1
|
Pérez-Saldívar M, Nakamura Y, Kiyotani K, Imoto S, Katayama K, Yamaguchi R, Miyano S, Martínez-Barnetche J, Godoy-Lozano EE, Ordoñez G, Sotelo J, González-Conchillos H, Martínez-Palomo A, Flores-Rivera J, Santos-Argumedo L, Sánchez-Salguero ES, Espinosa-Cantellano M. Comparative analysis of the B cell receptor repertoire during relapse and remission in patients with multiple sclerosis. Clin Immunol 2024; 269:110398. [PMID: 39551364 DOI: 10.1016/j.clim.2024.110398] [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: 09/26/2024] [Revised: 11/11/2024] [Accepted: 11/12/2024] [Indexed: 11/19/2024]
Abstract
Multiple sclerosis (MS) is a chronic, multifactorial, inflammatory and demyelinating disease of the central nervous system (CNS), which involves an autoimmune response against components of the myelin sheaths. Anti-B cell therapies have been proven to be successful in reducing relapses. Therefore, the study of B cells in both phases of the disease (relapse and remission) is of great importance. Here, we analyzed peripheral blood-cell BCR repertoire from 11 MS patients during a relapse phase and during remission, 6 patients with other inflammatory neurological diseases (OIND) and 10 healthy subjects (HCs), using next generation sequencing. In addition, immunoglobulins G, M, A and D were quantified in the serum of patients and controls, using ELISA. BCR repertoire of relapsing MS patients showed lower diversity, as well as a higher rate of somatic hypermutation compared to the other study groups. Within this group, the highest percentage of shared clonotypes was observed. IGHV4-32 gene was identified as a potential differential biomarker between MS and OIND, as well as IGL3-21 gene as a potential MS biomarker. On the other hand, an elevation of IgG and IgD was found in the serum of MS patients during remission, and the serum IgG was also elevated in MS patients during relapse. In conclusion, these results show the important role of B cells in the pathogenesis of the MS relapses and a new panorama on the analysis of the peripheral blood BCR repertoire to obtain diagnostic tools for MS. Furthermore, this work highlights the need of studies in diverse populations, since results reported in Caucasian populations may not coincide with the immunological course of MS patients in other latitudes, due to differences in genetic background and environmental exposures.
Collapse
Affiliation(s)
- Miriam Pérez-Saldívar
- Department of Infectomics and Molecular Pathogenesis, Center for Research and Advanced Studies (Cinvestav), Mexico City 07360, Mexico
| | - Yusuke Nakamura
- Project for Immunogenomics, Cancer Precision Medicine Center, Japanese Foundation for Cancer Research, Tokyo, Japan.
| | - Kazuma Kiyotani
- Project for Immunogenomics, Cancer Precision Medicine Center, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Seiya Imoto
- Human Genome Center, Institute of Medical Science, The University of Tokyo, Tokyo 108-8639, Japan
| | - Kotoe Katayama
- Human Genome Center, Institute of Medical Science, The University of Tokyo, Tokyo 108-8639, Japan
| | - Rui Yamaguchi
- Division of Cancer Systems Biology, Aichi Cancer Center Research Institute, Nagoya, Aichi 464-8681, Japan
| | - Satoru Miyano
- Human Genome Center, Institute of Medical Science, The University of Tokyo, Tokyo 108-8639, Japan
| | - Jesús Martínez-Barnetche
- Centro de Investigación Sobre Enfermedades Infecciosas-Instituto Nacional de Salud Pública, Cuernavaca, Morelos 62100, Mexico
| | | | - Graciela Ordoñez
- Department of Neuroimmunology, National Institute of Neurology and Neurosurgery "Manuel Velasco Suarez" (INNN), Mexico City 14269, Mexico
| | - Julio Sotelo
- Department of Neuroimmunology, National Institute of Neurology and Neurosurgery "Manuel Velasco Suarez" (INNN), Mexico City 14269, Mexico
| | - Hugo González-Conchillos
- Department of Infectomics and Molecular Pathogenesis, Center for Research and Advanced Studies (Cinvestav), Mexico City 07360, Mexico
| | - Adolfo Martínez-Palomo
- Department of Infectomics and Molecular Pathogenesis, Center for Research and Advanced Studies (Cinvestav), Mexico City 07360, Mexico
| | - José Flores-Rivera
- Clinical Laboratory of Neurodegenerative Diseases, National Institute of Neurology and Neurosurgery "Manuel Velasco Suarez" (INNN), Mexico City 14269, Mexico
| | - Leopoldo Santos-Argumedo
- Department of Molecular Biomedicine, Center for Research and Advanced Studies (Cinvestav), Mexico City 07360, Mexico
| | - Erick Saúl Sánchez-Salguero
- Department of Molecular Biomedicine, Center for Research and Advanced Studies (Cinvestav), Mexico City 07360, Mexico
| | - Martha Espinosa-Cantellano
- Department of Infectomics and Molecular Pathogenesis, Center for Research and Advanced Studies (Cinvestav), Mexico City 07360, Mexico.
| |
Collapse
|
2
|
Akpinar Adscheid S, Türeli AE, Günday-Türeli N, Schneider M. Nanotechnological approaches for efficient N2B delivery: from small-molecule drugs to biopharmaceuticals. BEILSTEIN JOURNAL OF NANOTECHNOLOGY 2024; 15:1400-1414. [PMID: 39559726 PMCID: PMC11572074 DOI: 10.3762/bjnano.15.113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Accepted: 10/22/2024] [Indexed: 11/20/2024]
Abstract
Central nervous system diseases negatively affect patients and society. Providing successful noninvasive treatments for these diseases is challenging because of the presence of the blood-brain barrier. While protecting the brain's homeostasis, the barrier limits the passage of almost all large-molecule drugs and most small-molecule drugs. A noninvasive method, nose-to-brain delivery (N2B delivery) has been proposed to overcome this challenge. By exploiting the direct anatomical interaction between the nose and the brain, the drugs can reach the target, the brain. Moreover, the drugs can be encapsulated into various drug delivery systems to enhance physicochemical characteristics and targeting success. Many preclinical data show that this strategy can effectively deliver biopharmaceuticals to the brain. Therefore, this review focuses on N2B delivery while giving examples of different drug delivery systems suitable for the applications. In addition, we emphasize the importance of the effective delivery of monoclonal antibodies and RNA and stress the recent literature tackling this challenge. While giving examples of nanotechnological approaches for the effective delivery of small or large molecules from the current literature, we highlight the preclinical studies and their results to prove the strategies' success and limitations.
Collapse
Affiliation(s)
- Selin Akpinar Adscheid
- MyBiotech GmbH; Industriestraße 1B, 66802 Überherrn, Germany
- Department of Pharmacy, Biopharmaceutics and Pharmaceutical Technology, PharmaScienceHub, Saarland University, Campus C4 1, Saarbrücken D-66123, Germany
| | | | | | - Marc Schneider
- Department of Pharmacy, Biopharmaceutics and Pharmaceutical Technology, PharmaScienceHub, Saarland University, Campus C4 1, Saarbrücken D-66123, Germany
| |
Collapse
|
3
|
Groh J, Simons M. White matter aging and its impact on brain function. Neuron 2024:S0896-6273(24)00767-0. [PMID: 39541972 DOI: 10.1016/j.neuron.2024.10.019] [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: 08/20/2024] [Revised: 09/16/2024] [Accepted: 10/18/2024] [Indexed: 11/17/2024]
Abstract
Aging has a detrimental impact on white matter, resulting in reduced volume, compromised structural integrity of myelinated axons, and an increase in white matter hyperintensities. These changes are closely linked to cognitive decline and neurological disabilities. The deterioration of myelin and its diminished ability to regenerate as we age further contribute to the progression of neurodegenerative disorders. Understanding these changes is crucial for devising effective disease prevention strategies. Here, we will discuss the structural alterations in white matter that occur with aging and examine the cellular and molecular mechanisms driving these aging-related transformations. We highlight how the progressive disruption of white matter may initiate a self-perpetuating cycle of inflammation and neural damage.
Collapse
Affiliation(s)
- Janos Groh
- Institute of Neuronal Cell Biology, Technical University Munich, Munich, Germany; German Center for Neurodegenerative Diseases (DZNE), Munich, Germany; Munich Cluster of Systems Neurology (SyNergy), Munich, Germany.
| | - Mikael Simons
- Institute of Neuronal Cell Biology, Technical University Munich, Munich, Germany; German Center for Neurodegenerative Diseases (DZNE), Munich, Germany; Munich Cluster of Systems Neurology (SyNergy), Munich, Germany; Institute for Stroke and Dementia Research, University Hospital of Munich, LMU Munich, Munich, Germany.
| |
Collapse
|
4
|
Dalla Costa G, Leocani L, Rodegher M, Chiveri L, Gradassi A, Comi G. An overview on disease modifying and symptomatic drug treatments for multiple sclerosis. Expert Rev Clin Pharmacol 2024:1-21. [PMID: 39376160 DOI: 10.1080/17512433.2024.2410393] [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: 05/03/2024] [Accepted: 09/25/2024] [Indexed: 10/09/2024]
Abstract
INTRODUCTION Multiple sclerosis (MS) is an inflammatory and degenerative autoimmune condition, resulting frequently in a disabling condition. Significant improvements of long-term prognosis have been recently achieved with an early and more aggressive use of disease modifying therapies (DMTs). Addressing the complexity of managing its progressive forms remains a significant challenge. AREAS COVERED This review provides an update on DMTs for relapsing-remitting MS (RRMS) and progressive MS and their efficacy, safety, and mechanism of action, emphasizing the critical role of biomarkers in optimizing treatment decisions. Moreover, some key information on drugs used to manage symptoms such as pain, fatigue, spasticity and urinary problems will be provided. The literature search was conducted using PubMed, Embase, and Cochrane Library databases covering the period from January 2000 to January 2024. EXPERT OPINION Major advances have been achieved in the treatment of RRMS. Treatment should start immediately as soon as the neurologist is confident with the diagnosis and its choice should be based on the prognostic profile and on the patient's propensity to accept drug-related risks. The therapeutic landscape for progressive MS is quite disappointing and necessitates further innovation. Personalized medicine, leveraging biomarker insights, holds promise for refining treatment efficacy and patient outcomes.
Collapse
Affiliation(s)
| | - Letizia Leocani
- Vita Salute San Raffaele University, Milan, Italy
- Department of Neurorehabilitation Sciences, Casa di Cura Igea, Milan, Italy
| | - Mariaemma Rodegher
- Department of Neurorehabilitation Sciences, Casa di Cura Igea, Milan, Italy
| | - Luca Chiveri
- Department of Neurorehabilitation Sciences, Casa di Cura Igea, Milan, Italy
| | | | - Giancarlo Comi
- Department of Neurorehabilitation Sciences, Casa di Cura Igea, Milan, Italy
| |
Collapse
|
5
|
Kirthivasan N, Cyster JG. Lymphoid tissue on the mind. Trends Immunol 2024; 45:325-326. [PMID: 38637201 DOI: 10.1016/j.it.2024.04.002] [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: 04/04/2024] [Accepted: 04/08/2024] [Indexed: 04/20/2024]
Abstract
To surveil an organ for pathogens, lymphoid structures need to sample antigens locally. The full set of lymphoid structures involved in surveilling for brain-tropic pathogens has not been defined. Through comprehensive imaging of the mouse meninges, a new study by Fitzpatrick et al. describes dural-associated lymphoid tissue (DALT) and its contribution to humoral responses following intranasal viral infection.
Collapse
Affiliation(s)
- Nikhita Kirthivasan
- Howard Hughes Medical Institute and Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94143, USA.
| | - Jason G Cyster
- Howard Hughes Medical Institute and Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94143, USA.
| |
Collapse
|
6
|
Delgado SR, Faissner S, Linker RA, Rammohan K. Key characteristics of anti-CD20 monoclonal antibodies and clinical implications for multiple sclerosis treatment. J Neurol 2024; 271:1515-1535. [PMID: 37906325 PMCID: PMC10973056 DOI: 10.1007/s00415-023-12007-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Revised: 09/13/2023] [Accepted: 09/14/2023] [Indexed: 11/02/2023]
Abstract
The recent success of anti-CD20 monoclonal antibody therapies in the treatment of multiple sclerosis (MS) has highlighted the role of B cells in the pathogenesis of MS. In people with MS, the inflammatory characteristics of B-cell activity are elevated, leading to increased pro-inflammatory cytokine release, diminished anti-inflammatory cytokine production and an accumulation of pathogenic B cells in the cerebrospinal fluid. Rituximab, ocrelizumab, ofatumumab, ublituximab and BCD-132 are anti-CD20 therapies that are either undergoing clinical development, or have been approved, for the treatment of MS. Despite CD20 being a common target for these therapies, differences have been reported in their mechanistic, pharmacological and clinical characteristics, which may have substantial clinical implications. This narrative review explores key characteristics of these therapies. By using clinical trial data and real-world evidence, we discuss their mechanisms of action, routes of administration, efficacy (in relation to B-cell kinetics), safety, tolerability and convenience of use. Clinicians, alongside patients and their families, should consider the aspects discussed in this review as part of shared decision-making discussions to improve outcomes and health-related quality of life for people living with MS.
Collapse
Affiliation(s)
- Silvia R Delgado
- Department of Neurology, Leonard M. Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Simon Faissner
- Department of Neurology, Ruhr-University Bochum, St Josef-Hospital, Bochum, Germany
| | - Ralf A Linker
- Department of Neurology, University Hospital Regensburg, Regensburg, Germany
| | - Kottil Rammohan
- Department of Neurology, Leonard M. Miller School of Medicine, University of Miami, Miami, FL, USA.
| |
Collapse
|
7
|
Algahtani H, Shirah B, Alqahtani A, Abdelghaffar N, Makki S. Half-dose ocrelizumab in selected patients with relapsing-remitting multiple sclerosis. Acta Neurol Belg 2024; 124:303-306. [PMID: 37301802 PMCID: PMC10257484 DOI: 10.1007/s13760-023-02303-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Accepted: 06/02/2023] [Indexed: 06/12/2023]
Affiliation(s)
- Hussein Algahtani
- Department of Medicine, Aseer Central Hospital, Abha, Saudi Arabia.
- Neurology Section, Department of Medicine, King Abdulaziz Medical City, P.O. Box: 12723, Jeddah, 21483, Saudi Arabia.
| | - Bader Shirah
- Department of Neuroscience, King Faisal Specialist Hospital & Research Centre, Jeddah, Saudi Arabia
| | | | - Nawal Abdelghaffar
- Department of Medicine, Aseer Central Hospital, Abha, Saudi Arabia
- Neurology Department, Kasr Al Ainy Hospital, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Samiah Makki
- College of Medicine, King Saud Bin Abdulaziz University for Health Sciences, Jeddah, Saudi Arabia
| |
Collapse
|
8
|
Boyko AN, Lasch NY, Guseva ME. [Optimal patient profile for ofatumumab treatment: analysis of Russian data from the Phase 3 study]. Zh Nevrol Psikhiatr Im S S Korsakova 2024; 124:37-42. [PMID: 39175238 DOI: 10.17116/jnevro202412407237] [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] [Indexed: 08/24/2024]
Abstract
The article provides an analysis of the features of the action of ofatumumab in subgroups of patients with multiple sclerosis (MS) who participated in phase 3 ASCLEPIOS I and II studies both in the general subgroup of 1882 patients and among 352 patients from the Russian Federation who participated in these studies. The results of the influence of age, gender, body weight of patients, as well as the basic level of disability on the EDSS scale, the presence of active foci on MRI and previously received therapy with drugs that alter the course of MS (PITRS) are presented. In a total group of 1.882 patients, a more positive effect of ofatumumab compared with teriflunomide was noted on the average annual incidence of exacerbations in men, younger people and with a mild baseline disability level - with a baseline EDSS level less than or equal to 3. In a subgroup of 352 patients from Russia, the same trends were noted, but dependencies were also revealed from the number of previously taken PITRS: a more significant difference was noted in patients with the lowest number of PITRS in the anamnesis. This feature was also confirmed by analyzing the secondary endpoints of the study: the number of active foci on MRI and the confirmed progression of disability according to the EDSS scale. Analysis in clinical subgroups makes it possible to clarify the profile of patients in whom the greatest clinical effect can be expected when using this new drug for the treatment of MS.
Collapse
Affiliation(s)
- A N Boyko
- Pirogov Russian National Research Medical University, Moscow, Russia
- Federal Center for Brain and Neurotechnology, Moscow, Russia
| | - N Yu Lasch
- Pirogov Russian National Research Medical University, Moscow, Russia
- Federal Center for Brain and Neurotechnology, Moscow, Russia
| | - M E Guseva
- Pirogov Russian National Research Medical University, Moscow, Russia
| |
Collapse
|
9
|
Hauser SL, Kappos L, Bar-Or A, Wiendl H, Paling D, Williams M, Gold R, Chan A, Milo R, Das Gupta A, Karlsson G, Sullivan R, Graham G, Merschhemke M, Häring DA, Vermersch P. The Development of Ofatumumab, a Fully Human Anti-CD20 Monoclonal Antibody for Practical Use in Relapsing Multiple Sclerosis Treatment. Neurol Ther 2023; 12:1491-1515. [PMID: 37450172 PMCID: PMC10444716 DOI: 10.1007/s40120-023-00518-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Accepted: 06/14/2023] [Indexed: 07/18/2023] Open
Abstract
The importance of B cells in multiple sclerosis (MS) has been demonstrated through the advent of B-cell-depleting anti-CD20 antibody therapies. Ofatumumab is the first fully human anti-CD20 monoclonal antibody (mAb) developed and tested for subcutaneous (SC) self-administration at monthly doses of 20 mg, and has been approved in the US, UK, EU, and other regions and countries worldwide for the treatment of relapsing MS. The development goal of ofatumumab was to obtain a highly efficacious anti-CD20 therapy, with a safety and tolerability profile that allows for self-administration by MS patients at home and a positive benefit-risk balance for use in the broad relapsing MS population. This development goal was enabled by the unique binding site, higher affinity to B cells, and higher potency of ofatumumab compared to previous anti-CD20 mAbs; these properties of ofatumumab facilitate rapid B-cell depletion and maintenance with a low dose at a low injection volume (20 mg/0.4 ml). The high potency in turn enables the selective targeting of B cells that reside in the lymphatic system via subcutaneous (SC) administration. Through a comprehensive dose-finding program in two phase 2 studies (one intravenous and one SC) and model simulations, it was found that safety and tolerability can be further improved, and the risk of systemic injection-related reactions (IRRs) minimized, by avoiding doses ≥ 30 mg, and by reaching initial and rapid B-cell depletion via stepwise weekly administration of ofatumumab at Weeks 0, 1, and 2 (instead of a single high dose). Once near-complete B-cell depletion is reached, it can be maintained by monthly doses of 20 mg/0.4 ml. Indeed, in phase 3 trials (ASCLEPIOS I/II), rapid and sustained near-complete B-cell depletion (largely independent of body weight, race and other factors) was observed with this dosing regimen, which resulted in superior efficacy of ofatumumab versus teriflunomide on relapse rates, disability worsening, neuronal injury (serum neurofilament light chain), and imaging outcomes. Likely due to its fully human nature, ofatumumab has a low immunogenic risk profile-only 2 of 914 patients receiving ofatumumab in ASCLEPIOS I/II developed anti-drug antibodies-and this may also underlie the infrequent IRRs (20% with ofatumumab vs. 15% with the placebo injection in the teriflunomide arm) that were mostly (99.8%) mild to moderate in severity. The overall rates of infections and serious infections in patients treated with ofatumumab were similar to those in patients treated with teriflunomide (51.6% vs. 52.7% and 2.5% vs. 1.8%, respectively). The benefit-risk profile of ofatumumab was favorable compared to teriflunomide in the broad RMS population, and also in the predefined subgroups of both recently diagnosed and/or treatment-naïve patients, as well as previously disease-modifying therapy-treated patients. Interim data from the ongoing extension study (ALITHIOS) have shown that long-term treatment with ofatumumab up to 4 years is well-tolerated in RMS patients, with no new safety risks identified. In parallel to the phase 3 trials in which SC administration was carried out with a pre-filled syringe, an autoinjector pen for more convenient self-administration of the ofatumumab 20 mg dose was developed and is available for use in clinical practice.
Collapse
Affiliation(s)
- Stephen L Hauser
- UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA.
| | - Ludwig Kappos
- Research Center for Clinical Neuroimmunology and Neuroscience Basel (RC2NB) and MS Center, and Departments of Medicine, Clinical Research, Biomedicine and Biomedical Engineering, University Hospital of Basel, University of Basel, Basel, Switzerland
| | - Amit Bar-Or
- Center for Neuroinflammation and Experimental Therapeutics and Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Heinz Wiendl
- Department of Neurology with Institute of Translational Neurology, University Hospital Münster, Münster, Germany
| | - David Paling
- Sheffield Institute of Translational Neuroscience, Sheffield Teaching Hospital NHS Foundation Trust, Sheffield, UK
| | - Mitzi Williams
- Joi Life Wellness Multiple Sclerosis Neurology Center, Atlanta, GA, USA
| | - Ralf Gold
- Department of Neurology, St Josef-Hospital/Ruhr-University Bochum, Bochum, Germany
| | - Andrew Chan
- Department of Neurology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Ron Milo
- Department of Neurology, Barzilai Medical Center, Ashkelon/Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | | | | | | | | | | | | | - Patrick Vermersch
- Univ. Lille, INSERM U1172 LilNCog, CHU Lille, FHU Precise, 59000, Lille, France
| |
Collapse
|
10
|
Wilhelm CR, Upadhye MA, Eschbacher KL, Karandikar NJ, Boyden AW. Proteolipid Protein-Induced Mouse Model of Multiple Sclerosis Requires B Cell-Mediated Antigen Presentation. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2023; 211:944-953. [PMID: 37548478 PMCID: PMC10528642 DOI: 10.4049/jimmunol.2200721] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Accepted: 07/18/2023] [Indexed: 08/08/2023]
Abstract
The pathogenic role B cells play in multiple sclerosis is underscored by the success of B cell depletion therapies. Yet, it remains unclear how B cells contribute to disease, although it is increasingly accepted that mechanisms beyond Ab production are involved. Better understanding of pathogenic interactions between B cells and autoreactive CD4 T cells will be critical for novel therapeutics. To focus the investigation on B cell:CD4 T cell interactions in vivo and in vitro, we previously developed a B cell-dependent, Ab-independent experimental autoimmune encephalomyelitis (EAE) mouse model driven by a peptide encompassing the extracellular domains of myelin proteolipid protein (PLPECD). In this study, we demonstrate that B cell depletion significantly inhibited PLPECD-induced EAE disease, blunted PLPECD-elicited delayed-type hypersensitivity reactions in vivo, and reduced CD4 T cell activation, proliferation, and proinflammatory cytokine production. Further, PLPECD-reactive CD4 T cells sourced from B cell-depleted donor mice failed to transfer EAE to naive recipients. Importantly, we identified B cell-mediated Ag presentation as the critical mechanism explaining B cell dependence in PLPECD-induced EAE, where bone marrow chimeric mice harboring a B cell-restricted MHC class II deficiency failed to develop EAE. B cells were ultimately observed to restimulate significantly higher Ag-specific proliferation from PLP178-191-reactive CD4 T cells compared with dendritic cells when provided PLPECD peptide in head-to-head cultures. We therefore conclude that PLPECD-induced EAE features a required pathogenic B cell-mediated Ag presentation function, providing for investigable B cell:CD4 T cell interactions in the context of autoimmune demyelinating disease.
Collapse
Affiliation(s)
- Connor R. Wilhelm
- Iowa City Veterans Affairs Medical Center
- Department of Pathology Graduate Program, University of Iowa, Iowa City, IA USA
| | - Mohit A. Upadhye
- Iowa City Veterans Affairs Medical Center
- Department of Pathology Graduate Program, University of Iowa, Iowa City, IA USA
| | | | - Nitin J. Karandikar
- Department of Pathology, University of Iowa Carver College of Medicine
- Iowa City Veterans Affairs Medical Center
- Department of Pathology Graduate Program, University of Iowa, Iowa City, IA USA
| | - Alexander W. Boyden
- Department of Pathology, University of Iowa Carver College of Medicine
- Iowa City Veterans Affairs Medical Center
| |
Collapse
|
11
|
Wang M, Liu C, Zou M, Niu Z, Zhu J, Jin T. Recent progress in epidemiology, clinical features, and therapy of multiple sclerosis in China. Ther Adv Neurol Disord 2023; 16:17562864231193816. [PMID: 37719665 PMCID: PMC10504852 DOI: 10.1177/17562864231193816] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2023] [Accepted: 07/24/2023] [Indexed: 09/19/2023] Open
Abstract
Multiple sclerosis (MS) is a demyelinating disease of the central nervous system characterized by inflammation, demyelination, and neurodegeneration. It mainly affects young adults, imposing a heavy burden on families and society. The epidemiology, clinical features, and management of MS are distinct among different countries. Although MS is a rare disease in China, there are 1.4 billion people in China, so the total number of MS patients is not small. Because of the lack of specific diagnostic biomarkers for MS, there is a high misdiagnosis rate in China, as in other regions. Due to different genetic backgrounds, the clinical manifestations of MS in Chinese are different from those in the West. Herein, this review aims to summarize the disease comprehensively, including clinical profile and the status of disease-modifying therapies in China based on published population-based observation and cohort studies, and also to compare with data from other countries and regions, thus providing help to develop diagnostic guideline and the novel therapeutic drugs. Meanwhile, we also discuss the problems and challenges we face, specifically for the diagnosis and treatment of MS in the middle- and low-income countries.
Collapse
Affiliation(s)
- Meng Wang
- Neuroscience Center, Department of Neurology, The First Hospital of Jilin University, Changchun, China
| | - Caiyun Liu
- Neuroscience Center, Department of Neurology, The First Hospital of Jilin University, Changchun, China
| | - Meijuan Zou
- Neuroscience Center, Department of Neurology, The First Hospital of Jilin University, Changchun, China
| | - Zixuan Niu
- Neuroscience Center, Department of Neurology, The First Hospital of Jilin University, Changchun, China
| | - Jie Zhu
- Neuroscience Center, Department of Neurology, The First Hospital of Jilin University, No. 1, Xinmin Street, Changchun 130021, China
- Division of Neurogeriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Karolinska University Hospital, Solna, Stockholm 171 64, Sweden
| | - Tao Jin
- Neuroscience Center, Department of Neurology, The First Hospital of Jilin University, No. 1, Xinmin Street, Changchun 130021, China
| |
Collapse
|
12
|
Xavier A, Maltby VE, Ewing E, Campagna MP, Burnard SM, Tegner JN, Slee M, Butzkueven H, Kockum I, Kular L, Jokubaitis VG, Kilpatrick T, Alfredsson L, Jagodic M, Ponsonby AL, Taylor BV, Scott RJ, Lea RA, Lechner-Scott J. DNA Methylation Signatures of Multiple Sclerosis Occur Independently of Known Genetic Risk and Are Primarily Attributed to B Cells and Monocytes. Int J Mol Sci 2023; 24:12576. [PMID: 37628757 PMCID: PMC10454485 DOI: 10.3390/ijms241612576] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Revised: 07/20/2023] [Accepted: 08/02/2023] [Indexed: 08/27/2023] Open
Abstract
Epigenetic mechanisms can regulate how DNA is expressed independently of sequence and are known to be associated with various diseases. Among those epigenetic mechanisms, DNA methylation (DNAm) is influenced by genotype and the environment, making it an important molecular interface for studying disease etiology and progression. In this study, we examined the whole blood DNA methylation profiles of a large group of people with (pw) multiple sclerosis (MS) compared to those of controls. We reveal that methylation differences in pwMS occur independently of known genetic risk loci and show that they more strongly differentiate disease (AUC = 0.85, 95% CI 0.82-0.89, p = 1.22 × 10-29) than known genetic risk loci (AUC = 0.72, 95% CI: 0.66-0.76, p = 9.07 × 10-17). We also show that methylation differences in MS occur predominantly in B cells and monocytes and indicate the involvement of cell-specific biological pathways. Overall, this study comprehensively characterizes the immune cell-specific epigenetic architecture of MS.
Collapse
Affiliation(s)
- Alexandre Xavier
- School of Biomedical Sciences and Pharmacy, Hunter Medical Research Institute, University of Newcastle, New Lambton Heights, NSW 2305, Australia; (A.X.); (S.M.B.); (R.J.S.)
| | - Vicki E. Maltby
- School of Medicine and Public Health, Hunter Medical Research Institute, University of Newcastle, New Lambton Heights, NSW 2305, Australia; (V.E.M.); (R.A.L.)
- Department of Neurology, John Hunter Hospital, New Lambton Heights, NSW 2305, Australia
| | - Ewoud Ewing
- Department of Clinical Neuroscience, Karolinska Institutet, Center for Molecular Medicine, Karolinska University Hospital, 17176 Stockholm, Sweden; (E.E.); (I.K.); (L.K.); (L.A.); (M.J.)
| | - Maria Pia Campagna
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC 3004, Australia; (M.P.C.); (H.B.); (V.G.J.)
| | - Sean M. Burnard
- School of Biomedical Sciences and Pharmacy, Hunter Medical Research Institute, University of Newcastle, New Lambton Heights, NSW 2305, Australia; (A.X.); (S.M.B.); (R.J.S.)
| | - Jesper N. Tegner
- Biological and Environmental Science and Engineering Division, King Abdullah University of Science and Technology (KAUST), Thuwal 23955-6900, Saudi Arabia;
- Computer, Electrical and Mathematical Sciences and Engineering Division, King Abdullah University of Science and Technology (KAUST), Thuwal 23955-6900, Saudi Arabia
- Unit of Computational Medicine, Department of Medicine, Center for Molecular Medicine, Karolinska Institutet, Karolinska University Hospital, L8:05, 17176 Stockholm, Sweden
- Science for Life Laboratory, Tomtebodavagen 23A, 17165 Solna, Sweden
| | - Mark Slee
- College of Medicine and Public Health, Flinders University, Bedford Park, SA 5042, Australia;
| | - Helmut Butzkueven
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC 3004, Australia; (M.P.C.); (H.B.); (V.G.J.)
- MSBase Foundation, Melbourne, VIC 3004, Australia
| | - Ingrid Kockum
- Department of Clinical Neuroscience, Karolinska Institutet, Center for Molecular Medicine, Karolinska University Hospital, 17176 Stockholm, Sweden; (E.E.); (I.K.); (L.K.); (L.A.); (M.J.)
| | - Lara Kular
- Department of Clinical Neuroscience, Karolinska Institutet, Center for Molecular Medicine, Karolinska University Hospital, 17176 Stockholm, Sweden; (E.E.); (I.K.); (L.K.); (L.A.); (M.J.)
| | | | - Vilija G. Jokubaitis
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC 3004, Australia; (M.P.C.); (H.B.); (V.G.J.)
| | - Trevor Kilpatrick
- Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Melbourne, VIC 3052, Australia; (T.K.); (A.-L.P.)
| | - Lars Alfredsson
- Department of Clinical Neuroscience, Karolinska Institutet, Center for Molecular Medicine, Karolinska University Hospital, 17176 Stockholm, Sweden; (E.E.); (I.K.); (L.K.); (L.A.); (M.J.)
| | - Maja Jagodic
- Department of Clinical Neuroscience, Karolinska Institutet, Center for Molecular Medicine, Karolinska University Hospital, 17176 Stockholm, Sweden; (E.E.); (I.K.); (L.K.); (L.A.); (M.J.)
| | - Anne-Louise Ponsonby
- Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Melbourne, VIC 3052, Australia; (T.K.); (A.-L.P.)
- National Centre for Epidemiology and Public Health, Australian National University, Canberra, ACT 2601, Australia
| | - Bruce V. Taylor
- Menzies Institute for Medical Research, University of Tasmania, Hobart, TAS 7000, Australia;
| | - Rodney J. Scott
- School of Biomedical Sciences and Pharmacy, Hunter Medical Research Institute, University of Newcastle, New Lambton Heights, NSW 2305, Australia; (A.X.); (S.M.B.); (R.J.S.)
- Department of Molecular Genetics, Pathology North, John Hunter Hospital, New Lambton Heights, NSW 2305, Australia
| | - Rodney A. Lea
- School of Medicine and Public Health, Hunter Medical Research Institute, University of Newcastle, New Lambton Heights, NSW 2305, Australia; (V.E.M.); (R.A.L.)
- Centre for Genomics and Personalised Health, School of Biomedical Science, Queensland University of Technology, Kelvin Grove, QLD 4059, Australia
| | - Jeannette Lechner-Scott
- School of Medicine and Public Health, Hunter Medical Research Institute, University of Newcastle, New Lambton Heights, NSW 2305, Australia; (V.E.M.); (R.A.L.)
- Department of Neurology, John Hunter Hospital, New Lambton Heights, NSW 2305, Australia
| |
Collapse
|
13
|
Tolaymat S, Sharma K, Kagzi Y, Sriwastava S. Anti-CD20 monoclonal antibody (mAb) therapy and colitis: A case series and review. Mult Scler Relat Disord 2023; 75:104763. [PMID: 37229799 DOI: 10.1016/j.msard.2023.104763] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2023] [Revised: 05/04/2023] [Accepted: 05/14/2023] [Indexed: 05/27/2023]
Abstract
The US Food and Drug Administration (FDA) recently issued a warning regarding ocrelizumab due to reports of colitis among patients taking this medication. Since it is the only FDA-approved therapy for primary progressive multiple sclerosis (PPMS), further research on this adverse event is necessary, and healthcare professionals should be informed of potential treatment options. In this review, we summarize the available data on the incidence of inflammatory colitis associated with anti-CD20 monoclonal antibodies (mAbs), such as ocrelizumab and rituximab, used in MS treatment. Although the exact pathophysiology of anti-CD20-induced colitis remains unknown, immunological dysregulation through treatment-mediated B-cell depletion has been proposed as a possible mechanism. Our study highlights the importance of clinicians being aware of this potential side effect, and patients taking these medications should be closely monitored for any new-onset gastrointestinal symptoms or diarrheal illness. Research indicates that prompt intervention with endoscopic examination and medical or surgical therapies can ensure timely and effective management, thus improving patient outcomes. However, large-scale studies are still needed to determine the associated risk factors and to establish definitive guidelines for the clinical evaluation of MS patients on anti-CD20 medications.
Collapse
Affiliation(s)
- Sarah Tolaymat
- Department of Neurology, School of Medicine, West Virginia University, Morgantown, WV, USA
| | - Kanika Sharma
- Division of Multiple Sclerosis and Neuroimmunology Department of Neurology, McGovern Medical School (UT Health), University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Yusuf Kagzi
- Mahatma Gandhi Memorial Medical College, Indore, India
| | - Shitiz Sriwastava
- Division of Multiple Sclerosis and Neuroimmunology Department of Neurology, McGovern Medical School (UT Health), University of Texas Health Science Center at Houston, Houston, TX, USA; West Virginia Clinical Transitional Science, Morgantown, WV, USA.
| |
Collapse
|
14
|
Techa-Angkoon P, Siritho S, Tisavipat N, Suansanae T. Current evidence of rituximab in the treatment of multiple sclerosis. Mult Scler Relat Disord 2023; 75:104729. [PMID: 37148577 DOI: 10.1016/j.msard.2023.104729] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 04/15/2023] [Accepted: 04/22/2023] [Indexed: 05/08/2023]
Abstract
Multiple sclerosis (MS) is a chronic inflammatory demyelinating disorder of the central nervous system. The immunopathology of MS involves both T and B lymphocytes. Rituximab is one of the anti-CD20 monoclonal antibody therapies which deplete B-cells. Although some anti-CD20 therapies have been approved by the Food and Drug Administration for treatment of MS, rituximab is used off-label. Several studies have shown that rituximab has a good efficacy and safety in MS, including certain specific patient conditions such as treatment-naïve patients, treatment-switching patients, and the Asian population. However, there are still questions about the optimal dose and duration of rituximab in MS due to the different dosing regimens used in each study. Moreover, many biosimilars have become available at a lower cost with comparable physicochemical properties, pharmacokinetics, pharmacodynamics, efficacy, safety, and immunogenicity. Thus, rituximab may be considered as a potential therapeutic option for patients without access to standard treatment. This narrative review summarized the evidence of both original and biosimilars of rituximab in MS treatment including pharmacokinetics, pharmacodynamics, clinical efficacy, safety, and dosing regimen.
Collapse
Affiliation(s)
- Phanutgorn Techa-Angkoon
- Division of Pharmacy Practice, Faculty of Pharmaceutical Sciences, Ubon Ratchathani University, Ubon Ratchathani, Thailand
| | - Sasitorn Siritho
- Division of Neurology, Department of Medicine, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand; Siriraj Neuroimmunology Center, Division of Neurology, Department of Medicine, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand; Bumrungrad International Hospital, Bangkok, Thailand
| | | | - Thanarat Suansanae
- Division of Clinical Pharmacy, Department of Pharmacy, Faculty of Pharmacy, Mahidol University, 447 Sri Ayutthaya Road, Ratchathewi, Bangkok 10400, Thailand.
| |
Collapse
|
15
|
Freeman L, Longbrake EE, Coyle PK, Hendin B, Vollmer T. High-Efficacy Therapies for Treatment-Naïve Individuals with Relapsing-Remitting Multiple Sclerosis. CNS Drugs 2022; 36:1285-1299. [PMID: 36350491 PMCID: PMC9645316 DOI: 10.1007/s40263-022-00965-7] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/05/2022] [Indexed: 11/11/2022]
Abstract
There are > 18 distinct disease-modifying therapy (DMT) options covering 10 mechanisms of action currently approved by the US Food and Drug Administration for the treatment of relapsing-remitting multiple sclerosis (RRMS). Given the multitude of available treatment options, and recent international consensus guidelines offering differing recommendations, there is broad heterogeneity in how the DMTs are used in clinical practice. Choosing a DMT for newly diagnosed patients with MS is currently a topic of significant debate in MS care. Historically, an escalation approach to DMT was used for newly diagnosed patients with RRMS. However, the evidence for clinical benefits of early treatment with high-efficacy therapies (HETs) in this population is emerging. In this review, we provide an overview of the DMT options and MS treatment strategies, and discuss the clinical benefits of HETs (including ofatumumab, ocrelizumab, natalizumab, alemtuzumab, and cladribine) in the early stages of MS, along with safety concerns associated with these DMTs. By minimizing the accumulation of neurological damage early in the disease course, early treatment with HETs may enhance long-term clinical outcomes over the lifetime of the patient.
Collapse
Affiliation(s)
- Léorah Freeman
- Department of Neurology, Dell Medical School, The University of Texas at Austin, 1601 Trinity St, Austin, TX, 78701, USA.
| | | | - Patricia K Coyle
- Department of Neurology, Stony Brook University Medical Center, Stony Brook, NY, USA
| | - Barry Hendin
- Banner, University Medicine Neurosciences Clinic, Phoenix, AZ, USA
| | - Timothy Vollmer
- Department of Neurology, University of Colorado, Anschutz Medical Campus, Aurora, CO, USA
| |
Collapse
|
16
|
Fissolo N, Pappolla A, Rio J, Villar LM, Perez-Hoyos S, Sanchez A, Gutierrez L, Montalban X, Comabella M. Serum Levels of CXCL13 Are Associated With Teriflunomide Response in Patients With Multiple Sclerosis. NEUROLOGY(R) NEUROIMMUNOLOGY & NEUROINFLAMMATION 2022; 10:10/1/e200050. [PMID: 36411079 PMCID: PMC9679885 DOI: 10.1212/nxi.0000000000200050] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Accepted: 09/07/2022] [Indexed: 11/22/2022]
Abstract
BACKGROUND AND OBJECTIVES To identify biomarkers associated with treatment response in patients with multiple sclerosis (MS) treated with the oral therapies teriflunomide, dimethyl fumarate (DMF), and fingolimod. METHODS Serum levels of IL-6, IL-17, TNF-α, granulocyte-macrophage colony-stimulating factor, IL-10, interferon-gamma (IFN-γ) IL-1β, and chemokine ligand 13 (CXCL13) were measured at baseline and 12 months with single molecule array (Simoa) assays in a cohort of patients with MS treated with teriflunomide (N = 19), DMF (N = 22), and fingolimod (N = 25) and classified into "no evidence of disease activity" (NEDA) and EDA patients after 1 year of treatment. RESULTS Serum CXCL13 and TNF-α levels were significantly decreased after treatment with teriflunomide in NEDA compared with EDA patients after 1 year of treatment (p = 0.008 for both cytokines). These findings were validated in an independent cohort of patients with MS treated with teriflunomide (N = 36) and serum CXCL13, and TNF-α levels were again significantly reduced in NEDA patients (p < 0.0001 for CXCL13 and p = 0.003 for TNF-α). CXCL13, but not TNF-α, showed good performance to classify NEDA and EDA patients according to a cut-off value of 9.64 pg/mL based on the change in CXCL13 levels between baseline and 12 months, with a sensitivity of 75% and specificity of 82% in the original cohort, and sensitivity of 65.4% and specificity of 60% in the validation cohort. DISCUSSION Altogether, these results point to CXCL13 as a treatment response biomarker to teriflunomide in relapsing-remitting patients with MS, and the change in CXCL13 levels during the first year of treatment can be used in clinical practice to identify optimal responders to teriflunomide.
Collapse
Affiliation(s)
- Nicolás Fissolo
- From the Servei de Neurologia, Centre d'Esclerosi Múltiple de Catalunya, Institut de Recerca Vall d'Hebron (N.F., A.P., J.R., L.G., X.M., M.C.); Departments of Immunology and Neurology (L.M.V.), Multiple Sclerosis Unit, Hospital Ramon y Cajal, (IRYCIS), Madrid; Statistics and Bioinformatics Unit, Vall d'Hebron Institut de Recerca (VHIR) (S.P.-H., A.S.); and Genetics, Microbiology and Statistics Department (A.S.), Universitat de Barcelona, Spain.
| | - Agustin Pappolla
- From the Servei de Neurologia, Centre d'Esclerosi Múltiple de Catalunya, Institut de Recerca Vall d'Hebron (N.F., A.P., J.R., L.G., X.M., M.C.); Departments of Immunology and Neurology (L.M.V.), Multiple Sclerosis Unit, Hospital Ramon y Cajal, (IRYCIS), Madrid; Statistics and Bioinformatics Unit, Vall d'Hebron Institut de Recerca (VHIR) (S.P.-H., A.S.); and Genetics, Microbiology and Statistics Department (A.S.), Universitat de Barcelona, Spain
| | - Jordi Rio
- From the Servei de Neurologia, Centre d'Esclerosi Múltiple de Catalunya, Institut de Recerca Vall d'Hebron (N.F., A.P., J.R., L.G., X.M., M.C.); Departments of Immunology and Neurology (L.M.V.), Multiple Sclerosis Unit, Hospital Ramon y Cajal, (IRYCIS), Madrid; Statistics and Bioinformatics Unit, Vall d'Hebron Institut de Recerca (VHIR) (S.P.-H., A.S.); and Genetics, Microbiology and Statistics Department (A.S.), Universitat de Barcelona, Spain
| | - Luisa M Villar
- From the Servei de Neurologia, Centre d'Esclerosi Múltiple de Catalunya, Institut de Recerca Vall d'Hebron (N.F., A.P., J.R., L.G., X.M., M.C.); Departments of Immunology and Neurology (L.M.V.), Multiple Sclerosis Unit, Hospital Ramon y Cajal, (IRYCIS), Madrid; Statistics and Bioinformatics Unit, Vall d'Hebron Institut de Recerca (VHIR) (S.P.-H., A.S.); and Genetics, Microbiology and Statistics Department (A.S.), Universitat de Barcelona, Spain
| | - Santiago Perez-Hoyos
- From the Servei de Neurologia, Centre d'Esclerosi Múltiple de Catalunya, Institut de Recerca Vall d'Hebron (N.F., A.P., J.R., L.G., X.M., M.C.); Departments of Immunology and Neurology (L.M.V.), Multiple Sclerosis Unit, Hospital Ramon y Cajal, (IRYCIS), Madrid; Statistics and Bioinformatics Unit, Vall d'Hebron Institut de Recerca (VHIR) (S.P.-H., A.S.); and Genetics, Microbiology and Statistics Department (A.S.), Universitat de Barcelona, Spain
| | - Alex Sanchez
- From the Servei de Neurologia, Centre d'Esclerosi Múltiple de Catalunya, Institut de Recerca Vall d'Hebron (N.F., A.P., J.R., L.G., X.M., M.C.); Departments of Immunology and Neurology (L.M.V.), Multiple Sclerosis Unit, Hospital Ramon y Cajal, (IRYCIS), Madrid; Statistics and Bioinformatics Unit, Vall d'Hebron Institut de Recerca (VHIR) (S.P.-H., A.S.); and Genetics, Microbiology and Statistics Department (A.S.), Universitat de Barcelona, Spain
| | - Lucía Gutierrez
- From the Servei de Neurologia, Centre d'Esclerosi Múltiple de Catalunya, Institut de Recerca Vall d'Hebron (N.F., A.P., J.R., L.G., X.M., M.C.); Departments of Immunology and Neurology (L.M.V.), Multiple Sclerosis Unit, Hospital Ramon y Cajal, (IRYCIS), Madrid; Statistics and Bioinformatics Unit, Vall d'Hebron Institut de Recerca (VHIR) (S.P.-H., A.S.); and Genetics, Microbiology and Statistics Department (A.S.), Universitat de Barcelona, Spain
| | - Xavier Montalban
- From the Servei de Neurologia, Centre d'Esclerosi Múltiple de Catalunya, Institut de Recerca Vall d'Hebron (N.F., A.P., J.R., L.G., X.M., M.C.); Departments of Immunology and Neurology (L.M.V.), Multiple Sclerosis Unit, Hospital Ramon y Cajal, (IRYCIS), Madrid; Statistics and Bioinformatics Unit, Vall d'Hebron Institut de Recerca (VHIR) (S.P.-H., A.S.); and Genetics, Microbiology and Statistics Department (A.S.), Universitat de Barcelona, Spain
| | - Manuel Comabella
- From the Servei de Neurologia, Centre d'Esclerosi Múltiple de Catalunya, Institut de Recerca Vall d'Hebron (N.F., A.P., J.R., L.G., X.M., M.C.); Departments of Immunology and Neurology (L.M.V.), Multiple Sclerosis Unit, Hospital Ramon y Cajal, (IRYCIS), Madrid; Statistics and Bioinformatics Unit, Vall d'Hebron Institut de Recerca (VHIR) (S.P.-H., A.S.); and Genetics, Microbiology and Statistics Department (A.S.), Universitat de Barcelona, Spain
| |
Collapse
|
17
|
DeMaio A, Mehrotra S, Sambamurti K, Husain S. The role of the adaptive immune system and T cell dysfunction in neurodegenerative diseases. J Neuroinflammation 2022; 19:251. [PMID: 36209107 PMCID: PMC9548183 DOI: 10.1186/s12974-022-02605-9] [Citation(s) in RCA: 59] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Accepted: 09/25/2022] [Indexed: 11/10/2022] Open
Abstract
The adaptive immune system and associated inflammation are vital in surveillance and host protection against internal and external threats, but can secondarily damage host tissues. The central nervous system is immune-privileged and largely protected from the circulating inflammatory pathways. However, T cell involvement and the disruption of the blood-brain barriers have been linked to several neurodegenerative diseases including Parkinson's disease, Alzheimer's disease, and multiple sclerosis. Under normal physiological conditions, regulatory T cells (Treg cells) dampen the inflammatory response of effector T cells. In the pathological states of many neurodegenerative disorders, the ability of Treg cells to mitigate inflammation is reduced, and a pro-inflammatory environment persists. This perspective review provides current knowledge on the roles of T cell subsets (e.g., effector T cells, Treg cells) in neurodegenerative and ocular diseases, including uveitis, diabetic retinopathy, age-related macular degeneration, and glaucoma. Many neurodegenerative and ocular diseases have been linked to immune dysregulation, but the cellular events and molecular mechanisms involved in such processes remain largely unknown. Moreover, the role of T cells in ocular pathologies remains poorly defined and limited literature is available in this area of research. Adoptive transfer of Treg cells appears to be a vital immunological approach to control ocular pathologies. Similarities in T cell dysfunction seen among non-ocular neurodegenerative diseases suggest that this area of research has a great potential to develop better therapeutic agents for ocular diseases and warrants further studies. Overall, this perspective review article provides significant information on the roles of T cells in numerous ocular and non-ocular neurodegenerative diseases.
Collapse
Affiliation(s)
- Alexa DeMaio
- Department of Ophthalmology, Storm Eye Institute, Room 713, Medical University of South Carolina, 167 Ashley Ave, SC, 29425, Charleston, USA
| | - Shikhar Mehrotra
- Department of Surgery, Hollings Cancer Center, Medical University of South Carolina, SC, 29425, Charleston, USA
| | - Kumar Sambamurti
- Department of Neuroscience, Medical University of South Carolina, SC, 29425, Charleston, USA
| | - Shahid Husain
- Department of Ophthalmology, Storm Eye Institute, Room 713, Medical University of South Carolina, 167 Ashley Ave, SC, 29425, Charleston, USA.
| |
Collapse
|
18
|
Chen TX, Fan YT, Peng BW. Distinct mechanisms underlying therapeutic potentials of CD20 in neurological and neuromuscular disease. Pharmacol Ther 2022; 238:108180. [DOI: 10.1016/j.pharmthera.2022.108180] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Revised: 03/16/2022] [Accepted: 03/28/2022] [Indexed: 10/18/2022]
|
19
|
A Targeted Literature Search and Phenomenological Review of Perspectives of People with Multiple Sclerosis and Healthcare Professionals of the Immunology of Disease-Modifying Therapies. Neurol Ther 2022; 11:955-979. [PMID: 35608740 PMCID: PMC9127487 DOI: 10.1007/s40120-022-00349-5] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Accepted: 03/25/2022] [Indexed: 11/29/2022] Open
Abstract
Introduction The mechanisms of action of disease-modifying therapies (DMTs) for multiple sclerosis (MS) are complex and involve an interplay of immune system components. People with MS (PwMS) may lack a clear understanding of the immunological pathways involved in MS and its treatment; effective communication between healthcare professionals (HCPs) and PwMS is needed to facilitate shared decision-making when discussing the disease and selecting DMTs and is particularly important in the coronavirus disease 2019 (COVID-19) era. Methods In this patient-authored two-part review, we performed a targeted literature search to assess the need for better communication between HCPs and PwMS regarding treatment selection, and also conducted a qualitative survey of four patient and care-partner authors to obtain insights regarding their understanding of and preferences for the treatment and management of MS. Results Following a search of the Embase and MEDLINE databases using Ovid in June 2020, an analysis of 40 journal articles and conference abstracts relating to patient empowerment and decision-making in DMT selection for MS showed a preference for safety and efficacy of treatments, followed by autonomy and convenience of administration. A need for better communication between HCPs and PwMS during treatment selection to improve patient satisfaction was also identified. The open survey responses from the patient authors revealed a need for greater involvement in decision-making processes and desire for improved communication and information tools. Conclusions This targeted literature search and phenomenological review confirms PwMS preferences for empowered decision-making in disease management and treatment selection, to optimize independence, safety, and efficacy. It also identifies an unmet need for improved communication and information tools that convey MS information in a relatable manner. Furthermore, this review seeks to address this unmet need by providing plain language figures and descriptions of MS immune mechanisms that can be used to facilitate discussions between HCPs and PwMS. Supplementary Information The online version contains supplementary material available at 10.1007/s40120-022-00349-5. In multiple sclerosis (MS), there are different cells in the immune system that contribute to the disease. The main cells in the immune system are T and B cells. People with MS (PwMS) might not be familiar with details about the immune system, and healthcare professionals might not always communicate details about how treatments work clearly to PwMS when choosing treatments with them. It is important for PwMS to have all the information they need to help make decisions about treatments. This information needs to be given in a way they can understand. This is especially important during the coronavirus disease 2019 (COVID-19) pandemic. In this paper, we first looked at what research has already been published about what is most important to PwMS when making treatment decisions. The existing research says that safety and effectiveness are the most important things and that PwMS prefer treatments that they can take themselves. PwMS also need better communication and information from doctors to make decisions and to help explain how MS treatments work in the body. Next, we gave a survey to the patients who are authors of this paper to ask about what is important to them when making treatment decisions. Their answers were very similar to the existing research. Overall, PwMS need better communication from healthcare professionals about the immune system. This paper also includes plain language descriptions and figures to help healthcare professionals explain and discuss the importance of the immune system in MS with PwMS.
Collapse
|
20
|
Baker D, MacDougall A, Kang AS, Schmierer K, Giovannoni G, Dobson R. Seroconversion following COVID-19 vaccination: can we optimize protective response in CD20-treated individuals? Clin Exp Immunol 2022; 207:263-271. [PMID: 35553629 PMCID: PMC9113152 DOI: 10.1093/cei/uxab015] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Revised: 10/28/2021] [Accepted: 11/05/2021] [Indexed: 12/14/2022] Open
Abstract
Although there is an ever-increasing number of disease-modifying treatments for relapsing multiple sclerosis (MS), few appear to influence coronavirus disease 2019 (COVID-19) severity. There is concern about the use of anti-CD20-depleting monoclonal antibodies, due to the apparent increased risk of severe disease following severe acute respiratory syndrome corona virus two (SARS-CoV-2) infection and inhibition of protective anti-COVID-19 vaccine responses. These antibodies are given as maintenance infusions/injections and cause persistent depletion of CD20+ B cells, notably memory B-cell populations that may be instrumental in the control of relapsing MS. However, they also continuously deplete immature and mature/naïve B cells that form the precursors for infection-protective antibody responses, thus blunting vaccine responses. Seroconversion and maintained SARS-CoV-2 neutralizing antibody levels provide protection from COVID-19. However, it is evident that poor seroconversion occurs in the majority of individuals following initial and booster COVID-19 vaccinations, based on standard 6 monthly dosing intervals. Seroconversion may be optimized in the anti-CD20-treated population by vaccinating prior to treatment onset or using extended/delayed interval dosing (3-6 month extension to dosing interval) in those established on therapy, with B-cell monitoring until (1-3%) B-cell repopulation occurs prior to vaccination. Some people will take more than a year to replete and therefore protection may depend on either the vaccine-induced T-cell responses that typically occur or may require prophylactic, or rapid post-infection therapeutic, antibody or small-molecule antiviral treatment to optimize protection against COVID-19. Further studies are warranted to demonstrate the safety and efficacy of such approaches and whether or not immunity wanes prematurely as has been observed in the other populations.
Collapse
Affiliation(s)
- David Baker
- The Blizard Institute, Barts and The London School of Medicine & Dentistry, Queen Mary University of London, London, UK
| | - Amy MacDougall
- Medical Statistics, London School of Hygiene and Tropical Medicine, London, UK
| | - Angray S Kang
- The Blizard Institute, Barts and The London School of Medicine & Dentistry, Queen Mary University of London, London, UK
- Centre for Oral Immunobiology and Regenerative Medicine, Dental Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Klaus Schmierer
- The Blizard Institute, Barts and The London School of Medicine & Dentistry, Queen Mary University of London, London, UK
- Clinical Board Medicine (Neuroscience), The Royal London Hospital, Barts Health NHS Trust, London, UK
| | - Gavin Giovannoni
- The Blizard Institute, Barts and The London School of Medicine & Dentistry, Queen Mary University of London, London, UK
- Clinical Board Medicine (Neuroscience), The Royal London Hospital, Barts Health NHS Trust, London, UK
| | - Ruth Dobson
- Clinical Board Medicine (Neuroscience), The Royal London Hospital, Barts Health NHS Trust, London, UK
- Preventive Neurology Unit, Wolfson Institute of Population Health, Queen Mary University of London, Barts and The London School of Medicine & Dentistry, London, UK
| |
Collapse
|
21
|
Zecca C, Gobbi C. Long-term treatment with anti-CD20 monoclonal antibodies is untenable because of risk: YES. Mult Scler 2022; 28:1173-1175. [PMID: 35411821 DOI: 10.1177/13524585221088734] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Affiliation(s)
- Chiara Zecca
- Multiple Sclerosis Center, Neurocenter of Southern Switzerland, Ente Ospedaliero Cantonale, Lugano, Switzerland/Faculty of Biomedical Sciences, Università della Svizzera Italiana, Lugano, Switzerland
| | - Claudio Gobbi
- Multiple Sclerosis Center, Neurocenter of Southern Switzerland, Ente Ospedaliero Cantonale, Lugano, Switzerland/Faculty of Biomedical Sciences, Università della Svizzera Italiana, Lugano, Switzerland
| |
Collapse
|
22
|
Zhang X, Sun Y, Wang N, Zhang Y, Xia Y, Liu Y. Immunomodulatory Treatment Strategies Targeting B Cells for Heart Failure. Front Pharmacol 2022; 13:854592. [PMID: 35350762 PMCID: PMC8957947 DOI: 10.3389/fphar.2022.854592] [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: 01/14/2022] [Accepted: 02/18/2022] [Indexed: 11/13/2022] Open
Abstract
Cardio-oncology, a nascent specialty, has evolved as a concerted strategy to address the cardiovascular complications of cancer therapies. On the other hand, emerging evidence has shown that some anti-tumor drugs, such as CD20-targeted rotuximab, also have markedly cardioprotective effects in addition to treating cancers. Rituximab is a CD20-targeted monoclonal antibody and kill tumor B-cells through antibody-mediated and antibody-independent pathways, indicating that B cells participate and promote the progression of cardiovascular diseases. In this review, we mainly present the evidence that B cells contribute to the development of hypertrophy, inflammation, and maladaptive tissue remodeling, with the aim of proposing novel immunomodulatory therapeutic strategies targeting B cells and their products for the treatment of heart failure.
Collapse
Affiliation(s)
- Xinxin Zhang
- Heart Failure and Structural Cardiology Division, First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Yuxi Sun
- Heart Failure and Structural Cardiology Division, First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Ning Wang
- Heart Failure and Structural Cardiology Division, First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Yanli Zhang
- Heart Failure and Structural Cardiology Division, First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Yunlong Xia
- Heart Failure and Structural Cardiology Division, First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Ying Liu
- Heart Failure and Structural Cardiology Division, First Affiliated Hospital of Dalian Medical University, Dalian, China
| |
Collapse
|
23
|
Abstract
Ocrelizumab (Ocrevus®) is an intravenously administered, humanized anti-CD20 monoclonal antibody approved for the treatment of adults with relapsing forms of multiple sclerosis (RMS) or primary progressive multiple sclerosis (PPMS). The efficacy of ocrelizumab in reducing relapse rates and disease activity in patients with RMS was demonstrated in pivotal trials (versus interferon β-1a) and supporting single-arm studies in specific subpopulations. In patients with PPMS, ocrelizumab reduced measures of clinical and MRI progression relative to placebo. Clinical benefits were maintained over ≥ 7.5 study years of treatment. Ocrelizumab was generally well tolerated and no new safety signals have emerged with long-term use. Extensive (albeit short-term) real-world data pertaining to ocrelizumab is consistent with that from clinical trials. Ocrelizumab provides the convenience of short, half-yearly infusions. Ocrelizumab continues to represent a generally well-tolerated, high-efficacy disease-modifying therapy (DMT) for RMS and is a valuable treatment for delaying disease progression in patients with PPMS (for whom there are currently no other approved DMTs).
Collapse
Affiliation(s)
- Yvette N Lamb
- Springer Nature, Mairangi Bay, Private Bag 65901, Auckland, 0754, New Zealand.
| |
Collapse
|
24
|
Liu J, Yang X, Pan J, Wei Z, Liu P, Chen M, Liu H. Single-Cell Transcriptome Profiling Unravels Distinct Peripheral Blood Immune Cell Signatures of RRMS and MOG Antibody-Associated Disease. Front Neurol 2022; 12:807646. [PMID: 35095746 PMCID: PMC8795627 DOI: 10.3389/fneur.2021.807646] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Accepted: 12/16/2021] [Indexed: 12/11/2022] Open
Abstract
Relapsing-remitting multiple sclerosis (RRMS) and myelin oligodendrocyte glycoprotein (MOG) antibody-associated disease (MOGAD) are inflammatory demyelinating diseases of the central nervous system (CNS). Due to the shared clinical manifestations, detection of disease-specific serum antibody of the two diseases is currently considered as the gold standard for the diagnosis; however, the serum antibody levels are unpredictable during different stages of the two diseases. Herein, peripheral blood single-cell transcriptome was used to unveil distinct immune cell signatures of the two diseases, with the aim to provide predictive discrimination. Single-cell RNA sequencing (scRNA-seq) was conducted on the peripheral blood from three subjects, i.e., one patient with RRMS, one patient with MOGAD, and one patient with healthy control. The results showed that the CD19+ CXCR4+ naive B cell subsets were significantly expanded in both RRMS and MOGAD, which was verified by flow cytometry. More importantly, RRMS single-cell transcriptomic was characterized by increased naive CD8+ T cells and cytotoxic memory-like Natural Killer (NK) cells, together with decreased inflammatory monocytes, whereas MOGAD exhibited increased inflammatory monocytes and cytotoxic CD8 effector T cells, coupled with decreased plasma cells and memory B cells. Collectively, our findings indicate that the two diseases exhibit distinct immune cell signatures, which allows for highly predictive discrimination of the two diseases and paves a novel avenue for diagnosis and therapy of neuroinflammatory diseases.
Collapse
Affiliation(s)
- Ju Liu
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xiaoyan Yang
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Jiali Pan
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Zhihua Wei
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Peidong Liu
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Min Chen
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Hongbo Liu
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- *Correspondence: Hongbo Liu
| |
Collapse
|
25
|
Yu H, Graham G, David OJ, Kahn JM, Savelieva M, Pigeolet E, Das Gupta A, Pingili R, Willi R, Ramanathan K, Kieseier BC, Häring DA, Bagger M, Soelberg Sørensen P. Population Pharmacokinetic-B Cell Modeling for Ofatumumab in Patients with Relapsing Multiple Sclerosis. CNS Drugs 2022; 36:283-300. [PMID: 35233753 PMCID: PMC8927028 DOI: 10.1007/s40263-021-00895-w] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/21/2021] [Indexed: 01/11/2023]
Abstract
BACKGROUND Ofatumumab, a fully human anti-CD20 monoclonal antibody indicated for the treatment of relapsing forms of multiple sclerosis (RMS), binds to a unique conformational epitope, thereby depleting B cells very efficiently and allowing subcutaneous administration at lower doses. OBJECTIVES The aims were to characterize the relationship between ofatumumab concentration and B cell levels, including the effect of covariates such as body weight, age, or baseline B cell count, and use simulations to confirm the chosen therapeutic dose. METHODS Graphical and regression analyses previously performed based on data from a dose-range finding study provided the B cell depletion target used in the present work. All available adult phase 2/3 data for ofatumumab in RMS patients were pooled to develop a population pharmacokinetics (PK)-B cell count model, using nonlinear mixed-effects modeling. The population PK-B cell model was used to simulate B cell depletion and repletion times and the effect of covariates on PK and B cell metrics, as well as the dose response across a range of subcutaneous ofatumumab monthly doses. RESULTS The final PK-B cell model was developed using data from 1486 patients. The predetermined B cell target was best achieved and sustained with the 20-mg dose regimen, with median B cell count reaching 8 cells/µL in 11 days and negligible repletion between doses. Only weight had a significant effect on PK, which did not translate into any clinically relevant effect on B cell levels. CONCLUSION The PK-B cell modeling confirms the dose chosen for the licensed ofatumumab regimen and demonstrates no requirement for dose adjustment based on adult patient characteristics.
Collapse
Affiliation(s)
- Huixin Yu
- Novartis Pharma AG, Postfach CH-4002, Basel, Switzerland
| | - Gordon Graham
- Novartis Pharma AG, Postfach CH-4002, Basel, Switzerland.
| | | | - Joseph M Kahn
- Novartis Pharmaceuticals Corporation, East Hanover, NJ, USA
| | | | | | | | | | - Roman Willi
- Novartis Pharma AG, Postfach CH-4002, Basel, Switzerland
| | | | - Bernd C Kieseier
- Novartis Pharma AG, Postfach CH-4002, Basel, Switzerland
- Department of Neurology, Medical Faculty, Heinrich-Heine University, Duesseldorf, Germany
| | | | - Morten Bagger
- Novartis Pharma AG, Postfach CH-4002, Basel, Switzerland
| | - Per Soelberg Sørensen
- Department of Neurology, Danish Multiple Sclerosis Center, University of Copenhagen, Rigshospitalet, Copenhagen, Denmark
| |
Collapse
|
26
|
Mado H, Kubicka-Bączyk K, Adamczyk-Sowa M. Anti-severe acute respiratory syndrome coronavirus-2 antibody responses following Pfizer-BioNTech vaccination in a patient with multiple sclerosis treated with ocrelizumab: a case report. J Int Med Res 2021; 49:3000605211044378. [PMID: 34528472 PMCID: PMC8451259 DOI: 10.1177/03000605211044378] [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] [Indexed: 11/25/2022] Open
Abstract
Patients with multiple sclerosis (MS) repeatedly receive therapies that cause B-lymphocyte depletion. This may lead to abnormal immune responses following coronavirus disease 2019 (COVID-19) vaccination, as has been suggested previously. We therefore evaluated post-vaccination immune responses in a patient with MS treated with ocrelizumab. The intervals between ocrelizumab infusions and vaccination were as recommended by the Section of Multiple Sclerosis and Neuroimmunology of the Polish Neurological Society. A reactive immune response was observed in this patient following vaccination. This suggests that appropriate intervals between ocrelizumab infusions and COVID-19 vaccinations may permit the generation of efficacious immune responses in patients receiving B-lymphocyte depleting therapies.
Collapse
Affiliation(s)
- Hubert Mado
- Department of Neurology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia in Katowice, Poland
| | - Katarzyna Kubicka-Bączyk
- Department of Neurology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia in Katowice, Poland
| | - Monika Adamczyk-Sowa
- Department of Neurology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia in Katowice, Poland
| |
Collapse
|
27
|
Lopez JA, Denkova M, Ramanathan S, Dale RC, Brilot F. Pathogenesis of autoimmune demyelination: from multiple sclerosis to neuromyelitis optica spectrum disorders and myelin oligodendrocyte glycoprotein antibody-associated disease. Clin Transl Immunology 2021; 10:e1316. [PMID: 34336206 PMCID: PMC8312887 DOI: 10.1002/cti2.1316] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 06/20/2021] [Accepted: 07/01/2021] [Indexed: 12/16/2022] Open
Abstract
Autoimmunity plays a significant role in the pathogenesis of demyelination. Multiple sclerosis (MS), neuromyelitis optica spectrum disorders (NMOSD) and myelin oligodendrocyte glycoprotein antibody‐associated disease (MOGAD) are now recognised as separate disease entities under the amalgam of human central nervous system demyelinating disorders. While these disorders share inherent similarities, investigations into their distinct clinical presentations and lesion pathologies have aided in differential diagnoses and understanding of disease pathogenesis. An interplay of various genetic and environmental factors contributes to each disease, many of which implicate an autoimmune response. The pivotal role of the adaptive immune system has been highlighted by the diagnostic autoantibodies in NMOSD and MOGAD, and the presence of autoreactive lymphocytes in MS lesions. While a number of autoantigens have been proposed in MS, recent emphasis on the contribution of B cells has shed new light on the well‐established understanding of T cell involvement in pathogenesis. This review aims to synthesise the clinical characteristics and pathological findings, discuss existing and emerging hypotheses regarding the aetiology of demyelination and evaluate recent pathogenicity studies involving T cells, B cells, and autoantibodies and their implications in human demyelination.
Collapse
Affiliation(s)
- Joseph A Lopez
- Brain Autoimmunity Group Kids Neuroscience Centre Kids Research at the Children's Hospital at Westmead Sydney NSW Australia.,Specialty of Child and Adolescent Health Faculty of Medicine and Health The University of Sydney Sydney NSW Australia
| | - Martina Denkova
- Brain Autoimmunity Group Kids Neuroscience Centre Kids Research at the Children's Hospital at Westmead Sydney NSW Australia.,School of Medical Sciences Faculty of Medicine and Health The University of Sydney Sydney NSW Australia
| | - Sudarshini Ramanathan
- Brain Autoimmunity Group Kids Neuroscience Centre Kids Research at the Children's Hospital at Westmead Sydney NSW Australia.,Sydney Medical School Faculty of Medicine and Health The University of Sydney Sydney NSW Australia.,Department of Neurology Concord Hospital Sydney NSW Australia
| | - Russell C Dale
- Brain Autoimmunity Group Kids Neuroscience Centre Kids Research at the Children's Hospital at Westmead Sydney NSW Australia.,Specialty of Child and Adolescent Health Faculty of Medicine and Health The University of Sydney Sydney NSW Australia.,Sydney Medical School Faculty of Medicine and Health The University of Sydney Sydney NSW Australia.,Brain and Mind Centre The University of Sydney Sydney NSW Australia
| | - Fabienne Brilot
- Brain Autoimmunity Group Kids Neuroscience Centre Kids Research at the Children's Hospital at Westmead Sydney NSW Australia.,Specialty of Child and Adolescent Health Faculty of Medicine and Health The University of Sydney Sydney NSW Australia.,School of Medical Sciences Faculty of Medicine and Health The University of Sydney Sydney NSW Australia.,Brain and Mind Centre The University of Sydney Sydney NSW Australia
| |
Collapse
|
28
|
Veroni C, Aloisi F. The CD8 T Cell-Epstein-Barr Virus-B Cell Trialogue: A Central Issue in Multiple Sclerosis Pathogenesis. Front Immunol 2021; 12:665718. [PMID: 34305896 PMCID: PMC8292956 DOI: 10.3389/fimmu.2021.665718] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Accepted: 06/07/2021] [Indexed: 12/11/2022] Open
Abstract
The cause and the pathogenic mechanisms leading to multiple sclerosis (MS), a chronic inflammatory disease of the central nervous system (CNS), are still under scrutiny. During the last decade, awareness has increased that multiple genetic and environmental factors act in concert to modulate MS risk. Likewise, the landscape of cells of the adaptive immune system that are believed to play a role in MS immunopathogenesis has expanded by including not only CD4 T helper cells but also cytotoxic CD8 T cells and B cells. Once the key cellular players are identified, the main challenge is to define precisely how they act and interact to induce neuroinflammation and the neurodegenerative cascade in MS. CD8 T cells have been implicated in MS pathogenesis since the 80's when it was shown that CD8 T cells predominate in MS brain lesions. Interest in the role of CD8 T cells in MS was revived in 2000 and the years thereafter by studies showing that CNS-recruited CD8 T cells are clonally expanded and have a memory effector phenotype indicating in situ antigen-driven reactivation. The association of certain MHC class I alleles with MS genetic risk implicates CD8 T cells in disease pathogenesis. Moreover, experimental studies have highlighted the detrimental effects of CD8 T cell activation on neural cells. While the antigens responsible for T cell recruitment and activation in the CNS remain elusive, the high efficacy of B-cell depleting drugs in MS and a growing number of studies implicate B cells and Epstein-Barr virus (EBV), a B-lymphotropic herpesvirus that is strongly associated with MS, in the activation of pathogenic T cells. This article reviews the results of human studies that have contributed to elucidate the role of CD8 T cells in MS immunopathogenesis, and discusses them in light of current understanding of autoreactivity, B-cell and EBV involvement in MS, and mechanism of action of different MS treatments. Based on the available evidences, an immunopathological model of MS is proposed that entails a persistent EBV infection of CNS-infiltrating B cells as the target of a dysregulated cytotoxic CD8 T cell response causing CNS tissue damage.
Collapse
Affiliation(s)
| | - Francesca Aloisi
- Department of Neuroscience, Istituto Superiore di Sanità, Rome, Italy
| |
Collapse
|
29
|
Erturk-Hasdemir D, Ochoa-Repáraz J, Kasper DL, Kasper LH. Exploring the Gut-Brain Axis for the Control of CNS Inflammatory Demyelination: Immunomodulation by Bacteroides fragilis' Polysaccharide A. Front Immunol 2021; 12:662807. [PMID: 34025663 PMCID: PMC8131524 DOI: 10.3389/fimmu.2021.662807] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Accepted: 04/16/2021] [Indexed: 12/12/2022] Open
Abstract
The symbiotic relationship between animals and their resident microorganisms has profound effects on host immunity. The human microbiota comprises bacteria that reside in the gastrointestinal tract and are involved in a range of inflammatory and autoimmune diseases. The gut microbiota's immunomodulatory effects extend to extraintestinal tissues, including the central nervous system (CNS). Specific symbiotic antigens responsible for inducing immunoregulation have been isolated from different bacterial species. Polysaccharide A (PSA) of Bacteroides fragilis is an archetypical molecule for host-microbiota interactions. Studies have shown that PSA has beneficial effects in experimental disease models, including experimental autoimmune encephalomyelitis (EAE), the most widely used animal model for multiple sclerosis (MS). Furthermore, in vitro stimulation with PSA promotes an immunomodulatory phenotype in human T cells isolated from healthy and MS donors. In this review, we discuss the current understanding of the interactions between gut microbiota and the host in the context of CNS inflammatory demyelination, the immunomodulatory roles of gut symbionts. More specifically, we also discuss the immunomodulatory effects of B. fragilis PSA in the gut-brain axis and its therapeutic potential in MS. Elucidation of the molecular mechanisms responsible for the microbiota's impact on host physiology offers tremendous promise for discovering new therapies.
Collapse
Affiliation(s)
| | | | - Dennis L. Kasper
- Department of Immunology, Harvard Medical School, Boston, MA, United States
| | - Lloyd H. Kasper
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth College, Hanover, NH, United States
| |
Collapse
|
30
|
Machlańska A, Helbig G, Chromik K, Zapała M, Zwiernik B, Selmaj K. Hemophagocytic lymphohistiocytosis associated with ocrelizumab treatment in a patient with multiple sclerosis. Mult Scler 2021; 27:1803-1805. [PMID: 33666121 DOI: 10.1177/1352458521993070] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
BACKGROUND Hemophagocytic lymphohistiocytosis (HLH) is a rarely recognized hyperinflammatory condition of high death risk. OBJECTIVE The objective was to describe a case of HLH in a patient with multiple sclerosis (MS) treated with ocrelizumab. METHODS Clinical observation, laboratory testing, and use of HLH-2004 criteria for HLH diagnosis. RESULTS A 32-year-old Caucasian female developed HLH during ocrelizumab treatment. She met six of the eight HLH criteria including fever, splenomegaly, cytopenia, hypertriglyceridemia and hypofibrinogenemia, high serum ferritin level, and low natural killer (NK) cells. CONCLUSION HLH should be considered in the differential diagnosis in MS patients displaying a fever and malaise syndrome following administration of ocrelizumab.
Collapse
Affiliation(s)
| | - Grzegorz Helbig
- Department of Hematology and Bone Marrow Transplantation, Silesian Medical University, Katowice, Poland
| | - Karolina Chromik
- Department of Hematology and Bone Marrow Transplantation, Silesian Medical University, Katowice, Poland
| | - Magdalena Zapała
- Department of Hematology and Bone Marrow Transplantation, Silesian Medical University, Katowice, Poland
| | - Bartosz Zwiernik
- Department of Hematology and Bone Marrow Transplantation, Silesian Medical University, Katowice, Poland
| | - Krzysztof Selmaj
- Center of Neurology, Lodz, Poland/ Department of Neurology, University of Warmia & Mazury Olsztyn, Poland
| |
Collapse
|
31
|
Fernández-Velasco JI, Kuhle J, Monreal E, Meca-Lallana V, Meca-Lallana J, Izquierdo G, Gascón-Giménez F, Sainz de la Maza S, Walo-Delgado PE, Maceski A, Rodríguez-Martín E, Roldán E, Villarrubia N, Saiz A, Blanco Y, Sánchez P, Carreón-Guarnizo E, Aladro Y, Brieva L, Íñiguez C, González-Suárez I, Rodríguez de Antonio LA, Masjuan J, Costa-Frossard L, Villar LM. Effect of Ocrelizumab in Blood Leukocytes of Patients With Primary Progressive MS. NEUROLOGY(R) NEUROIMMUNOLOGY & NEUROINFLAMMATION 2021; 8:e940. [PMID: 33408167 PMCID: PMC7862094 DOI: 10.1212/nxi.0000000000000940] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Accepted: 11/03/2020] [Indexed: 11/15/2022]
Abstract
OBJECTIVE To analyze the changes induced by ocrelizumab in blood immune cells of patients with primary progressive MS (PPMS). METHODS In this multicenter prospective study including 53 patients with PPMS who initiated ocrelizumab treatment, we determined effector, memory, and regulatory cells by flow cytometry at baseline and after 6 months of therapy. Wilcoxon matched paired tests were used to assess differences between baseline and 6 months' results. p Values were corrected using the Bonferroni test. RESULTS Ocrelizumab reduced the numbers of naive and memory B cells (p < 0.0001) and those of B cells producing interleukin (IL)-6, IL-10, granulocyte-macrophage colony-stimulating factor (GM-CSF), and tumor necrosis factor-alpha (TNFα) (p < 0.0001 in all cases). By contrast, the proportions of plasmablasts and B cells producing GM-CSF and TNFα increased significantly, suggesting the need for treatment continuation. We also observed a decrease in CD20+ T-cell numbers (p < 0.0001) and percentages (p < 0.0001), and a clear remodeling of the T-cell compartment characterized by relative increases of the naive/effector ratios in CD4+ (p = 0.002) and CD8+ (p = 0.002) T cells and relative decreases of CD4+ (p = 0.03) and CD8+ (p = 0.004) T cells producing interferon-gamma. Total monocyte numbers increased (p = 0.002), but no changes were observed in those producing inflammatory cytokines. The immunologic variations were associated with a reduction of serum neurofilament light chain (sNfL) levels (p = 0.008). The reduction was observed in patients with Gd-enhanced lesions at baseline and in Gd- patients with baseline sNfL >10 pg/mL. CONCLUSIONS In PPMS, effector B-cell depletion changed T-cell response toward a low inflammatory profile, resulting in decreased sNfL levels.
Collapse
Affiliation(s)
- José I. Fernández-Velasco
- From the Immunology Department (J.I.F.-V., P.E.W.-D., E.R.-M., E.R., N.V., L.M.V.), Ramon y Cajal University Hospital, Madrid, Spain; Neurologic Clinic and Policlinic (J.K., A.M.), Departments of Medicine, Biomedicine, and Clinical Research, University Hospital Basel, University of Basel, Switzerland; Neurology Department (E.M., S.S.d.l.M., J.M., L.C.-F.), Ramon y Cajal University Hospital, Madrid; Neurology Department (V.M.-L., P.S.), La Princesa University Hospital, Madrid; Multiple Sclerosis and Clinical Neuroimmunology Unit (J.M.-L., E.C.-G.), Virgen de la Arrixaca University Hospital, Murcia; Multiple Sclerosis Unit (G.I.), Vithas Nisa Sevilla Hospital; Neurology Department (F.G.-G.), Valencia Clinic University Hospital; Center of Neuroimmunology (A.S., Y.B.), Neurology Department, Clínic of Barcelona Hospital, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), and Institut de Neurociències, Universitat de Barcelona; Neurology Department (Y.A.), Getafe University Hospital, Madrid; Neurology Department (L.B.), Arnau de Vilanova Hospital, Lleida; Neurology Department (C.Í.), Lozano Blesa Clinic University Hospital, Zaragoza; Neurology Department (I.G.-S.), Alvaro Cunqueiro Hospital, Vigo; Neurology Department (L.A.R.d.A.), Fuenlabrada University Hospital, Madrid, Spain
| | - Jens Kuhle
- From the Immunology Department (J.I.F.-V., P.E.W.-D., E.R.-M., E.R., N.V., L.M.V.), Ramon y Cajal University Hospital, Madrid, Spain; Neurologic Clinic and Policlinic (J.K., A.M.), Departments of Medicine, Biomedicine, and Clinical Research, University Hospital Basel, University of Basel, Switzerland; Neurology Department (E.M., S.S.d.l.M., J.M., L.C.-F.), Ramon y Cajal University Hospital, Madrid; Neurology Department (V.M.-L., P.S.), La Princesa University Hospital, Madrid; Multiple Sclerosis and Clinical Neuroimmunology Unit (J.M.-L., E.C.-G.), Virgen de la Arrixaca University Hospital, Murcia; Multiple Sclerosis Unit (G.I.), Vithas Nisa Sevilla Hospital; Neurology Department (F.G.-G.), Valencia Clinic University Hospital; Center of Neuroimmunology (A.S., Y.B.), Neurology Department, Clínic of Barcelona Hospital, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), and Institut de Neurociències, Universitat de Barcelona; Neurology Department (Y.A.), Getafe University Hospital, Madrid; Neurology Department (L.B.), Arnau de Vilanova Hospital, Lleida; Neurology Department (C.Í.), Lozano Blesa Clinic University Hospital, Zaragoza; Neurology Department (I.G.-S.), Alvaro Cunqueiro Hospital, Vigo; Neurology Department (L.A.R.d.A.), Fuenlabrada University Hospital, Madrid, Spain
| | - Enric Monreal
- From the Immunology Department (J.I.F.-V., P.E.W.-D., E.R.-M., E.R., N.V., L.M.V.), Ramon y Cajal University Hospital, Madrid, Spain; Neurologic Clinic and Policlinic (J.K., A.M.), Departments of Medicine, Biomedicine, and Clinical Research, University Hospital Basel, University of Basel, Switzerland; Neurology Department (E.M., S.S.d.l.M., J.M., L.C.-F.), Ramon y Cajal University Hospital, Madrid; Neurology Department (V.M.-L., P.S.), La Princesa University Hospital, Madrid; Multiple Sclerosis and Clinical Neuroimmunology Unit (J.M.-L., E.C.-G.), Virgen de la Arrixaca University Hospital, Murcia; Multiple Sclerosis Unit (G.I.), Vithas Nisa Sevilla Hospital; Neurology Department (F.G.-G.), Valencia Clinic University Hospital; Center of Neuroimmunology (A.S., Y.B.), Neurology Department, Clínic of Barcelona Hospital, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), and Institut de Neurociències, Universitat de Barcelona; Neurology Department (Y.A.), Getafe University Hospital, Madrid; Neurology Department (L.B.), Arnau de Vilanova Hospital, Lleida; Neurology Department (C.Í.), Lozano Blesa Clinic University Hospital, Zaragoza; Neurology Department (I.G.-S.), Alvaro Cunqueiro Hospital, Vigo; Neurology Department (L.A.R.d.A.), Fuenlabrada University Hospital, Madrid, Spain
| | - Virginia Meca-Lallana
- From the Immunology Department (J.I.F.-V., P.E.W.-D., E.R.-M., E.R., N.V., L.M.V.), Ramon y Cajal University Hospital, Madrid, Spain; Neurologic Clinic and Policlinic (J.K., A.M.), Departments of Medicine, Biomedicine, and Clinical Research, University Hospital Basel, University of Basel, Switzerland; Neurology Department (E.M., S.S.d.l.M., J.M., L.C.-F.), Ramon y Cajal University Hospital, Madrid; Neurology Department (V.M.-L., P.S.), La Princesa University Hospital, Madrid; Multiple Sclerosis and Clinical Neuroimmunology Unit (J.M.-L., E.C.-G.), Virgen de la Arrixaca University Hospital, Murcia; Multiple Sclerosis Unit (G.I.), Vithas Nisa Sevilla Hospital; Neurology Department (F.G.-G.), Valencia Clinic University Hospital; Center of Neuroimmunology (A.S., Y.B.), Neurology Department, Clínic of Barcelona Hospital, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), and Institut de Neurociències, Universitat de Barcelona; Neurology Department (Y.A.), Getafe University Hospital, Madrid; Neurology Department (L.B.), Arnau de Vilanova Hospital, Lleida; Neurology Department (C.Í.), Lozano Blesa Clinic University Hospital, Zaragoza; Neurology Department (I.G.-S.), Alvaro Cunqueiro Hospital, Vigo; Neurology Department (L.A.R.d.A.), Fuenlabrada University Hospital, Madrid, Spain
| | - José Meca-Lallana
- From the Immunology Department (J.I.F.-V., P.E.W.-D., E.R.-M., E.R., N.V., L.M.V.), Ramon y Cajal University Hospital, Madrid, Spain; Neurologic Clinic and Policlinic (J.K., A.M.), Departments of Medicine, Biomedicine, and Clinical Research, University Hospital Basel, University of Basel, Switzerland; Neurology Department (E.M., S.S.d.l.M., J.M., L.C.-F.), Ramon y Cajal University Hospital, Madrid; Neurology Department (V.M.-L., P.S.), La Princesa University Hospital, Madrid; Multiple Sclerosis and Clinical Neuroimmunology Unit (J.M.-L., E.C.-G.), Virgen de la Arrixaca University Hospital, Murcia; Multiple Sclerosis Unit (G.I.), Vithas Nisa Sevilla Hospital; Neurology Department (F.G.-G.), Valencia Clinic University Hospital; Center of Neuroimmunology (A.S., Y.B.), Neurology Department, Clínic of Barcelona Hospital, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), and Institut de Neurociències, Universitat de Barcelona; Neurology Department (Y.A.), Getafe University Hospital, Madrid; Neurology Department (L.B.), Arnau de Vilanova Hospital, Lleida; Neurology Department (C.Í.), Lozano Blesa Clinic University Hospital, Zaragoza; Neurology Department (I.G.-S.), Alvaro Cunqueiro Hospital, Vigo; Neurology Department (L.A.R.d.A.), Fuenlabrada University Hospital, Madrid, Spain
| | - Guillermo Izquierdo
- From the Immunology Department (J.I.F.-V., P.E.W.-D., E.R.-M., E.R., N.V., L.M.V.), Ramon y Cajal University Hospital, Madrid, Spain; Neurologic Clinic and Policlinic (J.K., A.M.), Departments of Medicine, Biomedicine, and Clinical Research, University Hospital Basel, University of Basel, Switzerland; Neurology Department (E.M., S.S.d.l.M., J.M., L.C.-F.), Ramon y Cajal University Hospital, Madrid; Neurology Department (V.M.-L., P.S.), La Princesa University Hospital, Madrid; Multiple Sclerosis and Clinical Neuroimmunology Unit (J.M.-L., E.C.-G.), Virgen de la Arrixaca University Hospital, Murcia; Multiple Sclerosis Unit (G.I.), Vithas Nisa Sevilla Hospital; Neurology Department (F.G.-G.), Valencia Clinic University Hospital; Center of Neuroimmunology (A.S., Y.B.), Neurology Department, Clínic of Barcelona Hospital, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), and Institut de Neurociències, Universitat de Barcelona; Neurology Department (Y.A.), Getafe University Hospital, Madrid; Neurology Department (L.B.), Arnau de Vilanova Hospital, Lleida; Neurology Department (C.Í.), Lozano Blesa Clinic University Hospital, Zaragoza; Neurology Department (I.G.-S.), Alvaro Cunqueiro Hospital, Vigo; Neurology Department (L.A.R.d.A.), Fuenlabrada University Hospital, Madrid, Spain
| | - Francisco Gascón-Giménez
- From the Immunology Department (J.I.F.-V., P.E.W.-D., E.R.-M., E.R., N.V., L.M.V.), Ramon y Cajal University Hospital, Madrid, Spain; Neurologic Clinic and Policlinic (J.K., A.M.), Departments of Medicine, Biomedicine, and Clinical Research, University Hospital Basel, University of Basel, Switzerland; Neurology Department (E.M., S.S.d.l.M., J.M., L.C.-F.), Ramon y Cajal University Hospital, Madrid; Neurology Department (V.M.-L., P.S.), La Princesa University Hospital, Madrid; Multiple Sclerosis and Clinical Neuroimmunology Unit (J.M.-L., E.C.-G.), Virgen de la Arrixaca University Hospital, Murcia; Multiple Sclerosis Unit (G.I.), Vithas Nisa Sevilla Hospital; Neurology Department (F.G.-G.), Valencia Clinic University Hospital; Center of Neuroimmunology (A.S., Y.B.), Neurology Department, Clínic of Barcelona Hospital, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), and Institut de Neurociències, Universitat de Barcelona; Neurology Department (Y.A.), Getafe University Hospital, Madrid; Neurology Department (L.B.), Arnau de Vilanova Hospital, Lleida; Neurology Department (C.Í.), Lozano Blesa Clinic University Hospital, Zaragoza; Neurology Department (I.G.-S.), Alvaro Cunqueiro Hospital, Vigo; Neurology Department (L.A.R.d.A.), Fuenlabrada University Hospital, Madrid, Spain
| | - Susana Sainz de la Maza
- From the Immunology Department (J.I.F.-V., P.E.W.-D., E.R.-M., E.R., N.V., L.M.V.), Ramon y Cajal University Hospital, Madrid, Spain; Neurologic Clinic and Policlinic (J.K., A.M.), Departments of Medicine, Biomedicine, and Clinical Research, University Hospital Basel, University of Basel, Switzerland; Neurology Department (E.M., S.S.d.l.M., J.M., L.C.-F.), Ramon y Cajal University Hospital, Madrid; Neurology Department (V.M.-L., P.S.), La Princesa University Hospital, Madrid; Multiple Sclerosis and Clinical Neuroimmunology Unit (J.M.-L., E.C.-G.), Virgen de la Arrixaca University Hospital, Murcia; Multiple Sclerosis Unit (G.I.), Vithas Nisa Sevilla Hospital; Neurology Department (F.G.-G.), Valencia Clinic University Hospital; Center of Neuroimmunology (A.S., Y.B.), Neurology Department, Clínic of Barcelona Hospital, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), and Institut de Neurociències, Universitat de Barcelona; Neurology Department (Y.A.), Getafe University Hospital, Madrid; Neurology Department (L.B.), Arnau de Vilanova Hospital, Lleida; Neurology Department (C.Í.), Lozano Blesa Clinic University Hospital, Zaragoza; Neurology Department (I.G.-S.), Alvaro Cunqueiro Hospital, Vigo; Neurology Department (L.A.R.d.A.), Fuenlabrada University Hospital, Madrid, Spain
| | - Paulette E. Walo-Delgado
- From the Immunology Department (J.I.F.-V., P.E.W.-D., E.R.-M., E.R., N.V., L.M.V.), Ramon y Cajal University Hospital, Madrid, Spain; Neurologic Clinic and Policlinic (J.K., A.M.), Departments of Medicine, Biomedicine, and Clinical Research, University Hospital Basel, University of Basel, Switzerland; Neurology Department (E.M., S.S.d.l.M., J.M., L.C.-F.), Ramon y Cajal University Hospital, Madrid; Neurology Department (V.M.-L., P.S.), La Princesa University Hospital, Madrid; Multiple Sclerosis and Clinical Neuroimmunology Unit (J.M.-L., E.C.-G.), Virgen de la Arrixaca University Hospital, Murcia; Multiple Sclerosis Unit (G.I.), Vithas Nisa Sevilla Hospital; Neurology Department (F.G.-G.), Valencia Clinic University Hospital; Center of Neuroimmunology (A.S., Y.B.), Neurology Department, Clínic of Barcelona Hospital, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), and Institut de Neurociències, Universitat de Barcelona; Neurology Department (Y.A.), Getafe University Hospital, Madrid; Neurology Department (L.B.), Arnau de Vilanova Hospital, Lleida; Neurology Department (C.Í.), Lozano Blesa Clinic University Hospital, Zaragoza; Neurology Department (I.G.-S.), Alvaro Cunqueiro Hospital, Vigo; Neurology Department (L.A.R.d.A.), Fuenlabrada University Hospital, Madrid, Spain
| | - Aleksandra Maceski
- From the Immunology Department (J.I.F.-V., P.E.W.-D., E.R.-M., E.R., N.V., L.M.V.), Ramon y Cajal University Hospital, Madrid, Spain; Neurologic Clinic and Policlinic (J.K., A.M.), Departments of Medicine, Biomedicine, and Clinical Research, University Hospital Basel, University of Basel, Switzerland; Neurology Department (E.M., S.S.d.l.M., J.M., L.C.-F.), Ramon y Cajal University Hospital, Madrid; Neurology Department (V.M.-L., P.S.), La Princesa University Hospital, Madrid; Multiple Sclerosis and Clinical Neuroimmunology Unit (J.M.-L., E.C.-G.), Virgen de la Arrixaca University Hospital, Murcia; Multiple Sclerosis Unit (G.I.), Vithas Nisa Sevilla Hospital; Neurology Department (F.G.-G.), Valencia Clinic University Hospital; Center of Neuroimmunology (A.S., Y.B.), Neurology Department, Clínic of Barcelona Hospital, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), and Institut de Neurociències, Universitat de Barcelona; Neurology Department (Y.A.), Getafe University Hospital, Madrid; Neurology Department (L.B.), Arnau de Vilanova Hospital, Lleida; Neurology Department (C.Í.), Lozano Blesa Clinic University Hospital, Zaragoza; Neurology Department (I.G.-S.), Alvaro Cunqueiro Hospital, Vigo; Neurology Department (L.A.R.d.A.), Fuenlabrada University Hospital, Madrid, Spain
| | - Eulalia Rodríguez-Martín
- From the Immunology Department (J.I.F.-V., P.E.W.-D., E.R.-M., E.R., N.V., L.M.V.), Ramon y Cajal University Hospital, Madrid, Spain; Neurologic Clinic and Policlinic (J.K., A.M.), Departments of Medicine, Biomedicine, and Clinical Research, University Hospital Basel, University of Basel, Switzerland; Neurology Department (E.M., S.S.d.l.M., J.M., L.C.-F.), Ramon y Cajal University Hospital, Madrid; Neurology Department (V.M.-L., P.S.), La Princesa University Hospital, Madrid; Multiple Sclerosis and Clinical Neuroimmunology Unit (J.M.-L., E.C.-G.), Virgen de la Arrixaca University Hospital, Murcia; Multiple Sclerosis Unit (G.I.), Vithas Nisa Sevilla Hospital; Neurology Department (F.G.-G.), Valencia Clinic University Hospital; Center of Neuroimmunology (A.S., Y.B.), Neurology Department, Clínic of Barcelona Hospital, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), and Institut de Neurociències, Universitat de Barcelona; Neurology Department (Y.A.), Getafe University Hospital, Madrid; Neurology Department (L.B.), Arnau de Vilanova Hospital, Lleida; Neurology Department (C.Í.), Lozano Blesa Clinic University Hospital, Zaragoza; Neurology Department (I.G.-S.), Alvaro Cunqueiro Hospital, Vigo; Neurology Department (L.A.R.d.A.), Fuenlabrada University Hospital, Madrid, Spain
| | - Ernesto Roldán
- From the Immunology Department (J.I.F.-V., P.E.W.-D., E.R.-M., E.R., N.V., L.M.V.), Ramon y Cajal University Hospital, Madrid, Spain; Neurologic Clinic and Policlinic (J.K., A.M.), Departments of Medicine, Biomedicine, and Clinical Research, University Hospital Basel, University of Basel, Switzerland; Neurology Department (E.M., S.S.d.l.M., J.M., L.C.-F.), Ramon y Cajal University Hospital, Madrid; Neurology Department (V.M.-L., P.S.), La Princesa University Hospital, Madrid; Multiple Sclerosis and Clinical Neuroimmunology Unit (J.M.-L., E.C.-G.), Virgen de la Arrixaca University Hospital, Murcia; Multiple Sclerosis Unit (G.I.), Vithas Nisa Sevilla Hospital; Neurology Department (F.G.-G.), Valencia Clinic University Hospital; Center of Neuroimmunology (A.S., Y.B.), Neurology Department, Clínic of Barcelona Hospital, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), and Institut de Neurociències, Universitat de Barcelona; Neurology Department (Y.A.), Getafe University Hospital, Madrid; Neurology Department (L.B.), Arnau de Vilanova Hospital, Lleida; Neurology Department (C.Í.), Lozano Blesa Clinic University Hospital, Zaragoza; Neurology Department (I.G.-S.), Alvaro Cunqueiro Hospital, Vigo; Neurology Department (L.A.R.d.A.), Fuenlabrada University Hospital, Madrid, Spain
| | - Noelia Villarrubia
- From the Immunology Department (J.I.F.-V., P.E.W.-D., E.R.-M., E.R., N.V., L.M.V.), Ramon y Cajal University Hospital, Madrid, Spain; Neurologic Clinic and Policlinic (J.K., A.M.), Departments of Medicine, Biomedicine, and Clinical Research, University Hospital Basel, University of Basel, Switzerland; Neurology Department (E.M., S.S.d.l.M., J.M., L.C.-F.), Ramon y Cajal University Hospital, Madrid; Neurology Department (V.M.-L., P.S.), La Princesa University Hospital, Madrid; Multiple Sclerosis and Clinical Neuroimmunology Unit (J.M.-L., E.C.-G.), Virgen de la Arrixaca University Hospital, Murcia; Multiple Sclerosis Unit (G.I.), Vithas Nisa Sevilla Hospital; Neurology Department (F.G.-G.), Valencia Clinic University Hospital; Center of Neuroimmunology (A.S., Y.B.), Neurology Department, Clínic of Barcelona Hospital, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), and Institut de Neurociències, Universitat de Barcelona; Neurology Department (Y.A.), Getafe University Hospital, Madrid; Neurology Department (L.B.), Arnau de Vilanova Hospital, Lleida; Neurology Department (C.Í.), Lozano Blesa Clinic University Hospital, Zaragoza; Neurology Department (I.G.-S.), Alvaro Cunqueiro Hospital, Vigo; Neurology Department (L.A.R.d.A.), Fuenlabrada University Hospital, Madrid, Spain
| | - Albert Saiz
- From the Immunology Department (J.I.F.-V., P.E.W.-D., E.R.-M., E.R., N.V., L.M.V.), Ramon y Cajal University Hospital, Madrid, Spain; Neurologic Clinic and Policlinic (J.K., A.M.), Departments of Medicine, Biomedicine, and Clinical Research, University Hospital Basel, University of Basel, Switzerland; Neurology Department (E.M., S.S.d.l.M., J.M., L.C.-F.), Ramon y Cajal University Hospital, Madrid; Neurology Department (V.M.-L., P.S.), La Princesa University Hospital, Madrid; Multiple Sclerosis and Clinical Neuroimmunology Unit (J.M.-L., E.C.-G.), Virgen de la Arrixaca University Hospital, Murcia; Multiple Sclerosis Unit (G.I.), Vithas Nisa Sevilla Hospital; Neurology Department (F.G.-G.), Valencia Clinic University Hospital; Center of Neuroimmunology (A.S., Y.B.), Neurology Department, Clínic of Barcelona Hospital, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), and Institut de Neurociències, Universitat de Barcelona; Neurology Department (Y.A.), Getafe University Hospital, Madrid; Neurology Department (L.B.), Arnau de Vilanova Hospital, Lleida; Neurology Department (C.Í.), Lozano Blesa Clinic University Hospital, Zaragoza; Neurology Department (I.G.-S.), Alvaro Cunqueiro Hospital, Vigo; Neurology Department (L.A.R.d.A.), Fuenlabrada University Hospital, Madrid, Spain
| | - Yolanda Blanco
- From the Immunology Department (J.I.F.-V., P.E.W.-D., E.R.-M., E.R., N.V., L.M.V.), Ramon y Cajal University Hospital, Madrid, Spain; Neurologic Clinic and Policlinic (J.K., A.M.), Departments of Medicine, Biomedicine, and Clinical Research, University Hospital Basel, University of Basel, Switzerland; Neurology Department (E.M., S.S.d.l.M., J.M., L.C.-F.), Ramon y Cajal University Hospital, Madrid; Neurology Department (V.M.-L., P.S.), La Princesa University Hospital, Madrid; Multiple Sclerosis and Clinical Neuroimmunology Unit (J.M.-L., E.C.-G.), Virgen de la Arrixaca University Hospital, Murcia; Multiple Sclerosis Unit (G.I.), Vithas Nisa Sevilla Hospital; Neurology Department (F.G.-G.), Valencia Clinic University Hospital; Center of Neuroimmunology (A.S., Y.B.), Neurology Department, Clínic of Barcelona Hospital, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), and Institut de Neurociències, Universitat de Barcelona; Neurology Department (Y.A.), Getafe University Hospital, Madrid; Neurology Department (L.B.), Arnau de Vilanova Hospital, Lleida; Neurology Department (C.Í.), Lozano Blesa Clinic University Hospital, Zaragoza; Neurology Department (I.G.-S.), Alvaro Cunqueiro Hospital, Vigo; Neurology Department (L.A.R.d.A.), Fuenlabrada University Hospital, Madrid, Spain
| | - Pedro Sánchez
- From the Immunology Department (J.I.F.-V., P.E.W.-D., E.R.-M., E.R., N.V., L.M.V.), Ramon y Cajal University Hospital, Madrid, Spain; Neurologic Clinic and Policlinic (J.K., A.M.), Departments of Medicine, Biomedicine, and Clinical Research, University Hospital Basel, University of Basel, Switzerland; Neurology Department (E.M., S.S.d.l.M., J.M., L.C.-F.), Ramon y Cajal University Hospital, Madrid; Neurology Department (V.M.-L., P.S.), La Princesa University Hospital, Madrid; Multiple Sclerosis and Clinical Neuroimmunology Unit (J.M.-L., E.C.-G.), Virgen de la Arrixaca University Hospital, Murcia; Multiple Sclerosis Unit (G.I.), Vithas Nisa Sevilla Hospital; Neurology Department (F.G.-G.), Valencia Clinic University Hospital; Center of Neuroimmunology (A.S., Y.B.), Neurology Department, Clínic of Barcelona Hospital, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), and Institut de Neurociències, Universitat de Barcelona; Neurology Department (Y.A.), Getafe University Hospital, Madrid; Neurology Department (L.B.), Arnau de Vilanova Hospital, Lleida; Neurology Department (C.Í.), Lozano Blesa Clinic University Hospital, Zaragoza; Neurology Department (I.G.-S.), Alvaro Cunqueiro Hospital, Vigo; Neurology Department (L.A.R.d.A.), Fuenlabrada University Hospital, Madrid, Spain
| | - Ester Carreón-Guarnizo
- From the Immunology Department (J.I.F.-V., P.E.W.-D., E.R.-M., E.R., N.V., L.M.V.), Ramon y Cajal University Hospital, Madrid, Spain; Neurologic Clinic and Policlinic (J.K., A.M.), Departments of Medicine, Biomedicine, and Clinical Research, University Hospital Basel, University of Basel, Switzerland; Neurology Department (E.M., S.S.d.l.M., J.M., L.C.-F.), Ramon y Cajal University Hospital, Madrid; Neurology Department (V.M.-L., P.S.), La Princesa University Hospital, Madrid; Multiple Sclerosis and Clinical Neuroimmunology Unit (J.M.-L., E.C.-G.), Virgen de la Arrixaca University Hospital, Murcia; Multiple Sclerosis Unit (G.I.), Vithas Nisa Sevilla Hospital; Neurology Department (F.G.-G.), Valencia Clinic University Hospital; Center of Neuroimmunology (A.S., Y.B.), Neurology Department, Clínic of Barcelona Hospital, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), and Institut de Neurociències, Universitat de Barcelona; Neurology Department (Y.A.), Getafe University Hospital, Madrid; Neurology Department (L.B.), Arnau de Vilanova Hospital, Lleida; Neurology Department (C.Í.), Lozano Blesa Clinic University Hospital, Zaragoza; Neurology Department (I.G.-S.), Alvaro Cunqueiro Hospital, Vigo; Neurology Department (L.A.R.d.A.), Fuenlabrada University Hospital, Madrid, Spain
| | - Yolanda Aladro
- From the Immunology Department (J.I.F.-V., P.E.W.-D., E.R.-M., E.R., N.V., L.M.V.), Ramon y Cajal University Hospital, Madrid, Spain; Neurologic Clinic and Policlinic (J.K., A.M.), Departments of Medicine, Biomedicine, and Clinical Research, University Hospital Basel, University of Basel, Switzerland; Neurology Department (E.M., S.S.d.l.M., J.M., L.C.-F.), Ramon y Cajal University Hospital, Madrid; Neurology Department (V.M.-L., P.S.), La Princesa University Hospital, Madrid; Multiple Sclerosis and Clinical Neuroimmunology Unit (J.M.-L., E.C.-G.), Virgen de la Arrixaca University Hospital, Murcia; Multiple Sclerosis Unit (G.I.), Vithas Nisa Sevilla Hospital; Neurology Department (F.G.-G.), Valencia Clinic University Hospital; Center of Neuroimmunology (A.S., Y.B.), Neurology Department, Clínic of Barcelona Hospital, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), and Institut de Neurociències, Universitat de Barcelona; Neurology Department (Y.A.), Getafe University Hospital, Madrid; Neurology Department (L.B.), Arnau de Vilanova Hospital, Lleida; Neurology Department (C.Í.), Lozano Blesa Clinic University Hospital, Zaragoza; Neurology Department (I.G.-S.), Alvaro Cunqueiro Hospital, Vigo; Neurology Department (L.A.R.d.A.), Fuenlabrada University Hospital, Madrid, Spain
| | - Luis Brieva
- From the Immunology Department (J.I.F.-V., P.E.W.-D., E.R.-M., E.R., N.V., L.M.V.), Ramon y Cajal University Hospital, Madrid, Spain; Neurologic Clinic and Policlinic (J.K., A.M.), Departments of Medicine, Biomedicine, and Clinical Research, University Hospital Basel, University of Basel, Switzerland; Neurology Department (E.M., S.S.d.l.M., J.M., L.C.-F.), Ramon y Cajal University Hospital, Madrid; Neurology Department (V.M.-L., P.S.), La Princesa University Hospital, Madrid; Multiple Sclerosis and Clinical Neuroimmunology Unit (J.M.-L., E.C.-G.), Virgen de la Arrixaca University Hospital, Murcia; Multiple Sclerosis Unit (G.I.), Vithas Nisa Sevilla Hospital; Neurology Department (F.G.-G.), Valencia Clinic University Hospital; Center of Neuroimmunology (A.S., Y.B.), Neurology Department, Clínic of Barcelona Hospital, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), and Institut de Neurociències, Universitat de Barcelona; Neurology Department (Y.A.), Getafe University Hospital, Madrid; Neurology Department (L.B.), Arnau de Vilanova Hospital, Lleida; Neurology Department (C.Í.), Lozano Blesa Clinic University Hospital, Zaragoza; Neurology Department (I.G.-S.), Alvaro Cunqueiro Hospital, Vigo; Neurology Department (L.A.R.d.A.), Fuenlabrada University Hospital, Madrid, Spain
| | - Cristina Íñiguez
- From the Immunology Department (J.I.F.-V., P.E.W.-D., E.R.-M., E.R., N.V., L.M.V.), Ramon y Cajal University Hospital, Madrid, Spain; Neurologic Clinic and Policlinic (J.K., A.M.), Departments of Medicine, Biomedicine, and Clinical Research, University Hospital Basel, University of Basel, Switzerland; Neurology Department (E.M., S.S.d.l.M., J.M., L.C.-F.), Ramon y Cajal University Hospital, Madrid; Neurology Department (V.M.-L., P.S.), La Princesa University Hospital, Madrid; Multiple Sclerosis and Clinical Neuroimmunology Unit (J.M.-L., E.C.-G.), Virgen de la Arrixaca University Hospital, Murcia; Multiple Sclerosis Unit (G.I.), Vithas Nisa Sevilla Hospital; Neurology Department (F.G.-G.), Valencia Clinic University Hospital; Center of Neuroimmunology (A.S., Y.B.), Neurology Department, Clínic of Barcelona Hospital, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), and Institut de Neurociències, Universitat de Barcelona; Neurology Department (Y.A.), Getafe University Hospital, Madrid; Neurology Department (L.B.), Arnau de Vilanova Hospital, Lleida; Neurology Department (C.Í.), Lozano Blesa Clinic University Hospital, Zaragoza; Neurology Department (I.G.-S.), Alvaro Cunqueiro Hospital, Vigo; Neurology Department (L.A.R.d.A.), Fuenlabrada University Hospital, Madrid, Spain
| | - Inés González-Suárez
- From the Immunology Department (J.I.F.-V., P.E.W.-D., E.R.-M., E.R., N.V., L.M.V.), Ramon y Cajal University Hospital, Madrid, Spain; Neurologic Clinic and Policlinic (J.K., A.M.), Departments of Medicine, Biomedicine, and Clinical Research, University Hospital Basel, University of Basel, Switzerland; Neurology Department (E.M., S.S.d.l.M., J.M., L.C.-F.), Ramon y Cajal University Hospital, Madrid; Neurology Department (V.M.-L., P.S.), La Princesa University Hospital, Madrid; Multiple Sclerosis and Clinical Neuroimmunology Unit (J.M.-L., E.C.-G.), Virgen de la Arrixaca University Hospital, Murcia; Multiple Sclerosis Unit (G.I.), Vithas Nisa Sevilla Hospital; Neurology Department (F.G.-G.), Valencia Clinic University Hospital; Center of Neuroimmunology (A.S., Y.B.), Neurology Department, Clínic of Barcelona Hospital, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), and Institut de Neurociències, Universitat de Barcelona; Neurology Department (Y.A.), Getafe University Hospital, Madrid; Neurology Department (L.B.), Arnau de Vilanova Hospital, Lleida; Neurology Department (C.Í.), Lozano Blesa Clinic University Hospital, Zaragoza; Neurology Department (I.G.-S.), Alvaro Cunqueiro Hospital, Vigo; Neurology Department (L.A.R.d.A.), Fuenlabrada University Hospital, Madrid, Spain
| | - Luis A. Rodríguez de Antonio
- From the Immunology Department (J.I.F.-V., P.E.W.-D., E.R.-M., E.R., N.V., L.M.V.), Ramon y Cajal University Hospital, Madrid, Spain; Neurologic Clinic and Policlinic (J.K., A.M.), Departments of Medicine, Biomedicine, and Clinical Research, University Hospital Basel, University of Basel, Switzerland; Neurology Department (E.M., S.S.d.l.M., J.M., L.C.-F.), Ramon y Cajal University Hospital, Madrid; Neurology Department (V.M.-L., P.S.), La Princesa University Hospital, Madrid; Multiple Sclerosis and Clinical Neuroimmunology Unit (J.M.-L., E.C.-G.), Virgen de la Arrixaca University Hospital, Murcia; Multiple Sclerosis Unit (G.I.), Vithas Nisa Sevilla Hospital; Neurology Department (F.G.-G.), Valencia Clinic University Hospital; Center of Neuroimmunology (A.S., Y.B.), Neurology Department, Clínic of Barcelona Hospital, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), and Institut de Neurociències, Universitat de Barcelona; Neurology Department (Y.A.), Getafe University Hospital, Madrid; Neurology Department (L.B.), Arnau de Vilanova Hospital, Lleida; Neurology Department (C.Í.), Lozano Blesa Clinic University Hospital, Zaragoza; Neurology Department (I.G.-S.), Alvaro Cunqueiro Hospital, Vigo; Neurology Department (L.A.R.d.A.), Fuenlabrada University Hospital, Madrid, Spain
| | - Jaime Masjuan
- From the Immunology Department (J.I.F.-V., P.E.W.-D., E.R.-M., E.R., N.V., L.M.V.), Ramon y Cajal University Hospital, Madrid, Spain; Neurologic Clinic and Policlinic (J.K., A.M.), Departments of Medicine, Biomedicine, and Clinical Research, University Hospital Basel, University of Basel, Switzerland; Neurology Department (E.M., S.S.d.l.M., J.M., L.C.-F.), Ramon y Cajal University Hospital, Madrid; Neurology Department (V.M.-L., P.S.), La Princesa University Hospital, Madrid; Multiple Sclerosis and Clinical Neuroimmunology Unit (J.M.-L., E.C.-G.), Virgen de la Arrixaca University Hospital, Murcia; Multiple Sclerosis Unit (G.I.), Vithas Nisa Sevilla Hospital; Neurology Department (F.G.-G.), Valencia Clinic University Hospital; Center of Neuroimmunology (A.S., Y.B.), Neurology Department, Clínic of Barcelona Hospital, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), and Institut de Neurociències, Universitat de Barcelona; Neurology Department (Y.A.), Getafe University Hospital, Madrid; Neurology Department (L.B.), Arnau de Vilanova Hospital, Lleida; Neurology Department (C.Í.), Lozano Blesa Clinic University Hospital, Zaragoza; Neurology Department (I.G.-S.), Alvaro Cunqueiro Hospital, Vigo; Neurology Department (L.A.R.d.A.), Fuenlabrada University Hospital, Madrid, Spain
| | - Lucienne Costa-Frossard
- From the Immunology Department (J.I.F.-V., P.E.W.-D., E.R.-M., E.R., N.V., L.M.V.), Ramon y Cajal University Hospital, Madrid, Spain; Neurologic Clinic and Policlinic (J.K., A.M.), Departments of Medicine, Biomedicine, and Clinical Research, University Hospital Basel, University of Basel, Switzerland; Neurology Department (E.M., S.S.d.l.M., J.M., L.C.-F.), Ramon y Cajal University Hospital, Madrid; Neurology Department (V.M.-L., P.S.), La Princesa University Hospital, Madrid; Multiple Sclerosis and Clinical Neuroimmunology Unit (J.M.-L., E.C.-G.), Virgen de la Arrixaca University Hospital, Murcia; Multiple Sclerosis Unit (G.I.), Vithas Nisa Sevilla Hospital; Neurology Department (F.G.-G.), Valencia Clinic University Hospital; Center of Neuroimmunology (A.S., Y.B.), Neurology Department, Clínic of Barcelona Hospital, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), and Institut de Neurociències, Universitat de Barcelona; Neurology Department (Y.A.), Getafe University Hospital, Madrid; Neurology Department (L.B.), Arnau de Vilanova Hospital, Lleida; Neurology Department (C.Í.), Lozano Blesa Clinic University Hospital, Zaragoza; Neurology Department (I.G.-S.), Alvaro Cunqueiro Hospital, Vigo; Neurology Department (L.A.R.d.A.), Fuenlabrada University Hospital, Madrid, Spain
| | - Luisa M. Villar
- From the Immunology Department (J.I.F.-V., P.E.W.-D., E.R.-M., E.R., N.V., L.M.V.), Ramon y Cajal University Hospital, Madrid, Spain; Neurologic Clinic and Policlinic (J.K., A.M.), Departments of Medicine, Biomedicine, and Clinical Research, University Hospital Basel, University of Basel, Switzerland; Neurology Department (E.M., S.S.d.l.M., J.M., L.C.-F.), Ramon y Cajal University Hospital, Madrid; Neurology Department (V.M.-L., P.S.), La Princesa University Hospital, Madrid; Multiple Sclerosis and Clinical Neuroimmunology Unit (J.M.-L., E.C.-G.), Virgen de la Arrixaca University Hospital, Murcia; Multiple Sclerosis Unit (G.I.), Vithas Nisa Sevilla Hospital; Neurology Department (F.G.-G.), Valencia Clinic University Hospital; Center of Neuroimmunology (A.S., Y.B.), Neurology Department, Clínic of Barcelona Hospital, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), and Institut de Neurociències, Universitat de Barcelona; Neurology Department (Y.A.), Getafe University Hospital, Madrid; Neurology Department (L.B.), Arnau de Vilanova Hospital, Lleida; Neurology Department (C.Í.), Lozano Blesa Clinic University Hospital, Zaragoza; Neurology Department (I.G.-S.), Alvaro Cunqueiro Hospital, Vigo; Neurology Department (L.A.R.d.A.), Fuenlabrada University Hospital, Madrid, Spain
| |
Collapse
|
32
|
Clinical Perspectives on the Molecular and Pharmacological Attributes of Anti-CD20 Therapies for Multiple Sclerosis. CNS Drugs 2021; 35:985-997. [PMID: 34370283 PMCID: PMC8351586 DOI: 10.1007/s40263-021-00843-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/05/2021] [Indexed: 11/26/2022]
Abstract
Anti-CD20 therapies have demonstrated considerable efficacy in the treatment of relapsing multiple sclerosis, constituting a high-efficacy treatment approach for reducing relapse risk and mitigating disability progression. These therapies have been shown to strongly deplete circulating B cells and small subsets of CD3+ CD4 and CD8 T cells that express low levels of CD20. While the clinical profiles of the various anti-CD20 monoclonal antibodies used in treating multiple sclerosis are well-described in the literature, greater understanding of the implications of their distinct molecular and pharmacological attributes is needed. In this review, we focus on four anti-CD20 monoclonal antibodies-rituximab, ocrelizumab, ofatumumab, and ublituximab-that are currently used, approved, or in late-stage clinical development for the treatment of multiple sclerosis. We provide clinical perspectives on the potential implications of differences in molecular structures, target epitopes, dosing regimens, mechanisms and impact on B-cell depletion and reconstitution, immunogenicity, administration-related reactions, and infection risks.
Collapse
|
33
|
Baker D, Roberts CAK, Pryce G, Kang AS, Marta M, Reyes S, Schmierer K, Giovannoni G, Amor S. COVID-19 vaccine-readiness for anti-CD20-depleting therapy in autoimmune diseases. Clin Exp Immunol 2020; 202:149-161. [PMID: 32671831 PMCID: PMC7405500 DOI: 10.1111/cei.13495] [Citation(s) in RCA: 146] [Impact Index Per Article: 29.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Revised: 07/06/2020] [Accepted: 07/08/2020] [Indexed: 12/17/2022] Open
Abstract
Although most autoimmune diseases are considered to be CD4 T cell- or antibody-mediated, many respond to CD20-depleting antibodies that have limited influence on CD4 and plasma cells. This includes rituximab, oblinutuzumab and ofatumumab that are used in cancer, rheumatoid arthritis and off-label in a large number of other autoimmunities and ocrelizumab in multiple sclerosis. Recently, the COVID-19 pandemic created concerns about immunosuppression in autoimmunity, leading to cessation or a delay in immunotherapy treatments. However, based on the known and emerging biology of autoimmunity and COVID-19, it was hypothesised that while B cell depletion should not necessarily expose people to severe SARS-CoV-2-related issues, it may inhibit protective immunity following infection and vaccination. As such, drug-induced B cell subset inhibition, that controls at least some autoimmunities, would not influence innate and CD8 T cell responses, which are central to SARS-CoV-2 elimination, nor the hypercoagulation and innate inflammation causing severe morbidity. This is supported clinically, as the majority of SARS-CoV-2-infected, CD20-depleted people with autoimmunity have recovered. However, protective neutralizing antibody and vaccination responses are predicted to be blunted until naive B cells repopulate, based on B cell repopulation kinetics and vaccination responses, from published rituximab and unpublished ocrelizumab (NCT00676715, NCT02545868) trial data, shown here. This suggests that it may be possible to undertake dose interruption to maintain inflammatory disease control, while allowing effective vaccination against SARS-CoV-29, if and when an effective vaccine is available.
Collapse
Affiliation(s)
- D. Baker
- Blizard Institute, Barts and the London School of Medicine and DentistryQueen Mary University of LondonLondonUK
| | - C. A. K. Roberts
- Blizard Institute, Barts and the London School of Medicine and DentistryQueen Mary University of LondonLondonUK
| | - G. Pryce
- Blizard Institute, Barts and the London School of Medicine and DentistryQueen Mary University of LondonLondonUK
| | - A. S. Kang
- Blizard Institute, Barts and the London School of Medicine and DentistryQueen Mary University of LondonLondonUK
- Centre for Oral Immunobiology and Regenerative MedicineInstitute of Dentistry, Barts and The London School of Medicine and DentistryQueen Mary University of LondonLondonUK
| | - M. Marta
- Blizard Institute, Barts and the London School of Medicine and DentistryQueen Mary University of LondonLondonUK
- Clinical Board: Medicine (Neuroscience)The Royal London HospitalBarts Health NHS TrustLondonUK
| | - S. Reyes
- Blizard Institute, Barts and the London School of Medicine and DentistryQueen Mary University of LondonLondonUK
- Clinical Board: Medicine (Neuroscience)The Royal London HospitalBarts Health NHS TrustLondonUK
| | - K. Schmierer
- Blizard Institute, Barts and the London School of Medicine and DentistryQueen Mary University of LondonLondonUK
- Clinical Board: Medicine (Neuroscience)The Royal London HospitalBarts Health NHS TrustLondonUK
| | - G. Giovannoni
- Blizard Institute, Barts and the London School of Medicine and DentistryQueen Mary University of LondonLondonUK
- Clinical Board: Medicine (Neuroscience)The Royal London HospitalBarts Health NHS TrustLondonUK
| | - S. Amor
- Blizard Institute, Barts and the London School of Medicine and DentistryQueen Mary University of LondonLondonUK
- Pathology DepartmentAmsterdam UMCVUmc siteAmsterdamThe Netherlands
| |
Collapse
|
34
|
Yu X, Graner M, Kennedy PGE, Liu Y. The Role of Antibodies in the Pathogenesis of Multiple Sclerosis. Front Neurol 2020; 11:533388. [PMID: 33192968 PMCID: PMC7606501 DOI: 10.3389/fneur.2020.533388] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Accepted: 09/08/2020] [Indexed: 01/09/2023] Open
Abstract
The presence of persistent intrathecal oligoclonal immunoglobulin G (IgG) bands (OCBs) and lesional IgG deposition are seminal features of multiple sclerosis (MS) disease pathology. Despite extensive investigations, the role of antibodies, the products of mature CD19+ B cells, in disease development is still controversial and under significant debate. Recent success of B cell depletion therapies has revealed that CD20+ B cells contribute to MS pathogenesis via both antigen-presentation and T-cell-regulation. However, the limited efficacy of CD20+ B cell depletion therapies for the treatment of progressive MS indicates that additional mechanisms are involved. In this review, we present findings suggesting a potential pathological role for increased intrathecal IgGs, the relation of circulating antibodies to intrathecal IgGs, and the selective elevation of IgG1 and IgG3 subclasses in MS. We propose a working hypothesis that circulating B cells and antibodies contribute significantly to intrathecal IgGs, thereby exerting primary and pathogenic effects in MS development. Increased levels of IgG1 and IgG3 antibodies induce potent antibody-mediated cytotoxicity to central nervous system (CNS) cells and/or reduce the threshold required for antigen-driven antibody clustering leading to optimal activation of immune responses. Direct proof of the pathogenic roles of antibodies in MS may provide opportunities for novel blood biomarker identification as well as strategies for the development of effective therapeutic interventions.
Collapse
Affiliation(s)
- Xiaoli Yu
- Department of Neurosurgery, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Michael Graner
- Department of Neurosurgery, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Peter G E Kennedy
- Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, United Kingdom
| | - Yiting Liu
- Department of Neurology, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| |
Collapse
|
35
|
Yang Q, Wang G, Zhang F. Role of Peripheral Immune Cells-Mediated Inflammation on the Process of Neurodegenerative Diseases. Front Immunol 2020; 11:582825. [PMID: 33178212 PMCID: PMC7593572 DOI: 10.3389/fimmu.2020.582825] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Accepted: 09/08/2020] [Indexed: 12/12/2022] Open
Abstract
Neurodegenerative diseases are characterized by progressive loss of selectively vulnerable neuronal populations, which contrasts with selectively static loss of neurons due to toxic or metabolic disorders. The mechanisms underlying their progressive nature remain unknown. To date, a timely and well-controlled peripheral inflammatory reaction is verified to be essential for neurodegenerative diseases remission. The influence of peripheral inflammation on the central nervous system is closely related to immune cells activation in peripheral blood. The immune cells activation participated in the uncontrolled and prolonged inflammation that drives the chronic progression of neurodegenerative diseases. Thus, the dynamic modulation of this peripheral inflammatory reaction by interrupting the vicious cycle might become a disease-modifying therapeutic strategy for neurodegenerative diseases. This review focused on the role of peripheral immune cells on the pathological progression of neurodegenerative diseases.
Collapse
Affiliation(s)
- Qiuyu Yang
- Key Laboratory of Basic Pharmacology of Ministry of Education and Laboratory Animal Center and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, China
| | - Guoqing Wang
- Key Laboratory of Basic Pharmacology of Ministry of Education and Laboratory Animal Center and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, China
| | - Feng Zhang
- Key Laboratory of Basic Pharmacology of Ministry of Education and Laboratory Animal Center and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, China
| |
Collapse
|
36
|
Magliozzi R, Marastoni D, Calabrese M. The BAFF / APRIL system as therapeutic target in multiple sclerosis. Expert Opin Ther Targets 2020; 24:1135-1145. [PMID: 32900236 DOI: 10.1080/14728222.2020.1821647] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
INTRODUCTION The complex system of BAFF (B-cell-activating factor of the TNF family) and APRIL (A proliferation-inducing ligand) has been studied in animal models of autoimmune diseases such as those resembling human systemic lupus erythematosus and Sjogren's syndrome and multiple sclerosis (MS). Accumulating evidence suggests that BAFF and APRIL have a physiological role in B cell immunity regulation, however inappropriate production of these factors may represent a key event which disrupts immune tolerance which is associated with systemic autoimmune diseases. AREAS COVERED We provide an update on the latest studies of the BAFF/APRIL system in multiple sclerosis, as well as on related clinical trials. EXPERT OPINION Experimental and clinical evidence suggests that increased BAFF levels may interfere directly and indirectly with B cell immunity; this can lead to breakdown of immune tolerance, the production of autoantibodies and continuous local intracerebral inflammation and brain tissue destruction. A more comprehensive understanding of the cell/molecular mechanism immune reactions specifically regulated by BAFF/APRIL in MS would better elucidate the specific cell phenotype targeted by actual anti-BAFF/APRIL therapies; this may enable the identification of either specific biomarkers of MS subgroups that would benefit of anti-BAFF/APRIL treatments or new targets of MS-specific anti-BAFF/APRIL therapies.
Collapse
Affiliation(s)
- Roberta Magliozzi
- Neurology B, Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona , Verona, Italy
| | - Damiano Marastoni
- Neurology B, Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona , Verona, Italy
| | - Massimiliano Calabrese
- Neurology B, Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona , Verona, Italy
| |
Collapse
|
37
|
Affiliation(s)
- Jeffrey A Bluestone
- From the Sean N. Parker Autoimmune Research Laboratory (J.A.B.) and the Diabetes Center (J.A.B., M.A.), University of California, San Francisco, San Francisco
| | - Mark Anderson
- From the Sean N. Parker Autoimmune Research Laboratory (J.A.B.) and the Diabetes Center (J.A.B., M.A.), University of California, San Francisco, San Francisco
| |
Collapse
|
38
|
Mortales CL, Lee SU, Manousadjian A, Hayama KL, Demetriou M. N-Glycan Branching Decouples B Cell Innate and Adaptive Immunity to Control Inflammatory Demyelination. iScience 2020; 23:101380. [PMID: 32745987 PMCID: PMC7398982 DOI: 10.1016/j.isci.2020.101380] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Revised: 06/10/2020] [Accepted: 07/14/2020] [Indexed: 11/21/2022] Open
Abstract
B cell depletion potently reduces episodes of inflammatory demyelination in multiple sclerosis (MS), predominantly through loss of innate rather than adaptive immunity. However, molecular mechanisms controlling innate versus adaptive B cell function are poorly understood. N-glycan branching, via interactions with galectins, controls endocytosis and signaling of cell surface receptors to control cell function. Here we report that N-glycan branching in B cells dose dependently reduces pro-inflammatory innate responses by titrating decreases in Toll-like receptor-4 (TLR4) and TLR2 surface expression via endocytosis. In contrast, a minimal level of N-glycan branching maximizes surface retention of the B cell receptor (BCR) and the CD19 co-receptor to promote adaptive immunity. Branched N-glycans inhibit antigen presentation by B cells to reduce T helper cell-17 (TH17)/TH1 differentiation and inflammatory demyelination in mice. Thus, N-glycan branching negatively regulates B cell innate function while promoting/maintaining adaptive immunity via BCR, providing an attractive therapeutic target for MS.
Collapse
Affiliation(s)
- Christie-Lynn Mortales
- Department of Microbiology & Molecular Genetics, University of California, Irvine, CA 92617, USA
| | - Sung-Uk Lee
- Department of Neurology, University of California, Irvine, CA 92617, USA
| | - Armen Manousadjian
- Department of Neurology, University of California, Irvine, CA 92617, USA
| | - Ken L Hayama
- Department of Microbiology & Molecular Genetics, University of California, Irvine, CA 92617, USA
| | - Michael Demetriou
- Department of Neurology, University of California, Irvine, CA 92617, USA; Department of Microbiology & Molecular Genetics, University of California, Irvine, CA 92617, USA.
| |
Collapse
|
39
|
Baker D, Amor S, Kang AS, Schmierer K, Giovannoni G. The underpinning biology relating to multiple sclerosis disease modifying treatments during the COVID-19 pandemic. Mult Scler Relat Disord 2020; 43:102174. [PMID: 32464584 PMCID: PMC7214323 DOI: 10.1016/j.msard.2020.102174] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Revised: 04/29/2020] [Accepted: 04/30/2020] [Indexed: 02/08/2023]
Abstract
BACKGROUND SARS-CoV-2 viral infection causes COVID-19 that can result in severe acute respiratory distress syndrome (ARDS), which can cause significant mortality, leading to concern that immunosuppressive treatments for multiple sclerosis and other disorders have significant risks for both infection and ARDS. OBJECTIVE To examine the biology that potentially underpins immunity to the SARS-Cov-2 virus and the immunity-induced pathology related to COVID-19 and determine how this impinges on the use of current disease modifying treatments in multiple sclerosis. OBSERVATIONS Although information about the mechanisms of immunity are scant, it appears that monocyte/macrophages and then CD8 T cells are important in eliminating the SARS-CoV-2 virus. This may be facilitated via anti-viral antibody responses that may prevent re-infection. However, viral escape and infection of leucocytes to promote lymphopenia, apparent CD8 T cell exhaustion coupled with a cytokine storm and vascular pathology appears to contribute to the damage in ARDS. IMPLICATIONS In contrast to ablative haematopoietic stem cell therapy, most multiple-sclerosis-related disease modifying therapies do not particularly target the innate immune system and few have any major long-term impact on CD8 T cells to limit protection against COVID-19. In addition, few block the formation of immature B cells within lymphoid tissue that will provide antibody-mediated protection from (re)infection. However, adjustments to dosing schedules may help de-risk the chance of infection further and reduce the concerns of people with MS being treated during the COVID-19 pandemic.
Collapse
Key Words
- ace2, angiotensin converting enzyme two
- ards, acute respiratory distress syndrome
- asc, antibody secreting cells
- cns, central nervous system
- dmt, disease modifying therapies
- (hsct), haematopoietic stem cell therapy
- irt, immune reconstitution therapies
- ms, multiple sclerosis
- rbd, receptor binding domain
- rna, ribonucleic acid
- sars, severe acute respiratory syndrome
Collapse
Affiliation(s)
- David Baker
- Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, E1 2AT; United Kingdom.
| | - Sandra Amor
- Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, E1 2AT; United Kingdom; Pathology Department, VUmc, Amsterdam UMC, Amsterdam, The Netherlands.
| | - Angray S Kang
- Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, E1 2AT; United Kingdom; Centre for Oral Immunobiology and Regenerative Medicine, Institute of Dentistry, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, United Kingdom
| | - Klaus Schmierer
- Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, E1 2AT; United Kingdom; Clinical Board:Medicine (Neuroscience), The Royal London Hospital, Barts Health NHS Trust, London, United Kingdom
| | - Gavin Giovannoni
- Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, E1 2AT; United Kingdom; Clinical Board:Medicine (Neuroscience), The Royal London Hospital, Barts Health NHS Trust, London, United Kingdom
| |
Collapse
|
40
|
Sádaba MC, Rothhammer V, Muñoz Ú, Sebal C, Escudero E, Kivisäkk P, Garcia Sanchez MI, Izquierdo G, Hauser SL, Baranzini SE, Oksenberg JR, Álvarez-Lafuente R, Bakshi R, Weiner HL, Quintana FJ. Serum antibodies to phosphatidylcholine in MS. NEUROLOGY(R) NEUROIMMUNOLOGY & NEUROINFLAMMATION 2020; 7:e765. [PMID: 32518205 PMCID: PMC7309529 DOI: 10.1212/nxi.0000000000000765] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Accepted: 04/09/2020] [Indexed: 12/23/2022]
Abstract
OBJECTIVE To evaluate the value of serum immunoglobulin G (IgG) and immunoglobulin M (IgM) antibodies reactive with phosphatidylcholine (PC) and lactosylceramide (LC) as biomarkers in MS. METHODS We developed an ultrasensitive ELISA technique to analyze serum IgG and IgM antibodies to LC and PC, which we used to analyze samples from 362 patients with MS, 10 patients with non-MS myelin diseases (Non-MSMYDs), 11 patients with nonmyelin neurologic diseases (Non-MYNDs), and 80 controls. MS serum samples included clinically isolated syndrome (CIS, n = 17), relapsing-remitting MS (RRMS, n = 62), secondary progressive MS (SPMS, n = 50), primary progressive MS (PPMS, n = 37), and benign MS (BENMS, n = 36). RESULTS We detected higher levels of serum IgM antibodies to PC (IgM-PC) in MS than control samples; patients with CIS and RRMS showed higher IgM-PC levels than patients with SPMS, PPMS, and BENMS and controls. MS and control samples did not differ in serum levels of IgM antibodies reactive with LC, nor in IgG antibodies reactive with LC or PC. CONCLUSIONS Serum IgM-PC antibodies are elevated in patients with MS, particularly during the CIS and RRMS phases of the disease. Thus, serum IgM-PC is a candidate biomarker for early inflammatory stages of MS. CLASSIFICATION OF EVIDENCE This study provides Class III evidence that serum antibodies to PC are elevated in patients with MS. The study is rated Class III because of the case control design and the risk of spectrum bias: antibody levels in patients with MS were compared with healthy controls.
Collapse
Affiliation(s)
- Maria Cruz Sádaba
- From the Ann Romney Center for Neurologic Diseases (M.C.S., V.R., P.K., R.B., H.L.W., F.J.Q.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA; Facultad de Medicina (M.C.S., U.M., C.S., E.E.), Instituto de Medicina Molecular Aplicada (INMA), Universidad San Pablo-CEU, CEU Universities, Madrid; Molecular Biology Service and MS Unit (M.I.G.S., G.I.), University of Sevilla; Department of Neurology (S.L.H., S.E.B., J.R.O.), University of California, San Francisco; Instituto de Investigación Sanitaria San Carlos (IdISSC) (R.Á.-L.), Hospital Clínico San Carlos, Madrid, Spain; and The Broad Institute of Harvard and MIT (F.J.Q.), Cambridge, MA.
| | - Veit Rothhammer
- From the Ann Romney Center for Neurologic Diseases (M.C.S., V.R., P.K., R.B., H.L.W., F.J.Q.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA; Facultad de Medicina (M.C.S., U.M., C.S., E.E.), Instituto de Medicina Molecular Aplicada (INMA), Universidad San Pablo-CEU, CEU Universities, Madrid; Molecular Biology Service and MS Unit (M.I.G.S., G.I.), University of Sevilla; Department of Neurology (S.L.H., S.E.B., J.R.O.), University of California, San Francisco; Instituto de Investigación Sanitaria San Carlos (IdISSC) (R.Á.-L.), Hospital Clínico San Carlos, Madrid, Spain; and The Broad Institute of Harvard and MIT (F.J.Q.), Cambridge, MA
| | - Úrsula Muñoz
- From the Ann Romney Center for Neurologic Diseases (M.C.S., V.R., P.K., R.B., H.L.W., F.J.Q.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA; Facultad de Medicina (M.C.S., U.M., C.S., E.E.), Instituto de Medicina Molecular Aplicada (INMA), Universidad San Pablo-CEU, CEU Universities, Madrid; Molecular Biology Service and MS Unit (M.I.G.S., G.I.), University of Sevilla; Department of Neurology (S.L.H., S.E.B., J.R.O.), University of California, San Francisco; Instituto de Investigación Sanitaria San Carlos (IdISSC) (R.Á.-L.), Hospital Clínico San Carlos, Madrid, Spain; and The Broad Institute of Harvard and MIT (F.J.Q.), Cambridge, MA
| | - Cristina Sebal
- From the Ann Romney Center for Neurologic Diseases (M.C.S., V.R., P.K., R.B., H.L.W., F.J.Q.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA; Facultad de Medicina (M.C.S., U.M., C.S., E.E.), Instituto de Medicina Molecular Aplicada (INMA), Universidad San Pablo-CEU, CEU Universities, Madrid; Molecular Biology Service and MS Unit (M.I.G.S., G.I.), University of Sevilla; Department of Neurology (S.L.H., S.E.B., J.R.O.), University of California, San Francisco; Instituto de Investigación Sanitaria San Carlos (IdISSC) (R.Á.-L.), Hospital Clínico San Carlos, Madrid, Spain; and The Broad Institute of Harvard and MIT (F.J.Q.), Cambridge, MA
| | - Esther Escudero
- From the Ann Romney Center for Neurologic Diseases (M.C.S., V.R., P.K., R.B., H.L.W., F.J.Q.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA; Facultad de Medicina (M.C.S., U.M., C.S., E.E.), Instituto de Medicina Molecular Aplicada (INMA), Universidad San Pablo-CEU, CEU Universities, Madrid; Molecular Biology Service and MS Unit (M.I.G.S., G.I.), University of Sevilla; Department of Neurology (S.L.H., S.E.B., J.R.O.), University of California, San Francisco; Instituto de Investigación Sanitaria San Carlos (IdISSC) (R.Á.-L.), Hospital Clínico San Carlos, Madrid, Spain; and The Broad Institute of Harvard and MIT (F.J.Q.), Cambridge, MA
| | - Pia Kivisäkk
- From the Ann Romney Center for Neurologic Diseases (M.C.S., V.R., P.K., R.B., H.L.W., F.J.Q.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA; Facultad de Medicina (M.C.S., U.M., C.S., E.E.), Instituto de Medicina Molecular Aplicada (INMA), Universidad San Pablo-CEU, CEU Universities, Madrid; Molecular Biology Service and MS Unit (M.I.G.S., G.I.), University of Sevilla; Department of Neurology (S.L.H., S.E.B., J.R.O.), University of California, San Francisco; Instituto de Investigación Sanitaria San Carlos (IdISSC) (R.Á.-L.), Hospital Clínico San Carlos, Madrid, Spain; and The Broad Institute of Harvard and MIT (F.J.Q.), Cambridge, MA
| | - Maria Isabel Garcia Sanchez
- From the Ann Romney Center for Neurologic Diseases (M.C.S., V.R., P.K., R.B., H.L.W., F.J.Q.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA; Facultad de Medicina (M.C.S., U.M., C.S., E.E.), Instituto de Medicina Molecular Aplicada (INMA), Universidad San Pablo-CEU, CEU Universities, Madrid; Molecular Biology Service and MS Unit (M.I.G.S., G.I.), University of Sevilla; Department of Neurology (S.L.H., S.E.B., J.R.O.), University of California, San Francisco; Instituto de Investigación Sanitaria San Carlos (IdISSC) (R.Á.-L.), Hospital Clínico San Carlos, Madrid, Spain; and The Broad Institute of Harvard and MIT (F.J.Q.), Cambridge, MA
| | - Guillermo Izquierdo
- From the Ann Romney Center for Neurologic Diseases (M.C.S., V.R., P.K., R.B., H.L.W., F.J.Q.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA; Facultad de Medicina (M.C.S., U.M., C.S., E.E.), Instituto de Medicina Molecular Aplicada (INMA), Universidad San Pablo-CEU, CEU Universities, Madrid; Molecular Biology Service and MS Unit (M.I.G.S., G.I.), University of Sevilla; Department of Neurology (S.L.H., S.E.B., J.R.O.), University of California, San Francisco; Instituto de Investigación Sanitaria San Carlos (IdISSC) (R.Á.-L.), Hospital Clínico San Carlos, Madrid, Spain; and The Broad Institute of Harvard and MIT (F.J.Q.), Cambridge, MA
| | - Stephen L Hauser
- From the Ann Romney Center for Neurologic Diseases (M.C.S., V.R., P.K., R.B., H.L.W., F.J.Q.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA; Facultad de Medicina (M.C.S., U.M., C.S., E.E.), Instituto de Medicina Molecular Aplicada (INMA), Universidad San Pablo-CEU, CEU Universities, Madrid; Molecular Biology Service and MS Unit (M.I.G.S., G.I.), University of Sevilla; Department of Neurology (S.L.H., S.E.B., J.R.O.), University of California, San Francisco; Instituto de Investigación Sanitaria San Carlos (IdISSC) (R.Á.-L.), Hospital Clínico San Carlos, Madrid, Spain; and The Broad Institute of Harvard and MIT (F.J.Q.), Cambridge, MA
| | - Sergio E Baranzini
- From the Ann Romney Center for Neurologic Diseases (M.C.S., V.R., P.K., R.B., H.L.W., F.J.Q.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA; Facultad de Medicina (M.C.S., U.M., C.S., E.E.), Instituto de Medicina Molecular Aplicada (INMA), Universidad San Pablo-CEU, CEU Universities, Madrid; Molecular Biology Service and MS Unit (M.I.G.S., G.I.), University of Sevilla; Department of Neurology (S.L.H., S.E.B., J.R.O.), University of California, San Francisco; Instituto de Investigación Sanitaria San Carlos (IdISSC) (R.Á.-L.), Hospital Clínico San Carlos, Madrid, Spain; and The Broad Institute of Harvard and MIT (F.J.Q.), Cambridge, MA
| | - Jorge R Oksenberg
- From the Ann Romney Center for Neurologic Diseases (M.C.S., V.R., P.K., R.B., H.L.W., F.J.Q.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA; Facultad de Medicina (M.C.S., U.M., C.S., E.E.), Instituto de Medicina Molecular Aplicada (INMA), Universidad San Pablo-CEU, CEU Universities, Madrid; Molecular Biology Service and MS Unit (M.I.G.S., G.I.), University of Sevilla; Department of Neurology (S.L.H., S.E.B., J.R.O.), University of California, San Francisco; Instituto de Investigación Sanitaria San Carlos (IdISSC) (R.Á.-L.), Hospital Clínico San Carlos, Madrid, Spain; and The Broad Institute of Harvard and MIT (F.J.Q.), Cambridge, MA
| | - Roberto Álvarez-Lafuente
- From the Ann Romney Center for Neurologic Diseases (M.C.S., V.R., P.K., R.B., H.L.W., F.J.Q.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA; Facultad de Medicina (M.C.S., U.M., C.S., E.E.), Instituto de Medicina Molecular Aplicada (INMA), Universidad San Pablo-CEU, CEU Universities, Madrid; Molecular Biology Service and MS Unit (M.I.G.S., G.I.), University of Sevilla; Department of Neurology (S.L.H., S.E.B., J.R.O.), University of California, San Francisco; Instituto de Investigación Sanitaria San Carlos (IdISSC) (R.Á.-L.), Hospital Clínico San Carlos, Madrid, Spain; and The Broad Institute of Harvard and MIT (F.J.Q.), Cambridge, MA
| | - Rohit Bakshi
- From the Ann Romney Center for Neurologic Diseases (M.C.S., V.R., P.K., R.B., H.L.W., F.J.Q.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA; Facultad de Medicina (M.C.S., U.M., C.S., E.E.), Instituto de Medicina Molecular Aplicada (INMA), Universidad San Pablo-CEU, CEU Universities, Madrid; Molecular Biology Service and MS Unit (M.I.G.S., G.I.), University of Sevilla; Department of Neurology (S.L.H., S.E.B., J.R.O.), University of California, San Francisco; Instituto de Investigación Sanitaria San Carlos (IdISSC) (R.Á.-L.), Hospital Clínico San Carlos, Madrid, Spain; and The Broad Institute of Harvard and MIT (F.J.Q.), Cambridge, MA
| | - Howard L Weiner
- From the Ann Romney Center for Neurologic Diseases (M.C.S., V.R., P.K., R.B., H.L.W., F.J.Q.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA; Facultad de Medicina (M.C.S., U.M., C.S., E.E.), Instituto de Medicina Molecular Aplicada (INMA), Universidad San Pablo-CEU, CEU Universities, Madrid; Molecular Biology Service and MS Unit (M.I.G.S., G.I.), University of Sevilla; Department of Neurology (S.L.H., S.E.B., J.R.O.), University of California, San Francisco; Instituto de Investigación Sanitaria San Carlos (IdISSC) (R.Á.-L.), Hospital Clínico San Carlos, Madrid, Spain; and The Broad Institute of Harvard and MIT (F.J.Q.), Cambridge, MA
| | - Francisco J Quintana
- From the Ann Romney Center for Neurologic Diseases (M.C.S., V.R., P.K., R.B., H.L.W., F.J.Q.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA; Facultad de Medicina (M.C.S., U.M., C.S., E.E.), Instituto de Medicina Molecular Aplicada (INMA), Universidad San Pablo-CEU, CEU Universities, Madrid; Molecular Biology Service and MS Unit (M.I.G.S., G.I.), University of Sevilla; Department of Neurology (S.L.H., S.E.B., J.R.O.), University of California, San Francisco; Instituto de Investigación Sanitaria San Carlos (IdISSC) (R.Á.-L.), Hospital Clínico San Carlos, Madrid, Spain; and The Broad Institute of Harvard and MIT (F.J.Q.), Cambridge, MA.
| |
Collapse
|
41
|
Pistono C, Monti MC, Boiocchi C, Berzolari FG, Osera C, Mallucci G, Cuccia M, Pascale A, Montomoli C, Bergamaschi R. Response to oxidative stress of peripheral blood mononuclear cells from multiple sclerosis patients and healthy controls. Cell Stress Chaperones 2020; 25:81-91. [PMID: 31720998 PMCID: PMC6985352 DOI: 10.1007/s12192-019-01049-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Revised: 10/08/2019] [Accepted: 10/28/2019] [Indexed: 12/21/2022] Open
Abstract
The complex scenario of multiple sclerosis (MS) pathology involves several mechanisms, including oxidative stress response. The heat shock proteins (HSPs) are important for the protection of the cells; however, their role in MS is not clear. The present research is focused on the response of peripheral blood mononuclear cells (PBMCs) to oxidative stress and to the involvement of HSP70-2 (a protein coded by the HSPA1B gene, located in the MHC class III). To this aim, we challenged PBMCs from MS patients and healthy controls with hydrogen peroxide. Specifically, PBMCs mitochondrial activity, HSP70-2 protein expression and the production of intracellular reactive oxygen species were assessed. These parameters were also related to the HSP70-2 rs1061581 polymorphism, which is linked to the risk of developing MS. Moreover, mitochondrial activity and HSP70-2 protein levels were also related to disease severity. Overall, our results indicate that PBMCs, from both MS patients and healthy controls, may display a similar response towards an oxidative insult; within this context, HSP70-2 does not seem to be central in the protection of PBMCs. Nevertheless, the HSP70-2 rs1061581 polymorphism is related to ROS levels and appears to have a role in the different expression of HSP70-2 under oxidative stimulus.
Collapse
Affiliation(s)
- Cristiana Pistono
- Laboratory of Immunogenetics, Department of Biology and Biotechnology "L. Spallanzani", University of Pavia, Pavia, Italy.
- Laboratoire de Neurosciences Cognitives et Adaptatives (LNCA), CNRS/Université de Strasbourg, Faculté de psychologie, UMR, 7364, Strasbourg, France.
| | - Maria Cristina Monti
- Department of Public Health Experimental and Forensic Medicine, Unit of Biostatistics and Clinical Epidemiology, University of Pavia, Pavia, Italy
| | - Chiara Boiocchi
- Inter-Department Multiple Sclerosis Research Centre, National Neurological Institute "C. Mondino", Pavia, Italy
| | - Francesca Gigli Berzolari
- Department of Public Health Experimental and Forensic Medicine, Unit of Biostatistics and Clinical Epidemiology, University of Pavia, Pavia, Italy
| | - Cecilia Osera
- Department of Drug Sciences, Section of Pharmacology, University of Pavia, Pavia, Italy
| | - Giulia Mallucci
- Inter-Department Multiple Sclerosis Research Centre, National Neurological Institute "C. Mondino", Pavia, Italy
| | - Mariaclara Cuccia
- Laboratory of Immunogenetics, Department of Biology and Biotechnology "L. Spallanzani", University of Pavia, Pavia, Italy
| | - Alessia Pascale
- Department of Drug Sciences, Section of Pharmacology, University of Pavia, Pavia, Italy
| | - Cristina Montomoli
- Department of Public Health Experimental and Forensic Medicine, Unit of Biostatistics and Clinical Epidemiology, University of Pavia, Pavia, Italy
| | - Roberto Bergamaschi
- Inter-Department Multiple Sclerosis Research Centre, National Neurological Institute "C. Mondino", Pavia, Italy
| |
Collapse
|
42
|
Migotto MA, Mardon K, Orian J, Weckbecker G, Kneuer R, Bhalla R, Reutens DC. Efficient Distribution of a Novel Zirconium-89 Labeled Anti-cd20 Antibody Following Subcutaneous and Intravenous Administration in Control and Experimental Autoimmune Encephalomyelitis-Variant Mice. Front Immunol 2019; 10:2437. [PMID: 31681317 PMCID: PMC6813232 DOI: 10.3389/fimmu.2019.02437] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Accepted: 09/30/2019] [Indexed: 11/13/2022] Open
Abstract
Objective: To investigate the imaging and biodistribution of a novel zirconium-89 (89Zr)-labeled mouse anti-cd20 monoclonal antibody (mAb) in control and experimental autoimmune encephalomyelitis (EAE) mice following subcutaneous (s. c.) and intravenous (i.v.) administration. Background: Anti-cd20-mediated B-cell depletion using mAbs is a promising therapy for multiple sclerosis. Recombinant human myelin oligodendrocyte glycoprotein (rhMOG)-induced EAE involves B-cell-mediated inflammation and demyelination in mice. Design/Methods: C57BL/6J mice (n = 39) were EAE-induced using rhMOG. On Day 14 post EAE induction, 89Zr-labeled-anti-cd20 mAb was injected in control and EAE mice in the right lower flank (s.c.) or tail vein (i.v.). Positron emission tomography/computed tomography (PET/CT) imaging and gamma counting (ex vivo) were performed on Days 1, 3, and 7 to quantify tracer accumulation in the major organs, lymphatics, and central nervous system (CNS). A preliminary study was conducted in healthy mice to elucidate full and early kinetics of the tracer that were subsequently applied in the EAE and control mice study. Results:89Zr-labeled anti-cd20 mAb was effectively absorbed from s.c. and i.v. injection sites and distributed to all major organs in the EAE and control mice. There was a good correlation between in vivo PET/CT data and ex vivo quantification of biodistribution of the tracer. From gamma counting studies, initial tracer uptake within the lymphatic system was found to be higher in the draining lymph nodes (inguinal or subiliac and sciatic) following s.c. vs. i.v. administration; within the CNS a significantly higher tracer uptake was observed at 24 h in the cerebellum, cerebrum, and thoracic spinal cord (p < 0.05 for all) following s.c. vs. i.v. administration. Conclusions: The preclinical data suggest that initial tracer uptake was significantly higher in the draining lymph nodes (subiliac and sciatic) and parts of CNS (the cerebellum and cerebrum) when administered s.c. compared with i.v in EAE mice.
Collapse
Affiliation(s)
- Mary-Anne Migotto
- Centre for Advanced Imaging, The University of Queensland, Brisbane, QLD, Australia
| | - Karine Mardon
- Centre for Advanced Imaging, The University of Queensland, Brisbane, QLD, Australia.,National Imaging Facility, The University of Queensland, Brisbane, QLD, Australia
| | - Jacqueline Orian
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC, Australia
| | - Gisbert Weckbecker
- Novartis Institutes for BioMedical Research, Novartis Pharma AG, Basel, Switzerland
| | - Rainer Kneuer
- Novartis Institutes for BioMedical Research, Novartis Pharma AG, Basel, Switzerland
| | - Rajiv Bhalla
- Centre for Advanced Imaging, The University of Queensland, Brisbane, QLD, Australia.,ARC Training Centre for Innovation in Biomedical Imaging Technology, Brisbane, QLD, Australia
| | - David C Reutens
- Centre for Advanced Imaging, The University of Queensland, Brisbane, QLD, Australia.,ARC Training Centre for Innovation in Biomedical Imaging Technology, Brisbane, QLD, Australia
| |
Collapse
|
43
|
Kolahdouzan M, Futhey NC, Kieran NW, Healy LM. Novel Molecular Leads for the Prevention of Damage and the Promotion of Repair in Neuroimmunological Disease. Front Immunol 2019; 10:1657. [PMID: 31379852 PMCID: PMC6658885 DOI: 10.3389/fimmu.2019.01657] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Accepted: 07/03/2019] [Indexed: 11/20/2022] Open
Abstract
Neuroinflammation is a prominent pathological feature of all neuroimmunological diseases, including, but not limited to, multiple sclerosis (MS), myasthenia gravis, neuromyelitis optica, and Guillain–Barré syndrome. All currently-approved therapies for the treatment of these diseases focus on controlling or modulating the immune (innate and adaptive) responses to limit demyelination and neuronal damage. The primary purpose of this review is to detail the pre-clinical data and proposed mechanism of action of novel drugs currently in clinical trial, with a focus on novel compounds that promote repair and regeneration in the central nervous system (CNS). As the most recent advances have been made in the field of MS research, this review will focus primarily on this disease and its animal models. However, these compounds are likely to be effective for a range of indications with a neuroinflammatory component. Traditionally, MS was thought to proceed through two distinct phases. The first, predominantly inflammatory stage, is characterized by acute episodes of clinical relapse, followed by periods of partial or total recovery with an apparent absence of overall disease progression. In the vast majority of patients, this relapsing-remitting disease subsequently progresses into a second more chronic, neurodegenerative phase, which is characterized by oligodendrocyte damage and axonal destruction leading to brain atrophy and an accumulation of disability. Recent work has shown that rather than occurring independently, both the inflammatory and degenerative phases may run concurrently. This, combined with evidence that early therapeutic intervention slows accumulation of disability and delays progression, highlights the need for novel therapeutic approaches that promote repair and regeneration early in the disease trajectory. Such compounds may be used as monotherapies or in conjunction with classical anti-inflammatory therapies. This review will highlight novel therapies currently in clinical trial, and likely to appear in clinical practice in the near future, focusing on compounds that target the immune system and/or enhance endogenous repair mechanisms in the CNS.
Collapse
Affiliation(s)
- Mahshad Kolahdouzan
- Neuroimmunology Unit, Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montréal, QC, Canada
| | - Naomi C Futhey
- Neuroimmunology Unit, Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montréal, QC, Canada
| | - Nicholas W Kieran
- Neuroimmunology Unit, Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montréal, QC, Canada
| | - Luke M Healy
- Neuroimmunology Unit, Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montréal, QC, Canada
| |
Collapse
|
44
|
Abstract
Increasing evidence suggests that B cells contribute both to the regulation of normal autoimmunity and to the pathogenesis of immune mediated diseases, including multiple sclerosis (MS). B cells in MS are skewed toward a pro-inflammatory profile, and contribute to MS pathogenesis by antibody production, antigen presentation, T cells stimulation and activation, driving autoproliferation of brain-homing autoreactive CD4+ T cells, production of pro-inflammatory cytokines, and formation of ectopic meningeal germinal centers that drive cortical pathology and contribute to neurological disability. The recent interest in the key role of B cells in MS has been evoked by the profound anti-inflammatory effects of rituximab, a chimeric monoclonal antibody (mAb) targeting the B cell surface marker CD20, observed in relapsing-remitting MS. This has been reaffirmed by clinical trials with less immunogenic and more potent B cell-depleting mAbs targeting CD20 – ocrelizumab, ofatumumab and ublituximab. Ocrelizumab is also the first disease-modifying drug that has shown efficacy in primary-progressive MS, and is currently approved for both indications. Another promising approach is the inhibition of Bruton's tyrosine kinase, a key enzyme that mediates B cell activation and survival, by agents such as evobrutinib. On the other hand, targeting B cell cytokines with the fusion protein atacicept increased MS activity, highlighting the complex and not fully understood role of B cells and humoral immunity in MS. Finally, all other approved therapies for MS, some of which have been designed to target T cells, have some effects on the frequency, phenotype, or homing of B cells, which may contribute to their therapeutic activity.
Collapse
Affiliation(s)
- Ron Milo
- Ron Milo, Department of Neurology, Barzilai Medical Center, Ha-Histadrut St 2, Ashkelon 7308604, Israel,
| |
Collapse
|
45
|
Milo R. Therapies for multiple sclerosis targeting B cells. Croat Med J 2019; 60:87-98. [PMID: 31044580 PMCID: PMC6509632] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2019] [Accepted: 04/14/2019] [Indexed: 10/12/2023] Open
Abstract
Increasing evidence suggests that B cells contribute both to the regulation of normal autoimmunity and to the pathogenesis of immune mediated diseases, including multiple sclerosis (MS). B cells in MS are skewed toward a pro-inflammatory profile, and contribute to MS pathogenesis by antibody production, antigen presentation, T cells stimulation and activation, driving autoproliferation of brain-homing autoreactive CD4+ T cells, production of pro-inflammatory cytokines, and formation of ectopic meningeal germinal centers that drive cortical pathology and contribute to neurological disability. The recent interest in the key role of B cells in MS has been evoked by the profound anti-inflammatory effects of rituximab, a chimeric monoclonal antibody (mAb) targeting the B cell surface marker CD20, observed in relapsing-remitting MS. This has been reaffirmed by clinical trials with less immunogenic and more potent B cell-depleting mAbs targeting CD20 - ocrelizumab, ofatumumab and ublituximab. Ocrelizumab is also the first disease-modifying drug that has shown efficacy in primary-progressive MS, and is currently approved for both indications. Another promising approach is the inhibition of Bruton's tyrosine kinase, a key enzyme that mediates B cell activation and survival, by agents such as evobrutinib. On the other hand, targeting B cell cytokines with the fusion protein atacicept increased MS activity, highlighting the complex and not fully understood role of B cells and humoral immunity in MS. Finally, all other approved therapies for MS, some of which have been designed to target T cells, have some effects on the frequency, phenotype, or homing of B cells, which may contribute to their therapeutic activity.
Collapse
Affiliation(s)
- Ron Milo
- Ron Milo, Department of Neurology, Barzilai Medical Center, Ha-Histadrut St 2, Ashkelon 7308604, Israel,
| |
Collapse
|
46
|
Voo VTF, O'Brien T, Butzkueven H, Monif M. The role of vitamin D and P2X7R in multiple sclerosis. J Neuroimmunol 2019; 330:159-169. [PMID: 30908981 DOI: 10.1016/j.jneuroim.2019.03.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Revised: 02/11/2019] [Accepted: 03/05/2019] [Indexed: 12/13/2022]
Abstract
Multiple sclerosis (MS) is characterized by neuroinflammatory infiltrates and central nervous system demyelination. In the neuroinflammatory foci of MS there is increased expression of a purinergic receptor, P2X7R. Although implicated in the neuroinflammation, the exact role of P2X7R in the context of MS is unclear and forms the basis of this review. In this review, we also introduce the immunopathologies and inflammatory processes in MS, with a focus on P2X7R and the possible immunomodulatory role of vitamin D deficiency in this setting.
Collapse
Affiliation(s)
- Veronica Tsin Fong Voo
- Department of Physiology, The University of Melbourne, Melbourne, Australia; Department of Neuroscience, Monash University, Melbourne, Australia
| | - Terence O'Brien
- Department of Neuroscience, Monash University, Melbourne, Australia; Department of Neurology, Melbourne Health, Melbourne, Australia
| | | | - Mastura Monif
- Department of Physiology, The University of Melbourne, Melbourne, Australia; Department of Neuroscience, Monash University, Melbourne, Australia; Department of Neurology, Melbourne Health, Melbourne, Australia.
| |
Collapse
|
47
|
Regulatory B and T lymphocytes in multiple sclerosis: friends or foes? AUTOIMMUNITY HIGHLIGHTS 2018; 9:9. [PMID: 30415321 PMCID: PMC6230324 DOI: 10.1007/s13317-018-0109-x] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/04/2018] [Accepted: 10/17/2018] [Indexed: 12/12/2022]
Abstract
Current clinical experience with immunomodulatory agents and monoclonal antibodies in principle has established the benefit of depleting lymphocytic populations in relapsing–remitting multiple sclerosis (RRMS). B and T cells may exert multiple pro-inflammatory actions, but also possess regulatory functions making their role in RRMS pathogenesis much more complex. There is no clear correlation of Tregs and Bregs with clinical features of the disease. Herein, we discuss the emerging data on regulatory T and B cell subset distributions in MS and their roles in the pathophysiology of MS and its murine model, experimental autoimmune encephalomyelitis (EAE). In addition, we summarize the immunomodulatory properties of certain MS therapeutic agents through their effect on such regulatory cell subsets and their relevance to clinical outcomes.
Collapse
|
48
|
Han J, Zhu K, Zhang X, Harris RA. Enforced microglial depletion and repopulation as a promising strategy for the treatment of neurological disorders. Glia 2018; 67:217-231. [PMID: 30378163 PMCID: PMC6635749 DOI: 10.1002/glia.23529] [Citation(s) in RCA: 73] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2018] [Revised: 08/17/2018] [Accepted: 08/22/2018] [Indexed: 01/18/2023]
Abstract
Microglia are prominent immune cells in the central nervous system (CNS) and are critical players in both neurological development and homeostasis, and in neurological diseases when dysfunctional. Our previous understanding of the phenotypes and functions of microglia has been greatly extended by a dearth of recent investigations. Distinct genetically defined subsets of microglia are now recognized to perform their own independent functions in specific conditions. The molecular profiling of single microglial cells indicates extensively heterogeneous reactions in different neurological disorders, resulting in multiple potentials for crosstalk with other kinds of CNS cells such as astrocytes and neurons. In settings of neurological diseases it could thus be prudent to establish effective cell‐based therapies by targeting entire microglial networks. Notably, activated microglial depletion through genetic targeting or pharmacological therapies within a suitable time window can stimulate replenishment of the CNS niche with new microglia. Additionally, enforced repopulation through provision of replacement cells also represents a potential means of exchanging dysfunctional with functional microglia. In each setting the newly repopulated microglia might have the potential to resolve ongoing neuroinflammation. In this review, we aim to summarize the most recent knowledge of microglia and to highlight microglial depletion and subsequent repopulation as a promising cell replacement therapy. Although glial cell replacement therapy is still in its infancy and future translational studies are still required, the approach is scientifically sound and provides new optimism for managing the neurotoxicity and neuroinflammation induced by activated microglia.
Collapse
Affiliation(s)
- Jinming Han
- Applied Immunology and Immunotherapy, Department of Clinical NeuroscienceKarolinska Institutet, Center for Molecular Medicine, Karolinska University Hospital at SolnaStockholmSweden
| | - Keying Zhu
- Applied Immunology and Immunotherapy, Department of Clinical NeuroscienceKarolinska Institutet, Center for Molecular Medicine, Karolinska University Hospital at SolnaStockholmSweden
| | - Xing‐Mei Zhang
- Applied Immunology and Immunotherapy, Department of Clinical NeuroscienceKarolinska Institutet, Center for Molecular Medicine, Karolinska University Hospital at SolnaStockholmSweden
| | - Robert A. Harris
- Applied Immunology and Immunotherapy, Department of Clinical NeuroscienceKarolinska Institutet, Center for Molecular Medicine, Karolinska University Hospital at SolnaStockholmSweden
| |
Collapse
|
49
|
Negron A, Robinson RR, Stüve O, Forsthuber TG. The role of B cells in multiple sclerosis: Current and future therapies. Cell Immunol 2018; 339:10-23. [PMID: 31130183 DOI: 10.1016/j.cellimm.2018.10.006] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2018] [Revised: 10/19/2018] [Accepted: 10/20/2018] [Indexed: 02/07/2023]
Abstract
While it was long held that T cells were the primary mediators of multiple sclerosis (MS) pathogenesis, the beneficial effects observed in response to treatment with Rituximab (RTX), a monoclonal antibody (mAb) targeting CD20, shed light on a key contributor to MS that had been previously underappreciated: B cells. This has been reaffirmed by results from clinical trials testing the efficacy of subsequently developed B cell-depleting mAbs targeting CD20 as well as studies revisiting the effects of previous disease-modifying therapies (DMTs) on B cell subsets thought to modulate disease severity. In this review, we summarize current knowledge regarding the complex roles of B cells in MS pathogenesis and current and potential future B cell-directed therapies.
Collapse
Affiliation(s)
- Austin Negron
- Department of Biology, University of Texas at San Antonio, TX 78249, USA
| | - Rachel R Robinson
- Department of Biology, University of Texas at San Antonio, TX 78249, USA
| | - Olaf Stüve
- Department of Neurology and Neurotherapeutics, University of Texas Southwestern Medical Center, Dallas, TX, USA; Neurology Section, VA North Texas Health Care System, Medical Service, Dallas, TX, USA
| | | |
Collapse
|
50
|
Greenfield AL, Hauser SL. B-cell Therapy for Multiple Sclerosis: Entering an era. Ann Neurol 2018; 83:13-26. [PMID: 29244240 PMCID: PMC5876115 DOI: 10.1002/ana.25119] [Citation(s) in RCA: 165] [Impact Index Per Article: 23.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2017] [Revised: 12/01/2017] [Accepted: 12/09/2017] [Indexed: 12/12/2022]
Abstract
Monoclonal antibodies that target CD20 expressing B cells represent an important new treatment option for patients with multiple sclerosis (MS). B-cell-depleting therapy is highly effective against relapsing forms of the disease and is also the first treatment approach proven to protect against disability worsening in primary progressive MS. Moreover, evolving clinical experience with B-cell therapy, combined with a more sophisticated understanding of humoral immunity in preclinical models and in patients with MS, has led to major progress in deciphering the immune pathogenesis of MS. Here, we review the nuanced roles of B cells in MS autoimmunity, the clinical data supporting use of ocrelizumab and other anti-CD20 therapies in the treatment of MS, as well as safety and practical considerations for prescribing. Last, we summarize remaining unanswered questions regarding the proper role of anti-CD20 therapy in MS, its limitations, and the future landscape of B-cell-based approaches to treatment. Ann Neurol 2018;83:13-26.
Collapse
Affiliation(s)
- Ariele L. Greenfield
- Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, San Francisco, California
| | - Stephen L. Hauser
- Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, San Francisco, California
| |
Collapse
|