1
|
Franco-Fuquen P, Figueroa-Aguirre J, Martínez DA, Moreno-Cortes EF, Garcia-Robledo JE, Vargas-Cely F, Castro-Martínez DA, Almaini M, Castro JE. Cellular therapies in rheumatic and musculoskeletal diseases. J Transl Autoimmun 2025; 10:100264. [PMID: 39931050 PMCID: PMC11808717 DOI: 10.1016/j.jtauto.2024.100264] [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: 10/29/2024] [Revised: 12/12/2024] [Accepted: 12/13/2024] [Indexed: 02/13/2025] Open
Abstract
A substantial proportion of patients diagnosed with rheumatologic and musculoskeletal diseases (RMDs) exhibit resistance to conventional therapies or experience recurrent symptoms. These diseases, which include autoimmune disorders such as multiple sclerosis, rheumatoid arthritis, and systemic lupus erythematosus, are marked by the presence of autoreactive B cells that play a critical role in their pathogenesis. The persistence of these autoreactive B cells within lymphatic organs and inflamed tissues impairs the effectiveness of B-cell-depleting monoclonal antibodies like rituximab. A promising therapeutic approach involves using T cells genetically engineered to express chimeric antigen receptors (CARs) that target specific antigens. This strategy has demonstrated efficacy in treating B-cell malignancies by achieving long-term depletion of malignant and normal B cells. Preliminary data from patients with RMDs, particularly those with lupus erythematosus and dermatomyositis, suggest that CAR T-cells targeting CD19 can induce rapid and sustained depletion of circulating B cells, leading to complete clinical and serological responses in cases that were previously unresponsive to conventional therapies. This review will provide an overview of the current state of preclinical and clinical studies on the use of CAR T-cells and other cellular therapies for RMDs. Additionally, it will explore potential future applications of these innovative treatment modalities for managing patients with refractory and recurrent manifestations of these diseases.
Collapse
Affiliation(s)
- Pedro Franco-Fuquen
- Division of Hematology and Medical Oncology, Mayo Clinic, Phoenix, AZ, USA
- Cancer Research and Cellular Therapies Laboratory, Mayo Clinic, Phoenix, AZ, USA
| | - Juana Figueroa-Aguirre
- Division of Hematology and Medical Oncology, Mayo Clinic, Phoenix, AZ, USA
- Cancer Research and Cellular Therapies Laboratory, Mayo Clinic, Phoenix, AZ, USA
| | - David A. Martínez
- Division of Hematology and Medical Oncology, Mayo Clinic, Phoenix, AZ, USA
- Cancer Research and Cellular Therapies Laboratory, Mayo Clinic, Phoenix, AZ, USA
| | - Eider F. Moreno-Cortes
- Division of Hematology and Medical Oncology, Mayo Clinic, Phoenix, AZ, USA
- Cancer Research and Cellular Therapies Laboratory, Mayo Clinic, Phoenix, AZ, USA
| | - Juan E. Garcia-Robledo
- Division of Hematology and Medical Oncology, Mayo Clinic, Phoenix, AZ, USA
- Cancer Research and Cellular Therapies Laboratory, Mayo Clinic, Phoenix, AZ, USA
| | - Fabio Vargas-Cely
- Division of Hematology and Medical Oncology, Mayo Clinic, Phoenix, AZ, USA
- Cancer Research and Cellular Therapies Laboratory, Mayo Clinic, Phoenix, AZ, USA
| | | | - Mustafa Almaini
- Rheumatology, Allergy & Clinical Immunology Division, Mafraq Hospital, United Arab Emirates
| | - Januario E. Castro
- Division of Hematology and Medical Oncology, Mayo Clinic, Phoenix, AZ, USA
- Cancer Research and Cellular Therapies Laboratory, Mayo Clinic, Phoenix, AZ, USA
| |
Collapse
|
2
|
Carter LM, Ehrenstein MR, Vital EM. Evolution and trajectory of B-cell targeted therapies in rheumatic diseases. THE LANCET. RHEUMATOLOGY 2025; 7:e355-e367. [PMID: 40058377 DOI: 10.1016/s2665-9913(24)00338-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 10/29/2024] [Accepted: 10/30/2024] [Indexed: 04/29/2025]
Abstract
Aberrant B-cell function, which could arise from various underlying causes, is central to the pathogenesis of diverse autoimmune rheumatic diseases. B cells remain the only cell type to be specifically therapeutically targeted through depletion and have the only therapy with a routinely available flow cytometric biomarker of treatment. Since first use and subsequent licensing for rheumatoid arthritis, rituximab has had a transformative impact on patients globally and across the rheumatic diseases. Further insights from B-cell-activating factor (BAFF) blockade with belimumab in systemic lupus erythematosus have followed. Examination of B-cell depletion, clinical outcomes, and re-emergent disease after treatment have deepened our understanding of the identity, detailed phenotype, biology, and kinetics of the B-cell subsets that are central to disease. This Review reflects on 20 years of clinical and translational insights drawn from B-cell targeted therapies for adult autoimmune rheumatic diseases, and highlights how these therapies have informed an exciting new era of future therapeutic developments.
Collapse
Affiliation(s)
- Lucy Marie Carter
- Newcastle-upon-Tyne Hospitals NHS Foundation Trust, Freeman Hospital, Newcastle, UK; Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Leeds, UK
| | | | - Edward M Vital
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Leeds, UK; NIHR Leeds Biomedical Research Centre, Leeds Teaching Hospitals NHS Trust, Leeds, UK.
| |
Collapse
|
3
|
Mougiakakos D, Meyer EH, Schett G. CAR T cells in autoimmunity: game changer or stepping stone? Blood 2025; 145:1841-1849. [PMID: 39700499 DOI: 10.1182/blood.2024025413] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Revised: 11/19/2024] [Accepted: 12/11/2024] [Indexed: 12/21/2024] Open
Abstract
ABSTRACT The advent of chimeric antigen receptor (CAR) T cells has revolutionized the treatment landscape for hematologic malignancies, and emerging evidence suggests their potential in autoimmune diseases (AIDs). This article evaluates the early successes and future implications of B-cell-targeting CAR T-cell therapy in AIDs. Initial applications, particularly in refractory systemic lupus erythematosus, have demonstrated significant and durable clinical remissions, with accompanying evaluation of the immune system suggesting a so-called "reset" of innate inflammation and adaptive autoimmunity. This has generated widespread interest in expanding this therapeutic approach. CAR T cells offer unique advantages over other treatment modalities, including very deep B-cell depletion and unique therapeutic activity within inflamed tissues and associated lymphoid structures. However, the field must address key concerns, including long-term toxicity, particularly the risk of secondary malignancies, and future accessibility given the higher prevalence of AIDs compared with malignancies. Technological advances in cell therapy, such as next-generation CAR T cells, allogeneic off-the-shelf products, and alternative cell types, such as regulatory CAR T cells, are being explored in AIDs to improve efficacy and safety. In addition, bispecific antibodies are emerging as potential alternatives or complements to CAR T cells, potentially offering comparable efficacy without the need for complex logistics, lymphodepletion, and the risk of insertional mutagenesis. As the field evolves, cellular therapists will play a critical role in the multidisciplinary teams managing these complex cases. The transformative potential of CAR T cells in AIDs is undeniable, but careful consideration of safety, efficacy, and implementation is essential as this novel therapeutic approach moves forward.
Collapse
Affiliation(s)
- Dimitrios Mougiakakos
- Department of Hematology, Oncology, and Cell Therapy, Otto von Guericke University, Magdeburg, Germany
| | - Everett H Meyer
- Cellular Immune Tolerance Program, Blood and Marrow Transplantation and Cellular Therapy Division, Stanford School of Medicine, Stanford University, Stanford, CA
| | - Georg Schett
- Department of Medicine 3, Rheumatology and Immunology, Friedrich-Alexander University, Erlangen, Germany
| |
Collapse
|
4
|
Teisseyre M, Allinovi M, Audard V, Cremoni M, Belvederi G, Karamé A, Accinno M, Duquesne J, Sharma V, Fernandez C, Zorzi K, El Maï M, Brglez V, Benzaken S, Esnault VL, Vultaggio A, Kohli HS, Ramachandran R, Cirami CL, Seitz-Polski B. Obinutuzumab and Ofatumumab are More Effective Than Rituximab in the Treatment of Membranous Nephropathy Patients With Anti-Rituximab Antibodies. Kidney Int Rep 2025; 10:753-761. [PMID: 40225374 PMCID: PMC11993203 DOI: 10.1016/j.ekir.2024.12.012] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Revised: 11/20/2024] [Accepted: 12/09/2024] [Indexed: 04/15/2025] Open
Abstract
Introduction Although rituximab has significantly improved outcomes for patients with membranous nephropathy, response to treatment is not universal and drug resistance can occur. One mechanism of resistance is the occurrence of antidrug antibodies. Obinutuzumab and ofatumumab are humanized and human monoclonal antibodies, respectively, that target B cells. These treatments have been shown to be effective in membranous nephropathy. However, obinutuzumab and ofatumumab have never been compared with rituximab in the treatment of patients with membranous nephropathy with anti-rituximab antibodies. We aimed to compare the efficacy and safety of obinutuzumab and ofatumumab with rituximab in patients with membranous nephropathy with anti-rituximab antibodies. Methods This international retrospective multicenter study enrolled 34 patients with membranous nephropathy from 5 nephrology departments in France, India, and Italy. All the patients had previously developed anti-rituximab antibodies. Nineteen patients received rituximab, 12 received obinutuzumab, and 3 received ofatumumab. Results Patients treated with obinutuzumab or ofatumumab were more likely to achieve clinical remission than those treated with rituximab at month 6 (87% vs. 37%, P = 0.005) and month 12 (87% vs. 42%, P = 0.01). Patients treated with obinutuzumab or ofatumumab were more likely to achieve immunological remission and B-cell depletion at month 6 than the patients treated with rituximab (92% vs. 56%, P = 0.04 and 93% vs. 35%, P = 0.002, respectively). No serious adverse events were reported in the obinutuzumab or ofatumumab group. Conclusion Obinutuzumab and ofatumumab are more effective than rituximab in treating patients with membranous nephropathy with anti-rituximab antibodies. Anti-rituximab antibodies should be systematically monitored, to determine appropriate treatment.
Collapse
Affiliation(s)
- Maxime Teisseyre
- French Reference Center for Rare Diseases, Idiopathic Nephrotic Syndrome and Membranous Nephropathy, Nice University Hospital, Université Côte d’Azur, Nice, France
- Immunology Laboratory, Nice University Hospital, Université Côte d’Azur, Nice, France
- Department of Nephrology, Dialysis and Transplantation, Nice University Hospital, Université Côte d’Azur, Nice, France
- Unité de Recherche Clinique Côte d’Azur, Université Côte d’Azur, Nice, France
| | - Marco Allinovi
- Department of Nephrology, Dialysis and Transplantation, Careggi University Hospital, Florence, Italy
| | - Vincent Audard
- French Reference Center for Rare Diseases, Idiopathic Nephrotic Syndrome and Membranous Nephropathy, Assistance Publique des Hôpitaux de Paris, Henri-Mondor University Hospital, Créteil, France
- Department of Nephrology, Dialysis and Transplantation, Assistance Publique des Hôpitaux de Paris, Henri-Mondor University Hospital, Créteil, France
- Paris Est Créteil University, Institut National de la Santé et de la Recherche Médicale U955, Institut Mondor de Recherche Biomédicale, Créteil, France
| | - Marion Cremoni
- French Reference Center for Rare Diseases, Idiopathic Nephrotic Syndrome and Membranous Nephropathy, Nice University Hospital, Université Côte d’Azur, Nice, France
- Immunology Laboratory, Nice University Hospital, Université Côte d’Azur, Nice, France
- Department of Nephrology, Dialysis and Transplantation, Nice University Hospital, Université Côte d’Azur, Nice, France
- Unité de Recherche Clinique Côte d’Azur, Université Côte d’Azur, Nice, France
| | - Giulia Belvederi
- Department of Nephrology, Dialysis and Transplantation, Careggi University Hospital, Florence, Italy
| | - Alexandre Karamé
- Department of Nephrology and Dialysis, Néphropôle - Médipole Hôpital Privé, Lyon-Villeurbanne, Villeurbanne, France
| | - Matteo Accinno
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Julien Duquesne
- Department of Pharmacy, Nice University Hospital, Université Côte d’Azur, Nice, France
| | - Vinod Sharma
- Department of Nephrology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Céline Fernandez
- French Reference Center for Rare Diseases, Idiopathic Nephrotic Syndrome and Membranous Nephropathy, Nice University Hospital, Université Côte d’Azur, Nice, France
- Unité de Recherche Clinique Côte d’Azur, Université Côte d’Azur, Nice, France
| | - Kévin Zorzi
- French Reference Center for Rare Diseases, Idiopathic Nephrotic Syndrome and Membranous Nephropathy, Nice University Hospital, Université Côte d’Azur, Nice, France
- Department of Nephrology, Dialysis and Transplantation, Nice University Hospital, Université Côte d’Azur, Nice, France
- Unité de Recherche Clinique Côte d’Azur, Université Côte d’Azur, Nice, France
| | - Mounir El Maï
- Immunology Laboratory, Nice University Hospital, Université Côte d’Azur, Nice, France
| | - Vesna Brglez
- French Reference Center for Rare Diseases, Idiopathic Nephrotic Syndrome and Membranous Nephropathy, Nice University Hospital, Université Côte d’Azur, Nice, France
- Immunology Laboratory, Nice University Hospital, Université Côte d’Azur, Nice, France
- Unité de Recherche Clinique Côte d’Azur, Université Côte d’Azur, Nice, France
| | - Sylvia Benzaken
- Immunology Laboratory, Nice University Hospital, Université Côte d’Azur, Nice, France
| | - Vincent L.M. Esnault
- French Reference Center for Rare Diseases, Idiopathic Nephrotic Syndrome and Membranous Nephropathy, Nice University Hospital, Université Côte d’Azur, Nice, France
- Department of Nephrology, Dialysis and Transplantation, Nice University Hospital, Université Côte d’Azur, Nice, France
| | - Alessandra Vultaggio
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
- Immunoallergology Unit, Careggi University Hospital, Florence, Italy
| | - Harbir Singh Kohli
- Department of Nephrology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Raja Ramachandran
- Department of Nephrology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Calogero Lino Cirami
- Department of Nephrology, Dialysis and Transplantation, Careggi University Hospital, Florence, Italy
| | - Barbara Seitz-Polski
- French Reference Center for Rare Diseases, Idiopathic Nephrotic Syndrome and Membranous Nephropathy, Nice University Hospital, Université Côte d’Azur, Nice, France
- Immunology Laboratory, Nice University Hospital, Université Côte d’Azur, Nice, France
- Department of Nephrology, Dialysis and Transplantation, Nice University Hospital, Université Côte d’Azur, Nice, France
- Unité de Recherche Clinique Côte d’Azur, Université Côte d’Azur, Nice, France
| |
Collapse
|
5
|
Liu J, Zhao Y, Zhao H. Chimeric antigen receptor T-cell therapy in autoimmune diseases. Front Immunol 2024; 15:1492552. [PMID: 39628482 PMCID: PMC11611814 DOI: 10.3389/fimmu.2024.1492552] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2024] [Accepted: 10/28/2024] [Indexed: 12/06/2024] Open
Abstract
The administration of T cells that have been modified to carry chimeric antigen receptors (CARs) aimed at B cells has been an effective strategy in treating B cell malignancies. This breakthrough has spurred the creation of CAR T cells intended to specifically reduce or alter the faulty immune responses associated with autoimmune disorders. Early positive outcomes from clinical trials involving CAR T cells that target the B cell protein CD19 in patients suffering from autoimmune diseases driven by B cells have been reported. Additional strategies are being developed to broaden the use of CAR T cell therapy and enhance its safety in autoimmune conditions. These include employing chimeric autoantireceptors (CAAR) to specifically eliminate B cells that are reactive to autoantigens, and using regulatory T cells (Tregs) engineered to carry antigen-specific CARs for precise immune modulation. This discussion emphasizes key factors such as choosing the right target cell groups, designing CAR constructs, defining tolerable side effects, and achieving a lasting immune modification, all of which are critical for safely integrating CAR T cell therapy in treating autoimmune diseases.
Collapse
MESH Headings
- Humans
- Autoimmune Diseases/therapy
- Autoimmune Diseases/immunology
- Receptors, Chimeric Antigen/immunology
- Receptors, Chimeric Antigen/genetics
- Immunotherapy, Adoptive/methods
- Immunotherapy, Adoptive/adverse effects
- Animals
- T-Lymphocytes/immunology
- T-Lymphocytes/transplantation
- T-Lymphocytes, Regulatory/immunology
- B-Lymphocytes/immunology
- Receptors, Antigen, T-Cell/immunology
- Receptors, Antigen, T-Cell/genetics
- Autoantigens/immunology
- Antigens, CD19/immunology
Collapse
Affiliation(s)
- Jie Liu
- Department of Neurosurgery, the Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Yan Zhao
- Department of Respiratory, Lanzhou University Second Hospital, Lanzhou, Gansu, China
| | - Hai Zhao
- Department of Neurosurgery, the Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| |
Collapse
|
6
|
Chung JB, Brudno JN, Borie D, Kochenderfer JN. Chimeric antigen receptor T cell therapy for autoimmune disease. Nat Rev Immunol 2024; 24:830-845. [PMID: 38831163 DOI: 10.1038/s41577-024-01035-3] [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: 04/17/2024] [Indexed: 06/05/2024]
Abstract
Infusion of T cells engineered to express chimeric antigen receptors (CARs) that target B cells has proven to be a successful treatment for B cell malignancies. This success inspired the development of CAR T cells to selectively deplete or modulate the aberrant immune responses that underlie autoimmune disease. Promising results are emerging from clinical trials of CAR T cells targeting the B cell protein CD19 in patients with B cell-driven autoimmune diseases. Further approaches are being designed to extend the application and improve safety of CAR T cell therapy in the setting of autoimmunity, including the use of chimeric autoantibody receptors to selectively deplete autoantigen-specific B cells and the use of regulatory T cells engineered to express antigen-specific CARs for targeted immune modulation. Here, we highlight important considerations, such as optimal target cell populations, CAR construct design, acceptable toxicities and potential for lasting immune reset, that will inform the eventual safe adoption of CAR T cell therapy for the treatment of autoimmune diseases.
Collapse
Affiliation(s)
| | - Jennifer N Brudno
- Surgery Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | | | - James N Kochenderfer
- Surgery Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
7
|
Rispens T, Kuijpers TW, Killestein J, van Kempen ZLE, Bloem K. Cross-Reactivity of Antibodies to Rituximab with Other Therapeutic Anti-CD20 Antibodies. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 212:529-533. [PMID: 38149924 DOI: 10.4049/jimmunol.2300647] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Accepted: 12/06/2023] [Indexed: 12/28/2023]
Abstract
One reason for a lack of response to rituximab as well as infusion-related anaphylactic adverse events is the development of antidrug Abs to rituximab. Besides rituximab, a number of other therapeutic Abs targeting CD20 are nowadays available as alternatives. In this study, we investigated the potential cross-reactivity of (human) anti-rituximab Abs to three other anti-CD20 mAbs: ofatumumab, obinutuzumab, and ocrelizumab. In 25 cases of anti-rituximab Abs, cross-reactivity was examined using both direct binding assays and inhibition immunoassays. Although no cross-reactivity was observed to ofatumumab or obinutuzumab, 8 of 25 samples also showed reactivity toward ocrelizumab in at least one of the two assays. Furthermore, in three cases of anti-ocrelizumab Abs, cross-reactivity to rituximab was observed in an inhibition immunoassay, albeit not in a direct binding assay. Our results suggest that obinutuzumab or ofatumumab are safe anti-CD20 alternatives in case of the presence of anti-rituximab Abs. It is advisable to proceed cautiously if switching from rituximab to ocrelizumab (or vice versa) is considered in case these alternatives may not be available.
Collapse
Affiliation(s)
- Theo Rispens
- Department of Immunopathology, Sanquin Research and Landsteiner Laboratory, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, the Netherlands
- Amsterdam Institute for Infection and Immunity, Amsterdam, the Netherlands
| | - Taco W Kuijpers
- Amsterdam Institute for Infection and Immunity, Amsterdam, the Netherlands
- Department of Pediatric Immunology, Rheumatology and Infectious Disease, Amsterdam UMC, location AMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Joep Killestein
- Amsterdam UMC location Vrije Universiteit Amsterdam, Neurology, Amsterdam, the Netherlands
| | - Zoé L E van Kempen
- Amsterdam UMC location Vrije Universiteit Amsterdam, Neurology, Amsterdam, the Netherlands
| | - Karien Bloem
- Antibodies and Immunogenicity, Sanquin Diagnostic Services, Amsterdam, the Netherlands
| |
Collapse
|
8
|
Pezot M, Nocturne G, Belkhir R, Henry J, Pavy S, Seror R, Mariette X, Bitoun S. Obinutuzumab in patients with Sjogren's disease immunised against rituximab. Ann Rheum Dis 2024; 83:407-408. [PMID: 37945315 DOI: 10.1136/ard-2023-224999] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2023] [Accepted: 10/23/2023] [Indexed: 11/12/2023]
Affiliation(s)
- Mathilde Pezot
- Rheumatology Department, Université Paris-Saclay, Hôpital Bicêtre, Assistance Publique - Hôpitaux de Paris, FHU CARE, INSERM UMR1184, Le Kremlin Bicêtre, France
| | - Gaétane Nocturne
- Rheumatology Department, Université Paris-Saclay, Hôpital Bicêtre, Assistance Publique - Hôpitaux de Paris, FHU CARE, INSERM UMR1184, Le Kremlin Bicêtre, France
| | - Rakiba Belkhir
- Rheumatology Department, Université Paris-Saclay, Hôpital Bicêtre, Assistance Publique - Hôpitaux de Paris, FHU CARE, INSERM UMR1184, Le Kremlin Bicêtre, France
| | - Julien Henry
- Rheumatology Department, Université Paris-Saclay, Hôpital Bicêtre, Assistance Publique - Hôpitaux de Paris, FHU CARE, INSERM UMR1184, Le Kremlin Bicêtre, France
| | - Stephan Pavy
- Rheumatology Department, Université Paris-Saclay, Hôpital Bicêtre, Assistance Publique - Hôpitaux de Paris, FHU CARE, INSERM UMR1184, Le Kremlin Bicêtre, France
| | - Raphaèle Seror
- Rheumatology Department, Université Paris-Saclay, Hôpital Bicêtre, Assistance Publique - Hôpitaux de Paris, FHU CARE, INSERM UMR1184, Le Kremlin Bicêtre, France
| | - Xavier Mariette
- Rheumatology Department, Université Paris-Saclay, Hôpital Bicêtre, Assistance Publique - Hôpitaux de Paris, FHU CARE, INSERM UMR1184, Le Kremlin Bicêtre, France
| | - Samuel Bitoun
- Rheumatology Department, Université Paris-Saclay, Hôpital Bicêtre, Assistance Publique - Hôpitaux de Paris, FHU CARE, INSERM UMR1184, Le Kremlin Bicêtre, France
| |
Collapse
|
9
|
Devauchelle-Pensec V, Mariette X, Benyoussef AA, Boisrame S, Cochener B, Cornec D, Nocturne G, Gottenberg JE, Hachulla E, Labalette P, Le Guern V, M'Bwang Seppoh R, Morel J, Orliaguet M, Saraux A, Seror R, Costedoat-Chalumeau N. French national diagnostic and care protocol for Sjögren's disease. Rev Med Interne 2023; 44:423-457. [PMID: 37453854 DOI: 10.1016/j.revmed.2023.07.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Accepted: 07/03/2023] [Indexed: 07/18/2023]
Abstract
Sjögren's disease (SD), also known as Sjögren's syndrome (SS) or Gougerot-Sjögren's syndrome in France, is a rare systemic autoimmune disease in its primary form and is characterised by tropism for the exocrine glandular epithelia, particularly the salivary and lacrimal glands. The lymphocytic infiltration of these epithelia will clinically translate into a dry syndrome which, associated with fatigue and pain, constitutes the symptom triad of the disease. In about one third of patients, SD is associated with systemic complications that can affect the joints, skin, lungs, kidneys, central or peripheral nervous system, and lymphoid organs with an increased risk of B-cell lymphoma. SD affects women more frequently than men (9/1). The peak frequency is around the age of 50. However, the disease can occur at any age, with paediatric forms occurring even though they remain rare. SD can occur alone or in association with other systemic autoimmune diseases. In its isolated or primary form, the prevalence of SD is estimated to be between 1 per 1000 and 1 per 10,000 inhabitants. The most recent classification criteria were developed in 2016 by EULAR and ACR. The course and prognosis of the disease are highly variable and depend on the presence of systemic involvement and the severity of the dryness of the eyes and mouth. The current approach is therefore to identify at an early stage those patients most at risk of systemic complications or lymphoma, who require close follow-up. On the other hand, regular monitoring of the ophthalmological damage and of the dental status should be ensured to reduce the consequences.
Collapse
Affiliation(s)
- Valérie Devauchelle-Pensec
- Service de Rhumatologie, CHU de Brest, Inserm 1227, LBAI, Université de Bretagne Occidentale, Centre de Référence des Maladies Auto-Immunes Systémiques Rares du Nord et Nord-Ouest de France (CeRAINO), 29609 Brest cedex, France.
| | - Xavier Mariette
- Service de Rhumatologie, Hôpital Bicètre, AP-HP, Université Paris-Saclay, Paris, France
| | | | - Sylvie Boisrame
- UFR d'Odontologie, University of Western Brittany, CHU de Brest, 29200 Brest, France
| | | | - Divi Cornec
- Service de Rhumatologie, CHU de Brest, Inserm 1227, LBAI, Université de Bretagne Occidentale, Centre de Référence des Maladies Auto-Immunes Systémiques Rares du Nord et Nord-Ouest de France (CeRAINO), 29609 Brest cedex, France
| | - Gaëtane Nocturne
- Service de Rhumatologie, Hôpital Bicètre, AP-HP, Université Paris-Saclay, Paris, France
| | - Jacques Eric Gottenberg
- Service de Rhumatologie, Hôpitaux Universitaires de Strasbourg, RESO, Centre de Référence des Maladies Auto-Immunes Systémiques Rares Est Sud-Ouest, 67000 Strasbourg, France
| | - Eric Hachulla
- Service de Médecine Interne et Immunologie Clinique, Centre de Référence des Maladies Auto-Immunes Systémiques Rares du Nord et Nord-Ouest de France (CeRAINO), Université de Lille, Inserm, CHU Lille, U1286, INFINITE, Institute for Translational Research in Inflammation, Lille, France
| | - Pierre Labalette
- Service d'Ophtalmologie, Hôpital Huriez, CHU de Lille, rue Michel-Polonowski, 59000 Lille, France
| | | | | | - Jacques Morel
- Département de Rhumatologie, CHU de Montpellier, Hôpital Lapeyronie, Inserm, PhyMedExp, CNRS, Montpellier, France
| | - Marie Orliaguet
- UFR d'Odontologie, University of Western Brittany, CHU de Brest, 29200 Brest, France
| | - Alain Saraux
- Service de Rhumatologie, CHU de Brest, Inserm 1227, LBAI, Université de Bretagne Occidentale, Centre de Référence des Maladies Auto-Immunes Systémiques Rares du Nord et Nord-Ouest de France (CeRAINO), 29609 Brest cedex, France
| | - Raphaèle Seror
- Service de Rhumatologie, Hôpital Bicètre, AP-HP, Université Paris-Saclay, Paris, France
| | | |
Collapse
|
10
|
Wincup C, Dunn N, Ruetsch-Chelli C, Manouchehrinia A, Kharlamova N, Naja M, Seitz-Polski B, Isenberg DA, Fogdell-Hahn A, Ciurtin C, Jury EC. Anti-rituximab antibodies demonstrate neutralizing capacity, associate with lower circulating drug levels and earlier relapse in lupus. Rheumatology (Oxford) 2023; 62:2601-2610. [PMID: 36370065 PMCID: PMC10321108 DOI: 10.1093/rheumatology/keac608] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 09/21/2022] [Accepted: 10/18/2022] [Indexed: 11/13/2022] Open
Abstract
OBJECTIVES High rates of anti-drug antibodies (ADA) to rituximab have been demonstrated in patients undergoing treatment for SLE. However, little is known with regard to their long-term dynamics, impact on drug kinetics and subsequent implications for treatment response. In this study, we aimed to evaluate ADA persistence over time, impact on circulating drug levels, assess clinical outcomes and whether they are capable of neutralizing rituximab. METHODS Patients with SLE undergoing treatment with rituximab were recruited to this study (n = 35). Serum samples were collected across a follow-up period of 36 months following treatment (n = 114). Clinical and laboratory data were collected pre-treatment and throughout follow-up. ADA were detected via electrochemiluminescent immunoassays. A complement dependent cytotoxicity assay was used to determine neutralizing capacity of ADA in a sub-cohort of positive samples (n = 38). RESULTS ADA persisted over the 36-month study period in 64.3% of patients undergoing treatment and titres peaked earlier and remained higher in those who had previously been treated with rituximab when compared with than those who were previously treatment naive. ADA-positive samples had a significantly lower median drug level until six months post rituximab infusion (P = 0.0018). Patients with persistent ADA positivity showed a significant early improvement in disease activity followed by increased rates of relapse. In vitro analysis confirmed the neutralizing capacity of ADA to rituximab. CONCLUSIONS ADA to rituximab were common and persisted over the 36-month period of this study. They associated with earlier drug elimination, an increased rate of relapse and demonstrated neutralizing capacity in vitro.
Collapse
Affiliation(s)
- Chris Wincup
- Centre for Rheumatology Research, Division of Medicine, University College London, London, UK
| | - Nicky Dunn
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
- Center for Molecular Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Caroline Ruetsch-Chelli
- Laboratoire d’Immunologie, CHU de Nice, Université Côte d’Azur, Nice, France
- Centre Méditerranéen de Médecine Moléculaire (C3M), INSERM U1065, Université Côte d’Azur, Nice, France
- Unité de Recherche Clinique Côte d’Azur (UR2CA), Université Côte d’Azur, Nice, France
| | - Ali Manouchehrinia
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
- Center for Molecular Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Nastya Kharlamova
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
- Center for Molecular Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Meena Naja
- Centre for Adolescent Rheumatology Research, Division of Medicine, University College London, London, UK
| | - Barbara Seitz-Polski
- Laboratoire d’Immunologie, CHU de Nice, Université Côte d’Azur, Nice, France
- Unité de Recherche Clinique Côte d’Azur (UR2CA), Université Côte d’Azur, Nice, France
| | - David A Isenberg
- Centre for Rheumatology Research, Division of Medicine, University College London, London, UK
| | - Anna Fogdell-Hahn
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
- Center for Molecular Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Coziana Ciurtin
- Centre for Rheumatology Research, Division of Medicine, University College London, London, UK
- Centre for Adolescent Rheumatology Research, Division of Medicine, University College London, London, UK
| | - Elizabeth C Jury
- Centre for Rheumatology Research, Division of Medicine, University College London, London, UK
| |
Collapse
|
11
|
Cao S, Du J, Pan S, Zhang J, Xu S, Wei L, Tian Y. Case Report: Ofatumumab treatment in a patient with rituximab-intolerant refractory autoimmune GFAP astrocytopathy. Front Immunol 2023; 14:1164181. [PMID: 37223100 PMCID: PMC10200986 DOI: 10.3389/fimmu.2023.1164181] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2023] [Accepted: 04/24/2023] [Indexed: 05/25/2023] Open
Abstract
BACKGROUND Ofatumumab, a fully humanized anti-CD20 monoclonal antibody, has shown promising efficacy in limited cases of neuromyelitis optica spectrum disorder, but there is a lack of studies on its use in autoimmune glial fibrillary acidic protein (GFAP) astrocytopathy. We present a case of refractory GFAP astrocytopathy with poor response to conventional immunosuppressants and rituximab who responded well to subcutaneous ofatumumab. CASE REPORT The patient is a 36-year-old woman with a diagnosis of GFAP astrocytopathy and high disease activity. She experienced five relapses over three years despite immunosuppressive treatment with oral prednisone, azathioprine, mycophenolate mofetil, and intravenous rituximab. Additionally, her circulating B cells were not completely depleted during the second administration of rituximab and an allergic reaction occurred. Based on insufficient B cell depletion and allergic reaction to rituximab, subcutaneous ofatumumab was introduced. After twelve injections of ofatumumab without injection-related reactions, she had no further relapses and was sufficiently depleted of the circulating B cells. DISCUSSION This case illustrates the effective use and good tolerance of ofatumumab in GFAP astrocytopathy. Further studies are needed to investigate the efficacy and safety of ofatumumab in refractory GFAP astrocytopathy or those intolerant to rituximab.
Collapse
Affiliation(s)
- Shugang Cao
- Department of Neurology, Second Affiliated Hospital of Anhui Medical University, Hefei, China
- Department of Neurology, The Second People’s Hospital of Hefei, Hefei, China
| | - Jing Du
- Department of Neurology, Second Affiliated Hospital of Anhui Medical University, Hefei, China
- Department of Psychology, Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Sidi Pan
- Department of Neurology, First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Juanjuan Zhang
- Department of Neurology, First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Si Xu
- Department of Neurology, Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Ling Wei
- Department of Neurology, First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Yanghua Tian
- Department of Neurology, Second Affiliated Hospital of Anhui Medical University, Hefei, China
| |
Collapse
|
12
|
Bai Y, Li W, Yan C, Hou Y, Wang Q. Anti-rituximab antibodies in patients with refractory autoimmune nodopathy with anti-neurofascin-155 antibody. Front Immunol 2023; 14:1121705. [PMID: 37056784 PMCID: PMC10086195 DOI: 10.3389/fimmu.2023.1121705] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Accepted: 03/14/2023] [Indexed: 03/30/2023] Open
Abstract
BackgroundRecent studies have reported that similar to other IgG4 autoimmune diseases, such as muscle-specific kinase antibody-associated myasthenia gravis, most anti-neurofascin-155 (anti-NF155) nodopathies respond well to rituximab treatment, regardless of the dosage. However, there are still a few patients for which rituximab is ineffective for unknown reasons. Currently, there are no studies on the mechanism of ineffective treatment with rituximab.MethodsA 33-year-old Chinese man presenting with numbness, tremor, and muscle weakness for 4 years was recruited for this study. Anti-NF155 antibodies were identified by cell-based assay and confirmed by immunofluorescence assay on teased fibers. The anti-NF155 immunoglobulin (IgG) subclasses were also detected by immunofluorescence assay. Anti-rituximab antibodies (ARAs) were quantitatively analyzed using enzyme-linked immunosorbent assay (ELISA), and peripheral B cell counts were determined by flow cytometry.ResultsThe patient exhibited anti-NF155 IgG4-antibody positivity. After the first round of rituximab infusion, the patient showed stratified outcomes with improvements in numbness, muscle weakness and ambulation. However, after three rounds of rituximab infusion, the patient’s symptoms deteriorated, and the numbness, tremor and muscle weakness returned. No obvious improvement was found after plasma exchange and another round of rituximab treatment. 14 days after the last treatment with rituximab, ARAs were detected. And the titers gradually decreased on day 28 and 60 but remained higher than normal. Peripheral CD19+ B cell counts were less than 1% within the 2-month period following the final rituximab administration.ConclusionsIn this study, ARAs presented in a patient with anti-NF155 nodopathy undergoing rituximab treatment and showed an unfavorable impact on rituximab efficacy. This is the first case to report the occurrence of ARAs in patients with anti-NF155 antibodies. We suggest that ARAs should be tested early during the initial intervention, especially in patients who respond poorly to rituximab treatment. In addition, we believe it is necessary to investigate the association between ARAs and B cell counts, their effect on clinical efficacy, and their potential adverse reactions in a larger cohort of patients with anti-NF155 nodopathy.
Collapse
Affiliation(s)
- Yunfei Bai
- Department of Neurology, Qilu Hospital of Shandong University, Jinan, China
| | - Wei Li
- Department of Neurology, Qilu Hospital of Shandong University, Jinan, China
| | - Chuanzhu Yan
- Department of Neurology, Qilu Hospital of Shandong University, Jinan, China
- Department of Central Laboratory and Mitochondrial Medicine Laboratory, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, Qingdao, China
- Brain Science Research Institute, Shandong University, Jinan, China
| | - Ying Hou
- Department of Neurology, Qilu Hospital of Shandong University, Jinan, China
- *Correspondence: Qinzhou Wang, ; Ying Hou,
| | - Qinzhou Wang
- Department of Neurology, Qilu Hospital of Shandong University, Jinan, China
- *Correspondence: Qinzhou Wang, ; Ying Hou,
| |
Collapse
|
13
|
Konen FF, Möhn N, Witte T, Schefzyk M, Wiestler M, Lovric S, Hufendiek K, Schwenkenbecher P, Sühs KW, Friese MA, Klotz L, Pul R, Pawlitzki M, Hagin D, Kleinschnitz C, Meuth SG, Skripuletz T. Treatment of autoimmunity: The impact of disease-modifying therapies in multiple sclerosis and comorbid autoimmune disorders. Autoimmun Rev 2023; 22:103312. [PMID: 36924922 DOI: 10.1016/j.autrev.2023.103312] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Accepted: 03/09/2023] [Indexed: 03/17/2023]
Abstract
More than 10 disease-modifying therapies (DMT) are approved by the European Medicines Agency (EMA) and the US Food and Drug Administration (FDA) for the treatment of multiple sclerosis (MS) and new therapeutic options are on the horizon. Due to different underlying therapeutic mechanisms, a more individualized selection of DMTs in MS is possible, taking into account the patient's current situation. Therefore, concomitant treatment of various comorbid conditions, including autoimmune mediated disorders such as rheumatoid arthritis, should be considered in MS patients. Because the pathomechanisms of autoimmunity partially overlap, DMT could also treat concomitant inflammatory diseases and simplify the patient's treatment. In contrast, the exacerbation and even new occurrence of several autoimmune diseases have been reported as a result of immunomodulatory treatment of MS. To simplify treatment and avoid disease exacerbation, knowledge of the beneficial and adverse effects of DMT in other autoimmune disorders is critical. Therefore, we conducted a literature search and described the beneficial and adverse effects of approved and currently studied DMT in a large number of comorbid autoimmune diseases, including rheumatoid arthritis, ankylosing spondylitis, inflammatory bowel diseases, cutaneous disorders including psoriasis, Sjögren´s syndrome, systemic lupus erythematosus, systemic vasculitis, autoimmune hepatitis, and ocular autoimmune disorders. Our review aims to facilitate the selection of an appropriate DMT in patients with MS and comorbid autoimmune diseases.
Collapse
Affiliation(s)
- Franz Felix Konen
- Department of Neurology, Hannover Medical School, 30625 Hannover, Germany..
| | - Nora Möhn
- Department of Neurology, Hannover Medical School, 30625 Hannover, Germany..
| | - Torsten Witte
- Department of Rheumatology and Clinical Immunology, Hannover Medical School, 30625 Hannover, Germany..
| | - Matthias Schefzyk
- Department of Dermatology, Allergology and Venerology, Hannover Medical School, 30625 Hannover, Germany..
| | - Miriam Wiestler
- Department of Internal Medicine, Division of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, 30625 Hannover, Germany.
| | - Svjetlana Lovric
- Department of Nephrology and Hypertension, Hannover Medical School, 30625 Hannover, Germany.
| | - Karsten Hufendiek
- University Eye Hospital, Hannover Medical School, 30625 Hannover, Germany.
| | | | - Kurt-Wolfram Sühs
- Department of Neurology, Hannover Medical School, 30625 Hannover, Germany..
| | - Manuel A Friese
- Institute of Neuroimmunology and Multiple Sclerosis, Center for Molecular Neurobiology Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg 20251, Germany.
| | - Luisa Klotz
- Department of Neurology with Institute of Translational Neurology, University Hospital Muenster, 48149 Muenster, Germany.
| | - Refik Pul
- Department of Neurology, University Medicine Essen, Essen, Germany; Center for Translational Neuro- and Behavioral Sciences, University Hospital Essen, Essen 45147, Germany.
| | - Marc Pawlitzki
- Department of Neurology, Medical Faculty, Heinrich Heine University Dusseldorf, 40225 Dusseldorf, Germany.
| | - David Hagin
- Allergy and Clinical Immunology Unit, Department of Medicine, Tel-Aviv Sourasky Medical Center and Sackler Faculty of Medicine, University of Tel Aviv, 6 Weizmann St., Tel-Aviv 6423906, Israel.
| | - Christoph Kleinschnitz
- Department of Neurology, University Medicine Essen, Essen, Germany; Center for Translational Neuro- and Behavioral Sciences, University Hospital Essen, Essen 45147, Germany.
| | - Sven G Meuth
- Department of Neurology, Medical Faculty, Heinrich Heine University Dusseldorf, 40225 Dusseldorf, Germany.
| | - Thomas Skripuletz
- Department of Neurology, Hannover Medical School, 30625 Hannover, Germany..
| |
Collapse
|
14
|
Quartuccio L, Treppo E, Urso L, Del Frate G, Mescia F, Alberici F, Vaglio A, Emmi G. Unmet needs in ANCA-associated vasculitis: Physicians' and patients' perspectives. Front Immunol 2023; 14:1112899. [PMID: 36911748 PMCID: PMC9995379 DOI: 10.3389/fimmu.2023.1112899] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Accepted: 02/13/2023] [Indexed: 02/25/2023] Open
Abstract
In recent years, clinical research has increased significantly and therapies for antineutrophil cytoplasmic antibody (ANCA)-associated vasculitis have improved. However, there are still unanswered questions and unmet needs about AAV patients. The purpose of this review is to examine the frontiers of research related to emerging biomarkers eventually predicting relapse, and new therapeutic approaches, not to mention new quality of life assessment tools. Identifying predictors of relapse may help optimize therapeutic strategies, minimize disease recurrence, and reduce treatment-related side effects. In addition, it is important to recognize that patients may suffer long-term consequences of the disease and its treatment, which, although life-saving, is often associated with significant side effects. Our goal, therefore, is to highlight what has been achieved, the pitfalls, and what still needs to be done, comparing the views of physicians and patients.
Collapse
Affiliation(s)
- Luca Quartuccio
- Division of Rheumatology, Department of Medicine, University of Udine, Azienda Sanitaria Universitaria del Friuli Centrale, Udine, Italy
| | - Elena Treppo
- Division of Rheumatology, Department of Medicine, University of Udine, Azienda Sanitaria Universitaria del Friuli Centrale, Udine, Italy
| | - Livio Urso
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Giulia Del Frate
- Division of Rheumatology, Department of Medicine, University of Udine, Azienda Sanitaria Universitaria del Friuli Centrale, Udine, Italy
| | - Federica Mescia
- Nephrology Unit, University of Brescia, Azienda Socio Sanitaria Territoriale Spedali Civili, Brescia, Italy
| | - Federico Alberici
- Nephrology Unit, University of Brescia, Azienda Socio Sanitaria Territoriale Spedali Civili, Brescia, Italy
| | - Augusto Vaglio
- Nephrology and Dialysis Unit, Meyer Children’s Hospital, Florence, Italy
- Department of Biomedical, Experimental and Clinical Sciences, University of Florence, Florence, Italy
| | - Giacomo Emmi
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
- Centre for Inflammatory Diseases, Monash University Department of Medicine, Monash Medical Centre, Clayton, VIC, Australia
| |
Collapse
|
15
|
Blincoe A, Labrosse R, Abraham RS. Acquired B-cell deficiency secondary to B-cell-depleting therapies. J Immunol Methods 2022; 511:113385. [PMID: 36372267 DOI: 10.1016/j.jim.2022.113385] [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: 06/19/2022] [Revised: 09/26/2022] [Accepted: 10/17/2022] [Indexed: 11/13/2022]
Abstract
The advantage of the newer biological therapies is that the immunosuppressive effect is targeted, in contrast, to the standard, traditional immunomodulatory agents, which have a more global effect. However, there are unintended targets and consequences, even to these "precise" therapeutics, leading to acquired or secondary immunodeficiencies. Besides depleting specific cellular immune subsets, these biological agents, which include monoclonal antibodies against biologically relevant molecules, often have broader functional immune consequences, which become apparent over time. This review focuses on acquired B-cell immunodeficiency, secondary to the use of B-cell depleting therapeutic agents. Among the many adverse consequences of B-cell depletion is the risk of hypogammaglobulinemia, failure of B-cell recovery, impaired B-cell differentiation, and risk of infections. Factors, which modulate the outcomes of B-cell depleting therapies, include the intrinsic nature of the underlying disease, the concomitant use of other immunomodulatory agents, and the clinical status of the patient and other co-existing morbidities. This article seeks to explore the mechanism of action of B-cell depleting agents, the clinical utility and adverse effects of these therapies, and the relevance of systematic and serial laboratory immune monitoring in identifying patients at risk for developing immunological complications, and who may benefit from early intervention to mitigate the secondary consequences. Though these biological drugs are gaining widespread use, a harmonized approach to immune evaluation pre-and post-treatment has not yet gained traction across multiple clinical specialties, because of which, the true prevalence of these adverse events cannot be determined in the treated population, and a systematic and evidence-based dosing schedule cannot be developed. The aim of this review is to bring these issues into focus, and initiate a multi-specialty, data-driven approach to immune monitoring.
Collapse
Affiliation(s)
- Annaliesse Blincoe
- Department of Paediatric Immunology and Allergy, Starship Child Health, Auckland, NZ, New Zealand
| | - Roxane Labrosse
- Department of Pediatrics, CHU Sainte-Justine, University of Montreal, Montreal, Canada
| | - Roshini S Abraham
- Department of Pathology and Laboratory Medicine, Nationwide Children's Hospital, Columbus, OH, USA.
| |
Collapse
|
16
|
Pepple S, Arnold J, Vital EM, Rawstron AC, Pease CT, Dass S, Emery P, Md Yusof MY. Predicting Sustained Clinical Response to Rituximab in Moderate to Severe Systemic Manifestations of Primary Sjögren Syndrome. ACR Open Rheumatol 2022; 4:689-699. [PMID: 35666029 PMCID: PMC9374056 DOI: 10.1002/acr2.11466] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 04/22/2022] [Accepted: 05/06/2022] [Indexed: 12/25/2022] Open
Abstract
OBJECTIVE To assess outcomes of repeat rituximab cycles and identify predictors of sustained clinical response in systemic manifestations of primary Sjögren syndrome (pSS). METHODS An observational study was conducted in 40 rituximab-treated patients with pSS. Clinical response was defined as a 3-point or more reduction in the European League Against Rheumatism (EULAR) Sjögren Disease Activity Index (ESSDAI) at 6 months from baseline. Peripheral blood B cells were measured using highly sensitive flow cytometry. Predictors of sustained response (within two rituximab cycles) were analyzed using penalized logistic regression. RESULTS Thirty-eight out of 40 patients had moderate to severe systemic disease (ESSDAI >5). Main domains were articular (73%), mucocutaneous (23%), hematological (20%), and nervous system (18%). Twenty-eight out of 40 (70%) patients were on concomitant immunosuppressants. One hundred sixty-nine rituximab cycles were administered with a total follow-up of 165 patient-years. In cycle 1 (C1), 29/40 (73%) achieved ESSDAI response. Of C1 responders, 23/29 received retreatment on clinical relapse, and 15/23 (65%) responded. Of the 8/23 patients who lost response, these were due to secondary non-depletion and non-response (2NDNR; 4/23 [17%] as we previously observed in systemic lupus erythematosus with antirituximab antibodies, inefficacy = 2/23, and other side effects = 2/23). Within two cycles, 13/40 (33%) discontinued therapy. In multivariable analysis, concomitant immunosuppressant (odds ratio 7.16 [95% confidence interval: 1.37-37.35]) and achieving complete B-cell depletion (9.78 [1.32-72.25]) in C1 increased odds of response to rituximab. At 5 years, 57% of patients continued on rituximab. CONCLUSION Our data suggest that patients with pSS should be co-prescribed immunosuppressant with rituximab, and treatment should aim to achieve complete depletion. About one in six patients develop 2NDNR in repeat cycles. Humanized or type 2 anti-CD20 antibodies may improve clinical response in extra-glandular pSS.
Collapse
Affiliation(s)
- Sophanit Pepple
- Leeds Institute of Rheumatic and Musculoskeletal MedicineUniversity of LeedsLeedsUK
| | - Jack Arnold
- Leeds Institute of Rheumatic and Musculoskeletal MedicineUniversity of LeedsLeedsUK
| | - Edward M. Vital
- Leeds Institute of Rheumatic and Musculoskeletal MedicineUniversity of LeedsLeedsUK
- NIHR Leeds Biomedical Research CentreLeeds Teaching Hospitals NHS TrustLeedsUK
| | - Andrew C. Rawstron
- Haematological Malignancy Diagnostic ServiceLeeds Teaching Hospitals NHS TrustLeedsUK
| | - Colin T. Pease
- Leeds Institute of Rheumatic and Musculoskeletal MedicineUniversity of LeedsLeedsUK
| | - Shouvik Dass
- NIHR Leeds Biomedical Research CentreLeeds Teaching Hospitals NHS TrustLeedsUK
| | - Paul Emery
- Leeds Institute of Rheumatic and Musculoskeletal MedicineUniversity of LeedsLeedsUK
- NIHR Leeds Biomedical Research CentreLeeds Teaching Hospitals NHS TrustLeedsUK
| | - Md Yuzaiful Md Yusof
- Leeds Institute of Rheumatic and Musculoskeletal MedicineUniversity of LeedsLeedsUK
- NIHR Leeds Biomedical Research CentreLeeds Teaching Hospitals NHS TrustLeedsUK
| |
Collapse
|
17
|
Teisseyre M, Cremoni M, Boyer-Suavet S, Ruetsch C, Graça D, Esnault VLM, Brglez V, Seitz-Polski B. Advances in the Management of Primary Membranous Nephropathy and Rituximab-Refractory Membranous Nephropathy. Front Immunol 2022; 13:859419. [PMID: 35603210 PMCID: PMC9114510 DOI: 10.3389/fimmu.2022.859419] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Accepted: 03/22/2022] [Indexed: 02/06/2023] Open
Abstract
Primary membranous nephropathy (pMN) is an auto-immune disease characterized by auto-antibodies targeting podocyte antigens resulting in activation of complement and damage to the glomerular basement membrane. pMN is the most common cause of nephrotic syndrome in adults without diabetes. Despite a very heterogeneous course of the disease, the treatment of pMN has for many years been based on uniform management of all patients regardless of the severity of the disease. The identification of prognostic markers has radically changed the vision of pMN and allowed KDIGO guidelines to evolve in 2021 towards a more personalized management based on the assessment of the risk of progressive loss of kidney function. The recognition of pMN as an antibody-mediated autoimmune disease has rationalized the use immunosuppressive drugs such as rituximab. Rituximab is now a first line immunosuppressive therapy for patients with pMN with proven safety and efficacy achieving remission in 60-80% of patients. For the remaining 20-40% of patients, several mechanisms may explain rituximab resistance: (i) decreased rituximab bioavailability; (ii) immunization against rituximab; and (iii) chronic glomerular damage. The treatment of patients with rituximab-refractory pMN remains controversial and challenging. In this review, we provide an overview of recent advances in the management of pMN (according to the KDIGO 2021 guidelines), in the understanding of the pathophysiology of rituximab resistance, and in the management of rituximab-refractory pMN. We propose a treatment decision aid based on immunomonitoring to identify failures related to underdosing or immunization against rituximab to overcome treatment resistance.
Collapse
Affiliation(s)
- Maxime Teisseyre
- Centre de Référence Maladies Rares Syndrome Néphrotique Idiopathique, CHU de Nice, Université Côte d’Azur, Nice, France
- Unité de Recherche Clinique de la Côte d’Azur (UR2CA), Université Côte d’Azur, Nice, France
| | - Marion Cremoni
- Centre de Référence Maladies Rares Syndrome Néphrotique Idiopathique, CHU de Nice, Université Côte d’Azur, Nice, France
- Unité de Recherche Clinique de la Côte d’Azur (UR2CA), Université Côte d’Azur, Nice, France
| | - Sonia Boyer-Suavet
- Unité de Recherche Clinique de la Côte d’Azur (UR2CA), Université Côte d’Azur, Nice, France
| | - Caroline Ruetsch
- Laboratoire d’Immunologie, CHU de Nice, Université Côte d’Azur, Nice, France
| | - Daisy Graça
- Centre de Référence Maladies Rares Syndrome Néphrotique Idiopathique, CHU de Nice, Université Côte d’Azur, Nice, France
- Unité de Recherche Clinique de la Côte d’Azur (UR2CA), Université Côte d’Azur, Nice, France
| | - Vincent L. M. Esnault
- Centre de Référence Maladies Rares Syndrome Néphrotique Idiopathique, CHU de Nice, Université Côte d’Azur, Nice, France
- Service de Néphrologie-Dialyse-Transplantation, CHU de Nice, Université Côte d’Azur, Nice, France
| | - Vesna Brglez
- Centre de Référence Maladies Rares Syndrome Néphrotique Idiopathique, CHU de Nice, Université Côte d’Azur, Nice, France
- Unité de Recherche Clinique de la Côte d’Azur (UR2CA), Université Côte d’Azur, Nice, France
| | - Barbara Seitz-Polski
- Centre de Référence Maladies Rares Syndrome Néphrotique Idiopathique, CHU de Nice, Université Côte d’Azur, Nice, France
- Unité de Recherche Clinique de la Côte d’Azur (UR2CA), Université Côte d’Azur, Nice, France
- Laboratoire d’Immunologie, CHU de Nice, Université Côte d’Azur, Nice, France
- Service de Néphrologie-Dialyse-Transplantation, CHU de Nice, Université Côte d’Azur, Nice, France
| |
Collapse
|
18
|
Immunogenicity of Rituximab biosimilar GP2013 in chronic inflammatory rheumatic disorders in daily clinical practice. Semin Arthritis Rheum 2022; 52:151951. [PMID: 35038642 DOI: 10.1016/j.semarthrit.2022.151951] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Revised: 12/10/2021] [Accepted: 01/06/2022] [Indexed: 11/22/2022]
Abstract
OBJECTIVE To study in daily practice the risk of immunogenicity of patients treated with the biosimilar rituximab (RTX) GP2013 used for chronic inflammatory rheumatic disorders. METHODS A prospective monocentric routine care study was carried out between September 2018 and May 2021, including consecutive patients treated with the biosimilar RTX GP2013. Biosamples were taken before each infusion to quantify anti-RTX antibodies (ADAbs) and serum RTX trough levels by ELISA (Lisa Tracker Duo Rituximab, LTR005, Theradiag). RESULTS 168 GP2013-treated patients were included (129 who switched from originator RTX and 39 originator RTX naïve). The analysis of 602 samples identified 15 patients (8%) with positive ADAbs including 6 and 9 with transient and persistent ADAbs, respectively. The switch from originator RTX to GP2013 did not increase the risk of immunogenicity, with an incidence rate of 0.8 for 100 patient years. The frequency of persistent ADAs was higher in non-RA patients (5/56, 9% vs. 4/112, 3.5%). Patients with positive persistent ADAbs were more frequently non-caucasian (7/9, 78%, vs. 56/159, 35%, p<0.01) and all had detectable circulating B cells (vs. 40% in ADAb-negative patients, P<0.001). ADAb positivity was not associated with disease activity or RTX discontinuation but patients with ADAb titers >100 ng/mL experienced reduced treatment efficacy or severe infusion-related reaction. CONCLUSION Within the study duration, the immunogenicity of GP2013 is a rare event affecting the pharmacodynamics of RTX. Although development of ADAbs had no impact on treatment discontinuation, possible harmful consequences may be observed in patients with high antibody levels.
Collapse
|
19
|
Faustini F, Dunn N, Kharlamova N, Ryner M, Bruchfeld A, Malmström V, Fogdell-Hahn A, Gunnarsson I. First exposure to rituximab is associated to high rate of anti-drug antibodies in systemic lupus erythematosus but not in ANCA-associated vasculitis. Arthritis Res Ther 2021; 23:211. [PMID: 34389040 PMCID: PMC8361739 DOI: 10.1186/s13075-021-02589-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Accepted: 07/23/2021] [Indexed: 12/30/2022] Open
Abstract
Background Anti-drug antibodies (ADAs) can impact on the efficacy and safety of biologicals, today used to treat several chronic inflammatory conditions. Specific patient groups may be more prone to develop ADAs. Rituximab is routinely used for ANCA-associated vasculitis (AAV) and as off-label therapy for systemic lupus erythematosus (SLE), but data on occurrence and predisposing factors to ADAs in these diseases is limited. Objectives To elucidate the rate of occurrence, and risk factors for ADAs against rituximab in SLE and AAV. Methods ADAs were detected using a bridging electrochemiluminescent (ECL) immunoassay in sera from rituximab-naïve (AAV; n = 41 and SLE; n = 62) and rituximab-treated (AAV; n = 22 and SLE; n = 66) patients. Clinical data was retrieved from medical records. Disease activity was estimated by the SLE Disease Activity Index-2000 (SLEDAI-2 K) and the Birmingham Vasculitis Activity Score (BVAS). Results After first rituximab cycle, no AAV patients were ADA-positive compared to 37.8% of the SLE patients. Samples were obtained at a median (IQR) time of 5.5 (3.7–7.0) months (AAV), and 6.0 (5.0–7.0) months (SLE). ADA-positive SLE individuals were younger (34.0 (25.9–40.8) vs 44.3 (32.7–56.3) years, p = 0.002) and with more active disease (SLEDAI-2 K 14.0 (10.0–18.5) vs. 8.0 (6.0–14), p = 0.0017) and shorter disease duration (4.14 (1.18–10.08) vs 9.19 (5.71–16.93), p = 0.0097) compared to ADA-negative SLE. ADAs primarily occurred in nephritis patients, were associated with anti-dsDNA positivity but were not influenced by concomitant use of corticosteroids, cyclophosphamide or previous treatments. Despite overall reduction of SLEDAI-2 K (12.0 (7.0–16) to 4.0 (2.0–6.7), p < 0.0001), ADA-positive individuals still had higher SLEDAI-2 K (6.0 (4.0–9.0) vs 4.0 (2.0–6.0), p = 0.004) and their B cell count at 6 months follow-up was higher (CD19 + % 4.0 (0.5–10.0) vs 0.5 (0.4–1.0), p = 0.002). At retreatment, two ADA-positive SLE patients developed serum sickness (16.7%), and three had infusion reactions (25%) in contrast with one (5.2%) serum sickness in the ADA-negative group. Conclusions In contrast to AAV, ADAs were highly prevalent among rituximab-treated SLE patients already after the first course of treatment and were found to effect on both clinical and immunological responses. The high frequency in SLE may warrant implementations of ADA screening before retreatment and survey of immediate and late-onset infusion reactions.
Collapse
Affiliation(s)
- Francesca Faustini
- Department of Medicine Solna, Division of Rheumatology, Karolinska Institutet, Stockholm, Sweden. .,Unit of Rheumatology, Karolinska University Hospital, Stockholm, Sweden.
| | - Nicky Dunn
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden.,Center for Molecular Medicine, Stockholm, Sweden
| | - Nastya Kharlamova
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden.,Center for Molecular Medicine, Stockholm, Sweden
| | - Malin Ryner
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden.,Center for Molecular Medicine, Stockholm, Sweden
| | - Annette Bruchfeld
- Department of Health, Medicine and Caring Sciences, Linköping University, Linköping, Sweden.,University Hospital and Department of Renal Medicine, Karolinska University Hospital and CLINTEC Karolinska Institutet, Stockholm, Sweden
| | - Vivianne Malmström
- Department of Medicine Solna, Division of Rheumatology, Karolinska Institutet, Stockholm, Sweden.,Center for Molecular Medicine, Stockholm, Sweden
| | - Anna Fogdell-Hahn
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden.,Center for Molecular Medicine, Stockholm, Sweden
| | - Iva Gunnarsson
- Department of Medicine Solna, Division of Rheumatology, Karolinska Institutet, Stockholm, Sweden.,Unit of Rheumatology, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
20
|
Retamozo S, Sisó-Almirall A, Flores-Chávez A, Ramos-Casals M, Brito-Zerón P. An update of targeted therapeutic options for primary Sjögren syndrome: current status and future development. Expert Opin Pharmacother 2021; 22:2359-2371. [PMID: 34323636 DOI: 10.1080/14656566.2021.1951224] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
Introduction: Primary Sjögren syndrome (pSS) is a systemic autoimmune disease that may affect 3 in 1,000 people within the general population. The therapeutic scenario is complex, and no therapy has proved to be able to modify the natural course of the disease, nor to prevent the most severe systemic complications.Areas covered: Recently, the EULAR 2020 Recommendations for pSS have underlined the low level of evidence supporting efficacious therapeutic approaches, lacking a definition of specific treatment targets and being far from the 'disease modification' concept that is frequently used in other diseases. Herein, the authors review the status of current targeted therapies and provide the reader with their expert opinion.Expert opinion: The progress in discovering novel treatments for pSS seem to be focused on searching new biological therapies as highly-selective drugs that can be effective without the adverse effects related to the wide, nonspecific immunosuppression induced by the drugs currently used. Most likely, the more disruptive therapeutic approach in pSS that could be seen in a few years is the use of combination strategies targeting different etiopathogenic pathways.
Collapse
Affiliation(s)
- Soledad Retamozo
- Sjogren Syndrome Research Group (AGAUR), Department of Autoinmune Diseases, ICMiD, Hospital Clinic, Barcelona, Spain.,Rheumatology Department, Hospital Universitari Parc Taulí, Sabadell, Barcelona, Spain
| | - Antoni Sisó-Almirall
- Primary Care Centre Les Corts, Consorci d'Atenció Primària De Salut Barcelona Esquerra (CAPSBE), Barcelona, Spain.,Primary Healthcare Transversal Research Group, Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), Barcelona, Spain
| | | | - Manuel Ramos-Casals
- Sjogren Syndrome Research Group (AGAUR), Department of Autoinmune Diseases, ICMiD, Hospital Clinic, Barcelona, Spain.,Laboratory of Autoimmune Diseases Josep Font, IDIBAPS-CELLEX, Barcelona, Spain
| | - Pilar Brito-Zerón
- Laboratory of Autoimmune Diseases Josep Font, IDIBAPS-CELLEX, Barcelona, Spain.,Autoimmune Diseases Unit, Department of Medicine, Hospital CIMA-Sanitas, Barcelona, Spain
| |
Collapse
|
21
|
Parikh CR, Ponnampalam JK, Seligmann G, Coelewij L, Pineda-Torra I, Jury EC, Ciurtin C. Impact of immunogenicity on clinical efficacy and toxicity profile of biologic agents used for treatment of inflammatory arthritis in children compared to adults. Ther Adv Musculoskelet Dis 2021; 13:1759720X211002685. [PMID: 34188697 PMCID: PMC8212384 DOI: 10.1177/1759720x211002685] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Accepted: 02/12/2021] [Indexed: 12/15/2022] Open
Abstract
The treatment of inflammatory arthritis has been revolutionised by the
introduction of biologic treatments. Many biologic agents are currently licensed
for use in both paediatric and adult patients with inflammatory arthritis and
contribute to improved disease outcomes compared with the pre-biologic era.
However, immunogenicity to biologic agents, characterised by an immune reaction
leading to the production of anti-drug antibodies (ADAs), can negatively impact
the therapeutic efficacy of biologic drugs and induce side effects to treatment.
This review explores for the first time the impact of immunogenicity against all
licensed biologic treatments currently used in inflammatory arthritis across
age, and will examine any significant differences between ADA prevalence, titres
and timing of development, as well as ADA impact on therapeutic drug levels,
clinical efficacy and side effects between paediatric and adult patients. In
addition, we will investigate factors associated with differences in
immunogenicity across biologic agents used in inflammatory arthritis, and their
potential therapeutic implications.
Collapse
Affiliation(s)
- Chinar R Parikh
- Centre for Adolescent Rheumatology versus Arthritis, University College London, London, UK
| | - Jaya K Ponnampalam
- Centre for Adolescent Rheumatology versus Arthritis, University College London, London, UK
| | - George Seligmann
- Centre for Adolescent Rheumatology versus Arthritis, University College London, London, UK
| | - Leda Coelewij
- Centre for Rheumatology Research, University College London, London, UK
| | - Ines Pineda-Torra
- Centre for Cardiometabolic and Vascular Science, University College London, London, UK
| | - Elizabeth C Jury
- Centre for Rheumatology Research, University College London, London, UK
| | - Coziana Ciurtin
- Centre for Adolescent Rheumatology Versus Arthritis, University College London, 3rd Floor Central, 250 Euston Road, London NW1 2PG, UK
| |
Collapse
|
22
|
Orvain C, Boulch M, Bousso P, Allanore Y, Avouac J. Is there a place for CAR-T cells in the treatment of chronic autoimmune rheumatic diseases? Arthritis Rheumatol 2021; 73:1954-1965. [PMID: 34042325 DOI: 10.1002/art.41812] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2020] [Accepted: 05/11/2021] [Indexed: 11/09/2022]
Abstract
Chimeric-Antigen-Receptor T cell therapy or CAR-T cell is based on a specific targeting of tumor antigen leading to lysis and destruction of tumor cells development. CAR-T cells have demonstrated high potency for the management of B cell malignancies. This successful story was followed by the development of new CAR-T cell-derived constructions that have the ability to eradicate pathogenic B cells or restore tolerance. The objective of the herein manuscript is to review and discuss how the knowledge and technology generated by the use of CAR-T cells may be translated and integrated in the ongoing therapeutic strategies of autoimmune rheumatic diseases. To this end, we will introduce CAR-T cell technology, describe the meaningful achievements of CAR-T cells observed in onco-hematology and discuss preliminary data obtained with CAR-T cells and their derivative constructions in experimental models of autoimmune diseases. Then, we will focus on how CAR-T cell engineering is interfering with the pathogenesis of three chronic autoimmune rheumatic disorders - rheumatoid arthritis, systemic lupus erythematosus and systemic sclerosis - and discuss whether these constructs may permit to gain efficacy compared to current treatments and overcome their adverse events.
Collapse
Affiliation(s)
- Cindy Orvain
- INSERM U1016 and CNRS UMR8104, Institut Cochin, Paris, France
| | - Morgane Boulch
- Dynamics of Immune Responses Unit, Equipe Labellisée Ligue Contre le Cancer, Institut Pasteur, INSERM U1223, 75015, Paris, France
| | - Philippe Bousso
- Dynamics of Immune Responses Unit, Equipe Labellisée Ligue Contre le Cancer, Institut Pasteur, INSERM U1223, 75015, Paris, France
| | - Yannick Allanore
- INSERM U1016 and CNRS UMR8104, Institut Cochin, Paris, France.,Université de Paris, Université Paris Descartes, Paris, France.,Service de Rhumatologie, Hôpital Cochin, AP-HP.CUP, Paris, France
| | - Jérôme Avouac
- INSERM U1016 and CNRS UMR8104, Institut Cochin, Paris, France.,Université de Paris, Université Paris Descartes, Paris, France.,Service de Rhumatologie, Hôpital Cochin, AP-HP.CUP, Paris, France
| |
Collapse
|
23
|
[Rituximab immunomonitoring in autoimmune diseases: a support tool in clinical practice?]. Rev Med Interne 2021; 42:384-391. [PMID: 33678447 DOI: 10.1016/j.revmed.2021.02.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 01/08/2021] [Accepted: 02/06/2021] [Indexed: 11/23/2022]
Abstract
INTRODUCTION Immune monitoring of monoclonal antibodies is a helpful tool in optimizing the management of patients treated with TNF blockers, especially in gastroenterology. In contrast, studies evaluating the interest of such monitoring are lacking for other monoclonal antibodies used in autoimmune diseases, including rituximab despite its widespread use in the field for almost 15 years. Hence, we conducted a study whose goal was to describe the clinical and biological characteristics of all patients who had a rituximab immune monitoring. METHODS All the clinical, biological and therapeutic data attached to the demands (from 2015 onwards) we received for immune monitoring of rituximab (measurements of rituximab serum levels and anti-rituximab antibodies using the drug-sensitive assay LISA-TRACKER Duo Rituximab®), were retrospectively reviewed. Suspected cases of hypersensitivity and secondary non-response were included. RESULTS Several medical specialities (nephrology, haematology, neurology, rheumatology, internal medicine) were represented among the 18 records included in the study (out of 23 demands), 10 being suspected cases of hypersensitivity and 8 secondary non-responders. All 6 patients whose symptoms were consistent with the classical presentation of serum sickness, as well as half of the secondary non-responders, were positive for antirituximab antibodies. CONCLUSION This detailed real world case study illustrates the potential benefits of rituximab immune monitoring (especially anti-rituximab antibodies) in autoimmune diseases, suggesting it could be helpful in suspected cases of serum sickness, as well as secondary non-response (B-cell non-depletion being an early red flag). Larger and disease-specific studies are warranted to support these findings.
Collapse
|
24
|
Teisseyre M, Boyer-Suavet S, Crémoni M, Brglez V, Esnault V, Seitz-Polski B. Analysis and Management of Rituximab Resistance in PLA2R1-Associated Membranous Nephropathy. Kidney Int Rep 2021; 6:1183-1188. [PMID: 33912768 PMCID: PMC8071636 DOI: 10.1016/j.ekir.2021.01.022] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Revised: 01/12/2021] [Accepted: 01/18/2021] [Indexed: 12/19/2022] Open
Affiliation(s)
- Maxime Teisseyre
- Laboratoire d'Immunologie, CHU de Nice, Université Côte d'Azur, France.,Centre de Référence Maladies Rares Syndrome Néphrotique Idiopathique, CHU de Nice, Université Côte d'Azur, France
| | - Sonia Boyer-Suavet
- Centre de Référence Maladies Rares Syndrome Néphrotique Idiopathique, CHU de Nice, Université Côte d'Azur, France.,Service de Néphrologie-Dialyse-Transplantation, CHU de Nice, Université Côte d'Azur, France.,Unité de Recherche Clinique de la Côte d'Azur (UR2CA), Université Côte d'Azur, France
| | - Marion Crémoni
- Service de Néphrologie-Dialyse-Transplantation, CHU de Nice, Université Côte d'Azur, France
| | - Vesna Brglez
- Centre de Référence Maladies Rares Syndrome Néphrotique Idiopathique, CHU de Nice, Université Côte d'Azur, France.,Unité de Recherche Clinique de la Côte d'Azur (UR2CA), Université Côte d'Azur, France
| | - Vincent Esnault
- Service de Néphrologie-Dialyse-Transplantation, CHU de Nice, Université Côte d'Azur, France
| | - Barbara Seitz-Polski
- Laboratoire d'Immunologie, CHU de Nice, Université Côte d'Azur, France.,Centre de Référence Maladies Rares Syndrome Néphrotique Idiopathique, CHU de Nice, Université Côte d'Azur, France.,Service de Néphrologie-Dialyse-Transplantation, CHU de Nice, Université Côte d'Azur, France.,Unité de Recherche Clinique de la Côte d'Azur (UR2CA), Université Côte d'Azur, France
| |
Collapse
|
25
|
Abstract
It is now two decades since Rituximab was first used in the treatment of patients with systemic lupus erythematosus. There have been many challenges but in spite of failing to meet its primary endpoints in two clinical trials it is widely used for many aspects of lupus, its side-effects and the possibility that combining it with Benlysta may be of value. We also consider the proposal that it may provide a useful initial therapy. In this review, we consider the place of Rituximab in the treatment of lupus and anticipate how developments in fully-humanized anti-CD20 monoclonals may well extend the "therapeutic life" of B-cell depletion as a viable treatment option.
Collapse
Affiliation(s)
- Maria Leandro
- Centre for Rheumatology, Division of Medicine, University College London, London, UK.,Department of Rheumatology, University College London Hospitals NHS Foundation Trust, London, UK
| | - David A Isenberg
- Centre for Rheumatology, Division of Medicine, University College London, London, UK.,Department of Rheumatology, University College London Hospitals NHS Foundation Trust, London, UK
| |
Collapse
|
26
|
Wang X, Du W, Zhang X, Li P. The Influence of Different Disease States on Rituximab Pharmacokinetics. Curr Drug Metab 2020; 21:938-946. [PMID: 32682367 DOI: 10.2174/1389200221666200719004035] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 05/04/2020] [Accepted: 06/02/2020] [Indexed: 02/06/2023]
Abstract
BACKGROUND The anti-CD20 antibody rituximab, which promotes the selective depletion of CD20 positive B cells, was the first targeted therapy that was approved for the treatment of B-cell malignancies, and it is now widely prescribed in both malignant and non-malignant, immune-related diseases. However, the cause of its various clinical responses in certain diseases, have not been clearly elucidated. The variabilities in inter-individual pharmacokinetic and the emerging evidence of the relationships between pharmacokinetic and pharmacodynamic may provide a better understanding of this drug. METHODS We searched and summarized the latest published articles on rituximab pharmacokinetic profiles and the pharmacokinetic/pharmacodynamic models in different patient populations, including B-cell malignancies, rheumatoid arthritis, ANCA-associated vasculitis, and glomerular kidney diseases. RESULTS Most pharmacokinetic data are drawn from clinical studies in oncology clinical practice. Body weight, gender, and antigen-related factors are proven to be the key factors affecting rituximab pharmacokinetics. In addition, the positive exposure-response relations were reported, which provide encouraging evidence for individualized therapies. While in immune disorders, especially in the off-labeled indications, pharmacokinetic studies are quite limited. Compared with that in B-cell malignancies, the differences in the pharmacokinetic parameters may be attributed to the different pathogeneses of diseases, mechanisms of action and dosing strategies. However, the correlation between drug exposure and clinical outcomes remains unclear. CONCLUSION Here, we provide an overview of the complexities associated with rituximab pharmacokinetics and pharmacodynamics in different diseases. Although many influencing factors need to be verified in future studies, a better understanding of the relationships between pharmacokinetic and pharmacodynamic may assist in optimizing rituximab clinical practice.
Collapse
Affiliation(s)
- Xiaoxing Wang
- Department of Pharmacy, China-Japan Friendship Hospital, Beijing 100029, China
| | - Wenwen Du
- Department of Pharmacy, China-Japan Friendship Hospital, Beijing 100029, China
| | - Xianglin Zhang
- Department of Pharmacy, China-Japan Friendship Hospital, Beijing 100029, China
| | - Pengmei Li
- Department of Pharmacy, China-Japan Friendship Hospital, Beijing 100029, China
| |
Collapse
|
27
|
Boyer-Suavet S, Andreani M, Lateb M, Savenkoff B, Brglez V, Benzaken S, Bernard G, Nachman PH, Esnault V, Seitz-Polski B. Neutralizing Anti-Rituximab Antibodies and Relapse in Membranous Nephropathy Treated With Rituximab. Front Immunol 2020; 10:3069. [PMID: 31998325 PMCID: PMC6970431 DOI: 10.3389/fimmu.2019.03069] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Accepted: 12/16/2019] [Indexed: 02/06/2023] Open
Abstract
Membranous Nephropathy (MN) is an autoimmune disease associated with antibodies against podocyte proteins: M-type phospholipase A2 receptor (PLA2R1) or thrombospondin type-1 domain-containing 7A (THSD7A) in 70 and 3% of patients, respectively. Antibody titer is correlated with disease activity: rising during active disease and decreasing before remission. Therefore, decreasing PLA2R1-Antibodies titer has become an important goal of therapy. Rituximab a chimeric monoclonal antibody induces remission in 60-80% of primary MN patients. All monoclonal antibodies such as rituximab can elicit antidrug antibodies, which may interfere with therapeutic response. We aim to analyze the relevance of anti-rituximab antibodies on the outcome of MN after a first course of rituximab. Forty-four MN patients were included and treated with two 1 g infusions of rituximab at 2-weeks interval. Anti-rituximab antibodies, CD19 count, and clinical response were analyzed. Then, we (i) analyzed the association of anti-rituximab antibodies at month-6 with response to treatment: remission, relapse and the need for another rituximab course; (ii) confirmed if anti-rituximab antibodies could neutralize rituximab B-cells depletion; and (iii) tested whether anti-rituximab antibodies could cross-inhibit new humanized anti-CD20 therapies. Anti-rituximab antibodies were detected in 10 patients (23%). Seventeen patients received a second rituximab course after a median time of 12 months (7-12), following nine cases of resistance and eight relapses. Anti-rituximab antibodies were significantly associated with faster B-cell reconstitution at month-6 (75 [57-89] vs. 2 [0-41] cells/μl, p = 0.006), higher proteinuria 12 months after rituximab infusion (1.7 [0.7; 5.8] vs. 0.6 [0.2; 3.4], p = 0.03) and before treatment modification (3.5 [1.6; 7.1] vs. 1.7 [0.2; 1.7] p = 0.0004). Remission rate 6 months after rituximab was not different according to anti-rituximab status (p > 0.99) but the rate of relapse was significantly higher for patients with anti-rituximab antibodies (p < 0.001). These patients required more frequently a second course of rituximab infusions (7/10 vs. 10/34, p = 0.03). Anti-rituximab antibodies neutralized rituximab activity in 8/10 patients and cross-reacted with other humanized monoclonal antibodies in only two patients. Three patients with anti-rituximab antibodies were successfully treated with ofatumumab. Anti-rituximab antibodies could neutralize rituximab B cells cytotoxicity and impact clinical outcome of MN patients. Humanized anti-CD20 seems to be a satisfying therapeutic alternative for patients with anti-rituximab antibodies and resistant or relapsing MN.
Collapse
Affiliation(s)
- Sonia Boyer-Suavet
- Service de Néphrologie-Dialyse-Transplantation, CHU de Nice, Université Côte d'Azur, Nice, France.,CRMR SNI, CHU de Nice, Université Côte d'Azur, Nice, France
| | - Marine Andreani
- Service de Néphrologie-Dialyse-Transplantation, CHU de Nice, Université Côte d'Azur, Nice, France
| | - Maël Lateb
- Service de Néphrologie, Dialyse et Aphérèse Thérapeutique, CHR Metz-Thionville, Thionville, France
| | - Benjamin Savenkoff
- Service de Néphrologie, Dialyse et Aphérèse Thérapeutique, CHR Metz-Thionville, Thionville, France
| | - Vesna Brglez
- CRMR SNI, CHU de Nice, Université Côte d'Azur, Nice, France
| | - Sylvia Benzaken
- Laboratoire d'Immunologie, CHU de Nice, Université Côte d'Azur, Nice, France
| | - Ghislaine Bernard
- Laboratoire d'Immunologie, CHU de Nice, Université Côte d'Azur, Nice, France
| | - Patrick H Nachman
- Division of Renal Diseases and Hypertension, University of Minnesota, Minneapolis, MN, United States
| | - Vincent Esnault
- Service de Néphrologie-Dialyse-Transplantation, CHU de Nice, Université Côte d'Azur, Nice, France.,CRMR SNI, CHU de Nice, Université Côte d'Azur, Nice, France
| | - Barbara Seitz-Polski
- Service de Néphrologie-Dialyse-Transplantation, CHU de Nice, Université Côte d'Azur, Nice, France.,CRMR SNI, CHU de Nice, Université Côte d'Azur, Nice, France.,Laboratoire d'Immunologie, CHU de Nice, Université Côte d'Azur, Nice, France
| |
Collapse
|