1
|
Rutter-Locher Z, Kirkham BW, Bannister K, Bennett DL, Buckley CD, Taams LS, Denk F. An interdisciplinary perspective on peripheral drivers of pain in rheumatoid arthritis. Nat Rev Rheumatol 2024; 20:671-682. [PMID: 39242949 DOI: 10.1038/s41584-024-01155-z] [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: 08/05/2024] [Indexed: 09/09/2024]
Abstract
Pain is one of the most debilitating symptoms of rheumatoid arthritis (RA), and yet remains poorly understood, especially when pain occurs in the absence of synovitis. Without active inflammation, experts most often attribute joint pain to central nervous system dysfunction. However, advances in the past 5 years in both immunology and neuroscience research suggest that chronic pain in RA is also driven by a variety of abnormal interactions between peripheral neurons and mediators produced by resident cells in the local joint environment. In this Review, we discuss these novel insights from an interdisciplinary neuro-immune perspective. We outline a potential working model for the peripheral drivers of pain in RA, which includes autoantibodies, resident immune and mesenchymal cells and their interactions with different subtypes of peripheral sensory neurons. We also offer suggestions for how future collaborative research could be designed to accelerate analgesic drug development.
Collapse
Affiliation(s)
- Zoe Rutter-Locher
- Department of Rheumatology, Guy's Hospital, London, UK
- Centre for Inflammation Biology & Cancer Immunology, Department of Inflammation Biology, School of Immunology & Microbial Sciences, King's College London, London, UK
| | | | - Kirsty Bannister
- Wolfson Sensory Pain and Regeneration Centre (SPaRC), King's College London, London, UK
| | - David L Bennett
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| | | | - Leonie S Taams
- Centre for Inflammation Biology & Cancer Immunology, Department of Inflammation Biology, School of Immunology & Microbial Sciences, King's College London, London, UK.
| | - Franziska Denk
- Wolfson Sensory Pain and Regeneration Centre (SPaRC), King's College London, London, UK.
| |
Collapse
|
2
|
Schett G, Nagy G, Krönke G, Mielenz D. B-cell depletion in autoimmune diseases. Ann Rheum Dis 2024; 83:1409-1420. [PMID: 38777374 DOI: 10.1136/ard-2024-225727] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Accepted: 04/23/2024] [Indexed: 05/25/2024]
Abstract
B cells have a pivotal function in the pathogenesis of autoimmune diseases, such as rheumatoid arthritis, multiple sclerosis and systemic lupus erythematosus. In autoimmune disease, B cells orchestrate antigen presentation, cytokine production and autoantibody production, the latter via their differentiation into antibody-secreting plasmablasts and plasma cells. This article addresses the current therapeutic strategies to deplete B cells in order to ameliorate or potentially even cure autoimmune disease. It addresses the main target antigens in the B-cell lineage that are used for therapeutic approaches. Furthermore, it summarises the current evidence for successful treatment of autoimmune disease with monoclonal antibodies targeting B cells and the limitations and challenges of these approaches. Finally, the concept of deep B-cell depletion and immunological reset by chimeric antigen receptor T cells is discussed, as well as the lessons from this approach for better understanding the role of B cells in autoimmune disease.
Collapse
Affiliation(s)
- Georg Schett
- Department of Internal Medicine 3-Rheumatology and Immunology, Friedrich-Alexander-Universität Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
- Deutsches Zentrum Immuntherapie, Friedrich-Alexander-Universität Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
| | - György Nagy
- Division of Rheumatology and Clinical Immunology, Department of Internal Medicine and Oncology, Semmelweis University, Budapest, Hungary, Budapest, Hungary
- Heart and Vascular Center, Semmelweis University, Budapest, Hungary
- Hospital of the Hospitaller Order of Saint John of God, Budapest, Hungary
| | - Gerhard Krönke
- Department of Internal Medicine 3-Rheumatology and Immunology, Friedrich-Alexander-Universität Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
- Deutsches Zentrum Immuntherapie, Friedrich-Alexander-Universität Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
- Department of Rheumatology, Charite, Berlin, Germany
| | - Dirk Mielenz
- Division of Molecular Immunology, Department of Internal Medicine 3, Friedrich-Alexander-Universität Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Bayern, Germany
| |
Collapse
|
3
|
Frazzei G, Cramer SHM, Landewé RBM, Maijer KI, Gerlag DM, Tak PP, de Vries N, van Baarsen LGM, van Vollenhoven RF, Tas SW. The effect of rituximab on patient reported outcomes in the preclinical phase of rheumatoid arthritis: 2 year data from the PRAIRI study. RMD Open 2024; 10:e004622. [PMID: 39424405 PMCID: PMC11492957 DOI: 10.1136/rmdopen-2024-004622] [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: 06/05/2024] [Accepted: 10/04/2024] [Indexed: 10/21/2024] Open
Abstract
OBJECTIVES Early treatment of individuals at risk of developing rheumatoid arthritis (RA-risk) in the preclinical phase has the potential to positively impact both patients and society by preventing disease onset and improving patients' quality of life. The PRAIRI study was a randomised, double-blind, placebo-controlled trial with the B-cell depleting agent rituximab (RTX), which resulted in a significant delay of arthritis development of up to 12 months in seropositive RA-risk individuals. Here, we report our findings on patient-reported outcomes (PROs) in this study population. METHODS Seventy-eight RA-risk individuals were treated with one single dose of either placebo (PBO) or 1000 mg RTX plus 100 mg methylprednisolone (MP) and anti-histamines, regardless of treatment allocation, as co-medication. Data on quality of life were collected at baseline and 1, 4, 6, 12 and 24 months using established PRO questionnaires (visual analogue scale (VAS) pain, health assessment questionnaire disability index (HAQ-DI) score, EuroQol five dimension (EQ-5D) and both physical and mental component score of the 36-item short-form heath survey (SF-36)). RESULTS No significant changes in quality of life over a 2 year follow-up were observed in at-risk individuals treated with RTX compared to PBO given the PRO scores at 24 months (mean difference±SEM: HAQ score=0.07±0.16; EQ-5D=-0.02±0.05; VAS pain=11.11±7.40). Furthermore, no significant effect of treatment on perceived arthritis severity at the time of clinically manifest disease (arthritis) was found. CONCLUSION One single dose of RTX plus MP administered to RA-risk individuals does not have a meaningful and measurable positive effect on PROs after 2 years of follow-up and/or perceived disease severity at the time of arthritis development. TRIAL REGISTRATION NUMBER Trial registered at EU Clinical Trial Register, EudraCT Number: 2009-010955-29 (https://www.clinicaltrialsregister.eu/ctr-search/search?query=Prevention+of+RA+by+B+cell+directed+therapy).
Collapse
Affiliation(s)
- Giulia Frazzei
- Department of Rheumatology and Clinical Immunology, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, The Netherlands
- Amsterdam Rheumatology and Immunology Center, Amsterdam, The Netherlands
| | - Sophie H M Cramer
- Department of Rheumatology and Clinical Immunology, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Robert B M Landewé
- Department of Rheumatology and Clinical Immunology, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, The Netherlands
- Amsterdam Rheumatology and Immunology Center, Amsterdam, The Netherlands
- Rheumatology, Zuyderland MC, Heerlen, The Netherlands
| | - Karen I Maijer
- Dermatology, Tergooi Hospital, Hilversum, The Netherlands
| | - Danielle M Gerlag
- Department of Rheumatology and Clinical Immunology, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, The Netherlands
- UCB Pharma Ltd, Slough, UK
| | - Paul P Tak
- Department of Rheumatology and Clinical Immunology, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, The Netherlands
- Candel Therapeutics, Needham, Massachusetts, USA
- Research and Development, GSK, Stevenage, UK
| | - Niek de Vries
- Department of Rheumatology and Clinical Immunology, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, The Netherlands
- Amsterdam Rheumatology and Immunology Center, Amsterdam, The Netherlands
| | - Lisa G M van Baarsen
- Department of Rheumatology and Clinical Immunology, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, The Netherlands
- Amsterdam Rheumatology and Immunology Center, Amsterdam, The Netherlands
| | - Ronald F van Vollenhoven
- Department of Rheumatology and Clinical Immunology, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, The Netherlands
- Amsterdam Rheumatology and Immunology Center, Amsterdam, The Netherlands
| | - Sander W Tas
- Department of Rheumatology and Clinical Immunology, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, The Netherlands
- Amsterdam Rheumatology and Immunology Center, Amsterdam, The Netherlands
| |
Collapse
|
4
|
De Meyst E, Bertrand D, Joly J, Doumen M, Marchal A, Thelissen M, Neerinckx B, Westhovens R, Verschueren P. Treat-to-target fixed dose rituximab retreatment versus fixed interval retreatment with disease activity-guided rituximab dose optimisation for patients with rheumatoid arthritis: study protocol for a multicentre randomised controlled superiority trial focusing on long-term disease impact (RITUXERA). Trials 2024; 25:681. [PMID: 39407334 PMCID: PMC11476693 DOI: 10.1186/s13063-024-08542-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Accepted: 10/08/2024] [Indexed: 10/20/2024] Open
Abstract
BACKGROUND The optimal retreatment strategy with rituximab for rheumatoid arthritis (RA) remains a point of discussion. Depending on local guidelines, rituximab can either be administered at fixed intervals or when losing disease control, balancing therapeutic effectiveness with drug overexposure. However, treatment based on loss of disease control may significantly affect patients' lives, provoking uncertainty and potentially leading to progressive joint damage. Moreover, as low-dose rituximab proved to be effective in treating RA while decreasing toxicity, drug exposure may be limited by tapering down rituximab doses guided by disease activity. METHODS RITUXERA is a 104-week open-label multicentre randomised controlled superiority trial. In total, 134 patients with RA treated with rituximab will be 1:1 randomised when in need of retreatment (DAS28-CRP ≥ 3.2 with previous rituximab administration at least 24 weeks earlier) to either a treat-to-target-driven fixed dose retreatment strategy (usual care group) or fixed interval disease-activity guided dose optimisation strategy (experimental group). The usual care group will be retreated with fixed rituximab doses (1 × 1000 mg IV) in case of loss of disease control (DAS28-CRP ≥ 3.2). The experimental group will receive a 24-weekly rituximab treatment while tapering down the dose in a decreasing sequence if DAS28-CRP ≤ 3.2: 1 × 1000 mg IV (maximal dose), 1 × 500 mg IV, and 1 × 200 mg IV (minimal dose). If DAS28-CRP exceeds 3.2 at the six-monthly retreatment, patients will receive and remain on the previous effective dose. Study visits are planned every 12 weeks. Primary outcome is the comparison of longitudinal patient-reported disease impact over 104 weeks, measured with the Rheumatoid Arthritis Impact of Disease (RAID) instrument, analysed using a linear mixed model. Main secondary outcome is the comparison of longitudinal disease activity (DAS28-CRP) over 104 weeks. DISCUSSION The RITUXERA trial aims to explore the optimal retreatment strategy with rituximab for RA in terms of long-term patient-reported disease impact, by proposing a fixed interval disease activity-guided dose optimisation strategy as compared to a treat-to-target fixed dose strategy. TRIAL REGISTRATION CTIS 2023-506638-59-01 (registration date: 07 September 2023), ClinicalTrials.gov NCT06003283 (registration date: 17 August 2023).
Collapse
Affiliation(s)
- Elias De Meyst
- KU Leuven, Department of Development and Regeneration, Skeletal Biology and Engineering Research Centre, Herestraat 49, Leuven, 3000, Belgium.
- UZ Leuven, Rheumatology, Leuven, Belgium.
| | - Delphine Bertrand
- KU Leuven, Department of Development and Regeneration, Skeletal Biology and Engineering Research Centre, Herestraat 49, Leuven, 3000, Belgium
| | - Johan Joly
- UZ Leuven, Rheumatology, Leuven, Belgium
| | - Michaël Doumen
- KU Leuven, Department of Development and Regeneration, Skeletal Biology and Engineering Research Centre, Herestraat 49, Leuven, 3000, Belgium
- UZ Leuven, Rheumatology, Leuven, Belgium
| | | | | | - Barbara Neerinckx
- KU Leuven, Department of Development and Regeneration, Skeletal Biology and Engineering Research Centre, Herestraat 49, Leuven, 3000, Belgium
- UZ Leuven, Rheumatology, Leuven, Belgium
| | - René Westhovens
- KU Leuven, Department of Development and Regeneration, Skeletal Biology and Engineering Research Centre, Herestraat 49, Leuven, 3000, Belgium
- UZ Leuven, Rheumatology, Leuven, Belgium
| | - Patrick Verschueren
- KU Leuven, Department of Development and Regeneration, Skeletal Biology and Engineering Research Centre, Herestraat 49, Leuven, 3000, Belgium
- UZ Leuven, Rheumatology, Leuven, Belgium
| |
Collapse
|
5
|
Nakayama Y, Nagata W, Takeuchi Y, Fukui S, Fujita Y, Hosokawa Y, Ueno M, Ono K, Sumitomo S, Tabuchi Y, Nakanishi Y, Saito S, Ikeuchi H, Kawamori K, Sofue H, Doi G, Minami R, Hirota T, Minegishi K, Maeshima K, Motoyama R, Nakamura S, Suzuki S, Nishioka N, Wada TT, Onishi A, Nishimura K, Watanabe R, Yanai R, Kida T, Nishiwaki H, Yajima N, Kaneko Y, Tanaka E, Kawahito Y, Harigai M. Systematic review and meta-analysis for the 2024 update of the Japan College of Rheumatology clinical practice guidelines for the management of rheumatoid arthritis. Mod Rheumatol 2024; 34:1079-1094. [PMID: 38814660 DOI: 10.1093/mr/roae049] [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: 03/05/2024] [Revised: 05/03/2024] [Accepted: 05/15/2024] [Indexed: 05/31/2024]
Abstract
OBJECTIVES The aim of this article is to update evidence on the efficacy and safety of disease-modifying antirheumatic drugs (DMARDs) and provide information to the taskforce for the 2024 update of the Japan College of Rheumatology clinical practice guidelines for the management of rheumatoid arthritis (RA). METHODS We searched various databases for randomised controlled trials on RA published until June 2022, with no language restriction. For each of the 15 clinical questions, two independent reviewers screened the articles, evaluated the core outcomes, and performed meta-analyses. RESULTS Subcutaneous injection of methotrexate (MTX) showed similar efficacy to oral MTX in MTX-naïve RA patients. Ozoralizumab combined with MTX improved drug efficacy compared to the placebo in RA patients with inadequate response (IR) to conventional synthetic DMARD (csDMARD). Rituximab with and without concomitant csDMARDs showed similar efficacy to other biological DMARDs (bDMARDs) in bDMARD-IR RA patients. Combined Janus kinase inhibitors and MTX achieved similar clinical responses and equal safety during a 4-year period compared to tumour necrosis factor inhibitors in MTX-IR RA patients. Biosimilars showed efficacy equivalent to that of the original bDMARDs in csDMARD-IR and bDMARD-IR RA patients. CONCLUSIONS This systematic review provides latest evidence for the 2024 update of the Japan College of Rheumatology clinical practice guidelines for RA management.
Collapse
Affiliation(s)
- Yoichi Nakayama
- Department of Rheumatology and Clinical Immunology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Wataru Nagata
- Department of Pharmacology, National Defense Medical College, Tokorozawa, Japan
| | - Yoichi Takeuchi
- Department of Rheumatology and Nephrology, Japanese Red Cross Maebashi Hospital, Maebashi, Japan
| | - Sho Fukui
- Department of Emergency and General Medicine, Kyorin University School of Medicine, Tokyo, Japan
| | - Yuya Fujita
- The First Department of Internal Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Yohei Hosokawa
- Department of Clinical Immunology and Rheumatology, Hiroshima University Hospital, Hiroshima, Japan
| | - Masanobu Ueno
- The First Department of Internal Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Kumiko Ono
- Department of Joint Surgery, Research Hospital, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Shuji Sumitomo
- Department of Rheumatology, Kobe City Medical Center General Hospital, Kobe, Japan
| | - Yuya Tabuchi
- Department of Rheumatology and Clinical Immunology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Yuichiro Nakanishi
- Inflammation and Immunology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Shuntaro Saito
- Division of Rheumatology, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Hiroko Ikeuchi
- Department of Preventive Services, School of Public Health, Graduate School of Medicine and Faculty of Medicine, Kyoto University, Kyoto, Japan
| | - Kazutaka Kawamori
- Division of Rheumatology, Department of Medicine, Showa University School of Medicine, Tokyo, Japan
| | - Hideaki Sofue
- Inflammation and Immunology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Goro Doi
- Department of Internal Medicine, Kyushu University Beppu Hospital, Oita, Japan
| | - Runa Minami
- Department of Orthopaedic Surgery and Rheumatology, Otokoyama Hospital, Kyoto, Japan
| | - Tomoya Hirota
- Department of Infection and Rheumatology, University of Fukui Hospital, Fukui, Japan
| | - Kaoru Minegishi
- Department of Stem Cell and Immune Regulation, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | | | - Ryo Motoyama
- Division of Rheumatology, Department of Internal Medicine, Tokyo Women's Medical University School of Medicine, Tokyo, Japan
| | - Shohei Nakamura
- Division of Rheumatology, Department of Internal Medicine, Tokyo Women's Medical University School of Medicine, Tokyo, Japan
| | - Shotaro Suzuki
- Division of Rheumatology and Allergology, Department of Internal Medicine, St. Marianna University School of Medicine, Kawasaki, Japan
| | - Norihiro Nishioka
- Department of Preventive Services, School of Public Health, Graduate School of Medicine and Faculty of Medicine, Kyoto University, Kyoto, Japan
| | - Takuma Tsuzuki Wada
- Department of Rheumatology and Applied Immunology, Faculty of Medicine, Saitama Medical University, Saitama, Japan
| | - Akira Onishi
- Department of Advanced Medicine of Rheumatic Diseases, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Kenichi Nishimura
- Department of Pediatrics, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Ryu Watanabe
- Department of Clinical Immunology, Osaka Metropolitan University Graduate School of Medicine, Osaka, Japan
| | - Ryo Yanai
- Division of Rheumatology, Department of Medicine, Showa University School of Medicine, Tokyo, Japan
| | - Takashi Kida
- Inflammation and Immunology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Hiroki Nishiwaki
- Division of Nephrology, Department of Internal Medicine, Showa University Fujigaoka Hospital, Yokohama, Japan
| | - Nobuyuki Yajima
- Division of Rheumatology, Department of Medicine, Showa University School of Medicine, Tokyo, Japan
| | - Yuko Kaneko
- Division of Rheumatology, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Eiichi Tanaka
- Division of Rheumatology, Department of Internal Medicine, Tokyo Women's Medical University School of Medicine, Tokyo, Japan
| | - Yutaka Kawahito
- Inflammation and Immunology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Masayoshi Harigai
- Division of Rheumatology, Department of Internal Medicine, Tokyo Women's Medical University School of Medicine, Tokyo, Japan
| |
Collapse
|
6
|
Guffroy A, Jacquel L, Guffroy B, Martin T. CAR-T cells for treating systemic lupus erythematosus: A promising emerging therapy. Joint Bone Spine 2024; 91:105702. [PMID: 38336271 DOI: 10.1016/j.jbspin.2024.105702] [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: 12/11/2023] [Revised: 12/26/2023] [Accepted: 01/18/2024] [Indexed: 02/12/2024]
Abstract
Chimeric Antigen Receptor T-cell therapy (CAR-T), currently employed routinely for treating B-cell malignancies, has emerged as a groundbreaking approach in addressing severe autoimmune diseases, especially for systemic lupus erythematosus (SLE). The immunological rationale for targeting B lymphocytes in autoimmune diseases is well-established, demonstrating success in numerous autoantibody-mediated autoimmune conditions through targeted therapies over several years. However, this approach has often proven ineffective in the context of systemic lupus erythematosus. Recent data on CAR-T usage in lupus, revealed promising results including rapid and prolonged remission without treatment, highlighting the potential of CAR-T therapy in severe lupus cases. This article provides a comprehensive overview of CAR-T cells, tracing their evolution from hematological malignancies to their recent applications in autoimmune disorder, especially in lupus. Clinical trials within a regulated framework are now imperative to assess the procedural aspects in order to validate the considerable promise of CAR-T cell therapy in the field of autoimmune diseases. This includes evaluating safety and long-term efficacy and security of the procedure, the benefit-risk ratio in the field of autoimmunity, the availability and cost-related issues associated with this emerging cellular therapy procedure.
Collapse
Affiliation(s)
- Aurélien Guffroy
- Department of Clinical Immunology and Internal Medicine, National Reference Center for Systemic Autoimmune Diseases (CNR RESO), Tertiary Center for Primary Immunodeficiency, Strasbourg University Hospital, 67000 Strasbourg, France; University Strasbourg, INSERM UMR - S1109, Institut thématique interdisciplinaire (ITI) de Médecine de Précision de Strasbourg, Transplantex NG, Fédération Hospitalo-Universitaire OMICARE, Fédération de Médecine Translationnelle de Strasbourg (FMTS), 67000 Strasbourg, France.
| | - Léa Jacquel
- Department of Clinical Immunology and Internal Medicine, National Reference Center for Systemic Autoimmune Diseases (CNR RESO), Tertiary Center for Primary Immunodeficiency, Strasbourg University Hospital, 67000 Strasbourg, France; University Strasbourg, INSERM UMR - S1109, Institut thématique interdisciplinaire (ITI) de Médecine de Précision de Strasbourg, Transplantex NG, Fédération Hospitalo-Universitaire OMICARE, Fédération de Médecine Translationnelle de Strasbourg (FMTS), 67000 Strasbourg, France
| | - Blandine Guffroy
- Department of Hematology, ICANS (Institut for Cancer Strasbourg-Europe), Strasbourg, France
| | - Thierry Martin
- Department of Clinical Immunology and Internal Medicine, National Reference Center for Systemic Autoimmune Diseases (CNR RESO), Tertiary Center for Primary Immunodeficiency, Strasbourg University Hospital, 67000 Strasbourg, France; University Strasbourg, INSERM UMR - S1109, Institut thématique interdisciplinaire (ITI) de Médecine de Précision de Strasbourg, Transplantex NG, Fédération Hospitalo-Universitaire OMICARE, Fédération de Médecine Translationnelle de Strasbourg (FMTS), 67000 Strasbourg, France
| |
Collapse
|
7
|
Costanzo G, Ledda AG, Sambugaro G. State of the art: the treatment of systemic lupus erythematosus. Curr Opin Allergy Clin Immunol 2024; 24:266-273. [PMID: 38788118 DOI: 10.1097/aci.0000000000000996] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/26/2024]
Abstract
PURPOSE OF REVIEW Systemic lupus erythematosus (SLE) is a systemic autoimmune disease with dysregulated cells in the immune system. The disease affects organs like kidneys, nervous system, joints, and skin. To manage SLE effectively, novel treatments targeting immune system components have been developed. This review investigates the therapeutic potential of existing targeted therapies and explores future innovative approaches for well tolerated, personalized treatment. RECENT FINDINGS SLE treatment involves cytokine targets and specific immunologic pathways, with even small molecules involved. SUMMARY The advanced therapeutic options in SLE management give clinicians more tools to control disease activity according to personalized medicine.
Collapse
Affiliation(s)
- Giulia Costanzo
- Department of Medical Sciences and Public Health, University of Cagliari, Cagliari, Italy
| | | | | |
Collapse
|
8
|
Koh YR, Cummings KC. Newer Immunosuppressants for Rheumatologic Disease: Preoperative Considerations. Rheum Dis Clin North Am 2024; 50:545-557. [PMID: 38942584 DOI: 10.1016/j.rdc.2024.03.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/30/2024]
Abstract
With the advent of small-molecule immune modulators, recombinant fusion proteins, and monoclonal antibodies, treatment options for patients with rheumatic diseases are now broad. These agents carry significant risks and an individualized approach to each patient, balancing known risks and benefits, remains the most prudent course. This review summarizes the available immunosuppressant treatments, discusses their perioperative implications, and provides recommendations for their perioperative management.
Collapse
Affiliation(s)
- Ye Rin Koh
- Anesthesiology Institute, Cleveland Clinic, 9500 Euclid Avenue, E-31, Cleveland, OH 44195, USA
| | - Kenneth C Cummings
- Anesthesiology Institute, Cleveland Clinic, 9500 Euclid Avenue, E-31, Cleveland, OH 44195, USA.
| |
Collapse
|
9
|
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
|
10
|
Ruscitti P, Allanore Y, Baldini C, Barilaro G, Bartoloni Bocci E, Bearzi P, Bellis E, Berardicurti O, Biaggi A, Bombardieri M, Cantarini L, Cantatore FP, Caporali R, Caso F, Cervera R, Ciccia F, Cipriani P, Chatzis L, Colafrancesco S, Conti F, Corberi E, Costa L, Currado D, Cutolo M, D'Angelo S, Del Galdo F, Di Cola I, Di Donato S, Distler O, D'Onofrio B, Doria A, Fautrel B, Fasano S, Feist E, Fisher BA, Gabini M, Gandolfo S, Gatto M, Genovali I, Gerli R, Grembiale RD, Guggino G, Hoffmann-Vold AM, Iagnocco A, Iaquinta FS, Liakouli V, Manoussakis MN, Marino A, Mauro D, Montecucco C, Mosca M, Naty S, Navarini L, Occhialini D, Orefice V, Perosa F, Perricone C, Pilato A, Pitzalis C, Pontarini E, Prete M, Priori R, Rivellese F, Sarzi-Puttini P, Scarpa R, Sebastiani G, Selmi C, Shoenfeld Y, Triolo G, Trunfio F, Yan Q, Tzioufas AG, Giacomelli R. Tailoring the treatment of inflammatory rheumatic diseases by a better stratification and characterization of the clinical patient heterogeneity. Findings from a systematic literature review and experts' consensus. Autoimmun Rev 2024; 23:103581. [PMID: 39069240 DOI: 10.1016/j.autrev.2024.103581] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2024] [Accepted: 07/23/2024] [Indexed: 07/30/2024]
Abstract
Inflammatory rheumatic diseases are different pathologic conditions associated with a deregulated immune response, codified along a spectrum of disorders, with autoinflammatory and autoimmune diseases as two-end phenotypes of this continuum. Despite pathogenic differences, inflammatory rheumatic diseases are commonly managed with a limited number of immunosuppressive drugs, sometimes with partial evidence or transferring physicians' knowledge in different patients. In addition, several randomized clinical trials, enrolling these patients, did not meet the primary pre-established outcomes and these findings could be linked to the underlying molecular diversities along the spectrum of inflammatory rheumatic disorders. In fact, the resulting patient heterogeneity may be driven by differences in underlying molecular pathology also resulting in variable responses to immunosuppressive drugs. Thus, the identification of different clinical subsets may possibly overcome the major obstacles that limit the development more effective therapeutic strategies for these patients with inflammatory rheumatic diseases. This clinical heterogeneity could require a diverse therapeutic management to improve patient outcomes and increase the frequency of clinical remission. Therefore, the importance of better patient stratification and characterization is increasingly pointed out according to the precision medicine principles, also suggesting a new approach for disease treatment. In fact, based on a better proposed patient profiling, clinicians could more appropriately balance the therapeutic management. On these bases, we synthetized and discussed the available literature about the patient profiling in regard to therapy in the context of inflammatory rheumatic diseases, mainly focusing on randomized clinical trials. We provided an overview of the importance of a better stratification and characterization of the clinical heterogeneity of patients with inflammatory rheumatic diseases identifying this point as crucial in improving the management of these patients.
Collapse
Affiliation(s)
- Piero Ruscitti
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy.
| | - Yannick Allanore
- Rheumatology Department, Cochin Hospital, APHP, INSERM U1016, Université Paris Cité, Paris, France
| | - Chiara Baldini
- Rheumatology Unit, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Giuseppe Barilaro
- Department of Autoimmune Diseases, Reference Centre for Systemic Autoimmune Diseases, Vasculitis and Autoinflammatory Diseases of the Catalan and Spanish Health Systems, Member of ERN-ReCONNET/RITA, Hospital Clínic, University of Barcelona, Barcelona, Catalonia, Spain
| | - Elena Bartoloni Bocci
- Section of Rheumatology, Department of Medicine and Surgery, University of Perugia, Italy
| | - Pietro Bearzi
- Rheumatology and Clinical Immunology, Department of Medicine, School of Medicine, University of Rome "Campus Bio-Medico", 00128 Rome, Italy; Clinical and Research Section of Rheumatology and Clinical Immunology, Fondazione Policlinico Universitario Campus Bio-Medico, 00128 Rome, Italy
| | - Elisa Bellis
- Academic Rheumatology Centre, Dipartimento di Scienze Cliniche e Biologiche Università di Torino - AO Mauriziano di Torino, Turin, Italy
| | - Onorina Berardicurti
- Rheumatology and Clinical Immunology, Department of Medicine, School of Medicine, University of Rome "Campus Bio-Medico", 00128 Rome, Italy; Clinical and Research Section of Rheumatology and Clinical Immunology, Fondazione Policlinico Universitario Campus Bio-Medico, 00128 Rome, Italy
| | - Alice Biaggi
- Rheumatology and Clinical Immunology, Department of Medicine, School of Medicine, University of Rome "Campus Bio-Medico", 00128 Rome, Italy; Clinical and Research Section of Rheumatology and Clinical Immunology, Fondazione Policlinico Universitario Campus Bio-Medico, 00128 Rome, Italy
| | - Michele Bombardieri
- Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, Queen Mary University of London and Barts NIHR BRC & NHS Trust & National Institute for Health and Care Research (NIHR) Barts Biomedical Research Centre (BRC), London, UK
| | - Luca Cantarini
- Department of Medical Sciences, Surgery and Neurosciences, Research Center of Systemic Autoinflammatory Diseases and Behçet's Disease Clinic, University of Siena, Siena, Italy; Azienda Ospedaliero-Universitaria Senese [European Reference Network (ERN) for Rare Immunodeficiency, Autoinflammatory and Autoimmune Diseases (RITA) Center] Siena, Italy
| | - Francesco Paolo Cantatore
- Rheumatology Clinic, Department of Medical and Surgical Sciences, University of Foggia, 71122 Foggia, Italy
| | - Roberto Caporali
- Department of Clinical Sciences and Community Health, University of Milan, Paediatric Rheumatology Unit, and Clinical Rheumatology Unit, ASST Pini-CTO, Milan, Italy
| | - Francesco Caso
- Rheumatology Research Unit, Department of Clinical Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Ricard Cervera
- Department of Autoimmune Diseases, Reference Centre for Systemic Autoimmune Diseases, Vasculitis and Autoinflammatory Diseases of the Catalan and Spanish Health Systems, Member of ERN-ReCONNET/RITA, Hospital Clínic, University of Barcelona, Barcelona, Catalonia, Spain
| | - Francesco Ciccia
- Rheumatology Unit, Department of Precision Medicine, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Paola Cipriani
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| | - Loukas Chatzis
- Department of Pathophysiology, School of Medicine, National and Kapodistrian University of Athens, Greece
| | - Serena Colafrancesco
- Department of Internal Medicine and Medical Specialties, Rheumatology Unit, Sapienza University of Rome, Viale del Policlinico 155, 00185 Rome, Italy
| | - Fabrizio Conti
- Department of Internal Medicine and Medical Specialties, Rheumatology Unit, Sapienza University of Rome, Viale del Policlinico 155, 00185 Rome, Italy
| | - Erika Corberi
- Rheumatology and Clinical Immunology, Department of Medicine, School of Medicine, University of Rome "Campus Bio-Medico", 00128 Rome, Italy; Clinical and Research Section of Rheumatology and Clinical Immunology, Fondazione Policlinico Universitario Campus Bio-Medico, 00128 Rome, Italy
| | - Luisa Costa
- Rheumatology Research Unit, Department of Clinical Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Damiano Currado
- Rheumatology and Clinical Immunology, Department of Medicine, School of Medicine, University of Rome "Campus Bio-Medico", 00128 Rome, Italy; Clinical and Research Section of Rheumatology and Clinical Immunology, Fondazione Policlinico Universitario Campus Bio-Medico, 00128 Rome, Italy
| | - Maurizio Cutolo
- Laboratory of Experimental Rheumatology and Academic Division of Rheumatology, Department of Internal Medicine and Specialties, University of Genova Italy, IRCCS Polyclinic Hospital, Genova, Italy
| | - Salvatore D'Angelo
- Rheumatology Depatment of Lucania, San Carlo Hospital of Potenza and Madonna delle Grazie Hospital of Matera, Potenza, Italy
| | - Francesco Del Galdo
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Leeds, UK; NIHR Leeds Biomedical Research Centre, Leeds Teaching Hospitals NHS Trust, Leeds, UK
| | - Ilenia Di Cola
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| | - Stefano Di Donato
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Leeds, UK; NIHR Leeds Biomedical Research Centre, Leeds Teaching Hospitals NHS Trust, Leeds, UK
| | - Oliver Distler
- Department of Rheumatology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Bernardo D'Onofrio
- Department of Internal Medicine and Therapeutics, Università di Pavia, Division of Rheumatology, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Andrea Doria
- Rheumatology Unit, Department of Medicine (DIMED), University of Padua, Padua, Italy
| | - Bruno Fautrel
- Sorbonne Université - Assistance Publique Hôpitaux de Paris, INSERM UMRS 1136, Hôpital de La Pitié Salpêtrière, Paris, France
| | - Serena Fasano
- Rheumatology Unit, Department of Precision Medicine, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Eugen Feist
- Department of Rheumatology, Helios Fachklinik, Sophie-von-Boetticher-Straße 1, 39245, Vogelsang-Gommern, Germany; Charité - Universitätsmedizin Berlin, Medizinische Klinik mit Schwerpunkt Rheumatologie und Klinische Immunologie, Berlin, Germany
| | - Benjamin A Fisher
- Institute of Inflammation and Ageing, University Hospitals Birmingham, Birmingham, UK; Department of Rheumatology, National Institute for Health Research (NIHR), Birmingham Biomedical Research Centre, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
| | - Marco Gabini
- Rheumatology Unit, Santo Spirito Hospital, Pescara, Italy
| | - Saviana Gandolfo
- Unit of Rheumatology, San Giovanni Bosco Hospital, Naples, Italy
| | - Mariele Gatto
- Academic Rheumatology Centre, Dipartimento di Scienze Cliniche e Biologiche Università di Torino - AO Mauriziano di Torino, Turin, Italy
| | - Irene Genovali
- Rheumatology and Clinical Immunology, Department of Medicine, School of Medicine, University of Rome "Campus Bio-Medico", 00128 Rome, Italy; Clinical and Research Section of Rheumatology and Clinical Immunology, Fondazione Policlinico Universitario Campus Bio-Medico, 00128 Rome, Italy
| | - Roberto Gerli
- Section of Rheumatology, Department of Medicine and Surgery, University of Perugia, Italy
| | - Rosa Daniela Grembiale
- Rheumatology Research Unit, Dipartimento di Scienze della Salute, Università degli studi "Magna Graecia" di Catanzaro, Catanzaro, Italy
| | - Giuliana Guggino
- Rheumatology Unit, Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, University of Palermo, Italy
| | - Anna Maria Hoffmann-Vold
- Department of Rheumatology, University Hospital Zurich, University of Zurich, Zurich, Switzerland; Department of Rheumatology, Oslo University Hospital-Rikshospitalet, Oslo, Norway
| | - Annamaria Iagnocco
- Academic Rheumatology Centre, Dipartimento di Scienze Cliniche e Biologiche Università di Torino - AO Mauriziano di Torino, Turin, Italy
| | - Francesco Salvatore Iaquinta
- Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, Queen Mary University of London and Barts NIHR BRC & NHS Trust & National Institute for Health and Care Research (NIHR) Barts Biomedical Research Centre (BRC), London, UK
| | - Vasiliki Liakouli
- Rheumatology Unit, Department of Precision Medicine, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Menelaos N Manoussakis
- Department of Pathophysiology, School of Medicine, National and Kapodistrian University of Athens, Greece
| | - Annalisa Marino
- Rheumatology and Clinical Immunology, Department of Medicine, School of Medicine, University of Rome "Campus Bio-Medico", 00128 Rome, Italy; Clinical and Research Section of Rheumatology and Clinical Immunology, Fondazione Policlinico Universitario Campus Bio-Medico, 00128 Rome, Italy
| | - Daniele Mauro
- Rheumatology Unit, Department of Precision Medicine, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Carlomaurizio Montecucco
- Department of Internal Medicine and Therapeutics, Università di Pavia, Division of Rheumatology, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Marta Mosca
- Rheumatology Unit, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Saverio Naty
- Department of Health Sciences, "Magna Græcia" University of Catanzaro, 88100 Catanzaro, Italy
| | - Luca Navarini
- Rheumatology and Clinical Immunology, Department of Medicine, School of Medicine, University of Rome "Campus Bio-Medico", 00128 Rome, Italy; Clinical and Research Section of Rheumatology and Clinical Immunology, Fondazione Policlinico Universitario Campus Bio-Medico, 00128 Rome, Italy
| | - Daniele Occhialini
- Rheumatic and Systemic Autoimmune Diseases Unit, Department of Interdisciplinary Medicine (DIM), University of Bari Medical School, Italy
| | - Valeria Orefice
- Rheumatology Unit, San Camillo-Forlanini Hospital, Rome, Italy
| | - Federico Perosa
- Rheumatic and Systemic Autoimmune Diseases Unit, Department of Interdisciplinary Medicine (DIM), University of Bari Medical School, Italy
| | - Carlo Perricone
- Section of Rheumatology, Department of Medicine and Surgery, University of Perugia, Italy
| | - Andrea Pilato
- Rheumatology and Clinical Immunology, Department of Medicine, School of Medicine, University of Rome "Campus Bio-Medico", 00128 Rome, Italy; Clinical and Research Section of Rheumatology and Clinical Immunology, Fondazione Policlinico Universitario Campus Bio-Medico, 00128 Rome, Italy
| | - Costantino Pitzalis
- Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, Queen Mary University of London and Barts NIHR BRC & NHS Trust & National Institute for Health and Care Research (NIHR) Barts Biomedical Research Centre (BRC), London, UK; Department of Biomedical Sciences, Humanitas University, Milan, Italy
| | - Elena Pontarini
- Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, Queen Mary University of London and Barts NIHR BRC & NHS Trust & National Institute for Health and Care Research (NIHR) Barts Biomedical Research Centre (BRC), London, UK
| | - Marcella Prete
- Rheumatic and Systemic Autoimmune Diseases Unit, Department of Interdisciplinary Medicine (DIM), University of Bari Medical School, Italy
| | - Roberta Priori
- Department of Internal Medicine and Medical Specialties, Rheumatology Unit, Sapienza University of Rome, Viale del Policlinico 155, 00185 Rome, Italy
| | - Felice Rivellese
- Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, Queen Mary University of London and Barts NIHR BRC & NHS Trust & National Institute for Health and Care Research (NIHR) Barts Biomedical Research Centre (BRC), London, UK
| | - Piercarlo Sarzi-Puttini
- Rheumatology Department, ASST Fatebenefratelli Luigi Sacco University Hospital, Milan, Italy; Department of Clinical Sciences and Community Health, Università degli Studi di Milano, Milan, Italy
| | - Raffaele Scarpa
- Rheumatology Research Unit, Department of Clinical Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | | | - Carlo Selmi
- Department of Biomedical Sciences, Humanitas University, Milan, Italy
| | - Yehuda Shoenfeld
- Zabludovwicz autoimmunity center, Sheba medical center, Tel Hashomer Israel, Reichman University, Herzeliya, Israel
| | - Giovanni Triolo
- Rheumatology Unit, Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, University of Palermo, Italy
| | - Francesca Trunfio
- Rheumatology and Clinical Immunology, Department of Medicine, School of Medicine, University of Rome "Campus Bio-Medico", 00128 Rome, Italy; Clinical and Research Section of Rheumatology and Clinical Immunology, Fondazione Policlinico Universitario Campus Bio-Medico, 00128 Rome, Italy
| | - Qingran Yan
- Department of Rheumatology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Athanasios G Tzioufas
- Department of Pathophysiology, School of Medicine, National and Kapodistrian University of Athens, Greece
| | - Roberto Giacomelli
- Rheumatology and Clinical Immunology, Department of Medicine, School of Medicine, University of Rome "Campus Bio-Medico", 00128 Rome, Italy; Clinical and Research Section of Rheumatology and Clinical Immunology, Fondazione Policlinico Universitario Campus Bio-Medico, 00128 Rome, Italy
| |
Collapse
|
11
|
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
|
12
|
Mok TC, Mok CC. Non-TNF biologics and their biosimilars in rheumatoid arthritis. Expert Opin Biol Ther 2024; 24:599-613. [PMID: 38766765 DOI: 10.1080/14712598.2024.2358165] [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: 03/07/2024] [Accepted: 05/17/2024] [Indexed: 05/22/2024]
Abstract
INTRODUCTION Rheumatoid arthritis (RA) is a chronic inflammatory rheumatic disease that affects both the articular and extra-articular structures, leading to significant joint damage, disability and excess mortality. The treatment algorithm of RA has changed tremendously in the past 1-2 decades because of the emergence of novel biological therapies that target different mechanisms of action in addition to TNFα. AREAS COVERED This article summarizes the evidence and safety of the non-TNF biological DMARDs in the treatment of RA, including those that target B cells, T-cell co-stimulation, interleukin (IL)-6 and granulocyte-monocyte colony-stimulating factor (GM-CSF). The targeted synthetic DMARDs such as the Janus kinase inhibitors are not included. The availability of the less costly biosimilars has enabled more patients to receive biological therapy earlier in the course of the disease. The evidence for the non-TNF biosimilar compounds in RA is also reviewed. EXPERT OPINION There are unmet needs of developing novel therapeutic agents to enhance the response rate and provide more options for difficult-to-treat RA. These include the newer generation biologic and targeted synthetic DMARDs. A personalized treatment strategy in RA requires evaluation of the cellular, cytokine, genomic and transcriptomic profile that would predict treatment response to biologic or targeted DMARDs of different mechanisms of action.
Collapse
Affiliation(s)
- Tsz Ching Mok
- Department of Medicine, Ruttonjee Hospital, Hong Kong, China
| | - Chi Chiu Mok
- Department of Medicine, Tuen Mun Hospital, Hong Kong, China
| |
Collapse
|
13
|
Ali M, Benfante V, Di Raimondo D, Laudicella R, Tuttolomondo A, Comelli A. A Review of Advances in Molecular Imaging of Rheumatoid Arthritis: From In Vitro to Clinic Applications Using Radiolabeled Targeting Vectors with Technetium-99m. Life (Basel) 2024; 14:751. [PMID: 38929734 PMCID: PMC11204982 DOI: 10.3390/life14060751] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Revised: 05/24/2024] [Accepted: 06/07/2024] [Indexed: 06/28/2024] Open
Abstract
Rheumatoid arthritis (RA) is a systemic autoimmune disorder caused by inflammation of cartilaginous diarthrodial joints that destroys joints and cartilage, resulting in synovitis and pannus formation. Timely detection and effective management of RA are pivotal for mitigating inflammatory arthritis consequences, potentially influencing disease progression. Nuclear medicine using radiolabeled targeted vectors presents a promising avenue for RA diagnosis and response to treatment assessment. Radiopharmaceutical such as technetium-99m (99mTc), combined with single photon emission computed tomography (SPECT) combined with CT (SPECT/CT), introduces a more refined diagnostic approach, enhancing accuracy through precise anatomical localization, representing a notable advancement in hybrid molecular imaging for RA evaluation. This comprehensive review discusses existing research, encompassing in vitro, in vivo, and clinical studies to explore the application of 99mTc radiolabeled targeting vectors with SPECT imaging for RA diagnosis. The purpose of this review is to highlight the potential of this strategy to enhance patient outcomes by improving the early detection and management of RA.
Collapse
Affiliation(s)
- Muhammad Ali
- Ri.MED Foundation, Via Bandiera 11, 90133 Palermo, Italy; (M.A.); (A.C.)
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, Molecular and Clinical Medicine, University of Palermo, 90127 Palermo, Italy; (D.D.R.); (A.T.)
| | - Viviana Benfante
- Ri.MED Foundation, Via Bandiera 11, 90133 Palermo, Italy; (M.A.); (A.C.)
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, Molecular and Clinical Medicine, University of Palermo, 90127 Palermo, Italy; (D.D.R.); (A.T.)
| | - Domenico Di Raimondo
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, Molecular and Clinical Medicine, University of Palermo, 90127 Palermo, Italy; (D.D.R.); (A.T.)
| | - Riccardo Laudicella
- Nuclear Medicine Unit, Department of Biomedical and Dental Sciences and Morpho-Functional Imaging, Messina University, 98124 Messina, Italy;
| | - Antonino Tuttolomondo
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, Molecular and Clinical Medicine, University of Palermo, 90127 Palermo, Italy; (D.D.R.); (A.T.)
| | - Albert Comelli
- Ri.MED Foundation, Via Bandiera 11, 90133 Palermo, Italy; (M.A.); (A.C.)
- NBFC—National Biodiversity Future Center, 90133 Palermo, Italy
| |
Collapse
|
14
|
Bertrand D, Deprez A, Doumen M, De Cock D, Pazmino S, Marchal A, Thelissen M, Joly J, De Meyst E, Neerinckx B, Westhovens R, Verschueren P. Patients' and rheumatologists' perceptions on dose reduction of rituximab in rheumatoid arthritis. Musculoskeletal Care 2024; 22:e1893. [PMID: 38693680 DOI: 10.1002/msc.1893] [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: 04/11/2024] [Revised: 04/23/2024] [Accepted: 04/25/2024] [Indexed: 05/03/2024]
Abstract
OBJECTIVE The recommended dose of a rituximab course for the treatment of Rheumatoid Arthritis (RA) consists of two infusions of 1000 mg with a 2-week interval. Evidence is growing that a lower dose could be as effective. We aimed to investigate patients' and rheumatologists' perceptions on dose reduction of rituximab. METHODS Patients with RA treated with rituximab, and rheumatologists were invited for a qualitative study via individual semi-structured interviews. Participants were recruited based on purposive sampling to ensure diversity. Interviews were analysed according to the principles of grounded theory and the constant comparative method. RESULTS Sixteen patients and 13 rheumatologists were interviewed. Patients and rheumatologists perceived the benefits of rituximab dose reduction for reasons of safety and societal costs. Furthermore, available evidence for the effectiveness of lower doses was mentioned as an argument in favour, in addition to the possibility to tailor the dose based on the patients' clinical manifestations. However, patients and rheumatologists had concerns about the potential loss of effectiveness and quality of life. Moreover, some rheumatologists felt uncomfortable with dose reduction due to insufficient experience with rituximab in general. Patients and rheumatologists emphasised the importance of shared decision-making, underscoring the pivotal role of physicians in this process by explaining the reasoning behind dose reduction. CONCLUSION Although some concerns on effectiveness were perceived, both patients and rheumatologists saw potential benefits of dose reduction in terms of safety, societal costs, and application of a personalised approach. As a result, most rheumatologists and patients showed a willingness to consider dose reduction strategies.
Collapse
Affiliation(s)
- Delphine Bertrand
- Department of Development and Regeneration, Skeletal Biology and Engineering Research Center, KU Leuven, Leuven, Belgium
| | - Anke Deprez
- Department of Development and Regeneration, Skeletal Biology and Engineering Research Center, KU Leuven, Leuven, Belgium
- Department of Public Health and Primary Care, Environment and Health, KU Leuven, Leuven, Belgium
| | - Michaël Doumen
- Department of Development and Regeneration, Skeletal Biology and Engineering Research Center, KU Leuven, Leuven, Belgium
- Rheumatology, UZ Leuven, Leuven, Belgium
| | - Diederik De Cock
- Biostatistics and Medical Informatics Research Group, Department of Public Health, Faculty of Medicine and Pharmacy, Vrije Universiteit Brussel (VUB), Brussel, Belgium
| | - Sofia Pazmino
- Department of Development and Regeneration, Skeletal Biology and Engineering Research Center, KU Leuven, Leuven, Belgium
- Department of Chronic Diseases and Metabolism, Clinical and Experimental Endocrinology, KU Leuven, Leuven, Belgium
| | | | | | - Johan Joly
- Rheumatology, UZ Leuven, Leuven, Belgium
| | - Elias De Meyst
- Department of Development and Regeneration, Skeletal Biology and Engineering Research Center, KU Leuven, Leuven, Belgium
- Rheumatology, UZ Leuven, Leuven, Belgium
| | - Barbara Neerinckx
- Department of Development and Regeneration, Skeletal Biology and Engineering Research Center, KU Leuven, Leuven, Belgium
- Rheumatology, UZ Leuven, Leuven, Belgium
| | - René Westhovens
- Department of Development and Regeneration, Skeletal Biology and Engineering Research Center, KU Leuven, Leuven, Belgium
- Rheumatology, UZ Leuven, Leuven, Belgium
| | - Patrick Verschueren
- Department of Development and Regeneration, Skeletal Biology and Engineering Research Center, KU Leuven, Leuven, Belgium
- Rheumatology, UZ Leuven, Leuven, Belgium
| |
Collapse
|
15
|
Yang H, Liu C, Lin X, Li X, Zeng S, Gong Z, Xu Q, Li D, Li N. Wogonin inhibits the migration and invasion of fibroblast-like synoviocytes by targeting PI3K/AKT/NF-κB pathway in rheumatoid arthritis. Arch Biochem Biophys 2024; 755:109965. [PMID: 38552763 DOI: 10.1016/j.abb.2024.109965] [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: 10/05/2023] [Revised: 03/17/2024] [Accepted: 03/21/2024] [Indexed: 04/24/2024]
Abstract
BACKGROUND Rheumatoid arthritis (RA) is currently an autoimmune inflammatory disease with an unclear pathogenesis. Fibroblast-like synoviocytes (FLSs) have tumor-like properties, and their activation and secretion of pro-inflammatory factors are important factors in joint destruction. Wogonin (5,7-dihydroxy-8-methoxyflavone), a natural flavonoid isolated from Scutellaria baicalensis root, has been shown to have significant anti-inflammatory, anti-oxidative stress, and anti-tumor effects in a variety of diseases. However, the role of wogonin in RA has not yet been demonstrated. PURPOSE To investigate the inhibitory effect of wogonin on the invasive behavior of fibroblast-like synoviocytes and to explore the mechanism of action of wogonin in RA. METHODS CCK-8, EdU, cell migration and invasion, immunofluorescence staining, RT-qPCR, and protein blot analysis were used to study the inhibitory effects of wogonin on migration, invasion, and pro-inflammatory cytokine overexpression in the immortalized rheumatoid synovial cell line MH7A. The therapeutic effects of wogonin were validated in vivo using arthritis scores and histopathological evaluation of collagen-induced arthritis mice. RESULTS Wogonin inhibited the migration and invasion of MH7A cells, reduced the production of TNF-α, IL-1β, IL-6, MMP-3 and MMP-9, and increased the expression of IL-10. Moreover, wogonin also inhibited the myofibrillar differentiation of MH7A cells, increased the expression of E-cadherin (E-Cad) and decreased the expression of α-smooth muscle actin (α-SMA). In addition, wogonin treatment effectively ameliorated joint destruction in CIA mice. Further molecular mechanism studies showed that wogonin treatment significantly inhibited the activation of PI3K/AKT/NF-κB signaling pathway in TNF-α-induced arthritic FLSs. CONCLUSION Wogonin effectively inhibits migration, invasion and pro-inflammatory cytokine production of RA fibroblast-like synoviocytes through the PI3K/AKT/NF-κB pathway, and thus wogonin, as a natural flavonoid, has great potential for treating RA.
Collapse
Affiliation(s)
- Haixin Yang
- School of Traditional Chinese Medicine, Jinan University, 510632, Guangzhou, China.
| | - Cuizhen Liu
- The First Clinical College of Guangzhou University of Chinese Medicine, 510405, Guangzhou, China.
| | - Xiujuan Lin
- The First Clinical College of Guangzhou University of Chinese Medicine, 510405, Guangzhou, China.
| | - Xing Li
- Department of Rheumatology and Immunology, The Third Affiliated Hospital, Southern Medical University, 510630, Guangzhou, China.
| | - Shan Zeng
- Department of Rheumatology, The First Affiliated Hospital of Jinan University, 510632, Guangzhou, China.
| | - Zhaohui Gong
- Department of Cardiovascular, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, 510405, Guangzhou, China.
| | - Qiang Xu
- State Key Laboratory of Traditional Chinese Medicine Syndrome, Department of Rheumatology, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510405, China.
| | - Detang Li
- The First Clinical College of Guangzhou University of Chinese Medicine, 510405, Guangzhou, China; Department of Pharmacy, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510405, China; Guangdong Clinical Research Academy of Chinese Medicine, Guangzhou, 510405, China.
| | - Nan Li
- School of Traditional Chinese Medicine, Jinan University, 510632, Guangzhou, China.
| |
Collapse
|
16
|
Bass LE, Bonami RH. Factors Governing B Cell Recognition of Autoantigen and Function in Type 1 Diabetes. Antibodies (Basel) 2024; 13:27. [PMID: 38651407 PMCID: PMC11036271 DOI: 10.3390/antib13020027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Revised: 03/15/2024] [Accepted: 03/18/2024] [Indexed: 04/25/2024] Open
Abstract
Islet autoantibodies predict type 1 diabetes (T1D) but can be transient in murine and human T1D and are not thought to be directly pathogenic. Rather, these autoantibodies signal B cell activity as antigen-presenting cells (APCs) that present islet autoantigen to diabetogenic T cells to promote T1D pathogenesis. Disrupting B cell APC function prevents T1D in mouse models and has shown promise in clinical trials. Autoantigen-specific B cells thus hold potential as sophisticated T1D biomarkers and therapeutic targets. B cell receptor (BCR) somatic hypermutation is a mechanism by which B cells increase affinity for islet autoantigen. High-affinity B and T cell responses are selected in protective immune responses, but immune tolerance mechanisms are known to censor highly autoreactive clones in autoimmunity, including T1D. Thus, different selection rules often apply to autoimmune disease settings (as opposed to protective host immunity), where different autoantigen affinity ceilings are tolerated based on variations in host genetics and environment. This review will explore what is currently known regarding B cell signaling, selection, and interaction with T cells to promote T1D pathogenesis.
Collapse
Affiliation(s)
- Lindsay E. Bass
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA;
| | - Rachel H. Bonami
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA;
- Department of Medicine, Division of Rheumatology and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Vanderbilt Center for Immunobiology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Vanderbilt Institute for Infection, Immunology and Inflammation, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| |
Collapse
|
17
|
Romiti R, Hirayama ALDS, Porro AM, Gonçalves HDS, Miot LDB, Durães SMB, Marques SA. Infections in the era of immunobiologicals. An Bras Dermatol 2024; 99:167-180. [PMID: 38238209 PMCID: PMC10943328 DOI: 10.1016/j.abd.2023.08.004] [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: 04/05/2023] [Revised: 08/15/2023] [Accepted: 08/27/2023] [Indexed: 03/11/2024] Open
Abstract
Immunobiologicals represent an innovative therapeutic option in dermatology. They are indicated in severe and refractory cases of different diseases when there is contraindication, intolerance, or failure of conventional systemic therapy and in cases with significant impairment of patient quality of life. The main immunobiologicals used in dermatology basically include inhibitors of tumor necrosis factor-alpha (anti-TNF), inhibitors of interleukin-12 and -23 (anti-IL12/23), inhibitors of interleukin-17 and its receptor (anti-IL17), inhibitors of interleukin-23 (anti-IL23), rituximab (anti-CD20 antibody), dupilumab (anti-IL4/IL13) and intravenous immunoglobulin. Their immunomodulatory action may be associated with an increase in the risk of infections in the short and long term, and each case must be assessed individually, according to the risk inherent to the drug, the patient general condition, and the need for precautions. This article will discuss the main risks of infection associated with the use of immunobiologicals, addressing the risk in immunocompetent and immunosuppressed patients, vaccination, fungal infections, tuberculosis, leprosy, and viral hepatitis, and how to manage the patient in the most diverse scenarios.
Collapse
Affiliation(s)
- Ricardo Romiti
- Department of Dermatology, Hospital das Clínicas, Universidade de São Paulo, São Paulo, SP, Brazil
| | | | - Adriana Maria Porro
- Department of Dermatology, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, SP, Brazil
| | - Heitor de Sá Gonçalves
- State Health Secretariat of Ceará, Centro de Dermatologia Dona Libânia, Fortaleza, CE, Brazil
| | - Luciane Donida Bartoli Miot
- Department of Infectology, Dermatology, Imaging Diagnosis and Radiotherapy, Faculty of Medicine, Universidade Estadual Paulista, Botucatu, SP, Brazil
| | - Sandra Maria Barbosa Durães
- Department of Internal Medicine, Dermatology Unit, Faculty of Medicine, Universidade Federal Fluminense, Niterói, RJ, Brazil
| | - Silvio Alencar Marques
- Department of Infectology, Dermatology, Imaging Diagnosis and Radiotherapy, Faculty of Medicine, Universidade Estadual Paulista, Botucatu, SP, Brazil
| |
Collapse
|
18
|
Koh YR, Cummings KC. Newer Immunosuppressants for Rheumatologic Disease: Preoperative Considerations. Anesthesiol Clin 2024; 42:131-143. [PMID: 38278585 DOI: 10.1016/j.anclin.2023.07.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2024]
Abstract
With the advent of small-molecule immune modulators, recombinant fusion proteins, and monoclonal antibodies, treatment options for patients with rheumatic diseases are now broad. These agents carry significant risks and an individualized approach to each patient, balancing known risks and benefits, remains the most prudent course. This review summarizes the available immunosuppressant treatments, discusses their perioperative implications, and provides recommendations for their perioperative management.
Collapse
Affiliation(s)
- Ye Rin Koh
- Anesthesiology Institute, Cleveland Clinic, 9500 Euclid Avenue, E-31, Cleveland, OH 44195, USA
| | - Kenneth C Cummings
- Anesthesiology Institute, Cleveland Clinic, 9500 Euclid Avenue, E-31, Cleveland, OH 44195, USA.
| |
Collapse
|
19
|
Bertrand D, Deprez A, Doumen M, De Cock D, Pazmino S, Marchal A, Thelissen M, Joly J, De Meyst E, Neerinckx B, Westhovens R, Verschueren P. To flare or not to flare: patients' and rheumatologists' perceptions on the on-flare retreatment strategy of rituximab in rheumatoid arthritis. Ther Adv Musculoskelet Dis 2024; 16:1759720X241232268. [PMID: 38425577 PMCID: PMC10903194 DOI: 10.1177/1759720x241232268] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2023] [Accepted: 01/18/2024] [Indexed: 03/02/2024] Open
Abstract
Background Several retreatment strategies exist for rituximab in rheumatoid arthritis (RA). In some countries, reimbursement criteria require a loss of disease control for rituximab retreatment. Understanding the patients' and rheumatologists' perceptions regarding this retreatment strategy would be informative in identifying the optimal treatment administration schedule. Objectives This study aimed to uncover patients' and rheumatologists' perceptions regarding retreatment strategies of rituximab. Design Qualitative study - semi-structured interviews. Methods Patients with RA, treated with rituximab, and rheumatologists were invited to participate in a qualitative study consisting of individual, in-depth, semi-structured interviews. Interviews were analysed according to the Qualitative Analysis Guide of Leuven. Results A total of 16 patients and 13 rheumatologists were interviewed. Benefits (e.g. decreased risk of overtreatment, cost savings and long-lasting effectiveness of rituximab) and barriers (e.g. fluctuating disease activity, slow mode of action and increased glucocorticoid use) of on-flare retreatment were identified. To effectively treat on-flare, flares must first be identified timely. Both stakeholder groups acknowledged that patients are capable of recognizing flares. However, the patient's ability to discriminate between inflammatory and other types of pain was perceived as difficult. Furthermore, patients and rheumatologists stressed that patients must timely seek professional help in case of a flare, followed by a swift response from the rheumatologists. Remarkably, retreatment was approached in various ways among rheumatologists, and not always adhering strictly to the on-flare reimbursement criteria. Conclusion This study revealed that both stakeholder groups perceived the heterogeneity in recognition of and reaction to a flare as important in influencing the effectiveness of the on-flare retreatment strategy. Moreover, this study identified the benefits and barriers of treating on-flare, which could be informative for daily practice decisions.
Collapse
Affiliation(s)
- Delphine Bertrand
- Skeletal Biology and Engineering Research Center, KU Leuven Department of Development and Regeneration, Herestraat 49, Leuven 3000, Belgium
| | - Anke Deprez
- KU Leuven, Department of Development and Regeneration, Skeletal Biology and Engineering Research Center, Leuven, Belgium
- KU Leuven, Department of Public Health and Primary Care, Environment and Health, Leuven, Belgium
| | - Michaël Doumen
- KU Leuven, Department of Development and Regeneration, Skeletal Biology and Engineering Research Center, Leuven, Belgium
- UZ Leuven, Rheumatology, Leuven, Belgium
| | - Diederik De Cock
- KU Leuven, Department of Development and Regeneration, Skeletal Biology and Engineering Research Center, Leuven, Belgium
- Biostatistics and Medical Informatics Research Group, Department of Public Health Sciences, Vrije Universiteit Brussel (VUB), Brussels, Belgium
| | - Sofia Pazmino
- KU Leuven, Department of Development and Regeneration, Skeletal Biology and Engineering Research Center, Leuven, Belgium
- KU Leuven, Department of Chronic Diseases and Metabolism, Clinical and Experimental Endocrinology, Leuven, Belgium
| | | | | | - Johan Joly
- UZ Leuven, Rheumatology, Leuven, Belgium
| | - Elias De Meyst
- KU Leuven, Department of Development and Regeneration, Skeletal Biology and Engineering Research Center, Leuven, Belgium
- UZ Leuven, Rheumatology, Leuven, Belgium
| | - Barbara Neerinckx
- KU Leuven, Department of Development and Regeneration, Skeletal Biology and Engineering Research Center, Leuven, Belgium
- UZ Leuven, Rheumatology, Leuven, Belgium
| | - René Westhovens
- KU Leuven, Department of Development and Regeneration, Skeletal Biology and Engineering Research Center, Leuven, Belgium
- UZ Leuven, Rheumatology, Leuven, Belgium
| | - Patrick Verschueren
- KU Leuven, Department of Development and Regeneration, Skeletal Biology and Engineering Research Center, Leuven, Belgium
- UZ Leuven, Rheumatology, Leuven, Belgium
| |
Collapse
|
20
|
Jeong E, Hong H, Lee YA, Kim KS. Potential Rheumatoid Arthritis-Associated Interstitial Lung Disease Treatment and Computational Approach for Future Drug Development. Int J Mol Sci 2024; 25:2682. [PMID: 38473928 PMCID: PMC11154459 DOI: 10.3390/ijms25052682] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Revised: 02/19/2024] [Accepted: 02/20/2024] [Indexed: 03/14/2024] Open
Abstract
Rheumatoid arthritis (RA) is a systemic autoimmune disease characterized by swelling in at least one joint. Owing to an overactive immune response, extra-articular manifestations are observed in certain cases, with interstitial lung disease (ILD) being the most common. Rheumatoid arthritis-associated interstitial lung disease (RA-ILD) is characterized by chronic inflammation of the interstitial space, which causes fibrosis and the scarring of lung tissue. Controlling inflammation and pulmonary fibrosis in RA-ILD is important because they are associated with high morbidity and mortality. Pirfenidone and nintedanib are specific drugs against idiopathic pulmonary fibrosis and showed efficacy against RA-ILD in several clinical trials. Immunosuppressants and disease-modifying antirheumatic drugs (DMARDs) with anti-fibrotic effects have also been used to treat RA-ILD. Immunosuppressants moderate the overexpression of cytokines and immune cells to reduce pulmonary damage and slow the progression of fibrosis. DMARDs with mild anti-fibrotic effects target specific fibrotic pathways to regulate fibrogenic cellular activity, extracellular matrix homeostasis, and oxidative stress levels. Therefore, specific medications are required to effectively treat RA-ILD. In this review, the commonly used RA-ILD treatments are discussed based on their molecular mechanisms and clinical trial results. In addition, a computational approach is proposed to develop specific drugs for RA-ILD.
Collapse
Affiliation(s)
- Eunji Jeong
- Department of Medicine, College of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea;
| | - Hyunseok Hong
- Yale College, Yale University, New Haven, CT 06520, USA;
- Department of Clinical Pharmacology and Therapeutics, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Yeon-Ah Lee
- Division of Rheumatology, Department of Internal Medicine, Kyung Hee University Hospital, Seoul 02447, Republic of Korea;
| | - Kyoung-Soo Kim
- Department of Medicine, College of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea;
- Department of Clinical Pharmacology and Therapeutics, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
- East-West Bone & Joint Disease Research Institute, Kyung Hee University Hospital at Gangdong, Seoul 05278, Republic of Korea
| |
Collapse
|
21
|
Kwon S, Judson MA. Clinical Pharmacology in Sarcoidosis: How to Use and Monitor Sarcoidosis Medications. J Clin Med 2024; 13:1250. [PMID: 38592130 PMCID: PMC10932410 DOI: 10.3390/jcm13051250] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Revised: 02/11/2024] [Accepted: 02/13/2024] [Indexed: 04/10/2024] Open
Abstract
When sarcoidosis needs treatment, pharmacotherapy is usually required. Although glucocorticoids work reliably and relatively quickly for sarcoidosis, these drugs are associated with numerous significant side effects. Such side effects are common in sarcoidosis patients, as the disease frequently has a chronic course and glucocorticoid treatment courses are often prolonged. For these reasons, corticosteroid-sparing and corticosteroid-replacing therapies are often required for sarcoidosis. Unfortunately, many healthcare providers who care for sarcoidosis patients are not familiar with the use of these agents. In this manuscript, we provide a review of the pharmacotherapy of sarcoidosis. We discuss the mechanism of action, dosing, side-effect profile, approach to monitoring and patient counselling concerning glucocorticoids, and the common alternative drugs recommended for use in the recent European Respiratory Society (Lausanne, Switzerland) Sarcoidosis Treatment Guidelines. We also discuss the use of these agents in special situations including hepatic insufficiency, renal insufficiency, pregnancy, breastfeeding, vaccination, and drug-drug interactions. It is hoped that this manuscript will provide valuable practical guidance to clinicians who care for sarcoidosis patients.
Collapse
Affiliation(s)
- Sooyeon Kwon
- Samuel S. Stratton Veterans Affairs Medical Center, Albany, NY 12208, USA
| | - Marc A. Judson
- Division of Pulmonary and Critical Care Medicine, Albany Medical College, Albany, NY 12208, USA;
| |
Collapse
|
22
|
Lee EG, Oh JE. From neglect to spotlight: the underappreciated role of B cells in cutaneous inflammatory diseases. Front Immunol 2024; 15:1328785. [PMID: 38426103 PMCID: PMC10902158 DOI: 10.3389/fimmu.2024.1328785] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Accepted: 01/30/2024] [Indexed: 03/02/2024] Open
Abstract
The skin, covering our entire body as its largest organ, manifests enormous complexities and a profound interplay of systemic and local responses. In this heterogeneous domain, B cells were considered strangers. Yet, recent studies have highlighted their existence in the skin and their distinct role in modulating cutaneous immunity across various immune contexts. Accumulating evidence is progressively shedding light on the significance of B cells in maintaining skin health and in skin disorders. Herein, we integrate current insights on the systemic and local contributions of B cells in three prevalent inflammatory skin conditions: Pemphigus Vulgaris (PV), Systemic Lupus Erythematosus (SLE), and Atopic Dermatitis (AD), underscoring the previously underappreciated importance of B cells within skin immunity. Moreover, we address the potential adverse effects of current treatments used for skin diseases, emphasizing their unintentional consequences on B cells. These comprehensive approaches may pave the way for innovative therapeutic strategies that effectively address the intricate nature of skin disorders.
Collapse
Affiliation(s)
- Eun-Gang Lee
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
| | - Ji Eun Oh
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
- BioMedical Research Center, KAIST, Daejeon, Republic of Korea
| |
Collapse
|
23
|
Rispens T, Kuijpers TW, Killestein J, van Kempen ZLE, Bloem K. Cross-Reactivity of Antibodies to Rituximab with Other Therapeutic Anti-CD20 Antibodies. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 212:529-533. [PMID: 38149924 DOI: 10.4049/jimmunol.2300647] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Accepted: 12/06/2023] [Indexed: 12/28/2023]
Abstract
One reason for a lack of response to rituximab as well as infusion-related anaphylactic adverse events is the development of antidrug Abs to rituximab. Besides rituximab, a number of other therapeutic Abs targeting CD20 are nowadays available as alternatives. In this study, we investigated the potential cross-reactivity of (human) anti-rituximab Abs to three other anti-CD20 mAbs: ofatumumab, obinutuzumab, and ocrelizumab. In 25 cases of anti-rituximab Abs, cross-reactivity was examined using both direct binding assays and inhibition immunoassays. Although no cross-reactivity was observed to ofatumumab or obinutuzumab, 8 of 25 samples also showed reactivity toward ocrelizumab in at least one of the two assays. Furthermore, in three cases of anti-ocrelizumab Abs, cross-reactivity to rituximab was observed in an inhibition immunoassay, albeit not in a direct binding assay. Our results suggest that obinutuzumab or ofatumumab are safe anti-CD20 alternatives in case of the presence of anti-rituximab Abs. It is advisable to proceed cautiously if switching from rituximab to ocrelizumab (or vice versa) is considered in case these alternatives may not be available.
Collapse
Affiliation(s)
- Theo Rispens
- Department of Immunopathology, Sanquin Research and Landsteiner Laboratory, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, the Netherlands
- Amsterdam Institute for Infection and Immunity, Amsterdam, the Netherlands
| | - Taco W Kuijpers
- Amsterdam Institute for Infection and Immunity, Amsterdam, the Netherlands
- Department of Pediatric Immunology, Rheumatology and Infectious Disease, Amsterdam UMC, location AMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Joep Killestein
- Amsterdam UMC location Vrije Universiteit Amsterdam, Neurology, Amsterdam, the Netherlands
| | - Zoé L E van Kempen
- Amsterdam UMC location Vrije Universiteit Amsterdam, Neurology, Amsterdam, the Netherlands
| | - Karien Bloem
- Antibodies and Immunogenicity, Sanquin Diagnostic Services, Amsterdam, the Netherlands
| |
Collapse
|
24
|
Brown P, Pratt AG, Hyrich KL. Therapeutic advances in rheumatoid arthritis. BMJ 2024; 384:e070856. [PMID: 38233032 DOI: 10.1136/bmj-2022-070856] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/19/2024]
Abstract
Rheumatoid arthritis (RA) is one of the most common immune mediated inflammatory diseases. People with rheumatoid arthritis present with pain, swelling, and stiffness that typically affects symmetrically distributed small and large joints. Without effective treatment, significant joint damage, disability, and work loss develop, owing to chronic inflammation of the joint lining (synovium). Over the past 25 years, the management of this condition has been revolutionized, resulting in substantially higher levels of disease remission and better long term outcomes. This improvement reflects a paradigm shift towards early and aggressive pharmacological intervention coupled with a proliferation in treatment choice, in turn related to enhanced pathobiological understanding and the advent of new drugs for rheumatoid arthritis. Following an overview of these developments from a historical perspective, and with a general audience in mind, this review focuses on newer, targeted treatments in an ever evolving landscape. The review highlights ongoing areas of debate and unmet need, including the proportion of patients with persistent, difficult-to-treat disease, despite recent advances. Also discussed are personalized, strategic approaches to individual patients, the role for imaging in clinical decision making, and the goal of sustained, drug free remission and disease prevention in the future.
Collapse
Affiliation(s)
- Philip Brown
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK
- National Institute for Health and Care Research Newcastle Biomedical Research Centre, Newcastle University, Newcastle upon Tyne Hospitals and Cumbria, Northumberland; and Tyne and Wear NHS Foundation Trusts, Newcastle upon Tyne, UK
| | - Arthur G Pratt
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK
- National Institute for Health and Care Research Newcastle Biomedical Research Centre, Newcastle University, Newcastle upon Tyne Hospitals and Cumbria, Northumberland; and Tyne and Wear NHS Foundation Trusts, Newcastle upon Tyne, UK
| | - Kimme L Hyrich
- Centre for Musculoskeletal Research, University of Manchester, Manchester Academic Health Sciences Centre, Manchester, UK
- National Institute for Health and Care Research Manchester Biomedical Research Centre, Manchester University Hospitals NHS Foundation Trust, Manchester, UK
| |
Collapse
|
25
|
Liao TL, Chen IC, Chen HW, Tang KT, Huang WN, Chen YH, Chen YM. Exosomal microRNAs as biomarkers for viral replication in tofacitinib-treated rheumatoid arthritis patients with hepatitis C. Sci Rep 2024; 14:937. [PMID: 38195767 PMCID: PMC10776842 DOI: 10.1038/s41598-023-50963-y] [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: 08/03/2023] [Accepted: 12/28/2023] [Indexed: 01/11/2024] Open
Abstract
Notwithstanding recent advances in direct antiviral specialists (DAAs) for hepatitis C infection (HCV), it is yet a pervasive overall issue in patients with rheumatoid arthritis (RA). Exosomal microRNAs (miRNAs) is associated with HCV infection. However, it remains unknown how miRNAs respond following biologic disease-modifying antirheumatic drug (bDMARD) and targeted synthetic DMARD (tsDMARD) treatment in HCV patients with RA. We prospectively recruited RA patients taking anti-tumor necrosis factor-α (TNF-α) inhibitors rituximab (RTX) and tofacitinib. The serum hepatitis C viral load was measured using real-time quantitative reverse transcriptase PCR before and 6 months after bDMARD and tsDMARD therapy. HCV RNA replication activity was measured using an HCV-tricistronic replicon reporter system, and quantitative analysis of hsa-mir-122-5p and hsa-mir-155-5p in patients was performed using quantitative PCR. HCV RNA replication in hepatocytes was not affected by tofacitinib or TNF-α inhibitor treatment. Hsa-mir-155-5p and hsa-mir-122-5p were significantly expanded in RA patients with HCV as compared with those without HCV. We observed a dramatic increase in hsa-mir-122-5p and a decrease in hsa-mir-155-5p expression levels in patients taking RTX in comparison with other treatments. Finally, a reduction in hsa-mir-122-5p and an increase in hsa-mir-155-5p were observed in a time-dependent manner after tofacitinib and DAA therapy in RA-HCV patients. These results showed that hsa-mir-155-5p and hsa-mir-122-5p were significantly increased in RA-HCV patients as compared with those without HCV after taking tofacitinib. Hsa-mir-155-5p and hsa-mir-122-5p may be potential biomarkers for treatment efficacy in RA patients with HCV.
Collapse
Affiliation(s)
- Tsai-Ling Liao
- Department of Medical Research, Taichung Veterans General Hospital, Taichung, Taiwan
- Rong-Hsing Research Center for Translational Medicine, National Chung-Hsing University, Taichung, Taiwan
- Ph.D. Program in Translational Medicine, National Chung Hsing University, Taichung, Taiwan
| | - I-Chieh Chen
- Department of Medical Research, Taichung Veterans General Hospital, Taichung, Taiwan
| | - Hong-Wei Chen
- Department of Medical Research, Taichung Veterans General Hospital, Taichung, Taiwan
| | - Kuo-Tung Tang
- Rong-Hsing Research Center for Translational Medicine, National Chung-Hsing University, Taichung, Taiwan
- Division of Allergy, Immunology and Rheumatology, Department of Internal Medicine, Taichung Veterans General Hospital, 1650, Section 4, Taiwan Boulevard, Xitun Dist., Taichung, 407, Taiwan
- School of Medicine, National Yang-Ming Chiao Tung University, Taipei, Taiwan
| | - Wen-Nan Huang
- Division of Allergy, Immunology and Rheumatology, Department of Internal Medicine, Taichung Veterans General Hospital, 1650, Section 4, Taiwan Boulevard, Xitun Dist., Taichung, 407, Taiwan
- School of Medicine, National Yang-Ming Chiao Tung University, Taipei, Taiwan
- Department of Post-Baccalaureate Medicine, College of Medicine, National Chung Hsing University, Taichung, Taiwan
| | - Yi-Hsing Chen
- Division of Allergy, Immunology and Rheumatology, Department of Internal Medicine, Taichung Veterans General Hospital, 1650, Section 4, Taiwan Boulevard, Xitun Dist., Taichung, 407, Taiwan
- School of Medicine, National Yang-Ming Chiao Tung University, Taipei, Taiwan
- Department of Post-Baccalaureate Medicine, College of Medicine, National Chung Hsing University, Taichung, Taiwan
| | - Yi-Ming Chen
- Department of Medical Research, Taichung Veterans General Hospital, Taichung, Taiwan.
- Rong-Hsing Research Center for Translational Medicine, National Chung-Hsing University, Taichung, Taiwan.
- Ph.D. Program in Translational Medicine, National Chung Hsing University, Taichung, Taiwan.
- Division of Allergy, Immunology and Rheumatology, Department of Internal Medicine, Taichung Veterans General Hospital, 1650, Section 4, Taiwan Boulevard, Xitun Dist., Taichung, 407, Taiwan.
- School of Medicine, National Yang-Ming Chiao Tung University, Taipei, Taiwan.
- Department of Post-Baccalaureate Medicine, College of Medicine, National Chung Hsing University, Taichung, Taiwan.
- Precision Medicine Research Center, College of Medicine, National Chung Hsing University, Taichung, Taiwan.
| |
Collapse
|
26
|
Silva TL, Lopes CS, Silva MC, Ferreira FB, Barros HLS, Silva MF, Silva NM, Oliveira F, Mineo TWP, Mineo JR. Ileal inflammation is reduced due to treatment with a metalloprotease from BmooMP-α-I snake venom in an experimental model of Toxoplasma gondii infection. Parasitol Res 2023; 123:65. [PMID: 38133827 DOI: 10.1007/s00436-023-08033-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2023] [Accepted: 11/09/2023] [Indexed: 12/23/2023]
Abstract
The selection process for advanced therapies in patients with inflammatory bowel diseases (IBDs) must prioritize safety, especially when considering new biologic agents or oral molecule modulators. In C57BL/6 mice, oral infection with Toxoplasma gondii induces intestinal inflammation through excessive tumor necrosis factor (TNF) production, making TNF neutralization a potential therapeutic intervention. Considering this, the present study aimed to evaluate the therapeutic effects of BmooMP-α-I, a snake venom metalloprotease isolated from Bothrops moojeni, which could promote TNF hydrolysis, in treating T. gondii-induced ileitis. The results showed that C57BL/6 mice orally infected with 50 cysts of T. gondii from the Me49 strain and treated with BmooMP-α-I exhibited prolonged survival and improved morbidity scores. Additionally, the treatment ameliorated both the macroscopic and microscopic aspects of the intestine, reduced macrophage influx, and decreased the production of inflammatory mediators by mesenteric lymph node cells. These findings provide compelling experimental evidence supporting the ability of BmooMP-α-I to alleviate ileal inflammation. Considering that the currently available therapeutic protocols are not completely effective and often result in side effects, the exploration of alternative strategies involving novel therapeutic agents, as demonstrated in this study, has the potential to significantly enhance the quality of life for patients suffering from inflammatory bowel diseases.
Collapse
Affiliation(s)
- Tamires Lopes Silva
- Laboratory of Immunoparasitology "Dr. Mario Endsfeldz Camargo", Institute of Biomedical Sciences, Federal University of Uberlândia, Uberlândia, Minas Gerais, Brazil
| | - Carolina Salomão Lopes
- Laboratory of Immunoparasitology "Dr. Mario Endsfeldz Camargo", Institute of Biomedical Sciences, Federal University of Uberlândia, Uberlândia, Minas Gerais, Brazil
| | - Maraisa Cristina Silva
- Laboratory of Immunoparasitology "Dr. Mario Endsfeldz Camargo", Institute of Biomedical Sciences, Federal University of Uberlândia, Uberlândia, Minas Gerais, Brazil
| | - Flávia Batista Ferreira
- Laboratory of Immunoparasitology "Dr. Mario Endsfeldz Camargo", Institute of Biomedical Sciences, Federal University of Uberlândia, Uberlândia, Minas Gerais, Brazil
| | - Heber Leão Silva Barros
- Laboratory of Immunoparasitology "Dr. Mario Endsfeldz Camargo", Institute of Biomedical Sciences, Federal University of Uberlândia, Uberlândia, Minas Gerais, Brazil
| | - Mariana Ferreira Silva
- Laboratory of Immunoparasitology "Dr. Mario Endsfeldz Camargo", Institute of Biomedical Sciences, Federal University of Uberlândia, Uberlândia, Minas Gerais, Brazil
| | - Neide Maria Silva
- Laboratory of Immunopathology, Institute of Biomedical Sciences, Federal University of Uberlândia, Uberlândia, Minas Gerais, Brazil
| | - Fabio Oliveira
- Laboratory of Biophysics, Institute of Biomedical Sciences, Federal University of Uberlândia, Uberlândia, Minas Gerais, Brazil
- National Institute of Science and Technology in Nano-Biopharmaceutical (N-Biofar), Belo Horizonte, MG, Brazil
| | - Tiago Wilson Patriarca Mineo
- Laboratory of Immunoparasitology "Dr. Mario Endsfeldz Camargo", Institute of Biomedical Sciences, Federal University of Uberlândia, Uberlândia, Minas Gerais, Brazil
| | - José Roberto Mineo
- Laboratory of Immunoparasitology "Dr. Mario Endsfeldz Camargo", Institute of Biomedical Sciences, Federal University of Uberlândia, Uberlândia, Minas Gerais, Brazil.
| |
Collapse
|
27
|
Farisogullari B, Santos EJF, Dures E, Geenen R, Machado PM. Efficacy of pharmacological interventions: a systematic review informing the 2023 EULAR recommendations for the management of fatigue in people with inflammatory rheumatic and musculoskeletal diseases. RMD Open 2023; 9:e003349. [PMID: 38056919 DOI: 10.1136/rmdopen-2023-003349] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Accepted: 07/14/2023] [Indexed: 12/08/2023] Open
Abstract
OBJECTIVE To identify the best evidence on the efficacy of pharmacological interventions in reducing fatigue in people with inflammatory rheumatic and musculoskeletal diseases (I-RMDs) and to summarise their safety in the identified studies to inform European Alliance of Associations for Rheumatology recommendations for the management of fatigue in people with I-RMDs. METHODS Systematic review of adults with I-RMDs conducted according to the Cochrane Handbook. Search strategy ran in Medline, Embase, Cochrane Library, CINAHL Complete, PEDro, OTseeker and PsycINFO. Only randomised controlled trials (RCTs) or controlled clinical trials were eligible. Assessment of risk of bias, data extraction and synthesis performed by two reviewers independently and in duplicate. Data pooled in statistical meta-analyses. RESULTS From 4151 records, 455 were selected for full-text review, 99 fulfilled the inclusion criteria and 19 RCTs were included in meta-analyses. Adalimumab was superior to placebo in reducing fatigue at 12 and 52 weeks in rheumatoid arthritis (RA) (n=3 and 2 RCTs; mean difference (MD)= -3.03, p<0.001; MD=-2.25, p=0.03, respectively). Golimumab (n=2 RCTs; 24 weeks: MD=-5.27, p<0.001), baricitinib (n=2 RCTs; 24 weeks: MD=-4.06, p<0.001), sarilumab (n=2 RCTs; 24 weeks: MD=-3.15, p<0.001), tocilizumab (n=3 RCTs; 24 weeks: MD=-3.69, p<0.001) and tofacitinib (n=3 RCTs; 12 weeks: MD=-4.44, p<0.001) were also superior to placebo in reducing fatigue in RA. A dose/effect relationship was observed for sarilumab, tocilizumab and tofacitinib. In spondyloarthritis (excluding psoriatic arthritis), secukinumab was superior to placebo in reducing fatigue at 16 weeks (n=2 RCTs; MD=-4.15, p<0.001), with a dose/effect relationship also observed. The narrative results of the RCTs not included in the meta-analysis indicated that several other pharmacological interventions were efficacious in reducing fatigue, with reassuring safety results. CONCLUSIONS Several pharmacological interventions are efficacious and generally safe for managing fatigue in people with I-RMDs.
Collapse
Affiliation(s)
| | - Eduardo José Ferreira Santos
- School of Health, Polytechnic University, Viseu, Portugal
- Health Sciences Research Unit: Nursing (UICiSA:E), Coimbra, Portugal
| | - Emma Dures
- Academic Rheumatology, Bristol Royal Infirmary, Bristol, UK
- School of Health and Social Wellbeing, University of the West of England, Bristol, UK
| | - Rinie Geenen
- Department of Psychology, Utrecht University, Utrecht, The Netherlands
| | - Pedro M Machado
- Centre for Rheumatology & Department of Neuromuscular Diseases, University College London, London, UK
- National Institute for Health Research (NIHR) University College London Hospitals Biomedical Research Centre, University College London Hospitals NHS Foundation Trust, London, UK
- Department of Rheumatology, Northwick Park Hospital, London North West University Healthcare NHS Trust, London, UK
| |
Collapse
|
28
|
Hu S, Tao Y, Hu F, Liu X. Diminished LAG3 + B cells correlate with exacerbated rheumatoid arthritis. Ann Med 2023; 55:2208373. [PMID: 37143367 PMCID: PMC10165927 DOI: 10.1080/07853890.2023.2208373] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Revised: 03/08/2023] [Accepted: 04/24/2023] [Indexed: 05/06/2023] Open
Abstract
BACKGROUND Lymphocyte activation gene-3 (LAG3) positive B cells have been identified as a novel regulatory B cell subset, while the role of LAG3+ B cells in the pathogenesis of rheumatoid arthritis (RA) remains elusive. MATERIALS AND METHODS Peripheral blood mononuclear cells (PBMCs) from RA, osteoarthritis (OA) patients and healthy volunteers were collected for flow cytometry staining of LAG3+ B cells. Their correlation with RA patient clinical and immunological features were analyzed. Moreover, the frequencies of LAG3+ B cells in collagen-induced arthritis (CIA) mice and naive mice were also detected. RESULTS A significant decrease of LAG3+ B cells was observed in RA patients as compared with healthy individuals and OA patients. Notably, the frequencies of LAG3+ B cells were negatively correlated with tender joint count (r = -0.4301, p = .0157) and DAS28-ESR (r = -0.4018, p = .025) in RA patients. In CIA mice, LAG3+ B cell frequencies were also decreased and negatively correlated with the CIA arthritis score. CONCLUSIONS Impairment of LAG3+ B cells potentially contributes to RA development. Reconstituting LAG3+ B cells might provide novel therapeutic strategies for the persistent disease.Key messagesLAG3+ B cells have been identified as a novel regulatory B cell subset. However, its role in the pathogenesis of RA remains unknown.This study revealed the decreased frequency of LAG3+ B cells in RA patients. Notably, LAG3+ B cells were negatively correlated with RA disease activity including the tender joint count and DAS28-ESR.In CIA mice, LAG3+ B cell frequencies were also decreased and negatively correlated with the CIA arthritis score.Reconstitution of LAG3+ B cells might provide novel therapeutic strategies for disease perpetuation.
Collapse
Affiliation(s)
- Suiyuan Hu
- Department of Rheumatology and Immunology, Peking University People’s Hospital, Beijing Key Laboratory for Rheumatism Mechanism and Immune Diagnosis (BZ0135), Beijing, China
| | - Yuting Tao
- Department of Rheumatology and Immunology, Peking University People’s Hospital, Beijing Key Laboratory for Rheumatism Mechanism and Immune Diagnosis (BZ0135), Beijing, China
| | - Fanlei Hu
- Department of Rheumatology and Immunology, Peking University People’s Hospital, Beijing Key Laboratory for Rheumatism Mechanism and Immune Diagnosis (BZ0135), Beijing, China
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, China
- Department of Integration of Chinese and Western Medicine, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Xu Liu
- Department of Rheumatology and Immunology, Peking University People’s Hospital, Beijing Key Laboratory for Rheumatism Mechanism and Immune Diagnosis (BZ0135), Beijing, China
| |
Collapse
|
29
|
Clarke D, Sartor L, Do V, Manolios N, Swaminathan S, Samarawickrama C. Biologics in peripheral ulcerative keratitis. Semin Arthritis Rheum 2023; 63:152269. [PMID: 37776666 DOI: 10.1016/j.semarthrit.2023.152269] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2023] [Revised: 08/23/2023] [Accepted: 09/04/2023] [Indexed: 10/02/2023]
Abstract
Over the past two decades biologic therapies have seen a rapid uptake in the management of ocular inflammation. Peripheral ulcerative keratitis (PUK), once a harbinger of blindness and mortality in refractory rheumatological disease, is now increasingly being treated with these agents. We conducted a review to evaluate the evidence base for this application and to provide a road map for their clinical usage in PUK, including dosage and adverse effects. A literature search across Medline, Embase and Cochrane Database of Systematic Reviews was undertaken to identify all patients with PUK that were treated with a biologic in a peer viewed article. Overall, whilst the evidence base for biologic use in PUK was poor, reported cases demonstrate an increasingly powerful and effective role for biologics in refractory PUK. This was particularly the case for rituximab in PUK secondary to granulomatous with polyangiitis.
Collapse
Affiliation(s)
- Daniel Clarke
- Department of Ophthalmology, Royal Hobart Hospital, Tasmania, Australia; Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
| | - Lauren Sartor
- Department of Ophthalmology, Westmead Hospital, Sydney, NSW, Australia; Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
| | - Vu Do
- Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
| | - Nicholas Manolios
- Department of Ophthalmology, Westmead Hospital, Sydney, NSW, Australia; Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
| | - Sanjay Swaminathan
- Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia; Department of Rheumatology, Westmead Hospital, Westmead, Sydney, New South Wales, Australia; Department of Clinical Immunology and Immunopathology, Westmead Hospital, Sydney, New South Wales, Australia; Centre for Immunology and Allergy Research, The Westmead Institute for Medical Research, Sydney, New South Wales, Australia
| | - Chameen Samarawickrama
- Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia; Translational Ocular Research and Immunology Consortium, Sydney University, New South Wales, Australia.
| |
Collapse
|
30
|
Kim JEJ, Tung LT, Jiang RR, Yousefi M, Liang Y, Malo D, Vidal SM, Nijnik A. Dysregulation of B lymphocyte development in the SKG mouse model of rheumatoid arthritis. Immunology 2023; 170:553-566. [PMID: 37688495 DOI: 10.1111/imm.13691] [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: 06/01/2023] [Accepted: 08/22/2023] [Indexed: 09/11/2023] Open
Abstract
Rheumatoid arthritis is a chronic and systemic inflammatory disease that affects approximately 1% of the world's population and is characterised by joint inflammation, the destruction of articular cartilage and bone, and many potentially life-threatening extraarticular manifestations. B lymphocytes play a central role in the pathology of rheumatoid arthritis as the precursors of autoantibody secreting plasma cells, as highly potent antigen-presenting cells, and as a source of various inflammatory cytokines, however, the effects of rheumatoid arthritis on B lymphocyte development remain poorly understood. Here, we analyse B lymphocyte development in murine models of rheumatoid arthritis, quantifying all the subsets of B cell precursors in the bone marrow and splenic B cells using flow cytometry. We demonstrate a severe reduction in pre-B cells and immature B cells in the bone marrow of mice with active disease, despite no major effects on the mature naïve B cell numbers. The loss of B cell precursors in the bone marrow of the affected mice was associated with a highly significant reduction in the proportion of Ki67+ cells, indicating impaired cell proliferation, while the viability of the B cell precursors was not significantly affected. We also observed some mobilisation of the B cell precursor cells into the mouse spleen, demonstrated with flow cytometry and pre-B colony forming units assays. In summary, the current work demonstrates a severe dysregulation in B lymphocyte development in murine rheumatoid arthritis, with possible implications for B cell repertoire formation, tolerance induction, and disease mechanisms.
Collapse
Affiliation(s)
- Joo Eun June Kim
- Department of Physiology, McGill University, Montreal, Quebec, Canada
- McGill University Research Centre on Complex Traits, McGill University, Montreal, Quebec, Canada
| | - Lin Tze Tung
- Department of Physiology, McGill University, Montreal, Quebec, Canada
- McGill University Research Centre on Complex Traits, McGill University, Montreal, Quebec, Canada
| | - Roselyn R Jiang
- Department of Physiology, McGill University, Montreal, Quebec, Canada
- McGill University Research Centre on Complex Traits, McGill University, Montreal, Quebec, Canada
| | - Mitra Yousefi
- McGill University Research Centre on Complex Traits, McGill University, Montreal, Quebec, Canada
- Department of Human Genetics, McGill University, Montreal, Quebec, Canada
| | - Yue Liang
- Department of Physiology, McGill University, Montreal, Quebec, Canada
- McGill University Research Centre on Complex Traits, McGill University, Montreal, Quebec, Canada
| | - Danielle Malo
- McGill University Research Centre on Complex Traits, McGill University, Montreal, Quebec, Canada
- Department of Human Genetics, McGill University, Montreal, Quebec, Canada
- Department of Medicine, McGill University, Montreal, Quebec, Canada
| | - Silvia M Vidal
- McGill University Research Centre on Complex Traits, McGill University, Montreal, Quebec, Canada
- Department of Human Genetics, McGill University, Montreal, Quebec, Canada
- Department of Microbiology and Immunology, McGill University, Montreal, Quebec, Canada
| | - Anastasia Nijnik
- Department of Physiology, McGill University, Montreal, Quebec, Canada
- McGill University Research Centre on Complex Traits, McGill University, Montreal, Quebec, Canada
| |
Collapse
|
31
|
Cignarella A, Vegeto E, Bolego C, Trabace L, Conti L, Ortona E. Sex-oriented perspectives in immunopharmacology. Pharmacol Res 2023; 197:106956. [PMID: 37820857 DOI: 10.1016/j.phrs.2023.106956] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 09/27/2023] [Accepted: 10/08/2023] [Indexed: 10/13/2023]
Abstract
Several immunopharmacological agents are effective in the treatment of cancer and immune-mediated conditions, with a favorable impact on life expectancy and clinical outcomes for a large number of patients. Nevertheless, response variation and undesirable effects of these drugs represent major issues, and overall efficacy remains unpredictable. Males and females show a distinct difference in immune system responses, with females generally mounting stronger responses to a variety of stimuli. Therefore, exploring sex differences in the efficacy and safety of immunopharmacological agents would strengthen the practice of precision medicine. As a pharmacological target highlight, programmed cell death 1 ligand 1 (PD-L1) is the first functionally characterized ligand of the coinhibitory programmed death receptor 1 (PD-1). The PD-L1/PD-1 crosstalk plays an important role in the immune response and is relevant in cancer, infectious and autoimmune disease. Sex differences in the response to immune checkpoint inhibitors are well documented, with male patients responding better than female patients. Similarly, higher efficacy of and adherence to tumor necrosis factor inhibitors in chronic inflammatory conditions including rheumatoid arthritis and Crohn's disease have been reported in male patients. The pharmacological basis of sex-specific responses to immune system modulating drugs is actively investigated in other settings such as stroke and type 1 diabetes. Advances in therapeutics targeting the endothelium could soon be wielded against autoimmunity and metabolic disorders. Based on the established sexual dimorphism in immune-related pathophysiology and disease presentation, sex-specific immunopharmacological protocols should be integrated into clinical guidelines.
Collapse
Affiliation(s)
| | - Elisabetta Vegeto
- Department of Pharmaceutical Sciences, University of Milan, Milan, Italy
| | - Chiara Bolego
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova, Italy
| | - Luigia Trabace
- Department of Clinical and Experimental Medicine, University of Foggia, Foggia, Italy
| | - Lucia Conti
- Center for Gender-Specific Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Elena Ortona
- Center for Gender-Specific Medicine, Istituto Superiore di Sanità, Rome, Italy
| |
Collapse
|
32
|
Rivellese F, Nerviani A, Giorli G, Warren L, Jaworska E, Bombardieri M, Lewis MJ, Humby F, Pratt AG, Filer A, Gendi N, Cauli A, Choy E, McInnes I, Durez P, Edwards CJ, Buch MH, Gremese E, Taylor PC, Ng N, Cañete JD, Raizada S, McKay ND, Jadon D, Sainaghi PP, Stratton R, Ehrenstein MR, Ho P, Pereira JP, Dasgupta B, Gorman C, Galloway J, Chinoy H, van der Heijde D, Sasieni P, Barton A, Pitzalis C. Stratification of biological therapies by pathobiology in biologic-naive patients with rheumatoid arthritis (STRAP and STRAP-EU): two parallel, open-label, biopsy-driven, randomised trials. THE LANCET. RHEUMATOLOGY 2023; 5:e648-e659. [PMID: 38251532 DOI: 10.1016/s2665-9913(23)00241-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 08/29/2023] [Accepted: 09/05/2023] [Indexed: 01/23/2024]
Abstract
BACKGROUND Despite highly effective targeted therapies for rheumatoid arthritis, about 40% of patients respond poorly, and predictive biomarkers for treatment choices are lacking. We did a biopsy-driven trial to compare the response to rituximab, etanercept, and tocilizumab in biologic-naive patients with rheumatoid arthritis stratified for synovial B cell status. METHODS STRAP and STRAP-EU were two parallel, open-label, biopsy-driven, stratified, randomised, phase 3 trials done across 26 university centres in the UK and Europe. Biologic-naive patients aged 18 years or older with rheumatoid arthritis based on American College of Rheumatology (ACR)-European League Against Rheumatism classification criteria and an inadequate response to conventional synthetic disease-modifying antirheumatic drugs (DMARDs) were included. Following ultrasound-guided synovial biopsy, patients were classified as B cell poor or B cell rich according to synovial B cell signatures and randomly assigned (1:1:1) to intravenous rituximab (1000 mg at week 0 and week 2), subcutaneous tocilizumab (162 mg per week), or subcutaneous etanercept (50 mg per week). The primary outcome was the 16-week ACR20 response in the B cell-poor, intention-to-treat population (defined as all randomly assigned patients), with data pooled from the two trials, comparing etanercept and tocilizumab (grouped) versus rituximab. Safety was assessed in all patients who received at least one dose of study drug. These trials are registered with the EU Clinical Trials Register, 2014-003529-16 (STRAP) and 2017-004079-30 (STRAP-EU). FINDINGS Between June 8, 2015, and July 4, 2019, 226 patients were randomly assigned to etanercept (n=73), tocilizumab (n=74), and rituximab (n=79). Three patients (one in each group) were excluded after randomisation because they received parenteral steroids in the 4 weeks before recruitment. 168 (75%) of 223 patients in the intention-to-treat population were women and 170 (76%) were White. In the B cell-poor population, ACR20 response at 16 weeks (primary endpoint) showed no significant differences between etanercept and tocilizumab grouped together and rituximab (46 [60%] of 77 patients vs 26 [59%] of 44; odds ratio 1·02 [95% CI 0·47-2·17], p=0·97). No differences were observed for adverse events, including serious adverse events, which occurred in six (6%) of 102 patients in the rituximab group, nine (6%) of 108 patients in the etanercept group, and three (4%) of 73 patients in the tocilizumab group (p=0·53). INTERPRETATION In this biologic-naive population of patients with rheumatoid arthrtitis, the dichotomic classification into synovial B cell poor versus rich did not predict treatment response to B cell depletion with rituximab compared with alternative treatment strategies. However, the lack of response to rituximab in patients with a pauci-immune pathotype and the higher risk of structural damage progression in B cell-rich patients treated with rituximab warrant further investigations into the ability of synovial tissue analyses to inform disease pathogenesis and treatment response. FUNDING UK Medical Research Council and Versus Arthritis.
Collapse
Affiliation(s)
- Felice Rivellese
- Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, Queen Mary University of London, London, UK; Barts Health NHS Trust and Barts Biomedical Research Centre, National Institute for Health and Care Research (NIHR), London, UK
| | - Alessandra Nerviani
- Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, Queen Mary University of London, London, UK; Barts Health NHS Trust and Barts Biomedical Research Centre, National Institute for Health and Care Research (NIHR), London, UK
| | - Giovanni Giorli
- Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, Queen Mary University of London, London, UK
| | - Louise Warren
- Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, Queen Mary University of London, London, UK
| | - Edyta Jaworska
- Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, Queen Mary University of London, London, UK
| | - Michele Bombardieri
- Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, Queen Mary University of London, London, UK; Barts Health NHS Trust and Barts Biomedical Research Centre, National Institute for Health and Care Research (NIHR), London, UK
| | - Myles J Lewis
- Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, Queen Mary University of London, London, UK; Barts Health NHS Trust and Barts Biomedical Research Centre, National Institute for Health and Care Research (NIHR), London, UK
| | - Frances Humby
- Barts Health NHS Trust and Barts Biomedical Research Centre, National Institute for Health and Care Research (NIHR), London, UK; Rheumatology Department, Guy's and St Thomas' NHS Foundation Trust, London, UK
| | - Arthur G Pratt
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK; Directorate of Musculoskeletal Services, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
| | - Andrew Filer
- Rheumatology Research Group, Institute for Inflammation and Ageing, NIHR Birmingham Biomedical Research Centre and Clinical Research Facility, University of Birmingham, Queen Elizabeth Hospital, Birmingham, UK
| | - Nagui Gendi
- Basildon University Hospital, Basildon and Thurrock University NHS Hospitals Foundation Trust, Basildon, UK
| | - Alberto Cauli
- Rheumatology Unit, AOU and University of Cagliari, Monserrato, Italy; UOC of Radiology, Ospedale SS Trinità, ATS Cagliari, Italy
| | - Ernest Choy
- CREATE Centre, Cardiff University, Cardiff, UK; Department of Rheumatology, University Hospital of Wales, Cardiff, UK
| | - Iain McInnes
- Glasgow Clinical Research Facility, Glasgow Royal Infirmary, Glasgow, UK
| | - Patrick Durez
- Institute of Experimental and Clinical Research, Université Catholique de Louvain, Brussels, Belgium; Department of Rheumatology, Cliniques Universitaires Saint-Luc, Brussels, Belgium
| | - Christopher J Edwards
- NIHR Southampton Clinical Research Facility, University Hospital Southampton, Southampton, UK; Faculty of Medicine, University of Southampton, Southampton, UK
| | - Maya H Buch
- Centre for Musculoskeletal Research, Division of Musculoskeletal and Dermatological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK; NIHR Manchester Biomedical Research Centre, Manchester, UK
| | - Elisa Gremese
- Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Peter C Taylor
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, Botnar Research Centre, University of Oxford, Oxford, UK
| | - Nora Ng
- Rheumatology Department, Guy's and St Thomas' NHS Foundation Trust, London, UK
| | - Juan D Cañete
- Rheumatology Department, Hospital Clínic de Barcelona, Barcelona, Spain; Institut d'Investigacions Biomèdiques August Pí I Sunyer, Barcelona, Spain
| | - Sabrina Raizada
- New Cross Hospital and Cannock Chase Hospital, Royal Wolverhampton NHS Trust, Wolverhampton, UK
| | - Neil D McKay
- Edinburgh Rheumatology Research Group and Rheumatic Diseases Unit, NHS Lothian, Edinburgh, UK
| | - Deepak Jadon
- Department of Medicine, University of Cambridge, Cambridge, UK
| | - Pier Paolo Sainaghi
- Department of Rheumatology, University Eastern Piedmont and Maggiore della Carita Hospital, Novara, Italy
| | - Richard Stratton
- Royal Free Hospital, Royal Free London NHS Foundation Trust, London, UK
| | | | - Pauline Ho
- The Kellgren Centre for Rheumatology, Manchester Royal Infirmary, Manchester University NHS Foundation Trust, Manchester, UK
| | - Joaquim P Pereira
- Rheumatology Department, Hospital De Santa Maria, Centro Hospitalar Universitário Lisboa Norte, Lisbon, Portugal; Rheumatology Research Unit, Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - Bhaskar Dasgupta
- Rheumatology Department, Mid and South Essex University Hospitals NHS Foundation Trust, Southend University Hospital, Westcliff-on-Sea, UK
| | - Claire Gorman
- Department of Rheumatology, Homerton University Hospital, Homerton Healthcare NHS Foundation Trust, London, UK
| | - James Galloway
- King's College Hospital, King's College Hospital NHS Foundation Trust, London, UK
| | - Hector Chinoy
- Centre for Musculoskeletal Research, Division of Musculoskeletal and Dermatological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK; Department of Rheumatology, Salford Royal Hospital, Northern Care Alliance NHS Foundation Trust, Manchester Academic Health Science Centre, Salford, UK
| | | | - Peter Sasieni
- King's Clinical Trials Unit, Kings College London, London, UK
| | - Anne Barton
- Centre for Musculoskeletal Research, Division of Musculoskeletal and Dermatological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Costantino Pitzalis
- Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, Queen Mary University of London, London, UK; Barts Health NHS Trust and Barts Biomedical Research Centre, National Institute for Health and Care Research (NIHR), London, UK; IRCCS Humanitas Research Hospital, Milan, Italy.
| |
Collapse
|
33
|
Abduljaleel YR, Jwaid AH, Hussein AK. Evaluating the effect of ursodeoxycholic acid (UDCA) in comparison with dexamethasone and diclofenac in a rat model of rheumatoid arthritis. J Med Life 2023; 16:1519-1526. [PMID: 38313166 PMCID: PMC10835553 DOI: 10.25122/jml-2023-0107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Accepted: 05/08/2023] [Indexed: 02/06/2024] Open
Abstract
Ursodeoxycholic acid (UDCA) is known for its major effects on the liver, but its impact on autoimmune diseases is not well understood. This study aimed to assess the effectiveness of UDCA in controlling rheumatoid arthritis (RA) in an in vivo setting. Experimental RA was induced in rats using Freund's complete adjuvant, and the effects of UDCA (50,100 mg/kg) were compared to those of dexamethasone and diclofenac by measuring changes in paw size, IL-17, pro-inflammatory cytokines, oxidative stress (GSH, MDA), and radiological changes. The administration of UDCA resulted in decreased cartilage damage, reduced paw edema, and a decrease in the release of pro-inflammatory cytokines and oxidative stress. Additionally, X-ray joint alterations were observed in the UDCA-treated group compared to the dexamethasone and diclofenac groups. These results suggest that UDCA has anti-rheumatoid arthritis properties due to its ability to minimize oxidative stress and inflammation in arthritis-affected rats.
Collapse
Affiliation(s)
- Yamama Raad Abduljaleel
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Baghdad, Baghdad, Iraq
| | - Ahmed Hamed Jwaid
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Baghdad, Baghdad, Iraq
| | | |
Collapse
|
34
|
Schneider K, Arandjelovic S. Apoptotic cell clearance components in inflammatory arthritis. Immunol Rev 2023; 319:142-150. [PMID: 37507355 PMCID: PMC10615714 DOI: 10.1111/imr.13256] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2023] [Accepted: 07/18/2023] [Indexed: 07/30/2023]
Abstract
Rheumatoid arthritis (RA) is a chronic inflammatory disease of the synovial joints that affects ~1% of the human population. Joint swelling and bone erosion, hallmarks of RA, contribute to disability and, sometimes, loss of life. Mechanistically, disease is driven by immune dysregulation characterized by circulating autoantibodies, inflammatory mediators, tissue degradative enzymes, and metabolic dysfunction of resident stromal and recruited immune cells. Cell death by apoptosis has been therapeutically explored in animal models of RA due to the comparisons drawn between synovial hyperplasia and paucity of apoptosis in RA with the malignant transformation of cancer cells. Several efforts to induce cell death have shown benefits in reducing the development and/or severity of the disease. Apoptotic cells are cleared by phagocytes in a process known as efferocytosis, which differs from microbial phagocytosis in its "immuno-silent," or anti-inflammatory, nature. Failures in efferocytosis have been linked to autoimmune disease, whereas administration of apoptotic cells in RA models effectively inhibits inflammatory indices, likely though efferocytosis-mediated resolution-promoting mechanisms. However, the nature of signaling pathways elicited and the molecular identity of clearance mediators in RA are understudied. Furthermore, canonical efferocytosis machinery elements also play important non-canonical functions in homeostasis and pathology. Here, we discuss the roles of efferocytosis machinery components in models of RA and discuss their potential involvement in disease pathophysiology.
Collapse
Affiliation(s)
- Kevin Schneider
- University of Virginia, Center for Immunity, Inflammation and Regenerative Medicine, Department of Medicine, Charlottesville, VA, USA
| | - Sanja Arandjelovic
- University of Virginia, Center for Immunity, Inflammation and Regenerative Medicine, Department of Medicine, Charlottesville, VA, USA
| |
Collapse
|
35
|
Minopoulou I, Kleyer A, Yalcin-Mutlu M, Fagni F, Kemenes S, Schmidkonz C, Atzinger A, Pachowsky M, Engel K, Folle L, Roemer F, Waldner M, D'Agostino MA, Schett G, Simon D. Imaging in inflammatory arthritis: progress towards precision medicine. Nat Rev Rheumatol 2023; 19:650-665. [PMID: 37684361 DOI: 10.1038/s41584-023-01016-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/31/2023] [Indexed: 09/10/2023]
Abstract
Imaging techniques such as ultrasonography and MRI have gained ground in the diagnosis and management of inflammatory arthritis, as these imaging modalities allow a sensitive assessment of musculoskeletal inflammation and damage. However, these techniques cannot discriminate between disease subsets and are currently unable to deliver an accurate prediction of disease progression and therapeutic response in individual patients. This major shortcoming of today's technology hinders a targeted and personalized patient management approach. Technological advances in the areas of high-resolution imaging (for example, high-resolution peripheral quantitative computed tomography and ultra-high field MRI), functional and molecular-based imaging (such as chemical exchange saturation transfer MRI, positron emission tomography, fluorescence optical imaging, optoacoustic imaging and contrast-enhanced ultrasonography) and artificial intelligence-based data analysis could help to tackle these challenges. These new imaging approaches offer detailed anatomical delineation and an in vivo and non-invasive evaluation of the immunometabolic status of inflammatory reactions, thereby facilitating an in-depth characterization of inflammation. By means of these developments, the aim of earlier diagnosis, enhanced monitoring and, ultimately, a personalized treatment strategy looms closer.
Collapse
Affiliation(s)
- Ioanna Minopoulou
- Department of Internal Medicine 3, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
- Deutsches Zentrum Immuntherapie (DZI), Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Arnd Kleyer
- Department of Internal Medicine 3, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
- Deutsches Zentrum Immuntherapie (DZI), Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Melek Yalcin-Mutlu
- Department of Internal Medicine 3, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
- Deutsches Zentrum Immuntherapie (DZI), Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Filippo Fagni
- Department of Internal Medicine 3, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
- Deutsches Zentrum Immuntherapie (DZI), Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Stefan Kemenes
- Department of Internal Medicine 3, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
- Deutsches Zentrum Immuntherapie (DZI), Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Christian Schmidkonz
- Department of Nuclear Medicine, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
- Institute for Medical Engineering, University of Applied Sciences Amberg-Weiden, Weiden, Germany
| | - Armin Atzinger
- Department of Nuclear Medicine, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Milena Pachowsky
- Department of Internal Medicine 3, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
- Deutsches Zentrum Immuntherapie (DZI), Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
| | | | - Lukas Folle
- Pattern Recognition Lab, Friedrich-Alexander-University Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Frank Roemer
- Institute of Radiology, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
- Department of Radiology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Maximilian Waldner
- Deutsches Zentrum Immuntherapie (DZI), Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
- Department of Internal Medicine 1, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Maria-Antonietta D'Agostino
- Division of Rheumatology, Catholic University of the Sacred Heart, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
- Université Paris-Saclay, UVSQ, Inserm U1173, Infection et Inflammation, Laboratory of Excellence Inflamex, Montigny-Le-Bretonneux, France
| | - Georg Schett
- Department of Internal Medicine 3, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
- Deutsches Zentrum Immuntherapie (DZI), Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
| | - David Simon
- Department of Internal Medicine 3, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany.
- Deutsches Zentrum Immuntherapie (DZI), Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany.
| |
Collapse
|
36
|
Jia N, Gao Y, Li M, Liang Y, Li Y, Lin Y, Huang S, Lin Q, Sun X, He Q, Yao Y, Zhang B, Zhang Z, Zhang L. Metabolic reprogramming of proinflammatory macrophages by target delivered roburic acid effectively ameliorates rheumatoid arthritis symptoms. Signal Transduct Target Ther 2023; 8:280. [PMID: 37500654 PMCID: PMC10374631 DOI: 10.1038/s41392-023-01499-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2022] [Revised: 05/07/2023] [Accepted: 05/15/2023] [Indexed: 07/29/2023] Open
Abstract
Rheumatoid arthritis (RA) is a common chronic inflammatory disorder that usually affects joints. It was found that roburic acid (RBA), an ingredient from anti-RA herb Gentiana macrophylla Pall., displayed strong anti-inflammatory activity. However, its medical application is limited by its hydrophobicity, lack of targeting capability and unclear functional mechanism. Here, we constructed a pH responsive dual-target drug delivery system hitchhiking RBA (RBA-NPs) that targeted both CD44 and folate receptors, and investigated its pharmacological mechanism. In rat RA model, the nanocarriers effectively delivered RBA to inflammatory sites and significantly enhanced the therapeutic outcomes compared with free RBA, as well as strongly reducing inflammatory cytokine levels and promoting tissue repair. Following analysis revealed that M1 macrophages in the joints were reprogrammed to M2 phenotype by RBA. Since the balance of pro- and anti-inflammatory macrophages play important roles in maintaining immune homeostasis and preventing excessive inflammation in RA, this reprogramming is likely responsible for the anti-RA effect. Furthermore, we revealed that RBA-NPs drove M1-to-M2 phenotypic switch by down-regulating the glycolysis level via blocking ERK/HIF-1α/GLUT1 pathway. Thus, our work not only developed a targeting delivery system that remarkably improved the anti-RA efficiency of RBA, but also identified a potential molecular target to reversely reprogram macrophages though energy metabolism regulation.
Collapse
Affiliation(s)
- Na Jia
- Key Laboratory of Drug Targeting and Drug Delivery Systems, Ministry of Education, West China School of Pharmacy, College of Polymer Science and Engineering, State key Laboratory of Polymer Materials Engineering, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, 610041, P. R. China
| | - Yunzhen Gao
- Key Laboratory of Drug Targeting and Drug Delivery Systems, Ministry of Education, West China School of Pharmacy, College of Polymer Science and Engineering, State key Laboratory of Polymer Materials Engineering, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, 610041, P. R. China
| | - Min Li
- Key Laboratory of Drug Targeting and Drug Delivery Systems, Ministry of Education, West China School of Pharmacy, College of Polymer Science and Engineering, State key Laboratory of Polymer Materials Engineering, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, 610041, P. R. China
| | - Yi Liang
- Key Laboratory of Drug Targeting and Drug Delivery Systems, Ministry of Education, West China School of Pharmacy, College of Polymer Science and Engineering, State key Laboratory of Polymer Materials Engineering, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, 610041, P. R. China
| | - Yuwen Li
- Department of Pharmacy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Yunzhu Lin
- Key Laboratory of Drug Targeting and Drug Delivery Systems, Ministry of Education, West China School of Pharmacy, College of Polymer Science and Engineering, State key Laboratory of Polymer Materials Engineering, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, 610041, P. R. China
| | - Shiqi Huang
- Key Laboratory of Drug Targeting and Drug Delivery Systems, Ministry of Education, West China School of Pharmacy, College of Polymer Science and Engineering, State key Laboratory of Polymer Materials Engineering, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, 610041, P. R. China
| | - Qing Lin
- Key Laboratory of Drug Targeting and Drug Delivery Systems, Ministry of Education, West China School of Pharmacy, College of Polymer Science and Engineering, State key Laboratory of Polymer Materials Engineering, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, 610041, P. R. China
| | - Xun Sun
- Key Laboratory of Drug Targeting and Drug Delivery Systems, Ministry of Education, West China School of Pharmacy, College of Polymer Science and Engineering, State key Laboratory of Polymer Materials Engineering, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, 610041, P. R. China
| | - Qin He
- Key Laboratory of Drug Targeting and Drug Delivery Systems, Ministry of Education, West China School of Pharmacy, College of Polymer Science and Engineering, State key Laboratory of Polymer Materials Engineering, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, 610041, P. R. China
| | - Yuqin Yao
- Key Laboratory of Drug Targeting and Drug Delivery Systems, Ministry of Education, West China School of Pharmacy, College of Polymer Science and Engineering, State key Laboratory of Polymer Materials Engineering, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, 610041, P. R. China
| | - Ben Zhang
- Key Laboratory of Drug Targeting and Drug Delivery Systems, Ministry of Education, West China School of Pharmacy, College of Polymer Science and Engineering, State key Laboratory of Polymer Materials Engineering, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, 610041, P. R. China
| | - Zhirong Zhang
- Key Laboratory of Drug Targeting and Drug Delivery Systems, Ministry of Education, West China School of Pharmacy, College of Polymer Science and Engineering, State key Laboratory of Polymer Materials Engineering, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, 610041, P. R. China
| | - Ling Zhang
- Key Laboratory of Drug Targeting and Drug Delivery Systems, Ministry of Education, West China School of Pharmacy, College of Polymer Science and Engineering, State key Laboratory of Polymer Materials Engineering, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, 610041, P. R. China.
| |
Collapse
|
37
|
Shipa MRA, Di Cicco M, Balogh E, Nitu NA, Mainuddin MD, Bhadauria N, Mukerjee D, Roussou E. Drug-survival profiling of second-line biologic therapy in rheumatoid arthritis: Choice of another tumour necrosis factor inhibitor or a biologic of different mode of action? Mod Rheumatol 2023; 33:700-707. [PMID: 35920402 DOI: 10.1093/mr/roac086] [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: 06/01/2022] [Revised: 07/07/2022] [Accepted: 07/22/2022] [Indexed: 11/14/2022]
Abstract
OBJECTIVES To assess the best choice of second-line therapy between tumour necrosis factor-inhibitor (TNFi) and biologics of different-mode-of-action (BDMA-rituximab/tocilizumab/abatacept) in rheumatoid arthritis (RA) by evaluating drug-survival following discontinuation of the first-line TNFi. METHODS This retrospective drug-survival study was performed across two different hospitals by conventional-statistics and machine-learning approach. RESULTS From a total of 435 patients, 213 (48.9%; TNFi = 122, BDMA = 91) discontinued their second-line biologic {median drug-survival: TNFi, 27 months [95% confidence interval (95%CI) 22-32] vs BDMA, 37 months (95%CI 32-52)}. As a second-line biologic, BDMA was likely to reduce the risk of treatment-discontinuation [hazard-ratio (HR) 0.63, 95%CI 0.48-0.83] compared to TNFi, but only in seropositive-patients (HR 0.52, 95%CI 0.38-0.73), not in seronegative-RA. Drug-survival benefit of BDMA over TNFi was not observed if the seropositive-patients were previously exposed to monoclonal-TNFi (HR 0.77, 95%CI 0.49-1.22) versus soluble-TNFi (etanercept/biosimilars) or if the first-line TNFi was terminated within 23.9 months of initiation (HR 0.97, 95%CI 0.56-1.68). CONCLUSIONS BDMA, as a second-line biologic, is more likely to be sustained in seropositive-patients, particularly without prior exposure to monoclonal-TNFi. The drug-survival benefit of BDMA was not observed in seronegative-patients or if the first-line TNFi was stopped within 2 years.
Collapse
Affiliation(s)
- Muhammad R A Shipa
- Centre for Rheumatology, Division of Medicine, University College London, London, UK; Rayne Institute, 5 University St, Bloomsbury, London WC1E 6JF, UK
| | - Maria Di Cicco
- Department of Rheumatology, Barking Havering and Redbridge University Hospitals NHS Trust, London, UK
| | - Emese Balogh
- Department of Rheumatology, Barking Havering and Redbridge University Hospitals NHS Trust, London, UK
| | - Naila A Nitu
- Department of Rheumatology, North Middlesex University Trust, London, UK
| | - M D Mainuddin
- Department of Rheumatology, North Middlesex University Trust, London, UK
| | - Naveen Bhadauria
- Department of Rheumatology, North Middlesex University Trust, London, UK
| | - Dev Mukerjee
- Department of Rheumatology, North Middlesex University Trust, London, UK
| | - Euthalia Roussou
- Department of Rheumatology, Barking Havering and Redbridge University Hospitals NHS Trust, London, UK
| |
Collapse
|
38
|
Connolly CM, Paik JJ. Clinical pearls and promising therapies in myositis. Expert Rev Clin Immunol 2023; 19:797-811. [PMID: 37158055 PMCID: PMC10330909 DOI: 10.1080/1744666x.2023.2212162] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Accepted: 05/05/2023] [Indexed: 05/10/2023]
Abstract
INTRODUCTION Idiopathic inflammatory myopathies (IIMs) represent a diverse group of systemic autoimmune disorders with variable clinical manifestations and disease course. Currently, the challenges of IIMs are multifold, including difficulties in timely diagnosis owing to clinical heterogeneity, limited insights into disease pathogenesis, as well as a restricted number of available therapies. However, advances utilizing myositis-specific autoantibodies have facilitated the definition of subgroups as well as the prediction of clinical phenotypes, disease course, and response to treatment. AREAS COVERED Herein we provide an overview of the clinical presentations of dermatomyositis, anti-synthetase syndrome, immune-mediated necrotizing myopathy, and inclusion body myositis. We then provide an updated review of available and promising therapies for each of these disease groups. We synthesize current treatment recommendations in the context of case-based construct to facilitate application to patient care. Finally, we provide high-yield, clinical pearls relevant to each of the subgroups that can be incorporated into clinical reasoning. EXPERT OPINION There are many exciting developments on the horizon for IIM. As insights into pathogenesis evolve, the therapeutic armamentarium is expanding with many novel therapies in development, holding promise for more targeted treatment approaches.
Collapse
Affiliation(s)
- Caoilfhionn M. Connolly
- Division of Rheumatology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Julie J. Paik
- Division of Rheumatology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD
| |
Collapse
|
39
|
Boumaza X, Lafaurie M, Treiner E, Walter O, Pugnet G, Martin-Blondel G, Biotti D, Ciron J, Constantin A, Tauber M, Puisset F, Moulis G, Alric L, Renaudineau Y, Chauveau D, Sailler L. Infectious risk when prescribing rituximab in patients with hypogammaglobulinemia acquired in the setting of autoimmune diseases. Int Immunopharmacol 2023; 120:110342. [PMID: 37276827 DOI: 10.1016/j.intimp.2023.110342] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Revised: 05/03/2023] [Accepted: 05/12/2023] [Indexed: 06/07/2023]
Abstract
We conducted a single-centre retrospective cohort study in a French University Hospital between 2010 and 2018 to describe the risk of severe infectious event (SIE) within 2 years after the date of first rituximab infusion (T0) prescribed after the evidence of acquired hypogammaglobulinemia (gamma globulins [GG] ≤ 6 g/L) in the setting of autoimmune diseases (AID) other than rheumatoid arthritis. SIE occurred in 26 out of 121 included patients. Two years cumulative incidence rates were 12.7 % (95 % CI 5.1-23.9) in the multiple sclerosis/neuromyelitis optica spectrum disorder group (n = 48), 27.6 % (95 % CI 15.7-40.9) in the ANCA-associated vasculitis group (n = 48) and 30.6 % (95 % CI 13.1-50.3) in the 'other AID' group (n = 25). Median GG level at T0 was 5.3 g/l (IQR 4.1-5.6) in the 'SIE' group and 5.6 g/l (IQR 4.7-5.8) in the 'no SIE' group (p = 0.04). In regression analysis, risk of SIE increased with Charlson comorbidity index ≥ 3 (OR 2.77; 95 % CI 1.01-7.57), lung disease (OR 3.20; 95 % CI 1.27-7.99), GG < 4 g/L (OR 3.39; 95 % CI 1.02-11.19), concomitant corticosteroid therapy (OR 4.13; 95 % CI 1.63-10.44), previous cyclophosphamide exposure (OR 2.69; 95 % CI 1.10-6.61), a lymphocyte count < 1000 cells/µL (OR 2.86; 95 % CI 1.12-7.21) and absence of pneumococcal vaccination (OR 3.50; 95 % CI 1.41-8.70). These results may help to inform clinical decision when considering a treatment by rituximab in immunosuppressed AID patients with hypogammaglobulinemia.
Collapse
Affiliation(s)
- Xavier Boumaza
- Service de Médecine Interne Purpan, Centre Hospitalier Universitaire de Toulouse, France; Service des Maladies Infectieuses et Tropicales, Centre Hospitalier Universitaire de Toulouse, France.
| | - Margaux Lafaurie
- Service de Pharmacologie Médicale et Clinique, Centre Hospitalier Universitaire de Toulouse, France; Centre d'Investigation Clinique 1436, Equipe PEPSS, Centre Hospitalier Universitaire de Toulouse, INSERM, Toulouse, France
| | - Emmanuel Treiner
- Laboratoire d'Immunologie, Institut Fédératif de Biologie, Centre Hospitalier Universitaire de Toulouse, France; Centre de Physiopathologie de Toulouse-Purpan, Centre Hospitalier Universitaire de Toulouse, France
| | - Ondine Walter
- Service de Médecine Interne Purpan, Centre Hospitalier Universitaire de Toulouse, France; Centre d'Investigation Clinique 1436, Equipe PEPSS, Centre Hospitalier Universitaire de Toulouse, INSERM, Toulouse, France
| | - Gregory Pugnet
- Service de Médecine Interne et immunologie clinique Rangueil, Centre Hospitalier Universitaire de, Toulouse, France
| | - Guillaume Martin-Blondel
- Service des Maladies Infectieuses et Tropicales, Centre Hospitalier Universitaire de Toulouse, France; Institut Toulousain des Maladies Infectieuses et Inflammatoires (Infinity), INSERM UMR1291 - CNRS UMR5051 - Université Toulouse III, France
| | - Damien Biotti
- Service de Neurologie, Centre Hospitalier Universitaire de Toulouse, France
| | - Jonathan Ciron
- Service de Neurologie, Centre Hospitalier Universitaire de Toulouse, France
| | - Arnaud Constantin
- Service de Rhumatologie, Centre Hospitalier Universitaire de Toulouse, France
| | - Marie Tauber
- Service de Dermatologie, Centre Hospitalier Universitaire de Toulouse, France
| | - Florent Puisset
- Service de Pharmacologie, Institut Universitaire du Cancer Oncopole, France
| | - Guillaume Moulis
- Service de Médecine Interne Purpan, Centre Hospitalier Universitaire de Toulouse, France; Centre d'Investigation Clinique 1436, Equipe PEPSS, Centre Hospitalier Universitaire de Toulouse, INSERM, Toulouse, France
| | - Laurent Alric
- Service de Médecine Interne et immunologie clinique Rangueil, Centre Hospitalier Universitaire de, Toulouse, France
| | - Yves Renaudineau
- Laboratoire d'Immunologie, Institut Fédératif de Biologie, Centre Hospitalier Universitaire de Toulouse, France; Institut Toulousain des Maladies Infectieuses et Inflammatoires (Infinity), INSERM UMR1291 - CNRS UMR5051 - Université Toulouse III, France
| | - Dominique Chauveau
- Service de Néphrologie et Transplantation d'Organes, Centre de Référence Maladies Rénales Rares, Centre Hospitalier Universitaire de Toulouse, France
| | - Laurent Sailler
- Service de Médecine Interne Purpan, Centre Hospitalier Universitaire de Toulouse, France; Centre d'Investigation Clinique 1436, Equipe PEPSS, Centre Hospitalier Universitaire de Toulouse, INSERM, Toulouse, France
| |
Collapse
|
40
|
Cacciapaglia F, Stano S, Fornaro M, Iannone F. Subcutaneous immunoglobulin therapy for refractory skin thickening in rapidly progressive systemic sclerosis: A case report and literature review. JOURNAL OF SCLERODERMA AND RELATED DISORDERS 2023; 8:101-106. [PMID: 37287951 PMCID: PMC10242697 DOI: 10.1177/23971983221124180] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Accepted: 08/17/2022] [Indexed: 06/04/2024]
Abstract
The use of immunoglobulin is a therapeutic option with increasing evidence of efficacy for different rheumatologic autoimmune systemic diseases. Some studies concerning immunoglobulin use in systemic sclerosis have been published with encouraging results. We present the case of a young woman diagnosed with rapidly progressive diffuse cutaneous systemic sclerosis, refractory to therapy with methotrexate and rituximab, which presented a relevant skin improvement after one year of subcutaneous immunoglobulin (2 g/kg cumulative monthly dose, refracted in weekly administrations). Furthermore, a narrative literature review of the evidence for alternative treatments with a focus on immunoglobulin use for systemic sclerosis skin involvement was carried out.
Collapse
Affiliation(s)
- Fabio Cacciapaglia
- Rheumatology Unit, Department of Emergency and Transplantation of Organs (DETO), University of Bari Aldo Moro, Bari, Italy
| | - Stefano Stano
- Rheumatology Unit, Department of Emergency and Transplantation of Organs (DETO), University of Bari Aldo Moro, Bari, Italy
| | - Marco Fornaro
- Rheumatology Unit, Department of Emergency and Transplantation of Organs (DETO), University of Bari Aldo Moro, Bari, Italy
| | - Florenzo Iannone
- Rheumatology Unit, Department of Emergency and Transplantation of Organs (DETO), University of Bari Aldo Moro, Bari, Italy
| |
Collapse
|
41
|
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: 3] [Impact Index Per Article: 3.0] [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
|
42
|
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: 2] [Impact Index Per Article: 2.0] [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
|
43
|
Ansari MA, Nadeem A, Attia SM, Bakheet SA, Alasmari AF, Alomar HA, Al-Mazroua HA, Alhamed AS, Shahid M, Alqinyah M, Assiri MA, Al-Hamamah MA, Alassmrry YA, Ahmad SF. Rituximab exerts its anti-arthritic effects via inhibiting NF-κB/GM-CSF/iNOS signaling in B cells in a mouse model of collagen-induced arthritis. Heliyon 2023; 9:e16673. [PMID: 37274671 PMCID: PMC10238934 DOI: 10.1016/j.heliyon.2023.e16673] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 05/23/2023] [Accepted: 05/24/2023] [Indexed: 06/06/2023] Open
Abstract
Rheumatoidarthritis (RA) is an autoimmune disease characterized by uncontrolled joint inflammation and damage to bone and cartilage. B cells are known to play a crucial role in the pathogenesis and development of arthritis. Previous studies have found that B cells may be a potential target for treating RA. Rituximab, a monoclonal antibody targeting B cells, has induced long-term clinical responses in RA. Collagen-induced arthritis (CIA) mouse model is a widely studied autoimmune model of RA. CIA mouse model was used to investigate the effect of rituximab on the RA severity in the mice. Following induction of CIA, animals were treated with rituximab (250 mg/kg/week) intraperitoneally on the days 28, 35, 42, 49, 56, and 63 after collagen induction. We investigated the effect of rituximab on NF-κB p65, IκBα, GM-CSF, MCP-1, iNOS, TNF-α, and IL-6 cells in splenic CD19+ and CD45R+ B cells using flow cytometry. We also assessed the effect of rituximab on NF-κB p65, GM-CSF, IκBα, MCP-1, iNOS, TNF-α, and IL-6 at mRNA levels using RT-PCR analyses of knee tissues. Rituximab treatment significantly decreased CD19+NF-κB p65+, CD45R+NF-κB p65+, CD19+GM-CSF+, CD45R+GM-CSF+, CD19+MCP-1+, CD45R+MCP-1+, CD19+TNF-α+, CD45R+TNF-α+, CD19+iNOS+, CD45R+iNOS+, CD19+IL-6+, and CD45R+IL-6+, and increased CD45R+IκBα+ in spleen cells of CIA mice. We further observed that rituximab treatment downregulated NF-κB p65, GM-CSF, MCP-1, iNOS, TNF-α, and IL-6, whereas it upregulated IκBα, mRNA level. All these findings suggest that rituximab may be a novel therapeutic target for the treatment of RA.
Collapse
Affiliation(s)
- Mushtaq A. Ansari
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Ahmed Nadeem
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Sabry M. Attia
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Saleh A. Bakheet
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Abdullah F. Alasmari
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Hatun A. Alomar
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Haneen A. Al-Mazroua
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Abdullah S. Alhamed
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Mudassar Shahid
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Mohammed Alqinyah
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Mohammed A. Assiri
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Mohammed A. Al-Hamamah
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Yasseen A. Alassmrry
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Sheikh F. Ahmad
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| |
Collapse
|
44
|
Cannon L, Pan A, Kovalick L, Sarkissian A, Wu EY. Secondary immunodeficiencies and infectious considerations of biologic immunomodulatory therapies. Ann Allergy Asthma Immunol 2023; 130:718-726. [PMID: 36801438 PMCID: PMC10247415 DOI: 10.1016/j.anai.2023.02.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 02/02/2023] [Accepted: 02/13/2023] [Indexed: 02/19/2023]
Abstract
Biologic immunomodulatory medications have rapidly expanded in the previous decades, providing new treatment options for individuals with a spectrum of oncologic, allergic, rheumatologic, and neurologic conditions. Biologic therapies alter immune function and can impair key host defense mechanisms, resulting in secondary immunodeficiency and increased infectious risks. Biologic medications can increase general risk for upper respiratory tract infections but can also be associated with unique infectious risks owing to distinct mechanisms of action. With the widespread use of these medications, providers in every area of medicine will likely care for individuals receiving biologic therapies and understanding their potential infectious complications can help mitigate these risks. This practical review discusses the infectious implications of biologics by class of medication and provides recommendations regarding the examination and screening both before therapy initiation and while the patient is receiving the medication. With this knowledge and background, providers can reduce risk whereas patients receive the treatment benefits of these biologic medications.
Collapse
Affiliation(s)
- Laura Cannon
- Division of Pediatric Rheumatology, Department of Pediatrics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Alice Pan
- Division of Pediatric Rheumatology, Department of Pediatrics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina; Department of Pharmacy, UNC Health, Chapel Hill, North Carolina
| | - Leonard Kovalick
- Division of Pediatric Rheumatology, Department of Pediatrics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Aliese Sarkissian
- Division of Pediatric Rheumatology, Department of Pediatrics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Eveline Y Wu
- Division of Pediatric Rheumatology, Department of Pediatrics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina; Division of Pediatric Allergy/Immunology, Department of Pediatrics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina.
| |
Collapse
|
45
|
Mokbel A, Movahedi M, Philippopoulos E, Ojani P, Keystone EC. The Proportion of Patients With Rheumatoid Arthritis Achieving ACR20/50/70; Consistent Patterns of a 60/40/20 as Demonstrated by a Systematic Review and Meta-analysis. J Clin Rheumatol 2023; 29:183-189. [PMID: 36870081 DOI: 10.1097/rhu.0000000000001945] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/06/2023]
Abstract
OBJECTIVES We aimed to demonstrate that the proportion of rheumatoid arthritis patients achieving 20%/50%/70% improvement in American College of Rheumatology (ACR20/50/70) responses to Food and Drug Administration-approved biologic disease-modifying antirheumatic drugs (bDMARDs) after an inadequate response to methotrexate (MTX) and after failure of the first bDMARDs followed a consistent pattern. METHODS This systematic review and meta-analysis was performed in accordance with MECIR (Methodological Expectations for Cochrane Intervention Reviews) standards. Two separate groups of randomized controlled trials were included: the first group included studies with biologic-naive patients who added bDMARD to MTX as intervention arm compared with the placebo plus MTX group. The second group included biologic-irresponsive (IR) patients who used a second bDMARD plus MTX after the first bDMARD failure compared with placebo plus MTX group. Primary outcome was defined as the proportion of rheumatoid arthritis patients achieving ACR20/50/70 responses at 24 ± 6 weeks. RESULTS Twenty-one studies initiated between 1999 and 2017 were included: 15 studies for the biologic-naive group and 6 studies for the biologic-IR group. For the biologic-naive group, the proportions of patients achieving ACR20/50/70 were 61.4% (95% confidence interval [CI], 58.7%-64.1%), 37.8% (95% CI, 34.8%-40.8%), and 18.8% (95% CI, 16.1%-21.4%), respectively. For the biologic-IR group, proportions of patients achieving ACR20/50/70 were 48.5% (95% CI, 42.2%-54.8%), 27.3% (95% CI, 21.6%-33.0%), and 12.9% (95% CI, 11.3%-14.8%), respectively. CONCLUSION We were able to systematically demonstrate that ACR20/50/70 responses to biologic-naive follow a consistent pattern of 60%, 40%, and 20%, respectively. We also demonstrated that the ACR20/50/70 responses to a biologic IR follow a certain pattern of 50%, 25%, and 12.5%, respectively.
Collapse
|
46
|
Andreescu M. Risk of Infections Secondary to the Use of Targeted Therapies in Hematological Malignancies. Life (Basel) 2023; 13:1272. [PMID: 37374055 DOI: 10.3390/life13061272] [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: 04/26/2023] [Revised: 05/24/2023] [Accepted: 05/27/2023] [Indexed: 06/29/2023] Open
Abstract
Concurrent infections in hematological malignancies (HM) are major contributors to adverse clinical outcomes, including prolonged hospitalization and reduced life expectancy. Individuals diagnosed with HM are particularly susceptible to infectious pathogens due to immunosuppression, which can either be inherent to the hematological disorder or induced by specific therapeutic strategies. Over the years, the treatment paradigm for HM has witnessed a tremendous shift, from broad-spectrum treatment approaches to more specific targeted therapies. At present, the therapeutic landscape of HM is constantly evolving due to the advent of novel targeted therapies and the enhanced utilization of these agents for treatment purposes. By initiating unique molecular pathways, these agents hinder the proliferation of malignant cells, consequently affecting innate and adaptive immunity, which increases the risk of infectious complications. Due to the complexity of novel targeted therapies and their associated risks of infection, it often becomes a daunting task for physicians to maintain updated knowledge in their clinical practice. The situation is further aggravated by the fact that most of the initial clinical trials on targeted therapies provide inadequate information to determine the associated risk of infection. In such a scenario, a cumulative body of evidence is paramount in guiding clinicians regarding the infectious complications that can arise following targeted therapies. In this review, I summarize the recent knowledge on infectious complications arising in the context of targeted therapies for HM.
Collapse
Affiliation(s)
- Mihaela Andreescu
- Department of Clinical Sciences, Hematology, Faculty of Medicine, Titu Maiorescu University of Bucharest, 040051 Bucharest, Romania
- Department of Hematology, Colentina Clinical Hospital, 020125 Bucharest, Romania
| |
Collapse
|
47
|
Shirai T, Nakai A, Ando E, Fujimoto J, Leach S, Arimori T, Higo D, van Eerden FJ, Tulyeu J, Liu YC, Okuzaki D, Murayama MA, Miyata H, Nunomura K, Lin B, Tani A, Kumanogoh A, Ikawa M, Wing JB, Standley DM, Takagi J, Suzuki K. Celastrol suppresses humoral immune responses and autoimmunity by targeting the COMMD3/8 complex. Sci Immunol 2023; 8:eadc9324. [PMID: 37000855 DOI: 10.1126/sciimmunol.adc9324] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/03/2023]
Abstract
Celastrol, a bioactive molecule extracted from the
Tripterygium wilfordii
plant, has been shown to exhibit anti-inflammatory properties. However, its mechanism of action has not been fully elucidated. Here, we show that celastrol suppresses humoral immune responses and autoimmunity by disabling a protein complex consisting of copper metabolism MURR1 domain–containing (COMMD) 3 and COMMD8 (COMMD3/8 complex), a signaling adaptor for chemoattractant receptors. Having demonstrated the involvement of the COMMD3/8 complex in a mouse model of rheumatoid arthritis, we identified celastrol as a compound that covalently bound to and dissociated the COMMD3/8 complex. Celastrol inhibited B cell migration, reduced antibody responses, and blocked arthritis progression, recapitulating deficiency of the COMMD3/8 complex. These effects of celastrol were abolished in mice expressing a celastrol-resistant mutant of the COMMD3/8 complex. These findings establish that celastrol exerts immunosuppressive activity by targeting the COMMD3/8 complex. Our study suggests that the COMMD3/8 complex is a potentially druggable target in autoimmune diseases and points to celastrol as a lead pharmacologic candidate in this capacity.
Collapse
Affiliation(s)
- Taiichiro Shirai
- Laboratory of Immune Response Dynamics, WPI Immunology Frontier Research Center, Osaka University, Suita, Osaka, Japan
- Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - Akiko Nakai
- Laboratory of Immune Response Dynamics, WPI Immunology Frontier Research Center, Osaka University, Suita, Osaka, Japan
- Department of Immune Response Dynamics, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, Japan
| | - Emiko Ando
- Laboratory of Immune Response Dynamics, WPI Immunology Frontier Research Center, Osaka University, Suita, Osaka, Japan
| | - Jun Fujimoto
- Laboratory of Immune Response Dynamics, WPI Immunology Frontier Research Center, Osaka University, Suita, Osaka, Japan
- Department of Respiratory Medicine and Clinical Immunology, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - Sarah Leach
- Laboratory of Immune Response Dynamics, WPI Immunology Frontier Research Center, Osaka University, Suita, Osaka, Japan
| | - Takao Arimori
- Laboratory for Protein Synthesis and Expression, Institute for Protein Research, Osaka University, Suita, Osaka, Japan
| | - Daisuke Higo
- Thermo Fisher Scientific K.K., Yokohama, Kanagawa, Japan
| | - Floris J. van Eerden
- Laboratory of Systems Immunology, WPI Immunology Frontier Research Center, Osaka University, Suita, Osaka, Japan
- Department of Genome Informatics, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, Japan
| | - Janyerkye Tulyeu
- Laboratory of Human Single Cell Immunology, WPI Immunology Frontier Research Center, Osaka University, Suita, Osaka, Japan
- Center for Infectious Disease Education and Research, Osaka University, Suita, Osaka, Japan
| | - Yu-Chen Liu
- Laboratory of Human Immunology (Single Cell Genomics), WPI Immunology Frontier Research Center, Osaka University, Suita, Osaka, Japan
| | - Daisuke Okuzaki
- Center for Infectious Disease Education and Research, Osaka University, Suita, Osaka, Japan
- Laboratory of Human Immunology (Single Cell Genomics), WPI Immunology Frontier Research Center, Osaka University, Suita, Osaka, Japan
- Genome Information Research Center, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, Japan
| | - Masanori A. Murayama
- Department of Animal Models for Human Diseases, Institute of Biomedical Science, Kansai Medical University, Hirakata, Osaka, Japan
| | - Haruhiko Miyata
- Department of Experimental Genome Research, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, Japan
| | - Kazuto Nunomura
- Center for Supporting Drug Discovery and Life Science Research, Graduate School of Pharmaceutical Science, Osaka University, Suita, Osaka, Japan
| | - Bangzhong Lin
- Center for Supporting Drug Discovery and Life Science Research, Graduate School of Pharmaceutical Science, Osaka University, Suita, Osaka, Japan
| | - Akiyoshi Tani
- Center for Supporting Drug Discovery and Life Science Research, Graduate School of Pharmaceutical Science, Osaka University, Suita, Osaka, Japan
| | - Atsushi Kumanogoh
- Department of Respiratory Medicine and Clinical Immunology, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
- Center for Infectious Disease Education and Research, Osaka University, Suita, Osaka, Japan
- Laboratory of Immunopathology, WPI Immunology Frontier Research Center, Osaka University, Suita, Osaka, Japan
- Integrated Frontier Research for Medical Science Division, Institute for Open and Transdisciplinary Research Initiatives, Osaka University, Suita, Osaka, Japan
| | - Masahito Ikawa
- Center for Infectious Disease Education and Research, Osaka University, Suita, Osaka, Japan
- Department of Experimental Genome Research, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, Japan
| | - James B. Wing
- Laboratory of Human Single Cell Immunology, WPI Immunology Frontier Research Center, Osaka University, Suita, Osaka, Japan
- Center for Infectious Disease Education and Research, Osaka University, Suita, Osaka, Japan
| | - Daron M. Standley
- Laboratory of Systems Immunology, WPI Immunology Frontier Research Center, Osaka University, Suita, Osaka, Japan
- Department of Genome Informatics, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, Japan
- Center for Infectious Disease Education and Research, Osaka University, Suita, Osaka, Japan
| | - Junichi Takagi
- Laboratory for Protein Synthesis and Expression, Institute for Protein Research, Osaka University, Suita, Osaka, Japan
- Integrated Frontier Research for Medical Science Division, Institute for Open and Transdisciplinary Research Initiatives, Osaka University, Suita, Osaka, Japan
| | - Kazuhiro Suzuki
- Laboratory of Immune Response Dynamics, WPI Immunology Frontier Research Center, Osaka University, Suita, Osaka, Japan
- Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
- Department of Immune Response Dynamics, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, Japan
- Center for Infectious Disease Education and Research, Osaka University, Suita, Osaka, Japan
| |
Collapse
|
48
|
Bahap-Kara M, Duran E, Bayraktar-Ekincioglu A, Karadag O. Interchangeability and adverse events in originator-rituximab and its biosimilar (CT-P10) among rheumatic patients: a real-life experience. Intern Emerg Med 2023; 18:791-799. [PMID: 36826744 PMCID: PMC9951838 DOI: 10.1007/s11739-023-03222-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2022] [Accepted: 02/08/2023] [Indexed: 02/25/2023]
Abstract
Biosimilars offer cost-effective and safe treatment options both for patients and healthcare systems. CT-P10 is the first biosimilar of rituximab approved in Europe for use in all indications of originator rituximab (oRTX). This study aimed to provide real-life data on treatment changes and adverse events in patients who received oRTX or CT-P10. We retrospectively reviewed treatment-related adverse events [infusion-related reactions (IRRs), infections, hypogammaglobulinemia] in patients treated with at least one dose of oRTX (MabThera®) or CT-P10 (Truxima®) between 2020 and 2021 and had at least 6 months follow-up after rituximab infusion in a rheumatology clinic. The switches between oRTX and CT-P10 were performed according to drug availability at the hospital pharmacy at the time of infusion according to the local hospital procedure. Physicians were not involved in the decision of biosimilar selection. A total of 128 patients (CT-P10, n = 64; oRTX, n = 64) were included. CT-P10 was switched in 52 (40.6%) patients who had previously used oRTX, and 48 (37.5%) patients remained on oRTX. We demonstrated no difference between patients treated with oRTX or CT-P10 in the rates of IRRs, in which all reactions were grade 1 and 2. Comparable rates of infections (p > 0.05) and the rate of hypogammaglobulinemia (p > 0.05) were found in both groups with no significant difference. CT-P10 provides a safe treatment alternative in patients who require rituximab therapy. The rational use of biosimilars can be supported by evolving evidence on interchangeability and switching in real-life settings, which will help clinicians in decision-making.
Collapse
Affiliation(s)
- Melda Bahap-Kara
- grid.14442.370000 0001 2342 7339Department of Clinical Pharmacy, Faculty of Pharmacy, Hacettepe University, Sihhiye, Ankara, Turkey
| | - Emine Duran
- grid.14442.370000 0001 2342 7339Division of Rheumatology, Department of Internal Medicine, Faculty of Medicine, Hacettepe University, Ankara, Turkey
| | - Aygin Bayraktar-Ekincioglu
- grid.14442.370000 0001 2342 7339Department of Clinical Pharmacy, Faculty of Pharmacy, Hacettepe University, Sihhiye, Ankara, Turkey
| | - Omer Karadag
- grid.14442.370000 0001 2342 7339Division of Rheumatology, Department of Internal Medicine, Faculty of Medicine, Hacettepe University, Ankara, Turkey
| |
Collapse
|
49
|
Pombo-Suarez M, Sanchez-Piedra C, Gómez-Reino J, Lauper K, Mongin D, Iannone F, Pavelka K, Nordström DC, Inanc N, Codreanu C, Hyrich KL, Choquette D, Strangfeld A, Leeb BF, Rotar Z, Rodrigues A, Kristianslund EK, Kvien TK, Elkayam O, Lukina G, Bergstra SA, Finckh A, Courvoisier DS. After JAK inhibitor failure: to cycle or to switch, that is the question - data from the JAK-pot collaboration of registries. Ann Rheum Dis 2023; 82:175-181. [PMID: 36100351 DOI: 10.1136/ard-2022-222835] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
OBJECTIVES The expanded therapeutic arsenal in rheumatoid arthritis (RA) raises new clinical questions. The objective of this study is to compare the effectiveness of cycling Janus kinase inhibitors (JAKi) with switching to biologic disease-modifying antirheumatic drug (bDMARD) in patients with RA after failure to the first JAKi. METHODS This is a nested cohort study within data pooled from an international collaboration of 17 national registries (JAK-pot collaboration). Data from patients with RA with JAKi treatment failure and who were subsequently treated with either a second JAKi or with a bDMARD were prospectively collected. Differences in drug retention rates after second treatment initiation were assessed by log-rank test and Cox regression analysis adjusting for potential confounders. Change in Clinical Disease Activity Index (CDAI) over time was estimated using a linear regression model, adjusting for confounders. RESULTS 365 cycling and 1635 switching patients were studied. Cyclers were older and received a higher number of previous bDMARDs. Both strategies showed similar observed retention rates after 2 years of follow-up. However, adjusted analysis revealed that cycling was associated with higher retention (p=0.04). Among cyclers, when the first JAKi was discontinued due to an adverse event (AE), it was more likely that the second JAKi would also be stopped due to an AE. Improvement in CDAI over time was similar in both strategies. CONCLUSIONS After failing the first JAKi, cycling JAKi and switching to a bDMARD appear to have similar effectiveness. Caution is advised if an AE was the reason to stop the first JAKi.
Collapse
Affiliation(s)
- Manuel Pombo-Suarez
- Rheumatology Department, Complejo Hospitalario Universitario de Santiago de Compostela, Santiago de Compostela, Spain
| | | | - Juan Gómez-Reino
- Fundacion IDIS, Hospital Clinico Universitario, Santiago de Compostela, Spain
| | - Kim Lauper
- Division of Rheumatology, Department of Internal Medicine and Department of Medicine, Faculty of Medicine, Geneva University Hospitals, Geneva, Switzerland.,Centre for Epidemiology Versus Arthritis, Centre for Musculoskeletal Research, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
| | - Denis Mongin
- Division of Rheumatology, Department of Internal Medicine and Department of Medicine, Faculty of Medicine, Geneva University Hospitals, Geneva, Switzerland
| | | | - Karel Pavelka
- Department of Rheumatology, Charles University, Praha, Czech Republic
| | - Dan C Nordström
- ROB-FIN, Departments of Medicine and Rheumatology, Helsinki University Hospital and Helsinki University, Helsinki, Finland
| | - Nevsun Inanc
- Division of Rheumatology, Marmara University School of Medicine, Istanbul, Turkey
| | - Catalin Codreanu
- Rheumatology, Center of Rheumatic Diseases, University of Medicine and Pharmacy, Bucharest, Romania
| | - Kimme L Hyrich
- Centre for Epidemiology Versus Arthritis, Centre for Musculoskeletal Research, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, UK.,NIHR Manchester Biomedical Research Centre, Manchester University NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester, UK
| | - Denis Choquette
- Institut de Recherche en Rhumatologie de Montréal, University of Montreal, Montreal, Quebec, Canada
| | - Anja Strangfeld
- Epidemiology Unit, German Rheumatism Research Center (DRFZ), Berlin, Germany.,Department of Rheumatology and Clinical Immunology, Charité University Medicine, Berlin, Germany
| | - Burkhard F Leeb
- BioReg, Vienna, Austria.,Private Office, Hollabrunn, Austria
| | - Ziga Rotar
- Department of Rheumatology, University Medical Centre, Ljubljana, Slovenia.,Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Ana Rodrigues
- Unidade de Reumatologia, Hospital Lusiadas, Lisbon, Portugal
| | | | - Tore K Kvien
- Division of Rheumatology and Research, Diakonhjemmet Hospital, Oslo, Norway
| | - Ori Elkayam
- Department of Rheumatology, Tel Aviv Sourasky Medical Center, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Galina Lukina
- VA Nasonova Research Institute of Rheumatology, AS Loginov Moscow Clinical Scientific Center, RBITER, Institute of Rheumatology, Moscow, Russian Federation
| | - Sytske Anne Bergstra
- Department of Rheumatology, Leiden University Medical Center, Leiden, The Netherlands
| | - Axel Finckh
- Division of Rheumatology, Department of Internal Medicine and Department of Medicine, Faculty of Medicine, Geneva University Hospitals, Geneva, Switzerland
| | - Delphine Sophie Courvoisier
- Division of Rheumatology, Department of Internal Medicine and Department of Medicine, Faculty of Medicine, Geneva University Hospitals, Geneva, Switzerland
| |
Collapse
|
50
|
Harna B, Kalra P, Arya S, Jeyaraman N, Nallakumarasamy A, Jeyaraman M, Rajendran RL, Oh EJ, Khanna M, Rajendran UM, Chung HY, Ahn BC, Gangadaran P. Mesenchymal stromal cell therapy for patients with rheumatoid arthritis. Exp Cell Res 2023; 423:113468. [PMID: 36621669 DOI: 10.1016/j.yexcr.2023.113468] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 12/22/2022] [Accepted: 01/04/2023] [Indexed: 01/07/2023]
Abstract
Management of relapses and refractory rheumatoid arthritis (RA) patients is complex and difficult. Even after the administration of new biological disease-modifying anti-rheumatic drugs (DMARDs), only a few patients achieve the complete remission phase. DMARDs help only in modifying the disease activity, which sooner or later fails. They do not manage the disease at the patho-etiological level. There are some serious side effects as well as drug interaction with DMARDs. There are few subsets of RA patients who do not respond to DMARDs, reasons unknown. Mesenchymal stem cells (MSCs) provide a promising alternative, especially in such cases. This review elaborates on the studies pertaining to the application of MSCs in rheumatoid arthritis over the last two decades. A total of 14 studies (one review article) including 447 patients were included in the study. Most of the studies administered MSCs in refractory RA patients through the intravenous route with varied dosages and frequency of administration. MSCs help in RA treatment via various mechanisms including paracrine effects. All the studies depicted a better clinical outcome with minimal adverse events. The functional scores including the VAS scores improved significantly in all studies irrespective of dosage and source of MSCs. The majority of the studies depicted no complications. Although the use of MSCs in RA is still in the early stages requiring further refinement in the source of MSCs, dosage, and frequency. The role of MSCs in the management of RA has a promising prospect. MSCs target the RA at the molecular level and has the potential to manage refractory RA cases not responding to conventional treatment. Multicentric, large sample populations, and long-term studies are required to ascertain efficacy and safety.
Collapse
Affiliation(s)
- Bushu Harna
- Department of Orthopaedics, Maulana Azad Medical College, New Delhi, 110002, India; Indian Stem Cell Study Group (ISCSG) Association, Lucknow, 226010, Uttar Pradesh, India; Fellow in Orthopaedic Rheumatology, Dr. RML National Law University, Lucknow, 226010, Uttar Pradesh, India
| | - Pulkit Kalra
- Department of Orthopaedics, Maulana Azad Medical College, New Delhi, 110002, India
| | - Shivali Arya
- Department of Radiodiagnosis, Maulana Azad Medical College, New Delhi, 110002, India
| | - Naveen Jeyaraman
- Indian Stem Cell Study Group (ISCSG) Association, Lucknow, 226010, Uttar Pradesh, India; Fellow in Orthopaedic Rheumatology, Dr. RML National Law University, Lucknow, 226010, Uttar Pradesh, India; Fellow in Regenerative Interventional Orthobiologics, Dr. RML National Law University, Lucknow, 226010, Uttar Pradesh, India; Department of Orthopaedics, Rathimed Specialty Hospital, Chennai, 600040, Tamil Nadu, India
| | - Arulkumar Nallakumarasamy
- Indian Stem Cell Study Group (ISCSG) Association, Lucknow, 226010, Uttar Pradesh, India; Fellow in Orthopaedic Rheumatology, Dr. RML National Law University, Lucknow, 226010, Uttar Pradesh, India; Fellow in Regenerative Interventional Orthobiologics, Dr. RML National Law University, Lucknow, 226010, Uttar Pradesh, India; Department of Orthopaedics, All India Institute of Medical Sciences, Bhubaneswar, 751019, Odisha, India
| | - Madhan Jeyaraman
- Indian Stem Cell Study Group (ISCSG) Association, Lucknow, 226010, Uttar Pradesh, India; Department of Orthopaedics, ACS Medical College and Hospital, Dr MGR Educational and Research Institute, Chennai, 600056, Tamil Nadu, India; Department of Biotechnology, School of Engineering and Technology, Sharda University, Greater Noida, 201310, Uttar Pradesh, India; South Texas Orthopaedic Research Institute (STORI Inc.), Laredo, TX, 78045, USA.
| | - Ramya Lakshmi Rajendran
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu, 41944, South Korea
| | - Eun Jung Oh
- Department of Plastic and Reconstructive Surgery, CMRI, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu, 41944, South Korea
| | - Manish Khanna
- Indian Stem Cell Study Group (ISCSG) Association, Lucknow, 226010, Uttar Pradesh, India
| | | | - Ho Yun Chung
- Department of Plastic and Reconstructive Surgery, CMRI, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu, 41944, South Korea
| | - Byeong-Cheol Ahn
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu, 41944, South Korea; BK21 FOUR KNU Convergence Educational Program of Biomedical Sciences for Creative Future Talents, Department of Biomedical Science, School of Medicine, Kyungpook National University, Daegu, 41944, South Korea.
| | - Prakash Gangadaran
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu, 41944, South Korea; BK21 FOUR KNU Convergence Educational Program of Biomedical Sciences for Creative Future Talents, Department of Biomedical Science, School of Medicine, Kyungpook National University, Daegu, 41944, South Korea.
| |
Collapse
|