Subscribe to RSS
DOI: 10.1055/a-2687-1483
Therapie der rheumatoiden Arthritis – quo vadis?
Treatment of Rheumatoid Arthritis: Quo Vadis?Authors
Zusammenfassung
Die rheumatoide Arthritis (RA) ist eine chronisch-entzündliche Autoimmunerkrankung, deren Therapie sich durch frühzeitige Diagnose, zielgerichtete Behandlungsstrategien und den Einsatz von biologischen (bDMARDs) und zielgerichteten synthetischen krankheitsmodifizierenden Medikamenten (tsDMARDs) erheblich verbessert hat. Dennoch verbleibt ein relevanter Anteil von Patienten in einem schwer behandelbaren Zustand (difficult-to-treat RA), bei dem trotz moderner Therapien keine Remission oder geringe Krankheitsaktivität erreicht wird. Dieser Artikel analysiert neue immunmodulatorische Therapieansätze, die zum Ziel haben, diese Behandlungslücke zu schließen. Ein zentrales neues Konzept ist die Modulation von inhibitorischen Immun-Checkpoints. Der PD-1-Agonist Rosnilimab zeigte in einer Phase-2b-Studie bei mittelschwerer bis schwerer RA deutliche klinische Verbesserungen bei einem günstigen Sicherheitsprofil. Im Gegensatz zu klassischen Zytokinblockaden zielt Rosnilimab darauf ab, die T-Zell-Homöostase wiederherzustellen, indem es pathogene T-Zellen mit hohem PD-1-Gehalt moduliert sowie dezimiert und regulatorische T-Zellen induziert. Ein verwandter Wirkstoff, Peresolimab, bestätigte das Prinzip des PD-1-Agonismus, wurde aber nicht weiterentwickelt. Andere immunologische Zielmoleküle wie BTLA oder CD122 befinden sich derzeit in der präklinischen Testphase. Bispezifische Antikörper (bsAbs) stellen einen weiteren innovativen Therapiebereich dar. Es werden hauptsächlich zwei Ansätze verfolgt: 1. duale Zytokinblockade, zum Beispiel gegen TNF und IL-17A (ABT-122) oder TNF und IL-6 (V5–3), mit dem Ziel synergistischer Effekte bei redundanter Entzündungsaktivität; 2. T-Zell-Engager, die zytotoxische T-Zellen gegen autoreaktive B-Zellen (z. B. CD19xCD3, Blinatumomab) oder Plasmazellen (BCMAxCD3, Teclistamab) richten. Erste klinische Daten deuten auf eine deutliche Verringerung der Krankheitsaktivität und der pathogenen Autoantikörper hin. Andere bispezifische Moleküle, wie Imvotamab (CD20xCD3), werden derzeit erprobt. Antikörper-Glukokortikoid-Konjugate stellen ebenfalls einen interessanten therapeutischen Ansatz dar. Die CAR-T-Zelltherapie ist derzeit der intensivste therapeutische Ansatz. Erste Fallberichte zeigen, dass RA-Patienten nach einer Anti-CD19- oder CD20/CAR-T-Zelltherapie – ursprünglich gegen maligne Erkrankungen eingesetzt – eine medikamentenfreie Remission von mehr als einem Jahr erreichen können. Neben den B-Zellen rücken auch synoviale Fibroblasten (FAP-positive Zellen) als Zielstrukturen in den Fokus. In präklinischen Modellen zeigten FAP-CAR-T-Zellen eine gezielte Depletion von proinflammatorischen Stromazellen und eine Remission der Arthritis. Darüber hinaus werden regulatorische CAR-T-Zellen (CAR-Tregs) als neue Strategie zur Immunsuppression diskutiert. Diese neuen Therapien unterscheiden sich grundlegend in ihrer Zielstruktur, ihrem Wirkmechanismus und ihrer Anwendungsmodalität. Während PD-1-Agonisten eine Weiterentwicklung der biologischen Therapien darstellen, versprechen bispezifische Antikörper eine größere Zielgenauigkeit und CAR-T-Zellen potenziell kurative Wirkungen. Kosten, Komplexität und mögliche Nebenwirkungen sind jedoch die größten Herausforderungen. Parallel zur klinischen Entwicklung sind prädiktive Biomarker entscheidend, um personalisierte Behandlungsentscheidungen zu ermöglichen. Die RA-Therapie steht daher vor einem möglichen Paradigmenwechsel: weg von vorwiegend symptombekämpfenden Ansätzen hin zu immunologisch gezielten und potenziell kurativen Interventionen. Die nächsten Jahre werden zeigen, welche dieser innovativen Strategien ihren Weg in die klinische Praxis finden werden – mit dem langfristigen Ziel, eine Remission oder Heilung für alle Patienten zu erreichen.
Abstract
Rheumatoid arthritis (RA) is a chronic inflammatory autoimmune disease whose therapeutic management has improved significantly through early diagnosis, treat-to-target strategies, and the use of biologic (bDMARDs) and targeted synthetic (tsDMARDs) disease-modifying drugs. Nevertheless, a relevant proportion of patients remain in a difficult-to-treat state (difficult-to-treat RA), failing to achieve remission or low disease activity despite modern therapies. This article analyses new immunomodulatory therapeutic approaches with the aim of closing this treatment gap. A central new concept is the modulation of inhibitory immune checkpoints. The PD-1 agonist rosnilimab demonstrated significant clinical improvements with a favourable safety profile in a phase 2b study in moderate-to-severe RA. In contrast to classic cytokine blockades, rosnilimab aims to restore T-cell homeostasis by depleting pathogenic PD-1 high T cells and inducing regulatory T cells. A related agent, peresolimab, confirmed the principle of PD-1 agonism, but was not developed further. Other immunological targets, such as BTLA or CD122, are currently undergoing preclinical testing. Bispecific antibodies (bsAbs) represent another innovative field of therapy. Two main approaches are being pursued: (1) dual cytokine blockade – for example against TNF and IL-17A (ABT-122) or TNF and IL-6 (V5–3) – aimed at achieving synergistic effects in the presence of redundant inflammatory activity; (2) T-cell engagers that target cytotoxic T cells against autoreactive B cells (e. g. CD19xCD3, blinatumomab) or plasma cells (BCMAxCD3, teclistamab). Initial clinical data indicate significant reductions in both disease activity and pathogenic autoantibodies. Other bispecific molecules, such as imvotamab (CD20xCD3), are currently under clinical investigation. Antibody-glucocorticoid conjugates also represent a promising therapeutic approach. CAR T-cell therapy is currently the most intensive form of therapy. Case reports show that RA patients can achieve drug-free remission for more than a year after anti-CD19 or CD20/CAR T cell therapy, which was originally developed for malignant diseases. In addition to B cells, synovial fibroblasts (FAP-positive cells) are increasingly recognised as potential target structures. In preclinical models, FAP CAR T cells achieved targeted depletion of pro-inflammatory stromal cells and induced remission of arthritis. In addition, regulatory CAR T cells (CAR-Tregs) are being explored as a novel strategy for local immunosuppression in joints. These new therapies differ fundamentally in their target, mechanism of action, and mode of application. While PD-1 agonists represent an evolution of biological therapies, bispecific antibodies offer greater targeting precision, and CAR T cells hold the potential for curative effects. However, costs, complexity and potential side-effects are key challenges. Alongside clinical development, predictive biomarkers are crucial to enable personalised treatment decisions. RA therapy is therefore facing a potential paradigm shift: from predominantly symptom-controlling approaches towards immunologically targeted and potentially curative interventions. The next few years will show which of these innovative strategies will make their way into clinical practice, with the long-term goal of achieving remission or cure for all patients.
Schlüsselwörter
Rheumatoide Arthritis - Therapie - Modulation immunologischer - Checkpoints - Bispezifische Antikörper - CAR-T-ZelltherapieKeywords
rheumatoid arthritis - treatment - bispecific antibodies - CAR T cell therapy - immune checkpoint modulationPublication History
Received: 08 August 2025
Accepted: 11 August 2025
Article published online:
16 October 2025
© 2025. Thieme. All rights reserved.
Georg Thieme Verlag KG
Oswald-Hesse-Straße 50, 70469 Stuttgart, Germany
-
Literatur
- 1 Chiu YM, Lu YP, Lan JL. et al. Lifetime Risks, Life Expectancy, and Health Care Expenditures for Rheumatoid Arthritis: A Nationwide Cohort Followed Up From 2003 to 2016. Arthritis Rheumatol 2021; 73: 750-758
- 2 Takanashi S, Kaneko Y. Unmet Needs and Current Challenges of Rheumatoid Arthritis: Difficult-to-Treat Rheumatoid Arthritis and Late-Onset Rheumatoid Arthritis. Journal of clinical medicine 2024; 13
- 3 Roodenrijs NMT, de Hair MJH, van der Goes MC. et al. Characteristics of difficult-to-treat rheumatoid arthritis: results of an international survey. Annals of the rheumatic diseases 2018; 77: 1705-1709
- 4 Smolen JS, Landewé RBM, Bergstra SA. et al. EULAR recommendations for the management of rheumatoid arthritis with synthetic and biological disease-modifying antirheumatic drugs: 2022 update. Ann Rheum Dis 2023; 82: 3-18
- 5 Aaltonen KJ, Virkki LM, Malmivaara A. et al. Systematic review and meta-analysis of the efficacy and safety of existing TNF blocking agents in treatment of rheumatoid arthritis. PloS one 2012; 7: e30275
- 6 Avci AB, Feist E, Burmester GR. Targeting IL-6 or IL-6 Receptor in Rheumatoid Arthritis: What Have We Learned?. BioDrugs: clinical immunotherapeutics, biopharmaceuticals and gene therapy 2024; 38: 61-71
- 7 Emery P, Deodhar A, Rigby WF. et al. Efficacy and safety of different doses and retreatment of rituximab: a randomised, placebo-controlled trial in patients who are biological naive with active rheumatoid arthritis and an inadequate response to methotrexate (Study Evaluating Rituximab's Efficacy in MTX iNadequate rEsponders (SERENE)). Annals of the rheumatic diseases 2010; 69: 1629-1635
- 8 Kremer JM, Westhovens R, Leon M. et al. Treatment of rheumatoid arthritis by selective inhibition of T-cell activation with fusion protein CTLA4Ig. The. New England journal of medicine 2003; 349: 1907-1915
- 9 Kerschbaumer A, Sepriano A, Bergstra SA. et al. Efficacy of synthetic and biological DMARDs: a systematic literature review informing the 2022 update of the EULAR recommendations for the management of rheumatoid arthritis. Ann Rheum Dis 2023; 82: 95-106
- 10 Schipper LG, Vermeer M, Kuper HH. et al. A tight control treatment strategy aiming for remission in early rheumatoid arthritis is more effective than usual care treatment in daily clinical practice: a study of two cohorts in the Dutch Rheumatoid Arthritis Monitoring registry. Annals of the rheumatic diseases 2012; 71: 845-850
- 11 van Eijk IC, Nielen MM, van der Horst-Bruinsma I. et al. Aggressive therapy in patients with early arthritis results in similar outcome compared with conventional care: the STREAM randomized trial. Rheumatology (Oxford) 2012; 51: 686-694
- 12 Nagy G, Roodenrijs NMT, Welsing PM. et al. EULAR definition of difficult-to-treat rheumatoid arthritis. Annals of the rheumatic diseases 2021; 80: 31-35
- 13 Mullard A. Bispecific antibody takes a BiTE out of B cells in rheumatoid arthritis. Nature Reviews Drug Discovery. 23. 411 2024 Available from: https://www.nature.com/articles/d41573-024-00084-6 [Access Date 30.07.2025]
- 14 Kaufman MB. Positive Week 12 Results for Rosnilimab in Patients with RA. June 18. 2025 Available from https://www.the-rheumatologist.org/article/positive-week-12-results-for-rosnilimab-in-patients-with-ra/ [Access Date 30.07.2025]
- 15 Cush J. Targeting PD-1 in Rheumatoid Arthritis. RheumNow. 18 Feb. 2025 Available from https://rheumnow.com/news/targeting-pd-1-rheumatoid-arthritis [Access Date 30.07.2025]
- 16 Tuttle J, Drescher E, Simon-Campos JA. et al. A Phase 2 Trial of Peresolimab for Adults with Rheumatoid Arthritis. The. New England journal of medicine 2023; 388: 1853-1862
- 17 Osborne R. Lilly bid wilts as others forge on with PD-1 in RA. Nov. 7. 2024 Available from: https://www.bioworld.com/articles/714315-lilly-bid-wilts-as-others-forge-on-with-pd-1-in-ra?v=preview [Access Date 30.07.2025]
- 18 Anaptys Announces Phase 2b Trial of ANB032, a BTLA Agonist, Did Not Meet Primary or Secondary Endpoints in Atopic Dermatitis. Available from https://ir.anaptysbio.com/news-releases/news-release-details/anaptys-announces-phase-2b-trial-anb032-btla-agonist-did-not/ [Access Date 30.07.2025]
- 19 AnaptysBio Announces Portfolio Update Across Best-in-Class Immune Cell Modulating Antibodies. Available from https://ir.anaptysbio.com/news-releases/news-release-details/anaptysbio-announces-portfolio-update-across-best-class-immune/ [Access Date 30.07.2025]
- 20 Genovese MC, Weinblatt ME, Aelion JA. et al. ABT-122, a Bispecific Dual Variable Domain Immunoglobulin Targeting Tumor Necrosis Factor and Interleukin-17A, in Patients With Rheumatoid Arthritis With an Inadequate Response to Methotrexate: A Randomized, Double-Blind Study. Arthritis Rheumatol 2018; 70: 1710-1720
- 21 Biesemann N, Margerie D, Asbrand C. et al. Additive efficacy of a bispecific anti-TNF/IL-6 nanobody compound in translational models of rheumatoid arthritis. Science translational medicine 2023; 15: eabq4419
- 22 Upcoming Bispecific & Trispecific Antibodies Beyond Oncology. Jun 10, 2025 Available from https://www.delveinsight.com/blog/6-promising-bispecific-antibodies-beyond-oncology [Access Date 30.07.2025]
- 23 Bucci L, Hagen M, Rothe T. et al. Bispecific T cell engager therapy for refractory rheumatoid arthritis. Nature medicine 2024; 30: 1593-1601
- 24 Hicks L. Teclistamab Shows Promise in Refractory Autoimmune Diseases. September 04. 2024 Available from https://www.medscape.com/viewarticle/teclistamab-shows-promise-refractory-autoimmune-diseases-2024a1000g22 [Access Date 30.07.2025]
- 25 Hagen M, Bucci L, Boltz S. et al. BCMA-Targeted T-Cell-Engager Therapy for Autoimmune Disease. The. New England journal of medicine 2024; 391: 867-869
- 26 Alexander T, Kronke J, Cheng Q. et al. Teclistamab-Induced Remission in Refractory Systemic Lupus Erythematosus. The New England journal of medicine 2024; 391: 864-866
- 27 IGM Biosciences Announces FDA Clearance to Begin Clinical Studies of Imvotamab in Lupus and Rheumatoid Arthritis. Available from https://www.biospace.com/igm-biosciences-announces-fda-clearance-to-begin-clinical-studies-of-imvotamab-in-lupus-and-rheumatoid-arthritis [Access Date 30.07.2025]
- 28 Domingo-Gonzalez R, Baribaud I, Oyasu M. et al. Therapeutic potential of imvotamab, a CD20-targeted bispecific IgM T cell engager, for the treatment of refractory autoimmune disease patients. EULAR 2024;
- 29 Buttgereit F, Singhal A, Kivitz A. et al. Efficacy and Safety of ABBV-154 for the Treatment of Active Rheumatoid Arthritis: A Phase 2b, Randomized, Placebo-Controlled Trial. Arthritis Rheumatol 2025;
- 30 ABBV-154. Available from: https://www.guidetopharmacology.org/GRAC/LigandDisplayForward?ligandId=12918. [Access Date 30.07.2025]
- 31 Szabo D, Balogh A, Gopcsa L. et al. Sustained drug-free remission in rheumatoid arthritis associated with diffuse large B-cell lymphoma following tandem CD20-CD19-directed non-cryopreserved CAR-T cell therapy using zamtocabtagene autoleucel. RMD open 2024; 10
- 32 Mackensen A, Müller F, Mougiakakos D. et al. Anti-CD19 CAR T cell therapy for refractory systemic lupus erythematosus. Nat Med 2022; 28: 2124-2132
- 33 Kemble S, Mahony C, Smith C. et al. Targeting Fibroblasts in Inflammatory Disease Using Engineered T Cells. Arthritis Rheumatol 2023; 75 (suppl 9)
- 34 Yu J, Yang Y, Gu Z. et al. CAR immunotherapy in autoimmune diseases promises and challenges. Front Immunol 2024; 15: 1461102
- 35 Schubert ML, Schmitt M, Wang L. et al. Side-effect management of chimeric antigen receptor (CAR) T-cell therapy. Ann Oncol 2021; 32: 34-48
