1
|
Piroth L, Moretto F, Sixt T, Blot M. [Viral complications of biotherapies/targeted anti-inflammatory therapies]. Rev Med Interne 2025; 46:146-154. [PMID: 39947978 DOI: 10.1016/j.revmed.2024.12.007] [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: 03/27/2024] [Revised: 11/22/2024] [Accepted: 12/12/2024] [Indexed: 05/09/2025]
Abstract
By the end of the nineties, new immunomodulatory options impacting on the determinants of many immune-mediated diseases became available. These drugs were also called biologicals. Their use was associated with a significant improvement in the management of the patients and on their clinical evolution over time. On the other hand, their use was found to be also associated with an over-risk of infectious complications, in particular of viral origin, even though the savings of other at-risk treatments (e.g. corticosteroids or cyclophosphamide) allowed by these new therapies could have contributed to reduce it. These viral infections may be linked to an increased susceptibility to new infections because of impaired immunity and/or lower responsiveness to vaccination, to a higher risk of reactivation of latent infections, and to a higher severity than observed in the general population. Viruses mostly involved are respiratory (influenza, RSV, and SARS-CoV2), Varicella-Zoster, hepatitis B, or JC viruses, in particular. The viral risk depends not only on the type of biologicals, but also on the underlying disease, the associated comorbidities, the associated treatments, the epidemiological environment, and the individual and collective immunity. At an individual level, prevention and management of the infectious risk are of utmost importance in the global management of patients on biologicals.
Collapse
Affiliation(s)
- Lionel Piroth
- Département d'infectiologie, CHU de Dijon, 14, rue Gaffarel, 21079 Dijon, France; Inserm CIC 1432, université de Bourgogne, Bourgogne, France.
| | - Florian Moretto
- Département d'infectiologie, CHU de Dijon, 14, rue Gaffarel, 21079 Dijon, France
| | - Thibaut Sixt
- Département d'infectiologie, CHU de Dijon, 14, rue Gaffarel, 21079 Dijon, France
| | - Mathieu Blot
- Département d'infectiologie, CHU de Dijon, 14, rue Gaffarel, 21079 Dijon, France; Inserm CIC 1432, université de Bourgogne, Bourgogne, France
| |
Collapse
|
2
|
Picchianti-Diamanti A, Aiello A, De Lorenzo C, Migliori GB, Goletti D. Management of tuberculosis risk, screening and preventive therapy in patients with chronic autoimmune arthritis undergoing biotechnological and targeted immunosuppressive agents. Front Immunol 2025; 16:1494283. [PMID: 39963138 PMCID: PMC11830708 DOI: 10.3389/fimmu.2025.1494283] [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: 09/10/2024] [Accepted: 01/06/2025] [Indexed: 02/20/2025] Open
Abstract
Tuberculosis (TB) is the leading cause of death in the world from an infectious disease. Its etiologic agent, the Mycobacterium tuberculosis (Mtb), is a slow-growing bacterium that has coexisted in humans for thousands of years. According to the World Health Organization, 10.6 million new cases of TB and over 1 million deaths were reported in 2022. It is widely recognized that patients affected by chronic autoimmune arthritis such as rheumatoid arthritis (RA), psoriatic arthritis (PsA), and ankylosing spondylitis (AS) have an increased incidence rate of TB disease compared to the general population. As conceivable, the risk is associated with age ≥65 years and is higher in endemic regions, but immunosuppressive therapy plays a pivotal role. Several systematic reviews have analysed the impact of anti-TNF-α agents on the risk of TB in patients with chronic autoimmune arthritis, as well as for other biologic disease-modifying immunosuppressive anti-rheumatic drugs (bDMARDs) such as rituximab, abatacept, tocilizumab, ustekinumab, and secukinumab. However, the data are less robust compared to those available with TNF-α inhibitors. Conversely, data on anti-IL23 agents and JAK inhibitors (JAK-i), which have been more recently introduced for the treatment of RA and PsA/AS, are limited. TB screening and preventive therapy are recommended in Mtb-infected patients undergoing bDMARDs and targeted synthetic (ts)DMARDs. In this review, we evaluate the current evidence from randomized clinical trials, long-term extension studies, and real-life studies regarding the risk of TB in patients with RA, PsA, and AS treated with bDMARDs and tsDMARDs. According to the current evidence, TNF-α inhibitors carry the greatest risk of TB progression among bDMARDs and tsDMARDs, such as JAK inhibitors and anti-IL-6R agents. The management of TB screening and the updated preventive therapy are reported.
Collapse
Affiliation(s)
- Andrea Picchianti-Diamanti
- Department of Clinical and Molecular Medicine, “Sapienza” University, S. Andrea University Hospital, Rome, Italy
| | - Alessandra Aiello
- Translational Research Unit, National Institute for Infectious Diseases “Lazzaro Spallanzani”- Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Rome, Italy
| | - Chiara De Lorenzo
- Department of Clinical and Molecular Medicine, “Sapienza” University, S. Andrea University Hospital, Rome, Italy
| | - Giovanni Battista Migliori
- Istituti Clinici Scientifici Maugeri, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Tradate, Italy
| | - Delia Goletti
- Translational Research Unit, National Institute for Infectious Diseases “Lazzaro Spallanzani”- Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Rome, Italy
| |
Collapse
|
3
|
Kacar M, Al-Hakim A, Savic S. Sequelae of B-Cell Depleting Therapy: An Immunologist's Perspective. BioDrugs 2025; 39:103-130. [PMID: 39680306 DOI: 10.1007/s40259-024-00696-9] [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: 11/27/2024] [Indexed: 12/17/2024]
Abstract
B-cell depleting therapy (BCDT) has revolutionised the treatment of B-cell malignancies and autoimmune diseases by targeting specific B-cell surface antigens, receptors, ligands, and signalling pathways. This narrative review explores the mechanisms, applications, and complications of BCDT, focusing on the therapeutic advancements since the introduction of rituximab in 1997. Various monoclonal antibodies and kinase inhibitors are examined for their roles in depleting B cells through antibody-dependent and independent mechanisms. The off-target effects, such as hypogammaglobulinemia, infections, and cytokine release syndrome, are discussed, emphasising the need for immunologists to identify and help manage these complications. The increasing prevalence of BCDT has necessitated the involvement of clinical immunologists in addressing treatment-associated immunological abnormalities, including persistent hypogammaglobulinemia and neutropenia. We highlight the importance of considering underlying inborn errors of immunity (IEI) in patients presenting with these complications. Furthermore, we discuss the impact of BCDT on other immune cell populations and the challenges in predicting and managing long-term immunological sequelae. The potential for novel BCDT agents targeting the BAFF/APRIL-TACI/BCMA axis and B-cell receptor signalling pathways to treat autoimmune disorders is also explored, underscoring the rapidly evolving landscape of B-cell targeted therapies.
Collapse
Affiliation(s)
- Mark Kacar
- Department of Allergy, University Clinic Golnik, Golnik, Slovenia
- Department of Allergy and Clinical Immunology, St James' University Hospital, Leeds, UK
- Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Adam Al-Hakim
- Department of Allergy and Clinical Immunology, St James' University Hospital, Leeds, UK
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Leeds, UK
| | - Sinisa Savic
- Department of Allergy and Clinical Immunology, St James' University Hospital, Leeds, UK.
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Leeds, UK.
- NIHR Leeds Biomedical Research Centre, Leeds, UK.
| |
Collapse
|
4
|
Pecher AC, Hensen L, Lengerke C, Henes J. The Future of CAR T Therapeutics to Treat Autoimmune Disorders. Mol Diagn Ther 2024; 28:593-600. [PMID: 39078456 PMCID: PMC11349844 DOI: 10.1007/s40291-024-00730-0] [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] [Accepted: 07/08/2024] [Indexed: 07/31/2024]
Abstract
The concept of chimeric antigen receptor (CAR) T cell therapy emerged from cancer immunotherapy and has been rapidly adapted and developed for the treatment of autoimmune, especially B-cell-driven, diseases since the first publication of an article featuring a patient with systemic lupus erythematosus in 2021. Phase II studies are about to start, but up to now, only case reports and small series have been published. In contrast to hemato-oncological diseases, where an aggressive response to malignant cells and long-lasting persistence of CAR T cells has been aimed at and observed in many patients, this is not the case with autoimmune diseases but might not be necessary to control disease. Future studies will focus on the optimal target but also on the optimal level of immunogenicity. The latter can be influenced by numerous modulations that affect not only cytokine release but also regulation. In addition, there are potential applications in regulatory cells such as CAR regulatory T cells (Treg). The question of toxicity reduction must also be addressed, as long-term complications such as the potential development of malignant diseases, infections, or cytopenia must be considered even more critically in the area of autoimmune diseases than is the case for patients with oncologic diseases. Alternative antibody-based therapies using the same target (e.g., CD3/CD19 bispecific targeting antibodies) have not been used in these patients and might also be considered in the future. In conclusion, CAR T cell therapy represents a promising therapeutic approach for autoimmune diseases, offering a targeted strategy to modulate immune responses and restore immune tolerance.
Collapse
Affiliation(s)
- Ann-Christin Pecher
- Department of Internal Medicine II, Hematology, Oncology, Clinical Immunology, and Rheumatology, University Hospital Tübingen, Otfried-Mueller-Strasse 10, 72076, Tübingen, Germany.
| | - Luca Hensen
- Department of Internal Medicine II, Hematology, Oncology, Clinical Immunology, and Rheumatology, University Hospital Tübingen, Otfried-Mueller-Strasse 10, 72076, Tübingen, Germany
| | - Claudia Lengerke
- Department of Internal Medicine II, Hematology, Oncology, Clinical Immunology, and Rheumatology, University Hospital Tübingen, Otfried-Mueller-Strasse 10, 72076, Tübingen, Germany
| | - Jörg Henes
- Department of Internal Medicine II, Hematology, Oncology, Clinical Immunology, and Rheumatology, University Hospital Tübingen, Otfried-Mueller-Strasse 10, 72076, Tübingen, Germany
| |
Collapse
|
5
|
Ohno R, Nakamura A. Advancing autoimmune Rheumatic disease treatment: CAR-T Cell Therapies - Evidence, Safety, and future directions. Semin Arthritis Rheum 2024; 67:152479. [PMID: 38810569 DOI: 10.1016/j.semarthrit.2024.152479] [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: 03/09/2024] [Revised: 04/20/2024] [Accepted: 05/08/2024] [Indexed: 05/31/2024]
Abstract
INTRODUCTION Despite advancements in managing autoimmune rheumatic diseases (ARDs) with existing treatments, many patients still encounter challenges such as inadequate responses, difficulty in maintaining remission, and side effects. Chimeric Antigen Receptor (CAR) T-cell therapy, originally developed for cancer, has now emerged as a promising option for cases of refractory ARDs. METHODS A search of the literature was conducted to compose a narrative review exploring the current evidence, potential safety, limitations, potential modifications, and future directions of CAR-T cells in ARDs. RESULTS CAR-T cell therapy has been administered to patients with refractory ARDs, including systemic lupus erythematosus, antisynthetase syndrome, and systemic sclerosis, demonstrating significant improvement. Notable responses include enhanced clinical symptoms, reduced serum autoantibody titers, and sustained remissions in disease activity. Preclinical and in vitro studies using both animal and human samples also support the efficacy and elaborate on potential mechanisms of CAR-T cells against antineutrophil cytoplasmic antibody-associated vasculitis and rheumatoid arthritis. While cautious monitoring of adverse events, such as cytokine release syndrome, is crucial, the therapy appears to be highly tolerable. Nevertheless, challenges persist, including cost, durability due to potential CAR-T cell exhaustion, and manufacturing complexities, urging the development of innovative solutions to further enhance CAR-T cell therapy accessibility in ARDs. CONCLUSIONS CAR-T cell therapy for refractory ARDs has demonstrated high effectiveness. While no significant warning signs are currently reported, achieving a balance between therapeutic efficacy and safety is vital in adapting CAR-T cell therapy for ARDs. Moreover, there is significant potential for technological advancements to enhance the delivery of this treatment to patients, thereby ensuring safer and more effective disease control for patients.
Collapse
Affiliation(s)
- Ryunosuke Ohno
- Department of Medicine, Division of Rheumatology, Queen's University, Kingston, Ontario, Canada; Department of Medicine, Okayama University, Okayama, Japan
| | - Akihiro Nakamura
- Department of Medicine, Division of Rheumatology, Queen's University, Kingston, Ontario, Canada; Translational Institute of Medicine, School of Medicine, Queen's University, Ontario, Canada; Rheumatology Clinic, Kingston Health Science Centre, Kingston, Ontario, Canada.
| |
Collapse
|
6
|
Nguyen Y, Mariette X, Gottenberg JE, Iudici M, Morel J, Vittecoq O, Constantin A, Flipo RM, Schaeverbeke T, Sibilia J, Ravaud P, Porcher R, Seror R. Can efficacy and safety data from clinical trials of rituximab in RA be extrapolated? Insights from 1984 patients from the AIR-PR Registry. Rheumatology (Oxford) 2024; 63:1893-1901. [PMID: 37725356 DOI: 10.1093/rheumatology/kead495] [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: 06/26/2023] [Revised: 08/24/2023] [Accepted: 08/31/2023] [Indexed: 09/21/2023] Open
Abstract
OBJECTIVES To investigate whether the efficacy and safety data from drug-registration trials can be extrapolated to real-life RA patients receiving RTX. METHODS The 'AutoImmunity and Rituximab' (AIR-PR) registry is a French multicentre, prospective cohort of RA patients treated with RTX in a real-life setting. We compared treatment responses at 12 months and serious adverse events (AEs) between eligible and non-eligible patients, by retrieving the eligibility criteria of the three rituximab-registration trials. We determined critical eligibility criteria and modelled the benefit-risk ratio according to the number of fulfilled critical eligibility criteria. RESULTS Among 1984 RA patients, only 9-12% fulfilled all eligibility criteria. Non-eligible patients had fewer EULAR responses at 12 months (40.3% vs 46.9%, P = 0.044). Critical inclusion criteria included swollen joints count ≥4, tender joints count ≥4, CRP ≥15 mg/l and RF positivity. Critical exclusion criteria were age >80 years, RA-associated systemic diseases, ACR functional class IV, DMARD other than MTX and prednisone >10 mg/day. Only 20.8% fulfilled those critical eligibility criteria. During the first year, serious AEs occurred for 182 (9.2%) patients (70.3% serious infections) and patients with ≥1 critical exclusion criterion were at higher risk (hazard ratio 3.03; 95% CI 2.25-4.06; for ≥3 criteria vs 0). The incremental risk-benefit ratio decreased with the number of unmet critical inclusion criteria and of fulfilled exclusion criteria. CONCLUSION Few real-life RA patients were eligible for the drug-registration trials. Non-eligible patients had lower chance of response, and higher risk of serious AEs. Efficacy and safety data obtained from those trials may not be generalizable to RA patients receiving RTX in real-world clinical practice.
Collapse
Affiliation(s)
- Yann Nguyen
- Department of Rheumatology, Hôpital Bicêtre Assistance Publique-Hôpitaux de Paris, Le Kremlin-Bicêtre, France
- Center for Immunology of Viral Infections and Auto-immune Diseases (IMVA), Université Paris-Saclay, Institut pour la Santé et la Recherche Médicale (INSERM) UMR 1184, Le Kremlin Bicêtre, France
| | - Xavier Mariette
- Department of Rheumatology, Hôpital Bicêtre Assistance Publique-Hôpitaux de Paris, Le Kremlin-Bicêtre, France
- Center for Immunology of Viral Infections and Auto-immune Diseases (IMVA), Université Paris-Saclay, Institut pour la Santé et la Recherche Médicale (INSERM) UMR 1184, Le Kremlin Bicêtre, France
| | - Jacques E Gottenberg
- Rheumatology Department, Hôpitaux Universitaires de Strasbourg, Université de Strasbourg, Strasbourg, France
| | - Michele Iudici
- INSERM, UMR1153 Epidemiology and Statistics Sorbonne Paris Cité Research Centre (CRESS), Team METHODS, Paris, France
- Division of Rheumatology, Department of Internal Medicine Specialties, Geneva University Hospitals, Geneva, Switzerland
| | - Jacques Morel
- Rheumatology Department, CHU and University of Montpellier, PhyMedExp, Université de Montpellier, INSERM, CNRS, Montpellier, France
| | - Olivier Vittecoq
- Department of Rheumatology, Rouen University Hospital & Inserm U905, Rouen, France
| | - Arnaud Constantin
- Rheumatology Department, Purpan University Hospital, Paul Sabatier Toulouse III University, INSERM U1291 Infinity, Toulouse, France
| | - Rene-Marc Flipo
- Rheumatology Department, CHRU de Lille, Université de Lille, Lille, France
| | | | - Jean Sibilia
- Rheumatology Department, Hôpitaux Universitaires de Strasbourg, Université de Strasbourg, Strasbourg, France
| | - Philippe Ravaud
- INSERM, UMR1153 Epidemiology and Statistics Sorbonne Paris Cité Research Centre (CRESS), Team METHODS, Paris, France
- Assistance Publique des Hôpitaux de Paris (AP-HP), Hôpital Hôtel-Dieu, Center for Clinical Epidemiology, Paris, France
| | - Raphaël Porcher
- INSERM, UMR1153 Epidemiology and Statistics Sorbonne Paris Cité Research Centre (CRESS), Team METHODS, Paris, France
- Assistance Publique des Hôpitaux de Paris (AP-HP), Hôpital Hôtel-Dieu, Center for Clinical Epidemiology, Paris, France
| | - Raphaèle Seror
- Department of Rheumatology, Hôpital Bicêtre Assistance Publique-Hôpitaux de Paris, Le Kremlin-Bicêtre, France
- Center for Immunology of Viral Infections and Auto-immune Diseases (IMVA), Université Paris-Saclay, Institut pour la Santé et la Recherche Médicale (INSERM) UMR 1184, Le Kremlin Bicêtre, France
| |
Collapse
|
7
|
Hlavackova E, Krenova Z, Kerekes A, Slanina P, Vlkova M. B cell subsets reconstitution and immunoglobulin levels in children and adolescents with B non-Hodgkin lymphoma after treatment with single anti CD20 agent dose included in chemotherapeutic protocols: single center experience and review of the literature. Biomed Pap Med Fac Univ Palacky Olomouc Czech Repub 2024; 168:167-176. [PMID: 37227099 DOI: 10.5507/bp.2023.021] [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: 09/30/2022] [Accepted: 05/10/2023] [Indexed: 05/26/2023] Open
Abstract
BACKGROUND RTX, an anti-CD20 monoclonal antibody, added to chemotherapy has proven to be effective in children and adolescents with high-grade, high-risk and matured non-Hodgkin lymphoma. RTX leads to prompt CD19+ B lymphocyte depletion. However, despite preserved immunoglobulin production by long-lived plasmablasts after treatment, patients remain at risk of prolonged hypogammaglobulinemia. Further, there are few general guidelines for immunology laboratories and clinical feature monitoring after B cell-targeted therapies. The aim of this paper is to describe B cell reconstitution and immunoglobulin levels after pediatric B-NHL protocols, that included a single RTX dose and to review the literature. METHODS A retrospective single-center study on the impact of a single RTX dose included in a chemotherapeutic pediatric B Non-Hodgkin Lymphoma (B-NHL) treatment protocols. Immunology laboratory and clinical features were evaluated over an eight hundred days follow-up (FU) period, after completing B-NHL treatment. RESULTS Nineteen patients (fifteen Burkitt lymphoma, three Diffuse large B cell lymphoma, and one Marginal zone B cell lymphoma) fulfilled the inclusion criteria. Initiation of B cell subset reconstitution occurred a median of three months after B-NHL treatment. Naïve and transitional B cells declined over the FU in contrast to the marginal zone and the switched memory B cell increase. The percentage of patients with IgG, IgA, and IgM hypogammaglobulinemia declined consistently over the FU. Prolonged IgG hypogammaglobulinemia was detectable in 9%, IgM in 13%, and IgA in 25%. All revaccinated patients responded to protein-based vaccines by specific IgG antibody production increase. Following antibiotic prophylaxes, none of the patients with hypogammaglobulinemia manifested with either a severe or opportunistic infection course. CONCLUSION The addition of a single RTX dose to the chemotherapeutic treatment protocols was not shown to increase the risk of developing secondary antibody deficiency in B-NHL pediatric patients. Observed prolonged hypogammaglobulinemia remained clinically silent. However interdisciplinary agreement on regular long-term immunology FU after anti-CD20 agent treatment is required.
Collapse
Affiliation(s)
- Eva Hlavackova
- Department of Clinical Immunology and Allergology, St. Anne's University Hospital in Brno and Faculty of Medicine, Masaryk University, Brno, Czech Republic
- Department of Pediatric Oncology, University Hospital Brno and Faculty of Medicine, Masaryk University Brno, Czech Republic
| | - Zdenka Krenova
- Department of Pediatric Oncology, University Hospital Brno and Faculty of Medicine, Masaryk University Brno, Czech Republic
| | - Arpad Kerekes
- Department of Pediatric Oncology, University Hospital Brno and Faculty of Medicine, Masaryk University Brno, Czech Republic
| | - Peter Slanina
- Department of Clinical Immunology and Allergology, St. Anne's University Hospital in Brno and Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Marcela Vlkova
- Department of Clinical Immunology and Allergology, St. Anne's University Hospital in Brno and Faculty of Medicine, Masaryk University, Brno, Czech Republic
| |
Collapse
|
8
|
Rindi LV, Zaçe D, Braccialarghe N, Massa B, Barchi V, Iannazzo R, Fato I, De Maria F, Kontogiannis D, Malagnino V, Sarmati L, Iannetta M. Drug-Induced Progressive Multifocal Leukoencephalopathy (PML): A Systematic Review and Meta-Analysis. Drug Saf 2024; 47:333-354. [PMID: 38321317 DOI: 10.1007/s40264-023-01383-4] [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: 11/15/2023] [Indexed: 02/08/2024]
Abstract
INTRODUCTION Progressive multifocal leukoencephalopathy (PML) was first described among patients affected by hematological or solid tumors. Following the human immunodeficiency virus (HIV) epidemic, people living with HIV have represented most cases for more than a decade. With the diffusion of highly active antiretroviral therapy, this group progressively decreased in favor of patients undergoing treatment with targeted therapy/immunomodulators. In this systematic review and meta-analysis, the objective was to assess which drugs are most frequently related to PML development, and report the incidence of drug-induced PML through a meta-analytic approach. METHODS The electronic databases MEDLINE, EMBASE, ClinicalTrials.gov, Web of Science and the Canadian Agency for Drugs and Technologies in Health Database (CADTH) were searched up to May 10, 2022. Articles that reported the risk of PML development after treatment with immunomodulatory drugs, including patients of both sexes under the age of 80 years, affected by any pathology except HIV, primary immunodeficiencies or malignancies, were included in the review. The incidence of drug-induced PML was calculated based on PML cases and total number of patients observed per 100 persons and the observation time. Random-effect metanalyses were conducted for each drug reporting pooled incidence with 95% confidence intervals (CI) and median (interquartile range [IQR]) of the observation time. Heterogeneity was measured by I2 statistics. Publication bias was examined through funnel plots and Egger's test. RESULTS A total of 103 studies were included in the systematic review. In our analysis, we found no includible study reporting cases of PML during the course of treatment with ocrelizumab, vedolizumab, abrilumab, ontamalimab, teriflunomide, daclizumab, inebilizumab, basiliximab, tacrolimus, belimumab, infliximab, firategrast, disulone, azathioprine or danazole. Dalfampridine, glatiramer acetate, dimethyl fumarate and fingolimod show a relatively safe profile, although some cases of PML have been reported. The meta-analysis showed an incidence of PML cases among patients undergoing rituximab treatment for multiple sclerosis (MS) of 0.01 cases/100 persons (95% CI - 0.08 to 0.09; I2 = 20.4%; p = 0.25) for a median observation period of 23.5 months (IQR 22.1-42.1). Treatment of MS with natalizumab carried a PML risk of 0.33 cases/100 persons (95% CI 0.29-0.37; I2 = 50%; p = 0.003) for a median observation period of 44.1 months (IQR 28.4-60) and a mean number of doses of 36.3 (standard deviation [SD] ± 20.7). When comparing data about patients treated with standard interval dosing (SID) and extended interval dosing (EID), the latter appears to carry a smaller risk of PML, that is, 0.08 cases/100 persons (95% CI 0.0-0.15) for EID versus 0.3 cases/100 persons (95% CI 0.25-0.34) for SID. CONCLUSIONS A higher risk of drug-related PML in patients whose immune system is not additionally depressed by means of neoplasms, HIV or concomitant medications is found in the neurological field. This risk is higher in MS treatment, and specifically during long-term natalizumab therapy. While this drug is still routinely prescribed in this field, considering the efficacy in reducing MS relapses, in other areas it could play a smaller role, and be gradually replaced by other safer and more recently approved agents.
Collapse
Affiliation(s)
- Lorenzo Vittorio Rindi
- Department of Systems Medicine, Tor Vergata University, Via Montpellier, 1, 00133, Rome, Italy
| | - Drieda Zaçe
- Department of Systems Medicine, Tor Vergata University, Via Montpellier, 1, 00133, Rome, Italy
| | - Neva Braccialarghe
- Department of Systems Medicine, Tor Vergata University, Via Montpellier, 1, 00133, Rome, Italy
| | - Barbara Massa
- Department of Systems Medicine, Tor Vergata University, Via Montpellier, 1, 00133, Rome, Italy
| | - Virginia Barchi
- Department of Systems Medicine, Tor Vergata University, Via Montpellier, 1, 00133, Rome, Italy
| | - Roberta Iannazzo
- Department of Systems Medicine, Tor Vergata University, Via Montpellier, 1, 00133, Rome, Italy
| | - Ilenia Fato
- Department of Systems Medicine, Tor Vergata University, Via Montpellier, 1, 00133, Rome, Italy
| | - Francesco De Maria
- Department of Systems Medicine, Tor Vergata University, Via Montpellier, 1, 00133, Rome, Italy
| | - Dimitra Kontogiannis
- Department of Systems Medicine, Tor Vergata University, Via Montpellier, 1, 00133, Rome, Italy
| | - Vincenzo Malagnino
- Department of Systems Medicine, Tor Vergata University, Via Montpellier, 1, 00133, Rome, Italy
- Infectious Disease Clinic, Policlinico Tor Vergata, Viale Oxford, 81, 00133, Rome, Italy
| | - Loredana Sarmati
- Department of Systems Medicine, Tor Vergata University, Via Montpellier, 1, 00133, Rome, Italy
- Infectious Disease Clinic, Policlinico Tor Vergata, Viale Oxford, 81, 00133, Rome, Italy
| | - Marco Iannetta
- Department of Systems Medicine, Tor Vergata University, Via Montpellier, 1, 00133, Rome, Italy.
- Infectious Disease Clinic, Policlinico Tor Vergata, Viale Oxford, 81, 00133, Rome, Italy.
| |
Collapse
|
9
|
Storrer KM, Müller CDS, Pessoa MCDA, Pereira CADC. Connective tissue disease-associated interstitial lung disease. J Bras Pneumol 2024; 50:e20230132. [PMID: 38536980 PMCID: PMC11095924 DOI: 10.36416/1806-3756/e20230132] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Accepted: 11/22/2023] [Indexed: 05/18/2024] Open
Abstract
Connective tissue disease-associated interstitial lung disease (CTD-ILD) represents a group of systemic autoimmune disorders characterized by immune-mediated organ dysfunction. Systemic sclerosis, rheumatoid arthritis, idiopathic inflammatory myositis, and Sjögren's syndrome are the most common CTDs that present with pulmonary involvement, as well as with interstitial pneumonia with autoimmune features. The frequency of CTD-ILD varies according to the type of CTD, but the overall incidence is 15%, causing an important impact on morbidity and mortality. The decision of which CTD patient should be investigated for ILD is unclear for many CTDs. Besides that, the clinical spectrum can range from asymptomatic findings on imaging to respiratory failure and death. A significant proportion of patients will present with a more severe and progressive disease, and, for those, immunosuppression with corticosteroids and cytotoxic medications are the mainstay of pharmacological treatment. In this review, we summarized the approach to diagnosis and treatment of CTD-ILD, highlighting recent advances in therapeutics for the various forms of CTD.
Collapse
Affiliation(s)
| | | | | | - Carlos Alberto de Castro Pereira
- Disciplina de Pneumologia, Departamento de Medicina, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo (SP) Brasil
| |
Collapse
|
10
|
Rempenault C, Lukas C, Tardivon L, Daien CI, Combe B, Guilpain P, Morel J. Risk of severe infection associated with immunoglobulin deficiency under rituximab therapy in immune-mediated inflammatory disease. RMD Open 2024; 10:e003415. [PMID: 38296311 PMCID: PMC10836341 DOI: 10.1136/rmdopen-2023-003415] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Accepted: 11/17/2023] [Indexed: 02/05/2024] Open
Abstract
OBJECTIVES We evaluated the risk of severe infection in patients with immune-mediated inflammatory disease (IMID) treated with RTX and with Ig deficiency. METHODS This was an observational, retrospective single-centre study of patients undergoing treatment with at least one rituximab (RTX) infusion for an IMID until 31 May 2020. Patients were followed up for at least 12 months after the last infusion or until severe infection or death. Ig deficiency was classified as prevalent (before RTX) or acquired (normal Ig assay results before RTX but Ig deficiency during a follow-up). RESULTS Of 311 patients, 10.6% had prevalent and 19.6% acquired Ig deficiency. Prevalent Ig deficiency was related to concomitant treatment with glucocorticoids (GCs), in particular with a high daily dose at baseline; and acquired Ig deficiency to cumulative dose of RTX, mean Disease Activity Score in 28 joints (DAS28), immunosuppressor or GCs therapy at baseline, diabetes mellitus and obesity. Overall, 14.5% of patients had a severe infection during follow-up, which was numerically but not statistically more frequent in patients with prevalent Ig deficiency than normal Ig level. On multivariate analysis, risk of severe infection was associated with chronic pulmonary disease, GCs dose and mean DAS28-C reactive protein. In a time-dependent analysis, risk of severe infection was not associated with Ig deficiency, either acquired or prevalent (adjusted HR 1.04 (95% CI 0.5 to 2.3), p=0.92). CONCLUSION Risk of severe infection was not associated with RTX-induced Ig deficiency in patients with an IMID. RTX management should be discussed according to an individual assessment of the infectious risk, especially in patients with GC therapy or chronic lung disease.
Collapse
Affiliation(s)
| | - Cédric Lukas
- Rheumatology, CHU Montpellier, Montpellier, France
- UMR UA11 INSERM (IDESP), University of Montpellier, Montpellier, France
| | - Léa Tardivon
- Rheumatology, CHU Montpellier, Montpellier, France
| | - Claire Immediato Daien
- Rheumatology, CHU Montpellier, Montpellier, France
- INSERM, CNRS, University of Montpellier, Montpellier, France
| | | | - Philippe Guilpain
- Internal Medicine and Multi-Organic Diseases, CHU Montpellier, Montpellier, France
- IRMB, INSERM, University of Montpellier, Montpellier, France
| | - Jacques Morel
- Rheumatology, CHU Montpellier, Montpellier, France
- INSERM, CNRS, University of Montpellier, Montpellier, France
| |
Collapse
|
11
|
Omura S, Kida T, Noma H, Sunaga A, Kusuoka H, Kadoya M, Nakagomi D, Abe Y, Takizawa N, Nomura A, Kukida Y, Kondo N, Yamano Y, Yanagida T, Endo K, Hirata S, Matsui K, Takeuchi T, Ichinose K, Kato M, Yanai R, Matsuo Y, Shimojima Y, Nishioka R, Okazaki R, Takata T, Ito T, Moriyama M, Takatani A, Miyawaki Y, Ito-Ihara T, Yajima N, Kawaguchi T, Fukuda W, Kawahito Y. Association between hypogammaglobulinaemia and severe infections during induction therapy in ANCA-associated vasculitis: from J-CANVAS study. Rheumatology (Oxford) 2023; 62:3924-3931. [PMID: 36961329 DOI: 10.1093/rheumatology/kead138] [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: 10/14/2022] [Revised: 03/06/2023] [Accepted: 03/15/2023] [Indexed: 03/25/2023] Open
Abstract
OBJECTIVES To investigate the association between decreased serum IgG levels caused by remission-induction immunosuppressive therapy of antineutrophil cytoplasmic antibody-associated vasculitis (AAV) and the development of severe infections. METHODS We conducted a retrospective cohort study of patients with new-onset or severe relapsing AAV enrolled in the J-CANVAS registry, which was established at 24 referral sites in Japan. The minimum serum IgG levels up to 24 weeks and the incidence of severe infection up to 48 weeks after treatment initiation were evaluated. After multiple imputations for all explanatory variables, we performed the multivariate analysis using a Fine-Gray model to assess the association between low IgG (the minimum IgG levels <500 mg/dl) and severe infections. In addition, the association was expressed as a restricted cubic spline (RCS) and analysed by treatment subgroups. RESULTS Of 657 included patients (microscopic polyangiitis, 392; granulomatosis with polyangiitis, 139; eosinophilic granulomatosis with polyangiitis, 126), 111 (16.9%) developed severe infections. The minimum serum IgG levels were measured in 510 patients, of whom 77 (15.1%) had low IgG. After multiple imputations, the confounder-adjusted hazard ratio of low IgG for the incidence of severe infections was 1.75 (95% confidence interval: 1.03-3.00). The RCS revealed a U-shaped association between serum IgG levels and the incidence of severe infection with serum IgG 946 mg/dl as the lowest point. Subgroup analysis showed no obvious heterogeneity between treatment regimens. CONCLUSION Regardless of treatment regimens, low IgG after remission-induction treatment was associated with the development of severe infections up to 48 weeks after treatment initiation.
Collapse
Affiliation(s)
- Satoshi Omura
- Inflammation and Immunology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
- Center for Rheumatic Disease, Japanese Red Cross Society Kyoto Daiichi Hospital, Kyoto, Japan
| | - Takashi Kida
- Inflammation and Immunology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Hisashi Noma
- Department of Data Science, The Institute of Statistical Mathematics, Tokyo, Japan
| | - Atsuhiko Sunaga
- Inflammation and Immunology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
- Center for Rheumatic Disease, Japanese Red Cross Society Kyoto Daiichi Hospital, Kyoto, Japan
| | - Hiroaki Kusuoka
- Inflammation and Immunology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
- Center for Rheumatic Disease, Japanese Red Cross Society Kyoto Daiichi Hospital, Kyoto, Japan
| | - Masatoshi Kadoya
- Center for Rheumatic Disease, Japanese Red Cross Society Kyoto Daiichi Hospital, Kyoto, Japan
| | - Daiki Nakagomi
- Department of Rheumatology, University of Yamanashi Hospital, Yamanashi, Japan
| | - Yoshiyuki Abe
- Department of Internal Medicine and Rheumatology, Juntendo University, Tokyo, Japan
| | - Naoho Takizawa
- Department of Rheumatology, Chubu Rosai Hospital, Nagoya, Japan
| | - Atsushi Nomura
- Immuno-Rheumatology Center, St. Luke's International Hospital, Tokyo, Japan
| | - Yuji Kukida
- Department of Rheumatology, Japanese Red Cross Society Kyoto Daini Hospital, Kyoto, Japan
| | - Naoya Kondo
- Department of Nephrology, Kyoto Katsura Hospital, Kyoto, Japan
| | - Yasuhiko Yamano
- Department of Respiratory Medicine and Allergy, Tosei General Hospital, Aichi, Japan
| | - Takuya Yanagida
- Center for Rheumatic Disease, Japanese Red Cross Society Kyoto Daiichi Hospital, Kyoto, Japan
- Department of Hematology and Rheumatology, Kagoshima University Hospital, Kagoshima, Japan
| | - Koji Endo
- Department of General Internal Medicine, Tottori Prefectural Central Hospital, Tottori, Japan
| | - Shintaro Hirata
- Department of Clinical Immunology and Rheumatology, Hiroshima University Hospital, Hiroshima, Japan
| | - Kiyoshi Matsui
- Department of Diabetes, Endocrinology and Clinical Immunology, Hyogo Medical University School of Medicine, Hyogo, Japan
| | - Tohru Takeuchi
- Department of Internal Medicine (IV), Osaka Medical and Pharmaceutical University, Osaka, Japan
| | - Kunihiro Ichinose
- Department of Immunology and Rheumatology, Division of Advanced Preventive Medical Sciences, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
- Department of Rheumatology, Shimane University Faculty of Medicine, Shimane, Japan
| | - Masaru Kato
- Department of Rheumatology, Endocrinology and Nephrology, Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Ryo Yanai
- Division of Rheumatology, Department of Medicine, Showa University School of Medicine, Tokyo, Japan
| | - Yusuke Matsuo
- Department of Rheumatology, Tokyo Kyosai Hospital, Tokyo, Japan
- Department of Rheumatology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Yasuhiro Shimojima
- Department of Medicine (Neurology and Rheumatology), Shinshu University School of Medicine, Matsumoto, Japan
| | - Ryo Nishioka
- Department of Rheumatology, Graduate School of Medical Science, Kanazawa University, Kanazawa, Japan
| | - Ryota Okazaki
- Division of Respiratory Medicine and Rheumatology, Department of Multidisciplinary Internal Medicine, Faculty of Medicine, Tottori University, Yonago, Japan
| | - Tomoaki Takata
- Division of Gastroenterology and Nephrology, Tottori University, Yonago, Japan
| | - Takafumi Ito
- Division of Nephrology, Shimane University Hospital, Shimane, Japan
| | - Mayuko Moriyama
- Department of Rheumatology, Shimane University Faculty of Medicine, Shimane, Japan
| | - Ayuko Takatani
- Rheumatic Disease Center, Sasebo Chuo Hospital, Nagasaki, Japan
| | - Yoshia Miyawaki
- Department of Nephrology, Rheumatology, Endocrinology and Metabolism, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Toshiko Ito-Ihara
- The Clinical and Translational Research Center, University Hospital, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Nobuyuki Yajima
- Department of Rheumatology, Endocrinology and Nephrology, Graduate School of Medicine, Hokkaido University, Sapporo, Japan
- Department of Healthcare Epidemiology, Kyoto University Graduate School of Medicine and Public Health, Kyoto, Japan
- Center for Innovative Research for Communities and Clinical Excellence, Fukushima Medical University, Fukushima, Japan
| | - Takashi Kawaguchi
- Department of Practical Pharmacy, Tokyo University of Pharmacy and Life Sciences, Tokyo, Japan
| | - Wataru Fukuda
- Center for Rheumatic Disease, Japanese Red Cross Society Kyoto Daiichi Hospital, Kyoto, Japan
| | - Yutaka Kawahito
- Inflammation and Immunology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| |
Collapse
|
12
|
Alvarez E, Longbrake EE, Rammohan KW, Stankiewicz J, Hersh CM. Secondary hypogammaglobulinemia in patients with multiple sclerosis on anti-CD20 therapy: Pathogenesis, risk of infection, and disease management. Mult Scler Relat Disord 2023; 79:105009. [PMID: 37783194 DOI: 10.1016/j.msard.2023.105009] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Revised: 08/31/2023] [Accepted: 09/13/2023] [Indexed: 10/04/2023]
Abstract
Hypogammaglobulinemia is characterized by reduced serum immunoglobulin levels. Secondary hypogammaglobulinemia is of considerable interest to the practicing physician because it is a potential complication of some medications and may predispose patients to serious infections. Patients with multiple sclerosis (MS) treated with B-cell-depleting anti-CD20 therapies are particularly at risk of developing hypogammaglobulinemia. Among these patients, hypogammaglobulinemia has been associated with an increased risk of infections. The mechanism by which hypogammaglobulinemia arises with anti-CD20 therapies (ocrelizumab, ofatumumab, ublituximab, rituximab) remains unclear and does not appear to be simply due to the reduction in circulating B-cell levels. Further, despite the association between anti-CD20 therapies, hypogammaglobulinemia, and infections, there is currently no generally accepted monitoring and treatment approach among clinicians treating patients with MS. Here, we review the literature and discuss possible mechanisms of secondary hypogammaglobulinemia in patients with MS, hypogammaglobulinemia results in MS anti-CD20 therapy clinical trials, the risk of infection for patients with hypogammaglobulinemia, and possible strategies for disease management. We also include a suggested best-practice approach to specifically address secondary hypogammaglobulinemia in patients with MS treated with anti-CD20 therapies.
Collapse
Affiliation(s)
- Enrique Alvarez
- The Rocky Mountain MS Center at the University of Colorado Anschutz Medical Campus, Academic Office 1 Building, Room 5512, 12631 East 17th Avenue, B185, Aurora, CO 80045, United States
| | - Erin E Longbrake
- Department of Neurology, Yale School of Medicine, 6 Devine Street, Suite 2B, New Haven, CT 06473, United States
| | - Kottil W Rammohan
- Multiple Sclerosis Division, University of Miami Miller School of Medicine, 1120 NW 14th street, Suite 1322, Miami, FL 33136, United States
| | - James Stankiewicz
- Novartis Pharmaceuticals Corporation, East Hanover, NJ, United States
| | - Carrie M Hersh
- Cleveland Clinic Lou Ruvo Center for Brain Health, 888 W Bonneville Road, Las Vegas, NV 89106, United States.
| |
Collapse
|
13
|
Alhamadh MS, Alhowaish TS, Mathkour A, Altamimi B, Alheijani S, Alrashid A. Infection Risk, Mortality, and Hypogammaglobulinemia Prevalence and Associated Factors in Adults Treated with Rituximab: A Tertiary Care Center Experience. Clin Pract 2023; 13:1286-1302. [PMID: 37987416 PMCID: PMC10660466 DOI: 10.3390/clinpract13060115] [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: 09/13/2023] [Revised: 10/09/2023] [Accepted: 10/18/2023] [Indexed: 11/22/2023] Open
Abstract
BACKGROUND Rituximab is a human monoclonal antibody directed against the B-cell transmembrane protein CD20. Although well-tolerated, given its mechanism of action, rituximab can induce a state of severe immunosuppression, increasing the risk of opportunistic and fulminant infection and mortality. AIM To evaluate the risk of infection, mortality, and hypogammaglobulinemia and their associated factors among rituximab receivers. METHOD This was a single-center retrospective cohort study of adults treated with rituximab for various indications. Hypogammaglobulinemia was defined by a cut-off value below the normal limit (an IgG level of <7.51 g/L, an IgM level of <0.46 g/L, and/or an IgA level of <0.82 g/L). Patients who met the definition of hypogammaglobinemia solely based on IgA were excluded. Severe infection was defined as any infection that required intensive care unit admission. RESULTS A total of 137 adults with a mean age of 47.69 ± 18.86 years and an average BMI of 28.57 ± 6.55 kg/m2 were included. Hematological malignancies and connective tissue diseases were the most common primary diagnoses for which rituximab was used. More than half of the patients received the 375 mg/m2 dose. Rituximab's mean cumulative dose was 3216 ± 2282 mg, and the overall mortality rate was 22.6%. Hypogammaglobulinemia was diagnosed in 43.8% of the patients, and it was significantly more prevalent among males and the 375 mg/m2 and 500 mg doses. Hematological malignancy was the only predictor for infection. Patients with blood type AB or B, hematological malignancies, and corticosteroids had a significantly higher mortality rate. Receiving the 1000 mg dose and having a low CD19 were associated with a significantly lower risk of infection and mortality, respectively. CONCLUSIONS Hypogammaglobulinemia was diagnosed in 43.8% of the patients, and it was significantly more common among males and the 375 mg/m2 and 500 mg doses. Hematological malignancies were significantly associated with higher infection and mortality rates, while corticosteroids were significantly associated with a higher mortality. Since the culprit of mortality was infection, these findings highlight the critical need for more frequent immunological monitoring during rituximab treatment period to mitigate the burden of infection and identify candidates for immunoglobulin replacement.
Collapse
Affiliation(s)
- Moustafa S. Alhamadh
- College of Medicine, King Saud bin Abdulaziz University for Health Sciences, Ministry of the National Guard-Health Affairs, Riyadh 14611, Saudi Arabia
- King Abdullah International Medical Research Center, Ministry of the National Guard-Health Affairs, Riyadh 11481, Saudi Arabia; (T.S.A.)
| | - Thamer S. Alhowaish
- King Abdullah International Medical Research Center, Ministry of the National Guard-Health Affairs, Riyadh 11481, Saudi Arabia; (T.S.A.)
- Department of Neurology, King Abdulaziz Medical City, Ministry of the National Guard-Health Affairs, Riyadh 11426, Saudi Arabia
| | | | - Bayan Altamimi
- King Abdullah International Medical Research Center, Ministry of the National Guard-Health Affairs, Riyadh 11481, Saudi Arabia; (T.S.A.)
- Department of Medicine, Division of Rheumatology, King Abdulaziz Medical City, Ministry of the National Guard-Health Affairs, Riyadh 11426, Saudi Arabia
| | - Shahd Alheijani
- College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia
| | - Abdulrahman Alrashid
- College of Medicine, King Saud bin Abdulaziz University for Health Sciences, Ministry of the National Guard-Health Affairs, Riyadh 14611, Saudi Arabia
- King Abdullah International Medical Research Center, Ministry of the National Guard-Health Affairs, Riyadh 11481, Saudi Arabia; (T.S.A.)
- Department of Medicine, Division of Rheumatology, King Abdulaziz Medical City, Ministry of the National Guard-Health Affairs, Riyadh 11426, Saudi Arabia
| |
Collapse
|
14
|
Morel J. Infection prevention and vaccination in the rheumatic diseases. Joint Bone Spine 2023; 90:105568. [PMID: 36990142 DOI: 10.1016/j.jbspin.2023.105568] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/05/2022] [Indexed: 03/29/2023]
Abstract
Patients with chronic inflammatory rheumatisms (CIR) have a higher risk of infections compared to healthy subjects. Viral and bacterial pneumonia are the most frequent infections observed in CIR with targeted disease modifying anti-rheumatic drugs (DMARDs). Moreover, drugs used to treat CIR (especially biologic and synthetic targeted DMARDs) increase the risk of infection and expose CIR patients to opportunistic infections such as tuberculosis reactivation. To limit the risk of infection, the risk-benefit ratio should be evaluated for each patient based on their characteristics and comorbidities. To prevent infections, an initial pre-treatment work-up must be performed, especially before the initiation of conventional synthetic DMARDs or biological and synthetic targeted DMARDs. This pre-treatment assessment includes the case history, laboratory and radiology findings as well. The physician must make sure a patient's vaccinations are up-to-date. The vaccines recommended for patients with CIR being treated with conventional synthetic DMARDs, bDMARDs, tsDMARDs and/or steroids should be given. Patient education is also very important. During workshops, they learn how to manage their drug treatments in at-risk situations and learn which symptoms require treatment discontinuation.
Collapse
Affiliation(s)
- Jacques Morel
- Rheumatology Department, CHU and University of Montpellier, PhyMedExp, Inserm, CNRS, Montpellier, France.
| |
Collapse
|
15
|
Athni TS, Barmettler S. Hypogammaglobulinemia, late-onset neutropenia, and infections following rituximab. Ann Allergy Asthma Immunol 2023; 130:699-712. [PMID: 36706910 PMCID: PMC10247428 DOI: 10.1016/j.anai.2023.01.018] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 12/23/2022] [Accepted: 01/16/2023] [Indexed: 01/26/2023]
Abstract
Rituximab is a chimeric anti-CD20 monoclonal antibody that targets CD20-expressing B lymphocytes, has a well-defined efficacy and safety profile, and is broadly used to treat a wide array of diseases. In this review, we cover the mechanism of action of rituximab and focus on hypogammaglobulinemia and late-onset neutropenia-2 immune effects secondary to rituximab-and subsequent infection. We review risk factors and highlight key considerations for immunologic monitoring and clinical management of rituximab-induced secondary immune deficiencies. In patients treated with rituximab, monitoring for hypogammaglobulinemia and infections may help to identify the subset of patients at high risk for developing poor B cell reconstitution, subsequent infections, and adverse complications. These patients may benefit from early interventions such as vaccination, antibacterial prophylaxis, and immunoglobulin replacement therapy. Systematic evaluation of immunoglobulin levels and peripheral B cell counts by flow cytometry, both at baseline and periodically after therapy, is recommended for monitoring. In addition, in those patients with prolonged hypogammaglobulinemia and increased infections after rituximab use, immunologic evaluation for inborn errors of immunity may be warranted to further risk stratification, increase monitoring, and assist in therapeutic decision-making. As the immunologic effects of rituximab are further elucidated, personalized approaches to minimize the risk of adverse reactions while maximizing benefit will allow for improved care of patients with decreased morbidity and mortality.
Collapse
Affiliation(s)
| | - Sara Barmettler
- Allergy and Clinical Immunology Unit, Division of Rheumatology, Allergy, and Immunology, Massachusetts General Hospital, Boston, Massachusetts.
| |
Collapse
|
16
|
Dumusc A, Alromaih F, Perreau M, Hügle T, Zufferey P, Dan D. Real-life drug retention rate and safety of rituximab when treating rheumatic diseases: a single-centre Swiss retrospective cohort study. Arthritis Res Ther 2023; 25:91. [PMID: 37264414 DOI: 10.1186/s13075-023-03076-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Accepted: 05/25/2023] [Indexed: 06/03/2023] Open
Abstract
BACKGROUND In Switzerland, rituximab (RTX) is licenced for the treatment of rheumatoid arthritis (RA) and ANCA-associated vasculitis (AAV) but is frequently used off-label to treat other auto-immune diseases (AID), especially connective tissue diseases (CTD). We aimed to characterise the use of RTX in AID in a real-life Swiss setting and compare RTX retention rates and safety outcomes between patients treated for RA, CTD and AAV. METHODS A retrospective cohort study of patients who started RTX in the Rheumatology Department for RA or AID. The RTX retention rate was analysed using Kaplan-Meier survival curves. Occurrences of serious adverse events (SAE), low IgG levels and anti-drug antibodies (ADA) were reported. RESULTS Two hundred three patients were treated with RTX: 51.7% had RA, 29.6% CTD, 9.9% vasculitis and 8.9% other AIDs. The total observation time was 665 patient-years. RTX retention probability at 2 years (95%CI) was similar for RA and CTD 0.65 (0.55 to 0.73), 0.60 (0.47 to 0.72) and lower for vasculitis 0.25 (0.09 to 0.45). Survival curves for RTX retention matched closely (p = 0.97) between RA and CTD patients but were lower for patients with vasculitis due to a higher percentage of induced remission. Patients with vasculitis (95%) and CTD (75%) had a higher rate of concomitant glucocorticoid use than RA (60%). Moderate to severe hypogammaglobulinaemia was observed more frequently in patients with vasculitis (35%) than with RA (13%) or CTD (9%) and was associated with an increased risk of presenting a first infectious SAE (HR 2.01, 95% CI 1.04 to 3.91). The incidence rate of SAE was 23.3 SAE/100 patient-years (36% were infectious). When searched, ADAs were observed in 18% of the patients and were detected in 63% of infusions-related SAE. 10 patients died during RTX treatment and up to 12 months after the last RTX infusion, 50% from infection. CONCLUSION RTX retention rates are similar for patients with RA and CTD but lower for those with vasculitis due to more frequent remission. Patients treated with RTX for vasculitis present more SAE and infectious SAE than patients with RA and CTD, potentially due to a higher use of concomitant glucocorticoids and the occurrence of hypogammaglobulinaemia.
Collapse
Affiliation(s)
- Alexandre Dumusc
- Department of Rheumatology, Lausanne University Hospital, 1005, Lausanne, Switzerland.
- Faculty of Biology and Medicine, University of Lausanne, 1005, Lausanne, Switzerland.
| | - Fahad Alromaih
- Department of Rheumatology, Lausanne University Hospital, 1005, Lausanne, Switzerland
| | - Matthieu Perreau
- Faculty of Biology and Medicine, University of Lausanne, 1005, Lausanne, Switzerland
- Division of Immunology and Allergy, Lausanne University Hospital, 1005, Lausanne, Switzerland
| | - Thomas Hügle
- Department of Rheumatology, Lausanne University Hospital, 1005, Lausanne, Switzerland
- Faculty of Biology and Medicine, University of Lausanne, 1005, Lausanne, Switzerland
| | - Pascal Zufferey
- Department of Rheumatology, Lausanne University Hospital, 1005, Lausanne, Switzerland
- Faculty of Biology and Medicine, University of Lausanne, 1005, Lausanne, Switzerland
| | - Diana Dan
- Department of Rheumatology, Lausanne University Hospital, 1005, Lausanne, Switzerland
- Faculty of Biology and Medicine, University of Lausanne, 1005, Lausanne, Switzerland
| |
Collapse
|
17
|
Md Yusof MY, Arnold J, Saleem B, Vandevelde C, Dass S, Savic S, Vital EM, Emery P. Breakthrough SARS-CoV-2 infections and prediction of moderate-to-severe outcomes during rituximab therapy in patients with rheumatic and musculoskeletal diseases in the UK: a single-centre cohort study. THE LANCET. RHEUMATOLOGY 2023; 5:e88-e98. [PMID: 36712951 PMCID: PMC9873269 DOI: 10.1016/s2665-9913(23)00004-8] [Citation(s) in RCA: 35] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Background Concerns have been raised regarding the reduced immunogenicity of vaccines against COVID-19 in patients with autoimmune diseases treated with rituximab. However, the incidence and severity of breakthrough infections in unbiased samples of patients with specific rheumatic and musculoskeletal diseases are largely unknown. We aimed to assess the incidence of breakthrough SARS-CoV-2 infection, compare rates of moderate-to-severe COVID-19 with any severe infection event, and evaluate predictors of moderate-to-severe COVID-19 outcomes in patients treated with rituximab. Methods We did a retrospective cohort study in all rituximab-treated patients with rheumatic and musculoskeletal diseases in a single centre in Leeds, UK between March 1, 2020 (the index date), and April 1, 2022. Adults aged 18 years and older, who fulfilled classification criteria for established rheumatic and musculoskeletal diseases, and received therapy with at least one rituximab infusion between Sept 1, 2019 (6 months before the pandemic in the UK), and April 1, 2022, were eligible for inclusion in the study. SARS-CoV-2 infection was defined by antigen test or PCR. COVID-19 outcomes were categorised as mild (from ambulatory to hospitalised but not requiring oxygen support) or moderate-to-severe (hospitalised and requiring oxygen support or death). The primary outcome was breakthrough COVID-19 infection, which was defined as an infection occurring 14 days or more after the second vaccine dose. Predictors of moderate-to-severe COVID-19 outcomes were analysed using Cox regression proportional hazards. Findings Of the 1280 patients who were treated with at least one cycle of rituximab since Jan 1, 2002, 485 (38%) remained on rituximab therapy on April 1, 2022. Of these patients, 400 fulfilled all inclusion criteria and were included in our final analysis. The mean age at the index date was 58·9 years (SD 14·6), 288 (72%) of 400 patients were female and 112 (28%) were male, 333 (83%) were White, and 110 (28%) had two or more comorbidities. 272 (68%) of 400 patients had rheumatoid arthritis, 48 (12%) had systemic lupus erythematosus, 48 (12%) had anti-neutrophil cytoplasmic antibody-associated vasculitis, and 46 (12%) had other rheumatic and musculoskeletal diseases. During the study, 798 rituximab cycles were administered. Of the 398 (>99%) of 400 patients with vaccine data, 372 (93%) were fully vaccinated. Over the 774·6 patient-years of follow-up, there was an incremental increase in all SARS-CoV-2 severity types over the three pandemic phases (wild-type or alpha, delta, and omicron), but most infections were mild. The rates of moderate-to-severe COVID-19 were broadly similar across these three variant phases. Of 370 patients who were fully vaccinated and with complete data, 110 (30%) had all severity type breakthrough COVID-19, 16 (4%) had moderate-to-severe breakthrough COVID-19, and one (<1%) died. In the post-vaccination phase (after Dec 18, 2020), the incidence rates of all severity type and moderate-to-severe COVID-19 were substantially lower in those who were fully vaccinated compared with unvaccinated or partially vaccinated individuals (22·83 per 100 person-years [95% CI 18·94-27·52] in those who were fully vaccinated vs 89·46 per 100 person-years [52·98-151·05] in those who were partially vaccinated or unvaccinated for infections of all severities, and 3·32 per 100 person-years [2·03-5·42] in those who were fully vaccinated vs 25·56 per 100 person-years [9·59-68·10] in those who were partially vaccinated or unvaccinated for moderate-to-severe infections). The rate of moderate-to-severe COVID-19 was broadly similar to other severe infection events in this cohort (5·68 per 100 person-years [95% CI 4·22-7·63]). In multivariable Cox regression analysis, factors associated with an increased risk of moderate-to-severe COVID-19 were the number of comorbidities (hazard ratio 1·46 [95% CI 1·13-1·89]; p=0·0037) and hypogammaglobulinaemia (defined by a pre-rituximab IgG concentration of <6 g/L; 3·22 [1·27-8·19]; p=0·014). This risk was reduced with each vaccine dose received (0·49 [0·37-0·65]; p<0·0001). Other factors, including concomitant prednisolone use, rituximab-associated factors (eg, rituximab dose and time to vaccination since last rituximab dose), and vaccine-associated factors (eg, vaccine type and peripheral B-cell depletion) were not predictive of moderate-to-severe COVID-19 outcomes. Interpretation This study presented detailed analyses of rituximab-treated patients during various phases of the COVID-19 pandemic. In later stages of the pandemic, the SARS-CoV-2 breakthrough infection rate was high but severe COVID-19 rates were similar to any severe infection event rate in patients who were vaccinated. The risk-benefit ratio might still favour rituximab in vaccinated patients with severe rheumatic and musculoskeletal diseases who have few other treatment options. Increased vigilance is needed in the presence of comorbidities and hypogammaglobulinaemia for all infection types. Funding Wellcome Trust and Eli Lilly.
Collapse
Affiliation(s)
- Md Yuzaiful Md Yusof
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Chapel Allerton Hospital, Leeds, UK
- NIHR Leeds Biomedical Research Centre, Leeds Teaching Hospitals NHS Trust, Leeds, UK
| | - Jack Arnold
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Chapel Allerton Hospital, Leeds, UK
| | - Benazir Saleem
- Rheumatology Department, Leeds Teaching Hospitals NHS Trust, Leeds, UK
| | - Claire Vandevelde
- Rheumatology Department, Leeds Teaching Hospitals NHS Trust, Leeds, UK
| | - Shouvik Dass
- NIHR Leeds Biomedical Research Centre, Leeds Teaching Hospitals NHS Trust, Leeds, UK
- Rheumatology Department, Leeds Teaching Hospitals NHS Trust, Leeds, UK
| | - Sinisa Savic
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Chapel Allerton Hospital, Leeds, UK
- NIHR Leeds Biomedical Research Centre, Leeds Teaching Hospitals NHS Trust, Leeds, UK
| | - Edward M Vital
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Chapel Allerton Hospital, Leeds, UK
- NIHR Leeds Biomedical Research Centre, Leeds Teaching Hospitals NHS Trust, Leeds, UK
| | - Paul Emery
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Chapel Allerton Hospital, Leeds, UK
- NIHR Leeds Biomedical Research Centre, Leeds Teaching Hospitals NHS Trust, Leeds, UK
| |
Collapse
|
18
|
Makimoto K, Konno R, Kinoshita A, Kanzaki H, Suto S. Incidence of severe infection in patients with rheumatoid arthritis taking biological agents: a systematic review. JBI Evid Synth 2023; 21:835-885. [PMID: 36630204 DOI: 10.11124/jbies-22-00048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
OBJECTIVE The objective of this review was to estimate the population-based incidence of and determine the types of severe infection and deaths experienced by patients with rheumatoid arthritis taking biological agents. INTRODUCTION Since the late 1990s, various biological and synthetic drugs have been developed to treat rheumatoid arthritis. In recent years, the incidence of severe infection in patients with rheumatoid arthritis in Western nations has been determined by observational studies; however, no systematic review has been conducted on this topic. INCLUSION CRITERIA The following inclusion criteria were considered: i) observational studies on patients with rheumatoid arthritis treated with biological agents; ii) studies reporting the number of severe infections requiring hospitalization for treatment; iii) studies reporting person-years of observation data; and iv) studies based on rheumatoid arthritis registries, medical records from rheumatology centers, or insurance claim databases. METHODS PubMed, CINAHL, Embase, and Web of Science were searched to identify published studies. The reference lists of all studies selected for critical appraisal were screened for additional studies. Unpublished studies were searched on MedNar and OpenGrey databases. All the searches were updated on December 6, 2021. After removing the duplicates, 2 independent reviewers screened titles and abstracts against the inclusion criteria and then assessed full texts against the criteria. Two reviewers independently appraised the study and outcome levels for methodological quality using the critical appraisal instrument for cohort studies from JBI. Two reviewers extracted the relevant information related to severe infection and drugs. RESULTS Fifty-two studies from 21 countries reported severe infection rates associated with using 9 biologic agents. In total, 18,428 infections with 395,065 person-years of biologic drug exposure were included in the analysis. Thirty-five studies included infections in outpatients receiving intravenous antibiotic therapy. Fifteen studies reported the first episode of infection, and the remaining studies did not specify either the first or all of the episodes of infection. Inclusion of viral infection and/or opportunistic infection varied among studies. Fifteen studies reported the site of infection, and respiratory, skin/soft tissue, urinary tract infection, and sepsis/bacteremia were commonly reported. Ten studies reported the case fatality rates, ranging from 2.5% to 22.2%. Meta-analysis was conducted for 7 biologic agents and conventional disease-modifying antirheumatic drugs. The infection rate varied from 0.9 to 18.0/100 person-years. The meta-analysis revealed an infection rate of 4.2/100 person-years (95% CI 3.5-4.9) among patients receiving tumor necrosis factor inhibitors (heterogeneity 98.2%). The meta-analysis for the other 3 biologic agents revealed a point estimate of 5.5 to 8.7/100 person-years with high heterogeneity. Sensitivity analysis indicated that registry-based studies were less likely to have very low or very high infection rates compared with other data sources. The definition of infection, the patient composition of the cohorts, and the type of databases appeared to be the primary sources of clinical and methodological heterogeneity. SYSTEMATIC REVIEW REGISTRATION NUMBER PROSPERO CRD42020175137. CONCLUSIONS Due to high statistical heterogeneity, the meta-analysis was not suited to estimating a summary measure of the infection rate. Developing standardized data collection is necessary to compare infection rates across studies.
Collapse
Affiliation(s)
- Kiyoko Makimoto
- Osaka University, Suita, Osaka, Japan.,The Japan Centre for Evidence Based Practice: A JBI Centre of Excellence, Osaka University, Suita, Osaka, Japan
| | - Rie Konno
- Hyogo Medical University, Hypgo, Kobe, Japan
| | | | | | - Shunji Suto
- Nara Medical University, Kashihara, Nara, Japan
| |
Collapse
|
19
|
Opdam MAA, de Leijer JH, den Broeder N, Thurlings RM, van der Weele W, Nurmohamed MT, Kok MR, van Bon L, Ten Cate DF, Verhoef LM, den Broeder AA. Rituximab dose-dependent infection risk in rheumatoid arthritis is not mediated through circulating immunoglobulins, neutrophils or B cells. Rheumatology (Oxford) 2022; 62:330-334. [PMID: 35686851 DOI: 10.1093/rheumatology/keac318] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 05/20/2022] [Accepted: 05/20/2022] [Indexed: 12/27/2022] Open
Abstract
OBJECTIVES Rituximab (RTX) is a safe and effective treatment for RA. A dose-dependent infection risk was found in the REDO trial. Some studies associate RTX use with higher infection risks, possibly explained by low immunoglobulin levels and/or neutropenia. Additionally, a higher infection risk shortly after RTX infusion is reported. The objectives of this study were (i) to compare incidence rates of infections between doses and over time, and (ii) to assess B-cell counts, immunoglobulin levels, neutrophil counts and corticosteroid/disease modifying rheumatic drug use as mediating factors between RTX study dose and infection risk. METHODS Post hoc analyses of the REDO trial were performed. Infection incidence rates between RTX dosing groups and between time periods were compared using Poisson regression. A step-wise mediation analysis was performed to investigate if any of the factors mentioned above act as a mediator in the observed dose-dependent difference in infection risk. RESULTS The potential mediators that were investigated (circulating B-cell counts, immunoglobulin levels, neutrophil counts and drug use) did not explain the dose-dependent infection risk observed in the REDO trial. Additionally, a trend towards a time-dependent infection risk was found, with higher infection rates shortly after RTX infusion. CONCLUSIONS These secondary analyses of the REDO trial confirmed the observed dose-dependent infection risk. Additionally, we found that infection risks were higher shortly after RTX infusion. However, a mediating pathway was not found.
Collapse
Affiliation(s)
- Merel A A Opdam
- Department of Rheumatology, Sint Maartenskliniek.,Department of Rheumatology, Radboud Institute for Health Sciences, Radboudumc
| | | | | | | | | | | | - Marc R Kok
- Department of Rheumatology & Clinical Immunology, Maasstad Ziekenhuis, Rotterdam
| | - Lenny van Bon
- Department of Rheumatology, Hospital Group Twente, Almelo, the Netherlands
| | | | | | - Alfons A den Broeder
- Department of Rheumatology, Sint Maartenskliniek.,Department of Rheumatic Diseases, Radboudumc, Nijmegen
| |
Collapse
|
20
|
Koltsova EN, Lukina GV, Schmidt EI, Lytkina KA, Zhilyaev EV. Predictors of biologic disease modifying antirheumatic drugs withdrawal due to the development of adverse events in patients with rheumatoid arthritis. MODERN RHEUMATOLOGY JOURNAL 2022. [DOI: 10.14412/1996-7012-2022-6-26-31] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Currently, a large number of highly effective biologic disease modifying antirheumatic drugs (bDMARDs) and targeted synthetic DMARDs (tsDMARDs) are used for the treatment of rheumatoid arthritis (RA). However, in addition to effectiveness, it is necessary to evaluate the risk of adverse events (AEs) when using them.Objective: to determine the predictors of bDMARDs and tsDMARDs discontinuation due to AEs in patients with RA.Patients and methods. The study included 661 patients with RA who took bDMARDs and tsDMARDs. The search for predictors of targeted therapy discontinuation due to AEs was carried out in two stages. At the first stage, using the Kaplan-Meier method, we selected indicators that showed the greatest significant single-factor relationship with the duration of retention on therapy. At the second stage, significant independent indicators were obtained by iterative selection of variables within the multivariate proportional risk model according to Cox.Results and discussion. The presence of rheumatoid nodules (p<0.001), high doses of glucocorticoids (GC; p<0.001), low doses of methotrexate (MT; p=0.009) are significant independent factors for increasing the risk of drugs discontinuation due to the development of AEs. The type of bDMARDs/tsDMARD used also significantly correlated with the risk of discontinuation of therapy due to AEs. A relatively high risk of treatment discontinuation was observed with infliximab (IFN) and certolizumab pegol (CZP). Cancellation of IFN was associated with the occurrence of infusion reactions and infectious complications, and CZP was associated with infectious complications.Conclusion. An increase in the dose of MT and decrease in the use of GCs can help prevent the development of AEs leading to the abolition of biologics and tsDMARDs. Significant differences were found between bDMARDs in terms of the risk of their cancellation due to AEs.
Collapse
Affiliation(s)
| | - G. V. Lukina
- A.S. Loginov Moscow Clinical Scientific Center; V.A. Nasonova Research Institute of Rheumatology
| | | | | | - E. V. Zhilyaev
- European Medical Center; Russian Medical Academy of Continuing Professional Education, Ministry of Health of Russia; Pirogov Russian National Research Medical University
| |
Collapse
|
21
|
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
|
22
|
Vishnevetsky A, Kaplan TB, Levy M. Transitioning immunotherapy in neuromyelitis optica spectrum disorder – when and how to switch. Expert Opin Biol Ther 2022; 22:1393-1404. [DOI: 10.1080/14712598.2022.2145879] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Affiliation(s)
- Anastasia Vishnevetsky
- Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Tamara B. Kaplan
- Department of Neurology, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, USA
| | - Michael Levy
- Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| |
Collapse
|
23
|
Advancing Biologic Therapy for Refractory Autoimmune Hepatitis. Dig Dis Sci 2022; 67:4979-5005. [PMID: 35147819 DOI: 10.1007/s10620-021-07378-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Accepted: 12/27/2021] [Indexed: 01/05/2023]
Abstract
Biologic agents may satisfy an unmet clinical need for treatment of refractory autoimmune hepatitis. The goals of this review are to present the types and results of biologic therapy for refractory autoimmune hepatitis, indicate opportunities to improve and expand biologic treatment, and encourage comparative clinical trials. English abstracts were identified in PubMed by multiple search terms. Full-length articles were selected for review, and secondary and tertiary bibliographies were developed. Rituximab (monoclonal antibodies against CD20 on B cells), infliximab (monoclonal antibodies against tumor necrosis factor-alpha), low-dose recombinant interleukin 2 (regulatory T cell promoter), and belimumab (monoclonal antibodies against B cell activating factor) have induced laboratory improvement in small cohorts with refractory autoimmune hepatitis. Ianalumab (monoclonal antibodies against the receptor for B cell activating factor) is in clinical trial. These agents target critical pathogenic pathways, but they may also have serious side effects. Blockade of the B cell activating factor or its receptors may disrupt pivotal B and T cell responses, and recombinant interleukin 2 complexed with certain interleukin 2 antibodies may selectively expand the regulatory T cell population. A proliferation-inducing ligand that enhances T cell proliferation and survival is an unevaluated, potentially pivotal, therapeutic target. Fully human antibodies, expanded target options, improved targeting precision, more effective delivery systems, and biosimilar agents promise to improve efficacy, safety, and accessibility. In conclusion, biologic agents target key pathogenic pathways in autoimmune hepatitis, and early experiences in refractory disease encourage clarification of the preferred target, rigorous clinical trial, and comparative evaluations.
Collapse
|
24
|
Narváez J, Díaz Del Campo Fontecha P, Brito García N, Bonilla G, Aburto M, Castellví I, Cano-Jiménez E, Mena-Vázquez N, Nieto MA, Ortiz AM, Valenzuela C, Abad Hernández MÁ, Castrejón I, Correyero Plaza M, Francisco Hernández FM, Hernández Hernández MV, Rodríquez Portal JA. SER-SEPAR recommendations for the management of rheumatoid arthritis-related interstitial lung disease. Part 2: Treatment. REUMATOLOGIA CLINICA 2022; 18:501-512. [PMID: 36064885 DOI: 10.1016/j.reumae.2022.03.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Accepted: 03/15/2022] [Indexed: 06/15/2023]
Abstract
OBJECTIVE To develop multidisciplinary recommendations to improve the management of rheumatoid arthritis-related interstitial lung disease (RA-ILD). METHODS Clinical research questions relevant to the objective of the document were identified by a panel of rheumatologists and pneumologists selected based on their experience in the field. Systematic reviews of the available evidence were conducted, and evidence was graded according to the Scottish Intercollegiate Guidelines Network (SIGN) criteria. Specific recommendations were made. RESULTS Six PICO questions were selected, three of which analysed the safety and effectiveness of glucocorticoids, classical synthetic disease-modifying anti-rheumatic drugs (DMARDs) and other immunosuppressants, biological agents, targeted synthetic DMARDs, and antifibrotic therapies in the treatment of this complication. A total of 12 recommendations were formulated based on the evidence found and/or expert consensus. CONCLUSIONS We present the first official SER-SEPAR document with specific recommendations for RA-ILD management developed to resolve some common clinical questions, reduce clinical healthcare variability, and facilitate decision-making for patients.
Collapse
Affiliation(s)
- Javier Narváez
- Servicio de Reumatología, Hospital Universitario de Bellvitge, Hospitalet de Llobregat, Barcelona, Spain.
| | | | - Noé Brito García
- Unidad de Investigación, Sociedad Española de Reumatología, Madrid, Spain
| | - Gema Bonilla
- Servicio de Reumatología, Hospital Universitario La Paz, Madrid, Spain
| | - Myriam Aburto
- Servicio de Neumología, Hospital Universitario Galdakao-Usansolo, Bilbao, Spain
| | - Iván Castellví
- Servicio de Reumatología, Hospital Universitario de la Santa Creu i Sant Pau, Barcelona, Spain
| | | | - Natalia Mena-Vázquez
- Servicio de Reumatología, Hospital Regional Universitario de Málaga, Málaga, Spain
| | - M Asunción Nieto
- Servicio de Neumología, Hospital Clínico San Carlos, Madrid, Spain
| | - Ana María Ortiz
- Servicio de Reumatología, Hospital Universitario de La Princesa, Madrid, Spain
| | - Claudia Valenzuela
- Servicio de Neumología, Hospital Universitario de La Princesa, Madrid, Spain
| | | | - Isabel Castrejón
- Servicio de Reumatología, Hospital General Universitario Gregorio Marañón, Madrid, Spain
| | - María Correyero Plaza
- Servicio de Reumatología, Hospital Universitario Quironsalud de Pozuelo, Pozuelo de Alarcón, Madrid, Spain
| | | | | | | |
Collapse
|
25
|
Gottenberg JE, Chaudier A, Allenbach Y, Mekinian A, Amoura Z, Cacoub P, Cornec D, Hachulla E, Quartier P, Melki I, Richez C, Seror R, Terrier B, Devauchelle-Pensec V, Henry J, Gatfosse M, Bouillet L, Gaigneux E, Andre V, Baulier G, Saunier A, Desmurs M, Poulet A, Ete M, Bienvenu B, Truchetet ME, Michaud M, Larroche C, Dellal A, Leurs A, Ottaviani S, Nielly H, Vial G, Jaussaud R, Rouvière B, Jeandel PY, Guffroy A, Korganow AS, Jouvray M, Meyer A, Chatelus E, Sordet C, Felten R, Sibilia J, Litim-Ahmed-Yahia S, Kleinmann JF, Mariette X. Tolerance and efficacy of targeted therapies prescribed for off-label indications in refractory systemic autoimmune diseases: data of the first 100 patients enrolled in the TATA registry (TArgeted Therapy in Autoimmune Diseases). RMD Open 2022; 8:rmdopen-2022-002324. [PMID: 36319066 PMCID: PMC9628685 DOI: 10.1136/rmdopen-2022-002324] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Accepted: 07/19/2022] [Indexed: 11/07/2022] Open
Abstract
Objectives To assess the tolerance and efficacy of targeted therapies prescribed off-label in refractory low-prevalence autoimmune and inflammatory systemic diseases. Methods The TATA registry (TArgeted Therapy in Autoimmune Diseases) is a prospective, observational, national and independent cohort follow-up. The inclusion criteria in the registry are as follows: age >18 years; low-prevalence autoimmune and inflammatory systemic disease treated with off-label drugs started after 1 January 2019. Results Hundred (100) patients (79 women) were enrolled. The median age was 52.5 years (95% CI 49 to 56) and the median disease duration before enrolment was 5 years (3 to 7). The targeted therapies at enrolment were as follows: Janus kinase/signal transducers and activators of transcription inhibitors (44%), anti-interleukin (IL)-6R (22%), anti-IL-12/23, anti-IL-23 and anti-IL-17 (9%), anti-B cell activating factor of the tumour necrosis factor family (5%), abatacept (5%), other targeted treatments (9%) and combination of targeted treatments (6%). 73% of patients were receiving corticosteroid therapy at enrolment (median dose 10 mg/day). The current median follow-up time is 9 months (8 to 10). Safety: 11 serious infections (incidence rate of 14.8/100 patient-years) and 1 cancer (1.3 cancers/100 patient-years) were observed. Two patients died from severe COVID-19 (2.7 deaths/100 patient-years). Efficacy: the targeted treatment was considered effective by the clinician in 56% of patients and allowed, in responders, a median reduction of oral corticosteroids of 15 (9 to 21) mg/day, below 7.5 mg/day in 76% of patients, while 28% discontinued. Conclusion These initial results of the TATA registry confirm the diversity of targeted treatments prescribed off-label in refractory autoimmune diseases and their corticosteroid-sparing effect when effective. Tolerance was acceptable in these refractory patients with a long history of treatment with immunosuppressive drugs.
Collapse
Affiliation(s)
- Jacques-Eric Gottenberg
- Rheumatology, Hôpitaux universitaires de Strasbourg, Strasbourg, France,Rheumatology, East and South-West Reference Center, Strasbourg, France
| | - Aurore Chaudier
- Rheumatology, Hôpitaux universitaires de Strasbourg, Strasbourg, France,Rheumatology, East and South-West Reference Center, Strasbourg, France
| | - Yves Allenbach
- Department of Internal Medicine and Clinical Immunology, Hospital University Department: Inflammation, Immunopathology and Biotherapy (DHU i2B), University Hospital Pitié Salpêtrière, Paris, France,Internal Medicine, Ile-De-France Reference Center, Paris, France
| | - Arsène Mekinian
- Service de Médecine Interne, DHUi2B, Hospital Saint-Antoine, Paris, France
| | - Zahir Amoura
- Internal Medicine, University Hospital Pitié Salpêtrière, Paris, France,Internal Medicine, Lupus and SAPL Reference Center, Paris, France
| | - Patrice Cacoub
- Internal Medicine, Ile-De-France Reference Center, Paris, France,Service de médecine interne et immunologie clinique, Hopital Pitie-Salpetriere, Paris, France
| | - Divi Cornec
- Rhumatologie, Cavale Blanche Hospital, Brest, France,Rheumatology, North and North-West Reference Center, Brest, France
| | - Eric Hachulla
- Internal Medicine, Lille University School of Medicine, Lille, France,Internal Medicine, North and North-West Reference Center, Lille, France
| | - Pierre Quartier
- Pediatric Immuno-Hematology and Rheumatology Unit, Hopital universitaire Necker-Enfants malades, Paris, France,Paediatric, RAISE Reference Center, Paris, France
| | - Isabelle Melki
- Laboratory of Neurogenetics and Neuroinflammation, Imagine Institute, Paris, France,General Paediatrics, Infectious Diseases and Internal Medicine, Hopital Universitaire Robert Debre, Paris, France
| | - Christophe Richez
- Service de Rhumatologie, CHU Bordeaux GH Pellegrin, Bordeaux, France,Rheumatology, East and South-West Reference Center, Bordeaux, France
| | - Raphaele Seror
- Rheumatology, Hôpital Bicêtre, Le Kremlin-Bicetre, France,Rheumatology, Ile-De-France Reference Center, Le Kremlin-Bicetre, France
| | - Benjamin Terrier
- Internal Medicine, Ile-De-France Reference Center, Paris, France,Internal Medicine, Hospital Cochin, Paris, France
| | - Valérie Devauchelle-Pensec
- Rhumatologie, Cavale Blanche Hospital, Brest, France,Rheumatology, North and North-West Reference Center, Brest, France
| | - Julien Henry
- Rheumatology, Hôpital Bicêtre, Le Kremlin-Bicetre, France,Rheumatology, Ile-De-France Reference Center, Le Kremlin-Bicetre, France
| | - Marc Gatfosse
- Médecine Interne, Hôpital Saint-Antoine, Paris, France
| | - Laurence Bouillet
- Internal Medicine, University Hospital Centre Grenoble Alpes, Grenoble, France
| | - Emeline Gaigneux
- Rheumatology, Centre Hospitalier Départemental Vendée, La Roche-sur-Yon, France
| | - Vincent Andre
- Rheumatology, Centre Hospitalier Départemental Vendée, La Roche-sur-Yon, France
| | - Gildas Baulier
- Internal Medicine, Centre Hospitalier de Périgueux, Perigueux, France
| | - Aurélie Saunier
- Internal Medicine, Centre Hospitalier de Périgueux, Perigueux, France
| | - Marie Desmurs
- Rheumatology, Hospital Emile Muller, Mulhouse, France
| | - Antoine Poulet
- Internal Medicine, Saint Joseph Hospital, Marseille, France
| | | | - Boris Bienvenu
- Internal Medicine, Saint Joseph Hospital, Marseille, France
| | - Marie-Elise Truchetet
- Rheumatology, East and South-West Reference Center, Bordeaux, France,Rheumatology, University Hospital Centre Bordeaux, Bordeaux, France
| | - Martin Michaud
- Internal Medicine, Hopital Joseph Ducuing, Toulouse, France
| | - Claire Larroche
- Service de médecine interne, Hopital Avicenne, Bobigny, France
| | - Azeddine Dellal
- Rheumatology, Groupe Hospitalier Intercommunal Le Raincy-Montfermeil, Montfermeil, France
| | - Amélie Leurs
- Internal Medicine, Centre Hospitalier de Dunkerque, Dunkerque, France
| | | | - Hubert Nielly
- Internal Medicine, Begin Armed Forces Training Hospital, Paris, France
| | - Guillaume Vial
- Department of Internal Medicine and Clinical Immunology, CHU de Bordeaux, Bordeaux, France
| | | | | | | | - Aurelien Guffroy
- Rheumatology, East and South-West Reference Center, Strasbourg, France,Internal Medicine, Hôpitaux universitaires de Strasbourg, Strasbourg, France
| | - Anne-Sophie Korganow
- Rheumatology, East and South-West Reference Center, Strasbourg, France,Internal Medicine, Hôpitaux universitaires de Strasbourg, Strasbourg, France
| | - Mathieu Jouvray
- Internal Medicine, Begin Armed Forces Training Hospital, Paris, France
| | - Alain Meyer
- Rheumatology, Hôpitaux universitaires de Strasbourg, Strasbourg, France,Rheumatology, East and South-West Reference Center, Strasbourg, France
| | - Emmanuel Chatelus
- Rheumatology, Hôpitaux universitaires de Strasbourg, Strasbourg, France,Rheumatology, East and South-West Reference Center, Strasbourg, France
| | - Christelle Sordet
- Rheumatology, Hôpitaux universitaires de Strasbourg, Strasbourg, France,Rheumatology, East and South-West Reference Center, Strasbourg, France
| | - Renaud Felten
- Rheumatology, Hôpitaux universitaires de Strasbourg, Strasbourg, France,Rheumatology, East and South-West Reference Center, Strasbourg, France
| | - Jean Sibilia
- Rheumatology, Hôpitaux universitaires de Strasbourg, Strasbourg, France,Rheumatology, East and South-West Reference Center, Strasbourg, France
| | | | - Jean-Francois Kleinmann
- Rheumatology, Hôpitaux universitaires de Strasbourg, Strasbourg, France,Rheumatology, East and South-West Reference Center, Strasbourg, France
| | - Xavier Mariette
- Rheumatology, Hôpital Bicêtre, Le Kremlin-Bicetre, France,Rheumatology, Ile-De-France Reference Center, Le Kremlin-Bicetre, France
| |
Collapse
|
26
|
Abstract
Purpose of Review Rituximab has transformed the treatment of B-cell malignancies and rheumatoid arthritis in the past 2 decades. More recently, this anti-CD20 monoclonal antibody has seen increasing usage in the field of dermatology. This review highlights the evidence supporting its use in several important dermatologic conditions. Recent Findings Key recent findings include the 2018 FDA approval of rituximab for the treatment of moderate-to-severe pemphigus. Summary Data from randomized controlled trials have demonstrated the efficacy of rituximab in pemphigus, ANCA-associated vasculitis, and cryoglobulinemic vasculitis. More limited data suggests its use in recalcitrant cases of diseases such as pemphigoid, epidermolysis bullosa acquisita, and dermatomyositis. There is scarce evidence and mixed results for rituximab when studied in cutaneous polyarteritis nodosa and cutaneous lupus erythematosus.
Collapse
Affiliation(s)
- Connor Cole
- Division of Dermatology, Rush University Medical Center, Chicago, IL USA
| | - Kyle T. Amber
- Division of Dermatology, Rush University Medical Center, Chicago, IL USA
- Department of Internal Medicine, Rush University Medical Center, Chicago, IL USA
| |
Collapse
|
27
|
Goodman SM, Springer BD, Chen AF, Davis M, Fernandez DR, Figgie M, Finlayson H, George MD, Giles JT, Gilliland J, Klatt B, MacKenzie R, Michaud K, Miller A, Russell L, Sah A, Abdel MP, Johnson B, Mandl LA, Sculco P, Turgunbaev M, Turner AS, Yates A, Singh JA. 2022 American College of Rheumatology/American Association of Hip and Knee Surgeons Guideline for the Perioperative Management of Antirheumatic Medication in Patients With Rheumatic Diseases Undergoing Elective Total Hip or Total Knee Arthroplasty. J Arthroplasty 2022; 37:1676-1683. [PMID: 35732511 DOI: 10.1016/j.arth.2022.05.043] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
OBJECTIVE To develop updated American College of Rheumatology/American Association of Hip and Knee Surgeons guidelines for the perioperative management of disease-modifying medications for patients with rheumatic diseases, specifically those with inflammatory arthritis (IA) and those with systemic lupus erythematosus (SLE), undergoing elective total hip arthroplasty (THA) or elective total knee arthroplasty (TKA). METHODS We convened a panel of rheumatologists, orthopedic surgeons, and infectious disease specialists, updated the systematic literature review, and included currently available medications for the clinically relevant population, intervention, comparator, and outcomes (PICO) questions. We used the Grading of Recommendations Assessment, Development and Evaluation (GRADE) methodology to rate the quality of evidence and the strength of recommendations using a group consensus process. RESULTS This guideline updates the 2017 recommendations for perioperative use of disease-modifying antirheumatic therapy, including traditional disease-modifying antirheumatic drugs, biologic agents, targeted synthetic small-molecule drugs, and glucocorticoids used for adults with rheumatic diseases, specifically for the treatment of patients with IA, including rheumatoid arthritis and spondyloarthritis, those with juvenile idiopathic arthritis, or those with SLE who are undergoing elective THA or TKA. It updates recommendations regarding when to continue, when to withhold, and when to restart these medications and the optimal perioperative dosing of glucocorticoids. CONCLUSION This updated guideline includes recently introduced immunosuppressive medications to help decision-making by clinicians and patients regarding perioperative disease-modifying medication management for patients with IA and SLE at the time of elective THA or TKA.
Collapse
Affiliation(s)
- Susan M Goodman
- Hospital for Special Surgery, Weill Cornell Medicine, New York, New York.
| | | | | | | | - David R Fernandez
- Hospital for Special Surgery, Weill Cornell Medicine, New York, New York
| | - Mark Figgie
- Hospital for Special Surgery, Weill Cornell Medicine, New York, New York
| | - Heather Finlayson
- Multispecialty Physician Partners, LLC, Colorado Arthritis Associates, Lakewood, Colorado
| | | | | | - Jeremy Gilliland
- University of Utah and Veterans Affairs Medical Center, Salt Lake City
| | - Brian Klatt
- University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | - Ronald MacKenzie
- Hospital for Special Surgery, Weill Cornell Medicine, New York, New York
| | - Kaleb Michaud
- University of Nebraska Medical Center, Omaha, Nebraska, and Forward Databank, Wichita, Kansas
| | - Andy Miller
- Hospital for Special Surgery, Weill Cornell Medicine, New York, New York
| | - Linda Russell
- Hospital for Special Surgery, Weill Cornell Medicine, New York, New York
| | - Alexander Sah
- Sah Orthopaedic Associates, Institute for Joint Restoration, Freemont, California
| | | | | | - Lisa A Mandl
- Hospital for Special Surgery, Weill Cornell Medicine, New York, New York
| | - Peter Sculco
- Hospital for Special Surgery, Weill Cornell Medicine, New York, New York
| | | | - Amy S Turner
- American College of Rheumatology, Atlanta, Georgia
| | - Adolph Yates
- University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | - Jasvinder A Singh
- University of Alabama at Birmingham and Veterans Affairs Medical Center, Birmingham, Alabama
| |
Collapse
|
28
|
IgG-Based Bispecific Anti-CD95 Antibodies for the Treatment of B Cell-Derived Malignancies and Autoimmune Diseases. Cancers (Basel) 2022; 14:cancers14163941. [PMID: 36010934 PMCID: PMC9405798 DOI: 10.3390/cancers14163941] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Revised: 08/12/2022] [Accepted: 08/13/2022] [Indexed: 11/23/2022] Open
Abstract
Simple Summary Therapeutic antibodies have become a crucial cornerstone of the standard therapy for lymphoma and autoimmune diseases. However, the respective target antigens are also expressed on healthy B cells resulting in unspecific effects. In this article, we present a novel approach to selectively induce apoptosis in lymphoma cells and autoreactive B cells that express the CD95 death receptor. Therefore, we developed an improved IgG-based bispecific antibody format with favorable production properties and pharmacokinetics for CD20- and CD19-directed induction of apoptosis via CD95. We could show that our bispecific anti-CD95 antibodies are very efficient in the depletion of malignant and autoreactive B cells in vitro and in vivo. Therefore, our antibodies could help to provide a more selective therapy for patients with B cell-derived malignancies and autoimmune diseases. Abstract Antibodies against the B cell-specific antigens CD20 and CD19 have markedly improved the treatment of B cell-derived lymphoma and autoimmune diseases by depleting malignant and autoreactive B cells. However, since CD20 and CD19 are also expressed on healthy B cells, such antibodies lack disease specificity. Here, we optimize a previously developed concept that uses bispecific antibodies to induce apoptosis selectively in malignant and autoreactive B cells that express the death receptor CD95. We describe the development and characterization of bispecific antibodies with CD95xCD20 and CD95xCD19 specificity in a new IgG-based format. We could show that especially the CD95xCD20 antibody mediated a strong induction of apoptosis in malignant B cells in vitro. In vivo, the antibody was clearly superior to the previously used Fabsc format with identical specificities. In addition, both IgGsc antibodies depleted activated B cells in vitro, leading to a significant reduction in antibody production and cytokine secretion. The killing of resting B cells and hepatocytes that lack CD95 and CD20/CD19, respectively, was marginal. Thus, our results imply that bispecific anti-CD95 antibodies in the IgGsc format are an attractive tool for a more selective and efficient depletion of malignant as well as autoreactive B cells.
Collapse
|
29
|
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
|
30
|
Goodman SM, Springer BD, Chen AF, Davis M, Fernandez DR, Figgie M, Finlayson H, George MD, Giles JT, Gilliland J, Klatt B, MacKenzie R, Michaud K, Miller A, Russell L, Sah A, Abdel MP, Johnson B, Mandl LA, Sculco P, Turgunbaev M, Turner AS, Yates A, Singh JA. 2022 American College of Rheumatology/American Association of Hip and Knee Surgeons Guideline for the Perioperative Management of Antirheumatic Medication in Patients With Rheumatic Diseases Undergoing Elective Total Hip or Total Knee Arthroplasty. Arthritis Rheumatol 2022; 74:1464-1473. [PMID: 35722708 DOI: 10.1002/art.42140] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Revised: 03/18/2022] [Accepted: 04/07/2022] [Indexed: 11/12/2022]
Abstract
OBJECTIVE To develop updated guidelines for the perioperative management of disease-modifying medications for patients with rheumatic diseases, specifically those with inflammatory arthritis (IA) and those with systemic lupus erythematosus (SLE), undergoing elective total hip arthroplasty (THA) or elective total knee arthroplasty (TKA). METHODS We convened a panel of rheumatologists, orthopedic surgeons, and infectious disease specialists, updated the systematic literature review, and included currently available medications for the clinically relevant population, intervention, comparator, and outcomes (PICO) questions. We used the Grading of Recommendations Assessment, Development and Evaluation (GRADE) methodology to rate the quality of evidence and the strength of recommendations using a group consensus process. RESULTS This guideline updates the 2017 recommendations for perioperative use of disease-modifying antirheumatic therapy, including traditional disease-modifying antirheumatic drugs, biologic agents, targeted synthetic small-molecule drugs, and glucocorticoids used for adults with rheumatic diseases, specifically for the treatment of patients with IA, including rheumatoid arthritis and spondyloarthritis, those with juvenile idiopathic arthritis, or those with SLE who are undergoing elective THA or TKA. It updates recommendations regarding when to continue, when to withhold, and when to restart these medications and the optimal perioperative dosing of glucocorticoids. CONCLUSION This updated guideline includes recently introduced immunosuppressive medications to help decision-making by clinicians and patients regarding perioperative disease-modifying medication management for patients with IA and SLE at the time of elective THA or TKA.
Collapse
Affiliation(s)
- Susan M Goodman
- Hospital for Special Surgery, Weill Cornell Medicine, New York, New York
| | | | | | | | - David R Fernandez
- Hospital for Special Surgery, Weill Cornell Medicine, New York, New York
| | - Mark Figgie
- Hospital for Special Surgery, Weill Cornell Medicine, New York, New York
| | - Heather Finlayson
- Multispecialty Physician Partners, LLC, Colorado Arthritis Associates, Lakewood, Colorado
| | | | | | - Jeremy Gilliland
- University of Utah and Veterans Affairs Medical Center, Salt Lake City
| | - Brian Klatt
- University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | - Ronald MacKenzie
- Hospital for Special Surgery, Weill Cornell Medicine, New York, New York
| | - Kaleb Michaud
- University of Nebraska Medical Center, Omaha, Nebraska, and Forward Databank, Wichita, Kansas
| | - Andy Miller
- Hospital for Special Surgery, Weill Cornell Medicine, New York, New York
| | - Linda Russell
- Hospital for Special Surgery, Weill Cornell Medicine, New York, New York
| | - Alexander Sah
- Sah Orthopaedic Associates, Institute for Joint Restoration, Freemont, California
| | | | | | - Lisa A Mandl
- Hospital for Special Surgery, Weill Cornell Medicine, New York, New York
| | - Peter Sculco
- Hospital for Special Surgery, Weill Cornell Medicine, New York, New York
| | | | - Amy S Turner
- American College of Rheumatology, Atlanta, Georgia
| | - Adolph Yates
- University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | - Jasvinder A Singh
- University of Alabama at Birmingham and Veterans Affairs Medical Center, Birmingham, Alabama
| |
Collapse
|
31
|
Goodman SM, Springer BD, Chen AF, Davis M, Fernandez DR, Figgie M, Finlayson H, George MD, Giles JT, Gilliland J, Klatt B, MacKenzie R, Michaud K, Miller A, Russell L, Sah A, Abdel MP, Johnson B, Mandl LA, Sculco P, Turgunbaev M, Turner AS, Yates A, Singh JA. 2022 American College of Rheumatology/American Association of Hip and Knee Surgeons Guideline for the Perioperative Management of Antirheumatic Medication in Patients With Rheumatic Diseases Undergoing Elective Total Hip or Total Knee Arthroplasty. Arthritis Care Res (Hoboken) 2022; 74:1399-1408. [PMID: 35718887 DOI: 10.1002/acr.24893] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Revised: 03/18/2022] [Accepted: 04/07/2022] [Indexed: 11/11/2022]
Abstract
OBJECTIVE To develop updated guidelines for the perioperative management of disease-modifying medications for patients with rheumatic diseases, specifically those with inflammatory arthritis (IA) and those with systemic lupus erythematosus (SLE), undergoing elective total hip arthroplasty (THA) or elective total knee arthroplasty (TKA). METHODS We convened a panel of rheumatologists, orthopedic surgeons, and infectious disease specialists, updated the systematic literature review, and included currently available medications for the clinically relevant population, intervention, comparator, and outcomes (PICO) questions. We used the Grading of Recommendations Assessment, Development and Evaluation (GRADE) methodology to rate the quality of evidence and the strength of recommendations using a group consensus process. RESULTS This guideline updates the 2017 recommendations for perioperative use of disease-modifying antirheumatic therapy, including traditional disease-modifying antirheumatic drugs, biologic agents, targeted synthetic small-molecule drugs, and glucocorticoids used for adults with rheumatic diseases, specifically for the treatment of patients with IA, including rheumatoid arthritis and spondyloarthritis, those with juvenile idiopathic arthritis, or those with SLE who are undergoing elective THA or TKA. It updates recommendations regarding when to continue, when to withhold, and when to restart these medications and the optimal perioperative dosing of glucocorticoids. CONCLUSION This updated guideline includes recently introduced immunosuppressive medications to help decision-making by clinicians and patients regarding perioperative disease-modifying medication management for patients with IA and SLE at the time of elective THA or TKA.
Collapse
Affiliation(s)
- Susan M Goodman
- Hospital for Special Surgery, Weill Cornell Medicine, New York, New York
| | | | | | | | - David R Fernandez
- Hospital for Special Surgery, Weill Cornell Medicine, New York, New York
| | - Mark Figgie
- Hospital for Special Surgery, Weill Cornell Medicine, New York, New York
| | - Heather Finlayson
- Multispecialty Physician Partners, LLC, Colorado Arthritis Associates, Lakewood, Colorado
| | | | | | - Jeremy Gilliland
- University of Utah and Veterans Affairs Medical Center, Salt Lake City
| | - Brian Klatt
- University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | - Ronald MacKenzie
- Hospital for Special Surgery, Weill Cornell Medicine, New York, New York
| | - Kaleb Michaud
- University of Nebraska Medical Center, Omaha, Nebraska, and Forward Databank, Wichita, Kansas
| | - Andy Miller
- Hospital for Special Surgery, Weill Cornell Medicine, New York, New York
| | - Linda Russell
- Hospital for Special Surgery, Weill Cornell Medicine, New York, New York
| | - Alexander Sah
- Sah Orthopaedic Associates, Institute for Joint Restoration, Freemont, California
| | | | | | - Lisa A Mandl
- Hospital for Special Surgery, Weill Cornell Medicine, New York, New York
| | - Peter Sculco
- Hospital for Special Surgery, Weill Cornell Medicine, New York, New York
| | | | - Amy S Turner
- American College of Rheumatology, Atlanta, Georgia
| | - Adolph Yates
- University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | - Jasvinder A Singh
- University of Alabama at Birmingham and Veterans Affairs Medical Center, Birmingham, Alabama
| |
Collapse
|
32
|
Rituximab in myasthenia gravis: efficacy, associated infections and risk of induced hypogammaglobulinemia. Neuromuscul Disord 2022; 32:664-671. [DOI: 10.1016/j.nmd.2022.06.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Revised: 06/15/2022] [Accepted: 06/17/2022] [Indexed: 11/23/2022]
|
33
|
Redenbaugh V, Flanagan EP. Monoclonal Antibody Therapies Beyond Complement for NMOSD and MOGAD. Neurotherapeutics 2022; 19:808-822. [PMID: 35267170 PMCID: PMC9294102 DOI: 10.1007/s13311-022-01206-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/17/2022] [Indexed: 01/09/2023] Open
Abstract
Aquaporin-4 (AQP4)-IgG seropositive neuromyelitis optica spectrum disorders (AQP4-IgG seropositive NMOSD) and myelin oligodendrocyte glycoprotein (MOG)-IgG-associated disease (MOGAD) are inflammatory demyelinating disorders distinct from each other and from multiple sclerosis (MS).While anti-CD20 treatments can be used to treat MS and AQP4-IgG seropositive NMOSD, some MS medications are ineffective or could exacerbate AQP4-IgG seropositive NMOSD including beta-interferons, natalizumab, and fingolimod. AQP4-IgG seropositive NMOSD has a relapsing course in most cases, and preventative maintenance treatments should be started after the initial attack. Rituximab, eculizumab, inebilizumab, and satralizumab all have class 1 evidence for use in AQP4-IgG seropositive NMOSD, and the latter three have been approved by the US Food and Drug Administration (FDA). MOGAD is much more likely to be monophasic than AQP4-IgG seropositive NMOSD, and preventative therapy is usually reserved for those who have had a disease relapse. There is a lack of any class 1 evidence for MOGAD preventative treatment. Observational benefit has been suggested from oral immunosuppressants, intravenous immunoglobulin (IVIg), rituximab, and tocilizumab. Randomized placebo-controlled trials are urgently needed in this area.
Collapse
Affiliation(s)
- Vyanka Redenbaugh
- Department of Neurology, Mayo Clinic College of Medicine, Rochester, MN, 55905, USA
| | - Eoin P Flanagan
- Department of Neurology, Mayo Clinic College of Medicine, Rochester, MN, 55905, USA.
- Department of Laboratory Medicine and Pathology, Mayo Clinic College of Medicine, Rochester, MN, 55905, USA.
| |
Collapse
|
34
|
Krajnc N, Bsteh G, Berger T, Mares J, Hartung HP. Monoclonal Antibodies in the Treatment of Relapsing Multiple Sclerosis: an Overview with Emphasis on Pregnancy, Vaccination, and Risk Management. Neurotherapeutics 2022; 19:753-773. [PMID: 35378683 PMCID: PMC8978776 DOI: 10.1007/s13311-022-01224-9] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/14/2022] [Indexed: 01/10/2023] Open
Abstract
Monoclonal antibodies have become a mainstay in the treatment of patients with relapsing multiple sclerosis (RMS) and provide some benefit to patients with primary progressive MS. They are highly precise by specifically targeting molecules displayed on cells involved in distinct immune mechanisms of MS pathophysiology. They not only differ in the target antigen they recognize but also by the mode of action that generates their therapeutic effect. Natalizumab, an [Formula: see text]4[Formula: see text]1 integrin antagonist, works via binding to cell surface receptors, blocking the interaction with their ligands and, in that way, preventing the migration of leukocytes across the blood-brain barrier. On the other hand, the anti-CD52 monoclonal antibody alemtuzumab and the anti-CD20 monoclonal antibodies rituximab, ocrelizumab, ofatumumab, and ublituximab work via eliminating selected pathogenic cell populations. However, potential adverse effects may be serious and can necessitate treatment discontinuation. Most importantly, those are the risk for (opportunistic) infections, but also secondary autoimmune diseases or malignancies. Monoclonal antibodies also carry the risk of infusion/injection-related reactions, primarily in early phases of treatment. By careful patient selection and monitoring during therapy, the occurrence of these potentially serious adverse effects can be minimized. Monoclonal antibodies are characterized by a relatively long pharmacologic half-life and pharmacodynamic effects, which provides advantages such as permitting infrequent dosing, but also creates disadvantages regarding vaccination and family planning. This review presents an overview of currently available monoclonal antibodies for the treatment of RMS, including their mechanism of action, efficacy and safety profile. Furthermore, we provide practical recommendations for risk management, vaccination, and family planning.
Collapse
Affiliation(s)
- Nik Krajnc
- Department of Neurology, Medical University of Vienna, Vienna, Austria
| | - Gabriel Bsteh
- Department of Neurology, Medical University of Vienna, Vienna, Austria
| | - Thomas Berger
- Department of Neurology, Medical University of Vienna, Vienna, Austria
| | - Jan Mares
- Department of Neurology, Palacky University Olomouc, Olomouc, Czech Republic
| | - Hans-Peter Hartung
- Department of Neurology, Medical University of Vienna, Vienna, Austria.
- Department of Neurology, Palacky University Olomouc, Olomouc, Czech Republic.
- Department of Neurology, Medical Faculty, Heinrich-Heine University, Moorenstrasse 5, 40225, Düsseldorf, Germany.
- Brain and Mind Center, University of Sydney, Sydney, Australia.
| |
Collapse
|
35
|
Arnold J, Vital EM, Dass S, Aslam A, Rawstron AC, Savic S, Emery P, Md Yusof MY. A Personalized Rituximab Retreatment Approach Based on Clinical and B-Cell Biomarkers in ANCA-Associated Vasculitis. Front Immunol 2022; 12:803175. [PMID: 35095887 PMCID: PMC8789753 DOI: 10.3389/fimmu.2021.803175] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Accepted: 12/15/2021] [Indexed: 12/12/2022] Open
Abstract
Background Time to relapse after rituximab for the treatment of antineutrophil cytoplasmic antibody (ANCA)-associated vasculitis (AAV) is variable, and optimal retreatment strategy has remained unclear. In AAV following rituximab induction, the study objective was to evaluate clinical and B-cell predictors of relapse in order to develop a retreatment algorithm. Methods A retrospective observational study was conducted in 70 rituximab-treated ANCA-associated vasculitis patients followed up for over 10 years. Complete response (CR) was defined as Birmingham Vasculitis Activity Score v3.0 = 0. Retreatment was given on clinical relapse, defined as new features or worsening of persistent disease (not by biomarker status). Peripheral B-cell subsets were measured using highly sensitive flow cytometry. Predictors were tested using multivariable Cox regression. Results Median time to retreatment for cycles 1–5 were 84, 73, 67, 60, and 73 weeks. Over 467 patient-years follow-up, 158 relapses occurred in 60 patients; 16 (in 15 patients) were major (renal = 7, neurological = 4, ENT = 3, and respiratory = 2). The major-relapse rate was 3.4/100 patient-years. In multivariable analysis, concomitant immunosuppressant [HR, 0.48 (95% CI, 0.24–0.94)], achieving CR [0.24 (0.12–0.50)], and naïve B-cell repopulation at 6 months [0.43 (0.22–0.84)] were associated with longer time to relapse. Personalized retreatment using these three predictors in this cohort would have avoided an unnecessary fixed retreatment in 24% of patients. Area under the receiver operating characteristic for prediction of time to relapse was greater if guided by naïve B-cell repopulation than if previously evaluated ANCA and/or CD19+ cells return at 6 months had been used, 0.82 and 0.53, respectively. Conclusion Our findings suggest that all patients should be coprescribed oral immunosuppressant. Those with incomplete response or with absent naïve B cells should be retreated at 6 months. Patients with complete response and naïve repopulation should not receive fixed retreatment. This algorithm could reduce unnecessary retreatment and warrant investigation in clinical trials.
Collapse
Affiliation(s)
- Jack Arnold
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Chapel Allerton Hospital, Leeds, United Kingdom
| | - Edward M Vital
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Chapel Allerton Hospital, Leeds, United Kingdom.,National Institute for Health Research (NIHR) Leeds Biomedical Research Centre, Leeds Teaching Hospitals NHS Trust, Leeds, United Kingdom
| | - Shouvik Dass
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Chapel Allerton Hospital, Leeds, United Kingdom.,National Institute for Health Research (NIHR) Leeds Biomedical Research Centre, Leeds Teaching Hospitals NHS Trust, Leeds, United Kingdom
| | - Aamir Aslam
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Chapel Allerton Hospital, Leeds, United Kingdom.,National Institute for Health Research (NIHR) Leeds Biomedical Research Centre, Leeds Teaching Hospitals NHS Trust, Leeds, United Kingdom
| | - Andy C Rawstron
- Haematological Malignancy Diagnostic Service, Leeds Teaching Hospitals NHS Trust, Leeds, United Kingdom
| | - Sinisa Savic
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Chapel Allerton Hospital, Leeds, United Kingdom.,National Institute for Health Research (NIHR) Leeds Biomedical Research Centre, Leeds Teaching Hospitals NHS Trust, Leeds, United Kingdom
| | - Paul Emery
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Chapel Allerton Hospital, Leeds, United Kingdom.,National Institute for Health Research (NIHR) Leeds Biomedical Research Centre, Leeds Teaching Hospitals NHS Trust, Leeds, United Kingdom
| | - Md Yuzaiful Md Yusof
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Chapel Allerton Hospital, Leeds, United Kingdom.,National Institute for Health Research (NIHR) Leeds Biomedical Research Centre, Leeds Teaching Hospitals NHS Trust, Leeds, United Kingdom
| |
Collapse
|
36
|
Chisari CG, Sgarlata E, Arena S, Toscano S, Luca M, Patti F. Rituximab for the treatment of multiple sclerosis: a review. J Neurol 2022; 269:159-183. [PMID: 33416999 PMCID: PMC7790722 DOI: 10.1007/s00415-020-10362-z] [Citation(s) in RCA: 97] [Impact Index Per Article: 32.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Revised: 12/03/2020] [Accepted: 12/08/2020] [Indexed: 01/07/2023]
Abstract
In the last decades, evidence suggesting the direct or indirect involvement of B cells on multiple sclerosis (MS) pathogenesis has accumulated. The increased amount of data on the efficacy and safety of B-cell-depleting therapies from several studies has suggested the addition of these drugs as treatment options to the current armamentarium of disease modifying therapies (DMTs) for MS. Particularly, rituximab (RTX), a chimeric monoclonal antibody directed at CD20 positive B lymphocytes resulting in cell-mediated apoptosis, has been demonstrated to reduce inflammatory activity, incidence of relapses and new brain lesions on magnetic resonance imaging (MRI) in patients with relapsing-remitting MS (RRMS). Additional evidence also demonstrated that patients with progressive MS (PMS) may benefit from RTX, which also showed to be well tolerated, with acceptable safety risks and favorable cost-effectiveness profile.Despite these encouraging results, RTX is currently approved for non-Hodgkin's lymphoma, chronic lymphocytic leukemia, several forms of vasculitis and rheumatoid arthritis, while it can only be administered off-label for MS treatment. Between Northern European countries exist different rules for using not licensed drug for treating MS. The Sweden MS register reports a high rate (53.5%) of off-label RTX prescriptions in relation to other annually started DMTs to treat MS patients, while Danish and Norwegian neurologists have to use other anti-CD20 drugs, as ocrelizumab, in most of the cases.In this paper, we review the pharmacokinetics, pharmacodynamics, clinical efficacy, safety profile and cost effectiveness aspects of RTX for the treatment of MS. Particularly, with the approval of new anti-CD20 DMTs, the recent worldwide COVID-19 emergency and the possible increased risk of infection with this class of drugs, this review sheds light on the use of RTX as an alternative treatment option for MS management, while commenting the gaps of knowledge regarding this drug.
Collapse
Affiliation(s)
- Clara Grazia Chisari
- Department "GF Ingrassia", Section of Neurosciences, University of Catania, Catania, Italy
| | - Eleonora Sgarlata
- Department "GF Ingrassia", Section of Neurosciences, University of Catania, Catania, Italy
- Stroke Unit, Department of Medicine, Umberto I Hospital, Siracusa, Italy
| | - Sebastiano Arena
- Department "GF Ingrassia", Section of Neurosciences, University of Catania, Catania, Italy
| | - Simona Toscano
- Department "GF Ingrassia", Section of Neurosciences, University of Catania, Catania, Italy
| | - Maria Luca
- Department "GF Ingrassia", Section of Neurosciences, University of Catania, Catania, Italy
| | - Francesco Patti
- Department "GF Ingrassia", Section of Neurosciences, University of Catania, Catania, Italy.
| |
Collapse
|
37
|
Mathew J, Nair A, Chandhu AS, Zafar M, Vinodini G, Yadav B, Padiyar S, Ganapati A. Rituximab and COVID-19 infection in patients with autoimmune rheumatic diseases – A real-world study from India. INDIAN JOURNAL OF RHEUMATOLOGY 2022. [DOI: 10.4103/injr.injr_136_22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/25/2023] Open
|
38
|
Magliulo D, Wade SD, Kyttaris VC. Immunogenicity of SARS-CoV-2 vaccination in rituximab-treated patients: Effect of timing and immunologic parameters. Clin Immunol 2021; 234:108897. [PMID: 34848357 PMCID: PMC8627008 DOI: 10.1016/j.clim.2021.108897] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2021] [Revised: 11/24/2021] [Accepted: 11/24/2021] [Indexed: 12/12/2022]
Abstract
Rituximab (RTX), an important therapeutic option for patients with rheumatic diseases, has been shown to reduce immune responses to various vaccines. We asked whether following SARS-CoV-2 vaccination, response rates in RTX treated patients are reduced and whether specific patient characteristics influence the responses. We recruited patients on chronic RTX therapy undergoing anti-SARS-CoV2 vaccination and measured the post-vaccination anti-spike IgG antibody levels. The median time from pre-vaccination RTX infusion to vaccination and from vaccination to the post-vaccination RTX infusion was 20.5 weeks and 7.2 weeks respectively. Only 36.5% of patients developed measurable titers of IgG anti-SARS-CoV-2 spike antibody after vaccination. Hypogammaglobulinemia (IgG and/or IgM) but not timing of vaccination, B cell numbers, or concomitant immune suppressive medications, correlated with sero-negativity (p = 0.004). Our results underscore the fact that even after B cell reconstitution, RTX induced chronic hypogammaglobulinemia significantly impairs the ability of the immune system to respond to SARS-CoV-2 vaccination.
Collapse
Affiliation(s)
- Daniel Magliulo
- Division of Rheumatology, Beth Israel Deaconess Medical Center, Boston, MA, United States of America; Harvard Medical School, Boston, MA, United States of America
| | - Stefanie D Wade
- Division of Rheumatology, Beth Israel Deaconess Medical Center, Boston, MA, United States of America; Harvard Medical School, Boston, MA, United States of America
| | - Vasileios C Kyttaris
- Division of Rheumatology, Beth Israel Deaconess Medical Center, Boston, MA, United States of America; Harvard Medical School, Boston, MA, United States of America.
| |
Collapse
|
39
|
Smets I, Giovannoni G. Derisking CD20-therapies for long-term use. Mult Scler Relat Disord 2021; 57:103418. [PMID: 34902761 DOI: 10.1016/j.msard.2021.103418] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2021] [Revised: 09/15/2021] [Accepted: 11/20/2021] [Indexed: 11/16/2022]
Abstract
Anti-CD20 have quickly become the mainstay in the treatment of multiple sclerosis (MS) and other neuroinflammatory conditions. However, when they are used as a maintenance therapy the balance between risks and benefits changes. In this review, we suggested six steps to derisk anti-CD20. Firstly and secondly, adequate infectious screening followed by vaccinations before starting anti-CD20 are paramount. Third, family planning needs to be discussed upfront with every woman of childbearing age. Fourth, infusion reactions should be adequately managed to avoid treatment interruption. After repeated infusions, it becomes important to detect and prevent anti-CD20-related adverse events. Fifth, we recommended measuring immunoglobulin levels and reviewing vaccinations annually as well as counselling adequate fever management. For female patients, we emphasised the importance to engage with the local breast cancer screening programs. Sixth, to fundamentally derisk anti-CD20 therapies, we need evidence-based approaches to reduce dosing intervals and guide retreatment.
Collapse
Affiliation(s)
- Ide Smets
- Blizard Institute, Centre for Neuroscience, Surgery and Trauma, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, 4 Newark St, Whitechapel, London E1 2AT, United Kingdom; Clinical Board Medicine (Neuroscience), Royal London Hospital, Barts Health NHS Trust, Whitechapel Road, London E1 1FR, United Kingdom
| | - Gavin Giovannoni
- Blizard Institute, Centre for Neuroscience, Surgery and Trauma, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, 4 Newark St, Whitechapel, London E1 2AT, United Kingdom; Clinical Board Medicine (Neuroscience), Royal London Hospital, Barts Health NHS Trust, Whitechapel Road, London E1 1FR, United Kingdom.
| |
Collapse
|
40
|
Szepanowski F, Warnke C, Meyer Zu Hörste G, Mausberg AK, Hartung HP, Kleinschnitz C, Stettner M. Secondary Immunodeficiency and Risk of Infection Following Immune Therapies in Neurology. CNS Drugs 2021; 35:1173-1188. [PMID: 34657228 PMCID: PMC8520462 DOI: 10.1007/s40263-021-00863-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/13/2021] [Indexed: 12/13/2022]
Abstract
Secondary immunodeficiencies (SIDs) are acquired conditions that may occur as sequelae of immune therapy. In recent years a number of disease-modifying therapies (DMTs) has been approved for multiple sclerosis and related disorders such as neuromyelitis optica spectrum disorders, some of which are frequently also used in- or off-label to treat conditions such as chronic inflammatory demyelinating polyneuropathy (CIDP), myasthenia gravis, myositis, and encephalitis. In this review, we focus on currently available immune therapeutics in neurology to explore their specific modes of action that might contribute to SID, with particular emphasis on their potential to induce secondary antibody deficiency. Considering evidence from clinical trials as well as long-term observational studies related to the patients' immune status and risks of severe infections, we delineate long-term anti-CD20 therapy, with the greatest data availability for rituximab, as a major risk factor for the development of SID, particularly through secondary antibody deficiency. Alemtuzumab and cladribine have relevant effects on circulating B-cell counts; however, evidence for SID mediated by antibody deficiency appears limited and urgently warrants further systematic evaluation. To date, there has been no evidence suggesting that treatment with fingolimod, dimethyl fumarate, or natalizumab leads to antibody deficiency. Risk factors predisposing to development of SID include duration of therapy, increasing age, and pre-existing low immunoglobulin (Ig) levels. Prevention strategies of SID comprise awareness of risk factors, individualized treatment protocols, and vaccination concepts. Immune supplementation employing Ig replacement therapy might reduce morbidity and mortality associated with SIDs in neurological conditions. In light of the broad range of existing and emerging therapies, the potential for SID warrants urgent consideration among neurologists and other healthcare professionals.
Collapse
Affiliation(s)
- Fabian Szepanowski
- Department of Neurology and Center for Translational Neuro- and Behavioral Sciences (C-TNBS), University Medicine Essen, University of Duisburg-Essen, Hufelandstraße 55, 45147, Essen, Germany
| | - Clemens Warnke
- Department of Neurology, University of Cologne, Cologne, Germany
| | | | - Anne K Mausberg
- Department of Neurology and Center for Translational Neuro- and Behavioral Sciences (C-TNBS), University Medicine Essen, University of Duisburg-Essen, Hufelandstraße 55, 45147, Essen, Germany
| | - Hans-Peter Hartung
- Department of Neurology, Medical Faculty, University of Duesseldorf, Duesseldorf, Germany
- Brain and Mind Centre, University of Sydney, Sydney, NSW, Australia
- Medical University Vienna, Vienna, Austria
- Department of Neurology, Palacky University, Olomouc, Czech Republic
| | - Christoph Kleinschnitz
- Department of Neurology and Center for Translational Neuro- and Behavioral Sciences (C-TNBS), University Medicine Essen, University of Duisburg-Essen, Hufelandstraße 55, 45147, Essen, Germany
| | - Mark Stettner
- Department of Neurology and Center for Translational Neuro- and Behavioral Sciences (C-TNBS), University Medicine Essen, University of Duisburg-Essen, Hufelandstraße 55, 45147, Essen, Germany.
| |
Collapse
|
41
|
Evangelatos G, Fragoulis GE, Klavdianou K, Moschopoulou M, Vassilopoulos D, Iliopoulos A. Hypogammaglobulinemia after rituximab for rheumatoid arthritis is not rare and is related with good response: 13 years real-life experience. Rheumatology (Oxford) 2021; 60:2375-2382. [PMID: 33175958 DOI: 10.1093/rheumatology/keaa617] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Revised: 08/24/2020] [Indexed: 12/15/2022] Open
Abstract
OBJECTIVES Rituximab (RTX) use in the treatment of RA can be complicated by decrease in IgG, IgM or IgA levels (hypogammaglobulinemia-HGG). The aim of this study was to define the frequency of HGG in RA patients treated with RTX and to identify associations between its occurrence and patients' characteristics, disease outcomes and serious infections rate. METHODS RA patients treated with RTX in two rheumatology centers from January 2007 to January 2020 were retrospectively examined. Demographical, clinical and laboratory parameters were recorded at baseline and at last visit. RESULTS Eighty-three patients (84.3% females) with a mean age of 63.2 years were enrolled. They had baseline DAS28(CRP) of 5.2 (1.1) and received a median (range) of 8 (2-20) RTX cycles. A total of 43.4%, 24.1% and 31.3% developed 'any HGG', 'low IgG' and 'low IgM', respectively. Lower baseline IgG and IgM levels were predictors of 'low IgG' and 'low IgM' occurrence, respectively. Patients who developed 'low IgM' exhibited lower DAS28(CRP) and increased rates of remission and low disease activity compared with those with normal IgM levels. Patients who maintained normal IgG were receiving methotrexate more frequently. No differences were observed in serious infections rate among subgroups. CONCLUSION HGG occurred in 43% of RTX-treated patients. Patients who developed low IgG or low IgM had lower baseline levels than those who did not. Concomitant DMARD and corticosteroid therapy was not associated with HGG. Low IgM, but not low IgG, development was associated with better disease outcomes. HGG was not associated with an increased incidence of serious infections.
Collapse
Affiliation(s)
- Gerasimos Evangelatos
- Rheumatology Department, 417 Army Share Fund Hospital (NIMTS).,Rheumatology Unit, First Department of Propaedeutic Internal Medicine, School of Medicine, National and Kapodistrian University of Athens
| | - George E Fragoulis
- Rheumatology Unit, First Department of Propaedeutic Internal Medicine, School of Medicine, National and Kapodistrian University of Athens
| | - Kalliopi Klavdianou
- Department of Rheumatology, 'Asklepieion' General Hospital.,Joint Rheumatology Program, Clinical Immunology-Rheumatology Unit, 2nd Department of Medicine and Laboratory, School of Medicine, National and Kapodistrian University of Athens, Hippokration General Hospital
| | - Melina Moschopoulou
- Faculty of Pharmacy, National and Kapodistrian University of Athens, Athens, Greece
| | - Dimitrios Vassilopoulos
- Joint Rheumatology Program, Clinical Immunology-Rheumatology Unit, 2nd Department of Medicine and Laboratory, School of Medicine, National and Kapodistrian University of Athens, Hippokration General Hospital
| | | |
Collapse
|
42
|
Wade SD, Kyttaris VC. Rituximab-associated hypogammaglobulinemia in autoimmune rheumatic diseases: a single-center retrospective cohort study. Rheumatol Int 2021; 41:1115-1124. [PMID: 33811499 PMCID: PMC8019084 DOI: 10.1007/s00296-021-04847-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Accepted: 03/22/2021] [Indexed: 12/22/2022]
Abstract
B-cell targeted therapies, such as rituximab (RTX), are used widely in autoimmune rheumatic diseases (AIRD). RTX can cause hypogammaglobulinemia and predispose patients to infections. Herein, we asked whether the underlying diagnosis influences the risk for hypogammaglobulinemia in patients treated with RTX. All patients who received RTX infusions and carried a diagnosis of rheumatoid arthritis (RA), ANCA-associated vasculitis (AAV), or connective tissue disease (CTD) were included in this single-center retrospective cohort study. We used STATA® for analysis: Chi-square test was used for comparing categorical variables. Based on distribution, continuous variables were compared using the t test/ANOVA or the Wilcoxon/Kruskal-Wallis tests. Of the 163 patients who received RTX for an AIRD, 60 with pre- and post- RTX immunoglobulins were analyzed. A higher incidence of post-treatment hypogammaglobulinemia was seen in AAV (45%) compared to RA (22%) and CTD (9.1%) groups (p = 0.03). Glucocorticoid exposure of 10 mg or more was identified as a significant risk factor for hypogammaglobulinemia. Finally, we observed a higher number of clinically significant infections per person in the AAV group than in the RA and CTD groups. We observed an increased incidence of hypogammaglobulinemia in the RTX-treated AAV group, with almost half of patients developing post-RTX hypogammaglobulinemia. The rate of infections per person was highest in the AAV group. Screening immunoglobulins were not consistently measured pre- and post-RTX. Results highlight a need for increased awareness of the role of immunoglobulin measurement before maintenance doses of RTX, especially in patients with AAV and steroid exposure.
Collapse
Affiliation(s)
- Stefanie D Wade
- Division of Rheumatology and Clinical Immunology, Harvard Medical School, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Vasileios C Kyttaris
- Division of Rheumatology and Clinical Immunology, Harvard Medical School, Beth Israel Deaconess Medical Center, Boston, MA, USA.
| |
Collapse
|
43
|
Durmaz Y, Ilhanli I. Six month assessment of low dose rituximab in the treatment of rheumatoid arthritis during coronavirus disease 2019 (COVID-19) pandemic. THE EGYPTIAN RHEUMATOLOGIST 2021; 43:253-256. [PMID: 36277424 PMCID: PMC8101793 DOI: 10.1016/j.ejr.2021.05.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 05/01/2021] [Accepted: 05/03/2021] [Indexed: 11/22/2022]
Abstract
Aim of the work To evaluate the 6-month treatment responses to low dose rituximab (LDR) compared to standard dose rituximab (SDR) in rheumatoid arthritis (RA) patients whose treatments were disrupted due to the pandemic with increased disease activity and to examine the effect of LDR treatment on serum immunoglobulin (Ig) levels. Patients and methods Records were retrospectively analysed for 80 patients on SDR not admitted to the hospital due to fear of infection during pandemic, with increased disease activity and were resumed on LDR (500 mg intravenous RTX-infusion twice with 15 days intervals, and repeated for the second time in all patients after 6 months). Disease activity score (DAS-28) values were obtained. The Ig levels of the patients before and after rituximab treatment were calculated. Results The mean age of patients was 55.1 ± 13.1 years. They were 46 (57.5%) female and 34 (42.5%) male (F:M 1.4:1) with median disease duration of 13 (0.5-50) years. After the second dose of LDR, there was a significant decrease in the disease activity DAS28 (6.5 ± 1.01 to 3.2 ± 1.2, p < 0.0001) and acute phase reactants with a tendency to decrease in Ig levels. After LDR, 6 (7.5%) patients developed COVID-19 infection that did not require hospitalization. There was no difference between the Ig levels of patients with and without COVID-19 infection. Conclusions LDR is an effective treatment option in the treatment of RA. In our study, none of our patients developed severe COVID-19 infection requiring hospitalization, and LDR may be preferred during the COVID-19 pandemic period.
Collapse
Affiliation(s)
- Yunus Durmaz
- Division of Rheumatology, Department of Physical Medicine and Rehabilitation, Karabuk Training and Research Hospital, Karabuk, Turkey
| | - Ilker Ilhanli
- Department of Physical Medicine and Rehabilitation, Faculty of Medicine, Ondokuz Mayıs University, Samsun, Turkey
| |
Collapse
|
44
|
Smith TE, Kister I. Infection Mitigation Strategies for Multiple Sclerosis Patients on Oral and Monoclonal Disease-Modifying Therapies. Curr Neurol Neurosci Rep 2021; 21:36. [PMID: 34009478 PMCID: PMC8132488 DOI: 10.1007/s11910-021-01117-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/22/2021] [Indexed: 12/17/2022]
Abstract
PURPOSE OF REVIEW The newer, higher-efficacy disease-modifying therapies (DMTs) for multiple sclerosis (MS)-orals and monoclonals-have more profound immunomodulatory and immunosuppressive properties than the older, injectable therapies and require risk mitigation strategies to reduce the risk of serious infections. This review will provide a systematic framework for infectious risk mitigation strategies relevant to these therapies. RECENT FINDINGS We classify risk mitigation strategies according to the following framework: (1) screening and patient selection, (2) vaccinations, (3) antibiotic prophylaxis, (4) laboratory and MRI monitoring, (5) adjusting dose and frequency of DMT, and (6) behavioral modifications to limit the risk of infection. We systematically apply this framework to the infections for which risk mitigations are available: hepatitis B, herpetic infections, progressive multifocal leukoencephalopathy, and tuberculosis. We also discuss up-to-date recommendations regarding COVID-19 vaccinations for patients on DMTs. We offer a practical, comprehensive, DMT-specific framework of derisking strategies designed to minimize the risk of infections associated with the newer MS therapies.
Collapse
Affiliation(s)
- Tyler Ellis Smith
- Department of Neurology, NYU-Multiple Sclerosis Care Center, NYU School of Medicine, New York, NY, USA.
- , New York, NY, USA.
| | - Ilya Kister
- Department of Neurology, NYU-Multiple Sclerosis Care Center, NYU School of Medicine, New York, NY, USA
| |
Collapse
|
45
|
Jones JM, Faruqi AJ, Sullivan JK, Calabrese C, Calabrese LH. COVID-19 Outcomes in Patients Undergoing B Cell Depletion Therapy and Those with Humoral Immunodeficiency States: A Scoping Review. Pathog Immun 2021; 6:76-103. [PMID: 34056149 PMCID: PMC8150936 DOI: 10.20411/pai.v6i1.435] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Accepted: 04/26/2021] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND The role of humoral immunity has been well established in reducing infection risk and facilitating viral clearance in patients with COVID-19. However, the relationship between specific antibody responses and severity of COVID-19 is less well understood. METHODS To address this question and identify gaps in knowledge, we utilized the methodology of a scoping review to interrogate risk of infection and clinical outcomes of COVID-19 in patients with iatrogenic and inborn humoral immunodeficiency states based on existing literature. RESULTS Among patients with iatrogenic B-cell depletion, particularly with agents targeting CD20, our analysis found increased risk of severe COVID-19 and death across a range of underlying disease states. Among patients with humoral inborn errors of immunity with COVID-19, our synthesis found that patients with dysregulated humoral immunity, predominantly common variable immunodeficiency (CVID), may be more susceptible to severe COVID-19 than patients with humoral immunodeficiency states due to X-linked agammaglobulinemia and other miscellaneous forms of humoral immunodeficiency. There were insufficient data to appraise the risk of COVID-19 infection in both populations of patients. CONCLUSIONS Our work identifies potentially significant predictors of COVID-19 severity in patients with humoral immunodeficiency states and highlights the need for larger studies to control for clinical and biologic confounders of disease severity.
Collapse
Affiliation(s)
- Jessica M. Jones
- Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, Ohio
| | - Aiman J. Faruqi
- Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, Ohio
| | - James K. Sullivan
- Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, Ohio
| | - Cassandra Calabrese
- Cleveland Clinic, Department of Rheumatic and Immunologic Diseases, Cleveland, Ohio
| | - Leonard H. Calabrese
- Cleveland Clinic, Department of Rheumatic and Immunologic Diseases, Cleveland, Ohio
| |
Collapse
|
46
|
Kaestner S, Wolff HJ, Braeuer R, Kaestner P. Immunoglobulin deficiencies in treated patients suffering from rheumatoid arthritis. Rheumatol Int 2021; 41:1273-1280. [PMID: 33914121 DOI: 10.1007/s00296-021-04871-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Accepted: 04/15/2021] [Indexed: 11/25/2022]
Abstract
Immunoglobulins and antibodies to immunoglobulins (autoimmunoglobulins) have been identified to be implicated in the pathogenesis of rheumatoid arthritis (RA). Immunoglobulin deficiencies have been suggested to account for the increased risk of infections in RA patients. This study was carried out to determine the prevalence of immunoglobulin deficiencies in patients with RA and the identification of putative contributing factors. Immunoglobulin levels in blood samples of patients with rheumatoid arthritis were evaluated by an immunonephelometric assay. Demographic and disease related data (including age, sex, smoking habits, disease duration and activity, inflammatory markers) were assessed, and associations were identified by regression analysis. 539 patients were enrolled between 2011 and 2013. The most common immunoglobulin (Ig) deficiencies were those of IgM (24.5%) and IgG (19.9%). Most frequent deficiencies of subclasses were observed for IgG1 (42.3%), followed by IgG4 (10.4%), IgG2 (7.2%), and IgG3 (5.4%). Regression analyses revealed that deficiencies of IgM, IgG, IgG1, and IgG2 were more prevalent in older patients. In addition, smoking was associated with IgG2 deficiency, and IgA deficiency was associated with female sex. Occurrence of infections was significantly increased in patients with IgG, IgG2, and IgG4 deficiencies. RA patients displayed high rates of IgG and IgM deficiencies. In consequence, the assessment of immunoglobulin status should precede the application of immune modulating drugs to prevent a potential risk of infectious diseases. Prospective studies are needed to investigate the influence of immune modulating drugs on IgG and IgG subclass levels.
Collapse
Affiliation(s)
- Sara Kaestner
- Institute of Pathology, University Hospital Jena, 07743, Jena, Germany.
- Ambulantes Rheumazentrum Erfurt, Tschaikowskistraße 22, 99096, Erfurt, Germany.
| | - Hans-Juergen Wolff
- Ambulantes Rheumazentrum Erfurt, Tschaikowskistraße 22, 99096, Erfurt, Germany
| | - Rolf Braeuer
- Institute of Pathology, University Hospital Jena, 07743, Jena, Germany
| | - Peter Kaestner
- Ambulantes Rheumazentrum Erfurt, Tschaikowskistraße 22, 99096, Erfurt, Germany
| |
Collapse
|
47
|
Rituximab-induced hypogammaglobulinemia and infection risk in pediatric patients. J Allergy Clin Immunol 2021; 148:523-532.e8. [PMID: 33862010 DOI: 10.1016/j.jaci.2021.03.041] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Revised: 03/03/2021] [Accepted: 03/05/2021] [Indexed: 11/18/2022]
Abstract
BACKGROUND Rituximab is a B-cell depleting agent used in B-cell malignancies and autoimmune diseases. A subset of adult patients may develop prolonged and symptomatic hypogammaglobulinemia following rituximab treatment. However, this phenomenon has not been well delineated in the pediatric population. OBJECTIVES This study sought to determine the prevalence, risk factors, and clinical significance of hypogammaglobulinemia following rituximab therapy in children. METHODS This was a multicenter, retrospective cohort study that extracted clinical and immunological data from pediatric patients who received rituximab. RESULTS The cohort comprised 207 patients (median age, 12.0 years). Compared to baseline values, there was a significant increase in hypogammaglobulinemia post-rituximab therapy, with an increase in prevalence of hypo-IgG (28.7%-42.6%; P = .009), hypo-IgA (11.1%-20.4%; P = .02), and hypo-IgM (20.0%-62.0%; P < .0001). Additionally, low IgG levels at any time post-rituximab therapy were associated with a higher risk of serious infections (34.4% vs 18.9%; odds ratio, 2.3; 95% CI, 1.1-4.8; P = .03). Persistent IgG hypogammaglobulinemia was observed in 27 of 101 evaluable patients (26.7%). Significant risk factors for persistent IgG hypogammaglobulinemia included low IgG and IgA levels pre-rituximab therapy. Nine patients (4.3%) within the study were subsequently diagnosed with a primary immunodeficiency, 7 of which received rituximab for autoimmune cytopenias. CONCLUSIONS Hypogammaglobulinemia post-rituximab treatment is frequently diagnosed within the pediatric population. Low IgG levels are associated with a significant increase in serious infections, and underlying primary immunodeficiencies are relatively common in children receiving rituximab, thus highlighting the importance of immunologic monitoring both before and after rituximab therapy.
Collapse
|
48
|
Descamps E, Gorlier C, Ottaviani S, Palazzo E, Dieudé P, Forien M. Screening of dental and sinus infections in rheumatoid arthritis. Eur J Clin Invest 2021; 51:e13437. [PMID: 33089506 DOI: 10.1111/eci.13437] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Revised: 10/02/2020] [Accepted: 10/18/2020] [Indexed: 11/29/2022]
Abstract
BACKGROUND Rheumatoid arthritis (RA) is associated with increased risk of infections. Screening for oral (dental and/or sinus) infection could be proposed before biologic disease-modifying antirheumatic drugs (bDMARDs) initiation but is not systematically recommended. The aim of our study was to assess the prevalence of oral infection in RA patients requiring bDMARDs. MATERIALS AND METHODS This was a monocentric retrospective study. We included patients with RA and active disease requiring bDMARDs. Dental infection and sinusitis were assessed by a stomatologist and otorhinolaryngologist after clinical, panoramic dental X-ray and sinus CT evaluation. Factors associated with oral infections were analysed in uni- and multivariate models, estimating odds ratios (ORs) and 95% confidence intervals (CIs). RESULTS We included 223 RA patients (79.4% women, mean disease duration 8.9 ± 8.6 years). The mean age was 54.4 ± 10.9 years and mean Disease Activity Score in 28 joints 5.5 ± 2.6. Systematic dental screening revealed infection requiring treatment before bDMARDs initiation in 46 (20.9%) patients. Sinusitis was diagnosed by the otorhinolaryngologist in 33 (14.8%) patients. Among the 223 patients, 69 (30.9%) had dental and/or sinus infection. On univariate analysis, active smoking was associated with increased probability of oral infection (OR = 2.16 [95% CI 1.02-4.57], P = .038) and methotrexate with reduced probability (OR = 0.43 [95% CI 0.23-0.81], P = .006). On multivariate analysis, no RA variables were associated with oral infection. CONCLUSION In our study, asymptomatic oral infection was confirmed in one third of RA patients.
Collapse
Affiliation(s)
- Elise Descamps
- Rheumatology Department, DMU Locomotion, Bichat Hospital (APHP), Paris, France
| | - Clémence Gorlier
- Rheumatology Department, DMU Locomotion, Bichat Hospital (APHP), Paris, France
| | - Sébastien Ottaviani
- Rheumatology Department, DMU Locomotion, Bichat Hospital (APHP), Paris, France
| | - Elisabeth Palazzo
- Rheumatology Department, DMU Locomotion, Bichat Hospital (APHP), Paris, France
| | - Philippe Dieudé
- Rheumatology Department, DMU Locomotion, Bichat Hospital (APHP), Paris, France
| | - Marine Forien
- Rheumatology Department, DMU Locomotion, Bichat Hospital (APHP), Paris, France
| |
Collapse
|
49
|
Avouac A, Maarouf A, Stellmann JP, Rico A, Boutiere C, Demortiere S, Marignier R, Pelletier J, Audoin B. Rituximab-Induced Hypogammaglobulinemia and Infections in AQP4 and MOG Antibody-Associated Diseases. NEUROLOGY-NEUROIMMUNOLOGY & NEUROINFLAMMATION 2021; 8:8/3/e977. [PMID: 33722933 PMCID: PMC8054961 DOI: 10.1212/nxi.0000000000000977] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Accepted: 02/05/2021] [Indexed: 02/06/2023]
Abstract
Objective To determine the potential association between infections and rituximab (RTX)-induced hypogammaglobulinemia among patients with CNS inflammatory diseases. Methods We included in a prospective observational study all consecutive adults with aquaporin 4 (AQP4) or myelin oligodendrocyte glycoprotein (MOG) antibody–positive disorders treated with RTX. Dosing schedule was adapted to memory B-cell measurement. Results We included 48 patients (mean age 47 [SD: 14] years; 77% females; 31 AQP4 positive and 17 MOG positive). The median follow-up was 3.6 years (range: 0.9–8.1 years). The median number of RTX infusions was 8 (range: 2–14). The median dosing interval was 6 months (range: 1.7–13.7 months). Sixty-seven symptomatic infections (SIs) were observed in 26 of 48 (54%) patients, including 13 severe infections in 9 (19%). Urinary and lower respiratory tract infections were the most frequent, representing 42% and 21% of SI. At RTX onset, the immunoglobulin G (IgG) level was abnormal in 3 of 48 (6%) patients. After RTX, 15 (31%), 11 (23%), 3 (6%), and 0 of 48 patients showed sustained IgG level <7, <6, <4, and <2 g/L, respectively. On multivariate Cox proportional hazards analysis, the main variables explaining the risk of SI were the presence of urinary tract dysfunction (hazard ratio [HR] = 34, 95% CI 4–262, p < 0.001), the dosing intervals (HR = 0.98, 95% CI 0.97–0.99, p < 0.001), and the interaction between IgG level and urinary tract dysfunction (HR = 0.67, 95% CI 0.53–0.85, p < 0.005). IgG level <6 g/L during RTX was associated with male sex (HR = 4, 95% CI 1.4–11.4, p < 0.01) and previous immunosuppression (HR = 3.4, 95% CI 1.2–10, p < 0.05). Conclusions RTX used as maintenance therapy in CNS inflammatory diseases is frequently associated with reduced IgG level and increases the infection risk of the most vulnerable patients.
Collapse
Affiliation(s)
- Alexandre Avouac
- From the Service de Neurologie (A.A., A.M., C.B., S.D., J.P., B.A.), Pôle de Neurosciences Cliniques, and Département de Neuroradiologie (J.-P.S.), Hôpital de la Timone, APHM, Aix Marseille University; and Department of Neurology A (R.M.), Neurologic and Neurosurgical Hospital Pierre Wertheimer, Hospices Civils de Lyon, Bron, France
| | - Adil Maarouf
- From the Service de Neurologie (A.A., A.M., C.B., S.D., J.P., B.A.), Pôle de Neurosciences Cliniques, and Département de Neuroradiologie (J.-P.S.), Hôpital de la Timone, APHM, Aix Marseille University; and Department of Neurology A (R.M.), Neurologic and Neurosurgical Hospital Pierre Wertheimer, Hospices Civils de Lyon, Bron, France
| | - Jan-Patrick Stellmann
- From the Service de Neurologie (A.A., A.M., C.B., S.D., J.P., B.A.), Pôle de Neurosciences Cliniques, and Département de Neuroradiologie (J.-P.S.), Hôpital de la Timone, APHM, Aix Marseille University; and Department of Neurology A (R.M.), Neurologic and Neurosurgical Hospital Pierre Wertheimer, Hospices Civils de Lyon, Bron, France
| | - Audrey Rico
- From the Service de Neurologie (A.A., A.M., C.B., S.D., J.P., B.A.), Pôle de Neurosciences Cliniques, and Département de Neuroradiologie (J.-P.S.), Hôpital de la Timone, APHM, Aix Marseille University; and Department of Neurology A (R.M.), Neurologic and Neurosurgical Hospital Pierre Wertheimer, Hospices Civils de Lyon, Bron, France
| | - Clemence Boutiere
- From the Service de Neurologie (A.A., A.M., C.B., S.D., J.P., B.A.), Pôle de Neurosciences Cliniques, and Département de Neuroradiologie (J.-P.S.), Hôpital de la Timone, APHM, Aix Marseille University; and Department of Neurology A (R.M.), Neurologic and Neurosurgical Hospital Pierre Wertheimer, Hospices Civils de Lyon, Bron, France
| | - Sarah Demortiere
- From the Service de Neurologie (A.A., A.M., C.B., S.D., J.P., B.A.), Pôle de Neurosciences Cliniques, and Département de Neuroradiologie (J.-P.S.), Hôpital de la Timone, APHM, Aix Marseille University; and Department of Neurology A (R.M.), Neurologic and Neurosurgical Hospital Pierre Wertheimer, Hospices Civils de Lyon, Bron, France
| | - Romain Marignier
- From the Service de Neurologie (A.A., A.M., C.B., S.D., J.P., B.A.), Pôle de Neurosciences Cliniques, and Département de Neuroradiologie (J.-P.S.), Hôpital de la Timone, APHM, Aix Marseille University; and Department of Neurology A (R.M.), Neurologic and Neurosurgical Hospital Pierre Wertheimer, Hospices Civils de Lyon, Bron, France
| | - Jean Pelletier
- From the Service de Neurologie (A.A., A.M., C.B., S.D., J.P., B.A.), Pôle de Neurosciences Cliniques, and Département de Neuroradiologie (J.-P.S.), Hôpital de la Timone, APHM, Aix Marseille University; and Department of Neurology A (R.M.), Neurologic and Neurosurgical Hospital Pierre Wertheimer, Hospices Civils de Lyon, Bron, France
| | - Bertrand Audoin
- From the Service de Neurologie (A.A., A.M., C.B., S.D., J.P., B.A.), Pôle de Neurosciences Cliniques, and Département de Neuroradiologie (J.-P.S.), Hôpital de la Timone, APHM, Aix Marseille University; and Department of Neurology A (R.M.), Neurologic and Neurosurgical Hospital Pierre Wertheimer, Hospices Civils de Lyon, Bron, France.
| |
Collapse
|
50
|
Rymarz A, Matyjek A, Sułek-Jakóbczyk M, Mosakowska M, Niemczyk S. Impaired Kidney Function Associated with Increased Risk of Side Effects in Patients with Small Vessel Vasculitis Treated with Rituximab as an Induction Therapy. J Clin Med 2021; 10:jcm10040786. [PMID: 33669267 PMCID: PMC7920022 DOI: 10.3390/jcm10040786] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2020] [Revised: 02/06/2021] [Accepted: 02/08/2021] [Indexed: 11/16/2022] Open
Abstract
Rituximab (RTX), a monoclonal antibody against the CD20 molecule, is used as an induction therapy in the treatment of small vessel vasculitis (SVV). The aim of the study was to evaluate the efficacy and safety of RTX induction therapy for refractory SVV. A retrospective analysis of 20 patients treated with RTX for active SVV (BVAS/WG ≥ 3) was performed to assess the remission rate and the drug-related severe adverse events 6 months after therapy. The mean age of the studied population was 49 ± 13 years (50% female), 90% of which were PR3-ANCA positive. Complete remission was achieved in 85% of patients, and partial remission was achieved in a further 10% within 6 months after RTX infusions. The remission rate was not influenced by kidney function. Adverse events such as infections (25%), a late onset of neutropenia (10%) and severe hypogammaglobulinemia (5%) were noted. The patients who developed adverse events were older (42 ± 11 vs. 57 ± 12 years; p = 0.014) and had a higher serum creatinine level (1.3 mg/dL vs. 3.35 mg/dL; p = 0.044). Patients with a glomerular filtration rate (eGFR) lower than 30 mL/min/1.73 m2 had a nine-fold higher risk of side effects (OR 9.0, 95%CI: 1.14-71.0). In conclusion, RTX was highly effective as an induction therapy in patients with SVV. Advanced kidney failure with an eGFR lower than 30 mL/min/1.73 m2 was one of the risk factors for the occurrence of side effects.
Collapse
|