1
|
Etemadifar M, Nouri H, Sedaghat N, Ramezani A, Kargaran PK, Salari M, Kaveyee H. Anti-CD20 therapies for pediatric-onset multiple sclerosis: A systematic review. Mult Scler Relat Disord 2024; 91:105849. [PMID: 39243503 DOI: 10.1016/j.msard.2024.105849] [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: 02/04/2024] [Revised: 08/10/2024] [Accepted: 08/26/2024] [Indexed: 09/09/2024]
Abstract
BACKGROUND Pediatric-onset multiple sclerosis (POMS) cases, defined as multiple sclerosis (MS) with onset before the age of 18, represent between 3 and 5 % of all MS patients. Anti-CD20 drugs mainly rituximab, ocrelizumab, and ofatumumab are being widely used in adult-onset MS. Their use in POMS is also being increasingly considered by experts. OBJECTIVE to review the latest evidence on safety and efficacy of the use of anti-CD20 therapies in POMS. METHODS An extensive search was performed in PubMed, Scopus, and Web of Science databases until the end of July 1st, 2024. Two independent reviewers screened the articles, and collected data. 832 studies were screened using Preferred Reporting Items for Systematic Reviews and Meta-analyses (PRISMA) guidelines. RESULTS 12 studies on rituximab (328 patients) and 6 studies on ocrelizumab (106 patients) were synthesized. Using monoclonal antibodies in POMS patients has a noteworthy effect on reducing relapses and lesions and achieving no evidence of disease activity especially in highly active POMS patients. However, anti-CD20 therapies in MS are associated with potential adverse events (AEs). Additional data is required on the effect of anti-CD20 therapy on disability accrual. CONCLUSION Although anti-CD20 therapy is associated with some AEs, it can be provided in several circumstances, especially to patients with highly active disease, or ones resistant to platform therapies.
Collapse
Affiliation(s)
- Masoud Etemadifar
- Department of Neurosurgery, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Hosein Nouri
- School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran; Network of Immunity in Infection, Malignancy, and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Isfahan, Iran
| | - Nahad Sedaghat
- School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran; Network of Immunity in Infection, Malignancy, and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Isfahan, Iran
| | - Aryana Ramezani
- School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Parisa K Kargaran
- Department of Cardiovascular Medicine, Center for Regenerative Medicine, Mayo Clinic, Rochester, Rochester, MN, USA
| | - Mehri Salari
- Functional Neurosurgery Research Center, Shohada Tajrish Comprehensive Neurosurgical Center of Excellence, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hasan Kaveyee
- School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran.
| |
Collapse
|
2
|
Konen FF, Gingele S, Hümmert MW, Möhn N, Streichert AL, Kretschmer JR, Grote-Levi L, Nay S, Seeliger T, Ratuszny D, Jendretzky KF, Tkachenko D, Jacobs R, Skripuletz T, Schwenkenbecher P. Rapid depletion of CD20 + B and T cells following ofatumumab therapy onset. Mult Scler Relat Disord 2024; 91:105886. [PMID: 39299183 DOI: 10.1016/j.msard.2024.105886] [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: 07/08/2024] [Revised: 08/24/2024] [Accepted: 09/10/2024] [Indexed: 09/22/2024]
Abstract
BACKGROUND The humanized monoclonal anti-CD20-antibody ofatumumab is highly effective in treating relapsing multiple sclerosis (MS). OBJECTIVE This study aimed to investigate the immanent effect of ofatumumab on the peripheral immune system, particularly targeting B and T cells expressing CD20. METHODS Blood samples of 53 MS patients receiving ofatumumab were collected prior to first application and after one week, two weeks and three months. Multicolor flow cytometry was used to phenotype peripheral blood mononuclear cells, and immunoglobulin (Ig) concentrations were measured by nephelometry. RESULTS Among CD20+ lymphocytes, 13 % co-expressed CD3 (identifying them as CD3+CD20+ T lymphocytes), with a noticeable shift in the CD4/CD8-ratio towards CD8+ T cells. One week after administering ofatumumab, a significant reduction of CD20+ lymphocytes with complete depletion of CD3+CD20+ T lymphocytes was observed, persisting during the investigation period. During the treatment, IgM levels showed a slight but significant decrease, whereas IgA and IgG levels remained stable. CONCLUSION Ofatumumab effectively depletes CD20+ lymphocytes already after the first administration. This depletion affects not only B cells, but also a small proportion of T cells (CD3+CD20+), affirming the hypothesis that the anti-inflammatory effects of CD20+ cell depletion might extend to the reduction of CD3+CD20+ T lymphocytes.
Collapse
Affiliation(s)
- Franz Felix Konen
- Department of Neurology, Hannover Medical School, Carl-Neuberg-Str. 1, 30625 Hannover, Germany
| | - Stefan Gingele
- Department of Neurology, Hannover Medical School, Carl-Neuberg-Str. 1, 30625 Hannover, Germany
| | - Martin W Hümmert
- Department of Neurology, Hannover Medical School, Carl-Neuberg-Str. 1, 30625 Hannover, Germany
| | - Nora Möhn
- Department of Neurology, Hannover Medical School, Carl-Neuberg-Str. 1, 30625 Hannover, Germany
| | - Anna Lena Streichert
- Department of Neurology, Hannover Medical School, Carl-Neuberg-Str. 1, 30625 Hannover, Germany
| | - Julian Reza Kretschmer
- Department of Neurology, Hannover Medical School, Carl-Neuberg-Str. 1, 30625 Hannover, Germany
| | - Lea Grote-Levi
- Department of Neurology, Hannover Medical School, Carl-Neuberg-Str. 1, 30625 Hannover, Germany
| | - Sandra Nay
- Department of Neurology, Hannover Medical School, Carl-Neuberg-Str. 1, 30625 Hannover, Germany
| | - Tabea Seeliger
- Department of Neurology, Hannover Medical School, Carl-Neuberg-Str. 1, 30625 Hannover, Germany
| | - Dominica Ratuszny
- Department of Neurology, Hannover Medical School, Carl-Neuberg-Str. 1, 30625 Hannover, Germany
| | | | - Daria Tkachenko
- Department of Neurology, Hannover Medical School, Carl-Neuberg-Str. 1, 30625 Hannover, Germany
| | - Roland Jacobs
- Department of Rheumatology and Clinical Immunology, Hannover Medical School, Carl-Neuberg-Str. 1, 30625 Hannover, Germany
| | - Thomas Skripuletz
- Department of Neurology, Hannover Medical School, Carl-Neuberg-Str. 1, 30625 Hannover, Germany
| | - Philipp Schwenkenbecher
- Department of Neurology, Hannover Medical School, Carl-Neuberg-Str. 1, 30625 Hannover, Germany.
| |
Collapse
|
3
|
Rodriguez-Mogeda C, van Gool MM, van der Mast R, Nijland R, Keasberry Z, van de Bovekamp L, van Delft MA, Picon C, Reynolds R, Killestein J, Teunissen CE, de Vries HE, van Egmond M, Witte ME. Intrathecal IgG and IgM synthesis correlates with neurodegeneration markers and corresponds to meningeal B cell presence in MS. Sci Rep 2024; 14:25540. [PMID: 39462090 PMCID: PMC11513002 DOI: 10.1038/s41598-024-76969-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Accepted: 10/18/2024] [Indexed: 10/28/2024] Open
Abstract
Intrathecal synthesis of immunoglobulins (Igs) is a key hallmark of multiple sclerosis (MS). B cells are known to accumulate in the leptomeninges of MS patients and associate with pathology in the underlying cortex and a more severe disease course. However, the role of locally produced antibodies in MS brain pathology is poorly understood. Here, we quantified the protein levels of IgA, IgM, IgG and albumin in serum and cerebrospinal fluid (CSF) samples of 80 MS patients and 28 neurological controls to calculate Ig indices. In addition, we quantified presence of meningeal IgA+, IgM+ and IgG+ B cells in post-mortem brain tissue of 20 MS patients and 6 controls using immunostainings. IgM and IgG, but not IgA, indices were increased in CSF of MS patients compared to controls, with no observed differences between MS disease types. Both IgM and IgG indices correlated significantly with neurofilament light (NfL) levels in CSF, but not with clinical or radiological parameters of disease. Similarly, IgG+ and IgM+ B cells were increased in MS meninges compared to controls, whereas IgA+ B cells were not. Neuronal loss did not differ between sections with low or high IgA+, IgM+ and IgG+ B cells, but was increased in sections with high numbers of all CD19+ meningeal B cells. Similarly, high presence of CD19+ meningeal B cells and IgG+ meningeal B cells associated with increased microglial density in the underlying cortex. Taken together, intrathecal synthesis of IgG and IgM is elevated in MS, which corresponds to an increased number of IgG+ and IgM+ B cells in MS meninges. The significant correlation between intrathecal IgG and IgM production and NfL levels, and increased microglial activation in cortical areas adjacent to meningeal infiltrates with high levels of IgG+ B cells indicate a role for intrathecal IgM- and IgG-producing B cells in neuroinflammatory and degenerative processes in MS.
Collapse
Affiliation(s)
- Carla Rodriguez-Mogeda
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC Location VUmc, Amsterdam, The Netherlands.
- Amsterdam Neuroscience, Amsterdam UMC, Amsterdam, The Netherlands.
- MS Center Amsterdam, Amsterdam UMC Location VUmc, Amsterdam, The Netherlands.
| | - Melissa Mj van Gool
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC Location VUmc, Amsterdam, The Netherlands
- Amsterdam institute for Infection and Immunity, Amsterdam UMC, Amsterdam, The Netherlands
| | - Richard van der Mast
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC Location VUmc, Amsterdam, The Netherlands
- Amsterdam institute for Infection and Immunity, Amsterdam UMC, Amsterdam, The Netherlands
| | - Rutger Nijland
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC Location VUmc, Amsterdam, The Netherlands
| | - Zoë Keasberry
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC Location VUmc, Amsterdam, The Netherlands
| | - Lisanne van de Bovekamp
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC Location VUmc, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Amsterdam UMC, Amsterdam, The Netherlands
- MS Center Amsterdam, Amsterdam UMC Location VUmc, Amsterdam, The Netherlands
| | - Myrthe Am van Delft
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC Location VUmc, Amsterdam, The Netherlands
- Amsterdam institute for Infection and Immunity, Amsterdam UMC, Amsterdam, The Netherlands
| | - Carmen Picon
- Division of Neuroscience, Department of Brain Sciences, Imperial College London, Hammersmith Hospital Campus, Burlington Danes Building, Du Cane Road, London, W12 0NN, UK
| | - Richard Reynolds
- Division of Neuroscience, Department of Brain Sciences, Imperial College London, Hammersmith Hospital Campus, Burlington Danes Building, Du Cane Road, London, W12 0NN, UK
| | - Joep Killestein
- Amsterdam Neuroscience, Amsterdam UMC, Amsterdam, The Netherlands
- MS Center Amsterdam, Amsterdam UMC Location VUmc, Amsterdam, The Netherlands
- Department of Neurology, Amsterdam UMC Location VUmc, Amsterdam, The Netherlands
| | - Charlotte E Teunissen
- Amsterdam Neuroscience, Amsterdam UMC, Amsterdam, The Netherlands
- MS Center Amsterdam, Amsterdam UMC Location VUmc, Amsterdam, The Netherlands
- Neurochemistry Lab, Department of Laboratory Medicine Chemistry, Amsterdam UMC Location VUmc, Amsterdam, The Netherlands
| | - Helga E de Vries
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC Location VUmc, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Amsterdam UMC, Amsterdam, The Netherlands
- MS Center Amsterdam, Amsterdam UMC Location VUmc, Amsterdam, The Netherlands
| | - Marjolein van Egmond
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC Location VUmc, Amsterdam, The Netherlands
- Amsterdam institute for Infection and Immunity, Amsterdam UMC, Amsterdam, The Netherlands
- Department of Surgery, Amsterdam UMC Location VUmc, Amsterdam, The Netherlands
| | - Maarten E Witte
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC Location VUmc, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Amsterdam UMC, Amsterdam, The Netherlands
- MS Center Amsterdam, Amsterdam UMC Location VUmc, Amsterdam, The Netherlands
- Amsterdam institute for Infection and Immunity, Amsterdam UMC, Amsterdam, The Netherlands
| |
Collapse
|
4
|
Schett G, Nagy G, Krönke G, Mielenz D. B-cell depletion in autoimmune diseases. Ann Rheum Dis 2024; 83:1409-1420. [PMID: 38777374 DOI: 10.1136/ard-2024-225727] [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: 02/26/2024] [Accepted: 04/23/2024] [Indexed: 05/25/2024]
Abstract
B cells have a pivotal function in the pathogenesis of autoimmune diseases, such as rheumatoid arthritis, multiple sclerosis and systemic lupus erythematosus. In autoimmune disease, B cells orchestrate antigen presentation, cytokine production and autoantibody production, the latter via their differentiation into antibody-secreting plasmablasts and plasma cells. This article addresses the current therapeutic strategies to deplete B cells in order to ameliorate or potentially even cure autoimmune disease. It addresses the main target antigens in the B-cell lineage that are used for therapeutic approaches. Furthermore, it summarises the current evidence for successful treatment of autoimmune disease with monoclonal antibodies targeting B cells and the limitations and challenges of these approaches. Finally, the concept of deep B-cell depletion and immunological reset by chimeric antigen receptor T cells is discussed, as well as the lessons from this approach for better understanding the role of B cells in autoimmune disease.
Collapse
Affiliation(s)
- Georg Schett
- Department of Internal Medicine 3-Rheumatology and Immunology, Friedrich-Alexander-Universität Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
- Deutsches Zentrum Immuntherapie, Friedrich-Alexander-Universität Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
| | - György Nagy
- Division of Rheumatology and Clinical Immunology, Department of Internal Medicine and Oncology, Semmelweis University, Budapest, Hungary, Budapest, Hungary
- Heart and Vascular Center, Semmelweis University, Budapest, Hungary
- Hospital of the Hospitaller Order of Saint John of God, Budapest, Hungary
| | - Gerhard Krönke
- Department of Internal Medicine 3-Rheumatology and Immunology, Friedrich-Alexander-Universität Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
- Deutsches Zentrum Immuntherapie, Friedrich-Alexander-Universität Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
- Department of Rheumatology, Charite, Berlin, Germany
| | - Dirk Mielenz
- Division of Molecular Immunology, Department of Internal Medicine 3, Friedrich-Alexander-Universität Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Bayern, Germany
| |
Collapse
|
5
|
Aharoni R, Milo R, Arnon R. Glatiramer Acetate for the Treatment of Multiple Sclerosis: From First-Generation Therapy to Elucidation of Immunomodulation and Repair. Pharmacol Rev 2024; 76:1133-1158. [PMID: 39406508 DOI: 10.1124/pharmrev.124.000927] [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: 01/31/2024] [Revised: 08/14/2024] [Accepted: 08/20/2024] [Indexed: 10/18/2024] Open
Abstract
Multiple sclerosis (MS) is a chronic inflammatory demyelinating and neurodegenerative disease of the central nervous system (CNS), with a putative autoimmune origin and complex pathogenesis. Modification of the natural history of MS by reducing relapses and slowing disability accumulation was first attained in the 1990 s with the development of the first-generation disease-modifying therapies. Glatiramer acetate (GA), a copolymer of L-alanine, L-lysine, L-glutamic acid, and L-tyrosine, was discovered due to its ability to suppress the animal model of MS, experimental autoimmune encephalomyelitis. Extensive clinical trials and long-term assessments established the efficacy and the safety of GA. Furthermore, studies of the therapeutic processes induced by GA in animal models and in MS patients indicate that GA affects various levels of the innate and the adaptive immune response, generating deviation from proinflammatory to anti-inflammatory pathways. This includes competition for binding to antigen presenting cells; driving dendritic cells, monocytes, and B-cells toward anti-inflammatory responses; and stimulating T-helper 2 and T-regulatory cells. The immune cells stimulated by GA reach the CNS and secrete in situ anti-inflammatory cytokines alleviating the pathological processes. Furthermore, cumulative findings reveal that in addition to its immunomodulatory effect, GA promotes neuroprotective repair processes such as neurotrophic factors secretion, remyelination, and neurogenesis. This review aims to provide an overview of MS pathology diagnosis and treatment as well as the diverse mechanism of action of GA. SIGNIFICANCE STATEMENT: Understanding the complex MS immune pathogenesis provided multiple targets for therapeutic intervention, resulting in a plethora of agents, with various mechanisms of action, efficacy, and safety profiles. However, promoting repair beyond the body's limited spontaneous extent is still a major challenge. GA, one of the first approved disease-modifying therapies, induces diverse immunomodulatory effects. Furthermore, GA treatment results in elevated neurotrophic factors secretion, remyelination and neurogenesis, supporting the notion that immunomodulatory treatment can support in situ a growth-promoting and repair environment.
Collapse
Affiliation(s)
- Rina Aharoni
- Department of Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot, Israel (Ri.A., Ru.A.); and Department of Neurology, Barzilai Medical Center, Ashkelon, Israel, and Faculty of Health Sciences, Ben-Gurion University of the Negev, Be'er-Sheva, Israel (R.M.)
| | - Ron Milo
- Department of Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot, Israel (Ri.A., Ru.A.); and Department of Neurology, Barzilai Medical Center, Ashkelon, Israel, and Faculty of Health Sciences, Ben-Gurion University of the Negev, Be'er-Sheva, Israel (R.M.)
| | - Ruth Arnon
- Department of Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot, Israel (Ri.A., Ru.A.); and Department of Neurology, Barzilai Medical Center, Ashkelon, Israel, and Faculty of Health Sciences, Ben-Gurion University of the Negev, Be'er-Sheva, Israel (R.M.)
| |
Collapse
|
6
|
Wu W, Gupta S, Sagan SA, Moseley CE, Zamvil SS, Pak JE. High-yield production of recombinant human myelin oligodendrocyte glycoprotein in SHuffle bacteria without a refolding step. J Immunol Methods 2024; 534:113764. [PMID: 39406335 DOI: 10.1016/j.jim.2024.113764] [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: 07/23/2024] [Revised: 10/07/2024] [Accepted: 10/11/2024] [Indexed: 10/22/2024]
Abstract
Experimental autoimmune encephalomyelitis (EAE) is a model for central nervous system (CNS) autoimmune demyelinating diseases such as multiple sclerosis (MS) and MOG antibody-associated disease (MOGAD). Immunization with the extracellular domain of recombinant human MOG (rhMOG), which contains pathogenic antibody and T cell epitopes, induces B cell-dependent EAE for studies in mice. However, these studies have been hampered by rhMOG availability due to its insolubility when overexpressed in bacterial cells, and the requirement for inefficient denaturation and refolding. Here, we describe a new protocol for the high-yield production of soluble rhMOG in SHuffle cells, a commercially available E. coli strain engineered to facilitate disulfide bond formation in the cytoplasm. SHuffle cells can produce a soluble fraction of rhMOG yielding >100 mg/L. Analytical size exclusion chromatography multi-angle light scattering (SEC-MALS) and differential scanning fluorimetry of purified rhMOG reveals a homogeneous monomer with a high melting temperature, indicative of a well-folded protein. An in vitro proliferation assay establishes that purified rhMOG can be processed and recognized by T cells expressing a T cell receptor (TCR) specific for the immunodominant MOG35-55 peptide epitope. Lastly, immunization of wild-type, but not B cell deficient, mice with rhMOG resulted in robust induction of EAE, indicating a B cell-dependent induction. Our SHuffle cell method greatly simplifies rhMOG production by combining the high yield and speed of bacterial cell expression with enhanced disulfide bond formation and folding, which will enable further investigation of B cell-dependent EAE and expand human research of MOG in CNS demyelinating diseases.
Collapse
Affiliation(s)
- Wesley Wu
- Chan Zuckerberg Biohub - San Francisco, San Francisco, CA 94158, USA
| | - Sasha Gupta
- Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, CA 94158, USA
| | - Sharon A Sagan
- Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, CA 94158, USA
| | - Carson E Moseley
- Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, CA 94158, USA
| | - Scott S Zamvil
- Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, CA 94158, USA; Program in Immunology, University of California, San Francisco 94143, USA
| | - John E Pak
- Chan Zuckerberg Biohub - San Francisco, San Francisco, CA 94158, USA.
| |
Collapse
|
7
|
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
|
8
|
Ding Q, Wu Y, Triglia ET, Gommerman JL, Subramanian A, Kuchroo VK, Rothstein DM. TIM-4 Identifies Effector B Cells Expressing a RORγt-Driven Proinflammatory Cytokine Module That Promotes Immune Responsiveness. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.09.22.558524. [PMID: 37790513 PMCID: PMC10542535 DOI: 10.1101/2023.09.22.558524] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/05/2023]
Abstract
B cells can express pro-inflammatory cytokines that promote a wide variety of immune responses. Here we show that B cells expressing the phosphatidylserine receptor TIM-4, preferentially express IL-17A, as well as IL-22, IL-6, IL-1β, and GM-CSF - a collection of cytokines reminiscent of pathogenic Th17 cells. Expression of this proinflammatory module requires IL-23R signaling and selective expression of RORγt and IL-17A by TIM-4+ B cells. TIM-4+ B cell-derived-IL-17A not only enhances the severity of experimental autoimmune encephalomyelitis (EAE) and promotes allograft rejection, but also acts in an autocrine manner to prevent their conversion into IL-10-expressing B cells with regulatory function. Thus, IL-17A acts as an inflammatory mediator and also enforces the proinflammatory activity of TIM-4+ B cells. Thus, TIM-4 serves as a broad marker for RORγt+ effector B cells (Beff) and allows further study of the signals regulating Beff differentiation and effector molecule expression.
Collapse
Affiliation(s)
- Qing Ding
- Thomas E. Starzl Transplantation Institute; University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Yufan Wu
- Klarman Cell Observatory, Broad Institute of MIT and Harvard; Cambridge, MA, USA
| | - Elena Torlai Triglia
- Klarman Cell Observatory, Broad Institute of MIT and Harvard; Cambridge, MA, USA
| | | | - Ayshwarya Subramanian
- Klarman Cell Observatory, Broad Institute of MIT and Harvard; Cambridge, MA, USA
- The Broad Institute of MIT and Harvard; Cambridge, MA, USA
| | - Vijay K. Kuchroo
- Klarman Cell Observatory, Broad Institute of MIT and Harvard; Cambridge, MA, USA
- The Broad Institute of MIT and Harvard; Cambridge, MA, USA
- Gene Lay Institute of Immunology and Inflammation, Harvard Medical School and Brigham and Women’s Hospital, Boston, MA, USA
| | - David M. Rothstein
- Thomas E. Starzl Transplantation Institute; University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
| |
Collapse
|
9
|
Waede M, Voss LF, Kingo C, Moeller JB, Elkjaer ML, Illes Z. Longitudinal analysis of peripheral immune cells in patients with multiple sclerosis treated with anti-CD20 therapy. Ann Clin Transl Neurol 2024; 11:2657-2672. [PMID: 39279291 PMCID: PMC11514931 DOI: 10.1002/acn3.52182] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 07/17/2024] [Accepted: 08/03/2024] [Indexed: 09/18/2024] Open
Abstract
OBJECTIVE Anti-CD20 therapy is a highly effective treatment for multiple sclerosis (MS). In this study, we investigated MS-related changes in peripheral blood mononuclear cell (PBMC) subsets compared to healthy controls and longitudinal changes related to the treatment. METHODS Multicolor spectral flow cytometry analysis was performed on 78 samples to characterize disease- and treatment-related PBMC clusters. Blood samples from MS patients were collected at baseline and up to 8 months post-treatment, with three collection points after treatment initiation. Unsupervised clustering tools and manual gating were applied to identify subclusters of interest and quantify changes. RESULTS B cells were depleted from the periphery after anti-CD20 treatment as expected, and we observed an isolated acute, transitory drop in the proportion of natural killer (NK) and NKT cells among the main populations of PBMC (P = 0.03, P = 0.004). Major affected PBMC subpopulations were cytotoxic immune cells (NK, NKT, and CD8+ T cells), and we observed a higher proportion of cytotoxic cells with reduced brain-homing ability and a higher regulatory function as a long-term anti-CD20-related effect. Additionally, anti-CD20 therapy altered distributions of memory CD8+ T cells and reduced exhaustion markers in both CD4+ and CD8+ T cells. INTERPRETATION The findings of this study elucidate phenotypic clusters of NK and CD8+ T cells, which have previously been underexplored in the context of anti-CD20 therapy. Phenotypic modifications towards a more regulatory and controlled phenotype suggest that these subpopulations may play a critical and previously unrecognized role in mediating the therapeutic efficacy of anti-CD20 treatments.
Collapse
Affiliation(s)
- Mie Waede
- Department of NeurologyOdense University HospitalOdenseDenmark
- Department of Clinical ResearchUniversity of Southern DenmarkOdenseDenmark
- Department of Molecular MedicineUniversity of Southern DenmarkOdenseDenmark
| | - Lasse F. Voss
- Section for Experimental and Translational Immunology, Department of Health TechnologyTechnical University of DenmarkKongens LyngbyDenmark
| | - Christina Kingo
- Department of NeurologyOdense University HospitalOdenseDenmark
- Department of Clinical ResearchUniversity of Southern DenmarkOdenseDenmark
- Department of Molecular MedicineUniversity of Southern DenmarkOdenseDenmark
| | - Jesper B. Moeller
- Department of Molecular MedicineUniversity of Southern DenmarkOdenseDenmark
- Danish Institute for Advanced Study, University of Southern DenmarkOdenseDenmark
| | - Maria L. Elkjaer
- Department of NeurologyOdense University HospitalOdenseDenmark
- Department of Clinical ResearchUniversity of Southern DenmarkOdenseDenmark
- Department of Molecular MedicineUniversity of Southern DenmarkOdenseDenmark
- Institute for Computational Systems Biology, University of HamburgHamburgGermany
| | - Zsolt Illes
- Department of NeurologyOdense University HospitalOdenseDenmark
- Department of Clinical ResearchUniversity of Southern DenmarkOdenseDenmark
- Department of Molecular MedicineUniversity of Southern DenmarkOdenseDenmark
- BRIDGE – Brain Research Interdisciplinary Guided ExcellenceUniversity of Southern DenmarkOdenseDenmark
| |
Collapse
|
10
|
Wu HC, Gombolay GY, Yang JH, Graves JS, Christy A, Xiang XM. B-cell Depletion Therapy in Pediatric Neuroinflammatory Disease. Curr Neurol Neurosci Rep 2024; 24:479-494. [PMID: 39259430 DOI: 10.1007/s11910-024-01366-7] [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] [Accepted: 07/15/2024] [Indexed: 09/13/2024]
Abstract
PURPOSE OF REVIEW B-cell depletion therapy, including anti-CD20 and anti-CD19 therapies, is increasingly used for a variety of autoimmune and conditions, including those affecting the central nervous system. However, B-cell depletion therapy use can be complicated by adverse effects associated with administration and immunosuppression. This review aims to summarize the application of anti-CD20 and anti-CD19 therapies for the pediatric neurologist and neuroimmunologist. RECENT FINDINGS Most existing literature come from clinical trials with adult patients, although more recent studies are now capturing the effects of these therapies in children. The most common side effects include infusion related reactions and increased infection risk from immunosuppression. Several strategies can mitigate infusion related reactions. Increased infections due to persistent hypogammaglobulinemia can benefit from replacement immunoglobulin. B-cell depletion therapies can be safe and effective in pediatric patients. Anticipation and mitigation of common adverse effects through primary prevention strategies, close monitoring, and appropriate symptomatic management can improve safety and tolerability.
Collapse
Affiliation(s)
- Helen C Wu
- Department of Neurology, Oregon Health & Science University, Portland, OR, USA.
| | - Grace Y Gombolay
- Department of Pediatrics, Division of Pediatric Neurology, Children's Healthcare of Atlanta, Emory University School of Medicine, Atlanta, GA, USA
| | - Jennifer H Yang
- Department of Neurosciences, University of California San Diego, Rady Children's Hospital San Diego, San Diego, CA, USA
| | - Jennifer S Graves
- Department of Neurosciences, University of California San Diego, Rady Children's Hospital San Diego, San Diego, CA, USA
| | - Alison Christy
- Pediatric Neurology, Providence Health & Services, Portland, OR, USA
| | - Xinran M Xiang
- Department of Neurology, Oregon Health & Science University, Portland, OR, USA
- Pediatric Neurology, Oregon Health & Science University, Portland, OR, USA
| |
Collapse
|
11
|
Konitsioti AM, Prüss H, Laurent S, Fink GR, Heesen C, Warnke C. Chimeric antigen receptor T-cell therapy for autoimmune diseases of the central nervous system: a systematic literature review. J Neurol 2024; 271:6526-6542. [PMID: 39276207 PMCID: PMC11446985 DOI: 10.1007/s00415-024-12642-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2024] [Revised: 08/13/2024] [Accepted: 08/16/2024] [Indexed: 09/16/2024]
Abstract
IMPORTANCE B-cell-targeting monoclonal antibodies have demonstrated safety and efficacy in multiple sclerosis or anti-aquaporin-4 IgG positive neuromyelitis optica spectrum disorder. However, these therapies do not facilitate drug-free remission, which may become possible with cell-based therapies, including chimeric antigen receptor (CAR) T cells. CAR T-cell therapy holds promise for addressing other antibody-mediated CNS disorders, e.g., MOG-associated disease or autoimmune encephalitis. OBJECTIVE To provide an overview of the current clinical knowledge on CAR T-cell therapy in central nervous system autoimmunity. EVIDENCE REVIEW We searched PubMed, Embase, Google Scholar, PsycINFO, and clinicaltrials.gov using the terms 'CAR T cell' and 'multiple sclerosis/MS' or 'neuromyelitis optica/spectrum diseases/NMOSD' or 'MOG-associated disease/MOGAD 'or' autoimmune encephalitis' or 'neuroimmunology'. FINDINGS An ongoing phase I clinical trial has indicated the safety and benefits of anti-BCMA CAR T cells in 12 patients with AQP4-IgG seropositive neuromyelitis optica spectrum disorder. Case reports involving two individuals with progressive multiple sclerosis and one patient with stiff-person syndrome demonstrated a manageable safety profile following treatment with anti-CD19 CAR T cells. Recruitment has commenced for two larger studies in MS, and a phase I open-label basket study is underway to evaluate BCMA-directed CAR T cells in various antibody-associated inflammatory diseases, including MOG-associated disease. Preclinical research on NMDA receptor antibody autoimmune encephalitis treated with chimeric autoantibody receptor T cells generated promising data. CONCLUSIONS AND RELEVANCE There is minimal evidence of the benefits of CAR T-cell therapy in individuals with central nervous system-directed autoimmunity. Nevertheless, multicenter controlled clinical trials with a manageable safety profile appear feasible and are warranted due to very promising case experiences.
Collapse
Affiliation(s)
- Agni M Konitsioti
- Department of Neurology, University of Cologne, Kerpener Str. 62, 50937, Cologne, Germany.
- Faculty of Medicine, University Hospital Cologne, Cologne, Germany.
| | - Harald Prüss
- Department of Neurology and Experimental Neurology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Sarah Laurent
- Department of Neurology, University of Cologne, Kerpener Str. 62, 50937, Cologne, Germany
- Faculty of Medicine, University Hospital Cologne, Cologne, Germany
| | - Gereon R Fink
- Department of Neurology, University of Cologne, Kerpener Str. 62, 50937, Cologne, Germany
- Cognitive Neuroscience, Institute of Neuroscience and Medicine (INM3), Research Center Jülich, Jülich, Germany
- Faculty of Medicine, University Hospital Cologne, Cologne, Germany
| | - Christoph Heesen
- Department of Neurology, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
| | - Clemens Warnke
- Department of Neurology, University of Cologne, Kerpener Str. 62, 50937, Cologne, Germany.
- Faculty of Medicine, University Hospital Cologne, Cologne, Germany.
| |
Collapse
|
12
|
Intarakhao P, Laipasu T, Jitprapaikulsan J, Apiraksattayakul N, Kosiyakul P, Siritho S, Prayoonwiwat N, Ongphichetmetha T. Rituximab in secondary progressive multiple sclerosis: a meta-analysis. Ann Clin Transl Neurol 2024; 11:2707-2718. [PMID: 39186371 PMCID: PMC11514939 DOI: 10.1002/acn3.52186] [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: 05/14/2024] [Revised: 07/29/2024] [Accepted: 08/03/2024] [Indexed: 08/27/2024] Open
Abstract
OBJECTIVE To evaluate the efficacy of rituximab (RTX) in stabilizing disability progression in secondary progressive multiple sclerosis (SPMS). METHODS A systematic review was conducted, encompassing studies from inception to April 2023, utilizing the MEDLINE and EMBASE databases. Inclusion criteria comprised studies with a minimum of 3 SPMS patients receiving intravenous RTX in at least one infusion, with a follow-up duration of at least 6 months. Primary outcome measures included changes in Expanded Disability Status Scale (EDSS) scores. Mean differences in pre- and post-RTX EDSS scores were analyzed using a random-effects model. Meta-regression examined age at RTX initiation, pre-RTX EDSS scores, disease duration, and outcome reported time as variables. Secondary outcomes assessed changes in the annualized relapse rate (ARR). RESULTS Thirteen studies, involving 604 SPMS patients, met the inclusion criteria. Following a mean follow-up of 2 years, the mean difference in EDSS scores (ΔEDSS = EDSSpre-RTX - EDSSpost-RTX) was -0.21 (95% CI -0.51 to 0.08, p = 0.16), indicating no significant variation. Multivariable meta-regression identified significant associations between EDSS score mean difference and pre-RTX EDSS scores, disease duration at RTX initiation, and outcome reported time. However, age at RTX initiation showed no significant association. Pre- and post-RTX ARR data were available for 245 out of 604 SPMS patients across seven studies, revealing a mean difference in ARR (ΔARR = ARRpre-RTX - ARRpost-RTX) of 0.74 (95% CI 0.19-1.29, p = 0.008). INTERPRETATION RTX demonstrates efficacy in reducing relapse frequency and exhibits potential in stabilizing disability progression over a 2-year follow-up, particularly among individuals with shorter disease duration.
Collapse
Affiliation(s)
- Pasin Intarakhao
- Faculty of Medicine Siriraj HospitalMahidol UniversityBangkokThailand
| | - Taksaporn Laipasu
- Faculty of Medicine Siriraj HospitalMahidol UniversityBangkokThailand
| | - Jiraporn Jitprapaikulsan
- Siriraj Neuroimmunology Center, Division of Neurology, Department of Medicine, Faculty of Medicine Siriraj HospitalMahidol UniversityBangkokThailand
- Division of Neurology, Department of Medicine, Faculty of Medicine Siriraj HospitalMahidol UniversityBangkokThailand
| | - Natnasak Apiraksattayakul
- Siriraj Neuroimmunology Center, Division of Neurology, Department of Medicine, Faculty of Medicine Siriraj HospitalMahidol UniversityBangkokThailand
| | - Punchika Kosiyakul
- Siriraj Neuroimmunology Center, Division of Neurology, Department of Medicine, Faculty of Medicine Siriraj HospitalMahidol UniversityBangkokThailand
| | - Sasitorn Siritho
- Siriraj Neuroimmunology Center, Division of Neurology, Department of Medicine, Faculty of Medicine Siriraj HospitalMahidol UniversityBangkokThailand
- Bumrungrad International HospitalBangkokThailand
| | - Naraporn Prayoonwiwat
- Siriraj Neuroimmunology Center, Division of Neurology, Department of Medicine, Faculty of Medicine Siriraj HospitalMahidol UniversityBangkokThailand
- Division of Neurology, Department of Medicine, Faculty of Medicine Siriraj HospitalMahidol UniversityBangkokThailand
| | - Tatchaporn Ongphichetmetha
- Siriraj Neuroimmunology Center, Division of Neurology, Department of Medicine, Faculty of Medicine Siriraj HospitalMahidol UniversityBangkokThailand
- Division of Neurology, Department of Medicine, Faculty of Medicine Siriraj HospitalMahidol UniversityBangkokThailand
- Division of Clinical Epidemiology, Department of Research and Development, Faculty of Medicine Siriraj HospitalMahidol UniversityBangkokThailand
| |
Collapse
|
13
|
Lacagnina MJ, Willcox KF, Boukelmoune N, Bavencoffe A, Sankaranarayanan I, Barratt DT, Zuberi YA, Dayani D, Chavez MV, Lu JT, Farinotti AB, Shiers S, Barry AM, Mwirigi JM, Tavares-Ferreira D, Funk GA, Cervantes AM, Svensson CI, Walters ET, Hutchinson MR, Heijnen CJ, Price TJ, Fiore NT, Grace PM. B cells drive neuropathic pain-related behaviors in mice through IgG-Fc gamma receptor signaling. Sci Transl Med 2024; 16:eadj1277. [PMID: 39321269 PMCID: PMC11479571 DOI: 10.1126/scitranslmed.adj1277] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Revised: 03/06/2024] [Accepted: 09/03/2024] [Indexed: 09/27/2024]
Abstract
Neuroimmune interactions are essential for the development of neuropathic pain, yet the contributions of distinct immune cell populations have not been fully unraveled. Here, we demonstrate the critical role of B cells in promoting mechanical hypersensitivity (allodynia) after peripheral nerve injury in male and female mice. Depletion of B cells with a single injection of anti-CD20 monoclonal antibody at the time of injury prevented the development of allodynia. B cell-deficient (muMT) mice were similarly spared from allodynia. Nerve injury was associated with increased immunoglobulin G (IgG) accumulation in ipsilateral lumbar dorsal root ganglia (DRGs) and dorsal spinal cords. IgG was colocalized with sensory neurons and macrophages in DRGs and microglia in spinal cords. IgG also accumulated in DRG samples from human donors with chronic pain, colocalizing with a marker for macrophages and satellite glia. RNA sequencing revealed a B cell population in naive mouse and human DRGs. A B cell transcriptional signature was enriched in DRGs from human donors with neuropathic pain. Passive transfer of IgG from injured mice induced allodynia in injured muMT recipient mice. The pronociceptive effects of IgG are likely mediated through immune complexes interacting with Fc gamma receptors (FcγRs) expressed by sensory neurons, microglia, and macrophages, given that both mechanical allodynia and hyperexcitability of dissociated DRG neurons were abolished in nerve-injured FcγR-deficient mice. Consistently, the pronociceptive effects of IgG passive transfer were lost in FcγR-deficient mice. These data reveal that a B cell-IgG-FcγR axis is required for the development of neuropathic pain in mice.
Collapse
Affiliation(s)
- Michael J. Lacagnina
- Laboratories of Neuroimmunology, Department of Symptom Research, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Kendal F. Willcox
- Laboratories of Neuroimmunology, Department of Symptom Research, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Nabila Boukelmoune
- Laboratories of Neuroimmunology, Department of Symptom Research, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Alexis Bavencoffe
- Department of Integrative Biology and Pharmacology, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX 77225, USA
| | - Ishwarya Sankaranarayanan
- Department of Neuroscience and Center for Advanced Pain Studies, University of Texas at Dallas, Richardson, TX 75080, USA
| | - Daniel T. Barratt
- School of Biomedicine, University of Adelaide, Adelaide, SA 5005, Australia
- Davies Livestock Research Centre, University of Adelaide, Roseworthy, SA 5371, Australia
| | - Younus A. Zuberi
- Laboratories of Neuroimmunology, Department of Symptom Research, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Dorsa Dayani
- Laboratories of Neuroimmunology, Department of Symptom Research, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Melissa V. Chavez
- Laboratories of Neuroimmunology, Department of Symptom Research, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Jonathan T. Lu
- Laboratories of Neuroimmunology, Department of Symptom Research, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | | | - Stephanie Shiers
- Department of Neuroscience and Center for Advanced Pain Studies, University of Texas at Dallas, Richardson, TX 75080, USA
| | - Allison M. Barry
- Department of Neuroscience and Center for Advanced Pain Studies, University of Texas at Dallas, Richardson, TX 75080, USA
| | - Juliet M. Mwirigi
- Department of Neuroscience and Center for Advanced Pain Studies, University of Texas at Dallas, Richardson, TX 75080, USA
| | - Diana Tavares-Ferreira
- Department of Neuroscience and Center for Advanced Pain Studies, University of Texas at Dallas, Richardson, TX 75080, USA
| | | | | | - Camilla I. Svensson
- Department of Physiology and Pharmacology, Karolinska Institutet, 17177 Stockholm, Sweden
| | - Edgar T. Walters
- Department of Integrative Biology and Pharmacology, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX 77225, USA
| | - Mark R. Hutchinson
- School of Biomedicine, University of Adelaide, Adelaide, SA 5005, Australia
- Davies Livestock Research Centre, University of Adelaide, Roseworthy, SA 5371, Australia
- Australian Research Council Centre of Excellence for Nanoscale BioPhotonics, Adelaide, SA 5005, Australia
| | - Cobi J. Heijnen
- Department of Psychological Sciences, Rice University, Houston, TX 77005, USA
| | - Theodore J. Price
- Department of Neuroscience and Center for Advanced Pain Studies, University of Texas at Dallas, Richardson, TX 75080, USA
| | - Nathan T. Fiore
- Laboratories of Neuroimmunology, Department of Symptom Research, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Peter M. Grace
- Laboratories of Neuroimmunology, Department of Symptom Research, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| |
Collapse
|
14
|
Gallwitz M, Lindqvist I, Mulder J, Rasmusson AJ, Larsson A, Husén E, Borin J, van der Spek PJ, Sabbagh N, Widgren A, Bergquist J, Cervenka S, Burman J, Cunningham JL. Three cases with chronic obsessive compulsive disorder report gains in wellbeing and function following rituximab treatment. Mol Psychiatry 2024:10.1038/s41380-024-02750-y. [PMID: 39304742 DOI: 10.1038/s41380-024-02750-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Revised: 09/02/2024] [Accepted: 09/05/2024] [Indexed: 09/22/2024]
Abstract
Immunological aetiology is supported for a subgroup with obsessive compulsive disorder (OCD) and conceptualized as autoimmune OCD. The longitudinal clinical course is detailed for three severely ill cases with OCD and indications of immunological involvement with off-label rituximab treatment every six months. All cases showed clear and sustained gains regarding symptom burden and function for over 2.5 years. Brief Psychiatric Rating Scale and Yale-Brown Obsessive-Compulsive Inventory Scale scores decreased 67-100% and 44-92%, respectively. These complex cases, prior to rituximab, had very low functioning and disease duration has been eight, nine and 16 years respectively. All three patients had been unsuccessfully treated with at least two antidepressants or anxiolytics, one neuroleptic and cognitive behavioural therapy. Clinical phenotypes and findings were suggestive of possible autoimmune OCD. Indirect immunohistochemistry detected cerebral spinal fluid (CSF) antibodies in all three cases including a novel anti-neuronal staining pattern against mouse thalamic cells. Exploratory analyses of CSF markers and proteomics identified elevated levels of sCD27 and markers indicative of complement pathway activation when compared to CSF from healthy controls. Multidisciplinary collaboration, advanced clinical investigations and rituximab treatment are feasible in a psychiatric setting. The case histories provide a proof of principle for the newly proposed criteria for autoimmune OCD. The findings suggest that clinical red flags and biological measures may predict rituximab response in chronic treatment-resistant OCD. The report provides orientation that may inform the hypotheses and design of future treatment trials.
Collapse
Affiliation(s)
- Maike Gallwitz
- Department of Medical Sciences, Psychiatry, Uppsala University, Uppsala, Sweden
| | - Isa Lindqvist
- Department of Medical Sciences, Psychiatry, Uppsala University, Uppsala, Sweden
| | - Jan Mulder
- Department of Neuroscience, Karolinska Institute, Stockholm, Sweden
| | - Annica J Rasmusson
- Department of Medical Sciences, Psychiatry, Uppsala University, Uppsala, Sweden
| | - Anders Larsson
- Department of Medical Sciences, Clinical Chemistry, Uppsala University, Uppsala, Sweden
| | - Evelina Husén
- Department of Neuroscience, Karolinska Institute, Stockholm, Sweden
| | - Jesper Borin
- Department of Medical Sciences, Psychiatry, Uppsala University, Uppsala, Sweden
| | - Peter J van der Spek
- Department of Pathology and Clinical Bioinformatics, Erasmus MC, Rotterdam, The Netherlands
| | - Nour Sabbagh
- Department of Neuroscience, Karolinska Institute, Stockholm, Sweden
| | - Anna Widgren
- Department of Chemistry - BMC, Analytical Chemistry and Neurochemistry, Uppsala University, Uppsala, Sweden
| | - Jonas Bergquist
- Department of Chemistry - BMC, Analytical Chemistry and Neurochemistry, Uppsala University, Uppsala, Sweden
| | - Simon Cervenka
- Department of Medical Sciences, Psychiatry, Uppsala University, Uppsala, Sweden
- Centre for Psychiatry Research, Department of Clinical Neuroscience, Karolinska Institute and Stockholm Health Care Services, Region Stockholm, Stockholm, Sweden
| | - Joachim Burman
- Department of Medical Sciences, Translational Neurology, Uppsala University, Uppsala, Sweden
| | - Janet L Cunningham
- Department of Medical Sciences, Psychiatry, Uppsala University, Uppsala, Sweden.
| |
Collapse
|
15
|
Deffenbaugh JL, Jung KJ, Murphy SP, Liu Y, Rau CN, Petersen-Cherubini CL, Collins PL, Chung D, Lovett-Racke AE. Novel model of multiple sclerosis induced by EBV-like virus generates a unique B cell population. J Neuroimmunol 2024; 394:578408. [PMID: 39098102 DOI: 10.1016/j.jneuroim.2024.578408] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Revised: 06/21/2024] [Accepted: 07/21/2024] [Indexed: 08/06/2024]
Abstract
Epstein-Barr virus (EBV) is deemed a necessary, yet insufficient factor in the development of multiple sclerosis (MS). In this study, myelin basic protein-specific transgenic T cell receptor mice were infected with murid gammaherpesvirus 68 virus (MHV68), an EBV-like virus that infects mice, resulting in the onset neurological deficits at a significantly higher frequency than influenza or mock-infected mice. MHV68 infected mice exhibited signs including optic neuritis and ataxia which are frequently observed in MS patients but not in experimental autoimmune encephalomyelitis mice. MHV68-infected mice exhibited increased focal immune cell infiltration in the central nervous system. Single cell RNA sequencing identified the emergence of a population of B cells that express genes associated with antigen presentation and costimulation, indicating that gammaherpesvirus infection drives a distinct, pro-inflammatory transcriptional program in B cells that may promote autoreactive T cell responses in MS.
Collapse
Affiliation(s)
- Joshua L Deffenbaugh
- Department of Microbial Infection & Immunity, The Ohio State University Wexner Medical Center, USA.
| | - Kyeong-Joo Jung
- Department of Computer Science and Engineering, The Ohio State University, USA.
| | - Shawn P Murphy
- Department of Microbial Infection & Immunity, The Ohio State University Wexner Medical Center, USA.
| | - Yue Liu
- Department of Microbial Infection & Immunity, The Ohio State University Wexner Medical Center, USA.
| | - Christina N Rau
- Department of Microbial Infection & Immunity, The Ohio State University Wexner Medical Center, USA.
| | - Cora L Petersen-Cherubini
- Department of Microbial Infection & Immunity, The Ohio State University Wexner Medical Center, USA; Neuroscience Graduate Program, The Ohio State University, USA
| | - Patrick L Collins
- Department of Microbial Infection & Immunity, The Ohio State University Wexner Medical Center, USA.
| | - Dongjun Chung
- Department of Biomedical Informatics, The Ohio State University Wexner Medical Center, USA; Pelotonia Institute for Immuno-Oncology, The Ohio State University Wexner Medical Center, USA.
| | - Amy E Lovett-Racke
- Department of Microbial Infection & Immunity, The Ohio State University Wexner Medical Center, USA; Department of Neuroscience, The Ohio State University Wexner Medical Center, USA.
| |
Collapse
|
16
|
Raja P, Dhamija K, Samim MM, Saini J, Manjappaiah MG, Kandavel T, M N. A Real-World Experience of Rituximab: A Panacea in Therapy of Multiple Sclerosis in Low- and Middle-Income Setting. Clin Neuropharmacol 2024:00002826-990000000-00096. [PMID: 39258542 DOI: 10.1097/wnf.0000000000000612] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/12/2024]
Abstract
BACKGROUND Anti-CD20 monoclonal antibodies have received increasing attention in the past few years in the treatment of multiple sclerosis (MS). OBJECTIVES This study describes the (i) efficacy and safety of rituximab in people living with MS and (ii) assesses clinical and imaging outcomes following rituximab in MS. METHOD This is a chart review from the MS registry maintained at the institute from a University Hospital in South India. RESULT Eighty-three (M:F, 26:57) people living with MS received rituximab as immunomodulation between 2007 and 2022 with a median follow-up duration of 18 months. Fifty-nine (71%) were classified as relapsing-remitting MS, 16 (19%) were secondary progressive MS, and 8 (10%) were primary progressive MS. Seventy-two (87%) MS patients did not experience any relapse after receiving rituximab. In relapsing-remitting MS patients, the mean annualized recurrence rate dropped from 1.24 ± 1.19 to 0.16 ± 0.37. Infusion-related reaction occurred in 5 (6% of adverse events), urinary infections in 7 (8.4%), systemic infections in 3 (3%), Pneumocystis carinii pneumonia occurred in 1 (1%), and herpes zoster infection in 1 (1%) patient. Mortality was observed in 3 (3.5%) patients. While being on rituximab, 18 (22%) patients had mild COVID-19 illness and they all made complete recovery without any sequalae. CONCLUSIONS Rituximab is a safe, well-tolerated, easily accessible, inexpensive, and effective therapeutic option for people with MS. Rituximab showed both clinical and radiological improvement after a median follow-up of 1.5 years. None of our patients showed any severe COVID infection nor side effects after receiving COVID vaccination.
Collapse
Affiliation(s)
- Pritam Raja
- Department of Neurology, National Institute of Mental Health & Neurosciences, Bangalore, India
| | - Kamakshi Dhamija
- Department of Neurology, National Institute of Mental Health & Neurosciences, Bangalore, India
| | - M M Samim
- Department of Neurology, National Institute of Mental Health & Neurosciences, Bangalore, India
| | - Jitender Saini
- Department of Neuroimaging & Interventional Neuroradiology (NIIR), National Institute of Mental Health & Neurosciences, Bangalore, India, and
| | | | - Thennarasu Kandavel
- Department of Biostatistics National Institute of Mental Health & Neurosciences, Bangalore, India
| | - Netravathi M
- Department of Neurology, National Institute of Mental Health & Neurosciences, Bangalore, India
| |
Collapse
|
17
|
Liu ITT, Kesselheim AS. Drug Repurposing Via the Best Pharmaceuticals for Children Act. JAMA Pediatr 2024; 178:855-856. [PMID: 39073790 DOI: 10.1001/jamapediatrics.2024.2287] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 07/30/2024]
Abstract
This Viewpoint discusses the benefits and impediments of drug repurposing in the US and how the Best Pharmaceuticals for Children Act may serve as a successful legislative model to stimulate greater drug repurposing.
Collapse
Affiliation(s)
- Ian T T Liu
- Program on Regulation, Therapeutics, and Law, Division of Pharmacoepidemiology and Pharmacoeconomics, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Aaron S Kesselheim
- Program on Regulation, Therapeutics, and Law, Division of Pharmacoepidemiology and Pharmacoeconomics, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
18
|
Faust MA, Gibbs L, Oviedo JM, Cornwall DH, Fairfax KC, Zhou Z, Lamb TJ, Evavold BD. B Cells Influence Encephalitogenic T Cell Frequency to Myelin Oligodendrocyte Glycoprotein (MOG)38-49 during Full-length MOG Protein-Induced Demyelinating Disease. Immunohorizons 2024; 8:729-739. [PMID: 39330967 PMCID: PMC11447661 DOI: 10.4049/immunohorizons.2400069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Accepted: 08/27/2024] [Indexed: 09/28/2024] Open
Abstract
Although T cells are encephalitogenic during demyelinating disease, B cell-depleting therapies are a successful treatment for patients with multiple sclerosis. Murine models of demyelinating disease utilizing myelin epitopes, such as myelin oligodendrocyte glycoprotein (MOG)35-55, induce a robust CD4 T cell response but mitigate the contribution of pathological B cells. This limits their efficacy for investigating how B cell depletion affects T cells. Furthermore, induction of experimental autoimmune encephalomyelitis with a single CD4 T cell epitope does not reflect the breadth of epitopes observed in the clinic. To better model the adaptive immune response, mice were immunized with the full-length MOG protein or the MOG1-125 extracellular domain (ECD) and compared with MOG35-55. Mature MOG-reactive B cells were generated only by full-length MOG or ECD. The CNS-localized T cell response induced by full-length MOG is characterized by a reduction in frequency and the percentage of low-affinity T cells with reactivity toward the core epitope of MOG35-55. B cell depletion with anti-CD20 before full-length MOG-induced, but not ECD-induced, demyelinating disease restored T cell reactivity toward the immunodominant epitope of MOG35-55, suggesting the B cell-mediated control of encephalitogenic epitopes. Ultimately, this study reveals that anti-CD20 treatment can influence T cell epitopes found in the CNS during demyelinating disease.
Collapse
Affiliation(s)
- Michael A. Faust
- Division of Microbiology and Immunology, Department of Pathology, University of Utah, Salt Lake City, UT
| | - Lisa Gibbs
- Division of Microbiology and Immunology, Department of Pathology, University of Utah, Salt Lake City, UT
| | - Juan M. Oviedo
- Division of Microbiology and Immunology, Department of Pathology, University of Utah, Salt Lake City, UT
| | - Douglas H. Cornwall
- Division of Microbiology and Immunology, Department of Pathology, University of Utah, Salt Lake City, UT
| | - Keke C. Fairfax
- Division of Microbiology and Immunology, Department of Pathology, University of Utah, Salt Lake City, UT
| | - Zemin Zhou
- Division of Microbiology and Immunology, Department of Pathology, University of Utah, Salt Lake City, UT
| | - Tracey J. Lamb
- Division of Microbiology and Immunology, Department of Pathology, University of Utah, Salt Lake City, UT
| | - Brian D. Evavold
- Division of Microbiology and Immunology, Department of Pathology, University of Utah, Salt Lake City, UT
| |
Collapse
|
19
|
Karl AS, Klimas R, Katsimpoura M, Sgodzai M, Theile-Ochel S, Poser PL, Gisevius B, Faissner S, Salmen A, Nastos I, Gold R, Motte J. Quality of life and tolerability of B-cell directed therapy of multiple sclerosis with ofatumumab in a patient-centered real-world observational study. J Neurol 2024; 271:6080-6088. [PMID: 39039273 PMCID: PMC11377633 DOI: 10.1007/s00415-024-12581-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Revised: 07/10/2024] [Accepted: 07/12/2024] [Indexed: 07/24/2024]
Abstract
INTRODUCTION Ofatumumab (Kesimpta®) is a subcutaneous CD20-targeting antibody approved in Germany in 2021 for the treatment of relapsing multiple sclerosis (RMS). After careful instruction, patients can administer the treatment themselves. We previously reported data of 101 patients (Klimas et al. in Nervenarzt 94:923-933, 2023). The objective of this longitudinal study is to explore the tolerability and acceptability of ofatumumab from a patient perspective over a follow up period of 6 months. METHODS In this prospective observational real-world study, we report follow up data of 81 patients. We evaluated sociodemographic data, disease duration, duration and side effects of ofatumumab use, expanded disability status scale (EDSS), Beck Depression Inventory II (BDI-II), Short-Form 36 (SF-36), Fatigue Scale of Motor and Cognitive Functions (FSMC), and modified Multiple Sclerosis Functional Composite Test (MSFC). In addition, we asked for subjective treatment outcomes, such as impact on quality of life, walking distance, concentration, mood, medication adherence, fatigue and the subjective course of MS on a numerical rating scale (1 = very negative; 5 = very positive). Furthermore, treatment discontinuations were recorded. RESULTS The average duration of ofatumumab treatment was 10 months. In comparison to previous published data of our cohort, patients reported a significant increase in headache (10% up to 26%, p = 0.004) and limb pain (5% up to 26%, p < 0.001) as persistent side effects after the injections. More patients reported a very positive effect (p < 0.0001) on quality of life. 4 confirmed relapses occurred but no EDSS worsening, and no treatment discontinuations were documented during the observation period. DISCUSSION As previously described, our prospective study indicates that patients have a good tolerability of ofatumumab, precisely because of the mild and few side effects at the first administration. However, the longer the observation period, the more headaches and limb pain occurred after the injections. Despite this, patients' subjective quality of life improved. There were no discontinuations during the follow-up period, with the limitation of a high loss to follow-up.
Collapse
Affiliation(s)
- Anna-Sophia Karl
- Clinic for Neurology, St. Josef Hospital, Ruhr University Bochum, Gudrunstraße 56, 44791, Bochum, Germany
| | - Rafael Klimas
- Clinic for Neurology, St. Josef Hospital, Ruhr University Bochum, Gudrunstraße 56, 44791, Bochum, Germany.
| | - Melina Katsimpoura
- Clinic for Neurology, St. Josef Hospital, Ruhr University Bochum, Gudrunstraße 56, 44791, Bochum, Germany
| | - Melissa Sgodzai
- Clinic for Neurology, St. Josef Hospital, Ruhr University Bochum, Gudrunstraße 56, 44791, Bochum, Germany
| | - Simon Theile-Ochel
- Clinic for Neurology, St. Josef Hospital, Ruhr University Bochum, Gudrunstraße 56, 44791, Bochum, Germany
| | - Philip Lennart Poser
- Clinic for Neurology, St. Josef Hospital, Ruhr University Bochum, Gudrunstraße 56, 44791, Bochum, Germany
| | - Barbara Gisevius
- Clinic for Neurology, St. Josef Hospital, Ruhr University Bochum, Gudrunstraße 56, 44791, Bochum, Germany
| | - Simon Faissner
- Clinic for Neurology, St. Josef Hospital, Ruhr University Bochum, Gudrunstraße 56, 44791, Bochum, Germany
| | - Anke Salmen
- Clinic for Neurology, St. Josef Hospital, Ruhr University Bochum, Gudrunstraße 56, 44791, Bochum, Germany
| | - Ilias Nastos
- Specialist Practice for Neurology, Bochum, Germany
| | - Ralf Gold
- Clinic for Neurology, St. Josef Hospital, Ruhr University Bochum, Gudrunstraße 56, 44791, Bochum, Germany
| | - Jeremias Motte
- Clinic for Neurology, St. Josef Hospital, Ruhr University Bochum, Gudrunstraße 56, 44791, Bochum, Germany
| |
Collapse
|
20
|
Marti Z, Ruder J, Thomas OG, Bronge M, De La Parra Soto L, Grönlund H, Olsson T, Martin R. Enhanced and cross-reactive in vitro memory B cell response against Epstein-Barr virus nuclear antigen 1 in multiple sclerosis. Front Immunol 2024; 15:1334720. [PMID: 39257578 PMCID: PMC11385009 DOI: 10.3389/fimmu.2024.1334720] [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: 11/07/2023] [Accepted: 04/04/2024] [Indexed: 09/12/2024] Open
Abstract
Multiple sclerosis (MS) is a prototypical autoimmune disease of the central nervous system (CNS). In addition to CD4+ T cells, memory B cells are now recognized as a critical cell type in the disease. This is underlined by the fact that the best-characterized environmental risk factor for MS is the Epstein-Barr virus (EBV), which can infect and persist in memory B cells throughout life. Several studies have identified changes in anti-EBV immunity in patients with MS. Examples include elevated titers of anti-EBV nuclear antigen 1 (EBNA1) antibodies, interactions of these with the MS-associated HLA-DR15 haplotype, and molecular mimicry with MS autoantigens like myelin basic protein (MBP), anoctamin-2 (ANO2), glial cell adhesion molecule (GlialCAM), and alpha-crystallin B (CRYAB). In this study, we employ a simple in vitro assay to examine the memory B cell antibody repertoire in MS patients and healthy controls. We replicate previous serological data from MS patients demonstrating an increased secretion of anti-EBNA1380-641 IgG in cell culture supernatants, as well as a positive correlation of these levels with autoantibodies against GlialCAM262-416 and ANO21-275. For EBNA1380-641 and ANO21-275, we provide additional evidence suggesting antibody cross-reactivity between the two targets. Further, we show that two efficacious MS treatments - natalizumab (NAT) and autologous hematopoietic stem cell transplantation (aHSCT) - are associated with distinct changes in the EBNA1-directed B cell response and that these alterations can be attributed to the unique mechanisms of action of these therapies. Using an in vitro system, our study confirms MS-associated changes in the anti-EBNA1 memory B cell response, EBNA1380-641 antibody cross-reactivity with ANO21-275, and reveals treatment-associated changes in the immunoglobulin repertoire in MS.
Collapse
Affiliation(s)
- Zoe Marti
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
- Research and Development, Cellerys, Schlieren, Switzerland
- Department of Neuroimmunology and Multiple Sclerosis Research, University Hospital Zurich, Zurich, Switzerland
| | - Josefine Ruder
- Department of Neuroimmunology and Multiple Sclerosis Research, University Hospital Zurich, Zurich, Switzerland
| | - Olivia G Thomas
- Therapeutic Immune Design Unit, Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Mattias Bronge
- Therapeutic Immune Design Unit, Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
- Department of Neurology, Karolinska University Hospital, Stockholm, Sweden
| | - Lorenzo De La Parra Soto
- Therapeutic Immune Design Unit, Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
- Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
| | - Hans Grönlund
- Therapeutic Immune Design Unit, Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Tomas Olsson
- Neuroimmunology Unit, Department of Clinical Neurocience, Karolinska Institutet, Stockholm, Sweden
| | - Roland Martin
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
- Research and Development, Cellerys, Schlieren, Switzerland
- Department of Neuroimmunology and Multiple Sclerosis Research, University Hospital Zurich, Zurich, Switzerland
- Therapeutic Immune Design Unit, Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
21
|
Klehmet J, Bopp T. [Ofatumumab in the treatment of multiple sclerosis - A summary of preclinical and clinical data]. FORTSCHRITTE DER NEUROLOGIE-PSYCHIATRIE 2024. [PMID: 39159670 DOI: 10.1055/a-2365-0966] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/21/2024]
Abstract
BACKGROUND B-cell targeted therapies are highly effective in multiple sclerosis (MS). Most of these therapies are administered intravenously at long intervals. Ofatumumab, an anti-CD20 antibody that is administered subcutaneously at low doses on a monthly basis due to its high affinity to the target structure, became available for the treatment of MS in 2021. METHODS An overview of practice-relevant immunological and clinical data on ofatumumab is provided. RESULTS The high affinity of ofatumumab to the target structure allows low dose and low volume administration, with the release and absorption profile after subcutaneous application allowing for high concentrations in the lymph nodes and gradual depletion of B-cells. Rapid onset of action is achieved as well as B-cell repletion within a few months in case of discontinuation of therapy. Long-term data show stable IgG levels over up to four years and high efficacy with respect to relapse rate, progression, and cognition. According to current study data, the effect compared to teriflunomide is greater the earlier therapy is initiated. Ofatumumab has a specific B-cell depletion pattern. CD20 expressing B-cell progenitor cells in the bone marrow are preserved and therefore also the inducibility and differentiation of plasma cells. The formation of a humoral immunological memory is therefore possible. Four-year study data showed no abnormalities in the rate of severe infections or malignancies. CONCLUSIONS Ofatumumab is an innovative B-cell targeted therapy. It is highly effective with a good safety and tolerability profile, well controllable and maintains immunocompetence against pathogens.
Collapse
Affiliation(s)
- Juliane Klehmet
- Klinik für Neurologie, Zentrum für Multiple Sklerose, Jüdisches Krankenhaus Berlin, Berlin, Germany
| | - Tobias Bopp
- Institut für Immunologie, Universitätsmedizin der Johannes Gutenberg-Universität Mainz, Mainz, Germany
- Forschungszentrum für Immuntherapie (FZI), Universitätsmedizin der Johannes Gutenberg-Universität Mainz, Mainz, Germany
| |
Collapse
|
22
|
Stevenson M, Algarzae NK, Moussa C. Tyrosine kinases: multifaceted receptors at the intersection of several neurodegenerative disease-associated processes. FRONTIERS IN DEMENTIA 2024; 3:1458038. [PMID: 39221072 PMCID: PMC11361951 DOI: 10.3389/frdem.2024.1458038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Accepted: 07/31/2024] [Indexed: 09/04/2024]
Abstract
Tyrosine kinases (TKs) are catalytic enzymes activated by auto-phosphorylation that function by phosphorylating tyrosine residues on downstream substrates. Tyrosine kinase inhibitors (TKIs) have been heavily exploited as cancer therapeutics, primarily due to their role in autophagy, blood vessel remodeling and inflammation. This suggests tyrosine kinase inhibition as an appealing therapeutic target for exploiting convergent mechanisms across several neurodegenerative disease (NDD) pathologies. The overlapping mechanisms of action between neurodegeneration and cancer suggest that TKIs may play a pivotal role in attenuating neurodegenerative processes, including degradation of misfolded or toxic proteins, reduction of inflammation and prevention of fibrotic events of blood vessels in the brain. In this review, we will discuss the distinct roles that select TKs have been shown to play in various disease-associated processes, as well as identify TKs that have been explored as targets for therapeutic intervention and associated pharmacological agents being investigated as treatments for NDDs.
Collapse
Affiliation(s)
- Max Stevenson
- The Laboratory for Dementia and Parkinsonism, Translational Neurotherapeutics Program, Department of Neurology, Georgetown University Medical Center, Washington, DC, United States
| | - Norah K. Algarzae
- Department of Physiology, College of Medicine, King Saud University, Riyadh, Saudi Arabia
| | - Charbel Moussa
- The Laboratory for Dementia and Parkinsonism, Translational Neurotherapeutics Program, Department of Neurology, Georgetown University Medical Center, Washington, DC, United States
| |
Collapse
|
23
|
Gudesblatt M, Bumstead B, Buhse M, Zarif M, Morrow SA, Nicholas JA, Hancock LM, Wilken J, Weller J, Scott N, Gocke A, Lewin JB, Kaczmarek O, Mendoza JP, Golan D. De-escalation of Disease-Modifying Therapy for People with Multiple Sclerosis Due to Safety Considerations: Characterizing 1-Year Outcomes in 25 People Who Switched from Ocrelizumab to Diroximel Fumarate. Adv Ther 2024; 41:3059-3075. [PMID: 38861218 PMCID: PMC11263251 DOI: 10.1007/s12325-024-02902-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Accepted: 05/14/2024] [Indexed: 06/12/2024]
Abstract
INTRODUCTION Switching disease-modifying therapy (DMT) may be considered for relapsing-remitting multiple sclerosis (RRMS) if a patient's current therapy is no longer optimal. This was particularly important during the recent COVID-19 pandemic because of considerations around immune deficiency and impaired vaccine response associated with B cell-depleting DMTs. This real-world, single-center study aimed to evaluate change or decline in functional ability and overall disease stability in people with RRMS who were switched from B cell-depleting ocrelizumab (OCRE) to diroximel fumarate (DRF) because of safety concern related to the COVID-19 pandemic. METHODS Adults with RRMS were included if they had been clinically stable for ≥ 1 year on OCRE. Data collected at baseline and 1 year post switch included relapse rate, magnetic resonance imaging (MRI), blood work for assessment of peripheral immune parameters, the Cognitive Assessment Battery (CAB), optical coherence tomography (OCT), and patient-reported outcomes (PROs). RESULTS Participants (N = 25) had a mean (SD) age of 52 (9) years, and a mean (SD) duration of 26 (8) months' treatment with OCRE before the switch to DRF. Median washout duration since the last OCRE infusion was 7 months (range 4-18 months). No participants relapsed on DRF during follow-up, and all remained persistent on DRF after 1 year. There were no significant changes in peripheral immune parameters, other than an increase in the percentage of CD19+ cells 1 year after switching (p < 0.05). Similarly, there were no significant changes in CAB, OCT, and PROs. CONCLUSION These preliminary findings suggest that transition to DRF from OCRE may be an effective treatment option for people with RRMS who are clinically stable but may need to switch for reasons unrelated to effectiveness. Longer follow-up times on larger samples are needed to confirm these observations.
Collapse
Affiliation(s)
- Mark Gudesblatt
- NYU Langone South Shore Neurologic Associates, PC, 77 Medford Ave, Patchogue, NY, 11772, USA.
| | - Barbara Bumstead
- NYU Langone South Shore Neurologic Associates, PC, 77 Medford Ave, Patchogue, NY, 11772, USA
| | - Marijean Buhse
- NYU Langone South Shore Neurologic Associates, PC, 77 Medford Ave, Patchogue, NY, 11772, USA
| | - Myassar Zarif
- NYU Langone South Shore Neurologic Associates, PC, 77 Medford Ave, Patchogue, NY, 11772, USA
| | - Sarah A Morrow
- Department of Clinical Neurosciences, University of Calgary, Hotchkiss Brain Institute, Calgary, AB, Canada
| | - Jacqueline A Nicholas
- OhioHealth Multiple Sclerosis Center, Riverside Methodist Hospital, Columbus, OH, USA
| | - Laura M Hancock
- Neurological Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Jeffrey Wilken
- Washington Neuropsychology Research Group, Fairfax, VA, USA
- Department of Neurology, Georgetown University School of Medicine, Washington, DC, USA
| | - Joanna Weller
- NYU Langone South Shore Neurologic Associates, PC, 77 Medford Ave, Patchogue, NY, 11772, USA
| | | | | | | | - Olivia Kaczmarek
- NYU Langone South Shore Neurologic Associates, PC, 77 Medford Ave, Patchogue, NY, 11772, USA
| | | | - Daniel Golan
- Multiple Sclerosis and Neuroimmunology Center, Lady Davis Carmel Medical Center, Haifa, Israel
- Ruth and Bruce Rappaport Faculty of Medicine, Technion, Israel Institute of Technology, Haifa, Israel
| |
Collapse
|
24
|
Hallberg S, Evertsson B, Lillvall E, Boremalm M, de Flon P, Wang Y, Salzer J, Lycke J, Fink K, Frisell T, Al Nimer F, Svenningsson A. Hypogammaglobulinaemia during rituximab treatment in multiple sclerosis: A Swedish cohort study. Eur J Neurol 2024; 31:e16331. [PMID: 38794973 PMCID: PMC11236063 DOI: 10.1111/ene.16331] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 04/08/2024] [Accepted: 04/24/2024] [Indexed: 05/27/2024]
Abstract
BACKGROUND AND PURPOSE Mechanisms behind hypogammaglobulinaemia during rituximab treatment are poorly understood. METHODS In this register-based multi-centre retrospective cohort study of multiple sclerosis (MS) patients in Sweden, 2745 patients from six participating Swedish MS centres were identified via the Swedish MS registry and included between 14 March 2008 and 25 January 2021. The exposure was treatment with at least one dose of rituximab for MS or clinically isolated syndrome, including data on treatment duration and doses. The degree of yearly decrease in immunoglobulin G (IgG) and immunoglobulin M (IgM) levels was evaluated. RESULTS The mean decrease in IgG was 0.27 (95% confidence interval 0.17-0.36) g/L per year on rituximab treatment, slightly less in older patients, and without significant difference between sexes. IgG or IgM below the lower limit of normal (<6.7 or <0.27 g/L) was observed in 8.8% and 8.3% of patients, respectively, as nadir measurements. Six out of 2745 patients (0.2%) developed severe hypogammaglobulinaemia (IgG below 4.0 g/L) during the study period. Time on rituximab and accumulated dose were the main predictors for IgG decrease. Previous treatment with fingolimod and natalizumab, but not teriflunomide, dimethyl fumarate, interferons or glatiramer acetate, were significantly associated with lower baseline IgG levels by 0.80-1.03 g/L, compared with treatment-naïve patients. Switching from dimethyl fumarate or interferons was associated with an additional IgG decline of 0.14-0.19 g/L per year, compared to untreated. CONCLUSIONS Accumulated dose and time on rituximab treatment are associated with a modest but significant decline in immunoglobulin levels. Previous MS therapies may influence additional IgG decline.
Collapse
Affiliation(s)
- Susanna Hallberg
- Department of Clinical SciencesKarolinska Institutet, Danderyds SjukhusStockholmSweden
| | - Björn Evertsson
- Department of Clinical NeuroscienceKarolinska InstitutetStockholmSweden
| | - Ellen Lillvall
- Department of Clinical Neuroscience, Institute of Neuroscience and Physiology at Sahlgrenska AcademyUniversity of GothenburgGothenburgSweden
| | - Malin Boremalm
- Department of Clinical Science, NeurosciencesUmeå UniversityUmeåSweden
| | - Pierre de Flon
- Department of Clinical Sciences, Neurosciences, Unit of Neurology, ÖstersundUmeå UniversityUmeåSweden
| | - Yunzhang Wang
- Department of Clinical SciencesKarolinska Institutet, Danderyds SjukhusStockholmSweden
| | - Jonatan Salzer
- Department of Clinical Science, NeurosciencesUmeå UniversityUmeåSweden
| | - Jan Lycke
- Department of Clinical Neuroscience, Institute of Neuroscience and Physiology at Sahlgrenska AcademyUniversity of GothenburgGothenburgSweden
| | - Katharina Fink
- Department of Clinical NeuroscienceKarolinska InstitutetStockholmSweden
| | - Thomas Frisell
- Clinical Epidemiology Division, Department of Medicine SolnaKarolinska InstitutetStockholmSweden
| | - Faiez Al Nimer
- Department of Clinical NeuroscienceKarolinska InstitutetStockholmSweden
| | - Anders Svenningsson
- Department of Clinical SciencesKarolinska Institutet, Danderyds SjukhusStockholmSweden
| |
Collapse
|
25
|
Jacob A, Shatila AO, Inshasi J, Massouh J, Mir R, Noori S, Yamout B. Disease modifying treatment guidelines for multiple sclerosis in the United Arab Emirates. Mult Scler Relat Disord 2024; 88:105703. [PMID: 38924933 DOI: 10.1016/j.msard.2024.105703] [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: 01/21/2024] [Revised: 05/13/2024] [Accepted: 06/04/2024] [Indexed: 06/28/2024]
Abstract
The newly constituted National Multiple Sclerosis (MS) Society (NMSS)of the United Arab Emirates (UAE), set up a scientific committee to create a MS disease modifying treatment (DMT) guideline for UAE. The committee considered several unique features of the MS community in UAE including large number of expatriate population, wide variations in health insurance coverage, physician and patient preferences for DMT. The overall goal of the treatment guideline is to facilitate the most appropriate DMT to the widest number of patients. To this end it has adapted recommendations from various health systems and regulatory authorities into a pragmatic amalgamation of best practices from across the world. Importantly where data is unavailable or controversial, a common sense approach is taken rather than leave physicians and patients in limbo. The committee classifies MS into subcategories and suggests appropriate treatment choices. It recommends treatment of RIS and CIS with poor prognostic factors. It largely equates the efficacy and safety of DMT with similar mechanisms of action or drug classes e.g. ocrelizumab is similar to rituximab. It allows early switching of treatment for unambiguous disease activity and those with progression independent of relapses. Autologous hematopoietic stem cell transplantation can be offered to patients who fail one high efficacy DMT. Pragmatic guidance on switching and stopping DMT, DMT choices in pregnancy, lactation and pediatric MS have been included. It is expected that these guidelines will be updated periodically as new data becomes available.
Collapse
Affiliation(s)
- Anu Jacob
- Neurological Institute, Cleveland Clinic, Abu Dhabi, United Arab Emirates; The Walton Centre, Liverpool, United Kingdom.
| | - Ahmed Osman Shatila
- Department of Neurology, Sheikh Shakhbout Medical City Abu Dhabi, United Arab Emirates
| | - Jihad Inshasi
- Department of Neurology, Rashid Hospital and Dubai Medical College, Dubai Health Authority, Dubai, United Arab Emirates
| | - Joelle Massouh
- Neurology Institute and Multiple Sclerosis Centre, Harley Street Medical centre, Abu Dhabi, United Arab Emirates
| | - Ruquia Mir
- Abu Dhabi stem Cell Clinic, United Arab Emirates
| | - Suzan Noori
- University Hospital Sharjah, United Arab Emirates
| | - Bassem Yamout
- Neurology Institute and Multiple Sclerosis Centre, Harley Street Medical centre, Abu Dhabi, United Arab Emirates; American University of Beirut, Lebanon
| |
Collapse
|
26
|
Martin SJ, Guenette M, Oh J. Evaluating the Therapeutic Potential of Ublituximab in the Treatment of MS: Design, Development and Place in Therapy. Drug Des Devel Ther 2024; 18:3025-3042. [PMID: 39050801 PMCID: PMC11268567 DOI: 10.2147/dddt.s388410] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Accepted: 07/10/2024] [Indexed: 07/27/2024] Open
Abstract
B cells are critical to the pathogenesis of multiple sclerosis (MS), an autoimmune disease of the central nervous system. B cell depletion using anti-CD20 monoclonal antibodies (mAbs) has proven to be an extremely successful treatment strategy, with profound suppression of both clinical and radiological evidence of focal inflammatory disease. Several anti-CD20 mAbs are now licensed for use in MS, with ublituximab being the latest to gain regulatory approval. The unique properties of each of the anti-CD20 mAb may result in nuanced differences in timing, duration and depth of B cell depletion, with the potential for such differences to have a clinical relevance to both drug efficacy and adverse effects. In this review, we summarize the design, development, and current place in MS therapy for ublituximab.
Collapse
Affiliation(s)
- Sarah-Jane Martin
- Division of Neurology, Department of Medicine, St Michael’s Hospital, Toronto, Canada
- University of Glasgow, Glasgow, UK
| | - Melanie Guenette
- Division of Neurology, Department of Medicine, St Michael’s Hospital, Toronto, Canada
| | - Jiwon Oh
- Division of Neurology, Department of Medicine, St Michael’s Hospital, Toronto, Canada
| |
Collapse
|
27
|
Mustelin T, Andrade F. Autoimmunity: the neoantigen hypothesis. Front Immunol 2024; 15:1432985. [PMID: 38994353 PMCID: PMC11236689 DOI: 10.3389/fimmu.2024.1432985] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Accepted: 06/17/2024] [Indexed: 07/13/2024] Open
Affiliation(s)
- Tomas Mustelin
- Division of Rheumatology, Department of Medicine, University of Washington, Seattle, WA, United States
| | - Felipe Andrade
- Division of Rheumatology, The Johns Hopkins University School of Medicine, Baltimore, MD, United States
| |
Collapse
|
28
|
Fazazi MR, Doss PMIA, Pereira R, Fudge N, Regmi A, Joly-Beauparlant C, Akbar I, Yeola AP, Mailhot B, Baillargeon J, Grenier P, Bertrand N, Lacroix S, Droit A, Moore CS, Rojas OL, Rangachari M. Myelin-reactive B cells exacerbate CD4 + T cell-driven CNS autoimmunity in an IL-23-dependent manner. Nat Commun 2024; 15:5404. [PMID: 38926356 PMCID: PMC11208426 DOI: 10.1038/s41467-024-49259-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Accepted: 05/30/2024] [Indexed: 06/28/2024] Open
Abstract
B cells and T cells collaborate in multiple sclerosis (MS) pathogenesis. IgH[MOG] mice possess a B cell repertoire skewed to recognize myelin oligodendrocyte glycoprotein (MOG). Here, we show that upon immunization with the T cell-obligate autoantigen, MOG[35-55], IgH[MOG] mice develop rapid and exacerbated experimental autoimmune encephalomyelitis (EAE) relative to wildtype (WT) counterparts, characterized by aggregation of T and B cells in the IgH[MOG] meninges and by CD4+ T helper 17 (Th17) cells in the CNS. Production of the Th17 maintenance factor IL-23 is observed from IgH[MOG] CNS-infiltrating and meningeal B cells, and in vivo blockade of IL-23p19 attenuates disease severity in IgH[MOG] mice. In the CNS parenchyma and dura mater of IgH[MOG] mice, we observe an increased frequency of CD4+PD-1+CXCR5- T cells that share numerous characteristics with the recently described T peripheral helper (Tph) cell subset. Further, CNS-infiltrating B and Tph cells from IgH[MOG] mice show increased reactive oxygen species (ROS) production. Meningeal inflammation, Tph-like cell accumulation in the CNS and B/Tph cell production of ROS were all reduced upon p19 blockade. Altogether, MOG-specific B cells promote autoimmune inflammation of the CNS parenchyma and meninges in an IL-23-dependent manner.
Collapse
Affiliation(s)
- Mohamed Reda Fazazi
- axe Neurosciences, Centre de recherche du Centre hospitalier universitaire (CHU) de Québec - Université Laval, Pavillon CHUL, 2705 boul Laurier, Quebec City, G1V 4G2, QC, Canada
| | - Prenitha Mercy Ignatius Arokia Doss
- axe Neurosciences, Centre de recherche du Centre hospitalier universitaire (CHU) de Québec - Université Laval, Pavillon CHUL, 2705 boul Laurier, Quebec City, G1V 4G2, QC, Canada
| | - Resel Pereira
- Krembil Research Institute, University Health Network, Toronto, M5T 0S8, ON, Canada
| | - Neva Fudge
- Division of BioMedical Sciences, Memorial University of Newfoundland, St. John's, NL, A1B 3V6, Canada
- Department of Neurology, Faculty of Medicine, Memorial University of Newfoundland, St. John's, NL, A1B 3V6, Canada
| | - Aryan Regmi
- Krembil Research Institute, University Health Network, Toronto, M5T 0S8, ON, Canada
- Department of Immunology, University of Toronto, Toronto, M5S 1A1, ON, Canada
| | - Charles Joly-Beauparlant
- axe Endocrinologie et nephrologie, Centre de recherche du Centre hospitalier universitaire (CHU) de Québec - Université Laval, Pavillon CHUL, 2705 boul Laurier, Quebec City, QC, G1V 4G2, Canada
| | - Irshad Akbar
- axe Neurosciences, Centre de recherche du Centre hospitalier universitaire (CHU) de Québec - Université Laval, Pavillon CHUL, 2705 boul Laurier, Quebec City, G1V 4G2, QC, Canada
| | - Asmita Pradeep Yeola
- axe Neurosciences, Centre de recherche du Centre hospitalier universitaire (CHU) de Québec - Université Laval, Pavillon CHUL, 2705 boul Laurier, Quebec City, G1V 4G2, QC, Canada
| | - Benoit Mailhot
- axe Neurosciences, Centre de recherche du Centre hospitalier universitaire (CHU) de Québec - Université Laval, Pavillon CHUL, 2705 boul Laurier, Quebec City, G1V 4G2, QC, Canada
| | - Joanie Baillargeon
- axe Neurosciences, Centre de recherche du Centre hospitalier universitaire (CHU) de Québec - Université Laval, Pavillon CHUL, 2705 boul Laurier, Quebec City, G1V 4G2, QC, Canada
| | - Philippe Grenier
- axe Endocrinologie et nephrologie, Centre de recherche du Centre hospitalier universitaire (CHU) de Québec - Université Laval, Pavillon CHUL, 2705 boul Laurier, Quebec City, QC, G1V 4G2, Canada
| | - Nicolas Bertrand
- axe Endocrinologie et nephrologie, Centre de recherche du Centre hospitalier universitaire (CHU) de Québec - Université Laval, Pavillon CHUL, 2705 boul Laurier, Quebec City, QC, G1V 4G2, Canada
- Faculty of Pharmacy, Laval University, 1050 ave de la Médecine, Quebec City, QC, G1V 4G2, Canada
| | - Steve Lacroix
- axe Neurosciences, Centre de recherche du Centre hospitalier universitaire (CHU) de Québec - Université Laval, Pavillon CHUL, 2705 boul Laurier, Quebec City, G1V 4G2, QC, Canada
- Department of Molecular Medicine, Faculty of Medicine, Laval University, 1050 ave de la Médecine, Quebec City, QC, G1V 4G2, Canada
| | - Arnaud Droit
- axe Endocrinologie et nephrologie, Centre de recherche du Centre hospitalier universitaire (CHU) de Québec - Université Laval, Pavillon CHUL, 2705 boul Laurier, Quebec City, QC, G1V 4G2, Canada
- Department of Molecular Medicine, Faculty of Medicine, Laval University, 1050 ave de la Médecine, Quebec City, QC, G1V 4G2, Canada
| | - Craig S Moore
- Division of BioMedical Sciences, Memorial University of Newfoundland, St. John's, NL, A1B 3V6, Canada
- Department of Neurology, Faculty of Medicine, Memorial University of Newfoundland, St. John's, NL, A1B 3V6, Canada
| | - Olga L Rojas
- Krembil Research Institute, University Health Network, Toronto, M5T 0S8, ON, Canada
- Department of Immunology, University of Toronto, Toronto, M5S 1A1, ON, Canada
| | - Manu Rangachari
- axe Neurosciences, Centre de recherche du Centre hospitalier universitaire (CHU) de Québec - Université Laval, Pavillon CHUL, 2705 boul Laurier, Quebec City, G1V 4G2, QC, Canada.
- Department of Molecular Medicine, Faculty of Medicine, Laval University, 1050 ave de la Médecine, Quebec City, QC, G1V 4G2, Canada.
| |
Collapse
|
29
|
Georgieva T, Diddens J, Friedrich V, Lepennetier G, Brand RM, Lehmann-Horn K. Single-cell profiling indicates a high similarity between immune cells in the cerebrospinal fluid and in meningeal ectopic lymphoid tissue in experimental autoimmune encephalomyelitis. Front Immunol 2024; 15:1400641. [PMID: 38933267 PMCID: PMC11199773 DOI: 10.3389/fimmu.2024.1400641] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Accepted: 05/08/2024] [Indexed: 06/28/2024] Open
Abstract
Background and objectives B cell depleting anti-CD20 monoclonal antibodies (aCD20 mAbs) are highly effective in treatment of multiple sclerosis (MS) but fail to halt the formation of meningeal ectopic lymphoid tissue (mELT) in the murine model experimental autoimmune encephalomyelitis (EAE). While mELT can be examined in EAE, it is not accessible in vivo in MS patients. Our key objectives were to compare the immune cells in cerebrospinal fluid (CSF), which is accessible in patients, with those in mELT, and to study the effects of aCD20 mAbs on CSF and mELT in EAE. Methods Applying single cell RNA sequencing, we compared gene expression profiles in immune cells from (1) CSF with mELT and (2) aCD20 mAbs treated with control treated mice in a spontaneous 2D2xTh EAE model. Results The immune cell composition in CSF and mELT was very similar. Gene expression profiles and pathway enrichment analysis revealed no striking differences between the two compartments. aCD20 mAbs led not only to a virtually complete depletion of B cells in the CSF but also to a reduction of naïve CD4+ T cells and marked increase of macrophages. No remarkable differences in regulated genes or pathways were observed. Discussion Our results suggest that immune cells in the CSF may serve as a surrogate for mELT in EAE. Future studies are required to confirm this in MS patients. The observed increase of macrophages in B cell depleted CSF is a novel finding and requires verification in CSF of aCD20 mAbs treated MS patients. Due to unresolved technical challenges, we were unable to study the effects of aCD20 mAbs on mELT. This should be addressed in future studies.
Collapse
Affiliation(s)
| | | | | | | | | | - Klaus Lehmann-Horn
- Department of Neurology, TUM School of Medicine and Health, Technical University of Munich, Munich, Germany
| |
Collapse
|
30
|
Zhang C, Shi FD. Targeting B cells in neuromyelitis optica spectrum disorder. Lancet Neurol 2024; 23:549-550. [PMID: 38760085 DOI: 10.1016/s1474-4422(24)00115-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Accepted: 03/07/2024] [Indexed: 05/19/2024]
Affiliation(s)
- Chao Zhang
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Fu-Dong Shi
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin 300052, China; Department of Neurology, National Clinical Research Center for Neurological Diseases of China, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.
| |
Collapse
|
31
|
Khandait PD, Rohatgi S, Nirhale SP, Rao PM, Naphade PU. Rituximab Therapy for Immune-Mediated Neurological Diseases: Our Experience at a Tertiary Care Centre. Cureus 2024; 16:e62227. [PMID: 39006634 PMCID: PMC11244719 DOI: 10.7759/cureus.62227] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2024] [Accepted: 06/12/2024] [Indexed: 07/16/2024] Open
Abstract
INTRODUCTION Rituximab (RTX) is a monoclonal anti-CD20 chimeric antibody that inhibits B cell activity. However, it is an appealing substitute for traditional immunomodulatory drugs as a swiftly acting, targeted therapy with mounting evidence of efficacy and tolerance in numerous neuroinflammatory conditions. We discuss the scientific evidence for the use of RTX in neurological illnesses, as well as the dose, safety, and other practical elements of prescription. AIM This study aims to assess and correlate the effects of RTX on immune-mediated neurological disorders. OBJECTIVES The primary objective of this study is to determine the outcomes in patients treated with RTX for the following conditions: myasthenia gravis (MG), autoimmune encephalitis, multiple sclerosis (MS), neuromyelitis optica spectrum disorder (NMOSD), myelin oligodendrocyte glycoprotein antibody disease (MOGAD), immune-mediated peripheral neuropathy, and inflammatory muscle disease. The secondary objective is to assess adverse drug reactions in patients treated with RTX. METHODS This is a prospective observational study conducted at a tertiary care centre. The data were analyzed for the period from May 2022 to May 2024. Approval was obtained from the institutional ethics committee before commencing the study, and written informed consent was obtained from all patients. RESULTS AND CONCLUSIONS A total of 56 patients were included in the study. The distribution of patients according to diseases is as follows: MG (17), MS (11), NMOSD (10), MOGAD (7), immune-mediated peripheral neuropathy (6), autoimmune encephalitis (3), and inflammatory muscle disease (2). However, one patient was lost to follow-up in the autoimmune encephalitis group. All patients experienced improvements in symptoms, and no relapse episodes have been reported except for one patient who had a relapse in the inflammatory muscle disease group. During the infusion process, some adverse drug reactions, such as chills and rigors, were observed, and two patients experienced major side effects, such as Pott's disease and cryptogenic organizing pneumonia. Nevertheless, overall, rituximab shows promise as an off-label immunosuppressive treatment for the aforementioned neurological immune-mediated diseases.
Collapse
Affiliation(s)
- Pranit D Khandait
- Neurology, Dr. D. Y. Patil Medical College, Hospital & Research Centre, Dr. D. Y. Patil Vidyapeeth (Deemed to be University), Pune, IND
| | - Shalesh Rohatgi
- Neurology, Dr. D. Y. Patil Medical College, Hospital & Research Centre, Dr. D. Y. Patil Vidyapeeth (Deemed to be University), Pune, IND
| | - Satish P Nirhale
- Neurology, Dr. D. Y. Patil Medical College, Hospital & Research Centre, Dr. D. Y. Patil Vidyapeeth (Deemed to be University), Pune, IND
| | - Prajwal M Rao
- Neurology, Dr. D. Y. Patil Medical College, Hospital & Research Centre, Dr. D. Y. Patil Vidyapeeth (Deemed to be University), Pune, IND
| | - Pravin U Naphade
- Neurology, Dr. D. Y. Patil Medical College, Hospital & Research Centre, Dr. D. Y. Patil Vidyapeeth (Deemed to be University), Pune, IND
| |
Collapse
|
32
|
Alping P, Neovius M, Piehl F, Frisell T. Real-World Healthcare Cost Savings and Reduced Relapse Rate with Off-Label Rituximab versus Disease-Modifying Treatments Approved for Relapsing-Remitting Multiple Sclerosis: A Nationwide Cost-Effectiveness Study. Ann Neurol 2024; 95:1099-1111. [PMID: 38529711 DOI: 10.1002/ana.26914] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2023] [Revised: 02/28/2024] [Accepted: 03/04/2024] [Indexed: 03/27/2024]
Abstract
OBJECTIVE Although off-label use of rituximab is a common alternative to disease-modifying therapies (DMTs) approved for multiple sclerosis (MS) in several countries, the impact of this on treatment cost-effectiveness is not well known. METHODS We evaluated the relative cost-effectiveness of rituximab and MS-approved DMTs in a register-based cohort study of Swedish residents with relapsing-remitting MS, aged 18-65 years, starting treatment with rituximab, natalizumab, fingolimod, or dimethyl fumarate between January 2010 and July 2016, and followed through July 2021 (n = 5,924). By linking the population-based Swedish MS register to several Swedish health care and demographic registers, we estimated health care costs in relation to number of relapses, over 5 years from treatment start. Differences between treatments were estimated in inverse probability of treatment-weighted regression models, adjusting for a broad range of potential confounders covering demographics, medical history, and MS-related clinical characteristics. RESULTS Off-label rituximab was associated with both lower total health care costs (mean cost savings ranged $35,000-$66,000 vs. each approved DMT), and fewer relapses (mean number of prevented relapses ranged 0.12-0.22), per started therapy over 5 years. Results were robust to variations in discounting and pricing of health care visits, with the main driver of cost-savings being the price of the index drug itself. INTERPRETATION The cost-effectiveness of rituximab dominated the MS-approved alternatives. Off-label, low-dose rituximab should be considered for persons with MS and could reduce barriers to treatment, especially in resource-limited settings. ANN NEUROL 2024;95:1099-1111.
Collapse
Affiliation(s)
- Peter Alping
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
- Clinical Epidemiology Division, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
| | - Martin Neovius
- Clinical Epidemiology Division, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
| | - Fredrik Piehl
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Thomas Frisell
- Clinical Epidemiology Division, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
33
|
Brown JD, Muston BT, Massey J. Switching from natalizumab to an anti-CD20 monoclonal antibody in relapsing remitting multiple sclerosis: A systematic review. Mult Scler Relat Disord 2024; 86:105605. [PMID: 38640586 DOI: 10.1016/j.msard.2024.105605] [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: 11/22/2023] [Revised: 04/04/2024] [Accepted: 04/07/2024] [Indexed: 04/21/2024]
Abstract
BACKGROUND Use of natalizumab (NTZ) is precluded in many Multiple Sclerosis (MS) patients by the risk of progressive multifocal leukoencephalopathy (PML). Regardless, some patients may commence natalizumab for short term disease control in spite of being seropositive, and others may seroconvert whilst on treatment. In these circumstances, discontinuation of NTZ should not occur until a clear exit strategy is established to prevent post-NTZ disease reactivation, which often exceeds the severity of disease activity prior to NTZ treatment. The objective of this systematic review was to summarise the available evidence for CD20-monoclonal antibodies (CD20mAb) as a suitable NTZ exit strategy, and to identify whether a superior switch protocol can be established. METHODS In accordance with PRISMA guidelines, a total of 2393 references were extracted from a search of three online databases (PubMed, Scopus, MEDLINE). Following the application of inclusion/exclusion criteria, a total of 5 studies representing 331 patients were included. RESULTS The overall incidence of clinical relapse during washout periods ranging from 4.4-10.7 weeks was 0 %. The incidence of clinical relapse during two-year follow-up ranged from 1.8 % to 10 % for switches to all types of CD20 monoclonal antibody. The weighted mean for clinical relapse at 12 months was 8.8 %. Three studies reported an annualised relapse rate (ARR) ranging from 0.02-0.12 with a weighted mean ARR of 0.07. The overall incidence of PML during washout was 0 % and the overall incidence of PML within 6 months follow-up was 0.6 %. CONCLUSIONS This systematic review provides the first attempt at identifying a superior switch protocol in patients at risk of PML transitioning from NTZ to a CD20mAb. Our results indicate that CD20mAb's are a suitable transitional option for patients who discontinue NTZ, with our cohort demonstrating very low rates of carryover PML and low rates of clinical relapse. The most appropriate washout period is unclear due to confounding factors but is likely between 4 and 12 weeks.
Collapse
Affiliation(s)
| | - Benjamin T Muston
- Faculty of Medicine and Health, University of New South Wales, Sydney, Australia; The Collaborative Research Group (CORE), Sydney, Australia
| | - Jennifer Massey
- Faculty of Medicine and Health, University of New South Wales, Sydney, Australia; Neurology Department, St Vincent's Hospital Sydney, Australia
| |
Collapse
|
34
|
Bartak H, Fareh T, Ben Othman N, Viard D, Cohen M, Rocher F, Ewig E, Drici MD, Lebrun-Frenay C. Dental Adverse Effects of Anti-CD20 Therapies. Neurol Ther 2024; 13:917-930. [PMID: 38668835 PMCID: PMC11136893 DOI: 10.1007/s40120-024-00616-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Accepted: 04/03/2024] [Indexed: 05/30/2024] Open
Abstract
INTRODUCTION Over the past few years, anti-CD20 therapies like rituximab, ocrelizumab or ofatumumab have seen an increase in interest in the treatment of neurological autoimmune disorders such as multiple sclerosis (MS), neuromyelitis optica spectrum disorders (NMOSD), or resistant forms of generalized myasthenia gravis (MG). They are generally well-tolerated, but recent reports have highlighted severe dental disorders in patients undergoing anti-CD20 therapies. The aim was to describe a series of cases and to compare with the available scientific literature. METHODS We reviewed 6 patient cases with dental disorders during anti-CD20 therapy that were reported to the pharmacovigilance center. A disproportionality analysis was also conducted on Vigibase® for each anti-CD20 and each adverse effect described in the cases. RESULTS Six cases of dental and gingival conditions in relatively young patients were reported (median age: 40.5 years old [min: 34; max: 79]). Oral conditions were developed in four patients with MS treated with ocrelizumab and in two patients receiving rituximab (one patient with MG and one with NMOSD). The onset of oral conditions ranged from 10 days to 2 years after treatment initiation. Notably, all patients treated with ocrelizumab experienced gingival recession. Various dental pathologies were observed, including tooth loss, dental pain, caries, brittle teeth, dental fractures, dental abscesses, and periodontitis. Analysis of Vigibase® revealed 284 worldwide cases of dental and gingival conditions under ocrelizumab, 386 cases under rituximab, and 80 under ofatumumab. Significant associations were found between these therapies and dental pathologies, particularly tooth abscesses and infections. CONCLUSION To our knowledge, this is the first case series reporting dental conditions developed in patients long-term treated with anti-CD20 treatments. This issue, literature data, and Vigilyze® analysis might be considered a safety signal that necessitates being confirmed with more robust data, such as a retrospective study with a control group. Meanwhile, proactive measures are essential like frequent dental checkups and dental hygienic measures to prevent oral health problems associated with anti-CD20 therapies.
Collapse
Affiliation(s)
- Hélène Bartak
- Department of Pharmacology and Pharmacovigilance Center of Nice, University Hospital of Nice, Nice, France
| | - Tasnim Fareh
- Department of Pharmacology and Pharmacovigilance Center of Nice, University Hospital of Nice, Nice, France
| | - Nouha Ben Othman
- Department of Pharmacology and Pharmacovigilance Center of Nice, University Hospital of Nice, Nice, France
| | - Delphine Viard
- Department of Pharmacology and Pharmacovigilance Center of Nice, University Hospital of Nice, Nice, France
| | - Mikael Cohen
- Neurology MS Clinic, UMR2CA-URRIS, University Hospital of Nice, Nice, France
| | - Fanny Rocher
- Department of Pharmacology and Pharmacovigilance Center of Nice, University Hospital of Nice, Nice, France
| | - Elliot Ewig
- Department of Pharmacology and Pharmacovigilance Center of Nice, University Hospital of Nice, Nice, France
| | - Milou-Daniel Drici
- Department of Pharmacology and Pharmacovigilance Center of Nice, University Hospital of Nice, Nice, France.
| | | |
Collapse
|
35
|
Breu M, Sandesjö F, Milos R, Svoboda J, Salzer J, Schneider L, Reichelt JB, Bertolini A, Blaschek A, Fink K, Höftberger R, Lycke J, Rostásy K, Seidl R, Siegert S, Wickström R, Kornek B. Rituximab treatment in pediatric-onset multiple sclerosis. Eur J Neurol 2024; 31:e16228. [PMID: 38375947 PMCID: PMC11235651 DOI: 10.1111/ene.16228] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Revised: 12/05/2023] [Accepted: 01/15/2024] [Indexed: 02/21/2024]
Abstract
BACKGROUND AND PURPOSE Rituximab (RTX) is frequently used off-label in multiple sclerosis. However, studies on the risk-benefit profile of RTX in pediatric-onset multiple sclerosis are scarce. METHODS In this multicenter retrospective cohort study, patients with pediatric-onset multiple sclerosis from Sweden, Austria and Germany, who received RTX treatment were identified by chart review. Annualized relapse rates, Expanded Disability Status Scale scores and magnetic resonance imaging parameters (new T2 lesions and contrast-enhancing lesions) were assessed before and during RTX treatment. The proportion of patients who remained free from clinical and disease activity (NEDA-3) during RTX treatment was calculated. Side effects such as infusion-related reactions, infections and laboratory abnormalities were assessed. RESULTS Sixty-one patients received RTX during a median (interquartile range) follow-up period of 20.9 (35.6) months. The annualized relapse rate decreased from 0.6 (95% confidence interval [CI] 0.38-0.92) to 0.03 (95% CI 0.02-0.14). The annual rate of new T2 lesions decreased from 1.25 (95% CI 0.70-2.48) to 0.08 (95% CI 0.03-0.25) and annual rates of new contrast-enhancing lesions decreased from 0.86 (95% CI 0.30-3.96) to 0. Overall, 70% of patients displayed no evidence of disease activity (NEDA-3). Adverse events were observed in 67% of patients. Six patients discontinued treatment due to ongoing disease activity or adverse events. CONCLUSION Our study provides class IV evidence that RTX reduces clinical and radiological activity in pediatric-onset multiple sclerosis.
Collapse
Affiliation(s)
- Markus Breu
- Division of Pediatric Pulmonology, Allergology and Endocrinology, Department of Pediatrics and Adolescent MedicineMedical University of ViennaViennaAustria
| | - Fredrik Sandesjö
- Neuropediatric Unit, Department of Women's and Children's HealthKarolinska InstitutetStockholmSweden
| | - Ruxandra‐Iulia Milos
- Department of Biomedical Imaging and Image‐guided TherapyMedical University of ViennaViennaAustria
| | - Jan Svoboda
- Department of NeuroradiologyKarolinska University HospitalStockholmSweden
| | - Jonatan Salzer
- Department of Clinical Science, NeurosciencesUmeå UniversityUmeåSweden
| | - Lisa Schneider
- Division of Infectious Diseases, Department of Internal Medicine IMedical University of ViennaViennaAustria
| | - Julian Benedikt Reichelt
- Division of Pediatric Pulmonology, Allergology and Endocrinology, Department of Pediatrics and Adolescent MedicineMedical University of ViennaViennaAustria
- Department of NeurologyMedical University of ViennaViennaAustria
| | - Annikki Bertolini
- Department of Pediatric NeurologyUniversity Witten/Herdecke, Children's Hospital DattelnDattelnGermany
| | - Astrid Blaschek
- Paediatric Neurology and Developmental MedicineLudwig Maximilian University of Munich, Dr. von Hauner Children's HospitalMunichGermany
| | - Katharina Fink
- Department of Clinical NeuroscienceKarolinska InstitutetStockholmSweden
| | - Romana Höftberger
- Division of Neuropathology and Neurochemistry, Department of NeurologyMedical University of ViennaViennaAustria
- Comprehensive Center for Clinical Neurosciences and Mental HealthMedical University of ViennaViennaAustria
| | - Jan Lycke
- Department of Clinical Neuroscience, Institute of Neuroscience and Physiology, Sahlgrenska AcademyUniversity of GothenburgGothenburgSweden
| | - Kevin Rostásy
- Department of Pediatric NeurologyUniversity Witten/Herdecke, Children's Hospital DattelnDattelnGermany
| | - Rainer Seidl
- Division of Pediatric Pulmonology, Allergology and Endocrinology, Department of Pediatrics and Adolescent MedicineMedical University of ViennaViennaAustria
| | - Sandy Siegert
- Division of Pediatric Pulmonology, Allergology and Endocrinology, Department of Pediatrics and Adolescent MedicineMedical University of ViennaViennaAustria
| | - Ronny Wickström
- Neuropediatric Unit, Department of Women's and Children's HealthKarolinska InstitutetStockholmSweden
| | - Barbara Kornek
- Department of NeurologyMedical University of ViennaViennaAustria
- Comprehensive Center for Clinical Neurosciences and Mental HealthMedical University of ViennaViennaAustria
| |
Collapse
|
36
|
Graille-Avy L, Boutiere C, Rigollet C, Perriguey M, Rico A, Demortiere S, Durozard P, Hilezian F, Vely F, Bertault-Peres P, Pelletier J, Maarouf A, Audoin B. Effect of Prior Treatment With Fingolimod on Early and Late Response to Rituximab/Ocrelizumab in Patients With Multiple Sclerosis. NEUROLOGY(R) NEUROIMMUNOLOGY & NEUROINFLAMMATION 2024; 11:e200231. [PMID: 38626360 PMCID: PMC11090020 DOI: 10.1212/nxi.0000000000200231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Accepted: 02/02/2024] [Indexed: 04/18/2024]
Abstract
BACKGROUND AND OBJECTIVES Real-life studies noted that the risk of disease activity in multiple sclerosis (MS) after switching to rituximab (RTX) or ocrelizumab (OCR) may be unequal depending on prior disease-modifying therapy (DMT), with a higher risk associated with fingolimod (FING). METHODS We performed a retrospective analysis of a structured prospective data collection including all consecutive patients with relapsing MS who were prescribed RTX/OCR in the MS center of Marseille. Cox proportional hazards models were applied to clinical and MRI outcomes. RESULTS We included 321 patients with a median (interquartile range [IQR]) follow-up of 3.5 years (1.5-5) after RTX/OCR initiation. At the first RTX/OCR infusion, the mean (SD) age of patients was 37 (10) years, and the median (IQR) disease duration was 8 years (3-15): 68 patients did not receive treatment before RTX/OCR and 108 switched from FING, 47 from low efficacy therapy, and 98 from natalizumab. For statistical analysis, the group "FING" was divided into "short-FING" and "long-FING" groups according to the median value of the group's washout period (27 days). On Cox proportional hazards analysis, for only the "long-FING" group, the risk of relapse within the first 6 months of RTX/OCR was increased as compared with patients without previous DMT (hazard ratio [HR]: 8.78; 95% CI 1.72-44.86; p < 0.01). Previous DMT and washout period duration of FING had no effect on B-cell levels at 6 months. Beyond the first 6 months of RTX/OCR, age <40 years was associated with increased risk of relapse (HR: 3.93; 95% CI 1.30-11.89; p = 0.01), male sex with increased risk of new T2 lesions (HR: 2.26; 95% CI 1.08-4.74; p = 0.03), and EDSS ≥2 with increased risk of disability accumulation (HR: 3.01; 95% CI 1.34-6.74; p < 0.01). Previous DMT had no effect on the effectiveness of RTX/OCR beyond 6 months after initiation. DISCUSSION For patients switching from FING to RTX/OCR, the risk of disease reactivation within the first 6 months of treatment was increased as compared with patients with other DMT or no previous DMT only when the washout period exceeded 26 days. Neither FING nor other previous DMT reduced the effectiveness of RTX/OCR beyond the first 6 months of treatment.
Collapse
Affiliation(s)
- Lisa Graille-Avy
- From the APHM (L.G.-A., C.B., C.R., M.P., A.R., S.D., F.H., J.P., A.M., B.A.), Hôpital de la Timone, Pôle de Neurosciences Cliniques, Service de Neurologie; Aix-Marseille University (A.R., J.P., A.M., B.A.), CNRS, CRMBM, Marseille; Centre hospitalier d'Ajaccio (P.D.), Service de Neurologie, Ajaccio; APHM (F.V.), Hôpital de la Timone, Service d'immunologie, Marseille Immunopôle; Aix Marseille University (F.V.), CNRS, INSERM, CIML; and APHM (P.B.-P.), Hôpital de la Timone, Service Pharmacie, Marseille, France
| | - Clemence Boutiere
- From the APHM (L.G.-A., C.B., C.R., M.P., A.R., S.D., F.H., J.P., A.M., B.A.), Hôpital de la Timone, Pôle de Neurosciences Cliniques, Service de Neurologie; Aix-Marseille University (A.R., J.P., A.M., B.A.), CNRS, CRMBM, Marseille; Centre hospitalier d'Ajaccio (P.D.), Service de Neurologie, Ajaccio; APHM (F.V.), Hôpital de la Timone, Service d'immunologie, Marseille Immunopôle; Aix Marseille University (F.V.), CNRS, INSERM, CIML; and APHM (P.B.-P.), Hôpital de la Timone, Service Pharmacie, Marseille, France
| | - Camille Rigollet
- From the APHM (L.G.-A., C.B., C.R., M.P., A.R., S.D., F.H., J.P., A.M., B.A.), Hôpital de la Timone, Pôle de Neurosciences Cliniques, Service de Neurologie; Aix-Marseille University (A.R., J.P., A.M., B.A.), CNRS, CRMBM, Marseille; Centre hospitalier d'Ajaccio (P.D.), Service de Neurologie, Ajaccio; APHM (F.V.), Hôpital de la Timone, Service d'immunologie, Marseille Immunopôle; Aix Marseille University (F.V.), CNRS, INSERM, CIML; and APHM (P.B.-P.), Hôpital de la Timone, Service Pharmacie, Marseille, France
| | - Marine Perriguey
- From the APHM (L.G.-A., C.B., C.R., M.P., A.R., S.D., F.H., J.P., A.M., B.A.), Hôpital de la Timone, Pôle de Neurosciences Cliniques, Service de Neurologie; Aix-Marseille University (A.R., J.P., A.M., B.A.), CNRS, CRMBM, Marseille; Centre hospitalier d'Ajaccio (P.D.), Service de Neurologie, Ajaccio; APHM (F.V.), Hôpital de la Timone, Service d'immunologie, Marseille Immunopôle; Aix Marseille University (F.V.), CNRS, INSERM, CIML; and APHM (P.B.-P.), Hôpital de la Timone, Service Pharmacie, Marseille, France
| | - Audrey Rico
- From the APHM (L.G.-A., C.B., C.R., M.P., A.R., S.D., F.H., J.P., A.M., B.A.), Hôpital de la Timone, Pôle de Neurosciences Cliniques, Service de Neurologie; Aix-Marseille University (A.R., J.P., A.M., B.A.), CNRS, CRMBM, Marseille; Centre hospitalier d'Ajaccio (P.D.), Service de Neurologie, Ajaccio; APHM (F.V.), Hôpital de la Timone, Service d'immunologie, Marseille Immunopôle; Aix Marseille University (F.V.), CNRS, INSERM, CIML; and APHM (P.B.-P.), Hôpital de la Timone, Service Pharmacie, Marseille, France
| | - Sarah Demortiere
- From the APHM (L.G.-A., C.B., C.R., M.P., A.R., S.D., F.H., J.P., A.M., B.A.), Hôpital de la Timone, Pôle de Neurosciences Cliniques, Service de Neurologie; Aix-Marseille University (A.R., J.P., A.M., B.A.), CNRS, CRMBM, Marseille; Centre hospitalier d'Ajaccio (P.D.), Service de Neurologie, Ajaccio; APHM (F.V.), Hôpital de la Timone, Service d'immunologie, Marseille Immunopôle; Aix Marseille University (F.V.), CNRS, INSERM, CIML; and APHM (P.B.-P.), Hôpital de la Timone, Service Pharmacie, Marseille, France
| | - Pierre Durozard
- From the APHM (L.G.-A., C.B., C.R., M.P., A.R., S.D., F.H., J.P., A.M., B.A.), Hôpital de la Timone, Pôle de Neurosciences Cliniques, Service de Neurologie; Aix-Marseille University (A.R., J.P., A.M., B.A.), CNRS, CRMBM, Marseille; Centre hospitalier d'Ajaccio (P.D.), Service de Neurologie, Ajaccio; APHM (F.V.), Hôpital de la Timone, Service d'immunologie, Marseille Immunopôle; Aix Marseille University (F.V.), CNRS, INSERM, CIML; and APHM (P.B.-P.), Hôpital de la Timone, Service Pharmacie, Marseille, France
| | - Frederic Hilezian
- From the APHM (L.G.-A., C.B., C.R., M.P., A.R., S.D., F.H., J.P., A.M., B.A.), Hôpital de la Timone, Pôle de Neurosciences Cliniques, Service de Neurologie; Aix-Marseille University (A.R., J.P., A.M., B.A.), CNRS, CRMBM, Marseille; Centre hospitalier d'Ajaccio (P.D.), Service de Neurologie, Ajaccio; APHM (F.V.), Hôpital de la Timone, Service d'immunologie, Marseille Immunopôle; Aix Marseille University (F.V.), CNRS, INSERM, CIML; and APHM (P.B.-P.), Hôpital de la Timone, Service Pharmacie, Marseille, France
| | - Frederic Vely
- From the APHM (L.G.-A., C.B., C.R., M.P., A.R., S.D., F.H., J.P., A.M., B.A.), Hôpital de la Timone, Pôle de Neurosciences Cliniques, Service de Neurologie; Aix-Marseille University (A.R., J.P., A.M., B.A.), CNRS, CRMBM, Marseille; Centre hospitalier d'Ajaccio (P.D.), Service de Neurologie, Ajaccio; APHM (F.V.), Hôpital de la Timone, Service d'immunologie, Marseille Immunopôle; Aix Marseille University (F.V.), CNRS, INSERM, CIML; and APHM (P.B.-P.), Hôpital de la Timone, Service Pharmacie, Marseille, France
| | - Pierre Bertault-Peres
- From the APHM (L.G.-A., C.B., C.R., M.P., A.R., S.D., F.H., J.P., A.M., B.A.), Hôpital de la Timone, Pôle de Neurosciences Cliniques, Service de Neurologie; Aix-Marseille University (A.R., J.P., A.M., B.A.), CNRS, CRMBM, Marseille; Centre hospitalier d'Ajaccio (P.D.), Service de Neurologie, Ajaccio; APHM (F.V.), Hôpital de la Timone, Service d'immunologie, Marseille Immunopôle; Aix Marseille University (F.V.), CNRS, INSERM, CIML; and APHM (P.B.-P.), Hôpital de la Timone, Service Pharmacie, Marseille, France
| | - Jean Pelletier
- From the APHM (L.G.-A., C.B., C.R., M.P., A.R., S.D., F.H., J.P., A.M., B.A.), Hôpital de la Timone, Pôle de Neurosciences Cliniques, Service de Neurologie; Aix-Marseille University (A.R., J.P., A.M., B.A.), CNRS, CRMBM, Marseille; Centre hospitalier d'Ajaccio (P.D.), Service de Neurologie, Ajaccio; APHM (F.V.), Hôpital de la Timone, Service d'immunologie, Marseille Immunopôle; Aix Marseille University (F.V.), CNRS, INSERM, CIML; and APHM (P.B.-P.), Hôpital de la Timone, Service Pharmacie, Marseille, France
| | - Adil Maarouf
- From the APHM (L.G.-A., C.B., C.R., M.P., A.R., S.D., F.H., J.P., A.M., B.A.), Hôpital de la Timone, Pôle de Neurosciences Cliniques, Service de Neurologie; Aix-Marseille University (A.R., J.P., A.M., B.A.), CNRS, CRMBM, Marseille; Centre hospitalier d'Ajaccio (P.D.), Service de Neurologie, Ajaccio; APHM (F.V.), Hôpital de la Timone, Service d'immunologie, Marseille Immunopôle; Aix Marseille University (F.V.), CNRS, INSERM, CIML; and APHM (P.B.-P.), Hôpital de la Timone, Service Pharmacie, Marseille, France
| | - Bertrand Audoin
- From the APHM (L.G.-A., C.B., C.R., M.P., A.R., S.D., F.H., J.P., A.M., B.A.), Hôpital de la Timone, Pôle de Neurosciences Cliniques, Service de Neurologie; Aix-Marseille University (A.R., J.P., A.M., B.A.), CNRS, CRMBM, Marseille; Centre hospitalier d'Ajaccio (P.D.), Service de Neurologie, Ajaccio; APHM (F.V.), Hôpital de la Timone, Service d'immunologie, Marseille Immunopôle; Aix Marseille University (F.V.), CNRS, INSERM, CIML; and APHM (P.B.-P.), Hôpital de la Timone, Service Pharmacie, Marseille, France
| |
Collapse
|
37
|
Borghol AH, Bitar ER, Hanna A, Naim G, Rahal EA. The role of Epstein-Barr virus in autoimmune and autoinflammatory diseases. Crit Rev Microbiol 2024:1-21. [PMID: 38634723 DOI: 10.1080/1040841x.2024.2344114] [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: 01/16/2024] [Accepted: 04/11/2024] [Indexed: 04/19/2024]
Abstract
Epstein-Barr Virus (EBV), a dsDNA herpesvirus, is believed to play a significant role in exacerbating and potentially triggering autoimmune and autoinflammatory maladies. Around 90% of the world is infected with the virus, which establishes latency within lymphocytes. EBV is also known to cause infectious mononucleosis, a self-limited flu-like illness, in adolescents. EBV is often reactivated and it employs several mechanisms of evading the host immune system. It has also been implicated in inducing host immune dysfunction potentially resulting in exacerbation or triggering of inflammatory processes. EBV has therefore been linked to a number of autoimmune diseases, including systemic lupus erythematosus, multiple sclerosis, rheumatoid arthritis, and Sjögren's syndrome. The review examines the molecular mechanisms through which the virus alters host immune system components thus possibly resulting in autoimmune processes. Understanding the mechanisms underpinning EBV-associated autoimmunity is pivotal; however, the precise causal pathways remain elusive. Research on therapeutic agents and vaccines for EBV has been stagnant for a long number of years until recent advances shed light on potential therapeutic targets. The implications of EBV in autoimmunity underscore the importance of developing targeted therapeutic strategies and, potentially, vaccines to mitigate the autoimmune burden associated with this ubiquitous virus.
Collapse
Affiliation(s)
- Abdul Hamid Borghol
- Department of Experimental Pathology, Immunology and Microbiology, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
- Center for Infectious Diseases Research (CIDR), American University of Beirut, Beirut, Lebanon
| | - Elio R Bitar
- Department of Experimental Pathology, Immunology and Microbiology, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
- Center for Infectious Diseases Research (CIDR), American University of Beirut, Beirut, Lebanon
| | - Aya Hanna
- Department of Experimental Pathology, Immunology and Microbiology, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
- Center for Infectious Diseases Research (CIDR), American University of Beirut, Beirut, Lebanon
| | - Georges Naim
- Department of Experimental Pathology, Immunology and Microbiology, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
- Center for Infectious Diseases Research (CIDR), American University of Beirut, Beirut, Lebanon
| | - Elias A Rahal
- Department of Experimental Pathology, Immunology and Microbiology, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
- Center for Infectious Diseases Research (CIDR), American University of Beirut, Beirut, Lebanon
| |
Collapse
|
38
|
Ünlü S, Sánchez Navarro BG, Cakan E, Berchtold D, Meleka Hanna R, Vural S, Vural A, Meisel A, Fichtner ML. Exploring the depths of IgG4: insights into autoimmunity and novel treatments. Front Immunol 2024; 15:1346671. [PMID: 38698867 PMCID: PMC11063302 DOI: 10.3389/fimmu.2024.1346671] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Accepted: 02/29/2024] [Indexed: 05/05/2024] Open
Abstract
IgG4 subclass antibodies represent the rarest subclass of IgG antibodies, comprising only 3-5% of antibodies circulating in the bloodstream. These antibodies possess unique structural features, notably their ability to undergo a process known as fragment-antigen binding (Fab)-arm exchange, wherein they exchange half-molecules with other IgG4 antibodies. Functionally, IgG4 antibodies primarily block and exert immunomodulatory effects, particularly in the context of IgE isotype-mediated hypersensitivity reactions. In the context of disease, IgG4 antibodies are prominently observed in various autoimmune diseases combined under the term IgG4 autoimmune diseases (IgG4-AID). These diseases include myasthenia gravis (MG) with autoantibodies against muscle-specific tyrosine kinase (MuSK), nodo-paranodopathies with autoantibodies against paranodal and nodal proteins, pemphigus vulgaris and foliaceus with antibodies against desmoglein and encephalitis with antibodies against LGI1/CASPR2. Additionally, IgG4 antibodies are a prominent feature in the rare entity of IgG4 related disease (IgG4-RD). Intriguingly, both IgG4-AID and IgG4-RD demonstrate a remarkable responsiveness to anti-CD20-mediated B cell depletion therapy (BCDT), suggesting shared underlying immunopathologies. This review aims to provide a comprehensive exploration of B cells, antibody subclasses, and their general properties before examining the distinctive characteristics of IgG4 subclass antibodies in the context of health, IgG4-AID and IgG4-RD. Furthermore, we will examine potential therapeutic strategies for these conditions, with a special focus on leveraging insights gained from anti-CD20-mediated BCDT. Through this analysis, we aim to enhance our understanding of the pathogenesis of IgG4-mediated diseases and identify promising possibilities for targeted therapeutic intervention.
Collapse
Affiliation(s)
- Selen Ünlü
- Koç University Research Center for Translational Medicine (KUTTAM), İstanbul, Türkiye
- Koç University School of Medicine, Istanbul, Türkiye
| | - Blanca G. Sánchez Navarro
- Department of Neurology with Experimental Neurology, Integrated Myasthenia Gravis Center, Neuroscience Clinical Research Center, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Elif Cakan
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard University, Cambridge, MA, United States
| | - Daniel Berchtold
- Department of Neurology with Experimental Neurology, Integrated Myasthenia Gravis Center, Neuroscience Clinical Research Center, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Rafael Meleka Hanna
- Department of Neurology with Experimental Neurology, Integrated Myasthenia Gravis Center, Neuroscience Clinical Research Center, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Secil Vural
- Koç University Research Center for Translational Medicine (KUTTAM), İstanbul, Türkiye
- Department of Dermatology and Venereology, Koç University School of Medicine, İstanbul, Türkiye
| | - Atay Vural
- Koç University Research Center for Translational Medicine (KUTTAM), İstanbul, Türkiye
- Department of Neurology, Koç University School of Medicine, İstanbul, Türkiye
| | - Andreas Meisel
- Department of Neurology with Experimental Neurology, Integrated Myasthenia Gravis Center, Neuroscience Clinical Research Center, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Miriam L. Fichtner
- Koç University Research Center for Translational Medicine (KUTTAM), İstanbul, Türkiye
- Department of Neurology with Experimental Neurology, Integrated Myasthenia Gravis Center, Neuroscience Clinical Research Center, Charité Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
39
|
Elgenidy A, Abdelhalim NN, Al-kurdi MAM, Mohamed LA, Ghoneim MM, Fathy AW, Hassaan HK, Anan A, Alomari O. Hypogammaglobulinemia and infections in patients with multiple sclerosis treated with anti-CD20 treatments: a systematic review and meta-analysis of 19,139 multiple sclerosis patients. Front Neurol 2024; 15:1380654. [PMID: 38699050 PMCID: PMC11063306 DOI: 10.3389/fneur.2024.1380654] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Accepted: 04/03/2024] [Indexed: 05/05/2024] Open
Abstract
Background Recent years have seen the emergence of disease-modifying therapies in multiple sclerosis (MS), such as anti-cluster of differentiation 20 (anti-CD20) monoclonal antibodies, aiming to modulate the immune response and effectively manage MS. However, the relationship between anti-CD20 treatments and immunoglobulin G (IgG) levels, particularly the development of hypogammaglobulinemia and subsequent infection risks, remains a subject of scientific interest and variability. We aimed to investigate the intricate connection between anti-CD20 MS treatments, changes in IgG levels, and the associated risk of hypogammaglobulinemia and subsequent infections. Method PubMed, Scopus, Embase, Cochrane, and Web of Science databases have been searched for relevant studies. The "R" software utilized to analyze the occurrence of hypogammaglobulinemia, infections and mean differences in IgG levels pre- and post-treatment. The subgrouping analyses were done based on drug type and treatment duration. The assessment of heterogeneity utilized the I2 and chi-squared tests, applying the random effect model. Results Thirty-nine articles fulfilled our inclusion criteria and were included in our review which included a total of 20,501 MS patients. The overall prevalence rate of hypogammaglobulinemia was found to be 11% (95% CI: 0.08 to 0.15). Subgroup analysis based on drug type revealed varying prevalence rates, with rituximab showing the highest at 18%. Subgroup analysis based on drug usage duration revealed that the highest proportion of hypogammaglobulinemia occurred in individuals taking the drugs for 1 year or less (19%). The prevalence of infections in MS patients with a focus on different infection types stratified by the MS drug used revealed that pulmonary infections were the most prevalent (9%) followed by urinary tract infections (6%), gastrointestinal infections (2%), and skin and mucous membrane infections (2%). Additionally, a significant decrease in mean IgG levels after treatment compared to before treatment, with a mean difference of 0.57 (95% CI: 0.22 to 0.93). Conclusion This study provides a comprehensive analysis of the impact of anti-CD20 drugs on serum IgG levels in MS patients, exploring the prevalence of hypogammaglobulinemia, based on different drug types, treatment durations, and infection patterns. The identified rates and patterns offer a foundation for clinicians to consider in their risk-benefit. Systematic review registration https://www.crd.york.ac.uk/prospero/display_record.php?RecordID=518239, CRD42024518239.
Collapse
Affiliation(s)
- Anas Elgenidy
- Faculty of Medicine, Cairo University, Giza, Egypt
- Karl-Jaspers-Klinik, Bad Zwischenahn, Germany
| | | | | | | | | | | | | | - Ahmed Anan
- Faculty of Medicine, Suez Canal University, Ismailia, Egypt
| | - Omar Alomari
- Hamidiye International School of Medicine, University of Health Sciences, Istanbul, Türkiye
| |
Collapse
|
40
|
Freeman SA, Zéphir H. Anti-CD20 monoclonal antibodies in multiple sclerosis: Rethinking the current treatment strategy. Rev Neurol (Paris) 2024:S0035-3787(24)00474-0. [PMID: 38599976 DOI: 10.1016/j.neurol.2023.12.013] [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: 09/22/2023] [Revised: 12/17/2023] [Accepted: 12/22/2023] [Indexed: 04/12/2024]
Abstract
Anti-CD20 monoclonal antibodies are highly-effective B-cell-depleting therapies in multiple sclerosis (MS). These treatments have expanded the arsenal of highly effective disease-modifying therapies, and have changed the landscape in understanding the pathophysiology of MS and the natural course of the disease. Nevertheless, these treatments come at the cost of immunosuppression and risk of serious infections, diminished vaccination response and treatment-related secondary hypogammaglobulinemia. However, the COVID pandemic has given way to a possibility of readapting these therapies, with most notably extended dosing intervals. While these new strategies show efficacy in maintaining inflammatory MS disease control, and although it is tempting to speculate that tailoring CD20 therapies will reduce the negative outcomes of long-term immunosuppression, it is unknown whether they provide meaningful benefit in reducing the risk of treatment-related secondary hypogammaglobulinemia and serious infections. This review highlights the available anti-CD20 therapies that are available for treating MS patients, and sheds light on encouraging data, which propose that tailoring anti-CD20 monoclonal antibodies is the next step in rethinking the current treatment strategy.
Collapse
Affiliation(s)
- S A Freeman
- Department of Neurology, CRC-SEP, CHU of Toulouse, Toulouse, France; University Toulouse III, Inserm UMR1291, CHU Purpan, Toulouse Institute for Infectious and Inflammatory Diseases (INFINITY), 59000 Toulouse, France.
| | - H Zéphir
- Department of Neurology, CRC-SEP, CHU of Lille, Lille, France; University of Lille, Inserm, CHU of Lille, Laboratory of Neuroinflammation and Multiple Sclerosis (NEMESIS), U1172, Lille, France
| |
Collapse
|
41
|
Engler-Chiurazzi E. B cells and the stressed brain: emerging evidence of neuroimmune interactions in the context of psychosocial stress and major depression. Front Cell Neurosci 2024; 18:1360242. [PMID: 38650657 PMCID: PMC11033448 DOI: 10.3389/fncel.2024.1360242] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Accepted: 03/25/2024] [Indexed: 04/25/2024] Open
Abstract
The immune system has emerged as a key regulator of central nervous system (CNS) function in health and in disease. Importantly, improved understanding of immune contributions to mood disorders has provided novel opportunities for the treatment of debilitating stress-related mental health conditions such as major depressive disorder (MDD). Yet, the impact to, and involvement of, B lymphocytes in the response to stress is not well-understood, leaving a fundamental gap in our knowledge underlying the immune theory of depression. Several emerging clinical and preclinical findings highlight pronounced consequences for B cells in stress and MDD and may indicate key roles for B cells in modulating mood. This review will describe the clinical and foundational observations implicating B cell-psychological stress interactions, discuss potential mechanisms by which B cells may impact brain function in the context of stress and mood disorders, describe research tools that support the investigation of their neurobiological impacts, and highlight remaining research questions. The goal here is for this discussion to illuminate both the scope and limitations of our current understanding regarding the role of B cells, stress, mood, and depression.
Collapse
Affiliation(s)
- Elizabeth Engler-Chiurazzi
- Department of Neurosurgery and Neurology, Clinical Neuroscience Research Center, Tulane Brain Institute, Tulane University School of Medicine, New Orleans, LA, United States
| |
Collapse
|
42
|
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: 0] [Impact Index Per Article: 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
|
43
|
Mescia F, Salviani C, Tonoli M, Affatato S, Moratto D, Tedesco M, Guerini A, Gemmo A, Camoni M, Delbarba E, Zubani R, Garrafa E, Chiarini M, Gregorini G, Scolari F, Alberici F. Sustained post-rituximab B-cell depletion is common in ANCA-associated vasculitis and is affected by sex and renal function. Nephrol Dial Transplant 2024; 39:683-693. [PMID: 37673675 DOI: 10.1093/ndt/gfad197] [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: 05/16/2023] [Indexed: 09/08/2023] Open
Abstract
OBJECTIVE Despite the increasing use of rituximab in anti-neutrophil cytoplasmic antibody (ANCA)-associated vasculitis (AAV), it remains unclear what the optimal dosing is, especially for maintenance of remission. A deeper understanding of post-rituximab B-cell repopulation patterns may aid better-tailored treatment. METHODS This is a monocentric, retrospective study including ANCA-positive AAV patients receiving a single course of rituximab induction. CD19+ B cells were longitudinally monitored with flow cytometry. B-cell repopulation was defined as CD19+ >10 cells/μL. RESULTS Seventy-one patients were included, the majority with microscopic polyangiitis (75%), myeloperoxidase-ANCA positivity (75%) and with renal involvement (79%). During a median follow-up of 54 months since the first rituximab infusion, 44 patients (62%) repopulated B cells, with a median time to repopulation of 39 months (range 7-102). Patients experiencing B-cell depletion lasting longer than the overall median time to repopulation (39 months) exhibited a lower risk of flare and higher risk of serious infection. In multivariate Cox regression, higher estimated glomerular filtration rate (eGFR) [hazard ratio (HR) 1.84, 95% confidence interval (CI) 1.13-2.98 per 30 mL/min/1.73 m2 eGFR] and female sex (HR 2.70, 95% CI 1.37-5.31) were independent predictors of increased rate of B-cell repopulation. CONCLUSION A subset of AAV patients develop sustained post-rituximab B-cell depletion, which associates with reduced risk of flare and increased risk of serious infection in the long term. Preserved renal function and female sex are associated with faster B-cell repopulation. These observations further highlight the need to personalize immunosuppression to improve clinical outcomes.
Collapse
Affiliation(s)
- Federica Mescia
- Department of Medical and Surgical Specialties, Radiological Sciences and Public Health, University of Brescia, Brescia, Italy
- Nephrology Unit, Spedali Civili Hospital, ASST Spedali Civili of Brescia, Brescia, Italy
| | - Chiara Salviani
- Nephrology Unit, Spedali Civili Hospital, ASST Spedali Civili of Brescia, Brescia, Italy
| | - Mattia Tonoli
- Nephrology Unit, Spedali Civili Hospital, ASST Spedali Civili of Brescia, Brescia, Italy
| | - Stefania Affatato
- Nephrology Unit, Spedali Civili Hospital, ASST Spedali Civili of Brescia, Brescia, Italy
| | - Daniele Moratto
- Flow Cytometry Laboratory, Department of Laboratory Diagnostics, ASST Spedali Civili di Brescia, Brescia, Italy
| | - Martina Tedesco
- Department of Medical and Surgical Specialties, Radiological Sciences and Public Health, University of Brescia, Brescia, Italy
- Nephrology Unit, Spedali Civili Hospital, ASST Spedali Civili of Brescia, Brescia, Italy
| | - Alice Guerini
- Department of Medical and Surgical Specialties, Radiological Sciences and Public Health, University of Brescia, Brescia, Italy
- Nephrology Unit, Spedali Civili Hospital, ASST Spedali Civili of Brescia, Brescia, Italy
| | - Alessia Gemmo
- Department of Medical and Surgical Specialties, Radiological Sciences and Public Health, University of Brescia, Brescia, Italy
- Nephrology Unit, Spedali Civili Hospital, ASST Spedali Civili of Brescia, Brescia, Italy
| | - Marta Camoni
- Department of Medical and Surgical Specialties, Radiological Sciences and Public Health, University of Brescia, Brescia, Italy
- Nephrology Unit, Spedali Civili Hospital, ASST Spedali Civili of Brescia, Brescia, Italy
| | - Elisa Delbarba
- Nephrology Unit, Spedali Civili Hospital, ASST Spedali Civili of Brescia, Brescia, Italy
| | - Roberto Zubani
- Department of Medical and Surgical Specialties, Radiological Sciences and Public Health, University of Brescia, Brescia, Italy
- Nephrology Unit, Spedali Civili Hospital, ASST Spedali Civili of Brescia, Brescia, Italy
| | - Emirena Garrafa
- Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
- Department of Laboratory Diagnostics, ASST Spedali Civili, Brescia, Italy
| | - Marco Chiarini
- Flow Cytometry Laboratory, Department of Laboratory Diagnostics, ASST Spedali Civili di Brescia, Brescia, Italy
| | | | - Francesco Scolari
- Department of Medical and Surgical Specialties, Radiological Sciences and Public Health, University of Brescia, Brescia, Italy
- Nephrology Unit, Spedali Civili Hospital, ASST Spedali Civili of Brescia, Brescia, Italy
| | - Federico Alberici
- Department of Medical and Surgical Specialties, Radiological Sciences and Public Health, University of Brescia, Brescia, Italy
- Nephrology Unit, Spedali Civili Hospital, ASST Spedali Civili of Brescia, Brescia, Italy
| |
Collapse
|
44
|
Galetta K, Ham AS, Vishnevetsky A, Bhattacharyya S, Mateen FJ. Disease modifying therapy in the treatment of tumefactive multiple sclerosis: A retrospective cohort study. J Neuroimmunol 2024; 388:578299. [PMID: 38364529 DOI: 10.1016/j.jneuroim.2024.578299] [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: 11/15/2023] [Revised: 01/11/2024] [Accepted: 01/25/2024] [Indexed: 02/18/2024]
Abstract
Tumefactive multiple sclerosis (TMS) is characterized by large demyelinating brain lesions. This was a retrospective cohort study of 67 patients with TMS between January 2015-2023, examining different disease modifying therapy impact on expanded disability scale score change at follow-up. Median age was 36 with a female predominance. Mean EDSS was 3.3 ± 2.3 at TMS onset, 2.1 ± 1.9 at year one, and 2.1 ± 1.9 at last follow-up. A multilinear regression model found higher presentation EDSS and post-diagnosis non-B-cell high efficacy therapies were each independently associated with higher EDSS at last follow up. Further research is needed to determine the value of B-cell therapy in TMS.
Collapse
Affiliation(s)
- Kristin Galetta
- Department of Neurology, Stanford University, Palo Alto, CA, USA; Department of Neurology, Brigham and Women's Hospital, Boston, MA, USA.
| | - Andrew Siyoon Ham
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA.
| | | | | | - Farrah J Mateen
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA.
| |
Collapse
|
45
|
Wu X, Wang S, Xue T, Tan X, Li J, Chen Z, Wang Z. Disease-modifying therapy in progressive multiple sclerosis: a systematic review and network meta-analysis of randomized controlled trials. Front Neurol 2024; 15:1295770. [PMID: 38529035 PMCID: PMC10962394 DOI: 10.3389/fneur.2024.1295770] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Accepted: 02/06/2024] [Indexed: 03/27/2024] Open
Abstract
Background Currently, disease-modifying therapies (DMTs) for progressive multiple sclerosis (PMS) are widely used in clinical practice. At the same time, there are a variety of drug options for DMTs, but the effect of the drugs that can better relieve symptoms and improve the prognosis are still inconclusive. Objectives This systematic review aimed to evaluate the efficacy and safety of DMTs for PMS and to identify the best among these drugs. Methods MEDLINE, EMBASE, the Cochrane Library, and clinicaltrials.gov were systematically searched to identify relevant studies published before 30 January, 2023. We assessed the certainty of the evidence using the confidence in the network meta-analysis (CINeMA) framework. We estimated the summary risk ratio (RR) for dichotomous outcomes and mean differences (MD) for continuous outcomes with 95% credible intervals (CrIs). Results We included 18 randomized controlled trials (RCTs) involving 9,234 patients in the study. DMT can effectively control the disease progression of MS. Among them, mitoxantrone, siponimod, and ocrelizumab are superior to other drug options in delaying disease progression (high certainty). Mitoxantrone was the best (with high certainty) for mitigating deterioration (progression of disability). Ocrelizumab performed best on the pre- and post-treatment Timed 25-Foot Walk test (T25FW; low certainty), as did all other agents (RR range: 1.12-1.05). In the 9-Hole Peg Test (9HPT), natalizumab performed the best (high certainty), as did all other agents (RR range: 1.59-1.09). In terms of imaging, IFN-beta-1b performed better on the new T2 hypointense lesion on contrast, before and after treatment (high certainty), while siponimod performed best on the change from baseline in the total volume of lesions on T2-weighted image contrast before and after treatment (high certainty), and sWASO had the highest area under the curve (SUCRA) value (100%). In terms of adverse events (AEs), rituximab (RR 1.01), and laquinimod (RR 1.02) were more effective than the placebo (high certainty). In terms of serious adverse events (SAEs), natalizumab (RR 1.09), and ocrelizumab (RR 1.07) were safer than placebo (high certainty). Conclusion DMTs can effectively control disease progression and reduce disease deterioration during the treatment of PMS. Systematic review registration https://inplasy.com/?s=202320071, identifier: 202320071.
Collapse
Affiliation(s)
- Xin Wu
- Department of Neurosurgery and Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Shixin Wang
- Department of Neurosurgery and Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Tao Xue
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Xin Tan
- Department of Neurology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Suzhou, Jiangsu, China
| | - Jiaxuan Li
- Department of Neurosurgery and Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Zhouqing Chen
- Department of Neurosurgery and Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Zhong Wang
- Department of Neurosurgery and Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| |
Collapse
|
46
|
Yamout B, Al-Jumah M, Sahraian MA, Almalik Y, Khaburi JA, Shalaby N, Aljarallah S, Bohlega S, Dahdaleh M, Almahdawi A, Khoury SJ, Koussa S, Slassi E, Daoudi S, Aref H, Mrabet S, Zeineddine M, Zakaria M, Inshasi J, Gouider R, Alroughani R. Consensus recommendations for diagnosis and treatment of Multiple Sclerosis: 2023 revision of the MENACTRIMS guidelines. Mult Scler Relat Disord 2024; 83:105435. [PMID: 38245998 DOI: 10.1016/j.msard.2024.105435] [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: 10/26/2023] [Revised: 12/21/2023] [Accepted: 01/06/2024] [Indexed: 01/23/2024]
Abstract
With evolving diagnostic criteria and the advent of new oral and parenteral therapies for Multiple Sclerosis (MS), most current diagnostic and treatment algorithms need revision and updating. The diagnosis of MS relies on incorporating clinical and paraclinical findings to prove dissemination in space and time and exclude alternative diseases that can explain the findings at hand. The differential diagnostic workup should be guided by clinical and laboratory red flags to avoid unnecessary tests. Appropriate selection of MS therapies is critical to maximize patient benefit. The current guidelines review the current diagnostic criteria for MS and the scientific evidence supporting treatment of acute relapses, radiologically isolated syndrome, clinically isolated syndrome, relapsing remitting MS, progressive MS, pediatric cases and pregnant women. The purpose of these guidelines is to provide practical recommendations and algorithms for the diagnosis and treatment of MS based on current scientific evidence and clinical experience.
Collapse
Affiliation(s)
- B Yamout
- Neurology Institute and Multiple Sclerosis Center, Harley Street Medical Center, Abu Dhabi, United Arab Emirates.
| | - M Al-Jumah
- InterHealth hospital, Multiple Sclerosis Center, Riyadh, Saudi Arabia
| | - M A Sahraian
- Multiple Sclerosis Research Center, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Y Almalik
- Division of Neurology, College of Medicine, King Saud bin Abdulaziz University for Health Sciences, King Abdullah International Medical Research Center, National Guard Health Affairs, King Abdulaziz Medical City, Riyadh, Saudi Arabia
| | - J Al Khaburi
- Department of Neurology, The Royal Hospital, Sultanate of Oman
| | - N Shalaby
- Neurology Department, Kasr Al-Ainy School of Medicine, Cairo University, Cairo, Egypt
| | | | - S Bohlega
- King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | | | - A Almahdawi
- Consultant Neurologist, Neurology Unit, Baghdad Teaching Hospital, Medical City Complex, Iraq
| | - S J Khoury
- Nehme and Therese Tohme Multiple Sclerosis Center, American University of Beirut Medical Center, Beirut, Lebanon
| | - S Koussa
- Multiple Sclerosis Center, Geitaoui Lebanese University Hospital, Beirut, Lebanon
| | - E Slassi
- Hôpital Cheikh Khalifa Ibn Zaid, Casablanca, Morocco
| | - S Daoudi
- Hospital Center Nedir Mohamed, Faculty of Medicine, University Mouloud Mammeri Tizi-Ouzou, Algeria
| | - H Aref
- Neurology Department, Ain Shams University, Cairo, Egypt
| | - S Mrabet
- Department of Neurology, CIC, Razi Universitary Hospital, University of Tunis El Manar, Tunis, Tunisia
| | - M Zeineddine
- Middle East and North Africa Committee for Treatment and Research in Multiple Sclerosis (MENACTRIMS), Beirut, Lebanon
| | | | - J Inshasi
- Department of Neurology, Rashid Hospital and Dubai Medical College, Dubai Health Authority, Dubai, United Arab Emirates
| | - R Gouider
- Department of Neurology, CIC, Razi Universitary Hospital, University of Tunis El Manar, Tunis, Tunisia
| | - R Alroughani
- Amiri Hospital, Arabian Gulf Street, Sharq, Kuwait
| |
Collapse
|
47
|
Carlson AK, Amin M, Cohen JA. Drugs Targeting CD20 in Multiple Sclerosis: Pharmacology, Efficacy, Safety, and Tolerability. Drugs 2024; 84:285-304. [PMID: 38480630 PMCID: PMC10982103 DOI: 10.1007/s40265-024-02011-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/14/2024] [Indexed: 04/02/2024]
Abstract
Currently, there are four monoclonal antibodies (mAbs) that target the cluster of differentiation (CD) 20 receptor available to treat multiple sclerosis (MS): rituximab, ocrelizumab, ofatumumab, and ublituximab. B-cell depletion therapy has changed the therapeutic landscape of MS through robust efficacy on clinical manifestations and MRI lesion activity, and the currently available anti-CD20 mAb therapies for use in MS are a cornerstone of highly effective disease-modifying treatment. Ocrelizumab is currently the only therapy with regulatory approval for primary progressive MS. There are currently few data regarding the relative efficacy of these therapies, though several clinical trials are ongoing. Safety concerns applicable to this class of therapeutics relate primarily to immunogenicity and mechanism of action, and include infusion-related or injection-related reactions, development of hypogammaglobulinemia (leading to increased infection and malignancy risk), and decreased vaccine response. Exploration of alternative dose/dosing schedules might be an effective strategy for mitigating these risks. Future development of biosimilar medications might make these therapies more readily available. Although anti-CD20 mAb therapies have led to significant improvements in disease outcomes, CNS-penetrant therapies are still needed to more effectively address the compartmentalized inflammation thought to play an important role in disability progression.
Collapse
Affiliation(s)
- Alise K Carlson
- Mellen Center, Neurologic Institute, Cleveland Clinic, 9500 Euclid Ave U10, Cleveland, OH, 44195, USA
| | - Moein Amin
- Mellen Center, Neurologic Institute, Cleveland Clinic, 9500 Euclid Ave U10, Cleveland, OH, 44195, USA
| | - Jeffrey A Cohen
- Mellen Center, Neurologic Institute, Cleveland Clinic, 9500 Euclid Ave U10, Cleveland, OH, 44195, USA.
| |
Collapse
|
48
|
Peng Y, Yang H, Chen Q, Jin H, Xue YH, Du MQ, Liu S, Yao SY. An angel or a devil? Current view on the role of CD8 + T cells in the pathogenesis of myasthenia gravis. J Transl Med 2024; 22:183. [PMID: 38378668 PMCID: PMC10877804 DOI: 10.1186/s12967-024-04965-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Accepted: 02/07/2024] [Indexed: 02/22/2024] Open
Abstract
BACKGROUND Myasthenia gravis (MG) and the experimental autoimmune MG (EAMG) animal model are characterized by T-cell-induced and B-cell-dominated autoimmune diseases that affect the neuromuscular junction. Several subtypes of CD4+ T cells, including T helper (Th) 17 cells, follicular Th cells, and regulatory T cells (Tregs), contribute to the pathogenesis of MG. However, increasing evidence suggests that CD8+ T cells also play a critical role in the pathogenesis and treatment of MG. MAIN BODY Herein, we review the literature on CD8+ T cells in MG, focusing on their potential effector and regulatory roles, as well as on relevant evidence (peripheral, in situ, cerebrospinal fluid, and under different treatments), T-cell receptor usage, cytokine and chemokine expression, cell marker expression, and Treg, Tc17, CD3+CD8+CD20+ T, and CXCR5+ CD8+ T cells. CONCLUSIONS Further studies on CD8+ T cells in MG are necessary to determine, among others, the real pattern of the Vβ gene usage of autoantigen-specific CD8+ cells in patients with MG, real images of the physiology and function of autoantigen-specific CD8+ cells from MG/EAMG, and the subset of autoantigen-specific CD8+ cells (Tc1, Tc17, and IL-17+IFN-γ+CD8+ T cells). There are many reports of CD20-expressing T (or CD20 + T) and CXCR5+ CD8 T cells on autoimmune diseases, especially on multiple sclerosis and rheumatoid arthritis. Unfortunately, up to now, there has been no report on these T cells on MG, which might be a good direction for future studies.
Collapse
Affiliation(s)
- Yong Peng
- Department of Neurology, Affiliated First Hospital of Hunan Traditional Chinese Medical College, Zhuzhou, 412000, Hunan, China.
- Department of Neurology, The Third Affiliated Hospital of Hunan University of Chinese Medicine, Zhuzhou, 412000, Hunan, China.
| | - Huan Yang
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Quan Chen
- Department of Neurology, Affiliated First Hospital of Hunan Traditional Chinese Medical College, Zhuzhou, 412000, Hunan, China
- Department of Neurology, The Third Affiliated Hospital of Hunan University of Chinese Medicine, Zhuzhou, 412000, Hunan, China
| | - Hong Jin
- Department of Neurology, Affiliated First Hospital of Hunan Traditional Chinese Medical College, Zhuzhou, 412000, Hunan, China
- Department of Neurology, The Third Affiliated Hospital of Hunan University of Chinese Medicine, Zhuzhou, 412000, Hunan, China
| | - Ya-Hui Xue
- Department of Neurology, Affiliated First Hospital of Hunan Traditional Chinese Medical College, Zhuzhou, 412000, Hunan, China
- Department of Neurology, The Third Affiliated Hospital of Hunan University of Chinese Medicine, Zhuzhou, 412000, Hunan, China
| | - Miao-Qiao Du
- Department of Neurology, Affiliated First Hospital of Hunan Traditional Chinese Medical College, Zhuzhou, 412000, Hunan, China
- Department of Neurology, The Third Affiliated Hospital of Hunan University of Chinese Medicine, Zhuzhou, 412000, Hunan, China
| | - Shu Liu
- Department of Neurology, Affiliated First Hospital of Hunan Traditional Chinese Medical College, Zhuzhou, 412000, Hunan, China
- Department of Neurology, The Third Affiliated Hospital of Hunan University of Chinese Medicine, Zhuzhou, 412000, Hunan, China
| | - Shun-Yu Yao
- Department of Neurology, Affiliated First Hospital of Hunan Traditional Chinese Medical College, Zhuzhou, 412000, Hunan, China
- Department of Neurology, The Third Affiliated Hospital of Hunan University of Chinese Medicine, Zhuzhou, 412000, Hunan, China
| |
Collapse
|
49
|
Xu X, Han Y, Zhang B, Ren Q, Ma J, Liu S. Understanding immune microenvironment alterations in the brain to improve the diagnosis and treatment of diverse brain diseases. Cell Commun Signal 2024; 22:132. [PMID: 38368403 PMCID: PMC10874090 DOI: 10.1186/s12964-024-01509-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Accepted: 02/01/2024] [Indexed: 02/19/2024] Open
Abstract
Abnormal inflammatory states in the brain are associated with a variety of brain diseases. The dynamic changes in the number and function of immune cells in cerebrospinal fluid (CSF) are advantageous for the early prediction and diagnosis of immune diseases affecting the brain. The aggregated factors and cells in inflamed CSF may represent candidate targets for therapy. The physiological barriers in the brain, such as the blood‒brain barrier (BBB), establish a stable environment for the distribution of resident immune cells. However, the underlying mechanism by which peripheral immune cells migrate into the brain and their role in maintaining immune homeostasis in CSF are still unclear. To advance our understanding of the causal link between brain diseases and immune cell status, we investigated the characteristics of immune cell changes in CSF and the molecular mechanisms involved in common brain diseases. Furthermore, we summarized the diagnostic and treatment methods for brain diseases in which immune cells and related cytokines in CSF are used as targets. Further investigations of the new immune cell subtypes and their contributions to the development of brain diseases are needed to improve diagnostic specificity and therapy.
Collapse
Affiliation(s)
- Xiaotong Xu
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing, 100085, People's Republic of China
- University of Chinese Academy of Sciences, Beijing, 100049, People's Republic of China
| | - Yi Han
- Guang'an Men Hospital, China Academy of Chinese Medical Sciences, Beijing, 100053, People's Republic of China.
| | - Binlong Zhang
- Guang'an Men Hospital, China Academy of Chinese Medical Sciences, Beijing, 100053, People's Republic of China
| | - Quanzhong Ren
- JST Sarcopenia Research Centre, National Center for Orthopaedics, Beijing Research Institute of Traumatology and Orthopaedics, Beijing Jishuitan Hospital, Capital Medical University, Beijing, 100035, People's Republic of China
| | - Juan Ma
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing, 100085, People's Republic of China.
- University of Chinese Academy of Sciences, Beijing, 100049, People's Republic of China.
| | - Sijin Liu
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing, 100085, People's Republic of China
- University of Chinese Academy of Sciences, Beijing, 100049, People's Republic of China
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, People's Republic of China
| |
Collapse
|
50
|
Ongphichetmetha T, Jitprapaikulsan J, Siritho S, Rattanathamsakul N, Detweeratham T, Prayoonwiwat N. Efficacy and safety of rituximab in multiple sclerosis and neuromyelitis optica spectrum disorder. Sci Rep 2024; 14:3503. [PMID: 38347079 PMCID: PMC10861443 DOI: 10.1038/s41598-024-53838-y] [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: 11/23/2023] [Accepted: 02/06/2024] [Indexed: 02/15/2024] Open
Abstract
In Thailand, resource limitations lead many multiple sclerosis (MS) and neuromyelitis optica spectrum disorder (NMOSD) patients to use off-label immunosuppressants. This study assesses the efficacy and safety of rituximab (RTX) with a CD19-based reinfusion regimen among Thai MS and NMOSD patients. A retrospective review of patients at the Faculty of Medicine Siriraj Hospital from January 1994 to April 2023 was conducted. The primary outcome assessed was the change in annualized relapse rate (ARR) for patients using RTX for over a year. Secondary outcomes included changes in the Expanded Disability Status Scale (EDSS) scores, time to the first relapse after RTX initiation for patients using RTX for over a year, and an evaluation of the safety of RTX. The study encompassed 36 MS and 39 NMOSD patients. A majority of patients (91.7% of MS and 79.5% of NMOSD) experienced no relapses during a median follow-up of 30 months (Interquartile range [IQR] 20-46) and 31 months (IQR 23-41), respectively. The median ARR significantly decreased in both MS (from 0.77 [IQR 0.42-1.83] to 0 [IQR 0-0], p < 0.001) and NMOSD (from 0.92 [IQR 0.68-1.78] to 0 [IQR 0-0.17], p < 0.001) patients after switching to RTX, with no difference between those following a fixed 6-month time point regimen and a CD19-based reinfusion regimen. Median EDSS scores improved significantly at the last follow-up visit in both groups. The mean time to the first subsequent relapse was 8.3 ± 3.0 months in MS and 6.8 ± 1.7 months in NMOSD. Mild adverse drug reactions occurred in 44% of patients. RTX effectively prevents relapses in Thai MS and NMOSD patients, with no observed serious adverse drug reactions.
Collapse
Affiliation(s)
- Tatchaporn Ongphichetmetha
- Division of Neurology, Department of Medicine, Faculty of Medicine Siriraj Hospital, Mahidol University, 2 Wanglang Rd, Siriraj, Bangkok noi, Bangkok, 10700, Thailand
- Siriraj Neuroimmunology Center, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, 10700, Thailand
- Division of Clinical Epidemiology, Department of Research and Development, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, 10700, Thailand
| | - Jiraporn Jitprapaikulsan
- Division of Neurology, Department of Medicine, Faculty of Medicine Siriraj Hospital, Mahidol University, 2 Wanglang Rd, Siriraj, Bangkok noi, Bangkok, 10700, Thailand
- Siriraj Neuroimmunology Center, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, 10700, Thailand
| | - Sasitorn Siritho
- Siriraj Neuroimmunology Center, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, 10700, Thailand
- Bumrungrad International Hospital, Bangkok, 10110, Thailand
| | - Natthapon Rattanathamsakul
- Division of Neurology, Department of Medicine, Faculty of Medicine Siriraj Hospital, Mahidol University, 2 Wanglang Rd, Siriraj, Bangkok noi, Bangkok, 10700, Thailand
- Siriraj Neuroimmunology Center, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, 10700, Thailand
| | - Thammachet Detweeratham
- Division of Neurology, Department of Medicine, Faculty of Medicine Siriraj Hospital, Mahidol University, 2 Wanglang Rd, Siriraj, Bangkok noi, Bangkok, 10700, Thailand
| | - Naraporn Prayoonwiwat
- Division of Neurology, Department of Medicine, Faculty of Medicine Siriraj Hospital, Mahidol University, 2 Wanglang Rd, Siriraj, Bangkok noi, Bangkok, 10700, Thailand.
- Siriraj Neuroimmunology Center, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, 10700, Thailand.
| |
Collapse
|