising drug for preventing acute attacks in patients with NMOSD.
Collapse
Affiliation(s)
- Manabu Araki
- Multiple Sclerosis Center, National Center of Neurology and Psychiatry, Tokyo, Japan; Department of Immunology, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Tokyo, Japan.
| |
Collapse
|
21
|
Only Follow-Up of Memory B Cells Helps Monitor Rituximab Administration to Patients with Neuromyelitis Optica Spectrum Disorders. Neurol Ther 2018; 7:373-383. [PMID: 29881979 PMCID: PMC6283795 DOI: 10.1007/s40120-018-0101-4] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Indexed: 01/01/2023] Open
Abstract
Introduction Neuromyelitis optica spectrum disorders (NMOSD) are identified as a spectrum of inflammatory demyelinating disorders involving the brain, spinal cord and optic nerves. These disorders require early diagnosis and highly active immunosuppressive treatment. Rituximab (RTX) has demonstrated efficacy in limiting relapse in NMOSD when using several administration schedules. We questioned if the CD19+ CD27+ memory B cell count was a more reliable marker to monitor RTX administration than the RTX plasma level and CD19+ B cell count. Methods We analyzed 125 blood samples from 17 NMOSD patients treated with RTX and also measured the level of anti-aquaporine-4 antibodies (anti-AQP-4 Abs), human anti-chimeric antibodies to the murine fragment of RTX (HACA-RTX Abs), and the RTX concentration. Results The mean follow-up time of the cohort was 7.4 (2–16) years. All patients improved with a mean EDSS going from 4 (1–8.5) to 2.7 (1–5.5). The mean interval between RTX infusions was 9.6 months with identification of prolonged responders. Total CD19+ B cell detection with the routine technique did not correlate to re-emergence of CD19+ CD27+ memory B cells. The RTX residual concentration did not correlate with the CD19+ CD27+ memory B cell count or with anti-RTX antibody production. Conclusion In contrast to total CD19+ cell, detected with the routine technique, CD19+ CD27+ memory B cells are a reliable marker for biological relapse and allow a decrease in the frequency of infusions.
Collapse
|
22
|
Treatment of neuromyelitis optica with rituximab: a 2-year prospective multicenter study. J Neurol 2018; 265:917-925. [PMID: 29455361 DOI: 10.1007/s00415-018-8771-5] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2017] [Revised: 01/26/2018] [Accepted: 01/27/2018] [Indexed: 01/06/2023]
Abstract
OBJECTIVE Neuromyelitis optica (NMO) is a very severe autoimmune disorder of the central nervous system. It affects young subjects and has a poor prognosis both on a functional and vital level. Therefore, it is imperative to reduce the frequency of relapses. The purpose of this study was to evaluate the clinical and neuroradiological effectiveness of rituximab (RTX) on active forms of NMO. METHODS We conducted a 2-year open prospective multicenter study that included 32 patients treated with RTX at a dose of 375 mg/m2/week for 1 month. When the number of circulating CD19+ B cells reached 1%, a maintenance therapy was started, consisting of two infusions of 1 g of RTX, administered at a 15-day interval. The primary objective was to reduce the annual relapse rate (ARR), in comparison to that observed in the 2 years before treatment onset. RESULTS Rituximab administration reduced the ARR from 1.34 to 0.56 (p = 0.0005). The average Expanded Disability Status Scale (EDSS) score significantly improved by 1.1 point, from 5.9 (2-9) to 4.8 (0-9) after 2 years (p = 0.03). Anti-aquaporin-4 antibodies' level predicted treatment failure (p = 0.03). Frequency of Gad+ lesions in spinal cord decreased from 23.3 to 14.2%. RTX treatment did not prevent the death of three patients (treatment failure in two patients and acute myeloid leukemia in a patient previously treated with mitoxantrone). CONCLUSION Rituximab is clinically effective in active forms of NMO, although few patients are resistant to the treatment.
Collapse
|
23
|
Early relapse after RTX initiation in a patient with NMO/MS overlap syndrome: How long to conclude to a failure treatment? Mult Scler Relat Disord 2018; 20:220-222. [DOI: 10.1016/j.msard.2018.02.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2018] [Accepted: 02/03/2018] [Indexed: 11/22/2022]
|
24
|
Miyazaki Y, Niino M, Takahashi E, Suzuki M, Mizuno M, Hisahara S, Fukazawa T, Amino I, Nakano F, Nakamura M, Akimoto S, Minami N, Fujiki N, Doi S, Shimohama S, Terayama Y, Kikuchi S. Fingolimod induces BAFF and expands circulating transitional B cells without activating memory B cells and plasma cells in multiple sclerosis. Clin Immunol 2018; 187:95-101. [DOI: 10.1016/j.clim.2017.10.009] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2017] [Revised: 09/15/2017] [Accepted: 10/23/2017] [Indexed: 01/01/2023]
|
25
|
Abstract
PURPOSE OF REVIEW This article provides a practical approach for providers caring for patients with neuromyelitis optica (NMO) spectrum disorders. Clinical and imaging features, diagnostic criteria, treatment of acute exacerbations, chronic preventive therapy, and symptom management in NMO spectrum disorders are discussed. RECENT FINDINGS The rapid pace of research in NMO spectrum disorders has led to many recent advances. A broader understanding of the clinical spectrum of the disease as well as improvements in anti-aquaporin-4 antibody assays have led to recent revision of the diagnostic criteria. Several recent studies have expanded the knowledge base regarding the efficacy and safety of current therapies for NMO spectrum disorders. SUMMARY An NMO spectrum disorder is an inflammatory disorder affecting the central nervous system, previously thought to be closely related to multiple sclerosis but more recently demonstrated to represent a distinct clinical and pathophysiologic entity. As NMO spectrum disorders carry significant morbidity and, at times, mortality, prompt and accurate diagnosis followed by swift initiation of therapy for both treatment of acute exacerbations and prevention of further relapses is critical. This article provides a practical approach to the diagnosis and management of NMO spectrum disorders.
Collapse
|
26
|
Shosha E, Pittock SJ, Flanagan E, Weinshenker BG. Neuromyelitis optica spectrum disorders and pregnancy: Interactions and management. Mult Scler 2017; 23:1808-1817. [DOI: 10.1177/1352458517740215] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Neuromyelitis optica spectrum disorders (NMOSD) predominantly affect women who are of childbearing age. Understanding the interactions between pregnancy and NMOSD is important for clinical management. Aquaporin-4 (AQP4), the most common target antigen in NMOSD, is expressed on placenta in early pregnancy. A variety of immune and cytokine changes in pregnancy may impact pregnancy outcomes in NMOSD patients. Relapses continue during pregnancy and increase in frequency postpartum. Preeclampsia and fetal loss are more frequent in NMOSD than in controls. Transfer of AQP4-immunoglobulin G (IgG) from mother to baby occurs but appears not to cause disease. Several treatment options are relatively safe and mitigate the risk of relapse during pregnancy and postpartum. For patients with active NMOSD, it may be advisable to continue immunotherapy during pregnancy.
Collapse
Affiliation(s)
- Eslam Shosha
- Department of Neurology, Mayo Clinic, Rochester, MN, USA/College of Medicine, Al Majmaah University, Riyadh, Saudi Arabia
| | - Sean J Pittock
- Department of Neurology, Mayo Clinic, Rochester, MN, USA/ Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
| | - Eoin Flanagan
- Department of Neurology, Mayo Clinic, Rochester, MN, USA/ Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
| | | |
Collapse
|
27
|
Zabad RK, Stewart R, Healey KM. Pattern Recognition of the Multiple Sclerosis Syndrome. Brain Sci 2017; 7:brainsci7100138. [PMID: 29064441 PMCID: PMC5664065 DOI: 10.3390/brainsci7100138] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2017] [Revised: 09/29/2017] [Accepted: 10/17/2017] [Indexed: 12/13/2022] Open
Abstract
During recent decades, the autoimmune disease neuromyelitis optica spectrum disorder (NMOSD), once broadly classified under the umbrella of multiple sclerosis (MS), has been extended to include autoimmune inflammatory conditions of the central nervous system (CNS), which are now diagnosable with serum serological tests. These antibody-mediated inflammatory diseases of the CNS share a clinical presentation to MS. A number of practical learning points emerge in this review, which is geared toward the pattern recognition of optic neuritis, transverse myelitis, brainstem/cerebellar and hemispheric tumefactive demyelinating lesion (TDL)-associated MS, aquaporin-4-antibody and myelin oligodendrocyte glycoprotein (MOG)-antibody NMOSD, overlap syndrome, and some yet-to-be-defined/classified demyelinating disease, all unspecifically labeled under MS syndrome. The goal of this review is to increase clinicians’ awareness of the clinical nuances of the autoimmune conditions for MS and NMSOD, and to highlight highly suggestive patterns of clinical, paraclinical or imaging presentations in order to improve differentiation. With overlay in clinical manifestations between MS and NMOSD, magnetic resonance imaging (MRI) of the brain, orbits and spinal cord, serology, and most importantly, high index of suspicion based on pattern recognition, will help lead to the final diagnosis.
Collapse
Affiliation(s)
- Rana K Zabad
- Department of Neurological Sciences, University of Nebraska Medical Center College of Medicine, Omaha, NE 68198-8440, USA.
| | - Renee Stewart
- University of Nebraska Medical Center College of Nursing, Omaha, NE 68198-5330, USA.
| | - Kathleen M Healey
- Department of Neurological Sciences, University of Nebraska Medical Center College of Medicine, Omaha, NE 68198-8440, USA.
| |
Collapse
|
28
|
Dumitrascu OM, McKeon A, Zuniga L, Grill MF, Goodman BP. Opsoclonus-Myoclonus Syndrome during Rituximab Treatment for Autoimmune Autonomic Ganglionopathy. NEUROLOGY-NEUROIMMUNOLOGY & NEUROINFLAMMATION 2017; 4:e376. [PMID: 28740856 PMCID: PMC5507190 DOI: 10.1212/nxi.0000000000000376] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 02/03/2017] [Accepted: 03/23/2017] [Indexed: 01/04/2023]
Affiliation(s)
- Oana M Dumitrascu
- Department of Neurology (O.M.D., L.Z., M.F.G., B.P.G.), Mayo Clinic, Phoenix, AZ; and Department of Neurology (A.M.), Mayo Clinic, Rochester, MN
| | - Andrew McKeon
- Department of Neurology (O.M.D., L.Z., M.F.G., B.P.G.), Mayo Clinic, Phoenix, AZ; and Department of Neurology (A.M.), Mayo Clinic, Rochester, MN
| | - Leslie Zuniga
- Department of Neurology (O.M.D., L.Z., M.F.G., B.P.G.), Mayo Clinic, Phoenix, AZ; and Department of Neurology (A.M.), Mayo Clinic, Rochester, MN
| | - Marie F Grill
- Department of Neurology (O.M.D., L.Z., M.F.G., B.P.G.), Mayo Clinic, Phoenix, AZ; and Department of Neurology (A.M.), Mayo Clinic, Rochester, MN
| | - Brent P Goodman
- Department of Neurology (O.M.D., L.Z., M.F.G., B.P.G.), Mayo Clinic, Phoenix, AZ; and Department of Neurology (A.M.), Mayo Clinic, Rochester, MN
| |
Collapse
|
29
|
Araki M, Yamamura T. Neuromyelitis optica spectrum disorders: Emerging therapies. ACTA ACUST UNITED AC 2017. [DOI: 10.1111/cen3.12394] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Affiliation(s)
- Manabu Araki
- Multiple Sclerosis Center; National Institute of Neuroscience; National Center of Neurology and Psychiatry; Tokyo Japan
- Department of Immunology; National Institute of Neuroscience; National Center of Neurology and Psychiatry; Tokyo Japan
| | - Takashi Yamamura
- Multiple Sclerosis Center; National Institute of Neuroscience; National Center of Neurology and Psychiatry; Tokyo Japan
- Department of Immunology; National Institute of Neuroscience; National Center of Neurology and Psychiatry; Tokyo Japan
| |
Collapse
|
30
|
Kowarik MC, Astling D, Gasperi C, Wemlinger S, Schumann H, Dzieciatkowska M, Ritchie AM, Hemmer B, Owens GP, Bennett JL. CNS Aquaporin-4-specific B cells connect with multiple B-cell compartments in neuromyelitis optica spectrum disorder. Ann Clin Transl Neurol 2017; 4:369-380. [PMID: 28589164 PMCID: PMC5454399 DOI: 10.1002/acn3.418] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Revised: 03/25/2017] [Accepted: 03/28/2017] [Indexed: 12/29/2022] Open
Abstract
Objectives Neuromyelitis optica spectrum disorder (NMOSD) is a severe inflammatory disorder of the central nervous system (CNS) targeted against aquaporin‐4 (AQP4). The origin and trafficking of AQP4‐specific B cells in NMOSD remains unknown. Methods Peripheral (n = 7) and splenic B cells (n = 1) recovered from seven NMOSD patients were sorted into plasmablasts, naïve, memory, and CD27‐IgD‐ double negative (DN) B cells, and variable heavy chain (VH) transcriptome sequences were generated by deep sequencing. Peripheral blood (PB) VH repertoires were compared to the same patient's single‐cell cerebrospinal fluid (CSF) plasmablast (PB) VH transcriptome, CSF immunoglobulin (Ig) proteome, and serum Ig proteome. Recombinant antibodies were generated from paired CSF heavy‐ and light chains and tested for AQP4 reactivity. Results Approximately 9% of the CSF VH sequences aligned with PB memory B cells, DN B cells, and plasmablast VH sequences. AQP4‐specific VH sequences were observed in each peripheral B‐cell compartment. Lineage analysis of clonally related VH sequences indicates that CSF AQP4‐specific B cells are closely related to an expanded population of DN B cells that may undergo antigen‐specific B‐cell maturation within the CNS. CSF and serum Ig proteomes overlapped with the VH sequences from each B‐cell compartment; the majority of matches occurring between the PB VH sequences and serum Ig proteome. Interpretation During an acute NMOSD relapse, a dynamic exchange of B cells occurs between the periphery and CNS with AQP4‐specific CSF B cells emerging from postgerminal center memory B cells and plasmablasts. Expansion of the PB DN B‐cell compartment may be a potential biomarker of NMOSD activity.
Collapse
Affiliation(s)
- Markus C Kowarik
- Department of Neurology Klinikum rechts der Isar der Technischen Universität MünchenIsmaninger Str. 22 Munich 81675 Germany.,Munich Cluster for Systems Neurology (SyNergy) Munich Germany
| | - David Astling
- Biochemistry and Molecular Genetics University of Colorado Denver Colorado
| | - Christiane Gasperi
- Department of Neurology Klinikum rechts der Isar der Technischen Universität MünchenIsmaninger Str. 22 Munich 81675 Germany
| | - Scott Wemlinger
- Department of Neurology University of Colorado Denver Colorado
| | - Hannah Schumann
- Department of Neurology University of Colorado Denver Colorado
| | | | | | - Bernhard Hemmer
- Department of Neurology Klinikum rechts der Isar der Technischen Universität MünchenIsmaninger Str. 22 Munich 81675 Germany.,Munich Cluster for Systems Neurology (SyNergy) Munich Germany.,German Competence Network Multiple Sclerosis Einstein str. 1 Munich 81675 Germany
| | - Gregory P Owens
- Department of Neurology University of Colorado Denver Colorado
| | - Jeffrey L Bennett
- Department of Neurology University of Colorado Denver Colorado.,Department of Ophthalmology University of Colorado Denver Colorado.,Program in Neuroscience University of Colorado Denver Colorado
| |
Collapse
|
31
|
Affiliation(s)
- Aravind Ganesh
- Department of Clinical Neurosciences, University of Calgary, Canada; and Centre for Prevention of Stroke and Dementia, Nuffield Department of Clinical Neurosciences, University of Oxford, UK
| |
Collapse
|
32
|
Jarius S, Kleiter I, Ruprecht K, Asgari N, Pitarokoili K, Borisow N, Hümmert MW, Trebst C, Pache F, Winkelmann A, Beume LA, Ringelstein M, Stich O, Aktas O, Korporal-Kuhnke M, Schwarz A, Lukas C, Haas J, Fechner K, Buttmann M, Bellmann-Strobl J, Zimmermann H, Brandt AU, Franciotta D, Schanda K, Paul F, Reindl M, Wildemann B. MOG-IgG in NMO and related disorders: a multicenter study of 50 patients. Part 3: Brainstem involvement - frequency, presentation and outcome. J Neuroinflammation 2016; 13:281. [PMID: 27802825 PMCID: PMC5088671 DOI: 10.1186/s12974-016-0719-z] [Citation(s) in RCA: 168] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2016] [Accepted: 09/09/2016] [Indexed: 11/17/2022] Open
Abstract
Background Myelin oligodendrocyte glycoprotein antibodies (MOG-IgG) are present in a subset of aquaporin-4 (AQP4)-IgG-negative patients with optic neuritis (ON) and/or myelitis. Little is known so far about brainstem involvement in MOG-IgG-positive patients. Objective To investigate the frequency, clinical and paraclinical features, course, outcome, and prognostic implications of brainstem involvement in MOG-IgG-positive ON and/or myelitis. Methods Retrospective case study. Results Among 50 patients with MOG-IgG-positive ON and/or myelitis, 15 (30 %) with a history of brainstem encephalitis were identified. All were negative for AQP4-IgG. Symptoms included respiratory insufficiency, intractable nausea and vomiting (INV), dysarthria, dysphagia, impaired cough reflex, oculomotor nerve palsy and diplopia, nystagmus, internuclear ophthalmoplegia (INO), facial nerve paresis, trigeminal hypesthesia/dysesthesia, vertigo, hearing loss, balance difficulties, and gait and limb ataxia; brainstem involvement was asymptomatic in three cases. Brainstem inflammation was already present at or very shortly after disease onset in 7/15 (47 %) patients. 16/21 (76.2 %) brainstem attacks were accompanied by acute myelitis and/or ON. Lesions were located in the pons (11/13), medulla oblongata (8/14), mesencephalon (cerebral peduncles; 2/14), and cerebellar peduncles (5/14), were adjacent to the fourth ventricle in 2/12, and periaqueductal in 1/12; some had concomitant diencephalic (2/13) or cerebellar lesions (1/14). MRI or laboratory signs of blood-brain barrier damage were present in 5/12. Cerebrospinal fluid pleocytosis was found in 11/14 cases, with neutrophils in 7/11 (3-34 % of all CSF white blood cells), and oligoclonal bands in 4/14. Attacks were preceded by acute infection or vaccination in 5/15 (33.3 %). A history of teratoma was noted in one case. The disease followed a relapsing course in 13/15 (87 %); the brainstem was involved more than once in 6. Immunosuppression was not always effective in preventing relapses. Interferon-beta was followed by new attacks in two patients. While one patient died from central hypoventilation, partial or complete recovery was achieved in the remainder following treatment with high-dose steroids and/or plasma exchange. Brainstem involvement was associated with a more aggressive general disease course (higher relapse rate, more myelitis attacks, more frequently supratentorial brain lesions, worse EDSS at last follow-up). Conclusions Brainstem involvement is present in around one third of MOG-IgG-positive patients with ON and/or myelitis. Clinical manifestations are diverse and may include symptoms typically seen in AQP4-IgG-positive neuromyelitis optica, such as INV and respiratory insufficiency, or in multiple sclerosis, such as INO. As MOG-IgG-positive brainstem encephalitis may take a serious or even fatal course, particular attention should be paid to signs or symptoms of additional brainstem involvement in patients presenting with MOG-IgG-positive ON and/or myelitis.
Collapse
Affiliation(s)
- Sven Jarius
- Molecular Neuroimmunology Group, Department of Neurology, University Hospital Heidelberg, Otto Meyerhof Center, Im Neuenheimer Feld 350, 69120, Heidelberg, Germany.
| | - Ingo Kleiter
- Department of Neurology, Ruhr University Bochum, Bochum, Germany
| | - Klemens Ruprecht
- Department of Neurology, Charité - University Medicine Berlin, Berlin, Germany
| | - Nasrin Asgari
- Department of Neurology and Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | | | - Nadja Borisow
- Department of Neurology, NeuroCure Clinical Research Center and Clinical and Experimental Multiple Sclerosis Research Center, Charité University Medicine, Berlin, Germany.,Experimental and Clinical Research Center, Max Delbrueck Center for Molecular Medicine, Berlin, Germany
| | - Martin W Hümmert
- Department of Neurology, Hannover Medical School, Hannover, Germany
| | - Corinna Trebst
- Department of Neurology, Hannover Medical School, Hannover, Germany
| | - Florence Pache
- Department of Neurology, NeuroCure Clinical Research Center and Clinical and Experimental Multiple Sclerosis Research Center, Charité University Medicine, Berlin, Germany.,Experimental and Clinical Research Center, Max Delbrueck Center for Molecular Medicine, Berlin, Germany
| | | | | | | | - Oliver Stich
- Department of Neurology, Albert Ludwigs University, Freiburg, Germany
| | - Orhan Aktas
- Department of Neurology, Heinrich Heine University, Düsseldorf, Germany
| | - Mirjam Korporal-Kuhnke
- Molecular Neuroimmunology Group, Department of Neurology, University Hospital Heidelberg, Otto Meyerhof Center, Im Neuenheimer Feld 350, 69120, Heidelberg, Germany
| | - Alexander Schwarz
- Molecular Neuroimmunology Group, Department of Neurology, University Hospital Heidelberg, Otto Meyerhof Center, Im Neuenheimer Feld 350, 69120, Heidelberg, Germany
| | - Carsten Lukas
- Department of Neuroradiology, Ruhr University Bochum, Bochum, Germany
| | - Jürgen Haas
- Molecular Neuroimmunology Group, Department of Neurology, University Hospital Heidelberg, Otto Meyerhof Center, Im Neuenheimer Feld 350, 69120, Heidelberg, Germany
| | - Kai Fechner
- Institute of Experimental Immunology, affiliated to Euroimmun AG, Lübeck, Germany
| | - Mathias Buttmann
- Department of Neurology, Julius Maximilians University, Würzburg, Germany
| | - Judith Bellmann-Strobl
- Department of Neurology, NeuroCure Clinical Research Center and Clinical and Experimental Multiple Sclerosis Research Center, Charité University Medicine, Berlin, Germany.,Experimental and Clinical Research Center, Max Delbrueck Center for Molecular Medicine, Berlin, Germany
| | - Hanna Zimmermann
- Department of Neurology, Charité - University Medicine Berlin, Berlin, Germany
| | - Alexander U Brandt
- Department of Neurology, Charité - University Medicine Berlin, Berlin, Germany
| | | | - Kathrin Schanda
- Department of Neurology, Medical University Innsbruck, Innsbruck, Austria
| | - Friedemann Paul
- Department of Neurology, NeuroCure Clinical Research Center and Clinical and Experimental Multiple Sclerosis Research Center, Charité University Medicine, Berlin, Germany.,Experimental and Clinical Research Center, Max Delbrueck Center for Molecular Medicine, Berlin, Germany
| | - Markus Reindl
- Department of Neurology, Medical University Innsbruck, Innsbruck, Austria
| | - Brigitte Wildemann
- Molecular Neuroimmunology Group, Department of Neurology, University Hospital Heidelberg, Otto Meyerhof Center, Im Neuenheimer Feld 350, 69120, Heidelberg, Germany
| | | |
Collapse
|
33
|
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. [PMID: 27793206 PMCID: PMC5086042 DOI: 10.1186/s12974-016-0718-0] [Citation(s) in RCA: 638] [Impact Index Per Article: 79.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2016] [Accepted: 09/09/2016] [Indexed: 01/03/2023] Open
Abstract
BACKGROUND A subset of patients with neuromyelitis optica spectrum disorders (NMOSD) has been shown to be seropositive for myelin oligodendrocyte glycoprotein antibodies (MOG-IgG). OBJECTIVE To describe the epidemiological, clinical, radiological, cerebrospinal fluid (CSF), and electrophysiological features of a large cohort of MOG-IgG-positive patients with optic neuritis (ON) and/or myelitis (n = 50) as well as attack and long-term treatment outcomes. METHODS Retrospective multicenter study. RESULTS The sex ratio was 1:2.8 (m:f). Median age at onset was 31 years (range 6-70). The disease followed a multiphasic course in 80 % (median time-to-first-relapse 5 months; annualized relapse rate 0.92) and resulted in significant disability in 40 % (mean follow-up 75 ± 46.5 months), with severe visual impairment or functional blindness (36 %) and markedly impaired ambulation due to paresis or ataxia (25 %) as the most common long-term sequelae. Functional blindess in one or both eyes was noted during at least one ON attack in around 70 %. Perioptic enhancement was present in several patients. Besides acute tetra-/paraparesis, dysesthesia and pain were common in acute myelitis (70 %). Longitudinally extensive spinal cord lesions were frequent, but short lesions occurred at least once in 44 %. Fourty-one percent had a history of simultaneous ON and myelitis. Clinical or radiological involvement of the brain, brainstem, or cerebellum was present in 50 %; extra-opticospinal symptoms included intractable nausea and vomiting and respiratory insufficiency (fatal in one). CSF pleocytosis (partly neutrophilic) was present in 70 %, oligoclonal bands in only 13 %, and blood-CSF-barrier dysfunction in 32 %. Intravenous methylprednisolone (IVMP) and long-term immunosuppression were often effective; however, treatment failure leading to rapid accumulation of disability was noted in many patients as well as flare-ups after steroid withdrawal. Full recovery was achieved by plasma exchange in some cases, including after IVMP failure. Breakthrough attacks under azathioprine were linked to the drug-specific latency period and a lack of cotreatment with oral steroids. Methotrexate was effective in 5/6 patients. Interferon-beta was associated with ongoing or increasing disease activity. Rituximab and ofatumumab were effective in some patients. However, treatment with rituximab was followed by early relapses in several cases; end-of-dose relapses occurred 9-12 months after the first infusion. Coexisting autoimmunity was rare (9 %). Wingerchuk's 2006 and 2015 criteria for NMO(SD) and Barkhof and McDonald criteria for multiple sclerosis (MS) were met by 28 %, 32 %, 15 %, 33 %, respectively; MS had been suspected in 36 %. Disease onset or relapses were preceded by infection, vaccination, or pregnancy/delivery in several cases. CONCLUSION Our findings from a predominantly Caucasian cohort strongly argue against the concept of MOG-IgG denoting a mild and usually monophasic variant of NMOSD. The predominantly relapsing and often severe disease course and the short median time to second attack support the use of prophylactic long-term treatments in patients with MOG-IgG-positive ON and/or myelitis.
Collapse
|
34
|
Collongues N, de Seze J. An update on the evidence for the efficacy and safety of rituximab in the management of neuromyelitis optica. Ther Adv Neurol Disord 2016; 9:180-8. [PMID: 27134673 PMCID: PMC4811013 DOI: 10.1177/1756285616632653] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
Neuromyelitis optica spectrum disorders (NMOSDs) is a new concept which includes classical neuromyelitis optica (NMO) and partial forms of NMO such as recurrent optic neuritis with positive aquaporin-4 antibodies (AQP4) or brainstem symptoms (intractable hiccups or vomiting). This disease is clearly distinguished from multiple sclerosis (MS) and the therapeutic approach is clearly different. Rituximab is actually considered to be one of the most efficient treatments of NMOSD, even if class I studies are clearly lacking. In the present review, we describe the state of the art about rituximab treatment in NMOSD, including adults and children, plus its efficacy and tolerance and we also underline the questions that should be addressed in the near future.
Collapse
Affiliation(s)
- Nicolas Collongues
- Department of Neurology, University Hospitals of Strasbourg, Strasbourg, France
| | - Jérôme de Seze
- Department of Neurology, Hôpital de Hautepierre, 1 avenue Molière, 67098, Strasbourg Cedex, France
| |
Collapse
|
35
|
Nosadini M, Alper G, Riney CJ, Benson LA, Mohammad SS, Ramanathan S, Nolan M, Appleton R, Leventer RJ, Deiva K, Brilot F, Gorman MP, Waldman AT, Banwell B, Dale RC. Rituximab monitoring and redosing in pediatric neuromyelitis optica spectrum disorder. NEUROLOGY-NEUROIMMUNOLOGY & NEUROINFLAMMATION 2016; 3:e188. [PMID: 26819962 PMCID: PMC4723136 DOI: 10.1212/nxi.0000000000000188] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/24/2015] [Accepted: 09/29/2015] [Indexed: 01/01/2023]
Abstract
Objective: To study rituximab in pediatric neuromyelitis optica (NMO)/NMO spectrum disorders (NMOSD) and the relationship between rituximab, B cell repopulation, and relapses in order to improve rituximab monitoring and redosing. Methods: Multicenter retrospective study of 16 children with NMO/NMOSD receiving ≥2 rituximab courses. According to CD19 counts, events during rituximab were categorized as “repopulation,” “depletion,” or “depletion failure” relapses (repopulation threshold CD19 ≥10 × 106 cells/L). Results: The 16 patients (14 girls; mean age 9.6 years, range 1.8–15.3) had a mean of 6.1 events (range 1–11) during a mean follow-up of 6.1 years (range 1.6–13.6) and received a total of 76 rituximab courses (mean 4.7, range 2–9) in 42.6-year cohort treatment. Before rituximab, 62.5% had received azathioprine, mycophenolate mofetil, or cyclophosphamide. Mean time from rituximab to last documented B cell depletion and first repopulation was 4.5 and 6.8 months, respectively, with large interpatient variability. Earliest repopulations occurred with the lowest doses. Significant reduction between pre- and post-rituximab annualized relapse rate (ARR) was observed (p = 0.003). During rituximab, 6 patients were relapse-free, although 21 relapses occurred in 10 patients, including 13 “repopulation,” 3 “depletion,” and 4 “depletion failure” relapses. Of the 13 “repopulation” relapses, 4 had CD19 10–50 × 106 cells/L, 10 had inadequate monitoring (≤1 CD19 in the 4 months before relapses), and 5 had delayed redosing after repopulation detection. Conclusion: Rituximab is effective in relapse prevention, but B cell repopulation creates a risk of relapse. Redosing before B cell repopulation could reduce the relapse risk further. Classification of evidence: This study provides Class IV evidence that rituximab significantly reduces ARR in pediatric NMO/NMOSD. This study also demonstrates a relationship between B cell repopulation and relapses.
Collapse
Affiliation(s)
- Margherita Nosadini
- Neuroimmunology Group (M. Nosadini, S.S.M., S.R., F.B., R.C.D.), Institute for Neuroscience and Muscle Research, Children's Hospital at Westmead, University of Sydney, Australia; Paediatric Neurology Unit (M. Nosadini), Department of Paediatrics, University of Padua, Italy; Clinical Neuroimmunology Program (G.A.), Division of Child Neurology, Department of Pediatrics, Children's Hospital of Pittsburgh, University of Pittsburgh, PA; Neurology Department (C.J.R.), Lady Cilento Children's Hospital, University of Queensland, Australia; Pediatric Multiple Sclerosis and Related Diseases Program (L.A.B., M.P.G.), Boston Children's Hospital, Boston, MA; Department of Neurology (S.R.), Westmead Hospital, Sydney, Australia; Neurology Department (M. Nolan), Starship Children's Health, Auckland, New Zealand; The Roald Dahl EEG Unit (R.A.), Pediatric Neurosciences Foundation, Alder Hey Children's Hospital, Liverpool, UK; Department of Neurology (R.J.L.), Murdoch Childrens Research Institute and University of Melbourne Department of Paediatrics (R.J.L.), Royal Children's Hospital, Melbourne, Victoria, Australia; Assistance Publique-Hopitaux de Paris (K.D.), Hôpitaux Universitaires Paris-Sud, National Referral Center for Neuro-Inflammatory Diseases in Children (K.D.), Pediatric Neurology Department, and Université Paris-Sud (K.D.), Inserm U1012, Le Kremlin-Bicêtre, France; and Children's Hospital of Philadelphia (A.T.W., B.B.), University of Pennsylvania, Philadelphia
| | - Gulay Alper
- Neuroimmunology Group (M. Nosadini, S.S.M., S.R., F.B., R.C.D.), Institute for Neuroscience and Muscle Research, Children's Hospital at Westmead, University of Sydney, Australia; Paediatric Neurology Unit (M. Nosadini), Department of Paediatrics, University of Padua, Italy; Clinical Neuroimmunology Program (G.A.), Division of Child Neurology, Department of Pediatrics, Children's Hospital of Pittsburgh, University of Pittsburgh, PA; Neurology Department (C.J.R.), Lady Cilento Children's Hospital, University of Queensland, Australia; Pediatric Multiple Sclerosis and Related Diseases Program (L.A.B., M.P.G.), Boston Children's Hospital, Boston, MA; Department of Neurology (S.R.), Westmead Hospital, Sydney, Australia; Neurology Department (M. Nolan), Starship Children's Health, Auckland, New Zealand; The Roald Dahl EEG Unit (R.A.), Pediatric Neurosciences Foundation, Alder Hey Children's Hospital, Liverpool, UK; Department of Neurology (R.J.L.), Murdoch Childrens Research Institute and University of Melbourne Department of Paediatrics (R.J.L.), Royal Children's Hospital, Melbourne, Victoria, Australia; Assistance Publique-Hopitaux de Paris (K.D.), Hôpitaux Universitaires Paris-Sud, National Referral Center for Neuro-Inflammatory Diseases in Children (K.D.), Pediatric Neurology Department, and Université Paris-Sud (K.D.), Inserm U1012, Le Kremlin-Bicêtre, France; and Children's Hospital of Philadelphia (A.T.W., B.B.), University of Pennsylvania, Philadelphia
| | - Catherine J Riney
- Neuroimmunology Group (M. Nosadini, S.S.M., S.R., F.B., R.C.D.), Institute for Neuroscience and Muscle Research, Children's Hospital at Westmead, University of Sydney, Australia; Paediatric Neurology Unit (M. Nosadini), Department of Paediatrics, University of Padua, Italy; Clinical Neuroimmunology Program (G.A.), Division of Child Neurology, Department of Pediatrics, Children's Hospital of Pittsburgh, University of Pittsburgh, PA; Neurology Department (C.J.R.), Lady Cilento Children's Hospital, University of Queensland, Australia; Pediatric Multiple Sclerosis and Related Diseases Program (L.A.B., M.P.G.), Boston Children's Hospital, Boston, MA; Department of Neurology (S.R.), Westmead Hospital, Sydney, Australia; Neurology Department (M. Nolan), Starship Children's Health, Auckland, New Zealand; The Roald Dahl EEG Unit (R.A.), Pediatric Neurosciences Foundation, Alder Hey Children's Hospital, Liverpool, UK; Department of Neurology (R.J.L.), Murdoch Childrens Research Institute and University of Melbourne Department of Paediatrics (R.J.L.), Royal Children's Hospital, Melbourne, Victoria, Australia; Assistance Publique-Hopitaux de Paris (K.D.), Hôpitaux Universitaires Paris-Sud, National Referral Center for Neuro-Inflammatory Diseases in Children (K.D.), Pediatric Neurology Department, and Université Paris-Sud (K.D.), Inserm U1012, Le Kremlin-Bicêtre, France; and Children's Hospital of Philadelphia (A.T.W., B.B.), University of Pennsylvania, Philadelphia
| | - Leslie A Benson
- Neuroimmunology Group (M. Nosadini, S.S.M., S.R., F.B., R.C.D.), Institute for Neuroscience and Muscle Research, Children's Hospital at Westmead, University of Sydney, Australia; Paediatric Neurology Unit (M. Nosadini), Department of Paediatrics, University of Padua, Italy; Clinical Neuroimmunology Program (G.A.), Division of Child Neurology, Department of Pediatrics, Children's Hospital of Pittsburgh, University of Pittsburgh, PA; Neurology Department (C.J.R.), Lady Cilento Children's Hospital, University of Queensland, Australia; Pediatric Multiple Sclerosis and Related Diseases Program (L.A.B., M.P.G.), Boston Children's Hospital, Boston, MA; Department of Neurology (S.R.), Westmead Hospital, Sydney, Australia; Neurology Department (M. Nolan), Starship Children's Health, Auckland, New Zealand; The Roald Dahl EEG Unit (R.A.), Pediatric Neurosciences Foundation, Alder Hey Children's Hospital, Liverpool, UK; Department of Neurology (R.J.L.), Murdoch Childrens Research Institute and University of Melbourne Department of Paediatrics (R.J.L.), Royal Children's Hospital, Melbourne, Victoria, Australia; Assistance Publique-Hopitaux de Paris (K.D.), Hôpitaux Universitaires Paris-Sud, National Referral Center for Neuro-Inflammatory Diseases in Children (K.D.), Pediatric Neurology Department, and Université Paris-Sud (K.D.), Inserm U1012, Le Kremlin-Bicêtre, France; and Children's Hospital of Philadelphia (A.T.W., B.B.), University of Pennsylvania, Philadelphia
| | - Shekeeb S Mohammad
- Neuroimmunology Group (M. Nosadini, S.S.M., S.R., F.B., R.C.D.), Institute for Neuroscience and Muscle Research, Children's Hospital at Westmead, University of Sydney, Australia; Paediatric Neurology Unit (M. Nosadini), Department of Paediatrics, University of Padua, Italy; Clinical Neuroimmunology Program (G.A.), Division of Child Neurology, Department of Pediatrics, Children's Hospital of Pittsburgh, University of Pittsburgh, PA; Neurology Department (C.J.R.), Lady Cilento Children's Hospital, University of Queensland, Australia; Pediatric Multiple Sclerosis and Related Diseases Program (L.A.B., M.P.G.), Boston Children's Hospital, Boston, MA; Department of Neurology (S.R.), Westmead Hospital, Sydney, Australia; Neurology Department (M. Nolan), Starship Children's Health, Auckland, New Zealand; The Roald Dahl EEG Unit (R.A.), Pediatric Neurosciences Foundation, Alder Hey Children's Hospital, Liverpool, UK; Department of Neurology (R.J.L.), Murdoch Childrens Research Institute and University of Melbourne Department of Paediatrics (R.J.L.), Royal Children's Hospital, Melbourne, Victoria, Australia; Assistance Publique-Hopitaux de Paris (K.D.), Hôpitaux Universitaires Paris-Sud, National Referral Center for Neuro-Inflammatory Diseases in Children (K.D.), Pediatric Neurology Department, and Université Paris-Sud (K.D.), Inserm U1012, Le Kremlin-Bicêtre, France; and Children's Hospital of Philadelphia (A.T.W., B.B.), University of Pennsylvania, Philadelphia
| | - Sudarshini Ramanathan
- Neuroimmunology Group (M. Nosadini, S.S.M., S.R., F.B., R.C.D.), Institute for Neuroscience and Muscle Research, Children's Hospital at Westmead, University of Sydney, Australia; Paediatric Neurology Unit (M. Nosadini), Department of Paediatrics, University of Padua, Italy; Clinical Neuroimmunology Program (G.A.), Division of Child Neurology, Department of Pediatrics, Children's Hospital of Pittsburgh, University of Pittsburgh, PA; Neurology Department (C.J.R.), Lady Cilento Children's Hospital, University of Queensland, Australia; Pediatric Multiple Sclerosis and Related Diseases Program (L.A.B., M.P.G.), Boston Children's Hospital, Boston, MA; Department of Neurology (S.R.), Westmead Hospital, Sydney, Australia; Neurology Department (M. Nolan), Starship Children's Health, Auckland, New Zealand; The Roald Dahl EEG Unit (R.A.), Pediatric Neurosciences Foundation, Alder Hey Children's Hospital, Liverpool, UK; Department of Neurology (R.J.L.), Murdoch Childrens Research Institute and University of Melbourne Department of Paediatrics (R.J.L.), Royal Children's Hospital, Melbourne, Victoria, Australia; Assistance Publique-Hopitaux de Paris (K.D.), Hôpitaux Universitaires Paris-Sud, National Referral Center for Neuro-Inflammatory Diseases in Children (K.D.), Pediatric Neurology Department, and Université Paris-Sud (K.D.), Inserm U1012, Le Kremlin-Bicêtre, France; and Children's Hospital of Philadelphia (A.T.W., B.B.), University of Pennsylvania, Philadelphia
| | - Melinda Nolan
- Neuroimmunology Group (M. Nosadini, S.S.M., S.R., F.B., R.C.D.), Institute for Neuroscience and Muscle Research, Children's Hospital at Westmead, University of Sydney, Australia; Paediatric Neurology Unit (M. Nosadini), Department of Paediatrics, University of Padua, Italy; Clinical Neuroimmunology Program (G.A.), Division of Child Neurology, Department of Pediatrics, Children's Hospital of Pittsburgh, University of Pittsburgh, PA; Neurology Department (C.J.R.), Lady Cilento Children's Hospital, University of Queensland, Australia; Pediatric Multiple Sclerosis and Related Diseases Program (L.A.B., M.P.G.), Boston Children's Hospital, Boston, MA; Department of Neurology (S.R.), Westmead Hospital, Sydney, Australia; Neurology Department (M. Nolan), Starship Children's Health, Auckland, New Zealand; The Roald Dahl EEG Unit (R.A.), Pediatric Neurosciences Foundation, Alder Hey Children's Hospital, Liverpool, UK; Department of Neurology (R.J.L.), Murdoch Childrens Research Institute and University of Melbourne Department of Paediatrics (R.J.L.), Royal Children's Hospital, Melbourne, Victoria, Australia; Assistance Publique-Hopitaux de Paris (K.D.), Hôpitaux Universitaires Paris-Sud, National Referral Center for Neuro-Inflammatory Diseases in Children (K.D.), Pediatric Neurology Department, and Université Paris-Sud (K.D.), Inserm U1012, Le Kremlin-Bicêtre, France; and Children's Hospital of Philadelphia (A.T.W., B.B.), University of Pennsylvania, Philadelphia
| | - Richard Appleton
- Neuroimmunology Group (M. Nosadini, S.S.M., S.R., F.B., R.C.D.), Institute for Neuroscience and Muscle Research, Children's Hospital at Westmead, University of Sydney, Australia; Paediatric Neurology Unit (M. Nosadini), Department of Paediatrics, University of Padua, Italy; Clinical Neuroimmunology Program (G.A.), Division of Child Neurology, Department of Pediatrics, Children's Hospital of Pittsburgh, University of Pittsburgh, PA; Neurology Department (C.J.R.), Lady Cilento Children's Hospital, University of Queensland, Australia; Pediatric Multiple Sclerosis and Related Diseases Program (L.A.B., M.P.G.), Boston Children's Hospital, Boston, MA; Department of Neurology (S.R.), Westmead Hospital, Sydney, Australia; Neurology Department (M. Nolan), Starship Children's Health, Auckland, New Zealand; The Roald Dahl EEG Unit (R.A.), Pediatric Neurosciences Foundation, Alder Hey Children's Hospital, Liverpool, UK; Department of Neurology (R.J.L.), Murdoch Childrens Research Institute and University of Melbourne Department of Paediatrics (R.J.L.), Royal Children's Hospital, Melbourne, Victoria, Australia; Assistance Publique-Hopitaux de Paris (K.D.), Hôpitaux Universitaires Paris-Sud, National Referral Center for Neuro-Inflammatory Diseases in Children (K.D.), Pediatric Neurology Department, and Université Paris-Sud (K.D.), Inserm U1012, Le Kremlin-Bicêtre, France; and Children's Hospital of Philadelphia (A.T.W., B.B.), University of Pennsylvania, Philadelphia
| | - Richard J Leventer
- Neuroimmunology Group (M. Nosadini, S.S.M., S.R., F.B., R.C.D.), Institute for Neuroscience and Muscle Research, Children's Hospital at Westmead, University of Sydney, Australia; Paediatric Neurology Unit (M. Nosadini), Department of Paediatrics, University of Padua, Italy; Clinical Neuroimmunology Program (G.A.), Division of Child Neurology, Department of Pediatrics, Children's Hospital of Pittsburgh, University of Pittsburgh, PA; Neurology Department (C.J.R.), Lady Cilento Children's Hospital, University of Queensland, Australia; Pediatric Multiple Sclerosis and Related Diseases Program (L.A.B., M.P.G.), Boston Children's Hospital, Boston, MA; Department of Neurology (S.R.), Westmead Hospital, Sydney, Australia; Neurology Department (M. Nolan), Starship Children's Health, Auckland, New Zealand; The Roald Dahl EEG Unit (R.A.), Pediatric Neurosciences Foundation, Alder Hey Children's Hospital, Liverpool, UK; Department of Neurology (R.J.L.), Murdoch Childrens Research Institute and University of Melbourne Department of Paediatrics (R.J.L.), Royal Children's Hospital, Melbourne, Victoria, Australia; Assistance Publique-Hopitaux de Paris (K.D.), Hôpitaux Universitaires Paris-Sud, National Referral Center for Neuro-Inflammatory Diseases in Children (K.D.), Pediatric Neurology Department, and Université Paris-Sud (K.D.), Inserm U1012, Le Kremlin-Bicêtre, France; and Children's Hospital of Philadelphia (A.T.W., B.B.), University of Pennsylvania, Philadelphia
| | - Kumaran Deiva
- Neuroimmunology Group (M. Nosadini, S.S.M., S.R., F.B., R.C.D.), Institute for Neuroscience and Muscle Research, Children's Hospital at Westmead, University of Sydney, Australia; Paediatric Neurology Unit (M. Nosadini), Department of Paediatrics, University of Padua, Italy; Clinical Neuroimmunology Program (G.A.), Division of Child Neurology, Department of Pediatrics, Children's Hospital of Pittsburgh, University of Pittsburgh, PA; Neurology Department (C.J.R.), Lady Cilento Children's Hospital, University of Queensland, Australia; Pediatric Multiple Sclerosis and Related Diseases Program (L.A.B., M.P.G.), Boston Children's Hospital, Boston, MA; Department of Neurology (S.R.), Westmead Hospital, Sydney, Australia; Neurology Department (M. Nolan), Starship Children's Health, Auckland, New Zealand; The Roald Dahl EEG Unit (R.A.), Pediatric Neurosciences Foundation, Alder Hey Children's Hospital, Liverpool, UK; Department of Neurology (R.J.L.), Murdoch Childrens Research Institute and University of Melbourne Department of Paediatrics (R.J.L.), Royal Children's Hospital, Melbourne, Victoria, Australia; Assistance Publique-Hopitaux de Paris (K.D.), Hôpitaux Universitaires Paris-Sud, National Referral Center for Neuro-Inflammatory Diseases in Children (K.D.), Pediatric Neurology Department, and Université Paris-Sud (K.D.), Inserm U1012, Le Kremlin-Bicêtre, France; and Children's Hospital of Philadelphia (A.T.W., B.B.), University of Pennsylvania, Philadelphia
| | - Fabienne Brilot
- Neuroimmunology Group (M. Nosadini, S.S.M., S.R., F.B., R.C.D.), Institute for Neuroscience and Muscle Research, Children's Hospital at Westmead, University of Sydney, Australia; Paediatric Neurology Unit (M. Nosadini), Department of Paediatrics, University of Padua, Italy; Clinical Neuroimmunology Program (G.A.), Division of Child Neurology, Department of Pediatrics, Children's Hospital of Pittsburgh, University of Pittsburgh, PA; Neurology Department (C.J.R.), Lady Cilento Children's Hospital, University of Queensland, Australia; Pediatric Multiple Sclerosis and Related Diseases Program (L.A.B., M.P.G.), Boston Children's Hospital, Boston, MA; Department of Neurology (S.R.), Westmead Hospital, Sydney, Australia; Neurology Department (M. Nolan), Starship Children's Health, Auckland, New Zealand; The Roald Dahl EEG Unit (R.A.), Pediatric Neurosciences Foundation, Alder Hey Children's Hospital, Liverpool, UK; Department of Neurology (R.J.L.), Murdoch Childrens Research Institute and University of Melbourne Department of Paediatrics (R.J.L.), Royal Children's Hospital, Melbourne, Victoria, Australia; Assistance Publique-Hopitaux de Paris (K.D.), Hôpitaux Universitaires Paris-Sud, National Referral Center for Neuro-Inflammatory Diseases in Children (K.D.), Pediatric Neurology Department, and Université Paris-Sud (K.D.), Inserm U1012, Le Kremlin-Bicêtre, France; and Children's Hospital of Philadelphia (A.T.W., B.B.), University of Pennsylvania, Philadelphia
| | - Mark P Gorman
- Neuroimmunology Group (M. Nosadini, S.S.M., S.R., F.B., R.C.D.), Institute for Neuroscience and Muscle Research, Children's Hospital at Westmead, University of Sydney, Australia; Paediatric Neurology Unit (M. Nosadini), Department of Paediatrics, University of Padua, Italy; Clinical Neuroimmunology Program (G.A.), Division of Child Neurology, Department of Pediatrics, Children's Hospital of Pittsburgh, University of Pittsburgh, PA; Neurology Department (C.J.R.), Lady Cilento Children's Hospital, University of Queensland, Australia; Pediatric Multiple Sclerosis and Related Diseases Program (L.A.B., M.P.G.), Boston Children's Hospital, Boston, MA; Department of Neurology (S.R.), Westmead Hospital, Sydney, Australia; Neurology Department (M. Nolan), Starship Children's Health, Auckland, New Zealand; The Roald Dahl EEG Unit (R.A.), Pediatric Neurosciences Foundation, Alder Hey Children's Hospital, Liverpool, UK; Department of Neurology (R.J.L.), Murdoch Childrens Research Institute and University of Melbourne Department of Paediatrics (R.J.L.), Royal Children's Hospital, Melbourne, Victoria, Australia; Assistance Publique-Hopitaux de Paris (K.D.), Hôpitaux Universitaires Paris-Sud, National Referral Center for Neuro-Inflammatory Diseases in Children (K.D.), Pediatric Neurology Department, and Université Paris-Sud (K.D.), Inserm U1012, Le Kremlin-Bicêtre, France; and Children's Hospital of Philadelphia (A.T.W., B.B.), University of Pennsylvania, Philadelphia
| | - Amy T Waldman
- Neuroimmunology Group (M. Nosadini, S.S.M., S.R., F.B., R.C.D.), Institute for Neuroscience and Muscle Research, Children's Hospital at Westmead, University of Sydney, Australia; Paediatric Neurology Unit (M. Nosadini), Department of Paediatrics, University of Padua, Italy; Clinical Neuroimmunology Program (G.A.), Division of Child Neurology, Department of Pediatrics, Children's Hospital of Pittsburgh, University of Pittsburgh, PA; Neurology Department (C.J.R.), Lady Cilento Children's Hospital, University of Queensland, Australia; Pediatric Multiple Sclerosis and Related Diseases Program (L.A.B., M.P.G.), Boston Children's Hospital, Boston, MA; Department of Neurology (S.R.), Westmead Hospital, Sydney, Australia; Neurology Department (M. Nolan), Starship Children's Health, Auckland, New Zealand; The Roald Dahl EEG Unit (R.A.), Pediatric Neurosciences Foundation, Alder Hey Children's Hospital, Liverpool, UK; Department of Neurology (R.J.L.), Murdoch Childrens Research Institute and University of Melbourne Department of Paediatrics (R.J.L.), Royal Children's Hospital, Melbourne, Victoria, Australia; Assistance Publique-Hopitaux de Paris (K.D.), Hôpitaux Universitaires Paris-Sud, National Referral Center for Neuro-Inflammatory Diseases in Children (K.D.), Pediatric Neurology Department, and Université Paris-Sud (K.D.), Inserm U1012, Le Kremlin-Bicêtre, France; and Children's Hospital of Philadelphia (A.T.W., B.B.), University of Pennsylvania, Philadelphia
| | - Brenda Banwell
- Neuroimmunology Group (M. Nosadini, S.S.M., S.R., F.B., R.C.D.), Institute for Neuroscience and Muscle Research, Children's Hospital at Westmead, University of Sydney, Australia; Paediatric Neurology Unit (M. Nosadini), Department of Paediatrics, University of Padua, Italy; Clinical Neuroimmunology Program (G.A.), Division of Child Neurology, Department of Pediatrics, Children's Hospital of Pittsburgh, University of Pittsburgh, PA; Neurology Department (C.J.R.), Lady Cilento Children's Hospital, University of Queensland, Australia; Pediatric Multiple Sclerosis and Related Diseases Program (L.A.B., M.P.G.), Boston Children's Hospital, Boston, MA; Department of Neurology (S.R.), Westmead Hospital, Sydney, Australia; Neurology Department (M. Nolan), Starship Children's Health, Auckland, New Zealand; The Roald Dahl EEG Unit (R.A.), Pediatric Neurosciences Foundation, Alder Hey Children's Hospital, Liverpool, UK; Department of Neurology (R.J.L.), Murdoch Childrens Research Institute and University of Melbourne Department of Paediatrics (R.J.L.), Royal Children's Hospital, Melbourne, Victoria, Australia; Assistance Publique-Hopitaux de Paris (K.D.), Hôpitaux Universitaires Paris-Sud, National Referral Center for Neuro-Inflammatory Diseases in Children (K.D.), Pediatric Neurology Department, and Université Paris-Sud (K.D.), Inserm U1012, Le Kremlin-Bicêtre, France; and Children's Hospital of Philadelphia (A.T.W., B.B.), University of Pennsylvania, Philadelphia
| | - Russell C Dale
- Neuroimmunology Group (M. Nosadini, S.S.M., S.R., F.B., R.C.D.), Institute for Neuroscience and Muscle Research, Children's Hospital at Westmead, University of Sydney, Australia; Paediatric Neurology Unit (M. Nosadini), Department of Paediatrics, University of Padua, Italy; Clinical Neuroimmunology Program (G.A.), Division of Child Neurology, Department of Pediatrics, Children's Hospital of Pittsburgh, University of Pittsburgh, PA; Neurology Department (C.J.R.), Lady Cilento Children's Hospital, University of Queensland, Australia; Pediatric Multiple Sclerosis and Related Diseases Program (L.A.B., M.P.G.), Boston Children's Hospital, Boston, MA; Department of Neurology (S.R.), Westmead Hospital, Sydney, Australia; Neurology Department (M. Nolan), Starship Children's Health, Auckland, New Zealand; The Roald Dahl EEG Unit (R.A.), Pediatric Neurosciences Foundation, Alder Hey Children's Hospital, Liverpool, UK; Department of Neurology (R.J.L.), Murdoch Childrens Research Institute and University of Melbourne Department of Paediatrics (R.J.L.), Royal Children's Hospital, Melbourne, Victoria, Australia; Assistance Publique-Hopitaux de Paris (K.D.), Hôpitaux Universitaires Paris-Sud, National Referral Center for Neuro-Inflammatory Diseases in Children (K.D.), Pediatric Neurology Department, and Université Paris-Sud (K.D.), Inserm U1012, Le Kremlin-Bicêtre, France; and Children's Hospital of Philadelphia (A.T.W., B.B.), University of Pennsylvania, Philadelphia
| |
Collapse
|
36
|
Abstract
Neuromyelitis optica spectrum disorders (NMOSD) are important evolving entities, which have reached much attention in the recent years. NMOSD are characterized by inflammatory lesions in the optic nerves, spinal cord, and central parts of the brain, as well as an autoimmune process directed against aquaporin-4. As disability in NMOSD accumulates by inflammatory damage from attacks, both the treatment and prevention of attacks are decisive for the long-term outcome. NMOSD attacks are treated with high-dose intravenous corticosteroids and apheresis therapies, in particular therapeutic plasma exchange. In cases of incomplete remission, escalation of attack treatment is recommended. Preventive therapy is immunosuppressive and should by commenced as early as possible. Apart from classical immunosuppressants such as azathioprine and mycophenolate mofetil, repurposed biologicals are increasingly used. B-cell depletion with rituximab and other agents, inhibition of the interleukin-6 receptor with tocilizumab, and blockade of complement-mediated damage by eculizumab all are promising therapeutic strategies evaluated in randomized controlled trials. In this review, we will discuss present and future immunotherapies for NMOSD and also consider combination of treatments, plasma, cellular and other therapies. Current advances in immunopathological knowledge are translated into innovative concepts and begin a new era of NMOSD therapy.
Collapse
Affiliation(s)
- Ingo Kleiter
- Department of Neurology, St. Josef-Hospital, Ruhr-University, Bochum, Germany.
| | - Ralf Gold
- Department of Neurology, St. Josef-Hospital, Ruhr-University, Bochum, Germany
| |
Collapse
|
37
|
Abstract
Autoimmune myelopathies are a heterogeneous group of immune-mediated spinal cord disorders with a broad differential diagnosis. They encompass myelopathies with an immune attack on the spinal cord (e.g., aquaporin-4-IgG (AQP4-IgG) seropositive neuromyelitis optica (NMO) and its spectrum disorders (NMOSD)), myelopathies occurring with systemic autoimmune disorders (which may also be due to coexisting NMO/NMOSD), paraneoplastic autoimmune myelopathies, postinfectious autoimmune myelopathies (e.g., acute disseminated encephalomyelitis), and myelopathies thought to be immune-related (e.g., multiple sclerosis and spinal cord sarcoidosis). Spine magnetic resonance imaging is extremely useful in the evaluation of autoimmune myelopathies as the location of signal change, length of the lesion, gadolinium enhancement pattern, and evolution over time narrow the differential diagnosis considerably. The recent discovery of multiple novel neural-specific autoantibodies accompanying autoimmune myelopathies has improved their classification. These autoantibodies may be pathogenic (e.g., AQP4-IgG) or nonpathogenic and more reflective of a cytotoxic T-cell-mediated autoimmune response (collapsin response mediator protein-5(CRMP5)-IgG). The presence of an autoantibody may help guide cancer search, assist treatment decisions, and predict outcome/relapse. With paraneoplastic myelopathies the initial goal is detection and treatment of the underlying cancer. The aim of immunotherapy in all autoimmune myelopathies is to maximize reversibility, maintain benefits (while preventing relapse), and minimize side effects.
Collapse
|
38
|
Alexopoulos H, Biba A, Dalakas MC. Anti-B-Cell Therapies in Autoimmune Neurological Diseases: Rationale and Efficacy Trials. Neurotherapeutics 2016; 13:20-33. [PMID: 26566961 PMCID: PMC4720683 DOI: 10.1007/s13311-015-0402-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
B cells have an ever-increasing role in the etiopathology of a number of autoimmune neurological disorders, acting as antibody-producing cells and, most importantly, as sensors, coordinators, and regulators of the immune response. B cells, among other functions, regulate the T-cell activation process through their participation in antigen presentation and production of cytokines. The availability of monoclonal antibodies or fusion proteins against B-cell surface molecules or B-cell trophic factors bestows a rational approach for treating autoimmune neurological disorders, even when T cells are the main effector cells. This review summarizes basic aspects of B-cell biology, discusses the role(s) of B cells in neurological autoimmunity, and presents anti-B-cell drugs that are either currently on the market or are expected to be available in the near future for treating neurological autoimmune disorders.
Collapse
Affiliation(s)
- Harry Alexopoulos
- Neuroimmunology Unit, Department of Pathophysiology, Faculty of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Angie Biba
- Neuroimmunology Unit, Department of Pathophysiology, Faculty of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Marinos C Dalakas
- Department of Neurology, Thomas Jefferson University, Philadelphia, PA, USA.
| |
Collapse
|
39
|
Maurer MA, Tuller F, Gredler V, Berger T, Lutterotti A, Lünemann JD, Reindl M. Rituximab induces clonal expansion of IgG memory B-cells in patients with inflammatory central nervous system demyelination. J Neuroimmunol 2016; 290:49-53. [DOI: 10.1016/j.jneuroim.2015.11.006] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2015] [Revised: 11/09/2015] [Accepted: 11/12/2015] [Indexed: 11/24/2022]
|
40
|
Bennett JL, O'Connor KC, Bar-Or A, Zamvil SS, Hemmer B, Tedder TF, von Büdingen HC, Stuve O, Yeaman MR, Smith TJ, Stadelmann C. B lymphocytes in neuromyelitis optica. NEUROLOGY-NEUROIMMUNOLOGY & NEUROINFLAMMATION 2015; 2:e104. [PMID: 25977932 PMCID: PMC4426682 DOI: 10.1212/nxi.0000000000000104] [Citation(s) in RCA: 120] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/07/2015] [Accepted: 02/16/2015] [Indexed: 12/21/2022]
Abstract
Neuromyelitis optica (NMO) is an inflammatory autoimmune disorder of the CNS that predominantly affects the spinal cord and optic nerves. A majority (approximately 75%) of patients with NMO are seropositive for autoantibodies against the astrocyte water channel aquaporin-4 (AQP4). These autoantibodies are predominantly IgG1, and considerable evidence supports their pathogenicity, presumably by binding to AQP4 on CNS astrocytes, resulting in astrocyte injury and inflammation. Convergent clinical and laboratory-based investigations have indicated that B cells play a fundamental role in NMO immunopathology. Multiple mechanisms have been hypothesized: AQP4 autoantibody production, enhanced proinflammatory B cell and plasmablast activity, aberrant B cell tolerance checkpoints, diminished B cell regulatory function, and loss of B cell anergy. Accordingly, many current off-label therapies for NMO deplete B cells or modulate their activity. Understanding the role and mechanisms whereby B cells contribute to initiation, maintenance, and propagation of disease activity is important to advancing our understanding of NMO pathogenesis and developing effective disease-specific therapies.
Collapse
Affiliation(s)
- Jeffrey L Bennett
- Departments of Neurology and Ophthalmology and Neuroscience Program (J.L.B.), University of Colorado, Denver; Department of Neurology (K.C.O.), Yale University School of Medicine, New Haven, CT; Neuroimmunology Unit (A.B.-O.), Montreal Neurological Institute and Hospital, McGill University, Montreal, Quebec, Canada; Department of Neurology (S.S.Z., H.-C.v.B.), UCSF School of Medicine, San Francisco, CA; Department of Neurology (B.H.), Technische Universität München, Munich Cluster for Systems Neurology (SyNergy), Munich, Germany; Department of Immunology (T.F.T.), Duke University Medical Center, Durham, NC; Departments of Neurology and Neurotherapeutics (O.S.), University of Texas Southwestern Medical Center, Dallas, TX; Department of Medicine (M.R.Y.), Divisions of Molecular Medicine and Infectious Diseases, University of California, Los Angeles; Harbor-UCLA Medical Center (M.R.Y.), Torrance, CA; Departments of Ophthalmology and Visual Sciences and Internal Medicine (T.J.S.), University of Michigan Medical School, Ann Arbor; and Institute of Neuropathology (C.S.), University Medical Center, Göttingen, Germany
| | - Kevin C O'Connor
- Departments of Neurology and Ophthalmology and Neuroscience Program (J.L.B.), University of Colorado, Denver; Department of Neurology (K.C.O.), Yale University School of Medicine, New Haven, CT; Neuroimmunology Unit (A.B.-O.), Montreal Neurological Institute and Hospital, McGill University, Montreal, Quebec, Canada; Department of Neurology (S.S.Z., H.-C.v.B.), UCSF School of Medicine, San Francisco, CA; Department of Neurology (B.H.), Technische Universität München, Munich Cluster for Systems Neurology (SyNergy), Munich, Germany; Department of Immunology (T.F.T.), Duke University Medical Center, Durham, NC; Departments of Neurology and Neurotherapeutics (O.S.), University of Texas Southwestern Medical Center, Dallas, TX; Department of Medicine (M.R.Y.), Divisions of Molecular Medicine and Infectious Diseases, University of California, Los Angeles; Harbor-UCLA Medical Center (M.R.Y.), Torrance, CA; Departments of Ophthalmology and Visual Sciences and Internal Medicine (T.J.S.), University of Michigan Medical School, Ann Arbor; and Institute of Neuropathology (C.S.), University Medical Center, Göttingen, Germany
| | - Amit Bar-Or
- Departments of Neurology and Ophthalmology and Neuroscience Program (J.L.B.), University of Colorado, Denver; Department of Neurology (K.C.O.), Yale University School of Medicine, New Haven, CT; Neuroimmunology Unit (A.B.-O.), Montreal Neurological Institute and Hospital, McGill University, Montreal, Quebec, Canada; Department of Neurology (S.S.Z., H.-C.v.B.), UCSF School of Medicine, San Francisco, CA; Department of Neurology (B.H.), Technische Universität München, Munich Cluster for Systems Neurology (SyNergy), Munich, Germany; Department of Immunology (T.F.T.), Duke University Medical Center, Durham, NC; Departments of Neurology and Neurotherapeutics (O.S.), University of Texas Southwestern Medical Center, Dallas, TX; Department of Medicine (M.R.Y.), Divisions of Molecular Medicine and Infectious Diseases, University of California, Los Angeles; Harbor-UCLA Medical Center (M.R.Y.), Torrance, CA; Departments of Ophthalmology and Visual Sciences and Internal Medicine (T.J.S.), University of Michigan Medical School, Ann Arbor; and Institute of Neuropathology (C.S.), University Medical Center, Göttingen, Germany
| | - Scott S Zamvil
- Departments of Neurology and Ophthalmology and Neuroscience Program (J.L.B.), University of Colorado, Denver; Department of Neurology (K.C.O.), Yale University School of Medicine, New Haven, CT; Neuroimmunology Unit (A.B.-O.), Montreal Neurological Institute and Hospital, McGill University, Montreal, Quebec, Canada; Department of Neurology (S.S.Z., H.-C.v.B.), UCSF School of Medicine, San Francisco, CA; Department of Neurology (B.H.), Technische Universität München, Munich Cluster for Systems Neurology (SyNergy), Munich, Germany; Department of Immunology (T.F.T.), Duke University Medical Center, Durham, NC; Departments of Neurology and Neurotherapeutics (O.S.), University of Texas Southwestern Medical Center, Dallas, TX; Department of Medicine (M.R.Y.), Divisions of Molecular Medicine and Infectious Diseases, University of California, Los Angeles; Harbor-UCLA Medical Center (M.R.Y.), Torrance, CA; Departments of Ophthalmology and Visual Sciences and Internal Medicine (T.J.S.), University of Michigan Medical School, Ann Arbor; and Institute of Neuropathology (C.S.), University Medical Center, Göttingen, Germany
| | - Bernhard Hemmer
- Departments of Neurology and Ophthalmology and Neuroscience Program (J.L.B.), University of Colorado, Denver; Department of Neurology (K.C.O.), Yale University School of Medicine, New Haven, CT; Neuroimmunology Unit (A.B.-O.), Montreal Neurological Institute and Hospital, McGill University, Montreal, Quebec, Canada; Department of Neurology (S.S.Z., H.-C.v.B.), UCSF School of Medicine, San Francisco, CA; Department of Neurology (B.H.), Technische Universität München, Munich Cluster for Systems Neurology (SyNergy), Munich, Germany; Department of Immunology (T.F.T.), Duke University Medical Center, Durham, NC; Departments of Neurology and Neurotherapeutics (O.S.), University of Texas Southwestern Medical Center, Dallas, TX; Department of Medicine (M.R.Y.), Divisions of Molecular Medicine and Infectious Diseases, University of California, Los Angeles; Harbor-UCLA Medical Center (M.R.Y.), Torrance, CA; Departments of Ophthalmology and Visual Sciences and Internal Medicine (T.J.S.), University of Michigan Medical School, Ann Arbor; and Institute of Neuropathology (C.S.), University Medical Center, Göttingen, Germany
| | - Thomas F Tedder
- Departments of Neurology and Ophthalmology and Neuroscience Program (J.L.B.), University of Colorado, Denver; Department of Neurology (K.C.O.), Yale University School of Medicine, New Haven, CT; Neuroimmunology Unit (A.B.-O.), Montreal Neurological Institute and Hospital, McGill University, Montreal, Quebec, Canada; Department of Neurology (S.S.Z., H.-C.v.B.), UCSF School of Medicine, San Francisco, CA; Department of Neurology (B.H.), Technische Universität München, Munich Cluster for Systems Neurology (SyNergy), Munich, Germany; Department of Immunology (T.F.T.), Duke University Medical Center, Durham, NC; Departments of Neurology and Neurotherapeutics (O.S.), University of Texas Southwestern Medical Center, Dallas, TX; Department of Medicine (M.R.Y.), Divisions of Molecular Medicine and Infectious Diseases, University of California, Los Angeles; Harbor-UCLA Medical Center (M.R.Y.), Torrance, CA; Departments of Ophthalmology and Visual Sciences and Internal Medicine (T.J.S.), University of Michigan Medical School, Ann Arbor; and Institute of Neuropathology (C.S.), University Medical Center, Göttingen, Germany
| | - H-Christian von Büdingen
- Departments of Neurology and Ophthalmology and Neuroscience Program (J.L.B.), University of Colorado, Denver; Department of Neurology (K.C.O.), Yale University School of Medicine, New Haven, CT; Neuroimmunology Unit (A.B.-O.), Montreal Neurological Institute and Hospital, McGill University, Montreal, Quebec, Canada; Department of Neurology (S.S.Z., H.-C.v.B.), UCSF School of Medicine, San Francisco, CA; Department of Neurology (B.H.), Technische Universität München, Munich Cluster for Systems Neurology (SyNergy), Munich, Germany; Department of Immunology (T.F.T.), Duke University Medical Center, Durham, NC; Departments of Neurology and Neurotherapeutics (O.S.), University of Texas Southwestern Medical Center, Dallas, TX; Department of Medicine (M.R.Y.), Divisions of Molecular Medicine and Infectious Diseases, University of California, Los Angeles; Harbor-UCLA Medical Center (M.R.Y.), Torrance, CA; Departments of Ophthalmology and Visual Sciences and Internal Medicine (T.J.S.), University of Michigan Medical School, Ann Arbor; and Institute of Neuropathology (C.S.), University Medical Center, Göttingen, Germany
| | - Olaf Stuve
- Departments of Neurology and Ophthalmology and Neuroscience Program (J.L.B.), University of Colorado, Denver; Department of Neurology (K.C.O.), Yale University School of Medicine, New Haven, CT; Neuroimmunology Unit (A.B.-O.), Montreal Neurological Institute and Hospital, McGill University, Montreal, Quebec, Canada; Department of Neurology (S.S.Z., H.-C.v.B.), UCSF School of Medicine, San Francisco, CA; Department of Neurology (B.H.), Technische Universität München, Munich Cluster for Systems Neurology (SyNergy), Munich, Germany; Department of Immunology (T.F.T.), Duke University Medical Center, Durham, NC; Departments of Neurology and Neurotherapeutics (O.S.), University of Texas Southwestern Medical Center, Dallas, TX; Department of Medicine (M.R.Y.), Divisions of Molecular Medicine and Infectious Diseases, University of California, Los Angeles; Harbor-UCLA Medical Center (M.R.Y.), Torrance, CA; Departments of Ophthalmology and Visual Sciences and Internal Medicine (T.J.S.), University of Michigan Medical School, Ann Arbor; and Institute of Neuropathology (C.S.), University Medical Center, Göttingen, Germany
| | - Michael R Yeaman
- Departments of Neurology and Ophthalmology and Neuroscience Program (J.L.B.), University of Colorado, Denver; Department of Neurology (K.C.O.), Yale University School of Medicine, New Haven, CT; Neuroimmunology Unit (A.B.-O.), Montreal Neurological Institute and Hospital, McGill University, Montreal, Quebec, Canada; Department of Neurology (S.S.Z., H.-C.v.B.), UCSF School of Medicine, San Francisco, CA; Department of Neurology (B.H.), Technische Universität München, Munich Cluster for Systems Neurology (SyNergy), Munich, Germany; Department of Immunology (T.F.T.), Duke University Medical Center, Durham, NC; Departments of Neurology and Neurotherapeutics (O.S.), University of Texas Southwestern Medical Center, Dallas, TX; Department of Medicine (M.R.Y.), Divisions of Molecular Medicine and Infectious Diseases, University of California, Los Angeles; Harbor-UCLA Medical Center (M.R.Y.), Torrance, CA; Departments of Ophthalmology and Visual Sciences and Internal Medicine (T.J.S.), University of Michigan Medical School, Ann Arbor; and Institute of Neuropathology (C.S.), University Medical Center, Göttingen, Germany
| | - Terry J Smith
- Departments of Neurology and Ophthalmology and Neuroscience Program (J.L.B.), University of Colorado, Denver; Department of Neurology (K.C.O.), Yale University School of Medicine, New Haven, CT; Neuroimmunology Unit (A.B.-O.), Montreal Neurological Institute and Hospital, McGill University, Montreal, Quebec, Canada; Department of Neurology (S.S.Z., H.-C.v.B.), UCSF School of Medicine, San Francisco, CA; Department of Neurology (B.H.), Technische Universität München, Munich Cluster for Systems Neurology (SyNergy), Munich, Germany; Department of Immunology (T.F.T.), Duke University Medical Center, Durham, NC; Departments of Neurology and Neurotherapeutics (O.S.), University of Texas Southwestern Medical Center, Dallas, TX; Department of Medicine (M.R.Y.), Divisions of Molecular Medicine and Infectious Diseases, University of California, Los Angeles; Harbor-UCLA Medical Center (M.R.Y.), Torrance, CA; Departments of Ophthalmology and Visual Sciences and Internal Medicine (T.J.S.), University of Michigan Medical School, Ann Arbor; and Institute of Neuropathology (C.S.), University Medical Center, Göttingen, Germany
| | - Christine Stadelmann
- Departments of Neurology and Ophthalmology and Neuroscience Program (J.L.B.), University of Colorado, Denver; Department of Neurology (K.C.O.), Yale University School of Medicine, New Haven, CT; Neuroimmunology Unit (A.B.-O.), Montreal Neurological Institute and Hospital, McGill University, Montreal, Quebec, Canada; Department of Neurology (S.S.Z., H.-C.v.B.), UCSF School of Medicine, San Francisco, CA; Department of Neurology (B.H.), Technische Universität München, Munich Cluster for Systems Neurology (SyNergy), Munich, Germany; Department of Immunology (T.F.T.), Duke University Medical Center, Durham, NC; Departments of Neurology and Neurotherapeutics (O.S.), University of Texas Southwestern Medical Center, Dallas, TX; Department of Medicine (M.R.Y.), Divisions of Molecular Medicine and Infectious Diseases, University of California, Los Angeles; Harbor-UCLA Medical Center (M.R.Y.), Torrance, CA; Departments of Ophthalmology and Visual Sciences and Internal Medicine (T.J.S.), University of Michigan Medical School, Ann Arbor; and Institute of Neuropathology (C.S.), University Medical Center, Göttingen, Germany
| |
Collapse
|
41
|
Wong E, Vishwanath VA, Kister I. Rituximab in neuromyelitis optica: A review of literature. World J Neurol 2015; 5:39-46. [DOI: 10.5316/wjn.v5.i1.39] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2014] [Revised: 01/12/2015] [Accepted: 02/09/2015] [Indexed: 02/06/2023] Open
Abstract
Neuromyelitis optica spectrum disorders, or neuromyelitis optica (NMO), is an autoimmune disease of the central nervous system that must be distinguished from multiple sclerosis. Therapeutic approaches to relapse prevention in NMO include immunosuppressants and monoclonal antibodies. Rituximab, a monoclonal antibody that targets CD20 antigen expressed on the surface of pre-B, mature B-lymphocytes and a small subset of T-lymphocytes, has been widely used for the treatment of NMO. In this review, we aim to summarize global experience with rituximab in NMO. We identified 13 observational studies that involved a total of 209 NMO patients treated with rituximab. Majority of rituximab-treated patients evidenced stabilization or improvements in their disability scores compared to pre-treatment period and 66% of patients remained relapse-free during treatment period. Monitoring rituximab treatment response with CD19+ or CD27+ cell counts appears to improve treatment outcomes. We offer clinical pointers on rituximab use for NMO based on the literature and authors’ experience, and pose questions that would need to be addressed in future studies.
Collapse
|
42
|
Spadaro M, Gerdes LA, Mayer MC, Ertl-Wagner B, Laurent S, Krumbholz M, Breithaupt C, Högen T, Straube A, Giese A, Hohlfeld R, Lassmann H, Meinl E, Kümpfel T. Histopathology and clinical course of MOG-antibody-associated encephalomyelitis. Ann Clin Transl Neurol 2015; 2:295-301. [PMID: 25815356 PMCID: PMC4369279 DOI: 10.1002/acn3.164] [Citation(s) in RCA: 137] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2014] [Revised: 11/18/2014] [Accepted: 11/28/2014] [Indexed: 01/05/2023] Open
Abstract
We present histological, MRI, and clinical features of an adult patient with relapsing encephalomyelitis and antibodies against myelin oligodendrocyte glycoprotein (MOG). Furthermore, we report molecular details of the recognized epitope that is specific for human MOG. A brain biopsy revealed multiple sclerosis (MS)-type II pathology. Some features overlapped with both MS and neuromyelitis optica spectrum disorders (NMOSD), whereas others were distinct from both MS and NMOSD. Immunoadsorption and rituximab induced clinical stabilization. This case contributes a new, so far missing link in the emerging spectrum of MOG-antibody-associated encephalomyelitis.
Collapse
Affiliation(s)
- Melania Spadaro
- Institute of Clinical Neuroimmunology, Medical Campus Grosshadern, Ludwig Maximilian University Munich, Germany
| | - Lisa Ann Gerdes
- Institute of Clinical Neuroimmunology, Medical Campus Grosshadern, Ludwig Maximilian University Munich, Germany
| | - Marie C Mayer
- Institute of Clinical Neuroimmunology, Medical Campus Grosshadern, Ludwig Maximilian University Munich, Germany
| | - Birgit Ertl-Wagner
- Department of Radiology, Medical Campus Grosshadern, Ludwig Maximilian University Munich, Germany
| | - Sarah Laurent
- Institute of Clinical Neuroimmunology, Medical Campus Grosshadern, Ludwig Maximilian University Munich, Germany
| | - Markus Krumbholz
- Institute of Clinical Neuroimmunology, Medical Campus Grosshadern, Ludwig Maximilian University Munich, Germany
| | - Constanze Breithaupt
- Institute of Biochemistry and Biotechnology, Martin Luther University Halle-Wittenberg 06120, Halle, Germany
| | - Tobias Högen
- Department of Neurology, Medical Campus Grosshadern, Ludwig Maximilian University Munich, Germany
| | - Andreas Straube
- Department of Neurology, Medical Campus Grosshadern, Ludwig Maximilian University Munich, Germany
| | - Armin Giese
- Department of Neuropathology, Medical Campus Grosshadern, Ludwig Maximilian University Munich, Germany
| | - Reinhard Hohlfeld
- Institute of Clinical Neuroimmunology, Medical Campus Grosshadern, Ludwig Maximilian University Munich, Germany ; Munich Cluster for Systems Neurology (SyNergy) Munich, Germany
| | - Hans Lassmann
- Department for Neuroimmunology, Center for Brain Research Vienna, Austria
| | - Edgar Meinl
- Institute of Clinical Neuroimmunology, Medical Campus Grosshadern, Ludwig Maximilian University Munich, Germany
| | - Tania Kümpfel
- Institute of Clinical Neuroimmunology, Medical Campus Grosshadern, Ludwig Maximilian University Munich, Germany
| |
Collapse
|
43
|
Abstract
PURPOSE OF REVIEW Longitudinally extensive transverse myelitis (LETM) is a frequently devastating clinical syndrome which has come into focus for its association with neuromyelitis optica (NMO). Recent advances in the diagnosis of NMO have led to very sensitive and specific tests and advances in therapy for this disorder. LETM is not pathognomonic of NMO, therefore it is important to investigate for other causes of myelopathy in these patients. This review aims to discuss recent advances in NMO diagnosis and treatment, and to discuss the differential diagnosis in patients presenting with LETM. RECENT FINDINGS Fluorescence-activated cell sorting and cell binding assays for NMO-IgG are the most sensitive for detecting NMO spectrum disorders. Patients who have a clinical presentation of NMO, who have been tested with older ELISA or immunofluorescence assay and been found to be negative, should be retested with a fluorescence-activated cell sorting assay when available, particularly in the presence of recurrent LETM. Novel therapeutic strategies for LETM in the context of NMO include eculizumab, which could be considered in patients with active disease who have failed azathioprine and rituximab. Thorough investigation of patients with LETM who are negative for NMO-IgG may lead to an alternate cause for myelopathy. SUMMARY LETM is a heterogeneous condition. Novel treatment strategies are available for NMO, but other causes need to be excluded in NMO-IgG-seronegative patients.
Collapse
|
44
|
CD19 as a molecular target in CNS autoimmunity. Acta Neuropathol 2014; 128:177-90. [PMID: 24993505 DOI: 10.1007/s00401-014-1313-z] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2014] [Revised: 06/18/2014] [Accepted: 06/18/2014] [Indexed: 02/08/2023]
Abstract
Multiple sclerosis (MS) and neuromyelitis optica (NMO) are the most prevalent neuroinflammatory diseases of the central nervous system (CNS). The immunological cascade of these disorders is complex, and the exact spatial and temporal role of different immune cells is not fully understood. Although MS has been considered for many years to be primarily T cell driven, it is well established that B cells and the humoral immune response play an important role in its pathogenesis. This has long been evident from laboratory findings that include the presence of oligoclonal bands in the CSF. In NMO, the importance of the humoral immune system appears even more obvious as evidenced by pathogenic antibodies against aquaporin 4 (AQP4). Besides their capacity to mature into antibody-producing plasma cells, B cells are potent antigen-presenting cells to T lymphocytes and they can provide soluble factors for cell activation and differentiation to other immune-competent cells. In MS and NMO, there are substantial data from clinical trials that B cell depletion with CD20-directed agents is effective and relatively safe. Plasma cells, which produce antibodies against molecular targets expressed by the host, but which also provide humoral immune responses against pathogens, are not targeted by anti-CD20 therapies. Therefore, the depletion of CD19-expressing cells would offer potential advantages with regard to efficacy, but potentially higher risks with regard to infectious complications. This review will outline the rationale for CD19 as a molecular target in CNS autoimmunity. The current stage of drug development is illustrated. Potential safety concerns will be discussed.
Collapse
|
45
|
|
46
|
Jarius S, Wildemann B. Aquaporin-4 antibodies (NMO-IgG) as a serological marker of neuromyelitis optica: a critical review of the literature. Brain Pathol 2014; 23:661-83. [PMID: 24118483 DOI: 10.1111/bpa.12084] [Citation(s) in RCA: 185] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2013] [Accepted: 08/13/2013] [Indexed: 01/19/2023] Open
Abstract
Antibodies to aquaporin-4 (called NMO-IgG or AQP4-Ab) constitute a sensitive and highly specific serum marker of neuromyelitis optica (NMO) that can facilitate the differential diagnosis of NMO and classic multiple sclerosis. NMO-IgG/AQP4-Ab seropositive status has also important prognostic and therapeutic implications in patients with isolated longitudinally extensive myelitis (LETM) or optic neuritis (ON). In this article, we comprehensively review and critically appraise the existing literature on NMO-IgG/AQP4-Ab testing. All available immunoassays-including tissue-based (IHC), cell-based (ICC, FACS) and protein-based (RIPA, FIPA, ELISA, Western blotting) assays-and their differential advantages and disadvantages are discussed. Estimates for sensitivity, specificity, and positive and negative likelihood ratios are calculated for all published studies and accuracies of the various immunoassay techniques compared. Subgroup analyses are provided for NMO, LETM and ON, for relapsing vs. monophasic disease, and for various control groups (eg, MS vs. other controls). Numerous aspects of NMO-IgG/AQP4-Ab testing relevant for clinicians (eg, impact of antibody titers and longitudinal testing, indications for repeat testing, relevance of CSF testing and subclass analysis, NMO-IgG/AQP4-Ab in patients with rheumatic diseases) as well as technical aspects (eg, AQP4-M1 vs. AQP4-M23-based assays, intact AQP4 vs. peptide substrates, effect of storage conditions and freeze/thaw cycles) and pitfalls are discussed. Finally, recommendations for the clinical application of NMO-IgG/AQP4-Ab serology are given.
Collapse
Affiliation(s)
- Sven Jarius
- Division of Molecular Neuroimmunology, Department of Neurology, University of Heidelberg, Heidelberg, Germany
| | | |
Collapse
|
47
|
Affiliation(s)
- Lawrence Steinman
- Departments of Pediatrics, Neurology and Neurological Sciences, Stanford University, Stanford, California 94305;
| |
Collapse
|
48
|
Abstract
Neuromyelitis optica (NMO) is a chronic inflammatory disease of the central nervous system that affects the optic nerves and spinal cord resulting in visual impairment and myelopathy. There is a growing body of evidence that immunotherapeutic agents targeting T and B cell functions, as well as active elimination of proinflammatory molecules from the peripheral blood circulation, can attenuate disease progression. In this review, we discuss the immunotherapeutic options and the treatment strategies in NMO. We also analyze the pathogenic mechanisms of the disease in order to provide recommendations regarding treatments.
Collapse
|
49
|
Jarius S, Wildemann B. Aquaporin-4 antibodies (NMO-IgG) as a serological marker of neuromyelitis optica: a critical review of the literature. BRAIN PATHOLOGY (ZURICH, SWITZERLAND) 2013. [PMID: 24118483 DOI: 10.1111/bpa.12084"] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Antibodies to aquaporin-4 (called NMO-IgG or AQP4-Ab) constitute a sensitive and highly specific serum marker of neuromyelitis optica (NMO) that can facilitate the differential diagnosis of NMO and classic multiple sclerosis. NMO-IgG/AQP4-Ab seropositive status has also important prognostic and therapeutic implications in patients with isolated longitudinally extensive myelitis (LETM) or optic neuritis (ON). In this article, we comprehensively review and critically appraise the existing literature on NMO-IgG/AQP4-Ab testing. All available immunoassays-including tissue-based (IHC), cell-based (ICC, FACS) and protein-based (RIPA, FIPA, ELISA, Western blotting) assays-and their differential advantages and disadvantages are discussed. Estimates for sensitivity, specificity, and positive and negative likelihood ratios are calculated for all published studies and accuracies of the various immunoassay techniques compared. Subgroup analyses are provided for NMO, LETM and ON, for relapsing vs. monophasic disease, and for various control groups (eg, MS vs. other controls). Numerous aspects of NMO-IgG/AQP4-Ab testing relevant for clinicians (eg, impact of antibody titers and longitudinal testing, indications for repeat testing, relevance of CSF testing and subclass analysis, NMO-IgG/AQP4-Ab in patients with rheumatic diseases) as well as technical aspects (eg, AQP4-M1 vs. AQP4-M23-based assays, intact AQP4 vs. peptide substrates, effect of storage conditions and freeze/thaw cycles) and pitfalls are discussed. Finally, recommendations for the clinical application of NMO-IgG/AQP4-Ab serology are given.
Collapse
Affiliation(s)
- Sven Jarius
- Division of Molecular Neuroimmunology, Department of Neurology, University of Heidelberg, Heidelberg, Germany
| | | |
Collapse
|
50
|
Sato DK, Lana-Peixoto MA, Fujihara K, de Seze J. Clinical Spectrum and Treatment of Neuromyelitis Optica Spectrum Disorders: Evolution and Current Status. Brain Pathol 2013; 23:647-60. [DOI: 10.1111/bpa.12087] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2013] [Accepted: 08/15/2013] [Indexed: 12/18/2022] Open
Affiliation(s)
| | | | - Kazuo Fujihara
- Department of Multiple Sclerosis Therapeutics; Tohoku University Graduate School of Medicine; Sendai; Japan
| | - Jerome de Seze
- Department of Neurology; Strasbourg University and Clinical Investigation Center; Strasbourg Hospital; Strasbourg; France
| |
Collapse
|