1
|
Cree BAC, Berger JR, Greenberg B. The Evolution of Anti-CD20 Treatment for Multiple Sclerosis: Optimization of Antibody Characteristics and Function. CNS Drugs 2025; 39:545-564. [PMID: 40180777 PMCID: PMC12058931 DOI: 10.1007/s40263-025-01182-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/16/2025] [Indexed: 04/05/2025]
Abstract
B-cell depletion with CD20-targeted agents is commonly used for treatment of multiple sclerosis (MS), other autoimmune diseases, and certain hematologic malignancies. Initial apparent success with rituximab in MS and neuromyelitis optica spurred development of the anti-CD20 monoclonal antibody (mAb) therapies ocrelizumab, ofatumumab, and ublituximab as well as the anti-CD19 mAb inebilizumab. While each are effective at targeting and depleting B cells, structural differences translate into different mechanisms of action affecting maintenance of B-cell depletion and safety and tolerability. Although the anti-CD20 mAbs differ in degree of human versus mouse sequences as well as target CD20 epitope, these properties do not appear to substantially affect activity or tolerability. In contrast, an antibody-dependent cell-mediated cytotoxicity (ADCC) versus a complement-dependent cytotoxicity mechanism of action as well as subcutaneous versus intravenous administration may provide improved tolerability. Glycoengineering of the mAbs ublituximab and inebilizumab enhances ADCC and can overcome the reduced responses to mAb-mediated B-cell depletion associated with certain genetic polymorphisms. Other strategies for therapeutic targeting of CD20, including brain shuttle antibodies (e.g., RO7121932), bispecific antibodies, chimeric antigen receptor T-cell therapies, and antibody-drug conjugates, are in active clinical development and may be future treatment approaches in MS and other B-cell-mediated autoimmune diseases.
Collapse
Affiliation(s)
- Bruce A C Cree
- Department of Neurology, UCSF Weill Institute for Neurosciences, University of California San Francisco, 675 Nelson Rising Lane, #221C, San Francisco, CA, 94158, USA.
| | - Joseph R Berger
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Benjamin Greenberg
- Department of Neurology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| |
Collapse
|
2
|
Li J, Dan Y, Su W, Zhao M, Chen Z, Zhao Z. Immune-related biomarkers in the neuromyelitis optica spectrum disorder; pathogenesis and therapeutic approaches. Exp Eye Res 2025; 256:110395. [PMID: 40274184 DOI: 10.1016/j.exer.2025.110395] [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: 02/21/2025] [Revised: 04/06/2025] [Accepted: 04/21/2025] [Indexed: 04/26/2025]
Abstract
Neuromyelitis optica spectrum disorder (NMOSD) is an autoimmune inflammatory disease of the central nervous system that mostly affects the optic nerves and spinal cord. About eighty percent of patients have antibodies that are directed against the water channel aquaporin-4 (AQP4)-IgG, which is expressed on astrocytes. This protein was shown to be both a biomarker and a pathogenic cause of NMOSD. Researchers have discovered that antibodies against myelin oligodendrocyte glycoprotein (MOG) IgG can serve as a biomarker for a distinct condition known as MOG antibody-associated disease (MOGAD). This condition shares some similarities with AQP4-IgG-positive NMOSD, but it has distinct differences in terms of its underlying causes, clinical characteristics, response to treatment, and prognosis. Identifying AQP4 antibodies in the blood serum confirms the diagnosis of seropositive NMOSD. Nevertheless, it remains uncertain if there is a correlation between AQP4-IgG levels and disease activity, severity, responsiveness to medication, or long-term effects. Furthermore, there is still a need to establish and confirm biomarkers specifically for patients diagnosed with seronegative NMOSD. This study primarily examines the immunological aspects of NMOSD, which might have significant consequences for clinical practice. These implications include the possible use of new biomarkers to aid in the early and correct diagnosis of NMOSD, as well as the development of current treatment options to enhance the long-term prognosis of NMOSD patients.
Collapse
Affiliation(s)
- Jingyong Li
- Department of Laboratory Pathology and Blood Transfusion, People's Liberation Army Joint Logistic Support Force 945th Hospital, Yaan, 625000, China
| | - Ya Dan
- Department of Laboratory Pathology and Blood Transfusion, People's Liberation Army Joint Logistic Support Force 945th Hospital, Yaan, 625000, China
| | - Wei Su
- Department of Medical Imaging, The Second People's Hospital of Yaan, 625000, China
| | - Mingjun Zhao
- Department of Laboratory Pathology and Blood Transfusion, People's Liberation Army Joint Logistic Support Force 945th Hospital, Yaan, 625000, China
| | - Zhiguo Chen
- People's Liberation Army Joint Logistic Support Force Rehabilitation Center, Da Lian, Yaan, 625000, China
| | - Zhuyang Zhao
- Department of Laboratory Pathology and Blood Transfusion, People's Liberation Army Joint Logistic Support Force 945th Hospital, Yaan, 625000, China.
| |
Collapse
|
3
|
Weiner HL. Immune mechanisms and shared immune targets in neurodegenerative diseases. Nat Rev Neurol 2025; 21:67-85. [PMID: 39681722 DOI: 10.1038/s41582-024-01046-7] [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: 11/15/2024] [Indexed: 12/18/2024]
Abstract
The immune system plays a major part in neurodegenerative diseases. In some, such as multiple sclerosis, it is the primary driver of the disease. In others, such as Alzheimer disease, amyotrophic lateral sclerosis and Parkinson disease, it has an amplifying role. Immunotherapeutic approaches that target the adaptive and innate immune systems are being explored for the treatment of almost all neurological diseases, and the targets and approaches are often common across diseases. Microglia are the primary immune cells in the brain that contribute to disease pathogenesis, and are consequently a common immune target for therapy. Other therapeutic approaches target components of the peripheral immune system, such as regulatory T cells and monocytes, which in turn act within the CNS. This Review considers in detail how microglia, monocytes and T cells contribute to the pathogenesis of multiple sclerosis, Alzheimer disease, amyotrophic lateral sclerosis and Parkinson disease, and their potential as shared therapeutic targets across these diseases. The microbiome is also highlighted as an emerging therapeutic target that indirectly modulates the immune system. Therapeutic approaches being developed to target immune function in neurodegenerative diseases are discussed, highlighting how immune-based approaches developed to treat one disease could be applicable to multiple other neurological diseases.
Collapse
Affiliation(s)
- Howard L Weiner
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
4
|
Proschinger S, Belen S, Adammek F, Schlagheck ML, Rademacher A, Schenk A, Warnke C, Bloch W, Zimmer P. Sportizumab - Multimodal progressive exercise over 10 weeks decreases Th17 frequency and CD49d expression on CD8 + T cells in relapsing-remitting multiple sclerosis: A randomized controlled trial. Brain Behav Immun 2025; 124:397-408. [PMID: 39675643 DOI: 10.1016/j.bbi.2024.12.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Revised: 12/07/2024] [Accepted: 12/12/2024] [Indexed: 12/17/2024] Open
Abstract
BACKGROUND Multiple Sclerosis (MS) represents a neuroinflammatory autoimmune disease characterized by the predominance of circulating T cell subsets with proinflammatory characteristics and increased central nervous system (CNS)-homing potential. Substantial evidence confirms various beneficial effects of chronic exercise interventions in MS, but it is unknown how long-term multi-modal intense exercise affects MS-associated lymphocytes that are commonly targeted by medication in persons with relapsing remitting MS (pwRRMS). METHODS A total of 45 participants with defined RRMS were randomized to either the exercise (n = 22) or passive waitlist-control group (n = 23). A 10-week intervention consisting of progressive resistance and strength-endurance exercises was applied (3x/week à 60 min). Blood was drawn before (T1) and after (T2) the intervention period. Flow cytometry was used for phenotyping lymphocyte subsets. RESULTS Relative protein expression of CD49d within CD8+ T cells, quantified via mean fluorescence intensity (MFI), is significantly associated with the Expanded Disability Status Scale (p = 0.007, r = 0.440), decreased in the exercise group (p = 0.001) only, and was significantly lower in the exercise compared to the control group at T2 (p < 0.001). T helper (Th) 17 cell frequency decreased only in the exercise group (p < 0.001). CD8+CD20+ T cell frequency was significantly lower in the exercise compared to the control group at T2 (p = 0.003), without showing significant time effects. CONCLUSION The 10-week multimodal exercise intervention mainly affected circulating T cells harboring a pathophysiological phenotype in MS. The findings of a decreased frequency of pathogenic Th17 cells and the reduced CNS-homing potential of CD8+ T cells, indicated by reduced CD49d MFI, substantiate the positive effects of exercise on cellular biomarkers involved in disease activity and progression in MS. To confirm exercise-mediated beneficial effects on both disease domains, clinical endpoints (i.e., relapse rate, lesion formation, EDSS score) should be assessed together with these cellular and molecular markers in studies with a larger sample size and a duration of six to twelve months or longer.
Collapse
Affiliation(s)
- Sebastian Proschinger
- TU Dortmund University, Institute for Sport and Sport Science, Division of Performance and Health (Sports Medicine), 44227 Dortmund, Germany
| | - Sergen Belen
- TU Dortmund University, Institute for Sport and Sport Science, Division of Performance and Health (Sports Medicine), 44227 Dortmund, Germany; Department for Molecular and Cellular Sports Medicine, Institute of Cardiovascular Research and Sports Medicine, German Sport University Cologne, Cologne, Germany
| | - Frederike Adammek
- TU Dortmund University, Institute for Sport and Sport Science, Division of Performance and Health (Sports Medicine), 44227 Dortmund, Germany
| | - Marit Lea Schlagheck
- TU Dortmund University, Institute for Sport and Sport Science, Division of Performance and Health (Sports Medicine), 44227 Dortmund, Germany
| | | | - Alexander Schenk
- TU Dortmund University, Institute for Sport and Sport Science, Division of Performance and Health (Sports Medicine), 44227 Dortmund, Germany
| | - Clemens Warnke
- Department of Neurology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Wilhelm Bloch
- Department for Molecular and Cellular Sports Medicine, Institute of Cardiovascular Research and Sports Medicine, German Sport University Cologne, Cologne, Germany
| | - Philipp Zimmer
- TU Dortmund University, Institute for Sport and Sport Science, Division of Performance and Health (Sports Medicine), 44227 Dortmund, Germany.
| |
Collapse
|
5
|
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
|
6
|
Mittl K, Hayashi F, Dandekar R, Schubert RD, Gerdts J, Oshiro L, Loudermilk R, Greenfield A, Augusto DG, Ramesh A, Tran E, Koshal K, Kizer K, Dreux J, Cagalingan A, Schustek F, Flood L, Moore T, Kirkemo LL, Cooper T, Harms M, Gomez R, Sibener L, Cree BAC, Hauser SL, Hollenbach JA, Gee M, Wilson MR, Zamvil SS, Sabatino JJ. Antigen specificity of clonally-enriched CD8+ T cells in multiple sclerosis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.07.611010. [PMID: 39282370 PMCID: PMC11398516 DOI: 10.1101/2024.09.07.611010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 09/19/2024]
Abstract
CD8+ T cells are the dominant lymphocyte population in multiple sclerosis (MS) lesions where they are highly clonally expanded. The clonal identity, function, and antigen specificity of CD8+ T cells in MS are not well understood. Here we report a comprehensive single-cell RNA-seq and T cell receptor (TCR)-seq analysis of the cerebrospinal fluid (CSF) and blood from a cohort of treatment-naïve MS patients and control participants. A small subset of highly expanded and activated CD8+ T cells were enriched in the CSF in MS that displayed high activation, cytotoxicity and tissue-homing transcriptional profiles. Using a combination of unbiased and targeted antigen discovery approaches, MS-derived CD8+ T cell clonotypes recognizing Epstein-Barr virus (EBV) antigens and multiple novel mimotopes were identified. These findings shed vital insight into the role of CD8+ T cells in MS and pave the way towards disease biomarkers and therapeutic targets.
Collapse
|
7
|
Ikeguchi R, Kanda N, Kobayashi M, Masui K, Nitta M, Misu T, Muragaki Y, Kawamata T, Shibata N, Kitagawa K, Shimizu Y. CNS B cell infiltration in tumefactive anti-myelin oligodendrocyte glycoprotein antibody-associated disease. Mult Scler J Exp Transl Clin 2024; 10:20552173241301011. [PMID: 39651331 PMCID: PMC11622319 DOI: 10.1177/20552173241301011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Accepted: 10/30/2024] [Indexed: 12/11/2024] Open
Abstract
Background Few studies have examined B cells among patients with anti-myelin oligodendrocyte glycoprotein (MOG) antibody-associated disease (MOGAD), including brain pathology. Objective To describe cases of tumefactive MOGAD with B-cell dominant central nervous system (CNS) infiltration. Methods In this study, we reviewed three cases with clinical and brain histopathological features with tumefactive MOGAD. Results Forty-nine cases of tumefactive brain lesions (TBL) between January 2003 and December 2023 were included; of these, seven had MOGAD. Three underwent a brain biopsy. B-cell dominant CNS infiltration was observed in two cases. In two cases with B-cell dominant CNS infiltration, symptoms included fever, headache, nausea, somnolence, and focal neurological deficits. Cerebrospinal fluid examination revealed both mild pleocytosis and negative oligoclonal IgG bands. Magnetic resonance imaging of the brain revealed large abnormal lesions extending from the basal ganglia to the parietotemporal lobe in both cases. These cases showed a good response to steroids; however, one case relapsed. Brain pathology showed demyelination and perivascular lymphocytic infiltration. One showed small vessel vasculitis. Deposition of the activated complement component was absent or rarely observed. Loss of MOG was observed in two cases. Conclusion MOGAD could exhibit B-cell dominant CNS infiltration and small vessel vasculitis. MOGAD should be considered in differential diagnosis of TBL.
Collapse
Affiliation(s)
- Ryotaro Ikeguchi
- Department of Neurology, Tokyo Women's Medical University, Tokyo, Japan
| | - Natsuki Kanda
- Department of Neurology, Tokyo Women's Medical University, Tokyo, Japan
| | - Masaki Kobayashi
- Department of Neurology, Tokyo Women's Medical University, Tokyo, Japan
| | - Kenta Masui
- Department of Pathology, Tokyo Women's Medical University, Tokyo, Japan
| | - Masayuki Nitta
- Depertment of Neurosurgery, Tokyo Women's Medical University, Tokyo, Japan
| | - Tatsuro Misu
- Department of Neurology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Yoshihiro Muragaki
- Depertment of Neurosurgery, Tokyo Women's Medical University, Tokyo, Japan
| | - Takakazu Kawamata
- Depertment of Neurosurgery, Tokyo Women's Medical University, Tokyo, Japan
| | - Noriyuki Shibata
- Department of Pathology, Tokyo Women's Medical University, Tokyo, Japan
| | - Kazuo Kitagawa
- Department of Neurology, Tokyo Women's Medical University, Tokyo, Japan
| | - Yuko Shimizu
- Department of Neurology, Tokyo Women's Medical University, Tokyo, Japan
| |
Collapse
|
8
|
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
|
9
|
Arneth B. Current Knowledge about CD3 +CD20 + T Cells in Patients with Multiple Sclerosis. Int J Mol Sci 2024; 25:8987. [PMID: 39201672 PMCID: PMC11354236 DOI: 10.3390/ijms25168987] [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: 05/24/2024] [Revised: 08/16/2024] [Accepted: 08/18/2024] [Indexed: 09/03/2024] Open
Abstract
Multiple sclerosis (MS) is a disease of the central nervous system (CNS) characterized by inflammation and autoimmune responses. This review explores the participation of T cells, particularly certain CD3+CD20+ T cells, in the clinical manifestations of MS and highlights their presence in diagnosed patients. These T cells show aberrant expression of CD20, normally considered a B-cell marker. In this review, relevant journal articles available in PubMed and CINAHL were identified by employing diverse search terms, such as MS, CD3+CD20+ T cells, the incidence and significance of CD3+CD20+ T cells in MS patients, and the impact of rituximab treatment. The search was limited to articles published in the ten-year period from 2014 to 2024. The results of this review suggest that most scholars agree on the presence of CD3+CD20+ T cells in cerebrospinal fluid. Emerging concepts relate to the fundamental role of CD20-expressing T cells in determining the target and efficacy of MS therapeutics and the presence of T cells in the cerebrospinal fluid of MS patients. The results clearly show that CD20+ T cells indicate disease chronicity and high disease activity.
Collapse
Affiliation(s)
- Borros Arneth
- Institute of Laboratory Medicine and Pathobiochemistry, Molecular Diagnostics, Hospital of the Universities of Giessen and Marburg (UKGM), Justus Liebig University Giessen, Feulgenstr. 12, 35392 Giessen, Germany;
- Institute of Laboratory Medicine and Pathobiochemistry, Molecular Diagnostics, Hospital of the Universities of Giessen and Marburg (UKGM), Philipps University Marburg, Baldinger Str., 35043 Marburg, Germany
| |
Collapse
|
10
|
Stys PK, Tsutsui S, Gafson AR, ‘t Hart BA, Belachew S, Geurts JJG. New views on the complex interplay between degeneration and autoimmunity in multiple sclerosis. Front Cell Neurosci 2024; 18:1426231. [PMID: 39161786 PMCID: PMC11330826 DOI: 10.3389/fncel.2024.1426231] [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: 05/01/2024] [Accepted: 06/14/2024] [Indexed: 08/21/2024] Open
Abstract
Multiple sclerosis (MS) is a frequently disabling neurological disorder characterized by symptoms, clinical signs and imaging abnormalities that typically fluctuate over time, affecting any level of the CNS. Prominent lymphocytic inflammation, many genetic susceptibility variants involving immune pathways, as well as potent responses of the neuroinflammatory component to immunomodulating drugs, have led to the natural conclusion that this disease is driven by a primary autoimmune process. In this Hypothesis and Theory article, we discuss emerging data that cast doubt on this assumption. After three decades of therapeutic experience, what has become clear is that potent immune modulators are highly effective at suppressing inflammatory relapses, yet exhibit very limited effects on the later progressive phase of MS. Moreover, neuropathological examination of MS tissue indicates that degeneration, CNS atrophy, and myelin loss are most prominent in the progressive stage, when lymphocytic inflammation paradoxically wanes. Finally, emerging clinical observations such as "progression independent of relapse activity" and "silent progression," now thought to take hold very early in the course, together argue that an underlying "cytodegenerative" process, likely targeting the myelinating unit, may in fact represent the most proximal step in a complex pathophysiological cascade exacerbated by an autoimmune inflammatory overlay. Parallels are drawn with more traditional neurodegenerative disorders, where a progressive proteopathy with prion-like propagation of toxic misfolded species is now known to play a key role. A potentially pivotal contribution of the Epstein-Barr virus and B cells in this process is also discussed.
Collapse
Affiliation(s)
- Peter K. Stys
- Department of Clinical Neurosciences, Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Shigeki Tsutsui
- Department of Clinical Neurosciences, Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Arie R. Gafson
- Biogen Digital Health, Biogen, Cambridge, MA, United States
| | - Bert A. ‘t Hart
- Department of Anatomy and Neurosciences, Amsterdam University Medical Centers (location VUmc), Amsterdam, Netherlands
| | - Shibeshih Belachew
- TheraPanacea, Paris, France
- Indivi (DBA of Healios AG), Basel, Switzerland
| | - Jeroen J. G. Geurts
- Department of Anatomy and Neurosciences, Amsterdam University Medical Centers (location VUmc), Amsterdam, Netherlands
| |
Collapse
|
11
|
Bodansky A, Mettelman RC, Sabatino JJ, Vazquez SE, Chou J, Novak T, Moffitt KL, Miller HS, Kung AF, Rackaityte E, Zamecnik CR, Rajan JV, Kortbawi H, Mandel-Brehm C, Mitchell A, Wang CY, Saxena A, Zorn K, Yu DJL, Pogorelyy MV, Awad W, Kirk AM, Asaki J, Pluvinage JV, Wilson MR, Zambrano LD, Campbell AP, Thomas PG, Randolph AG, Anderson MS, DeRisi JL. Molecular mimicry in multisystem inflammatory syndrome in children. Nature 2024; 632:622-629. [PMID: 39112696 PMCID: PMC11324515 DOI: 10.1038/s41586-024-07722-4] [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: 06/20/2023] [Accepted: 06/14/2024] [Indexed: 08/16/2024]
Abstract
Multisystem inflammatory syndrome in children (MIS-C) is a severe, post-infectious sequela of SARS-CoV-2 infection1,2, yet the pathophysiological mechanism connecting the infection to the broad inflammatory syndrome remains unknown. Here we leveraged a large set of samples from patients with MIS-C to identify a distinct set of host proteins targeted by patient autoantibodies including a particular autoreactive epitope within SNX8, a protein involved in regulating an antiviral pathway associated with MIS-C pathogenesis. In parallel, we also probed antibody responses from patients with MIS-C to the complete SARS-CoV-2 proteome and found enriched reactivity against a distinct domain of the SARS-CoV-2 nucleocapsid protein. The immunogenic regions of the viral nucleocapsid and host SNX8 proteins bear remarkable sequence similarity. Consequently, we found that many children with anti-SNX8 autoantibodies also have cross-reactive T cells engaging both the SNX8 and the SARS-CoV-2 nucleocapsid protein epitopes. Together, these findings suggest that patients with MIS-C develop a characteristic immune response to the SARS-CoV-2 nucleocapsid protein that is associated with cross-reactivity to the self-protein SNX8, demonstrating a mechanistic link between the infection and the inflammatory syndrome, with implications for better understanding a range of post-infectious autoinflammatory diseases.
Collapse
Affiliation(s)
- Aaron Bodansky
- Department of Pediatrics, Division of Critical Care, University of California San Francisco, San Francisco, CA, USA
| | - Robert C Mettelman
- Department of Host-Microbe Interactions, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Joseph J Sabatino
- Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA, USA
- Department of Neurology, University of California San Francisco, San Francisco, CA, USA
| | - Sara E Vazquez
- Department of Biochemistry and Biophysics, University of California San Francisco, San Francisco, CA, USA
| | - Janet Chou
- Division of Immunology, Department of Pediatrics, Boston, MA, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Tanya Novak
- Department of Anesthesiology, Critical Care and Pain Medicine, Boston Children's Hospital, Boston, MA, USA
- Department of Anesthesia, Harvard Medical School, Boston, MA, USA
| | - Kristin L Moffitt
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
- Department of Pediatric, Division of Infectious Diseases, Boston Children's Hospital, Boston, MA, USA
| | - Haleigh S Miller
- Department of Biochemistry and Biophysics, University of California San Francisco, San Francisco, CA, USA
- Biological and Medical Informatics Program, University of California San Francisco, San Francisco, CA, USA
| | - Andrew F Kung
- Department of Biochemistry and Biophysics, University of California San Francisco, San Francisco, CA, USA
- Biological and Medical Informatics Program, University of California San Francisco, San Francisco, CA, USA
| | - Elze Rackaityte
- Department of Biochemistry and Biophysics, University of California San Francisco, San Francisco, CA, USA
| | - Colin R Zamecnik
- Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA, USA
- Department of Neurology, University of California San Francisco, San Francisco, CA, USA
| | - Jayant V Rajan
- Department of Biochemistry and Biophysics, University of California San Francisco, San Francisco, CA, USA
| | - Hannah Kortbawi
- Department of Biochemistry and Biophysics, University of California San Francisco, San Francisco, CA, USA
- Medical Scientist Training Program, University of California San Francisco, San Francisco, CA, USA
| | - Caleigh Mandel-Brehm
- Department of Biochemistry and Biophysics, University of California San Francisco, San Francisco, CA, USA
| | | | | | - Aditi Saxena
- Chan Zuckerberg Biohub SF, San Francisco, CA, USA
| | - Kelsey Zorn
- Department of Biochemistry and Biophysics, University of California San Francisco, San Francisco, CA, USA
| | - David J L Yu
- Diabetes Center, School of Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Mikhail V Pogorelyy
- Department of Host-Microbe Interactions, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Walid Awad
- Department of Host-Microbe Interactions, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Allison M Kirk
- Department of Host-Microbe Interactions, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - James Asaki
- Biomedical Sciences Program, University of California San Francisco, San Francisco, CA, USA
| | - John V Pluvinage
- Department of Neurology, University of California San Francisco, San Francisco, CA, USA
| | - Michael R Wilson
- Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA, USA
- Department of Neurology, University of California San Francisco, San Francisco, CA, USA
| | - Laura D Zambrano
- COVID-19 Response Team and Coronavirus and Other Respiratory Viruses Division, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Angela P Campbell
- COVID-19 Response Team and Coronavirus and Other Respiratory Viruses Division, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Paul G Thomas
- Department of Host-Microbe Interactions, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Adrienne G Randolph
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
- Department of Anesthesiology, Critical Care and Pain Medicine, Boston Children's Hospital, Boston, MA, USA
- Department of Anesthesia, Harvard Medical School, Boston, MA, USA
| | - Mark S Anderson
- Diabetes Center, School of Medicine, University of California San Francisco, San Francisco, CA, USA.
- Department of Medicine, Division of Endocrinology and Metabolism, University of California San Francisco, San Francisco, CA, USA.
| | - Joseph L DeRisi
- Department of Biochemistry and Biophysics, University of California San Francisco, San Francisco, CA, USA.
- Chan Zuckerberg Biohub SF, San Francisco, CA, USA.
| |
Collapse
|
12
|
Prapas P, Anagnostouli M. Macrophages and HLA-Class II Alleles in Multiple Sclerosis: Insights in Therapeutic Dynamics. Int J Mol Sci 2024; 25:7354. [PMID: 39000461 PMCID: PMC11242320 DOI: 10.3390/ijms25137354] [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: 06/15/2024] [Revised: 07/01/2024] [Accepted: 07/02/2024] [Indexed: 07/16/2024] Open
Abstract
Antigen presentation is a crucial mechanism that drives the T cell-mediated immune response and the development of Multiple Sclerosis (MS). Genetic alterations within the highly variable Major Histocompatibility Complex Class II (MHC II) have been proven to result in significant changes in the molecular basis of antigen presentation and the clinical course of patients with both Adult-Onset MS (AOMS) and Pediatric-Onset MS (POMS). Among the numerous polymorphisms of the Human Leucocyte Antigens (HLA), within MHC II complex, HLA-DRB1*15:01 has been labeled, in Caucasian ethnic groups, as a high-risk allele for MS due to the ability of its structure to increase affinity to Myelin Basic Protein (MBP) epitopes. This characteristic, among others, in the context of the trimolecular complex or immunological synapsis, provides the foundation for autoimmunity triggered by environmental or endogenous factors. As with all professional antigen presenting cells, macrophages are characterized by the expression of MHC II and are often implicated in the formation of MS lesions. Increased presence of M1 macrophages in MS patients has been associated both with progression and onset of the disease, each involving separate but similar mechanisms. In this critical narrative review, we focus on macrophages, discussing how HLA genetic alterations can promote dysregulation of this population's homeostasis in the periphery and the Central Nervous System (CNS). We also explore the potential interconnection in observed pathological macrophage mechanisms and the function of the diverse structure of HLA alleles in neurodegenerative CNS, seen in MS, by comparing available clinical with molecular data through the prism of HLA-immunogenetics. Finally, we discuss available and experimental pharmacological approaches for MS targeting the trimolecular complex that are based on cell phenotype modulation and HLA genotype involvement and try to reveal fertile ground for the potential development of novel drugs.
Collapse
Affiliation(s)
- Petros Prapas
- Research Immunogenetics Laboratory, First Department of Neurology, Aeginition University Hospital, School of Medicine, National and Kapodistrian University of Athens, Vas. Sofias 72-74, 11528 Athens, Greece
| | - Maria Anagnostouli
- Research Immunogenetics Laboratory, First Department of Neurology, Aeginition University Hospital, School of Medicine, National and Kapodistrian University of Athens, Vas. Sofias 72-74, 11528 Athens, Greece
- Multiple Sclerosis and Demyelinating Diseases Unit, Center of Expertise for Rare Demyelinating and Autoimmune Diseases of CNS, First Department of Neurology, School of Medicine, National and Kapodistrian University of Athens NKUA, Aeginition University Hospital, Vas. Sofias 72-74, 11528 Athens, Greece
| |
Collapse
|
13
|
Abbadessa G, Lepore MT, Bruzzaniti S, Piemonte E, Miele G, Signoriello E, Perna F, De Falco C, Lus G, Matarese G, Bonavita S, Galgani M. Ocrelizumab Alters Cytotoxic Lymphocyte Function While Reducing EBV-Specific CD8 + T-Cell Proliferation in Patients With Multiple Sclerosis. NEUROLOGY(R) NEUROIMMUNOLOGY & NEUROINFLAMMATION 2024; 11:e200250. [PMID: 38662990 PMCID: PMC11087045 DOI: 10.1212/nxi.0000000000200250] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Accepted: 03/08/2024] [Indexed: 05/12/2024]
Abstract
BACKGROUND AND OBJECTIVES The role of B cells in the pathogenic events leading to relapsing multiple sclerosis (R-MS) has only been recently elucidated. A pivotal step in defining this role has been provided by therapeutic efficacy of anti-CD20 monoclonal antibodies. Indeed, treatment with anti-CD20 can also alter number and function of other immune cells not directly expressing CD20 on their cell surface, whose activities can contribute to unknown aspects influencing therapeutic efficacy. We examined the phenotype and function of cytotoxic lymphocytes and Epstein-Barr virus (EBV)-specific immune responses in people with R-MS before and after ocrelizumab treatment. METHODS In this prospective study, we collected blood samples from people with R-MS (n = 41) before and 6 and 12 months after initiating ocrelizumab to assess the immune phenotype and the indirect impact on cytotoxic functions of CD8+ T and NK cells. In addition, we evaluated the specific anti-EBV proliferative responses of both CD8+ T and NK lymphocytes as surrogate markers of anti-EBV activity. RESULTS We observed that while ocrelizumab depleted circulating B cells, it also reduced the expression of activation and migratory markers on both CD8+ T and NK cells as well as their in vitro cytotoxic activity. A comparable pattern in the modulation of immune molecules by ocrelizumab was observed in cytotoxic cells even when patients with R-MS were divided into groups based on their prior disease-modifying treatment. These effects were accompanied by a significant and selective reduction of CD8+ T-cell proliferation in response to EBV antigenic peptides. DISCUSSION Taken together, our findings suggest that ocrelizumab-while depleting B cells-affects the cytotoxic function of CD8+ and NK cells, whose reduced cross-activity against myelin antigens might also contribute to its therapeutic efficacy during MS.
Collapse
MESH Headings
- Humans
- Antibodies, Monoclonal, Humanized/pharmacology
- Female
- Adult
- Male
- Herpesvirus 4, Human/immunology
- CD8-Positive T-Lymphocytes/drug effects
- CD8-Positive T-Lymphocytes/immunology
- Middle Aged
- Immunologic Factors/pharmacology
- Multiple Sclerosis, Relapsing-Remitting/drug therapy
- Multiple Sclerosis, Relapsing-Remitting/immunology
- Multiple Sclerosis, Relapsing-Remitting/blood
- T-Lymphocytes, Cytotoxic/drug effects
- T-Lymphocytes, Cytotoxic/immunology
- Prospective Studies
- Cell Proliferation/drug effects
- Killer Cells, Natural/drug effects
- Killer Cells, Natural/immunology
- Lymphocyte Activation/drug effects
Collapse
Affiliation(s)
- Gianmarco Abbadessa
- From the Department of Advanced Medical and Surgical Sciences (G.A., G. Miele, E.S., G.L., S. Bonavita), University of Campania Luigi Vanvitelli; Istituto per l'Endocrinologia e l'Oncologia Sperimentale "G.Salvatore" - Consiglio Nazionale delle Ricerche (M.T.L., S. Bruzzaniti, G. Matarese, M.G.); Department of Molecular Medicine and Medical Biotechnologies (E.P., G. Matarese, M.G.); Department of Clinical Medicine and Surgery (F.P.), University of Naples "Federico II"; and UOC Biochimica Clinica - Ospedali dei Colli (C.D.F.), Naples, Italy
| | - Maria Teresa Lepore
- From the Department of Advanced Medical and Surgical Sciences (G.A., G. Miele, E.S., G.L., S. Bonavita), University of Campania Luigi Vanvitelli; Istituto per l'Endocrinologia e l'Oncologia Sperimentale "G.Salvatore" - Consiglio Nazionale delle Ricerche (M.T.L., S. Bruzzaniti, G. Matarese, M.G.); Department of Molecular Medicine and Medical Biotechnologies (E.P., G. Matarese, M.G.); Department of Clinical Medicine and Surgery (F.P.), University of Naples "Federico II"; and UOC Biochimica Clinica - Ospedali dei Colli (C.D.F.), Naples, Italy
| | - Sara Bruzzaniti
- From the Department of Advanced Medical and Surgical Sciences (G.A., G. Miele, E.S., G.L., S. Bonavita), University of Campania Luigi Vanvitelli; Istituto per l'Endocrinologia e l'Oncologia Sperimentale "G.Salvatore" - Consiglio Nazionale delle Ricerche (M.T.L., S. Bruzzaniti, G. Matarese, M.G.); Department of Molecular Medicine and Medical Biotechnologies (E.P., G. Matarese, M.G.); Department of Clinical Medicine and Surgery (F.P.), University of Naples "Federico II"; and UOC Biochimica Clinica - Ospedali dei Colli (C.D.F.), Naples, Italy
| | - Erica Piemonte
- From the Department of Advanced Medical and Surgical Sciences (G.A., G. Miele, E.S., G.L., S. Bonavita), University of Campania Luigi Vanvitelli; Istituto per l'Endocrinologia e l'Oncologia Sperimentale "G.Salvatore" - Consiglio Nazionale delle Ricerche (M.T.L., S. Bruzzaniti, G. Matarese, M.G.); Department of Molecular Medicine and Medical Biotechnologies (E.P., G. Matarese, M.G.); Department of Clinical Medicine and Surgery (F.P.), University of Naples "Federico II"; and UOC Biochimica Clinica - Ospedali dei Colli (C.D.F.), Naples, Italy
| | - Giuseppina Miele
- From the Department of Advanced Medical and Surgical Sciences (G.A., G. Miele, E.S., G.L., S. Bonavita), University of Campania Luigi Vanvitelli; Istituto per l'Endocrinologia e l'Oncologia Sperimentale "G.Salvatore" - Consiglio Nazionale delle Ricerche (M.T.L., S. Bruzzaniti, G. Matarese, M.G.); Department of Molecular Medicine and Medical Biotechnologies (E.P., G. Matarese, M.G.); Department of Clinical Medicine and Surgery (F.P.), University of Naples "Federico II"; and UOC Biochimica Clinica - Ospedali dei Colli (C.D.F.), Naples, Italy
| | - Elisabetta Signoriello
- From the Department of Advanced Medical and Surgical Sciences (G.A., G. Miele, E.S., G.L., S. Bonavita), University of Campania Luigi Vanvitelli; Istituto per l'Endocrinologia e l'Oncologia Sperimentale "G.Salvatore" - Consiglio Nazionale delle Ricerche (M.T.L., S. Bruzzaniti, G. Matarese, M.G.); Department of Molecular Medicine and Medical Biotechnologies (E.P., G. Matarese, M.G.); Department of Clinical Medicine and Surgery (F.P.), University of Naples "Federico II"; and UOC Biochimica Clinica - Ospedali dei Colli (C.D.F.), Naples, Italy
| | - Francesco Perna
- From the Department of Advanced Medical and Surgical Sciences (G.A., G. Miele, E.S., G.L., S. Bonavita), University of Campania Luigi Vanvitelli; Istituto per l'Endocrinologia e l'Oncologia Sperimentale "G.Salvatore" - Consiglio Nazionale delle Ricerche (M.T.L., S. Bruzzaniti, G. Matarese, M.G.); Department of Molecular Medicine and Medical Biotechnologies (E.P., G. Matarese, M.G.); Department of Clinical Medicine and Surgery (F.P.), University of Naples "Federico II"; and UOC Biochimica Clinica - Ospedali dei Colli (C.D.F.), Naples, Italy
| | - Chiara De Falco
- From the Department of Advanced Medical and Surgical Sciences (G.A., G. Miele, E.S., G.L., S. Bonavita), University of Campania Luigi Vanvitelli; Istituto per l'Endocrinologia e l'Oncologia Sperimentale "G.Salvatore" - Consiglio Nazionale delle Ricerche (M.T.L., S. Bruzzaniti, G. Matarese, M.G.); Department of Molecular Medicine and Medical Biotechnologies (E.P., G. Matarese, M.G.); Department of Clinical Medicine and Surgery (F.P.), University of Naples "Federico II"; and UOC Biochimica Clinica - Ospedali dei Colli (C.D.F.), Naples, Italy
| | - G Lus
- From the Department of Advanced Medical and Surgical Sciences (G.A., G. Miele, E.S., G.L., S. Bonavita), University of Campania Luigi Vanvitelli; Istituto per l'Endocrinologia e l'Oncologia Sperimentale "G.Salvatore" - Consiglio Nazionale delle Ricerche (M.T.L., S. Bruzzaniti, G. Matarese, M.G.); Department of Molecular Medicine and Medical Biotechnologies (E.P., G. Matarese, M.G.); Department of Clinical Medicine and Surgery (F.P.), University of Naples "Federico II"; and UOC Biochimica Clinica - Ospedali dei Colli (C.D.F.), Naples, Italy
| | - Giuseppe Matarese
- From the Department of Advanced Medical and Surgical Sciences (G.A., G. Miele, E.S., G.L., S. Bonavita), University of Campania Luigi Vanvitelli; Istituto per l'Endocrinologia e l'Oncologia Sperimentale "G.Salvatore" - Consiglio Nazionale delle Ricerche (M.T.L., S. Bruzzaniti, G. Matarese, M.G.); Department of Molecular Medicine and Medical Biotechnologies (E.P., G. Matarese, M.G.); Department of Clinical Medicine and Surgery (F.P.), University of Naples "Federico II"; and UOC Biochimica Clinica - Ospedali dei Colli (C.D.F.), Naples, Italy
| | - Simona Bonavita
- From the Department of Advanced Medical and Surgical Sciences (G.A., G. Miele, E.S., G.L., S. Bonavita), University of Campania Luigi Vanvitelli; Istituto per l'Endocrinologia e l'Oncologia Sperimentale "G.Salvatore" - Consiglio Nazionale delle Ricerche (M.T.L., S. Bruzzaniti, G. Matarese, M.G.); Department of Molecular Medicine and Medical Biotechnologies (E.P., G. Matarese, M.G.); Department of Clinical Medicine and Surgery (F.P.), University of Naples "Federico II"; and UOC Biochimica Clinica - Ospedali dei Colli (C.D.F.), Naples, Italy
| | - Mario Galgani
- From the Department of Advanced Medical and Surgical Sciences (G.A., G. Miele, E.S., G.L., S. Bonavita), University of Campania Luigi Vanvitelli; Istituto per l'Endocrinologia e l'Oncologia Sperimentale "G.Salvatore" - Consiglio Nazionale delle Ricerche (M.T.L., S. Bruzzaniti, G. Matarese, M.G.); Department of Molecular Medicine and Medical Biotechnologies (E.P., G. Matarese, M.G.); Department of Clinical Medicine and Surgery (F.P.), University of Naples "Federico II"; and UOC Biochimica Clinica - Ospedali dei Colli (C.D.F.), Naples, Italy
| |
Collapse
|
14
|
Peng BJ, Alvarado A, Cassim H, Guarneri S, Wong S, Willis J, SantaMaria J, Martynchuk A, Stratton V, Patel D, Chen CC, Li Y, Binder GK, Dryer-Minnerly R, Lee J, Basu S. Preclinical specificity & activity of a fully human 41BB-expressing anti-CD19 CART- therapy for treatment-resistant autoimmune disease. Mol Ther Methods Clin Dev 2024; 32:101267. [PMID: 38883975 PMCID: PMC11176803 DOI: 10.1016/j.omtm.2024.101267] [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: 10/02/2023] [Accepted: 05/16/2024] [Indexed: 06/18/2024]
Abstract
Over 4% of the global population is estimated to live with autoimmune disease, necessitating immunosuppressive treatment that is often chronic, not curative, and carries associated risks. B cells have emerged as key players in disease pathogenesis, as evidenced by partial responsiveness to B cell depletion by antibody-based therapies. However, these treatments often have transient effects due to incomplete depletion of tissue-resident B cells. Chimeric antigen receptor (CAR) T cells targeting B cells have demonstrated efficacy in refractory systemic lupus erythematosus. To this end, we developed an anti-CD19 CAR T cell product candidate, CABA-201, containing a clinically evaluated fully human CD19 binder (IC78) with a 4-1BB costimulatory domain and CD3 zeta stimulation domain for treatment refractory autoimmune disease. Here, we demonstrate specific cytotoxic activity of CABA-201 against CD19+ Nalm6 cells with no off-target effects on primary human cells. Novel examination of CABA-201 generated from primary T cells from multiple patients with autoimmune disease displayed robust CAR surface expression and effective elimination of the intended target autologous CD19+ B cells in vitro. Together, these findings support the tolerability and activity of CABA-201 for clinical development in patients with autoimmune disease.
Collapse
Affiliation(s)
- Binghao J Peng
- Department of Cellular and Molecular Immunology, Cabaletta Bio, Philadelphia, PA 19130, USA
| | - Andrea Alvarado
- Department of Cellular and Molecular Immunology, Cabaletta Bio, Philadelphia, PA 19130, USA
| | - Hangameh Cassim
- Department of Cellular and Molecular Immunology, Cabaletta Bio, Philadelphia, PA 19130, USA
| | - Soprina Guarneri
- Department of Protein and Molecular Biology, Cabaletta Bio, Philadelphia, PA 19130, USA
| | - Steven Wong
- Department of Protein and Molecular Biology, Cabaletta Bio, Philadelphia, PA 19130, USA
| | - Jonathan Willis
- Department of Analytical Development, Cabaletta Bio, Philadelphia, PA 19130, USA
| | - Julia SantaMaria
- Department of Analytical Development, Cabaletta Bio, Philadelphia, PA 19130, USA
| | - Ashley Martynchuk
- Department of Manufacturing, Science, and Technologies, Cabaletta Bio, Philadelphia, PA 19130, USA
| | - Victoria Stratton
- Department of Manufacturing, Science, and Technologies, Cabaletta Bio, Philadelphia, PA 19130, USA
| | - Darshil Patel
- Department of Protein and Molecular Biology, Cabaletta Bio, Philadelphia, PA 19130, USA
| | - Chien-Chung Chen
- Department of Analytical Development, Cabaletta Bio, Philadelphia, PA 19130, USA
| | - Yan Li
- Department of Manufacturing, Science, and Technologies, Cabaletta Bio, Philadelphia, PA 19130, USA
| | | | | | - Jinmin Lee
- Department of Cellular and Molecular Immunology, Cabaletta Bio, Philadelphia, PA 19130, USA
| | - Samik Basu
- Cabaletta Bio, Philadelphia, PA 19130, USA
| |
Collapse
|
15
|
Aliyu M, Zohora FT, Ceylan A, Hossain F, Yazdani R, Azizi G. Immunopathogenesis of multiple sclerosis: molecular and cellular mechanisms and new immunotherapeutic approaches. Immunopharmacol Immunotoxicol 2024; 46:355-377. [PMID: 38634438 DOI: 10.1080/08923973.2024.2330642] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Accepted: 03/09/2024] [Indexed: 04/19/2024]
Abstract
BACKGROUND Multiple sclerosis (MS) is a central nervous system (CNS) demyelinating autoimmune disease with increasing global prevalence. It predominantly affects females, especially those of European descent. The interplay between environmental factors and genetic predisposition plays a crucial role in MS etiopathogenesis. METHODS We searched recent relevant literature on reputable databases, which include, PubMed, Embase, Web of Science, Scopus, and ScienceDirect using the following keywords: multiple sclerosis, pathogenesis, autoimmunity, demyelination, therapy, and immunotherapy. RESULTS Various animal models have been employed to investigate the MS etiopathogenesis and therapeutics. Autoreactive T cells within the CNS recruit myeloid cells through chemokine expression, leading to the secretion of inflammatory cytokines driving the MS pathogenesis, resulting in demyelination, gliosis, and axonal loss. Key players include T cell lymphocytes (CD4+ and CD8+), B cells, and neutrophils. Signaling dysregulation in inflammatory pathways and the immunogenetic basis of MS are essential considerations for any successful therapy to MS. Data indicates that B cells and neutrophils also have significant roles in MS, despite the common belief that T cells are essential. High neutrophil-to-lymphocyte ratios correlate with MS severity, indicating their contribution to disease progression. Dysregulated signaling pathways further exacerbate MS progression. CONCLUSION MS remains incurable, but disease-modifying therapies, monoclonal antibodies, and immunomodulatory drugs offer hope for patients. Research on the immunogenetics and immunoregulatory functions of gut microbiota is continuing to provide light on possible treatment avenues. Understanding the complex interplay between genetic predisposition, environmental factors, and immune dysregulation is critical for developing effective treatments for MS.
Collapse
Affiliation(s)
- Mansur Aliyu
- Department of Immunology, School of Public Health, Tehran University of Medical Sciences, International Campus, TUMS-IC, Tehran, Iran
- Department of Medical Microbiology, Faculty of Clinical Science, College of Health Sciences, Bayero University, Kano, Nigeria
| | - Fatema Tuz Zohora
- Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Subang Jaya, Malaysia
| | - Ayca Ceylan
- Medical Faculty, Department of Pediatrics, Division of Immunology and Allergy, Selcuk University, Konya, Turkey
| | - Fariha Hossain
- Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Subang Jaya, Malaysia
| | - Reza Yazdani
- Department of Neurology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Gholamreza Azizi
- Department of Neurology, Thomas Jefferson University, Philadelphia, PA, USA
- Non-communicable Diseases Research Center, Alborz University of Medical Sciences, Karaj, Iran
| |
Collapse
|
16
|
Friedli C, Krajnc N, Hammer HN, Marti S, Zrzavy T, Evangelopoulos ME, Kapsali I, Rommer P, Berger T, Chan A, Bsteh G, Hoepner R. Different lymphocyte counts of multiple sclerosis patients treated with ofatumumab and ocrelizumab: A retrospective observational study. J Cent Nerv Syst Dis 2024; 16:11795735241249644. [PMID: 38711956 PMCID: PMC11072073 DOI: 10.1177/11795735241249644] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Accepted: 03/08/2024] [Indexed: 05/08/2024] Open
Abstract
Introduction Patients with Multiple Sclerosis (pwMS) treated with anti-CD20 (cluster of differentiation) monoclonal antibodies (mAbs) such as ocrelizumab (OCR) and ofatumumab (OFA) show a reduction mainly of B-lymphocytes, but also other lymphocyte subsets can be affected by these treatments. There is limited data on differences between lymphocyte subset counts of pwMS after treatment initiation with OCR or OFA. Objective To compare lymphocyte subset counts after treatment initiation in pwMS treated with OCR and OFA. Methods We analyzed 22 pwMS initiated on OFA and 56 sex-, age- and MS course matched pwMS initiated on OCR from 2 prospectively collected observational MS databases (Bern [n: OFA 14, OCR 44] and Vienna [n: OFA 8, OCR 12]) statistically comparing lymphocyte subset counts (Mann Whitney Test). Results We found that pwMS treated with OCR showed a stronger reduction of CD20 B-lymphocytes (P = .001), and a trend towards lower counts of CD8+ T cells (P = .056) compared to pwMS treated with OFA, whereas reduction of total lymphocyte, CD4+ lymphocyte and NK cell count was equally distributed between both treatments. Conclusion Different effects on lymphocyte subpopulations appear to be present in pwMS after treatment initiation with different anti-CD20 mAbs. Further studies are needed to determine potential effects on anti-CD20 treatment efficacy as well as treatment associated risks such as failed vaccinations and infections.
Collapse
Affiliation(s)
- Christoph Friedli
- Department of Neurology, Inselspital, Bern University Hospital and University of Bern, Switzerland
- Department of Neurology, Waikato Hospital, Hamilton, New Zealand
| | - Nik Krajnc
- Comprehensive Center for Clinical Neurosciences and Mental Health, Medical University of Vienna, Vienna, Austria
- Department of Neurology, Medical University of Vienna, Wien, Austria
| | - Helly N. Hammer
- Department of Neurology, Inselspital, Bern University Hospital and University of Bern, Switzerland
| | - Stefanie Marti
- Department of Neurology, Inselspital, Bern University Hospital and University of Bern, Switzerland
| | - Tobias Zrzavy
- Comprehensive Center for Clinical Neurosciences and Mental Health, Medical University of Vienna, Vienna, Austria
- Department of Neurology, Medical University of Vienna, Wien, Austria
| | - Maria E. Evangelopoulos
- Department of Neurology, Eginition University Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Ioanna Kapsali
- Department of Neurology, Eginition University Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Paulus Rommer
- Comprehensive Center for Clinical Neurosciences and Mental Health, Medical University of Vienna, Vienna, Austria
- Department of Neurology, Medical University of Vienna, Wien, Austria
| | - Thomas Berger
- Comprehensive Center for Clinical Neurosciences and Mental Health, Medical University of Vienna, Vienna, Austria
- Department of Neurology, Medical University of Vienna, Wien, Austria
| | - Andrew Chan
- Department of Neurology, Inselspital, Bern University Hospital and University of Bern, Switzerland
| | - Gabriel Bsteh
- Comprehensive Center for Clinical Neurosciences and Mental Health, Medical University of Vienna, Vienna, Austria
- Department of Neurology, Medical University of Vienna, Wien, Austria
| | - Robert Hoepner
- Department of Neurology, Inselspital, Bern University Hospital and University of Bern, Switzerland
| |
Collapse
|
17
|
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
|
18
|
Yazdanpanah E, Dadfar S, Shadab A, Orooji N, Nemati M, Pazoki A, Esmaeili S, Baharlou R, Haghmorad D. Berberine: A natural modulator of immune cells in multiple sclerosis. Immun Inflamm Dis 2024; 12:e1213. [PMID: 38477663 PMCID: PMC10936236 DOI: 10.1002/iid3.1213] [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: 08/25/2023] [Revised: 02/26/2024] [Accepted: 03/01/2024] [Indexed: 03/14/2024] Open
Abstract
Berberine is a benzylisoquinoline alkaloid found in such plants as Berberis vulgaris, Berberis aristata, and others, revealing a variety of pharmacological properties as a result of interacting with different cellular and molecular targets. Recent studies have shown the immunomodulatory effects of Berberine which result from its impacts on immune cells and immune response mediators such as diverse T lymphocyte subsets, dendritic cells (DCs), and different inflammatory cytokines. Multiple sclerosis (MS) is a chronic disabling and neurodegenerative disease of the central nervous system (CNS) characterized by the recruitment of autoreactive T cells into the CNS causing demyelination, axonal damage, and oligodendrocyte loss. There have been considerable changes discovered in MS regards to the function and frequency of T cell subsets such as Th1 cells, Th17 cells, Th2 cells, Treg cells, and DCs. In the current research, we reviewed the outcomes of in vitro, experimental, and clinical investigations concerning the modulatory effects that Berberine provides on the function and numbers of T cell subsets and DCs, as well as important cytokines that are involved in MS.
Collapse
Affiliation(s)
| | - Sepehr Dadfar
- Department of Immunology, School of MedicineSemnan University of Medical SciencesSemnanIran
| | - Alireza Shadab
- Department of Immunology, School of MedicineSemnan University of Medical SciencesSemnanIran
| | - Niloufar Orooji
- Department of Immunology, School of MedicineSemnan University of Medical SciencesSemnanIran
| | - MohammadHossein Nemati
- Department of Immunology, School of MedicineSemnan University of Medical SciencesSemnanIran
| | - Alireza Pazoki
- Department of Immunology, School of MedicineSemnan University of Medical SciencesSemnanIran
| | | | - Rasoul Baharlou
- Department of Immunology, School of MedicineSemnan University of Medical SciencesSemnanIran
- Cancer Research CenterSemnan University of Medical SciencesSemnanIran
| | - Dariush Haghmorad
- Department of Immunology, School of MedicineSemnan University of Medical SciencesSemnanIran
- Cancer Research CenterSemnan University of Medical SciencesSemnanIran
| |
Collapse
|
19
|
Ipavec N, Rogić Vidaković M, Markotić A, Pavelin S, Buljubašić Šoda M, Šoda J, Dolić K, Režić Mužinić N. Treated and Untreated Primary Progressive Multiple Sclerosis: Walkthrough Immunological Changes of Monocytes and T Regulatory Cells. Biomedicines 2024; 12:464. [PMID: 38398067 PMCID: PMC10887021 DOI: 10.3390/biomedicines12020464] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Revised: 02/05/2024] [Accepted: 02/16/2024] [Indexed: 02/25/2024] Open
Abstract
The objective of this study was to investigate regulatory T cells (Tregs) and monocytes; specifically, the expression of CTLA-4 (CD152) and FOXP3+ in CD4+CD25+ Tregs and the expression of CD40+ and CD192+ monocyte subpopulations in subjects with primary progressive multiple sclerosis (PPMS). Immunological analysis was conducted on peripheral blood samples collected from the 28 PPMS subjects (15 treated with ocrelizumab and 13 untreated PPMS subjects) and 10 healthy control subjects (HCs). The blood samples were incubated with antihuman CD14, CD16, CD40, and CD192 antibodies for monocytes and antihuman CD4, CD25, FOXP3, and CTLA-4 antibodies for lymphocytes. The study results showed that in comparison to HCs both ocrelizumab treated (N = 15) and untreated (N = 13) PPMS subjects had significantly increased percentages of CTLA-4+ and FOXP3+ in CD4+CD25+ Tregs. Further, ocrelizumab treated PPMS subjects, compared to the untreated ones, had significantly decreased percentages of CD192+ and CD40+ nonclassical monocytes. Increased percentages of CTLA-4+ and FOXP3+ in CD4+CD25+ Tregs in both ocrelizumab treated and untreated PPMS subjects indicates the suppressive (inhibitory) role of Tregs in abnormal immune responses in PPMS subjects. Decreased percentages of CD40+ and CD192+ non-classical CD14+CD16++ monocytes for treated compared to untreated PPMS subjects suggests a possible role for ocrelizumab in dampening CNS inflammation.
Collapse
Affiliation(s)
- Nina Ipavec
- Transfusion Medicine Division, University Hospital of Split, 21000 Split, Croatia;
| | - Maja Rogić Vidaković
- Laboratory for Human and Experimental Neurophysiology, Department of Neuroscience, School of Medicine, University of Split, 21000 Split, Croatia
| | - Anita Markotić
- Department of Medical Chemistry and Biochemistry, School of Medicine, University of Split, 21000 Split, Croatia;
| | - Sanda Pavelin
- Department of Neurology, University Hospital of Split, 21000 Split, Croatia;
| | | | - Joško Šoda
- Signal Processing, Analysis, Advanced Diagnostics Research and Education Laboratory (SPAADREL), Department for Marine Electrical Engineering and Information Technologies, Faculty of Maritime Studies, University of Split, 21000 Split, Croatia;
| | - Krešimir Dolić
- Department of Interventional and Diagnostic Radiology, University Hospital of Split, 21000 Split, Croatia;
- Department of Radiology, School of Medicine, University of Split, 21000 Split, Croatia
| | - Nikolina Režić Mužinić
- Department of Medical Chemistry and Biochemistry, School of Medicine, University of Split, 21000 Split, Croatia;
| |
Collapse
|
20
|
Shirani A, Stuve O, Cross AH. Role of B Cells in Relapsing-Remitting and Progressive Multiple Sclerosis and Long-Term Effects of B Cell Depletion. Neurol Clin 2024; 42:137-153. [PMID: 37980111 DOI: 10.1016/j.ncl.2023.06.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2023]
Abstract
Depletion of circulating B lymphocytes using anti-CD20 monoclonal antibodies (mAbs) greatly reduces inflammatory activity in relapsing multiple sclerosis (RMS); it reduces progression to a lesser extent in nonrelapsing progressive MS. Mechanisms whereby anti-CD20 mAbs reduce MRI and clinical relapse activity in people with RMS are still being elucidated. Anti-CD20 agents do not fully protect from nonrelapsing disease progression, possibly due to their inability to cross the blood-brain barrier and inability to ameliorate the full extent of biology of MS progression. Anti-CD20 mAbs have a relatively favorable safety profile, at least in the short-term. Long-term safety studies are still needed.
Collapse
Affiliation(s)
- Afsaneh Shirani
- Division of Multiple Sclerosis, Department of Neurological Sciences, University of Nebraska Medical Center, 988440 Nebraska Medical Center, Omaha, NE 68198-8440, USA
| | - Olaf Stuve
- Department of Neurology, University of Texas Southwestern Medical Center, 6000 Harry Hines Boulevard, Dallas, TX 75390-8813, USA
| | - Anne H Cross
- Department of Neurology, Washington University School of Medicine in St. Louis, 660 South Euclid Avenue, CB 8111, St Louis, MO 63110, USA.
| |
Collapse
|
21
|
Suliman BA. Potential clinical implications of molecular mimicry-induced autoimmunity. Immun Inflamm Dis 2024; 12:e1178. [PMID: 38415936 PMCID: PMC10832321 DOI: 10.1002/iid3.1178] [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: 11/04/2023] [Revised: 01/16/2024] [Accepted: 01/18/2024] [Indexed: 02/29/2024] Open
Abstract
BACKGROUND Molecular mimicry is hypothesized to be a mechanism by which autoimmune diseases are triggered. It refers to sequence or structural homology between foreign antigens and self-antigens, which can activate cross-reactive lymphocytes that attack host tissues. Elucidating the role of molecular mimicry in human autoimmunity could have important clinical implications. OBJECTIVE To review evidence for the role of molecular mimicry in major autoimmune diseases and discuss potential clinical implications. METHODS Comprehensive literature review of clinical trials, observational studies, animal models, and immunology studies on molecular mimicry in multiple sclerosis, type 1 diabetes, rheumatoid arthritis, lupus, Guillain-Barre syndrome, autoimmune myocarditis, and primary biliary cirrhosis published from 2000-2023. RESULTS Substantial indirect evidence supports molecular mimicry as a contributor to loss of self-tolerance in several autoimmune conditions. Proposed microbial triggers include Epstein-Barr virus, coxsackievirus, Campylobacter jejuni, and bacterial commensals. Key mechanisms involve cross-reactive T cells and autoantibodies induced by epitope homology between microbial and self-antigens. Perpetuation of autoimmunity involves epitope spreading, inflammatory mediators, and genetic factors. CONCLUSIONS Molecular mimicry plausibly explains initial stages of autoimmune pathogenesis induced by infection or microbiota disturbances. Understanding mimicry antigens and pathways could enable improved prediction, monitoring, and antigen-specific immunotherapy for autoimmune disorders. However, definitive proof of causation in humans remains limited. Further research should focus on establishing clinical evidence and utility.
Collapse
Affiliation(s)
- Bandar A Suliman
- Department of Clinical Laboratory Sciences, College of Applied Medical SciencesTaibah UniversityMadinahSaudi Arabia
| |
Collapse
|
22
|
Arneth B. Regulatory T Cells in Multiple Sclerosis Diagnostics-What Do We Know So Far? J Pers Med 2023; 14:29. [PMID: 38248730 PMCID: PMC10821144 DOI: 10.3390/jpm14010029] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Revised: 12/10/2023] [Accepted: 12/19/2023] [Indexed: 01/23/2024] Open
Abstract
BACKGROUND Multiple sclerosis (MS) is an autoimmune disorder that affects the central nervous system (CNS) through inflammation. MS symptoms become acute if the disease progresses to the relapsing phase. AIM This review aimed to evaluate the role played by regulatory T cells (Tregs) in the pathogenesis of MS. METHODS This review used scholarly journal articles obtained from PubMed, PsycINFO, and CINAHL with different search parameters such as 'regulatory T cells', 'multiple sclerosis', and 'current knowledge'. The process of searching for articles was limited to those that had publication dates falling between 2010 and 2020. RESULTS Tregs play a role in the pathogenesis of MS. This conclusion is supported by animal disease models and environmental factors that can underlie Treg alterations in MS. Despite the knowledge of the role played by Tregs in MS pathogenesis, the specific subsets of Tregs involved in MS development remain incompletely understood. DISCUSSION This review provides an essential link between Tregs and MS activity. Targeting Tregs could be an efficient way to establish new treatment methods for MS management. CONCLUSION MS is a complex condition affecting many people worldwide. Research has shown that Tregs can influence MS development and progression. More investigations are needed to understand how Tregs affect the pathogenesis of MS.
Collapse
Affiliation(s)
- Borros Arneth
- Institute of Laboratory Medicine and Pathobiochemistry, Philipps University Marburg, 35043 Marburg, Germany;
- Institute of Laboratory Medicine and Pathobiochemistry, Justus Liebig University Giessen, 35392 Giessen, Germany
- Hospital of the Universities of Giessen and Marburg, 35392 Giessen, Germany
| |
Collapse
|
23
|
Thomas OG, Olsson T. Mimicking the brain: Epstein-Barr virus and foreign agents as drivers of neuroimmune attack in multiple sclerosis. Front Immunol 2023; 14:1304281. [PMID: 38022632 PMCID: PMC10655090 DOI: 10.3389/fimmu.2023.1304281] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Accepted: 10/23/2023] [Indexed: 12/01/2023] Open
Abstract
T cells have an essential role in adaptive immunity against pathogens and cancer, but failure of thymic tolerance mechanisms can instead lead to escape of T cells with the ability to attack host tissues. Multiple sclerosis (MS) occurs when structures such as myelin and neurons in the central nervous system (CNS) are the target of autoreactive immune responses, resulting in lesions in the brain and spinal cord which cause varied and episodic neurological deficits. A role for autoreactive T cell and antibody responses in MS is likely, and mounting evidence implicates Epstein-Barr virus (EBV) in disease mechanisms. In this review we discuss antigen specificity of T cells involved in development and progression of MS. We examine the current evidence that these T cells can target multiple antigens such as those from pathogens including EBV and briefly describe other mechanisms through which viruses could affect disease. Unravelling the complexity of the autoantigen T cell repertoire is essential for understanding key events in the development and progression of MS, with wider implications for development of future therapies.
Collapse
Affiliation(s)
- Olivia G. Thomas
- Therapeutic Immune Design, Centre for Molecular Medicine, Department of Clinical Neuroscience, Karolinska Institute, Stockholm, Sweden
- Neuroimmunology Unit, Department of Clinical Neuroscience, Centre for Molecular Medicine, Karolinska Institute, Stockholm, Sweden
| | - Tomas Olsson
- Therapeutic Immune Design, Centre for Molecular Medicine, Department of Clinical Neuroscience, Karolinska Institute, Stockholm, Sweden
| |
Collapse
|
24
|
Groß-Albenhausen E, Weier A, Velten M, Heider T, Chunder R, Kuerten S. Immune monitoring of SARS-CoV-2-specific T cell and B cell responses in patients with multiple sclerosis treated with ocrelizumab. Front Immunol 2023; 14:1254128. [PMID: 37841269 PMCID: PMC10569464 DOI: 10.3389/fimmu.2023.1254128] [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: 07/06/2023] [Accepted: 08/30/2023] [Indexed: 10/17/2023] Open
Abstract
Introduction Since the development of the coronavirus disease (COVID-19) pandemic caused by severe acute respiratory syndrome coronavirus type 2 (SARS-CoV-2), there has been significant interest in determining the effectiveness of SARS-CoV-2 vaccines in patients under immunomodulatory or immunosuppressive therapies. The aim of this study was to evaluate the impact of ocrelizumab, a monoclonal anti-CD20 antibody, on SARS-CoV-2-specific T cell and B cell responses in patients with relapsing-remitting multiple sclerosis (RRMS). Methods To this end, peripheral blood mononuclear cells (PBMCs) were isolated from n = 23 patients with RRMS. Of these patients, n = 17 were tested before (time point t0) and one month after (time point t1) their first dose of ocrelizumab. In addition, we studied n = 9 RRMS patients that got infected with SARS-CoV-2 over the course of ocrelizumab therapy (time point t2). PBMCs were also isolated from n = 19 age- and gender-matched healthy controls (HCs) after vaccination or infection with SARS-CoV-2, respectively. Interferon-γ (IFN-γ)/interleukin-2 (IL-2) and granzyme B (GzB)/perforin (PFN) double-color enzyme-linked immunospot (ELISPOT) assays or single-color ELISPOT assays were performed to measure SARS-CoV-2 antigen-specific T cell and B cell responses. Anti-viral antibody titers were quantified in the serum by chemiluminescence immunoassay. Results Our data indicate a significant difference in the SARS-CoV-2 specific IFN-γ (P = 0.0119) and PFN (P = 0.0005) secreting T cell compartment in the MS cohort at t0 compared to HCs. Following the first dose of ocrelizumab treatment, a significant decrease in the number of SARS-CoV-2 spike protein-specific B cells was observed (P = 0.0012). Infection with SARS-CoV-2 in MS patients under ocrelizumab therapy did not significantly alter their existing immune response against the virus. Kaplan-Meier survival analysis suggested that the spike S1 protein-specific immunoglobulin (Ig)G response might be a key parameter for predicting the probability of (re)infection with SARS-CoV-2. Discussion Our results call for a critical discussion regarding appropriate vaccination intervals and potential biomarkers for the prediction of (re)infection with SARS-CoV-2 in patients with MS receiving ocrelizumab. Unique identifier DRKS00029110; URL: http://apps.who.int/trialsearch/.
Collapse
Affiliation(s)
- Elina Groß-Albenhausen
- Institute of Neuroanatomy, Faculty of Medicine, University of Bonn and University Hospital Bonn, Bonn, Germany
| | - Alicia Weier
- Institute of Neuroanatomy, Faculty of Medicine, University of Bonn and University Hospital Bonn, Bonn, Germany
| | - Markus Velten
- Department of Anesthesiology and Intensive Care Medicine, University Medical Center Bonn, Bonn, Germany
| | - Thorsten Heider
- Clinic for Neurology, Klinikum St. Marien Amberg, Amberg, Germany
| | - Rittika Chunder
- Institute of Neuroanatomy, Faculty of Medicine, University of Bonn and University Hospital Bonn, Bonn, Germany
| | - Stefanie Kuerten
- Institute of Neuroanatomy, Faculty of Medicine, University of Bonn and University Hospital Bonn, Bonn, Germany
| |
Collapse
|
25
|
Wilhelm CR, Upadhye MA, Eschbacher KL, Karandikar NJ, Boyden AW. Proteolipid Protein-Induced Mouse Model of Multiple Sclerosis Requires B Cell-Mediated Antigen Presentation. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2023; 211:944-953. [PMID: 37548478 PMCID: PMC10528642 DOI: 10.4049/jimmunol.2200721] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Accepted: 07/18/2023] [Indexed: 08/08/2023]
Abstract
The pathogenic role B cells play in multiple sclerosis is underscored by the success of B cell depletion therapies. Yet, it remains unclear how B cells contribute to disease, although it is increasingly accepted that mechanisms beyond Ab production are involved. Better understanding of pathogenic interactions between B cells and autoreactive CD4 T cells will be critical for novel therapeutics. To focus the investigation on B cell:CD4 T cell interactions in vivo and in vitro, we previously developed a B cell-dependent, Ab-independent experimental autoimmune encephalomyelitis (EAE) mouse model driven by a peptide encompassing the extracellular domains of myelin proteolipid protein (PLPECD). In this study, we demonstrate that B cell depletion significantly inhibited PLPECD-induced EAE disease, blunted PLPECD-elicited delayed-type hypersensitivity reactions in vivo, and reduced CD4 T cell activation, proliferation, and proinflammatory cytokine production. Further, PLPECD-reactive CD4 T cells sourced from B cell-depleted donor mice failed to transfer EAE to naive recipients. Importantly, we identified B cell-mediated Ag presentation as the critical mechanism explaining B cell dependence in PLPECD-induced EAE, where bone marrow chimeric mice harboring a B cell-restricted MHC class II deficiency failed to develop EAE. B cells were ultimately observed to restimulate significantly higher Ag-specific proliferation from PLP178-191-reactive CD4 T cells compared with dendritic cells when provided PLPECD peptide in head-to-head cultures. We therefore conclude that PLPECD-induced EAE features a required pathogenic B cell-mediated Ag presentation function, providing for investigable B cell:CD4 T cell interactions in the context of autoimmune demyelinating disease.
Collapse
Affiliation(s)
- Connor R. Wilhelm
- Iowa City Veterans Affairs Medical Center
- Department of Pathology Graduate Program, University of Iowa, Iowa City, IA USA
| | - Mohit A. Upadhye
- Iowa City Veterans Affairs Medical Center
- Department of Pathology Graduate Program, University of Iowa, Iowa City, IA USA
| | | | - Nitin J. Karandikar
- Department of Pathology, University of Iowa Carver College of Medicine
- Iowa City Veterans Affairs Medical Center
- Department of Pathology Graduate Program, University of Iowa, Iowa City, IA USA
| | - Alexander W. Boyden
- Department of Pathology, University of Iowa Carver College of Medicine
- Iowa City Veterans Affairs Medical Center
| |
Collapse
|
26
|
von Essen MR, Talbot J, Hansen RHH, Chow HH, Lundell H, Siebner HR, Sellebjerg F. Intrathecal CD8 +CD20 + T Cells in Primary Progressive Multiple Sclerosis. NEUROLOGY(R) NEUROIMMUNOLOGY & NEUROINFLAMMATION 2023; 10:e200140. [PMID: 37369602 DOI: 10.1212/nxi.0000000000200140] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Accepted: 05/15/2023] [Indexed: 06/29/2023]
Abstract
BACKGROUND AND OBJECTIVE Despite accumulating evidence of intrathecal inflammation in patients with primary progressive multiple sclerosis (PPMS), immunomodulatory and suppressive treatment strategies have proven unsuccessful. With this study, we investigated the involvement of CD20+ T cells and the effect of dimethyl fumarate on CD20+ T cells in PPMS. METHODS The main outcomes in this observational, case-control study were flow cytometry assessments of blood and CSF CD20+ T cells and ELISA measurements of myelin basic protein and neurofilament light chain in untreated patients with PPMS and patients treated for 48 weeks with dimethyl fumarate or placebo. MRI measures included new and enlarging T2-weighted lesions over 48 weeks and lesion, normal-appearing white matter, cortical, and thalamic volume. RESULTS Assessing CD20+ T cells in patients with PPMS and controls showed an increased percentage of CD20+ T cells in the blood of untreated patients and a strong enrichment in the CSF. In addition, a higher frequency of CD8+CD20+ T cells in the CSF correlated with a higher concentration of myelin basic protein and T2-weighted lesion volume and with a lower normal-appearing white matter and thalamus volume. Furthermore, CD8+CD20+ T cells were associated with the development of new T2 lesions. After 48 weeks of treatment with dimethyl fumarate, total T cells in CSF were reduced; however, CD20+ T cells were unaffected. DISCUSSION This study shows an association between intrathecal CD8+CD20+ T cells, white matter injury, and thalamic atrophy in PPMS, suggesting a role of CD8+CD20+ T cells in the immunopathogenesis of PPMS. The results also suggest that limited efficacy of dimethyl fumarate in PPMS may, at least partly, be a consequence of failure to suppress CD8+CD20+ T cells in CSF.
Collapse
Affiliation(s)
- Marina Rode von Essen
- From the Danish Multiple Sclerosis Center (M.R.E., J.T., R.H.H.H., H.H.C., F.S.), Department of Neurology, Copenhagen University Hospital - Rigshospitalet, Glostrup; Danish Research Centre for Magnetic Resonance (H.L., H.R.S.), Copenhagen University Hospital - Amager and Hvidovre; Department of Clinical Medicine (H.R.S.), University of Copenhagen; and Department of Neurology (H.R.S.), Copenhagen University Hospital - Bispebjerg and Frederiksberg, Denmark.
| | - Jacob Talbot
- From the Danish Multiple Sclerosis Center (M.R.E., J.T., R.H.H.H., H.H.C., F.S.), Department of Neurology, Copenhagen University Hospital - Rigshospitalet, Glostrup; Danish Research Centre for Magnetic Resonance (H.L., H.R.S.), Copenhagen University Hospital - Amager and Hvidovre; Department of Clinical Medicine (H.R.S.), University of Copenhagen; and Department of Neurology (H.R.S.), Copenhagen University Hospital - Bispebjerg and Frederiksberg, Denmark
| | - Rikke Holm Holm Hansen
- From the Danish Multiple Sclerosis Center (M.R.E., J.T., R.H.H.H., H.H.C., F.S.), Department of Neurology, Copenhagen University Hospital - Rigshospitalet, Glostrup; Danish Research Centre for Magnetic Resonance (H.L., H.R.S.), Copenhagen University Hospital - Amager and Hvidovre; Department of Clinical Medicine (H.R.S.), University of Copenhagen; and Department of Neurology (H.R.S.), Copenhagen University Hospital - Bispebjerg and Frederiksberg, Denmark
| | - Helene Højsgaard Chow
- From the Danish Multiple Sclerosis Center (M.R.E., J.T., R.H.H.H., H.H.C., F.S.), Department of Neurology, Copenhagen University Hospital - Rigshospitalet, Glostrup; Danish Research Centre for Magnetic Resonance (H.L., H.R.S.), Copenhagen University Hospital - Amager and Hvidovre; Department of Clinical Medicine (H.R.S.), University of Copenhagen; and Department of Neurology (H.R.S.), Copenhagen University Hospital - Bispebjerg and Frederiksberg, Denmark
| | - Henrik Lundell
- From the Danish Multiple Sclerosis Center (M.R.E., J.T., R.H.H.H., H.H.C., F.S.), Department of Neurology, Copenhagen University Hospital - Rigshospitalet, Glostrup; Danish Research Centre for Magnetic Resonance (H.L., H.R.S.), Copenhagen University Hospital - Amager and Hvidovre; Department of Clinical Medicine (H.R.S.), University of Copenhagen; and Department of Neurology (H.R.S.), Copenhagen University Hospital - Bispebjerg and Frederiksberg, Denmark
| | - Hartwig Roman Siebner
- From the Danish Multiple Sclerosis Center (M.R.E., J.T., R.H.H.H., H.H.C., F.S.), Department of Neurology, Copenhagen University Hospital - Rigshospitalet, Glostrup; Danish Research Centre for Magnetic Resonance (H.L., H.R.S.), Copenhagen University Hospital - Amager and Hvidovre; Department of Clinical Medicine (H.R.S.), University of Copenhagen; and Department of Neurology (H.R.S.), Copenhagen University Hospital - Bispebjerg and Frederiksberg, Denmark
| | - Finn Sellebjerg
- From the Danish Multiple Sclerosis Center (M.R.E., J.T., R.H.H.H., H.H.C., F.S.), Department of Neurology, Copenhagen University Hospital - Rigshospitalet, Glostrup; Danish Research Centre for Magnetic Resonance (H.L., H.R.S.), Copenhagen University Hospital - Amager and Hvidovre; Department of Clinical Medicine (H.R.S.), University of Copenhagen; and Department of Neurology (H.R.S.), Copenhagen University Hospital - Bispebjerg and Frederiksberg, Denmark
| |
Collapse
|
27
|
Jiang N, Malone M, Chizari S. Antigen-specific and cross-reactive T cells in protection and disease. Immunol Rev 2023; 316:120-135. [PMID: 37209375 PMCID: PMC10524458 DOI: 10.1111/imr.13217] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2023] [Revised: 04/25/2023] [Accepted: 04/27/2023] [Indexed: 05/22/2023]
Abstract
Human T cells have a diverse T-cell receptor (TCR) repertoire that endows them with the ability to identify and defend against a broad spectrum of antigens. The universe of possible antigens that T cells may encounter, however, is even larger. To effectively surveil such a vast universe, the T-cell repertoire must adopt a high degree of cross-reactivity. Likewise, antigen-specific and cross-reactive T-cell responses play pivotal roles in both protective and pathological immune responses in numerous diseases. In this review, we explore the implications of these antigen-driven T-cell responses, with a particular focus on CD8+ T cells, using infection, neurodegeneration, and cancer as examples. We also summarize recent technological advances that facilitate high-throughput profiling of antigen-specific and cross-reactive T-cell responses experimentally, as well as computational biology approaches that predict these interactions.
Collapse
Affiliation(s)
- Ning Jiang
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, 19104
- Institute for Immunology, University of Pennsylvania, Philadelphia, PA, 19104
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, 19104
- Center for Cellular Immunotherapies, University of Pennsylvania, Philadelphia, PA, 19104
- Institute for RNA Innovation, University of Pennsylvania, Philadelphia, PA, 19104
| | - Michael Malone
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, 19104
| | - Shahab Chizari
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, 19104
| |
Collapse
|
28
|
Otero-Romero S, Lebrun-Frénay C, Reyes S, Amato MP, Campins M, Farez M, Filippi M, Hacohen Y, Hemmer B, Juuti R, Magyari M, Oreja-Guevara C, Siva A, Vukusic S, Tintoré M. ECTRIMS/EAN consensus on vaccination in people with multiple sclerosis: Improving immunization strategies in the era of highly active immunotherapeutic drugs. Mult Scler 2023; 29:904-925. [PMID: 37293841 PMCID: PMC10338708 DOI: 10.1177/13524585231168043] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Revised: 01/30/2023] [Accepted: 03/19/2023] [Indexed: 06/10/2023]
Abstract
BACKGROUND With the new highly active drugs available for people with multiple sclerosis (pwMS), vaccination becomes an essential part of the risk management strategy. OBJECTIVE To develop a European evidence-based consensus for the vaccination strategy of pwMS who are candidates for disease-modifying therapies (DMTs). METHODS This work was conducted by a multidisciplinary working group using formal consensus methodology. Clinical questions (defined as population, interventions, and outcomes) considered all authorized DMTs and vaccines. A systematic literature search was conducted and quality of evidence was defined according to the Oxford Centre for Evidence-Based Medicine Levels of Evidence. The recommendations were formulated based on the quality of evidence and the risk-benefit balance. RESULTS Seven questions, encompassing vaccine safety, vaccine effectiveness, global vaccination strategy and vaccination in sub-populations (pediatric, pregnant women, elderly and international travelers) were considered. A narrative description of the evidence considering published studies, guidelines, and position statements is presented. A total of 53 recommendations were agreed by the working group after three rounds of consensus. CONCLUSION This first European consensus on vaccination in pwMS proposes the best vaccination strategy according to current evidence and expert knowledge, with the goal of homogenizing the immunization practices in pwMS.
Collapse
Affiliation(s)
- Susana Otero-Romero
- Department of Preventive Medicine and Epidemiology, Vall d’Hebron Barcelona Hospital, Barcelona, Spain Multiple Sclerosis Centre of Catalonia (Cemcat), Vall d’Hebron Barcelona Hospital, Barcelona, Spain
| | | | - Saúl Reyes
- Fundación Santa Fe de Bogotá, Bogotá, Colombia School of Medicine, Universidad de los Andes, Bogotá, Colombia Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Maria Pia Amato
- Department NEUROFARBA, University of Florence, Florence, Italy IRCCS Fondazione Don Carlo Gnocchi, Florence, Italy
| | - Magda Campins
- Department of Preventive Medicine and Epidemiology, Vall d’Hebron Barcelona Hospital, Barcelona, Spain
| | - Mauricio Farez
- Centro para la Investigación de Enfermedades Neuroinmunológicas (CIEN), FLENI, Buenos Aires, Argentina
| | - Massimo Filippi
- Neuroimaging Research Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy Neurology Unit, Neurorehabilitation Unit, and Neurophysiology Service, IRCCS San Raffaele Scientific Institute, Milan, Italy Vita-Salute San Raffaele University, Milan, Italy
| | - Yael Hacohen
- Department of Paediatric Neurology, Great Ormond Street Hospital for Children, London, UK Department of Neuroinflammation, Queen Square Multiple Sclerosis Centre, UCL Institute of Neurology, London, UK
| | - Bernhard Hemmer
- Department of Neurology, Klinikum Rechts der Isar, School of Medicine, Technical University of Munich, Munich, Germany Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Rosa Juuti
- Multiple Sclerosis International Federation, London, UK
| | - Melinda Magyari
- Department of Neurology, Danish Multiple Sclerosis Center and the Danish Multiple Sclerosis Registry, Rigshospitalet and University of Copenhagen, Copenhagen, Denmark
| | - Celia Oreja-Guevara
- Department of Neurology, Hospital Clínico San Carlos, IdISSC, Departamento de Medicina, Universidad Complutense, Madrid, Spain
| | - Aksel Siva
- Department of Neurology, School of Medicine, Istanbul University Cerrahpasa, Cerrahpasa, Istanbul, Turkey
| | - Sandra Vukusic
- Service de Neurologie, Sclérose en Plaques, Pathologies de la Myéline et Neuro-inflammation, Centre de Référence des Maladies Inflammatoires Rares du Cerveau et de la Moelle, Hôpital Neurologique Pierre Wertheimer, Hospices Civils de Lyon, Lyon, France
- Centre des Neurosciences de Lyon, Observatoire Français de la Sclérose en Plaques, INSERM 1028 et CNRS UMR5292, Lyon, France Université Claude Bernard Lyon 1, Faculté de Médecine Lyon Est, Lyon, France
| | - Mar Tintoré
- Multiple Sclerosis Centre of Catalonia (Cemcat), Vall d’Hebron Barcelona Hospital, Barcelona, Spain
| |
Collapse
|
29
|
Laurent SA, Strauli NB, Eggers EL, Wu H, Michel B, Demuth S, Palanichamy A, Wilson MR, Sirota M, Hernandez RD, Cree BAC, Herman AE, von Büdingen HC. Effect of Ocrelizumab on B- and T-Cell Receptor Repertoire Diversity in Patients With Relapsing Multiple Sclerosis From the Randomized Phase III OPERA Trial. NEUROLOGY(R) NEUROIMMUNOLOGY & NEUROINFLAMMATION 2023; 10:e200118. [PMID: 37094998 PMCID: PMC10136682 DOI: 10.1212/nxi.0000000000200118] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Accepted: 02/22/2023] [Indexed: 04/26/2023]
Abstract
BACKGROUND AND OBJECTIVES The B cell-depleting anti-CD20 antibody ocrelizumab (OCR) effectively reduces MS disease activity and slows disability progression. Given the role of B cells as antigen-presenting cells, the primary goal of this study was to evaluate the effect of OCR on the T-cell receptor repertoire diversity. METHODS To examine whether OCR substantially alters the molecular diversity of the T-cell receptor repertoire, deep immune repertoire sequencing (RepSeq) of CD4+ and CD8+ T-cell receptor β-chain variable regions was performed on longitudinal blood samples. The IgM and IgG heavy chain variable region repertoire was also analyzed to characterize the residual B-cell repertoire under OCR treatment. RESULTS Peripheral blood samples for RepSeq were obtained from 8 patients with relapsing MS enrolled in the OPERA I trial over a period of up to 39 months. Four patients each were treated with OCR or interferon β1-a during the double-blind period of OPERA I. All patients received OCR during the open-label extension. The diversity of the CD4+/CD8+ T-cell repertoires remained unaffected in OCR-treated patients. The expected OCR-associated B-cell depletion was mirrored by reduced B-cell receptor diversity in peripheral blood and a shift in immunoglobulin gene usage. Despite deep B-cell depletion, longitudinal persistence of clonally related B-cells was observed. DISCUSSION Our data illustrate that the diversity of CD4+/CD8+ T-cell receptor repertoires remained unaltered in OCR-treated patients with relapsing MS. Persistence of a highly diverse T-cell repertoire suggests that aspects of adaptive immunity remain intact despite extended anti-CD20 therapy. TRIAL REGISTRATION INFORMATION This is a substudy (BE29353) of the OPERA I (WA21092; NCT01247324) trial. Date of registration, November 23, 2010; first patient enrollment, August 31, 2011.
Collapse
Affiliation(s)
- Sarah A Laurent
- From the Department of Neurology (S.A.L., E.L.E., H.W., B.M., S.D., A.P., M.R.W., B.A.C.C., H.-C.B.), Weill Institute for Neurosciences; Biomedical Sciences Graduate Program (N.B.S.); Bakar Computational Health Sciences Institute and Department of Pediatrics (M.S.); Department of Bioengineering and Therapeutic Sciences (R.D.H.), University of California, San Francisco, CA; Department of Human Genetics (R.D.H.), McGill University, Montreal, QC, Canada; and OMNI Biomarker Development (A.E.H.), Genentech, Inc., South San Francisco, CA
| | - Nicolas B Strauli
- From the Department of Neurology (S.A.L., E.L.E., H.W., B.M., S.D., A.P., M.R.W., B.A.C.C., H.-C.B.), Weill Institute for Neurosciences; Biomedical Sciences Graduate Program (N.B.S.); Bakar Computational Health Sciences Institute and Department of Pediatrics (M.S.); Department of Bioengineering and Therapeutic Sciences (R.D.H.), University of California, San Francisco, CA; Department of Human Genetics (R.D.H.), McGill University, Montreal, QC, Canada; and OMNI Biomarker Development (A.E.H.), Genentech, Inc., South San Francisco, CA
| | - Erica L Eggers
- From the Department of Neurology (S.A.L., E.L.E., H.W., B.M., S.D., A.P., M.R.W., B.A.C.C., H.-C.B.), Weill Institute for Neurosciences; Biomedical Sciences Graduate Program (N.B.S.); Bakar Computational Health Sciences Institute and Department of Pediatrics (M.S.); Department of Bioengineering and Therapeutic Sciences (R.D.H.), University of California, San Francisco, CA; Department of Human Genetics (R.D.H.), McGill University, Montreal, QC, Canada; and OMNI Biomarker Development (A.E.H.), Genentech, Inc., South San Francisco, CA
| | - Hao Wu
- From the Department of Neurology (S.A.L., E.L.E., H.W., B.M., S.D., A.P., M.R.W., B.A.C.C., H.-C.B.), Weill Institute for Neurosciences; Biomedical Sciences Graduate Program (N.B.S.); Bakar Computational Health Sciences Institute and Department of Pediatrics (M.S.); Department of Bioengineering and Therapeutic Sciences (R.D.H.), University of California, San Francisco, CA; Department of Human Genetics (R.D.H.), McGill University, Montreal, QC, Canada; and OMNI Biomarker Development (A.E.H.), Genentech, Inc., South San Francisco, CA
| | - Brady Michel
- From the Department of Neurology (S.A.L., E.L.E., H.W., B.M., S.D., A.P., M.R.W., B.A.C.C., H.-C.B.), Weill Institute for Neurosciences; Biomedical Sciences Graduate Program (N.B.S.); Bakar Computational Health Sciences Institute and Department of Pediatrics (M.S.); Department of Bioengineering and Therapeutic Sciences (R.D.H.), University of California, San Francisco, CA; Department of Human Genetics (R.D.H.), McGill University, Montreal, QC, Canada; and OMNI Biomarker Development (A.E.H.), Genentech, Inc., South San Francisco, CA
| | - Stanislas Demuth
- From the Department of Neurology (S.A.L., E.L.E., H.W., B.M., S.D., A.P., M.R.W., B.A.C.C., H.-C.B.), Weill Institute for Neurosciences; Biomedical Sciences Graduate Program (N.B.S.); Bakar Computational Health Sciences Institute and Department of Pediatrics (M.S.); Department of Bioengineering and Therapeutic Sciences (R.D.H.), University of California, San Francisco, CA; Department of Human Genetics (R.D.H.), McGill University, Montreal, QC, Canada; and OMNI Biomarker Development (A.E.H.), Genentech, Inc., South San Francisco, CA
| | - Arumugam Palanichamy
- From the Department of Neurology (S.A.L., E.L.E., H.W., B.M., S.D., A.P., M.R.W., B.A.C.C., H.-C.B.), Weill Institute for Neurosciences; Biomedical Sciences Graduate Program (N.B.S.); Bakar Computational Health Sciences Institute and Department of Pediatrics (M.S.); Department of Bioengineering and Therapeutic Sciences (R.D.H.), University of California, San Francisco, CA; Department of Human Genetics (R.D.H.), McGill University, Montreal, QC, Canada; and OMNI Biomarker Development (A.E.H.), Genentech, Inc., South San Francisco, CA
| | - Michael R Wilson
- From the Department of Neurology (S.A.L., E.L.E., H.W., B.M., S.D., A.P., M.R.W., B.A.C.C., H.-C.B.), Weill Institute for Neurosciences; Biomedical Sciences Graduate Program (N.B.S.); Bakar Computational Health Sciences Institute and Department of Pediatrics (M.S.); Department of Bioengineering and Therapeutic Sciences (R.D.H.), University of California, San Francisco, CA; Department of Human Genetics (R.D.H.), McGill University, Montreal, QC, Canada; and OMNI Biomarker Development (A.E.H.), Genentech, Inc., South San Francisco, CA
| | - Marina Sirota
- From the Department of Neurology (S.A.L., E.L.E., H.W., B.M., S.D., A.P., M.R.W., B.A.C.C., H.-C.B.), Weill Institute for Neurosciences; Biomedical Sciences Graduate Program (N.B.S.); Bakar Computational Health Sciences Institute and Department of Pediatrics (M.S.); Department of Bioengineering and Therapeutic Sciences (R.D.H.), University of California, San Francisco, CA; Department of Human Genetics (R.D.H.), McGill University, Montreal, QC, Canada; and OMNI Biomarker Development (A.E.H.), Genentech, Inc., South San Francisco, CA
| | - Ryan D Hernandez
- From the Department of Neurology (S.A.L., E.L.E., H.W., B.M., S.D., A.P., M.R.W., B.A.C.C., H.-C.B.), Weill Institute for Neurosciences; Biomedical Sciences Graduate Program (N.B.S.); Bakar Computational Health Sciences Institute and Department of Pediatrics (M.S.); Department of Bioengineering and Therapeutic Sciences (R.D.H.), University of California, San Francisco, CA; Department of Human Genetics (R.D.H.), McGill University, Montreal, QC, Canada; and OMNI Biomarker Development (A.E.H.), Genentech, Inc., South San Francisco, CA
| | - Bruce Anthony Campbell Cree
- From the Department of Neurology (S.A.L., E.L.E., H.W., B.M., S.D., A.P., M.R.W., B.A.C.C., H.-C.B.), Weill Institute for Neurosciences; Biomedical Sciences Graduate Program (N.B.S.); Bakar Computational Health Sciences Institute and Department of Pediatrics (M.S.); Department of Bioengineering and Therapeutic Sciences (R.D.H.), University of California, San Francisco, CA; Department of Human Genetics (R.D.H.), McGill University, Montreal, QC, Canada; and OMNI Biomarker Development (A.E.H.), Genentech, Inc., South San Francisco, CA
| | - Ann E Herman
- From the Department of Neurology (S.A.L., E.L.E., H.W., B.M., S.D., A.P., M.R.W., B.A.C.C., H.-C.B.), Weill Institute for Neurosciences; Biomedical Sciences Graduate Program (N.B.S.); Bakar Computational Health Sciences Institute and Department of Pediatrics (M.S.); Department of Bioengineering and Therapeutic Sciences (R.D.H.), University of California, San Francisco, CA; Department of Human Genetics (R.D.H.), McGill University, Montreal, QC, Canada; and OMNI Biomarker Development (A.E.H.), Genentech, Inc., South San Francisco, CA
| | - H-Christian von Büdingen
- From the Department of Neurology (S.A.L., E.L.E., H.W., B.M., S.D., A.P., M.R.W., B.A.C.C., H.-C.B.), Weill Institute for Neurosciences; Biomedical Sciences Graduate Program (N.B.S.); Bakar Computational Health Sciences Institute and Department of Pediatrics (M.S.); Department of Bioengineering and Therapeutic Sciences (R.D.H.), University of California, San Francisco, CA; Department of Human Genetics (R.D.H.), McGill University, Montreal, QC, Canada; and OMNI Biomarker Development (A.E.H.), Genentech, Inc., South San Francisco, CA.
| |
Collapse
|
30
|
Pfeuffer S, Rolfes L, Ingwersen J, Pul R, Kleinschnitz K, Korsen M, Räuber S, Ruck T, Schieferdecker S, Willison AG, Aktas O, Kleinschnitz C, Hartung HP, Kappos L, Meuth SG. Effect of Previous Disease-Modifying Therapy on Treatment Effectiveness for Patients Treated With Ocrelizumab. NEUROLOGY(R) NEUROIMMUNOLOGY & NEUROINFLAMMATION 2023; 10:10/3/e200104. [PMID: 37041077 PMCID: PMC10091366 DOI: 10.1212/nxi.0000000000200104] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Accepted: 01/27/2023] [Indexed: 04/13/2023]
Abstract
BACKGROUND AND OBJECTIVES B cell-depleting antibodies were proven as effective strategy for the treatment of relapsing multiple sclerosis (RMS). The monoclonal antibody ocrelizumab was approved in 2017 in the United States and in 2018 in the European Union, but despite proven efficacy in randomized, controlled clinical trials, its effectiveness in the real-world setting remains to be fully elucidated. In particular, most study patients were treatment naive or switched from injectable therapies, whereas oral substances or monoclonal antibodies made up >1% of previous treatments. METHODS We evaluated ocrelizumab-treated patients with RMS enrolled in the prospective cohorts at the University Hospitals Duesseldorf and Essen, Germany. Epidemiologic data at baseline were compared, and Cox proportional hazard models were applied to evaluate outcomes. RESULTS Two hundred eighty patients were included (median age: 37 years, 35% male patients). Compared with using ocrelizumab as a first-line treatment, its use as a third-line therapy increased hazard ratios (HRs) for relapse and disability progression, whereas differences between first- vs second-line and second- vs third-line remained smaller. We stratified patients according to their last previous disease-modifying treatment and here identified fingolimod (FTY) (45 patients, median age 40 years, 33% male patients) as a relevant risk factor for ongoing relapse activity despite 2nd-line (HR: 3.417 [1.007-11.600]) or 3rd-line (HR: 5.903 [2.489-13.999]) ocrelizumab treatment, disability worsening (2nd line: HR: 3.571 [1.013-12.589]; 3rd line: HR: 4.502 [1.728-11.729]), and occurrence of new/enlarging MRI lesions (2nd line: HR: 1.939 [0.604-6.228]; 3rd line: HR: 4.627 [1.982-10.802]). Effects were persistent throughout the whole follow-up. Neither peripheral B-cell repopulation nor immunoglobulin G levels were associated with rekindling disease activity. DISCUSSION Our prospectively collected observational data suggest suboptimal effectiveness of ocrelizumab in patients switching from FTY compared with those switching from other substances or having been treatment naive. These findings support previous studies indicating abated effectiveness of immune cell-depleting therapies following FTY treatment in patients with RMS. CLASSIFICATION OF EVIDENCE This study provides Class IV evidence that for patients with RMS, previous treatment with FTY compared with previous treatment with other immunomodulating therapies decreases the effectiveness of ocrelizumab.
Collapse
Affiliation(s)
- Steffen Pfeuffer
- From the Department of Neurology (S.P.), University Hospital Giessen and Marburg, Justus-Liebig-University Giessen; Department of Neurology (L.R., J.I., M.K., S.R., T.R., S.S., A.G.W., O.A., H.-P.H., S.G.M.), University Hospital Duesseldorf, Germany; Brain and Mind Center (H.-P.H.), University of Sydney, NSW, Australia; Department of Neurology (H.-P.H.), Palacky University, Olomouc, Czech Republic; Department of Neurology (H.-P.H.), Medical University of Vienna, Austria; Department of Neurology and Centre for Translational Neuro- and Behavioural Sciences (C-TNBS) (R.P., K.K., C.K.), University Hospital Essen, Germany; and Neurologic Clinic and Policlinic (L.K.), Departments of Medicine, Biomedicine and Clinical Research, University Hospital Basel, University of Basel, Switzerland.
| | - Leoni Rolfes
- From the Department of Neurology (S.P.), University Hospital Giessen and Marburg, Justus-Liebig-University Giessen; Department of Neurology (L.R., J.I., M.K., S.R., T.R., S.S., A.G.W., O.A., H.-P.H., S.G.M.), University Hospital Duesseldorf, Germany; Brain and Mind Center (H.-P.H.), University of Sydney, NSW, Australia; Department of Neurology (H.-P.H.), Palacky University, Olomouc, Czech Republic; Department of Neurology (H.-P.H.), Medical University of Vienna, Austria; Department of Neurology and Centre for Translational Neuro- and Behavioural Sciences (C-TNBS) (R.P., K.K., C.K.), University Hospital Essen, Germany; and Neurologic Clinic and Policlinic (L.K.), Departments of Medicine, Biomedicine and Clinical Research, University Hospital Basel, University of Basel, Switzerland
| | - Jens Ingwersen
- From the Department of Neurology (S.P.), University Hospital Giessen and Marburg, Justus-Liebig-University Giessen; Department of Neurology (L.R., J.I., M.K., S.R., T.R., S.S., A.G.W., O.A., H.-P.H., S.G.M.), University Hospital Duesseldorf, Germany; Brain and Mind Center (H.-P.H.), University of Sydney, NSW, Australia; Department of Neurology (H.-P.H.), Palacky University, Olomouc, Czech Republic; Department of Neurology (H.-P.H.), Medical University of Vienna, Austria; Department of Neurology and Centre for Translational Neuro- and Behavioural Sciences (C-TNBS) (R.P., K.K., C.K.), University Hospital Essen, Germany; and Neurologic Clinic and Policlinic (L.K.), Departments of Medicine, Biomedicine and Clinical Research, University Hospital Basel, University of Basel, Switzerland
| | - Refik Pul
- From the Department of Neurology (S.P.), University Hospital Giessen and Marburg, Justus-Liebig-University Giessen; Department of Neurology (L.R., J.I., M.K., S.R., T.R., S.S., A.G.W., O.A., H.-P.H., S.G.M.), University Hospital Duesseldorf, Germany; Brain and Mind Center (H.-P.H.), University of Sydney, NSW, Australia; Department of Neurology (H.-P.H.), Palacky University, Olomouc, Czech Republic; Department of Neurology (H.-P.H.), Medical University of Vienna, Austria; Department of Neurology and Centre for Translational Neuro- and Behavioural Sciences (C-TNBS) (R.P., K.K., C.K.), University Hospital Essen, Germany; and Neurologic Clinic and Policlinic (L.K.), Departments of Medicine, Biomedicine and Clinical Research, University Hospital Basel, University of Basel, Switzerland
| | - Konstanze Kleinschnitz
- From the Department of Neurology (S.P.), University Hospital Giessen and Marburg, Justus-Liebig-University Giessen; Department of Neurology (L.R., J.I., M.K., S.R., T.R., S.S., A.G.W., O.A., H.-P.H., S.G.M.), University Hospital Duesseldorf, Germany; Brain and Mind Center (H.-P.H.), University of Sydney, NSW, Australia; Department of Neurology (H.-P.H.), Palacky University, Olomouc, Czech Republic; Department of Neurology (H.-P.H.), Medical University of Vienna, Austria; Department of Neurology and Centre for Translational Neuro- and Behavioural Sciences (C-TNBS) (R.P., K.K., C.K.), University Hospital Essen, Germany; and Neurologic Clinic and Policlinic (L.K.), Departments of Medicine, Biomedicine and Clinical Research, University Hospital Basel, University of Basel, Switzerland
| | - Melanie Korsen
- From the Department of Neurology (S.P.), University Hospital Giessen and Marburg, Justus-Liebig-University Giessen; Department of Neurology (L.R., J.I., M.K., S.R., T.R., S.S., A.G.W., O.A., H.-P.H., S.G.M.), University Hospital Duesseldorf, Germany; Brain and Mind Center (H.-P.H.), University of Sydney, NSW, Australia; Department of Neurology (H.-P.H.), Palacky University, Olomouc, Czech Republic; Department of Neurology (H.-P.H.), Medical University of Vienna, Austria; Department of Neurology and Centre for Translational Neuro- and Behavioural Sciences (C-TNBS) (R.P., K.K., C.K.), University Hospital Essen, Germany; and Neurologic Clinic and Policlinic (L.K.), Departments of Medicine, Biomedicine and Clinical Research, University Hospital Basel, University of Basel, Switzerland
| | - Saskia Räuber
- From the Department of Neurology (S.P.), University Hospital Giessen and Marburg, Justus-Liebig-University Giessen; Department of Neurology (L.R., J.I., M.K., S.R., T.R., S.S., A.G.W., O.A., H.-P.H., S.G.M.), University Hospital Duesseldorf, Germany; Brain and Mind Center (H.-P.H.), University of Sydney, NSW, Australia; Department of Neurology (H.-P.H.), Palacky University, Olomouc, Czech Republic; Department of Neurology (H.-P.H.), Medical University of Vienna, Austria; Department of Neurology and Centre for Translational Neuro- and Behavioural Sciences (C-TNBS) (R.P., K.K., C.K.), University Hospital Essen, Germany; and Neurologic Clinic and Policlinic (L.K.), Departments of Medicine, Biomedicine and Clinical Research, University Hospital Basel, University of Basel, Switzerland
| | - Tobias Ruck
- From the Department of Neurology (S.P.), University Hospital Giessen and Marburg, Justus-Liebig-University Giessen; Department of Neurology (L.R., J.I., M.K., S.R., T.R., S.S., A.G.W., O.A., H.-P.H., S.G.M.), University Hospital Duesseldorf, Germany; Brain and Mind Center (H.-P.H.), University of Sydney, NSW, Australia; Department of Neurology (H.-P.H.), Palacky University, Olomouc, Czech Republic; Department of Neurology (H.-P.H.), Medical University of Vienna, Austria; Department of Neurology and Centre for Translational Neuro- and Behavioural Sciences (C-TNBS) (R.P., K.K., C.K.), University Hospital Essen, Germany; and Neurologic Clinic and Policlinic (L.K.), Departments of Medicine, Biomedicine and Clinical Research, University Hospital Basel, University of Basel, Switzerland
| | - Simon Schieferdecker
- From the Department of Neurology (S.P.), University Hospital Giessen and Marburg, Justus-Liebig-University Giessen; Department of Neurology (L.R., J.I., M.K., S.R., T.R., S.S., A.G.W., O.A., H.-P.H., S.G.M.), University Hospital Duesseldorf, Germany; Brain and Mind Center (H.-P.H.), University of Sydney, NSW, Australia; Department of Neurology (H.-P.H.), Palacky University, Olomouc, Czech Republic; Department of Neurology (H.-P.H.), Medical University of Vienna, Austria; Department of Neurology and Centre for Translational Neuro- and Behavioural Sciences (C-TNBS) (R.P., K.K., C.K.), University Hospital Essen, Germany; and Neurologic Clinic and Policlinic (L.K.), Departments of Medicine, Biomedicine and Clinical Research, University Hospital Basel, University of Basel, Switzerland
| | - Alice Grizzel Willison
- From the Department of Neurology (S.P.), University Hospital Giessen and Marburg, Justus-Liebig-University Giessen; Department of Neurology (L.R., J.I., M.K., S.R., T.R., S.S., A.G.W., O.A., H.-P.H., S.G.M.), University Hospital Duesseldorf, Germany; Brain and Mind Center (H.-P.H.), University of Sydney, NSW, Australia; Department of Neurology (H.-P.H.), Palacky University, Olomouc, Czech Republic; Department of Neurology (H.-P.H.), Medical University of Vienna, Austria; Department of Neurology and Centre for Translational Neuro- and Behavioural Sciences (C-TNBS) (R.P., K.K., C.K.), University Hospital Essen, Germany; and Neurologic Clinic and Policlinic (L.K.), Departments of Medicine, Biomedicine and Clinical Research, University Hospital Basel, University of Basel, Switzerland
| | - Orhan Aktas
- From the Department of Neurology (S.P.), University Hospital Giessen and Marburg, Justus-Liebig-University Giessen; Department of Neurology (L.R., J.I., M.K., S.R., T.R., S.S., A.G.W., O.A., H.-P.H., S.G.M.), University Hospital Duesseldorf, Germany; Brain and Mind Center (H.-P.H.), University of Sydney, NSW, Australia; Department of Neurology (H.-P.H.), Palacky University, Olomouc, Czech Republic; Department of Neurology (H.-P.H.), Medical University of Vienna, Austria; Department of Neurology and Centre for Translational Neuro- and Behavioural Sciences (C-TNBS) (R.P., K.K., C.K.), University Hospital Essen, Germany; and Neurologic Clinic and Policlinic (L.K.), Departments of Medicine, Biomedicine and Clinical Research, University Hospital Basel, University of Basel, Switzerland
| | - Christoph Kleinschnitz
- From the Department of Neurology (S.P.), University Hospital Giessen and Marburg, Justus-Liebig-University Giessen; Department of Neurology (L.R., J.I., M.K., S.R., T.R., S.S., A.G.W., O.A., H.-P.H., S.G.M.), University Hospital Duesseldorf, Germany; Brain and Mind Center (H.-P.H.), University of Sydney, NSW, Australia; Department of Neurology (H.-P.H.), Palacky University, Olomouc, Czech Republic; Department of Neurology (H.-P.H.), Medical University of Vienna, Austria; Department of Neurology and Centre for Translational Neuro- and Behavioural Sciences (C-TNBS) (R.P., K.K., C.K.), University Hospital Essen, Germany; and Neurologic Clinic and Policlinic (L.K.), Departments of Medicine, Biomedicine and Clinical Research, University Hospital Basel, University of Basel, Switzerland
| | - Hans-Peter Hartung
- From the Department of Neurology (S.P.), University Hospital Giessen and Marburg, Justus-Liebig-University Giessen; Department of Neurology (L.R., J.I., M.K., S.R., T.R., S.S., A.G.W., O.A., H.-P.H., S.G.M.), University Hospital Duesseldorf, Germany; Brain and Mind Center (H.-P.H.), University of Sydney, NSW, Australia; Department of Neurology (H.-P.H.), Palacky University, Olomouc, Czech Republic; Department of Neurology (H.-P.H.), Medical University of Vienna, Austria; Department of Neurology and Centre for Translational Neuro- and Behavioural Sciences (C-TNBS) (R.P., K.K., C.K.), University Hospital Essen, Germany; and Neurologic Clinic and Policlinic (L.K.), Departments of Medicine, Biomedicine and Clinical Research, University Hospital Basel, University of Basel, Switzerland
| | - Ludwig Kappos
- From the Department of Neurology (S.P.), University Hospital Giessen and Marburg, Justus-Liebig-University Giessen; Department of Neurology (L.R., J.I., M.K., S.R., T.R., S.S., A.G.W., O.A., H.-P.H., S.G.M.), University Hospital Duesseldorf, Germany; Brain and Mind Center (H.-P.H.), University of Sydney, NSW, Australia; Department of Neurology (H.-P.H.), Palacky University, Olomouc, Czech Republic; Department of Neurology (H.-P.H.), Medical University of Vienna, Austria; Department of Neurology and Centre for Translational Neuro- and Behavioural Sciences (C-TNBS) (R.P., K.K., C.K.), University Hospital Essen, Germany; and Neurologic Clinic and Policlinic (L.K.), Departments of Medicine, Biomedicine and Clinical Research, University Hospital Basel, University of Basel, Switzerland
| | - Sven G Meuth
- From the Department of Neurology (S.P.), University Hospital Giessen and Marburg, Justus-Liebig-University Giessen; Department of Neurology (L.R., J.I., M.K., S.R., T.R., S.S., A.G.W., O.A., H.-P.H., S.G.M.), University Hospital Duesseldorf, Germany; Brain and Mind Center (H.-P.H.), University of Sydney, NSW, Australia; Department of Neurology (H.-P.H.), Palacky University, Olomouc, Czech Republic; Department of Neurology (H.-P.H.), Medical University of Vienna, Austria; Department of Neurology and Centre for Translational Neuro- and Behavioural Sciences (C-TNBS) (R.P., K.K., C.K.), University Hospital Essen, Germany; and Neurologic Clinic and Policlinic (L.K.), Departments of Medicine, Biomedicine and Clinical Research, University Hospital Basel, University of Basel, Switzerland
| |
Collapse
|
31
|
Fong CC, Spencer J, Howlett-Prieto Q, Feng X, Reder AT. Adaptive and innate immune responses in multiple sclerosis with anti-CD20 therapy: Gene expression and protein profiles. Front Neurol 2023; 14:1158487. [PMID: 37168665 PMCID: PMC10166068 DOI: 10.3389/fneur.2023.1158487] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Accepted: 03/24/2023] [Indexed: 05/13/2023] Open
Abstract
Background Anti-CD20 is a highly effective therapy for multiple sclerosis (MS), a disease with multiple abnormalities in function of B and T cells and innate immune cells. Anti-CD20 therapy depletes B cells, which alters antibody production and has diverse effects on B cell immunity. These changes potentially affect immunity beyond B cells in MS. Objective Determine if anti-CD20 therapy effects non-B cell, as well as B cell, gene expression, and serum protein levels. Methods Samples were collected from 10 healthy controls and from clinically stable relapsing-remitting MS - 10 untreated, 9 interferon-β-treated, and 15 ocrelizumab-treated patients were studied before, and 2 weeks and 6 months after, the first anti-CD20 infusion. Peripheral blood mononuclear cells (PBMC) were analyzed with sensitive, 135,000-transcript RNA expression microarrays, using stringent criteria. Gene expression was compared to 43 MS-relevant serum immune and neurotrophic proteins, using multiplex protein assays. Results Anti-CD20 therapy reduced expression of 413 total genes and 185 B-cell-regulated genes at 2 weeks vs. pre-therapy. Expression of 19 (15%) of these B cell genes returned toward baseline by 6 months, including genes for the B cell activation protein, CD79A, and for immunoglobulin A, D, and G heavy chains. Expression pathways for Th17 and CD4 regulatory T-cell (Treg) development, differentiation, and proliferation also quieted. In contrast, expression increased in Th1 and myeloid cell antiviral, pro-inflammatory, and toll-like receptor (TLR) gene pathways. Conclusion These findings have clinical implications. B cell gene expression diminishes 2 weeks after anti-CD20 antibody infusion, but begins to rebound by 6 months. This suggests that the optimum time for vaccination is soon before reinfusion of anti-CD20 therapy. In addition, at 6 months, there is enhanced Th1 cell gene expression and induction of innate immune response genes and TLR expression, which can enhance anti-viral and anti-tumor immunity. This may compensate for diminished B cell gene expression after therapy. These data suggest that anti-CD20 therapy has dynamic effect on B cells and causes a compensatory rise in Th1 and myeloid immunity.
Collapse
Affiliation(s)
| | | | | | - Xuan Feng
- Department of Neurology, University of Chicago Medicine, Chicago, IL, United States
| | - Anthony T. Reder
- Department of Neurology, University of Chicago Medicine, Chicago, IL, United States
| |
Collapse
|
32
|
Mader S, Ho S, Wong HK, Baier S, Winklmeier S, Riemer C, Rübsamen H, Fernandez IM, Gerhards R, Du C, Chuquisana O, Lünemann JD, Lux A, Nimmerjahn F, Bradl M, Kawakami N, Meinl E. Dissection of complement and Fc-receptor-mediated pathomechanisms of autoantibodies to myelin oligodendrocyte glycoprotein. Proc Natl Acad Sci U S A 2023; 120:e2300648120. [PMID: 36943883 PMCID: PMC10068779 DOI: 10.1073/pnas.2300648120] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Accepted: 02/08/2023] [Indexed: 03/23/2023] Open
Abstract
Autoantibodies against myelin oligodendrocyte glycoprotein (MOG) have recently been established to define a new disease entity, MOG-antibody-associated disease (MOGAD), which is clinically overlapping with multiple sclerosis. MOG-specific antibodies (Abs) from patients are pathogenic, but the precise effector mechanisms are currently still unknown and no therapy is approved for MOGAD. Here, we determined the contributions of complement and Fc-receptor (FcR)-mediated effects in the pathogenicity of MOG-Abs. Starting from a recombinant anti-MOG (mAb) with human IgG1 Fc, we established MOG-specific mutant mAbs with differential FcR and C1q binding. We then applied selected mutants of this MOG-mAb in two animal models of experimental autoimmune encephalomyelitis. First, we found MOG-mAb-induced demyelination was mediated by both complement and FcRs about equally. Second, we found that MOG-Abs enhanced activation of cognate MOG-specific T cells in the central nervous system (CNS), which was dependent on FcR-, but not C1q-binding. The identification of complement-dependent and -independent pathomechanisms of MOG-Abs has implications for therapeutic strategies in MOGAD.
Collapse
Affiliation(s)
- Simone Mader
- Institute of Clinical Neuroimmunology, Biomedical Center and University Hospital, Ludwig-Maximilians-Universität München, 82152Planegg-Martinsried, Germany
| | - Samantha Ho
- Institute of Clinical Neuroimmunology, Biomedical Center and University Hospital, Ludwig-Maximilians-Universität München, 82152Planegg-Martinsried, Germany
- Graduate School of Systemic Neuroscience, Ludwig-Maximilians-Universität München, 82152Planegg-Martinsried, Germany
| | - Hoi Kiu Wong
- Institute of Clinical Neuroimmunology, Biomedical Center and University Hospital, Ludwig-Maximilians-Universität München, 82152Planegg-Martinsried, Germany
| | - Selia Baier
- Institute of Clinical Neuroimmunology, Biomedical Center and University Hospital, Ludwig-Maximilians-Universität München, 82152Planegg-Martinsried, Germany
| | - Stephan Winklmeier
- Institute of Clinical Neuroimmunology, Biomedical Center and University Hospital, Ludwig-Maximilians-Universität München, 82152Planegg-Martinsried, Germany
| | - Carolina Riemer
- Chair of Genetics, Department of Biology, Friedrich Alexander University of Erlangen-Nürnberg, 91058Erlangen, Germany
| | - Heike Rübsamen
- Institute of Clinical Neuroimmunology, Biomedical Center and University Hospital, Ludwig-Maximilians-Universität München, 82152Planegg-Martinsried, Germany
| | - Iris Marti Fernandez
- Institute of Clinical Neuroimmunology, Biomedical Center and University Hospital, Ludwig-Maximilians-Universität München, 82152Planegg-Martinsried, Germany
| | - Ramona Gerhards
- Institute of Clinical Neuroimmunology, Biomedical Center and University Hospital, Ludwig-Maximilians-Universität München, 82152Planegg-Martinsried, Germany
| | - Cuilian Du
- Institute of Clinical Neuroimmunology, Biomedical Center and University Hospital, Ludwig-Maximilians-Universität München, 82152Planegg-Martinsried, Germany
| | - Omar Chuquisana
- Department of Neurology with Institute of Translational Neurology, University Hospital Münster, 48149Münster, Germany
| | - Jan D. Lünemann
- Department of Neurology with Institute of Translational Neurology, University Hospital Münster, 48149Münster, Germany
| | - Anja Lux
- Chair of Genetics, Department of Biology, Friedrich Alexander University of Erlangen-Nürnberg, 91058Erlangen, Germany
| | - Falk Nimmerjahn
- Chair of Genetics, Department of Biology, Friedrich Alexander University of Erlangen-Nürnberg, 91058Erlangen, Germany
- Medical Immunology Campus Erlangen, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen91058, Germany
| | - Monika Bradl
- Department of Neuroimmunology, Center for Brain Research, Medical University of Vienna, 1090Vienna, Austria
| | - Naoto Kawakami
- Institute of Clinical Neuroimmunology, Biomedical Center and University Hospital, Ludwig-Maximilians-Universität München, 82152Planegg-Martinsried, Germany
| | - Edgar Meinl
- Institute of Clinical Neuroimmunology, Biomedical Center and University Hospital, Ludwig-Maximilians-Universität München, 82152Planegg-Martinsried, Germany
| |
Collapse
|
33
|
Ho S, Oswald E, Wong HK, Vural A, Yilmaz V, Tüzün E, Türkoğlu R, Straub T, Meinl I, Thaler F, Kümpfel T, Meinl E, Mader S. Ocrelizumab Treatment Modulates B-Cell Regulating Factors in Multiple Sclerosis. NEUROLOGY(R) NEUROIMMUNOLOGY & NEUROINFLAMMATION 2023; 10:e200083. [PMID: 36702538 PMCID: PMC9880874 DOI: 10.1212/nxi.0000000000200083] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Accepted: 11/22/2022] [Indexed: 01/28/2023]
Abstract
BACKGROUND AND OBJECTIVES Antibodies to CD20 efficiently reduce new relapses in multiple sclerosis (MS), and ocrelizumab has been shown to be effective also in primary progressive MS. Although anti-CD20 treatments efficiently deplete B cells in blood, some B cells and CD20- plasma cells persist in lymphatic organs and the inflamed CNS; their survival is regulated by the B cell-activating factor (BAFF)/A proliferation-inducing ligand (APRIL) system. The administration of a soluble receptor for BAFF and APRIL, atacicept, unexpectedly worsened MS. Here, we explored the long-term effects of ocrelizumab on immune cell subsets as well as on cytokines and endogenous soluble receptors comprising the BAFF-APRIL system. METHODS We analyzed immune cell subsets and B cell-regulating factors longitudinally for up to 2.5 years in patients with MS treated with ocrelizumab. In a second cohort, we determined B-cell regulatory factors in the CSF before and after ocrelizumab. We quantified the cytokines BAFF and APRIL along with their endogenous soluble receptors soluble B-cell maturation antigen (sBCMA) and soluble transmembrane activator and calcium-modulator and cyclophilin ligand (CAML) interactor (sTACI) using enzyme-linked immunosorbent assays (ELISAs). In addition, we established an in-house ELISA to measure sTACI-BAFF complexes. RESULTS Ocrelizumab treatment of people with MS persistently depleted B cells and CD20+ T cells. This treatment enhanced BAFF and reduced the free endogenous soluble receptor and decoy sTACI in both serum and CSF. Levels of sTACI negatively correlated with BAFF levels. Reduction of sTACI was associated with formation of sTACI-BAFF complexes. DISCUSSION We describe a novel effect of anti-CD20 therapy on the BAFF-APRIL system, namely reduction of sTACI. Because sTACI is a decoy for APRIL, its reduction may enhance local APRIL activity, thereby promoting regulatory IgA+ plasma cells and astrocytic interleukin (IL)-10 production. Thus, reducing sTACI might contribute to the beneficial effect of anti-CD20 as exogenous sTACI (atacicept) worsened MS. CLASSIFICATION OF EVIDENCE This study provides Class IV evidence that endogenous sTACI in blood and CSF is decreased after ocrelizumab treatment.
Collapse
Affiliation(s)
- Samantha Ho
- From the Institute of Clinical Neuroimmunology (S.H., E.O., H.K.W., A.V., I.M., F.T., T.K., E.M., S.M.), Biomedical Center and University Hospitals, Ludwig-Maximilians-Universität München; Graduate School of Systemic Neurosciences (S.H.), Ludwig-Maximilians-Universität München, Germany; Department of Neurology (A.V.), Koc University School of Medicine; Department of Neuroscience (V.Y., E.T.), Aziz Sancar Institute of Experimental Medicine, Istanbul University; Department of Neurology (R.T.), Haydarpasa Numune Education and Research Hospital, Istanbul, Türkiye; Core Facility Bioinformatics (T.S.), Biomedical Center, Ludwig-Maximilians-Universität München, Germany; Munich Cluster for Systems Neurology (SyNergy) (F.T.), Germany
| | - Eva Oswald
- From the Institute of Clinical Neuroimmunology (S.H., E.O., H.K.W., A.V., I.M., F.T., T.K., E.M., S.M.), Biomedical Center and University Hospitals, Ludwig-Maximilians-Universität München; Graduate School of Systemic Neurosciences (S.H.), Ludwig-Maximilians-Universität München, Germany; Department of Neurology (A.V.), Koc University School of Medicine; Department of Neuroscience (V.Y., E.T.), Aziz Sancar Institute of Experimental Medicine, Istanbul University; Department of Neurology (R.T.), Haydarpasa Numune Education and Research Hospital, Istanbul, Türkiye; Core Facility Bioinformatics (T.S.), Biomedical Center, Ludwig-Maximilians-Universität München, Germany; Munich Cluster for Systems Neurology (SyNergy) (F.T.), Germany
| | - Hoi Kiu Wong
- From the Institute of Clinical Neuroimmunology (S.H., E.O., H.K.W., A.V., I.M., F.T., T.K., E.M., S.M.), Biomedical Center and University Hospitals, Ludwig-Maximilians-Universität München; Graduate School of Systemic Neurosciences (S.H.), Ludwig-Maximilians-Universität München, Germany; Department of Neurology (A.V.), Koc University School of Medicine; Department of Neuroscience (V.Y., E.T.), Aziz Sancar Institute of Experimental Medicine, Istanbul University; Department of Neurology (R.T.), Haydarpasa Numune Education and Research Hospital, Istanbul, Türkiye; Core Facility Bioinformatics (T.S.), Biomedical Center, Ludwig-Maximilians-Universität München, Germany; Munich Cluster for Systems Neurology (SyNergy) (F.T.), Germany
| | - Atay Vural
- From the Institute of Clinical Neuroimmunology (S.H., E.O., H.K.W., A.V., I.M., F.T., T.K., E.M., S.M.), Biomedical Center and University Hospitals, Ludwig-Maximilians-Universität München; Graduate School of Systemic Neurosciences (S.H.), Ludwig-Maximilians-Universität München, Germany; Department of Neurology (A.V.), Koc University School of Medicine; Department of Neuroscience (V.Y., E.T.), Aziz Sancar Institute of Experimental Medicine, Istanbul University; Department of Neurology (R.T.), Haydarpasa Numune Education and Research Hospital, Istanbul, Türkiye; Core Facility Bioinformatics (T.S.), Biomedical Center, Ludwig-Maximilians-Universität München, Germany; Munich Cluster for Systems Neurology (SyNergy) (F.T.), Germany
| | - Vuslat Yilmaz
- From the Institute of Clinical Neuroimmunology (S.H., E.O., H.K.W., A.V., I.M., F.T., T.K., E.M., S.M.), Biomedical Center and University Hospitals, Ludwig-Maximilians-Universität München; Graduate School of Systemic Neurosciences (S.H.), Ludwig-Maximilians-Universität München, Germany; Department of Neurology (A.V.), Koc University School of Medicine; Department of Neuroscience (V.Y., E.T.), Aziz Sancar Institute of Experimental Medicine, Istanbul University; Department of Neurology (R.T.), Haydarpasa Numune Education and Research Hospital, Istanbul, Türkiye; Core Facility Bioinformatics (T.S.), Biomedical Center, Ludwig-Maximilians-Universität München, Germany; Munich Cluster for Systems Neurology (SyNergy) (F.T.), Germany
| | - Erdem Tüzün
- From the Institute of Clinical Neuroimmunology (S.H., E.O., H.K.W., A.V., I.M., F.T., T.K., E.M., S.M.), Biomedical Center and University Hospitals, Ludwig-Maximilians-Universität München; Graduate School of Systemic Neurosciences (S.H.), Ludwig-Maximilians-Universität München, Germany; Department of Neurology (A.V.), Koc University School of Medicine; Department of Neuroscience (V.Y., E.T.), Aziz Sancar Institute of Experimental Medicine, Istanbul University; Department of Neurology (R.T.), Haydarpasa Numune Education and Research Hospital, Istanbul, Türkiye; Core Facility Bioinformatics (T.S.), Biomedical Center, Ludwig-Maximilians-Universität München, Germany; Munich Cluster for Systems Neurology (SyNergy) (F.T.), Germany
| | - Recai Türkoğlu
- From the Institute of Clinical Neuroimmunology (S.H., E.O., H.K.W., A.V., I.M., F.T., T.K., E.M., S.M.), Biomedical Center and University Hospitals, Ludwig-Maximilians-Universität München; Graduate School of Systemic Neurosciences (S.H.), Ludwig-Maximilians-Universität München, Germany; Department of Neurology (A.V.), Koc University School of Medicine; Department of Neuroscience (V.Y., E.T.), Aziz Sancar Institute of Experimental Medicine, Istanbul University; Department of Neurology (R.T.), Haydarpasa Numune Education and Research Hospital, Istanbul, Türkiye; Core Facility Bioinformatics (T.S.), Biomedical Center, Ludwig-Maximilians-Universität München, Germany; Munich Cluster for Systems Neurology (SyNergy) (F.T.), Germany
| | - Tobias Straub
- From the Institute of Clinical Neuroimmunology (S.H., E.O., H.K.W., A.V., I.M., F.T., T.K., E.M., S.M.), Biomedical Center and University Hospitals, Ludwig-Maximilians-Universität München; Graduate School of Systemic Neurosciences (S.H.), Ludwig-Maximilians-Universität München, Germany; Department of Neurology (A.V.), Koc University School of Medicine; Department of Neuroscience (V.Y., E.T.), Aziz Sancar Institute of Experimental Medicine, Istanbul University; Department of Neurology (R.T.), Haydarpasa Numune Education and Research Hospital, Istanbul, Türkiye; Core Facility Bioinformatics (T.S.), Biomedical Center, Ludwig-Maximilians-Universität München, Germany; Munich Cluster for Systems Neurology (SyNergy) (F.T.), Germany
| | - Ingrid Meinl
- From the Institute of Clinical Neuroimmunology (S.H., E.O., H.K.W., A.V., I.M., F.T., T.K., E.M., S.M.), Biomedical Center and University Hospitals, Ludwig-Maximilians-Universität München; Graduate School of Systemic Neurosciences (S.H.), Ludwig-Maximilians-Universität München, Germany; Department of Neurology (A.V.), Koc University School of Medicine; Department of Neuroscience (V.Y., E.T.), Aziz Sancar Institute of Experimental Medicine, Istanbul University; Department of Neurology (R.T.), Haydarpasa Numune Education and Research Hospital, Istanbul, Türkiye; Core Facility Bioinformatics (T.S.), Biomedical Center, Ludwig-Maximilians-Universität München, Germany; Munich Cluster for Systems Neurology (SyNergy) (F.T.), Germany
| | - Franziska Thaler
- From the Institute of Clinical Neuroimmunology (S.H., E.O., H.K.W., A.V., I.M., F.T., T.K., E.M., S.M.), Biomedical Center and University Hospitals, Ludwig-Maximilians-Universität München; Graduate School of Systemic Neurosciences (S.H.), Ludwig-Maximilians-Universität München, Germany; Department of Neurology (A.V.), Koc University School of Medicine; Department of Neuroscience (V.Y., E.T.), Aziz Sancar Institute of Experimental Medicine, Istanbul University; Department of Neurology (R.T.), Haydarpasa Numune Education and Research Hospital, Istanbul, Türkiye; Core Facility Bioinformatics (T.S.), Biomedical Center, Ludwig-Maximilians-Universität München, Germany; Munich Cluster for Systems Neurology (SyNergy) (F.T.), Germany
| | - Tania Kümpfel
- From the Institute of Clinical Neuroimmunology (S.H., E.O., H.K.W., A.V., I.M., F.T., T.K., E.M., S.M.), Biomedical Center and University Hospitals, Ludwig-Maximilians-Universität München; Graduate School of Systemic Neurosciences (S.H.), Ludwig-Maximilians-Universität München, Germany; Department of Neurology (A.V.), Koc University School of Medicine; Department of Neuroscience (V.Y., E.T.), Aziz Sancar Institute of Experimental Medicine, Istanbul University; Department of Neurology (R.T.), Haydarpasa Numune Education and Research Hospital, Istanbul, Türkiye; Core Facility Bioinformatics (T.S.), Biomedical Center, Ludwig-Maximilians-Universität München, Germany; Munich Cluster for Systems Neurology (SyNergy) (F.T.), Germany
| | - Edgar Meinl
- From the Institute of Clinical Neuroimmunology (S.H., E.O., H.K.W., A.V., I.M., F.T., T.K., E.M., S.M.), Biomedical Center and University Hospitals, Ludwig-Maximilians-Universität München; Graduate School of Systemic Neurosciences (S.H.), Ludwig-Maximilians-Universität München, Germany; Department of Neurology (A.V.), Koc University School of Medicine; Department of Neuroscience (V.Y., E.T.), Aziz Sancar Institute of Experimental Medicine, Istanbul University; Department of Neurology (R.T.), Haydarpasa Numune Education and Research Hospital, Istanbul, Türkiye; Core Facility Bioinformatics (T.S.), Biomedical Center, Ludwig-Maximilians-Universität München, Germany; Munich Cluster for Systems Neurology (SyNergy) (F.T.), Germany.
| | - Simone Mader
- From the Institute of Clinical Neuroimmunology (S.H., E.O., H.K.W., A.V., I.M., F.T., T.K., E.M., S.M.), Biomedical Center and University Hospitals, Ludwig-Maximilians-Universität München; Graduate School of Systemic Neurosciences (S.H.), Ludwig-Maximilians-Universität München, Germany; Department of Neurology (A.V.), Koc University School of Medicine; Department of Neuroscience (V.Y., E.T.), Aziz Sancar Institute of Experimental Medicine, Istanbul University; Department of Neurology (R.T.), Haydarpasa Numune Education and Research Hospital, Istanbul, Türkiye; Core Facility Bioinformatics (T.S.), Biomedical Center, Ludwig-Maximilians-Universität München, Germany; Munich Cluster for Systems Neurology (SyNergy) (F.T.), Germany.
| |
Collapse
|
34
|
Garcia A, Dugast E, Shah S, Morille J, Lebrun-Frenay C, Thouvenot E, De Sèze J, Le Page E, Vukusic S, Maurousset A, Berger E, Casez O, Labauge P, Ruet A, Raposo C, Bakdache F, Buffels R, Le Frère F, Nicot A, Wiertlewski S, Gourraud PA, Berthelot L, Laplaud D. Immune Profiling Reveals the T-Cell Effect of Ocrelizumab in Early Relapsing-Remitting Multiple Sclerosis. NEUROLOGY(R) NEUROIMMUNOLOGY & NEUROINFLAMMATION 2023; 10:10/3/e200091. [PMID: 36810163 PMCID: PMC9944617 DOI: 10.1212/nxi.0000000000200091] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Accepted: 12/12/2022] [Indexed: 02/23/2023]
Abstract
BACKGROUND AND OBJECTIVES Ocrelizumab (OCR), a humanized anti-CD20 monoclonal antibody, is highly efficient in patients with relapsing-remitting multiple sclerosis (RR-MS). We assessed early cellular immune profiles and their association with disease activity at treatment start and under therapy, which may provide new clues on the mechanisms of action of OCR and on the disease pathophysiology. METHODS A first group of 42 patients with an early RR-MS, never exposed to disease-modifying therapy, was included in 11 centers participating to an ancillary study of the ENSEMBLE trial (NCT03085810) to evaluate the effectiveness and safety of OCR. The phenotypic immune profile was comprehensively assessed by multiparametric spectral flow cytometry at baseline and after 24 and 48 weeks of OCR treatment on cryopreserved peripheral blood mononuclear cells and analyzed in relation to disease clinical activity. A second group of 13 untreated patients with RR-MS was included for comparative analysis of peripheral blood and CSF. The transcriptomic profile was assessed by single-cell qPCRs of 96 genes of immunologic interest. RESULTS Using an unbiased analysis, we found that OCR as an effect on 4 clusters of CD4+ T cells: one corresponding to naive CD4+ T cells was increased, the other clusters corresponded to effector memory (EM) CD4+CCR6- T cells expressing homing and migration markers, 2 of them also expressing CCR5 and were decreased by the treatment. Of interest, one CD8+ T-cell cluster was decreased by OCR corresponding to EM CCR5-expressing T cells with high expression of the brain homing markers CD49d and CD11a and correlated with the time elapsed since the last relapse. These EM CD8+CCR5+ T cells were enriched in the CSF of patients with RR-MS and corresponded to activated and cytotoxic cells. DISCUSSION Our study provides novel insights into the mode of action of anti-CD20, pointing toward the role of EM T cells, particularly a subset of CD8 T cells expressing CCR5.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - David Laplaud
- From the CHU Nantes (A.G., E.D., S.S., J.M., A.N., S.W., P.-A.G., L.B., D.L.), Nantes Université, INSERM UMR1064, Center for Research in Transplantation and Translational Immunology (CR2TI); CRCSEP (C.L.-F.), CHU de Nice Pasteur 2, Université Nice Côte d'Azur UR2CA URRIS; Service de Neurologie (E.T.), CHU de Nîmes, Institut de Génomique Fonctionnelle, Université de Montpellier, CNRS, INSERM; Service de Neurologie et Centre d'Investigation Clinique (J.D.S.), CHU de Strasbourg; Service de Neurologie (E.L.P.), CHU Pontchaillou, Rennes; Université de Lyon (S.V.), Université Claude Bernard Lyon 1; Service de Neurologie (S.V.), sclérose en plaques, pathologies de la Myéline et Neuro-inflammation, Hôpital Neurologique Pierre Wertheimer, Hospices Civils de Lyon, Bron; Observatoire Français de la Sclérose en Plaques (S.V.), Centre de Recherche en Neurosciences de Lyon; EUGENE DEVIC EDMUS Foundation Against Multiple sclerosis (S.V.), state-approved Foundation, Bron; Service de Neurologie (A.M.), CHU Bretonneau, Tours; Service de Neurologie (E.B.), CHU de Besançon; Service de Neurologie (O.C.), CHU de Grenoble; Service de Neurologie (P.L.), CHU de Montpellier, Montpellier; Service de Neurologie (A.R.), CHU de Bordeaux; Université de Bordeaux (A.R.), INSERM, Neurocentre Magendie; F. Hoffmann-La Roche Ltd (C.R., F.B., R.B.) CIC INSERM 1413 (F.L.F., S.W., D.L.), Nantes; CHU Nantes (S.W., D.L.), Nantes Université, Service de Neurologie; and CHU Nantes (P.-A.G.), Nantes Université, Clinique des données, France.
| |
Collapse
|
35
|
Dybowski S, Torke S, Weber MS. Targeting B Cells and Microglia in Multiple Sclerosis With Bruton Tyrosine Kinase Inhibitors: A Review. JAMA Neurol 2023; 80:404-414. [PMID: 36780171 DOI: 10.1001/jamaneurol.2022.5332] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/14/2023]
Abstract
Importance Currently, disease-modifying therapies for multiple sclerosis (MS) use 4 mechanisms of action: immune modulation, suppressing immune cell proliferation, inhibiting immune cell migration, or cellular depletion. Over the last decades, the repertoire substantially increased because of the conceptual progress that not only T cells but also B cells play an important pathogenic role in MS, fostered by the empirical success of B cell-depleting antibodies against the surface molecule CD20. Notwithstanding this advance, a continuous absence of B cells may harbor safety risks, such as a decline in the endogenous production of immunoglobulins. Accordingly, novel B cell-directed MS therapies are in development, such as inhibitors targeting Bruton tyrosine kinase (BTK). Observations BTK is centrally involved in the B cell receptor-mediated activation of B cells, one key requirement in the development of autoreactive B cells, but also in the activation of myeloid cells, such as macrophages and microglia. Various compounds in development differ in their binding mode, selectivity and specificity, relative inhibitory concentration, and potential to enter the central nervous system. The latter may be important in assessing whether BTK inhibition is a promising strategy to control inflammatory circuits within the brain, the key process that is assumed to drive MS progression. Accordingly, clinical trials using BTK inhibitors are currently conducted in patients with relapsing-remitting MS as well as progressive MS, so far generating encouraging data regarding efficacy and safety. Conclusions and Relevance While the novel approach of targeting BTK is highly promising, several questions remain unanswered, such as the long-term effects of using BTK inhibitors in the treatment of inflammatory CNS disease. Potential changes in circulating antibody levels should be evaluated and compared with B cell depletion. Also important is the potential of BTK inhibitors to enter the CNS, which depends on the given compound. Remaining questions involve where BTK inhibitors fit in the landscape of MS therapeutics. A comparative analysis of their distinct properties is necessary to identify which inhibitors may be used in relapsing vs progressive forms of MS as well as to clarify which agent may be most suitable for sequential use after anti-CD20 treatment.
Collapse
Affiliation(s)
- Sarah Dybowski
- Institute of Neuropathology, University Medical Center, Göttingen, Germany
| | - Sebastian Torke
- Institute of Neuropathology, University Medical Center, Göttingen, Germany.,Experimental and Clinical Research Center, Charité-Universitätsmedizin Berlin, Berlin, Germany.,Max-Delbrück-Center for Molecular Medicine, Berlin, Germany
| | - Martin S Weber
- Institute of Neuropathology, University Medical Center, Göttingen, Germany.,Department of Neurology, University Medical Center, Göttingen, Germany.,Fraunhofer Institute for Translational Medicine and Pharmacology, Göttingen, Germany
| |
Collapse
|
36
|
Learning multiple sclerosis immunopathogenesis from anti-CD20 therapy. Proc Natl Acad Sci U S A 2023; 120:e2221544120. [PMID: 36719925 PMCID: PMC9962916 DOI: 10.1073/pnas.2221544120] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
|
37
|
Mathias A, Pantazou V, Perriot S, Canales M, Jones S, Oberholster L, Moulin M, Fenwick C, Bernard-Valnet R, Théaudin M, Pot C, Du Pasquier RA. Ocrelizumab Impairs the Phenotype and Function of Memory CD8 + T Cells: A 1-Year Longitudinal Study in Patients With Multiple Sclerosis. NEUROLOGY(R) NEUROIMMUNOLOGY & NEUROINFLAMMATION 2023; 10:10/2/e200084. [PMID: 36717268 PMCID: PMC9887539 DOI: 10.1212/nxi.0000000000200084] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Accepted: 11/22/2022] [Indexed: 02/01/2023]
Abstract
BACKGROUND AND OBJECTIVE Depleting CD20+ B cells is the primary mechanism by which ocrelizumab (OCRE) is efficient in persons with multiple sclerosis (pwMS). However, the exact role of OCRE on other immune cell subsets directly or indirectly remains elusive. The purpose of this study is to characterize the dynamics of peripheral immune cells of pwMS on OCRE. METHODS We collected blood samples from 38 pwMS before OCRE onset (T0) and at 6 and 12 months (T6, T12) after initiation. To cover the immune cell diversity, using mass cytometry time of flight, we designed a 38-parameter panel to analyze B, T, and innate immune cell markers and CNS migratory markers. In parallel, viral-specific CD8+ T-cell responses were assessed by the quantification of interferon-γ secretion using the enzyme-linked immunospot assay on cytomegalovirus, Epstein-Barr virus, and influenza stimulations. RESULTS Beside B-cell depletion, we observed a loss in memory CD8+CD20+ and central memory CD8+ T cells but not in CD4+CD20+ T cells already at T6 and T12 (p < 0.001). The loss of memory CD8+ T cells correlated with a lower CXCR3 expression (p < 0.001) and CNS-related LFA-1 integrin expression (p < 0.001) as well as a reduced antiviral cellular immune response observed at both time points (p < 0.001). Of note, we did not observe major changes in the phenotype of the other cell types studied. Seven of 38 (18.4%) patients in our cohort presented with infections while on OCRE; 4 of which were switched from dimethyl fumarate. Finally, using a mixed linear model on mass cytometry data, we demonstrated that the immunomodulation induced by previous disease-modifying therapies (DMTs) was prolonged over the period of the study. DISCUSSION In addition to its well-known role on B cells, our data suggest that OCRE also acts on CD8+ T cells by depleting the memory compartment. These changes in CD8+ T cells may be an asset in the action of OCRE on MS course but might also contribute to explain the increased occurrence of infections in these patients. Finally, although more data are needed to confirm this observation, it suggests that clinicians should pay a special attention to an increased infection risk in pwMS switched from other DMTs to OCRE.
Collapse
Affiliation(s)
- Amandine Mathias
- From the Laboratories of Neuroimmunology (A.M., V.P., S.P., M.C., S.J., L.O., C.P., R.A.D), Neuroscience Research Center, Department of Clinical Neurosciences, Lausanne University Hospital and University of Lausanne, Switzerland; Service of Neurology (V.P., R.B.-V., M.T., C.P., R.A.D.), Department of Clinical Neurosciences, Lausanne University Hospital and University of Lausanne, Switzerland; Paris Brain Institute (V.P.), Lubetzki-Stankoff group of Myelination, France; Service of Immunology and Allergy (M.M., C.F.), Department of Medicine, Lausanne University Hospital and University of Lausanne, Switzerland.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Shinoda K, Li R, Rezk A, Mexhitaj I, Patterson KR, Kakara M, Zuroff L, Bennett JL, von Büdingen HC, Carruthers R, Edwards KR, Fallis R, Giacomini PS, Greenberg BM, Hafler DA, Ionete C, Kaunzner UW, Lock CB, Longbrake EE, Pardo G, Piehl F, Weber MS, Ziemssen T, Jacobs D, Gelfand JM, Cross AH, Cameron B, Musch B, Winger RC, Jia X, Harp CT, Herman A, Bar-Or A. Differential effects of anti-CD20 therapy on CD4 and CD8 T cells and implication of CD20-expressing CD8 T cells in MS disease activity. Proc Natl Acad Sci U S A 2023; 120:e2207291120. [PMID: 36634138 PMCID: PMC9934304 DOI: 10.1073/pnas.2207291120] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
A small proportion of multiple sclerosis (MS) patients develop new disease activity soon after starting anti-CD20 therapy. This activity does not recur with further dosing, possibly reflecting deeper depletion of CD20-expressing cells with repeat infusions. We assessed cellular immune profiles and their association with transient disease activity following anti-CD20 initiation as a window into relapsing disease biology. Peripheral blood mononuclear cells from independent discovery and validation cohorts of MS patients initiating ocrelizumab were assessed for phenotypic and functional profiles using multiparametric flow cytometry. Pretreatment CD20-expressing T cells, especially CD20dimCD8+ T cells with a highly inflammatory and central nervous system (CNS)-homing phenotype, were significantly inversely correlated with pretreatment MRI gadolinium-lesion counts, and also predictive of early disease activity observed after anti-CD20 initiation. Direct removal of pretreatment proinflammatory CD20dimCD8+ T cells had a greater contribution to treatment-associated changes in the CD8+ T cell pool than was the case for CD4+ T cells. Early disease activity following anti-CD20 initiation was not associated with reconstituting CD20dimCD8+ T cells, which were less proinflammatory compared with pretreatment. Similarly, this disease activity did not correlate with early reconstituting B cells, which were predominantly transitional CD19+CD24highCD38high with a more anti-inflammatory profile. We provide insights into the mode-of-action of anti-CD20 and highlight a potential role for CD20dimCD8+ T cells in MS relapse biology; their strong inverse correlation with both pretreatment and early posttreatment disease activity suggests that CD20-expressing CD8+ T cells leaving the circulation (possibly to the CNS) play a particularly early role in the immune cascades involved in relapse development.
Collapse
Affiliation(s)
- Koji Shinoda
- aDepartment of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA19104
- bCenter for Neuroinflammation and Experimental Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA19104
| | - Rui Li
- aDepartment of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA19104
- bCenter for Neuroinflammation and Experimental Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA19104
| | - Ayman Rezk
- aDepartment of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA19104
- bCenter for Neuroinflammation and Experimental Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA19104
| | - Ina Mexhitaj
- aDepartment of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA19104
- bCenter for Neuroinflammation and Experimental Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA19104
| | - Kristina R. Patterson
- aDepartment of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA19104
- bCenter for Neuroinflammation and Experimental Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA19104
| | - Mihir Kakara
- aDepartment of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA19104
- bCenter for Neuroinflammation and Experimental Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA19104
| | - Leah Zuroff
- aDepartment of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA19104
- bCenter for Neuroinflammation and Experimental Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA19104
| | - Jeffrey L. Bennett
- cDepartments of Neurology and Ophthalmology, Programs in Neuroscience and Immunology, University of Colorado School of Medicine, Aurora, CO80045
| | | | - Robert Carruthers
- eDepartment of Medicine, University of British Columbia, Vancouver, BCV6T 2B5, Canada
| | - Keith R. Edwards
- fMultiple Sclerosis Center of Northeastern New York, Comprehensive MS Care Center Affiliated with the National MS Society, Latham, NY12110
| | - Robert Fallis
- gDepartment of Neurology, Ohio State University Medical Center, Columbus, OH43210
| | - Paul S. Giacomini
- hDepartment of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, QCH3A 2B4, Canada
| | - Benjamin M. Greenberg
- iDepartment of Neurology, University of Texas Southwestern Medical Center, Dallas, TX75390
| | - David A. Hafler
- jDepartments of Neurology and Immunobiology, Yale School of Medicine, New Haven, CT06510
| | - Carolina Ionete
- kDepartment of Neurology, University of Massachusetts Medical School, Worcester, MA01655
| | - Ulrike W. Kaunzner
- lJudith Jaffe Multiple Sclerosis Center, Weill Cornell Medicine, New York, NY10021
| | - Christopher B. Lock
- mDepartment of Neurology and Neurological Sciences, Stanford University, Palo Alto, CA94304
| | | | - Gabriel Pardo
- oOklahoma Medical Research Foundation, Multiple Sclerosis Center of Excellence, Oklahoma City, OK73104
| | - Fredrik Piehl
- pDepartment of Clinical Neuroscience, Karolinska Institute, SE-171 76Stockholm, Sweden
- qDepartment of Neurology, Karolinska University Hospital, SE-171 77Stockholm, Sweden
- rNeuroimmunology Unit, Center for Molecular Medicine, Karolinska University Hospital, Karolinska Institute, SE-171 77Stockholm, Sweden
| | - Martin S. Weber
- sInstitute of Neuropathology, University Medical Center, 37075Göttingen, Germany
- tDepartment of Neurology, University Medical Center, 37075Göttingen, Germany
- uFraunhofer-Institute for Translational Medicine and Pharmackology ITMP, 37075Göttingen, Germany
| | - Tjalf Ziemssen
- vDepartment of Neurology, Center of Clinical Neuroscience, University Hospital Carl Gustav Carus, Technical University of Dresden, 01307Dresden, Germany
| | - Dina Jacobs
- aDepartment of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA19104
- bCenter for Neuroinflammation and Experimental Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA19104
| | - Jeffrey M. Gelfand
- wWeill Institute for Neurosciences, University of California, San Francisco, CA94158
- xDepartment of Neurology, University of California, San Francisco, CA94158
| | - Anne H. Cross
- yDepartment of Neurology, Washington University School of Medicine, Saint Louis, MO63110
| | | | - Bruno Musch
- zGenentech, Inc., South San Francisco, CA94080
| | | | | | | | - Ann Herman
- zGenentech, Inc., South San Francisco, CA94080
| | - Amit Bar-Or
- aDepartment of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA19104
- bCenter for Neuroinflammation and Experimental Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA19104
- aaChildren's Hospital of Philadelphia, University of Pennsylvania, Philadelphia, PA19104
- 1To whom correspondence may be addressed.
| |
Collapse
|
39
|
de Sèze J, Maillart E, Gueguen A, Laplaud DA, Michel L, Thouvenot E, Zephir H, Zimmer L, Biotti D, Liblau R. Anti-CD20 therapies in multiple sclerosis: From pathology to the clinic. Front Immunol 2023; 14:1004795. [PMID: 37033984 PMCID: PMC10076836 DOI: 10.3389/fimmu.2023.1004795] [Citation(s) in RCA: 49] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Accepted: 02/13/2023] [Indexed: 04/11/2023] Open
Abstract
The immune system plays a significant role in multiple sclerosis. While MS was historically thought to be T cell-mediated, multiple pieces of evidence now support the view that B cells are essential players in multiple sclerosis pathogenic processes. High-efficacy disease-modifying therapies that target the immune system have emerged over the past two decades. Anti-CD20 monoclonal antibodies selectively deplete CD20+ B and CD20+ T cells and efficiently suppress inflammatory disease activity. These monotherapies prevent relapses, reduce new or active magnetic resonance imaging brain lesions, and lessen disability progression in patients with relapsing multiple sclerosis. Rituximab, ocrelizumab, and ofatumumab are currently used in clinical practice, while phase III clinical trials for ublituximab have been recently completed. In this review, we compare the four anti-CD20 antibodies in terms of their mechanisms of action, routes of administration, immunological targets, and pharmacokinetic properties. A deeper understanding of the individual properties of these molecules in relation to their efficacy and safety profiles is critical for their use in clinical practice.
Collapse
Affiliation(s)
- Jérôme de Sèze
- Department of Neurology, Hôpital de Hautepierre, Clinical Investigation Center, Institut National de la Santé et de la Recherche Médicale (INSERM), Strasbourg, France
- Fédération de Médecine Translationelle, Institut National de la Santé et de la Recherche Médicale (INSERM), Strasbourg, France
- *Correspondence: Jérôme de Sèze,
| | - Elisabeth Maillart
- Department of Neurology, Pitié Salpêtrière Hospital, Paris, France
- Centre de Ressources et de Compétences Sclérose en Plaques, Paris, France
| | - Antoine Gueguen
- Department of Neurology, Rothschild Ophthalmologic Foundation, Paris, France
| | - David A. Laplaud
- Department of Neurology, Centre Hospitalier Universitaire (CHU) Nantes, Nantes Université, Institut National de la Santé et de la Recherche Médicale (INSERM), Centre d’Investigation Clinique (CIC), Center for Research in Transplantation and Translational Immunology, UMR, UMR1064, Nantes, France
| | - Laure Michel
- Clinical Neuroscience Centre, CIC_P1414 Institut National de la Santé et de la Recherche Médicale (INSERM), Rennes University Hospital, Rennes University, Rennes, France
- Microenvironment, Cell Differentiation, Immunology and Cancer Unit, Institut National de la Santé et de la Recherche Médicale (INSERM), Rennes I University, French Blood Agency, Rennes, France
- Neurology Department, Rennes University Hospital, Rennes, France
| | - Eric Thouvenot
- Department of Neurology, Centre Hospitalier Universitaire (CHU) Nîmes, University of Montpellier, Nîmes, France
- Institut de Génomique Fonctionnelle, UMR, Institut National de la Santé et de la Recherche Médicale (INSERM), University of Montpellier, Montpellier, France
| | - Hélène Zephir
- University of Lille, Institut National de la Santé et de la Recherche Médicale (INSERM) U1172, Centre Hospitalier Universitaire (CHU), Lille, France
| | - Luc Zimmer
- Université Claude Bernard Lyon 1, Hospices Civils de Lyon, Institut National de la Santé et de la Recherche Médicale (INSERM), CNRS, Lyon Neuroscience Research Center, Lyon, France
| | - Damien Biotti
- Centre Ressources et Compétences Sclérose En Plaques (CRC-SEP) and Department of Neurology, Centre Hospitalier Universitaire (CHU) Toulouse Purpan – Hôpital Pierre-Paul Riquet, Toulouse, France
| | - Roland Liblau
- Toulouse Institute for Infectious and Inflammatory Diseases (Infinity), University of Toulouse, CNRS, Institut National de la Santé et de la Recherche Médicale (INSERM), UPS, Toulouse, France
- Department of Immunology, Toulouse University Hospital, Toulouse, France
| |
Collapse
|
40
|
Rui Y, Eppler HB, Yanes AA, Jewell CM. Tissue-Targeted Drug Delivery Strategies to Promote Antigen-Specific Immune Tolerance. Adv Healthc Mater 2023; 12:e2202238. [PMID: 36417578 PMCID: PMC9992113 DOI: 10.1002/adhm.202202238] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 11/15/2022] [Indexed: 11/27/2022]
Abstract
During autoimmunity or organ transplant rejection, the immune system attacks host or transplanted tissue, causing debilitating inflammation for millions of patients. There is no cure for most of these diseases. Further, available therapies modulate inflammation through nonspecific pathways, reducing symptoms but also compromising patients' ability to mount healthy immune responses. Recent preclinical advances to regulate immune dysfunction with vaccine-like antigen specificity reveal exciting opportunities to address the root cause of autoimmune diseases and transplant rejection. Several of these therapies are currently undergoing clinical trials, underscoring the promise of antigen-specific tolerance. Achieving antigen-specific tolerance requires precision and often combinatorial delivery of antigen, cytokines, small molecule drugs, and other immunomodulators. This can be facilitated by biomaterial technologies, which can be engineered to orient and display immunological cues, protect against degradation, and selectively deliver signals to specific tissues or cell populations. In this review, some key immune cell populations involved in autoimmunity and healthy immune tolerance are described. Opportunities for drug delivery to immunological organs are discussed, where specialized tissue-resident immune cells can be programmed to respond in unique ways toward antigens. Finally, cell- and biomaterial-based therapies to induce antigen-specific immune tolerance that are currently undergoing clinical trials are highlighted.
Collapse
Affiliation(s)
- Yuan Rui
- Fischell Department of BioengineeringUniversity of MarylandCollege ParkMD20742USA
| | - Haleigh B. Eppler
- Fischell Department of BioengineeringUniversity of MarylandCollege ParkMD20742USA
- Biological Sciences Training ProgramUniversity of MarylandCollege ParkMD20742USA
| | - Alexis A. Yanes
- Fischell Department of BioengineeringUniversity of MarylandCollege ParkMD20742USA
| | - Christopher M. Jewell
- Fischell Department of BioengineeringUniversity of MarylandCollege ParkMD20742USA
- Biological Sciences Training ProgramUniversity of MarylandCollege ParkMD20742USA
- US Department of Veterans AffairsVA Maryland Health Care SystemBaltimoreMD21201USA
- Robert E. Fischell Institute for Biomedical DevicesCollege ParkMD20742USA
- Department of Microbiology and ImmunologyUniversity of Maryland Medical SchoolBaltimoreMD21201USA
- Marlene and Stewart Greenebaum Cancer CenterBaltimoreMD21201USA
| |
Collapse
|
41
|
Vakrakou AG, Paschalidis N, Pavlos E, Giannouli C, Karathanasis D, Tsipota X, Velonakis G, Stadelmann-Nessler C, Evangelopoulos ME, Stefanis L, Kilidireas C. Specific myeloid signatures in peripheral blood differentiate active and rare clinical phenotypes of multiple sclerosis. Front Immunol 2023; 14:1071623. [PMID: 36761741 PMCID: PMC9905713 DOI: 10.3389/fimmu.2023.1071623] [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: 10/16/2022] [Accepted: 01/03/2023] [Indexed: 01/26/2023] Open
Abstract
Current understanding of Multiple Sclerosis (MS) pathophysiology implicates perturbations in adaptive cellular immune responses, predominantly T cells, in Relapsing-Remitting forms (RRMS). Nevertheless, from a clinical perspective MS is a heterogeneous disease reflecting the heterogeneity of involved biological systems. This complexity requires advanced analysis tools at the single-cell level to discover biomarkers for better patient-group stratification. We designed a novel 44-parameter mass cytometry panel to interrogate predominantly the role of effector and regulatory subpopulations of peripheral blood myeloid subsets along with B and T-cells (excluding granulocytes) in MS, assessing three different patient cohorts: RRMS, PPMS (Primary Progressive) and Tumefactive MS patients (TMS) (n=10, 8, 14 respectively). We further subgrouped our cohort into inactive or active disease stages to capture the early underlying events in disease pathophysiology. Peripheral blood analysis showed that TMS cases belonged to the spectrum of RRMS, whereas PPMS cases displayed different features. In particular, TMS patients during a relapse stage were characterized by a specific subset of CD11c+CD14+ CD33+, CD192+, CD172+-myeloid cells with an alternative phenotype of monocyte-derived macrophages (high arginase-1, CD38, HLA-DR-low and endogenous TNF-a production). Moreover, TMS patients in relapse displayed a selective CD4 T-cell lymphopenia of cells with a Th2-like polarised phenotype. PPMS patients did not display substantial differences from healthy controls, apart from a trend toward higher expansion of NK cell subsets. Importantly, we found that myeloid cell populations are reshaped under effective disease-modifying therapy predominantly with glatiramer acetate and to a lesser extent with anti-CD20, suggesting that the identified cell signature represents a specific therapeutic target in TMS. The expanded myeloid signature in TMS patients was also confirmed by flow cytometry. Serum neurofilament light-chain levels confirmed the correlation of this myeloid cell signature with indices of axonal injury. More in-depth analysis of myeloid subsets revealed an increase of a subset of highly cytolytic and terminally differentiated NK cells in PPMS patients with leptomeningeal enhancement (active-PPMS), compared to those without (inactive-PPMS). We have identified previously uncharacterized subsets of circulating myeloid cells and shown them to correlate with distinct disease forms of MS as well as with specific disease states (relapse/remission).
Collapse
Affiliation(s)
- Aigli G Vakrakou
- Demyelinating Diseases Unit, 1st Department of Neurology, School of Medicine, Aeginition Hospital, National and Kapodistrian University of Athens, Athens, Greece.,Department of Neuropathology, University of Göttingen Medical Center, Göttingen, Germany
| | - Nikolaos Paschalidis
- Mass Cytometry-CyTOF Laboratory, Center for Clinical Research, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| | - Eleftherios Pavlos
- Center for Clinical Research, Experimental Surgery and Translational Research Biomedical Research Foundation of the Academy of Athens, Athens, Greece.,Division of Basic Sciences, University of Crete Medical School, Heraklion, Greece
| | - Christina Giannouli
- Center for Clinical Research, Experimental Surgery and Translational Research Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| | - Dimitris Karathanasis
- Demyelinating Diseases Unit, 1st Department of Neurology, School of Medicine, Aeginition Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Xristina Tsipota
- Demyelinating Diseases Unit, 1st Department of Neurology, School of Medicine, Aeginition Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Georgios Velonakis
- Research Unit of Radiology, 2nd Department of Radiology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | | | - Maria-Eleftheria Evangelopoulos
- Demyelinating Diseases Unit, 1st Department of Neurology, School of Medicine, Aeginition Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Leonidas Stefanis
- Demyelinating Diseases Unit, 1st Department of Neurology, School of Medicine, Aeginition Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Constantinos Kilidireas
- Demyelinating Diseases Unit, 1st Department of Neurology, School of Medicine, Aeginition Hospital, National and Kapodistrian University of Athens, Athens, Greece.,Department of Neurology, Henry Dunant Hospital Center, Athens, Greece
| |
Collapse
|
42
|
Clarkson BDS, Johnson RK, Bingel C, Lothaller C, Howe CL. Preservation of antigen-specific responses in cryopreserved CD4 + and CD8 + T cells expanded with IL-2 and IL-7. J Transl Autoimmun 2022; 5:100173. [PMID: 36467614 PMCID: PMC9713293 DOI: 10.1016/j.jtauto.2022.100173] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Revised: 10/31/2022] [Accepted: 11/20/2022] [Indexed: 11/27/2022] Open
Abstract
Objectives We sought to develop medium throughput standard operating procedures for screening cryopreserved human peripheral blood mononuclear cells (PBMCs) for CD4+ and CD8+ T cell responses to potential autoantigens. Methods Dendritic cells were loaded with a peptide cocktail from ubiquitous viruses or full-length viral protein antigens and cocultured with autologous T cells. We measured expression of surface activation markers on T cells by flow cytometry and cytometry by time of flight 24-72 h later. We tested responses among T cells freshly isolated from healthy control PBMCs, cryopreserved T cells, and T cells derived from a variety of T cell expansion protocols. We also compared the transcriptional profile of CD8+ T cells rested with interleukin (IL)7 for 48 h after 1) initial thawing, 2) expansion, and 3) secondary cryopreservation/thawing of expanded cells. To generate competent antigen presenting cells from PBMCs, we promoted differentiation of PBMCs into dendritic cells with granulocyte macrophage colony stimulating factor and IL-4. Results We observed robust dendritic cell differentiation from human PBMCs treated with 50 ng/mL GM-CSF and 20 ng/mL IL-4 in as little as 3 days. Dendritic cell purity was substantially increased by magnetically enriching for CD14+ monocytes prior to differentiation. We also measured antigen-dependent T cell activation in DC-T cell cocultures. However, polyclonal expansion of T cells with anti-CD3/antiCD28 abolished antigen-dependent upregulation of CD69 in our assay despite minimal transcriptional differences between rested CD8+ T cells before and after expansion. Furthermore, resting these expanded T cells in IL-2, IL-7 or IL-15 did not restore the antigen dependent responses. In contrast, T cells that were initially expanded with IL-2 + IL-7 rather than plate bound anti-CD3 + anti-CD28 retained responsiveness to antigen stimulation and these responses strongly correlated with responses measured at initial thawing. Significance While screening techniques for potential pathological autoantibodies have come a long way, comparable full-length protein target assays for screening patient T cells at medium throughput are noticeably lacking due to technical hurdles. Here we advance techniques that should have broad applicability to translational studies investigating cell mediated immunity in infectious or autoimmune diseases. Future studies are aimed at investigating possible CD8+ T cell autoantigens in MS and other CNS autoimmune diseases.
Collapse
Affiliation(s)
- Benjamin DS. Clarkson
- Department of Neurology, Mayo Clinic, Rochester, MN, 55905, USA,Center for Multiple Sclerosis and Autoimmune Neurology, Mayo Clinic, Rochester, MN, 55905, USA,Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, 55905, USA,Corresponding author. Mayo Clinic, Guggenheim 1521C, 200 First Street SW, Rochester, MN, 55905.
| | | | - Corinna Bingel
- Clinical Cooperation Unit Pediatric Oncology, German Cancer Research Center, Heidelberg, Germany
| | | | - Charles L. Howe
- Department of Neurology, Mayo Clinic, Rochester, MN, 55905, USA,Center for Multiple Sclerosis and Autoimmune Neurology, Mayo Clinic, Rochester, MN, 55905, USA,Division of Experimental Neurology, Mayo Clinic, Rochester, MN, 55905, USA,Department of Immunology, Mayo Clinic, Rochester, MN, 55905, USA
| |
Collapse
|
43
|
Liu R, Du S, Zhao L, Jain S, Sahay K, Rizvanov A, Lezhnyova V, Khaibullin T, Martynova E, Khaiboullina S, Baranwal M. Autoreactive lymphocytes in multiple sclerosis: Pathogenesis and treatment target. Front Immunol 2022; 13:996469. [PMID: 36211343 PMCID: PMC9539795 DOI: 10.3389/fimmu.2022.996469] [Citation(s) in RCA: 53] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2022] [Accepted: 09/08/2022] [Indexed: 11/13/2022] Open
Abstract
Multiple sclerosis (MS) is a chronic inflammatory disease of the central nervous system (CNS) characterized by destruction of the myelin sheath structure. The loss of myelin leads to damage of a neuron’s axon and cell body, which is identified as brain lesions on magnetic resonance image (MRI). The pathogenesis of MS remains largely unknown. However, immune mechanisms, especially those linked to the aberrant lymphocyte activity, are mainly responsible for neuronal damage. Th1 and Th17 populations of lymphocytes were primarily associated with MS pathogenesis. These lymphocytes are essential for differentiation of encephalitogenic CD8+ T cell and Th17 lymphocyte crossing the blood brain barrier and targeting myelin sheath in the CNS. B-lymphocytes could also contribute to MS pathogenesis by producing anti-myelin basic protein antibodies. In later studies, aberrant function of Treg and Th9 cells was identified as contributing to MS. This review summarizes the aberrant function and count of lymphocyte, and the contributions of these cell to the mechanisms of MS. Additionally, we have outlined the novel MS therapeutics aimed to amend the aberrant function or counts of these lymphocytes.
Collapse
Affiliation(s)
- Rongzeng Liu
- Department of Immunology, School of Basic Medical Sciences, Henan University of Science and Technology, Luoyang, China
| | - Shushu Du
- Department of Immunology, School of Basic Medical Sciences, Henan University of Science and Technology, Luoyang, China
| | - Lili Zhao
- Department of Immunology, School of Basic Medical Sciences, Henan University of Science and Technology, Luoyang, China
| | - Sahil Jain
- Department of Biochemistry and Molecular Biology, Faculty of Life Sciences, Tel-Aviv University, Tel-Aviv, Israel
| | - Kritika Sahay
- Department of Biotechnology, Thapar Institute of Engineering and Technology, Patiala, India
| | - Albert Rizvanov
- Gene and cell Department, Kazan Federal University, Kazan, Russia
| | - Vera Lezhnyova
- Gene and cell Department, Kazan Federal University, Kazan, Russia
| | - Timur Khaibullin
- Neurological Department, Republican Clinical Neurological Center, Kazan, Russia
| | | | - Svetlana Khaiboullina
- Gene and cell Department, Kazan Federal University, Kazan, Russia
- *Correspondence: Svetlana Khaiboullina, ; Manoj Baranwal, ;
| | - Manoj Baranwal
- Department of Biotechnology, Thapar Institute of Engineering and Technology, Patiala, India
- *Correspondence: Svetlana Khaiboullina, ; Manoj Baranwal, ;
| |
Collapse
|
44
|
Lee AYS. CD20 + T cells: an emerging T cell subset in human pathology. Inflamm Res 2022; 71:1181-1189. [PMID: 35951029 PMCID: PMC9616751 DOI: 10.1007/s00011-022-01622-x] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 07/23/2022] [Accepted: 07/27/2022] [Indexed: 11/13/2022] Open
Abstract
Introduction Although CD20 is classically a B cell marker, in the last three decades, dim expression has been noted on a subset of T cells as well that has been independently verified by a number of groups. Our understanding of these cells and their function is not well established. Methods A thorough review of original articles on CD20+T cells was undertaken of Pubmed by using combination of phrases including “CD20+”, “CD20-positive” and “T cells”. Articles in English were considered, and there was no time restriction. Results CD20+T cells express the standard T cell markers and, in comparison to CD20¯ T cells, appear to express greater inflammatory cytokines and markers of effector function. Although the ontogeny of these cells is still being established, the current theory is that CD20 may be acquired by trogocytosis from B cells. CD20+T cells may be found in healthy controls and in a wide range of pathologies including autoimmune diseases, haematological and non-haematological malignancies and human immunodeficiency virus (HIV) infections. One of the best studied diseases where these cells are found is multiple sclerosis (MS) where a number of therapeutic interventions, including anti-CD20 depletion, have been shown to effectively deplete these cells. Conclusion This review summarises the latest understanding of CD20+T cells, their presence in various diseases, their putative function and how they may be an ongoing target of CD20-depleting agents. Unfortunately, our understanding of these cells is still at its infancy and ongoing study in a wider range of pathologies is required.
Collapse
Affiliation(s)
- Adrian Y S Lee
- Department of Clinical Immunology, Westmead Hospital, Hawkesbury Road, Westmead, NSW, 2145, Australia. .,Department of Immunopathology, ICPMR and NSW Health Pathology, Westmead Hospital, Westmead, NSW, Australia. .,Department of Medicine, Westmead Clinical School, The University of Sydney, Westmead, NSW, Australia.
| |
Collapse
|
45
|
von Essen MR, Hansen RH, Højgaard C, Ammitzbøll C, Wiendl H, Sellebjerg F. Ofatumumab Modulates Inflammatory T Cell Responses and Migratory Potential in Patients With Multiple Sclerosis. NEUROLOGY(R) NEUROIMMUNOLOGY & NEUROINFLAMMATION 2022; 9:9/4/e200004. [PMID: 35672145 PMCID: PMC9272791 DOI: 10.1212/nxi.0000000000200004] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Accepted: 04/20/2022] [Indexed: 12/24/2022]
Abstract
BACKGROUND AND OBJECTIVES The anti-CD20 antibody ofatumumab is an efficacious therapy for multiple sclerosis (MS) through depletion of B cells. The purpose of this study was to examine the derivative effects of B cell depletion on the peripheral immune system and a direct treatment effect on T cells expressing CD20. METHODS Frequency and absolute numbers of peripheral leukocytes of treatment-naive patients with relapsing-remitting MS (RRMS) and patients treated with ofatumumab for a mean of 482 days were assessed in this observational study by flow cytometry. In addition, effector function and CNS migration of T cells using a human in vitro blood-brain barrier (BBB) assay were analyzed. RESULTS This study showed that ofatumumab treatment of patients with RRMS increased the control of effector T cells and decreased T cell autoreactivity. It also showed that ofatumumab reduced the level of peripheral CD20+ T cells and that the observed decrease in CNS-migratory capacity of T cells was caused by the depletion of CD20+ T cells. Finally, our study pointed out a bias in the measurement of CD20+ cells due to a steric hindrance between the treatment antibody and the flow cytometry antibody. DISCUSSION The substantial ofatumumab-induced alteration in the T cell compartment including a severely decreased CNS-migratory capacity of T cells could partly be attributed to the depletion of CD20+ T cells. Therefore, we propose that depletion of CD20+ T cells contributes to the positive treatment effect of ofatumumab and suggests that ofatumumab therapy should be considered a B cell and CD20+ T cell depletion therapy. CLASSIFICATION OF EVIDENCE This study provides Class IV evidence that compared with treatment-naive patients, ofatumumab treatment of patients with RRMS decreases peripheral CD20+ T cells, increases effector T cell control, and decreases T cell autoreactivity.
Collapse
Affiliation(s)
- Marina Rode von Essen
- From the Danish Multiple Sclerosis Center (M.R.E., R.H.H., C.H., C.A., F.S.), Department of Neurology, Rigshospitalet, University of Copenhagen, Glostrup, Denmark; and Department of Neurology with Institute of Translational Neurology (H.W.), University Hospital Münster, Germany.
| | - Rikke Holm Hansen
- From the Danish Multiple Sclerosis Center (M.R.E., R.H.H., C.H., C.A., F.S.), Department of Neurology, Rigshospitalet, University of Copenhagen, Glostrup, Denmark; and Department of Neurology with Institute of Translational Neurology (H.W.), University Hospital Münster, Germany
| | - Camilla Højgaard
- From the Danish Multiple Sclerosis Center (M.R.E., R.H.H., C.H., C.A., F.S.), Department of Neurology, Rigshospitalet, University of Copenhagen, Glostrup, Denmark; and Department of Neurology with Institute of Translational Neurology (H.W.), University Hospital Münster, Germany
| | - Cecilie Ammitzbøll
- From the Danish Multiple Sclerosis Center (M.R.E., R.H.H., C.H., C.A., F.S.), Department of Neurology, Rigshospitalet, University of Copenhagen, Glostrup, Denmark; and Department of Neurology with Institute of Translational Neurology (H.W.), University Hospital Münster, Germany
| | - Heinz Wiendl
- From the Danish Multiple Sclerosis Center (M.R.E., R.H.H., C.H., C.A., F.S.), Department of Neurology, Rigshospitalet, University of Copenhagen, Glostrup, Denmark; and Department of Neurology with Institute of Translational Neurology (H.W.), University Hospital Münster, Germany
| | - Finn Sellebjerg
- From the Danish Multiple Sclerosis Center (M.R.E., R.H.H., C.H., C.A., F.S.), Department of Neurology, Rigshospitalet, University of Copenhagen, Glostrup, Denmark; and Department of Neurology with Institute of Translational Neurology (H.W.), University Hospital Münster, Germany
| |
Collapse
|
46
|
Boziki M, Bakirtzis C, Sintila SA, Kesidou E, Gounari E, Ioakimidou A, Tsavdaridou V, Skoura L, Fylaktou A, Nikolaidou V, Stangou M, Nikolaidis I, Giantzi V, Karafoulidou E, Theotokis P, Grigoriadis N. Ocrelizumab in Patients with Active Primary Progressive Multiple Sclerosis: Clinical Outcomes and Immune Markers of Treatment Response. Cells 2022; 11:cells11121959. [PMID: 35741088 PMCID: PMC9222195 DOI: 10.3390/cells11121959] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Revised: 06/14/2022] [Accepted: 06/15/2022] [Indexed: 02/04/2023] Open
Abstract
Ocrelizumab is a B-cell-depleting monoclonal antibody approved for the treatment of relapsing-remitting multiple sclerosis (RRMS) and active primary progressive MS (aPPMS). This prospective, uncontrolled, open-label, observational study aimed to assess the efficacy of ocrelizumab in patients with aPPMS and to dissect the clinical, radiological and laboratory attributes of treatment response. In total, 22 patients with aPPMS followed for 24 months were included. The primary efficacy outcome was the proportion of patients with optimal response at 24 months, defined as patients free of relapses, free of confirmed disability accumulation (CDA) and free of T1 Gd-enhancing lesions and new/enlarging T2 lesions on the brain and cervical MRI. In total, 14 (63.6%) patients and 13 patients (59.1%) were classified as responders at 12 and 24 months, respectively. Time exhibited a significant effect on mean absolute and normalized gray matter cerebellar volume (F = 4.342, p = 0.23 and F = 4.279, p = 0.024, respectively). Responders at 24 months exhibited reduced peripheral blood ((%) of CD19+ cells) plasmablasts compared to non-responders at the 6-month point estimate (7.69 ± 4.4 vs. 22.66 ± 7.19, respectively, p = 0.043). Response to ocrelizumab was linked to lower total and gray matter cerebellar volume loss over time. Reduced plasmablast depletion was linked for the first time to sub-optimal response to ocrelizumab in aPPMS.
Collapse
Affiliation(s)
- Marina Boziki
- Multiple Sclerosis Center of the 2nd Neurological University Department, School of Medicine, Aristotle University of Thessaloniki, AHEPA General University Hospital, 54636 Thessaloniki, Greece; (M.B.); (C.B.); (S.-A.S.); (E.K.); (I.N.); (V.G.); (E.K.); (P.T.)
| | - Christos Bakirtzis
- Multiple Sclerosis Center of the 2nd Neurological University Department, School of Medicine, Aristotle University of Thessaloniki, AHEPA General University Hospital, 54636 Thessaloniki, Greece; (M.B.); (C.B.); (S.-A.S.); (E.K.); (I.N.); (V.G.); (E.K.); (P.T.)
| | - Styliani-Aggeliki Sintila
- Multiple Sclerosis Center of the 2nd Neurological University Department, School of Medicine, Aristotle University of Thessaloniki, AHEPA General University Hospital, 54636 Thessaloniki, Greece; (M.B.); (C.B.); (S.-A.S.); (E.K.); (I.N.); (V.G.); (E.K.); (P.T.)
| | - Evangelia Kesidou
- Multiple Sclerosis Center of the 2nd Neurological University Department, School of Medicine, Aristotle University of Thessaloniki, AHEPA General University Hospital, 54636 Thessaloniki, Greece; (M.B.); (C.B.); (S.-A.S.); (E.K.); (I.N.); (V.G.); (E.K.); (P.T.)
| | - Evdoxia Gounari
- Microbiology Laboratory, Department of Immunology, AHEPA University Hospital, 54636 Thessaloniki, Greece; (E.G.); (A.I.); (V.T.); (L.S.)
| | - Aliki Ioakimidou
- Microbiology Laboratory, Department of Immunology, AHEPA University Hospital, 54636 Thessaloniki, Greece; (E.G.); (A.I.); (V.T.); (L.S.)
| | - Vasiliki Tsavdaridou
- Microbiology Laboratory, Department of Immunology, AHEPA University Hospital, 54636 Thessaloniki, Greece; (E.G.); (A.I.); (V.T.); (L.S.)
| | - Lemonia Skoura
- Microbiology Laboratory, Department of Immunology, AHEPA University Hospital, 54636 Thessaloniki, Greece; (E.G.); (A.I.); (V.T.); (L.S.)
| | - Asimina Fylaktou
- National Peripheral Histocompatibility Center, Immunology Department, Hippokration General Hospital, 54642 Thessaloniki, Greece; (A.F.); (V.N.)
| | - Vasiliki Nikolaidou
- National Peripheral Histocompatibility Center, Immunology Department, Hippokration General Hospital, 54642 Thessaloniki, Greece; (A.F.); (V.N.)
| | - Maria Stangou
- Department of Nephrology, Medical School, Aristotle University of Thessaloniki, Hippokration Hospital, 54642 Thessaloniki, Greece;
| | - Ioannis Nikolaidis
- Multiple Sclerosis Center of the 2nd Neurological University Department, School of Medicine, Aristotle University of Thessaloniki, AHEPA General University Hospital, 54636 Thessaloniki, Greece; (M.B.); (C.B.); (S.-A.S.); (E.K.); (I.N.); (V.G.); (E.K.); (P.T.)
| | - Virginia Giantzi
- Multiple Sclerosis Center of the 2nd Neurological University Department, School of Medicine, Aristotle University of Thessaloniki, AHEPA General University Hospital, 54636 Thessaloniki, Greece; (M.B.); (C.B.); (S.-A.S.); (E.K.); (I.N.); (V.G.); (E.K.); (P.T.)
| | - Eleni Karafoulidou
- Multiple Sclerosis Center of the 2nd Neurological University Department, School of Medicine, Aristotle University of Thessaloniki, AHEPA General University Hospital, 54636 Thessaloniki, Greece; (M.B.); (C.B.); (S.-A.S.); (E.K.); (I.N.); (V.G.); (E.K.); (P.T.)
| | - Paschalis Theotokis
- Multiple Sclerosis Center of the 2nd Neurological University Department, School of Medicine, Aristotle University of Thessaloniki, AHEPA General University Hospital, 54636 Thessaloniki, Greece; (M.B.); (C.B.); (S.-A.S.); (E.K.); (I.N.); (V.G.); (E.K.); (P.T.)
| | - Nikolaos Grigoriadis
- Multiple Sclerosis Center of the 2nd Neurological University Department, School of Medicine, Aristotle University of Thessaloniki, AHEPA General University Hospital, 54636 Thessaloniki, Greece; (M.B.); (C.B.); (S.-A.S.); (E.K.); (I.N.); (V.G.); (E.K.); (P.T.)
- Special Unit for Biomedical Research and Education, School of Medicine, Aristotle University of Thessaloniki, 54636 Thessaloniki, Greece
- Correspondence:
| |
Collapse
|
47
|
Dorcet G, Migné H, Biotti D, Bost C, Lerebours F, Ciron J, Treiner E. Early B cells repopulation in multiple sclerosis patients treated with rituximab is not predictive of a risk of relapse or clinical progression. J Neurol 2022; 269:5443-5453. [PMID: 35652942 PMCID: PMC9159933 DOI: 10.1007/s00415-022-11197-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Revised: 05/12/2022] [Accepted: 05/15/2022] [Indexed: 11/29/2022]
Abstract
BACKGROUND It is currently unknown whether early B cell reconstitution (EBR) in MS patients under rituximab is associated with a risk of relapse or progression. OBJECTIVES Analyzing EBR in rituximab-treated patients and its putative association with clinical findings. METHODS Prospective lymphocytes immunophenotyping was performed in a monocentric cohort of MS patients treated by rituximab for 2 years. EBR was defined when B cells concentration was > 5 cells/mm3. B cell subsets were retrospectively associated with clinical data. Clinical and radiological monitoring included relapses, EDSS (Expanded Disability Status Scale), SDMT (Symbol Digit Modalities Test), and MRI. RESULTS 182 patients were analyzed (61 remitting-relapsing and 121 progressive-active). 38.5% experienced EBR at least once, but very few (7/182) showed systematic reconstitution. Most patients remained stable upon treatment, regardless of the occurrence of EBR. Dynamics of B cell reconstitution featured increased naïve/transitional B cells, and decreased memory subsets. Homeostasis of the B cell compartment differed at baseline between patients experiencing or not EBR upon treatment. In patients with EBR, reciprocal dynamics of transitional and pro-inflammatory double-negative B cell subsets was associated with better response to rituximab treatment. CONCLUSION EBR is common in rituximab-treated MS patients and is not associated with clinical disease activity. EBR in the peripheral blood may reflect regulatory immunological phenomena in subgroup of patients.
Collapse
Affiliation(s)
- Guillaume Dorcet
- Department of Neurology, CRC-SEP, University Hospital of Toulouse, Toulouse, France.,INSERM U1291-CNRS 5051, INFINITy, Toulouse, France
| | - Hugo Migné
- Immunology Laboratory, Biology Department, University Hospital of Toulouse, Toulouse, France
| | - Damien Biotti
- Department of Neurology, CRC-SEP, University Hospital of Toulouse, Toulouse, France.,INSERM U1291-CNRS 5051, INFINITy, Toulouse, France
| | - Chloé Bost
- INSERM U1291-CNRS 5051, INFINITy, Toulouse, France.,Immunology Laboratory, Biology Department, University Hospital of Toulouse, Toulouse, France
| | - Fleur Lerebours
- Department of Neurology, CRC-SEP, University Hospital of Toulouse, Toulouse, France
| | - Jonathan Ciron
- Department of Neurology, CRC-SEP, University Hospital of Toulouse, Toulouse, France.,INSERM U1291-CNRS 5051, INFINITy, Toulouse, France
| | - Emmanuel Treiner
- INSERM U1291-CNRS 5051, INFINITy, Toulouse, France. .,Immunology Laboratory, Biology Department, University Hospital of Toulouse, Toulouse, France.
| |
Collapse
|
48
|
Abstract
Multiple sclerosis (MS) is a chronic autoimmune demyelinating disease of the central nervous system (CNS) that often progresses to severe disability. Previous studies have highlighted the role of T cells in disease pathophysiology; however, the success of B-cell-targeted therapies has led to an increased interest in how B cells contribute to disease immunopathology. In this review, we summarize evidence of B-cell involvement in MS disease mechanisms, starting with pathology and moving on to review aspects of B cell immunobiology potentially relevant to MS. We describe current theories of critical B cell contributions to the inflammatory CNS milieu in MS, namely (i) production of autoantibodies, (ii) antigen presentation, (iii) production of proinflammatory cytokines (bystander activation), and (iv) EBV involvement. In the second part of the review, we summarize medications that have targeted B cells in patients with MS and their current position in the therapeutic armamentarium based on clinical trials and real-world data. Covered therapeutic strategies include the targeting of surface molecules such as CD20 (rituximab, ocrelizumab, ofatumumab, ublituximab) and CD19 (inebilizumab), and molecules necessary for B-cell activation such as B cell activating factor (BAFF) (belimumab) and Bruton's Tyrosine Kinase (BTK) (evobrutinib). We finally discuss the use of B-cell-targeted therapeutics in pregnancy.
Collapse
|
49
|
Bronge M, Högelin KA, Thomas OG, Ruhrmann S, Carvalho-Queiroz C, Nilsson OB, Kaiser A, Zeitelhofer M, Holmgren E, Linnerbauer M, Adzemovic MZ, Hellström C, Jelcic I, Liu H, Nilsson P, Hillert J, Brundin L, Fink K, Kockum I, Tengvall K, Martin R, Tegel H, Gräslund T, Al Nimer F, Guerreiro-Cacais AO, Khademi M, Gafvelin G, Olsson T, Grönlund H. Identification of four novel T cell autoantigens and personal autoreactive profiles in multiple sclerosis. SCIENCE ADVANCES 2022; 8:eabn1823. [PMID: 35476434 PMCID: PMC9045615 DOI: 10.1126/sciadv.abn1823] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Accepted: 02/17/2022] [Indexed: 05/29/2023]
Abstract
Multiple sclerosis (MS) is an inflammatory disease of the central nervous system (CNS), in which pathological T cells, likely autoimmune, play a key role. Despite its central importance, the autoantigen repertoire remains largely uncharacterized. Using a novel in vitro antigen delivery method combined with the Human Protein Atlas library, we screened for T cell autoreactivity against 63 CNS-expressed proteins. We identified four previously unreported autoantigens in MS: fatty acid-binding protein 7, prokineticin-2, reticulon-3, and synaptosomal-associated protein 91, which were verified to induce interferon-γ responses in MS in two cohorts. Autoreactive profiles were heterogeneous, and reactivity to several autoantigens was MS-selective. Autoreactive T cells were predominantly CD4+ and human leukocyte antigen-DR restricted. Mouse immunization induced antigen-specific responses and CNS leukocyte infiltration. This represents one of the largest systematic efforts to date in the search for MS autoantigens, demonstrates the heterogeneity of autoreactive profiles, and highlights promising targets for future diagnostic tools and immunomodulatory therapies in MS.
Collapse
Affiliation(s)
- Mattias Bronge
- Therapeutic Immune Design, Department of Clinical Neuroscience, Karolinska Institutet, Center for Molecular Medicine, 171 76 Stockholm, Sweden
| | - Klara Asplund Högelin
- Neuroimmunology Unit, Department of Clinical Neuroscience, Center for Molecular Medicine, Karolinska Institutet, 171 76 Stockholm, Sweden
| | - Olivia G. Thomas
- Therapeutic Immune Design, Department of Clinical Neuroscience, Karolinska Institutet, Center for Molecular Medicine, 171 76 Stockholm, Sweden
| | - Sabrina Ruhrmann
- Therapeutic Immune Design, Department of Clinical Neuroscience, Karolinska Institutet, Center for Molecular Medicine, 171 76 Stockholm, Sweden
| | - Claudia Carvalho-Queiroz
- Therapeutic Immune Design, Department of Clinical Neuroscience, Karolinska Institutet, Center for Molecular Medicine, 171 76 Stockholm, Sweden
| | - Ola B. Nilsson
- Therapeutic Immune Design, Department of Clinical Neuroscience, Karolinska Institutet, Center for Molecular Medicine, 171 76 Stockholm, Sweden
| | - Andreas Kaiser
- Therapeutic Immune Design, Department of Clinical Neuroscience, Karolinska Institutet, Center for Molecular Medicine, 171 76 Stockholm, Sweden
| | - Manuel Zeitelhofer
- Division of Vascular Biology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 171 77 Stockholm, Sweden
| | - Erik Holmgren
- Therapeutic Immune Design, Department of Clinical Neuroscience, Karolinska Institutet, Center for Molecular Medicine, 171 76 Stockholm, Sweden
| | - Mathias Linnerbauer
- Neuroimmunology Unit, Department of Clinical Neuroscience, Center for Molecular Medicine, Karolinska Institutet, 171 76 Stockholm, Sweden
| | - Milena Z. Adzemovic
- Neuroimmunology Unit, Department of Clinical Neuroscience, Center for Molecular Medicine, Karolinska Institutet, 171 76 Stockholm, Sweden
| | - Cecilia Hellström
- Division of Affinity Proteomics, Department of Protein Science, SciLifeLab, KTH–Royal Institute of Technology, 171 65 Solna, Sweden
| | - Ivan Jelcic
- Neuroimmunology and MS Research Section (NIMS), Neurology Clinic, University of Zürich, University Hospital Zürich, 8091 Zürich, Switzerland
| | - Hao Liu
- Department of Protein Science, KTH–Royal Institute of Technology, 114 21 Stockholm, Sweden
| | - Peter Nilsson
- Division of Affinity Proteomics, Department of Protein Science, SciLifeLab, KTH–Royal Institute of Technology, 171 65 Solna, Sweden
| | - Jan Hillert
- Department of Clinical Neuroscience, Division of Neurology, Karolinska Institutet, Karolinska University Hospital, 171 76 Stockholm, Sweden
| | - Lou Brundin
- Department of Clinical Neuroscience, Division of Neurology, Karolinska Institutet, Karolinska University Hospital, 171 76 Stockholm, Sweden
| | - Katharina Fink
- Department of Clinical Neuroscience, Division of Neurology, Karolinska Institutet, Karolinska University Hospital, 171 76 Stockholm, Sweden
| | - Ingrid Kockum
- Neuroimmunology Unit, Department of Clinical Neuroscience, Center for Molecular Medicine, Karolinska Institutet, 171 76 Stockholm, Sweden
| | - Katarina Tengvall
- Neuroimmunology Unit, Department of Clinical Neuroscience, Center for Molecular Medicine, Karolinska Institutet, 171 76 Stockholm, Sweden
- Science for Life Laboratory, Department of Medical Biochemistry and Microbiology, Uppsala University, 752 37 Uppsala, Sweden
| | - Roland Martin
- Neuroimmunology and MS Research Section (NIMS), Neurology Clinic, University of Zürich, University Hospital Zürich, 8091 Zürich, Switzerland
| | - Hanna Tegel
- Human Protein Atlas, Department of Protein Science, KTH–Royal Institute of Technology, Stockholm, Sweden
| | - Torbjörn Gräslund
- Department of Protein Science, KTH–Royal Institute of Technology, 114 21 Stockholm, Sweden
| | - Faiez Al Nimer
- Neuroimmunology Unit, Department of Clinical Neuroscience, Center for Molecular Medicine, Karolinska Institutet, 171 76 Stockholm, Sweden
| | - André Ortlieb Guerreiro-Cacais
- Neuroimmunology Unit, Department of Clinical Neuroscience, Center for Molecular Medicine, Karolinska Institutet, 171 76 Stockholm, Sweden
| | - Mohsen Khademi
- Neuroimmunology Unit, Department of Clinical Neuroscience, Center for Molecular Medicine, Karolinska Institutet, 171 76 Stockholm, Sweden
| | - Guro Gafvelin
- Therapeutic Immune Design, Department of Clinical Neuroscience, Karolinska Institutet, Center for Molecular Medicine, 171 76 Stockholm, Sweden
| | - Tomas Olsson
- Neuroimmunology Unit, Department of Clinical Neuroscience, Center for Molecular Medicine, Karolinska Institutet, 171 76 Stockholm, Sweden
| | - Hans Grönlund
- Therapeutic Immune Design, Department of Clinical Neuroscience, Karolinska Institutet, Center for Molecular Medicine, 171 76 Stockholm, Sweden
| |
Collapse
|
50
|
Stathopoulos P, Dalakas MC. Evolution of Anti-B Cell Therapeutics in Autoimmune Neurological Diseases. Neurotherapeutics 2022; 19:691-710. [PMID: 35182380 PMCID: PMC9294112 DOI: 10.1007/s13311-022-01196-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/31/2022] [Indexed: 02/08/2023] Open
Abstract
B cells have an ever-increasing role in the etiopathology of a number of autoimmune neurological disorders, acting as antigen-presenting cells facilitating antibody production but also as sensors, coordinators, and regulators of the immune response. In particular, B cells can regulate the T cell activation process through their participation in antigen presentation, production of proinflammatory cytokines (bystander activation or suppression), and contribution to ectopic lymphoid aggregates. Such an important interplay between B and T cells makes therapeutic depletion of B cells an attractive treatment strategy. The last decade, anti-B cell therapies using monoclonal antibodies against B cell surface molecules have evolved into a rational approach for successfully treating autoimmune neurological disorders, even when T cells seem to be the main effector cells. The paper summarizes basic aspects of B cell biology, discusses the roles of B cells in neurological autoimmunities, and highlights how the currently available or under development anti-B cell therapeutics exert their action in the wide spectrum and immunologically diverse neurological disorders. The efficacy of the various anti-B cell therapies and practical issues on induction and maintenance therapy is specifically detailed for the treatment of patients with multiple sclerosis, neuromyelitis-spectrum disorders, autoimmune encephalitis and hyperexcitability CNS disorders, autoimmune neuropathies, myasthenia gravis, and inflammatory myopathies. The success of anti-B cell therapies in inducing long-term remission in IgG4 neuroautoimmunities is also highlighted pointing out potential biomarkers for follow-up infusions.
Collapse
Affiliation(s)
- Panos Stathopoulos
- 1st Department of Neurology, National and Kapodistrian University of Athens, Athens, Greece
| | - Marinos C Dalakas
- Thomas Jefferson University, Philadelphia, PA, USA.
- Neuroimmunology Unit, National and Kapodistrian University of Athens, Athens, Greece.
| |
Collapse
|