1
|
Harigai M, Kaneko Y, Tanaka E, Hirata S, Kameda H, Kaneko K, Kishimoto M, Kohno M, Kojima M, Kojima T, Morinobu A, Nakajima A, Sugihara T, Fusama M, Yajima N, Yanai R, Kawahito Y. 2024 Update of the Japan College of Rheumatology Clinical Practice Guidelines for the Management of Rheumatoid Arthritis: Secondary publication. Mod Rheumatol 2025; 35:387-401. [PMID: 39820350 DOI: 10.1093/mr/roaf006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2024] [Revised: 12/28/2024] [Accepted: 01/15/2025] [Indexed: 01/19/2025]
Abstract
OBJECTIVES The aim of this study is to update the Japan College of Rheumatology Clinical Practice Guidelines for the Management of Rheumatoid Arthritis (CPG for RA). METHODS The recommendations were developed based on the evidence published until the end of June 2022 using the Grading of Recommendations Assessment, Development, and Evaluation. The steering committee, CPG panel, systematic review (SR) group, and SR support team were organised. RESULTS The treatment goal and drug treatment algorithm required no modifications; however, the footnotes of the drug treatment algorithm were modified. SR of 21 new or updated recommendations for subcutaneous methotrexate (n = 1), biological disease-modifying antirheumatic drugs (n = 1), rituximab (n = 5), Janus kinase inhibitors (n = 6), biosimilars (n = 2), older patients (n = 4), and pregnancy and lactation (n = 2) was conducted. The recommendations for comorbidities and surgery and rehabilitation remained unchanged from the 2020 CPG for RA. CONCLUSIONS The 2024 CPG for RA, which provide recommendations that reflect the current healthcare environment for rheumatoid arthritis in Japan, can be used effectively as a tool for shared decision-making between rheumatologists and patients in the treatment of RA.
Collapse
Affiliation(s)
- Masayoshi Harigai
- Division of Rheumatology, Department of Internal Medicine, Tokyo Women's Medical University School of Medicine, Shinjuku-ku, Tokyo, Japan
| | - Yuko Kaneko
- Division of Rheumatology, Department of Internal Medicine, Keio University School of Medicine, Shinjuku-ku, Tokyo, Japan
| | - Eiichi Tanaka
- Division of Rheumatology, Department of Internal Medicine, Tokyo Women's Medical University School of Medicine, Shinjuku-ku, Tokyo, Japan
| | - Shintaro Hirata
- Department of Clinical Immunology and Rheumatology, Hiroshima University Hospital, Hiroshima, Japan
| | - Hideto Kameda
- Division of Rheumatology, Department of Internal Medicine, Faculty of Medicine, Toho University (Ohashi Medical Center), Meguro-ku, Tokyo, Japan
| | - Kayoko Kaneko
- Division of Maternal medicine, Center for Maternal-Fetal, Neonatal and Reproductive Medicine, National Center for Child Health and Development, Setagaya-ku, Tokyo, Japan
| | - Mitsumasa Kishimoto
- Department of Nephrology and Rheumatology, Kyorin University School of Medicine, Mitaka, Tokyo, Japan
| | - Masataka Kohno
- Inflammation and Immunology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto-shi, Kyoto, Japan
| | - Masayo Kojima
- Graduate School of Medical Sciences, Nagoya City University, Nagoya, Japan
| | - Toshihisa Kojima
- Orthopaedic Surgery and Rheumatology, National Hospital Organization, Nagoya Medical Center, Nagoya-shi, Aichi, Japan
| | - Akio Morinobu
- Department of Rheumatology and Clinical Immunology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Ayako Nakajima
- Department of Rheumatology, Mie University Graduate School of Medicine, Tsu, Mie, Japan
| | - Takahiko Sugihara
- Division of Rheumatology, Department of Internal Medicine, Toho University School of Medicine, Ota-ku, Tokyo, Japan
| | - Mie Fusama
- Health Sciences Department of Nursing, Kansai University of International Studies, Miki, Hyogo, Japan
| | - Nobuyuki Yajima
- Division of Rheumatology, Department of Medicine, Showa University School of Medicine, Shinagawa-ku, Tokyo, Japan
| | - Ryo Yanai
- Division of Rheumatology, Department of Medicine, Showa University School of Medicine, Shinagawa-ku, Tokyo, Japan
| | - Yutaka Kawahito
- Inflammation and Immunology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto-shi, Kyoto, Japan
| |
Collapse
|
2
|
Blockmans D, Penn SK, Setty AR, Schmidt WA, Rubbert-Roth A, Hauge EM, Keen HI, Ishii T, Khalidi N, Dejaco C, Cid MC, Hellmich B, Liu M, Zhao W, Lagunes I, Romero AB, Wung PK, Merkel PA. A Phase 3 Trial of Upadacitinib for Giant-Cell Arteritis. N Engl J Med 2025. [PMID: 40174237 DOI: 10.1056/nejmoa2413449] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/04/2025]
Abstract
BACKGROUND Giant-cell arteritis is a systemic vasculitis with limited treatment options. The efficacy and safety of upadacitinib - a selective Janus kinase (JAK) inhibitor that blocks the signaling of several cytokines, including interleukin-6 and interferon-γ - are unknown in patients with giant-cell arteritis. METHODS We randomly assigned patients with new-onset or relapsing giant-cell arteritis, in a 2:1:1 ratio, to receive upadacitinib at a dose of 15 mg or 7.5 mg orally once daily plus a 26-week glucocorticoid taper or placebo plus a 52-week glucocorticoid taper. The primary end point was sustained remission at week 52, defined by the absence of signs or symptoms of giant-cell arteritis from week 12 through week 52 and adherence to the protocol-specified glucocorticoid taper. RESULTS A total of 209 patients received upadacitinib at a dose of 15 mg, 107 received upadacitinib at a dose of 7.5 mg, and 112 received placebo; 70% of the patients had new-onset giant-cell arteritis. Upadacitinib at a dose of 15 mg showed superiority over placebo with respect to the primary end point (46.4% [95% confidence interval {CI}, 39.6 to 53.2] vs. 29.0% [95% CI, 20.6 to 37.5]; P = 0.002). Upadacitinib at a dose of 15 mg was superior to placebo in the analysis of the hierarchically prespecified and multiplicity-controlled key secondary end points of sustained complete remission, time to a disease flare, cumulative glucocorticoid exposure, and patient-reported outcomes. Upadacitinib at a dose of 7.5 mg was not superior to placebo with respect to the primary end point (41.1% [95% CI, 31.8 to 50.4]). Safety outcomes during the treatment period of 52 weeks were similar in the upadacitinib and placebo groups. Although cardiovascular risk is a potential concern with a JAK inhibitor, no major adverse cardiovascular events occurred in the upadacitinib groups. CONCLUSIONS In patients with giant-cell arteritis, upadacitinib at a dose of 15 mg - but not 7.5 mg - with a 26-week glucocorticoid taper showed efficacy superior to that of placebo with a 52-week glucocorticoid taper. (Funded by AbbVie; SELECT-GCA ClinicalTrials.gov number, NCT03725202.).
Collapse
Affiliation(s)
- Daniel Blockmans
- Department of General Internal Medicine, University Hospitals Leuven, Leuven, Belgium
- Department of Microbiology, Immunology, and Transplantation, KU Leuven, Leuven, Belgium
| | | | | | - Wolfgang A Schmidt
- Immanuel Krankenhaus Berlin, Medical Center for Rheumatology Berlin-Buch, Berlin
| | - Andrea Rubbert-Roth
- Division of Rheumatology and Immunology, Cantonal Hospital St. Gallen, St. Gallen, Switzerland
| | - Ellen M Hauge
- Department of Rheumatology, Aarhus University Hospital, Aarhus, Denmark
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Helen I Keen
- University of Western Australia Medical School, Fiona Stanley Hospital, Murdoch, Australia
| | - Tomonori Ishii
- Division of Hematology and Rheumatology, Tohoku Medical and Pharmaceutical University, Sendai, Japan
| | - Nader Khalidi
- St. Joseph's Healthcare, McMaster University, Hamilton, ON, Canada
| | - Christian Dejaco
- Department of Rheumatology, Hospital of Brunico, Südtiroler Sanitätsbetrieb - Azienda Sanitaria dell'Alto Adige, Brunico, Italy
- Medical University of Graz, Department of Rheumatology, Graz, Austria
| | - Maria C Cid
- Department of Autoimmune Diseases, European Reference Network RITA, Hospital Clinic, University of Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer, Barcelona
| | - Bernhard Hellmich
- Medius Kliniken, Teaching Hospital University of Tübingen, Department of Internal Medicine, Rheumatology, Pneumology, Nephrology and Diabetology, Kirchheim unter Teck, Germany
| | | | | | | | | | | | - Peter A Merkel
- Division of Rheumatology, Department of Medicine, and the Division of Epidemiology, Department of Biostatistics, Epidemiology, and Informatics, University of Pennsylvania, Philadelphia
| |
Collapse
|
3
|
Saraux A, Carvajal Alegria G, Dernis E, Roux C, Richez C, Tison A, Quere B, Jousse-Joulin S, Guellec D, Marhadour T, Kervarrec P, Cornec D, Le Henaff C, Lesven S, Nowak E, Souki A, Devauchelle-Pensec V. Baricitinib in early polymyalgia rheumatica (BACHELOR): a randomised, double-blind, placebo-controlled, parallel-group trial. THE LANCET. RHEUMATOLOGY 2025; 7:e233-e242. [PMID: 39818228 DOI: 10.1016/s2665-9913(24)00270-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 08/20/2024] [Accepted: 08/29/2024] [Indexed: 01/18/2025]
Abstract
BACKGROUND Moderate doses of glucocorticoids result in improvements in nearly all patients with polymyalgia rheumatica, but related adverse events are common in older individuals. We aimed to evaluate whether treatment with baricitinib (a Janus kinase 1/2 inhibitor) results in disease control without the use of oral glucocorticoids in people with recent-onset polymyalgia rheumatica. METHODS We conducted a randomised, double-blind, placebo-controlled, parallel-group trial at six expert centres in France. Participants with recent (<6 months) polymyalgia rheumatica naive to glucocorticoids and a C-reactive protein polymyalgia rheumatica activity score (CRP PMR-AS) of more than 17 were randomly assigned (1:1), with stratification by hospital, to receive either 4 mg baricitinib orally or placebo (with oral glucocorticoids as rescue treatment in the event of high disease activity) for 12 weeks, followed by 2 mg baricitinib or placebo for another 12 weeks. Subdeltoid glucocorticoid injections at week 1 and week 4 were permitted. Participants, investigators, outcome assessors, and sponsor personnel were masked to group assignments. The primary outcome was a CRP PMR-AS of 10 or less at week 12 without oral glucocorticoid use from week 1 to week 12, analysed in all randomly assigned participants who did not withdraw before first treatment administration. Participants were followed up for 36 weeks. An individual with lived experience of polymyalgia rheumatica was involved in the study design. The trial was registered on ClinicalTrials.gov, NCT04027101, and is complete. FINDINGS We assessed 39 individuals for eligibility between Dec 1, 2020, and Aug 30, 2023. 34 participants (22 women and 12 men) were randomly assigned; 18 participants were assigned to the baricitinib group and 16 participants were assigned to the placebo group. One person allocated to placebo withdrew before the first infusion and was not included in analyses. The primary endpoint was reached at week 12 by 14 (78%) of 18 participants in the baricitinib group and two (13%) of 15 participants in the placebo group (relative risk 5·8, 95% CI 3·2-10·6; crude p=0·0004; adjusted p<0·0001). The most common adverse events were musculoskeletal and connective tissue disorders (13 [72%] of 18 participants in the baricitinib group and four [25%] of 16 in the placebo group. There were no deaths and no major adverse cardiovascular events in either study group. INTERPRETATION This study suggests that, compared with placebo, individuals with polymyalgia rheumatica receiving 4 mg baricitinib are less likely to need oral glucocorticoids to have low disease activity at week 12 of treatment without any new safety signals. FUNDING CHU Brest and Eli Lilly.
Collapse
Affiliation(s)
- Alain Saraux
- Department of Rheumatology, Université de Bretagne Occidentale, CHU Brest, INSERM (U1227), LabEx IGO Brest, France.
| | | | | | - Christian Roux
- Service de rhumatologie, CHU de Nice, IbV, Université Cote d'Azur, Nice, France
| | - Christophe Richez
- Service de Rhumatologie, National Reference Center for Systemic Autoimmune Rare Diseases, Bordeaux University Hospital, Hôpital Pellegrin, Bordeaux, France; CNRS, Immunoconcept, UMR 5164, Bordeaux University, Bordeaux, France
| | - Alice Tison
- Department of Rheumatology, Université de Bretagne Occidentale, CHU Brest, INSERM (U1227), LabEx IGO Brest, France
| | - Baptiste Quere
- Department of Rheumatology, Université de Bretagne Occidentale, CHU Brest, INSERM (U1227), LabEx IGO Brest, France
| | - Sandrine Jousse-Joulin
- Department of Rheumatology, Université de Bretagne Occidentale, CHU Brest, INSERM (U1227), LabEx IGO Brest, France
| | - Dewi Guellec
- Department of Rheumatology, Université de Bretagne Occidentale, CHU Brest, INSERM (U1227), LabEx IGO Brest, France
| | - Thierry Marhadour
- Department of Rheumatology, Université de Bretagne Occidentale, CHU Brest, INSERM (U1227), LabEx IGO Brest, France
| | - Patrice Kervarrec
- Department of Rheumatology, Université de Bretagne Occidentale, CHU Brest, INSERM (U1227), LabEx IGO Brest, France
| | - Divi Cornec
- Department of Rheumatology, Université de Bretagne Occidentale, CHU Brest, INSERM (U1227), LabEx IGO Brest, France
| | | | - Sandra Lesven
- Clinical Research and Innovation Department, INSERM CIC 0502, CHU Brest, Brest, France
| | - Emmanuel Nowak
- Clinical Research and Innovation Department, INSERM CIC 0502, CHU Brest, Brest, France
| | - Aghiles Souki
- Clinical Research and Innovation Department, INSERM CIC 0502, CHU Brest, Brest, France
| | - Valérie Devauchelle-Pensec
- Department of Rheumatology, Université de Bretagne Occidentale, CHU Brest, INSERM (U1227), LabEx IGO Brest, France
| |
Collapse
|
4
|
Kanda R, Miyazaki Y, Nakayamada S, Fukuyo S, Kubo S, Miyagawa I, Yamaguchi A, Satoh-Kanda Y, Ohkubo N, Todoroki Y, Tanaka H, Ueno M, Nagayasu A, Fujita Y, Aritomi T, Kusaka K, Sakai H, Matsunaga S, Nohara H, Tanaka Y. Effective Second-Line b/tsDMARDs for Patients with Rheumatoid Arthritis Unresponsive to First-Line b/tsDMARDs from the FIRST Registry. Rheumatol Ther 2025; 12:353-369. [PMID: 40025347 PMCID: PMC11920512 DOI: 10.1007/s40744-025-00747-9] [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: 11/22/2024] [Accepted: 01/23/2025] [Indexed: 03/04/2025] Open
Abstract
INTRODUCTION For patients with rheumatoid arthritis (RA) unresponsive to first-line biologic/targeted synthetic disease-modifying antirheumatic drugs (b/tsDMARDs), the selection of second-line b/tsDMARDs is crucial to prevent progression to difficult-to-treat rheumatoid arthritis (D2TRA). However, indicators for selection are lacking. This study aimed to identify optimal second-line b/tsDMARDs among the phase III treatment strategies based on European League Against Rheumatism (EULAR) RA management recommendations. METHODS A total of 687 RA patients treated with second-line b/tsDMARDs (tumor necrosis factor inhibitor (n = 246), interleukin-6 receptor inhibitor [n = 195], cytotoxic T-lymphocyte-associated protein 4 immunoglobulin [n = 119], and Janus kinase inhibitor [n = 127]) were enrolled between October 2013 and April 2023. Rates of patients achieving Clinical Disease Activity Index (CDAI) remission and CDAI low disease activity (LDA), changes in CDAI, persistence rates, and adverse events within 24 weeks after treatment initiation were compared among the four groups. Propensity score-based inverse probability of treatment weighting (PS-IPTW) was used to minimize selection bias. RESULTS After PS-IPTW adjustment, the Janus kinase inhibitor (JAKi) group had the highest persistence rate among the four groups. At 24 weeks, the JAKi group showed the greatest improvement in CDAI and the highest CDAI remission rate. Among patients treated with JAKi as second-line b/tsDMARDs, upadacitinib (UPA) was most likely to achieve CDAI remission at 24 weeks. The comparison between the UPA group (n = 32) and the non-UPA JAKi group (tofacitinib and baricitinib [n = 95]) showed comparable persistence rates but significantly lower CDAI scores and higher CDAI remission rate at 24 weeks in the UPA group. No significant difference was noted in the overall incidence of adverse events among the four groups treated with b/tsDMARDs or between the groups treated with JAKi. CONCLUSIONS Selecting JAKi, especially UPA, may effectively improve the disease activity for RA patients unresponsive to first-line b/tsDMARDs. Further large-scale studies are needed to clarify the efficacy and safety of UPA. TRIAL REGISTRATION FIRST registry (approval number#04-23): October 2013, retrospectively registered.
Collapse
Affiliation(s)
- Ryuichiro Kanda
- First Department of Internal Medicine, School of Medicine, University of Occupational and Environmental Health, 1-1 Iseigaoka, Yahata-nishi-ku, Kitakyushu, Fukuoka, 807-8555, Japan
| | - Yusuke Miyazaki
- First Department of Internal Medicine, School of Medicine, University of Occupational and Environmental Health, 1-1 Iseigaoka, Yahata-nishi-ku, Kitakyushu, Fukuoka, 807-8555, Japan
| | - Shingo Nakayamada
- First Department of Internal Medicine, School of Medicine, University of Occupational and Environmental Health, 1-1 Iseigaoka, Yahata-nishi-ku, Kitakyushu, Fukuoka, 807-8555, Japan
| | - Shunsuke Fukuyo
- First Department of Internal Medicine, School of Medicine, University of Occupational and Environmental Health, 1-1 Iseigaoka, Yahata-nishi-ku, Kitakyushu, Fukuoka, 807-8555, Japan
| | - Satoshi Kubo
- First Department of Internal Medicine, School of Medicine, University of Occupational and Environmental Health, 1-1 Iseigaoka, Yahata-nishi-ku, Kitakyushu, Fukuoka, 807-8555, Japan
- Department of Molecular Targeted Therapies, School of Medicine, University of Occupational and Environmental Health, 1-1 Iseigaoka, Yahata-nishi-ku, Kitakyushu, Fukuoka, 807-8555, Japan
| | - Ippei Miyagawa
- First Department of Internal Medicine, School of Medicine, University of Occupational and Environmental Health, 1-1 Iseigaoka, Yahata-nishi-ku, Kitakyushu, Fukuoka, 807-8555, Japan
| | - Ayako Yamaguchi
- First Department of Internal Medicine, School of Medicine, University of Occupational and Environmental Health, 1-1 Iseigaoka, Yahata-nishi-ku, Kitakyushu, Fukuoka, 807-8555, Japan
- Department of Laboratory and Transfusion Medicine, Hospital of the University of the Occupational and Environmental Health, 1-1 Iseigaoka, Yahata-nishi-ku, Kitakyushu, Fukuoka, 807-8555, Japan
| | - Yurie Satoh-Kanda
- First Department of Internal Medicine, School of Medicine, University of Occupational and Environmental Health, 1-1 Iseigaoka, Yahata-nishi-ku, Kitakyushu, Fukuoka, 807-8555, Japan
| | - Naoaki Ohkubo
- First Department of Internal Medicine, School of Medicine, University of Occupational and Environmental Health, 1-1 Iseigaoka, Yahata-nishi-ku, Kitakyushu, Fukuoka, 807-8555, Japan
- Department of Environmental Epidemiology, University of Occupational and Environmental Health, 1-1 Iseigaoka, Yahata-nishi-ku, Kitakyushu, Fukuoka, 807-8555, Japan
| | - Yasuyuki Todoroki
- First Department of Internal Medicine, School of Medicine, University of Occupational and Environmental Health, 1-1 Iseigaoka, Yahata-nishi-ku, Kitakyushu, Fukuoka, 807-8555, Japan
- Department of Molecular Targeted Therapies, School of Medicine, University of Occupational and Environmental Health, 1-1 Iseigaoka, Yahata-nishi-ku, Kitakyushu, Fukuoka, 807-8555, Japan
| | - Hiroaki Tanaka
- First Department of Internal Medicine, School of Medicine, University of Occupational and Environmental Health, 1-1 Iseigaoka, Yahata-nishi-ku, Kitakyushu, Fukuoka, 807-8555, Japan
| | - Masanobu Ueno
- First Department of Internal Medicine, School of Medicine, University of Occupational and Environmental Health, 1-1 Iseigaoka, Yahata-nishi-ku, Kitakyushu, Fukuoka, 807-8555, Japan
| | - Atsushi Nagayasu
- First Department of Internal Medicine, School of Medicine, University of Occupational and Environmental Health, 1-1 Iseigaoka, Yahata-nishi-ku, Kitakyushu, Fukuoka, 807-8555, Japan
| | - Yuya Fujita
- First Department of Internal Medicine, School of Medicine, University of Occupational and Environmental Health, 1-1 Iseigaoka, Yahata-nishi-ku, Kitakyushu, Fukuoka, 807-8555, Japan
| | - Takafumi Aritomi
- First Department of Internal Medicine, School of Medicine, University of Occupational and Environmental Health, 1-1 Iseigaoka, Yahata-nishi-ku, Kitakyushu, Fukuoka, 807-8555, Japan
- Department of Laboratory and Transfusion Medicine, Hospital of the University of the Occupational and Environmental Health, 1-1 Iseigaoka, Yahata-nishi-ku, Kitakyushu, Fukuoka, 807-8555, Japan
| | - Katsuhide Kusaka
- First Department of Internal Medicine, School of Medicine, University of Occupational and Environmental Health, 1-1 Iseigaoka, Yahata-nishi-ku, Kitakyushu, Fukuoka, 807-8555, Japan
| | - Hidenori Sakai
- First Department of Internal Medicine, School of Medicine, University of Occupational and Environmental Health, 1-1 Iseigaoka, Yahata-nishi-ku, Kitakyushu, Fukuoka, 807-8555, Japan
| | - Satsuki Matsunaga
- First Department of Internal Medicine, School of Medicine, University of Occupational and Environmental Health, 1-1 Iseigaoka, Yahata-nishi-ku, Kitakyushu, Fukuoka, 807-8555, Japan
| | - Hirotsugu Nohara
- First Department of Internal Medicine, School of Medicine, University of Occupational and Environmental Health, 1-1 Iseigaoka, Yahata-nishi-ku, Kitakyushu, Fukuoka, 807-8555, Japan
| | - Yoshiya Tanaka
- First Department of Internal Medicine, School of Medicine, University of Occupational and Environmental Health, 1-1 Iseigaoka, Yahata-nishi-ku, Kitakyushu, Fukuoka, 807-8555, Japan.
| |
Collapse
|
5
|
Mikaeili B, Alqahtani ZA, Hincapie AL, Guo JJ. Safety of Janus kinase inhibitors in rheumatoid arthritis: a disproportionality analysis using FAERS database from 2012 to 2022. Clin Rheumatol 2025; 44:1467-1474. [PMID: 39945946 PMCID: PMC11993438 DOI: 10.1007/s10067-025-07360-9] [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: 10/15/2024] [Revised: 01/24/2025] [Accepted: 01/27/2025] [Indexed: 04/13/2025]
Abstract
INTRODUCTION/OBJECTIVE Janus kinase (JAK) inhibitors have expanded treatment options for rheumatoid arthritis (RA), particularly for patients unresponsive to traditional disease-modifying antirheumatic drugs (DMARDs). However, safety concerns necessitate a thorough post-market evaluation. This study is aimed at comparing the safety profiles of tofacitinib, baricitinib, and upadacitinib using adverse event (AE) reports from the FDA Adverse Event Reporting System (FAERS). METHODS A retrospective disproportionality analysis was performed using the FAERS data from 2012 to 2022. The AE reports were categorized into cardiovascular, cancer, respiratory, gastrointestinal, musculoskeletal, and arthralgia-related events. Proportional reporting ratio (PRR) and reporting odds ratios (RORs) with 95% confidence intervals (CIs) were calculated to identify significant safety signals. RESULTS Of 273,657 AE reports, tofacitinib had the most (227,144), with increased musculoskeletal-related events (ROR = 1.53, 95% CI 1.49-1.57) and a reduced cancer risk (ROR = 0.44, 95% CI 0.41-0.47). Baricitinib (9305 reports) showed the highest risk of cardiovascular events (ROR = 1.63, 95% CI 1.50-1.78) and cancer (ROR = 2.17, 95% CI 1.83-2.58). Upadacitinib (37,208 reports) had elevated risks for respiratory events (ROR = 2.04, 95% CI 1.88-2.21) and cancer (ROR = 2.24, 95% CI 2.05-2.43). CONCLUSION The distinct safety profiles of these JAK inhibitors suggest that baricitinib poses higher cardiovascular and cancer risks, whereas upadacitinib increases the risk of respiratory and gastrointestinal events. Tofacitinib may be safer for patients with a history of cancer but requires monitoring for musculoskeletal AEs. Personalized risk assessments are critical for safe use of JAK inhibitors. Key Points • This study provides a comprehensive post-market safety assessment of three JAK inhibitors-tofacitinib, baricitinib, and upadacitinib-using the FAERS data from 2012 to 2022. • Distinct safety profiles were identified, with baricitinib showing a higher risk of cardiovascular events and cancer, while upadacitinib posed an increased risk of respiratory and gastrointestinal events. • Tofacitinib demonstrated a lower cancer risk than other JAK inhibitors but was associated with more musculoskeletal-related adverse events. • These findings emphasize the importance of personalized risk assessment and vigilant monitoring when prescribing JAK inhibitors for rheumatoid arthritis.
Collapse
Affiliation(s)
- Bahar Mikaeili
- Division of Pharmacy Practice & Administrative Sciences, James L. Winkle College of Pharmacy, University of Cincinnati, Cincinnati, OH, USA.
| | - Zuhair A Alqahtani
- Division of Pharmacy Practice & Administrative Sciences, James L. Winkle College of Pharmacy, University of Cincinnati, Cincinnati, OH, USA
- College of Pharmacy, Health Economics and Outcome Research, King Saud University, Riyadh, Saudi Arabia
| | - Ana L Hincapie
- Division of Pharmacy Practice & Administrative Sciences, James L. Winkle College of Pharmacy, University of Cincinnati, Cincinnati, OH, USA
| | - Jeff Jianfei Guo
- Division of Pharmacy Practice & Administrative Sciences, James L. Winkle College of Pharmacy, University of Cincinnati, Cincinnati, OH, USA
| |
Collapse
|
6
|
Ludwig MP, Wilson JR, Galbraith MD, Bhandari N, Dunn LN, Black JC, Sullivan KD. NF-κB signaling directs a program of transient amplifications at innate immune response genes. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.11.641929. [PMID: 40161744 PMCID: PMC11952383 DOI: 10.1101/2025.03.11.641929] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
The cellular response to pathogens involves an intricate response directed by key innate immune signaling pathways which is characterized by cell-to-cell heterogeneity. How this heterogeneity is established and regulated remains unclear. We describe a program of transient site-specific gains (TSSG) producing extrachromosomal DNA (ecDNA) of immune-related genes in response to innate immune signaling. Activation of NF-κB drives TSSG of the interferon receptor gene cluster through inducible recruitment of the transcription factor RelA and the pre-replication complex member MCM2 to an epigenetically regulated TSSG control element. Targeted recruitment of RelA or p300 are sufficient to induce TSSG formation. RelA and MCM2 specify a program of TSSG for at least six and as many as 179 regions enriched in innate immune response genes. Identification of this program reveals regulated production of ecDNA as a mechanism of heterogeneity in the host response.
Collapse
Affiliation(s)
- Michael P. Ludwig
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- These Authors Contributed Equally
| | - Jason R. Wilson
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- These Authors Contributed Equally
| | - Matthew D. Galbraith
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- RNA Bioscience Initiative, University of Colorado Anschutz Medical Campus; Aurora, CO, USA
| | - Nirajan Bhandari
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Lauren N. Dunn
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Joshua C. Black
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Kelly D. Sullivan
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- RNA Bioscience Initiative, University of Colorado Anschutz Medical Campus; Aurora, CO, USA
- Department of Pediatrics, Section of Developmental Biology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- Lead Contact
| |
Collapse
|
7
|
Hui CW, Wu WC, Tong TK, Shiu C, Ng HL, Leung SO. Discovery of a new anti-γc antibody in clinical development for the treatment of autoimmune diseases. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2025:vkae020. [PMID: 40163668 DOI: 10.1093/jimmun/vkae020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Accepted: 11/11/2024] [Indexed: 04/02/2025]
Abstract
Autoimmune disease refers to a condition when the immune system anomalously attacks its own body and healthy cells. Although the exact causes of autoimmune diseases are unknown, it is recognized that excessive or aberrant cytokine responses contribute significantly to the development of autoimmunity. Among them, the common gamma c chain (γc) cytokines driven signaling cascade plays an indispensable role in driving pathogenic immune responses in patients with autoimmune diseases. Thus, we hypothesize that the development of an antibody targeting γc receptor could serve as a potential approach for treating autoimmune diseases and fulfil the unmet medical needs in this area. Here, we demonstrate that a humanized anti-γc antibody, hC2, could show high binding affinity to the human γc receptor and suppress 6 γc cytokines (interleukin [IL]-2, IL-4, IL-7, IL-9, IL-15 and IL-21)-driven STAT phosphorylation, leading to inhibition of autoimmunity and activation in B, T, and natural killer cell lines. Similar inhibitory effects were observed in the human peripheral blood mononuclear cell culture. Moreover, administration of hC2 could reduce expansion and tissue infiltration of T helper and cytotoxic T cells, leading to attenuation of damages to skin, liver, and kidney in the humanized xenograft mouse model. The current study demonstrates the potential of γc blockades for the treatment of T cell-mediated autoimmune diseases and chronic graft-versus-host disease. Anti-γc antibody hC2 might offer a more efficacious therapy compared with antibodies targeting a single γc cytokine and safer therapy than JAK inhibitors to fulfill the unmet medical needs in the autoimmune diseases in the future.
Collapse
Affiliation(s)
- Chin Wai Hui
- SinoMab BioScience Limited, Pak Shek Kok, New Territories, Hong Kong, China
| | - Wai Chung Wu
- SinoMab BioScience Limited, Pak Shek Kok, New Territories, Hong Kong, China
| | - Tak Keung Tong
- SinoMab BioScience Limited, Pak Shek Kok, New Territories, Hong Kong, China
| | - Carol Shiu
- SinoMab BioScience Limited, Pak Shek Kok, New Territories, Hong Kong, China
| | - Hoi Lam Ng
- SinoMab BioScience Limited, Pak Shek Kok, New Territories, Hong Kong, China
| | - Shui On Leung
- SinoMab BioScience Limited, Pak Shek Kok, New Territories, Hong Kong, China
| |
Collapse
|
8
|
Nash P, Kerschbaumer A, Konzett V, Aletaha D, Dörner T, Fleischmann R, McInnes I, Primdahl J, Sattar N, Tanaka Y, Trauner M, Winthrop K, de Wit M, Askling J, Baraliakos X, Boehncke WH, Emery P, Gossec L, Isaacs JD, Krauth M, Lee EB, Maksymowych W, Pope J, Scholte-Voshaar M, Schreiber K, Schreiber S, Stamm T, Taylor PC, Takeuchi T, Tam LS, Van den Bosch F, Westhovens R, Zeitlinger M, Smolen JS. Expert consensus statement on the treatment of immune-mediated inflammatory diseases with Janus kinase inhibitors: 2024 update. Ann Rheum Dis 2025:S0003-4967(25)00181-5. [PMID: 40037995 DOI: 10.1016/j.ard.2025.01.032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Revised: 11/30/2024] [Accepted: 12/16/2024] [Indexed: 03/06/2025]
Abstract
In light of the introduction of new Janus kinase inhibitors (JAKi), new indications for JAKi and recent safety considerations that have arisen since the preceding consensus statement on JAKi therapy, a multidisciplinary taskforce was assembled, encompassing patients, health care professionals, and clinicians with expertise in JAKi therapy across specialties. This taskforce, informed by two comprehensive systematic literature reviews, undertook the objective to update the previous expert consensus for using JAKi developed in 2019. The taskforce deliberated on overarching principles, indications, dosage and comedication strategies, warnings and contraindications, screening protocols, monitoring recommendations, and adverse effect profiles. The methodology was based on the European Alliance of Associations for Rheumatology standard operating procedures, with voting on these important elements. Furthermore, an updated research agenda was proposed. The task force did not address when a JAKi should be prescribed but rather considerations once this decision has been made. This update aimed to equip clinicians with the necessary knowledge and guidance for the efficient and safe administration of this expanding and significant class of drugs.
Collapse
Affiliation(s)
- Peter Nash
- Griffith University School of Medicine, Nathan, Brisbane, QLD, Australia
| | - Andreas Kerschbaumer
- Division of Rheumatology, Department of Medicine 3, Medical University of Vienna, Vienna, Austria
| | - Victoria Konzett
- Division of Rheumatology, Department of Medicine 3, Medical University of Vienna, Vienna, Austria
| | - Daniel Aletaha
- Division of Rheumatology, Department of Medicine 3, Medical University of Vienna, Vienna, Austria
| | - Thomas Dörner
- Department of Medicine/Rheumatology and Clinical Immunology, Charité University Hospital, Berlin, Germany
| | - Roy Fleischmann
- Metroplex Clinical Research Center and University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Iain McInnes
- Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, UK
| | - Jette Primdahl
- Department of Regional Health Research, University of Southern Denmark, Odense, Denmark
| | - Naveed Sattar
- School of Cardiovascular and Metabolic Health, University of Glasgow, UK
| | - Yoshiya Tanaka
- First Department of Internal Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Michael Trauner
- Division of Gastroenterology and Hepatology, Department of Medicine 3, Medical University of Vienna, Vienna, Austria
| | - Kevin Winthrop
- Division of Infectious Diseases and School of Public Health, Oregon Health and Science University, Portland, OR, USA
| | - Maarten de Wit
- Medical Humanities, Amsterdam University Medical Centre, Amsterdam, Netherlands
| | - Johan Askling
- Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
| | | | - Wolf-Henning Boehncke
- Division of Dermatology and Venereology, Geneva University Hospitals, Geneva, Switzerland; Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Paul Emery
- Leeds NIHR Biomedical Research Centre, LTHT, Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Leeds, UK
| | - Laure Gossec
- Sorbonne Université, INSERM, Institut Pierre Louis d'Epidémiologie et de Santé Publique, Paris France, AP-HP, Pitié-Salpêtrière Hospital, Rheumatology Department, Paris, France
| | - John D Isaacs
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UKNIHR Newcastle Biomedical Research Centre and Musculoskeletal Unit, Newcastle upon Tyne Hospitals, Newcastle upon Tyne, UK
| | - Maria Krauth
- Division of Haematology and Haemostaseology, Department of Medicine 1, Medical University of Vienna, Vienna, Austria
| | - Eun Bong Lee
- Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Walter Maksymowych
- Medicine, University of Alberta Faculty of Medicine and Dentistry, Edmonton, AB, Canada
| | - Janet Pope
- Medicine, Division of Rheumatology, The University of Western Ontario, London, ON, Canada
| | - Marieke Scholte-Voshaar
- Department of Pharmacy, Sint Maartenskliniek, Netherlands; Department of Pharmacy, Radboudumc, Nijmegen, Netherlands
| | - Karen Schreiber
- Danish Centre for Expertise in Rheumatology (CeViG), Danish Hospital for Rheumatic Diseases, Sønderborg, Denmark; Department of Regional Health Research (IRS), University of Southern Denmark, Odense, Denmark; Thrombosis and Haemostasis, Guy's and St Thomas' NHS Foundation Trust, London, UK
| | - Stefan Schreiber
- Department Internal Medicine I, University Hospital Schleswig-Holstein, Kiel University, Kiel, Germany
| | - Tanja Stamm
- Section for Outcomes Research, Center for Medical Statistics, Informatics, and Intelligent Systems, Medical University of Vienna, Vienna, Austria
| | - Peter C Taylor
- Botnar Research Centre, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, UK
| | - Tsutomu Takeuchi
- Division of Rheumatology, Department of Internal Medicine, Keio University School of Medicine, Tokyo, and Saitama Medical University, Saitama, Japan
| | - Lai-Shan Tam
- Rheumatology, Department of Medicine and Therapeutics, Chinese University of Hong Kong Shaw College, New Territories, Hong Kong
| | - Filip Van den Bosch
- VIB-UGent Center for Inflammation Research, Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium; Ghent University Hospital, Department of Rheumatology, Ghent, Belgium
| | - Rene Westhovens
- Department of Development and Regeneration, Skeletal Biology and Engineering Research Center, KU Leuven, Leuven, Belgium
| | - Markus Zeitlinger
- Department of Clinical Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Josef S Smolen
- Division of Rheumatology, Department of Medicine 3, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
9
|
Huang F, Hu D, Fan H, Hu B, Liu Y, Dong W, Liu X, Li Y, Yan D, Ding R, Niu S, Chen L, Nie X, Fang Y. Efficacy and Safety of Janus Kinase Inhibitors in Patients with Vitiligo: A Systematic Review and Meta-Analysis. Clin Pharmacol Ther 2025; 117:659-669. [PMID: 39713946 PMCID: PMC11835431 DOI: 10.1002/cpt.3538] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Accepted: 12/08/2024] [Indexed: 12/24/2024]
Abstract
Although several case reports and small clinical trials have reported promising outcomes with Janus kinase (JAK) inhibitors for vitiligo, high-quality evidence and guidelines are lacking. We evaluated the efficacy and safety of JAK inhibitors for the treatment of vitiligo using a meta-analysis of randomized controlled trials (RCTs). We searched the PubMed, Embase, and Cochrane Library databases up to August 2023, with additional studies from ClinicalTrials.gov and company websites. We assessed outcomes, including percentage improvement in total vitiligo area score index (TVASI) and facial vitiligo area score index (FVASI); the proportion of patients achieving 50% improvement in TVASI (TVASI50) and 50% and 75% improvement in FVASI (FVASI50 and FVASI75); the risk of treatment-emergent adverse events (TEAEs), serious adverse events (SAEs), infections, and skin-related adverse events (AEs). Five studies with 1,550 participants were included. JAK inhibitors were associated with a higher proportion of TVASI50 (relative risk [RR] 2.67, 95% confidence interval [CI] 1.24-5.78) and FVASI75 (RR 3.97, 95%CI 2.62-6.02) responders than placebo. JAK inhibitors significantly increased the risk of skin-related AEs (RR 1.96, 95% CI 1.29-2.98) compared with placebo. However, the risk of TEAEs, SAEs, and infections was not significantly different between the JAK inhibitor and placebo groups. Subgroup analysis showed that JAK1 and JAK1/2 inhibitors were more effective than JAK3 inhibitors. However, there was insufficient evidence to suggest that the route of administration affects the efficacy and safety of JAK inhibitors in vitiligo. These findings indicate that JAK inhibitors are effective in repigmentation and well tolerated in patients with vitiligo.
Collapse
Affiliation(s)
- Fan Huang
- Clinical Trial InstitutionPeking University People's HospitalBeijingChina
- Department of Pharmacy Administration and Clinical Pharmacy, School of Pharmaceutical SciencesPeking UniversityBeijingChina
| | - Dingyuan Hu
- Clinical Trial InstitutionPeking University People's HospitalBeijingChina
- Department of Pharmacy Administration and Clinical Pharmacy, School of Pharmaceutical SciencesPeking UniversityBeijingChina
| | - Huaying Fan
- Clinical Trial InstitutionPeking University People's HospitalBeijingChina
| | - Binyi Hu
- Clinical Trial InstitutionPeking University People's HospitalBeijingChina
- Department of Clinical PharmacyXuzhou Medical UniversityXuzhouChina
| | - Yian Liu
- Clinical Trial InstitutionPeking University People's HospitalBeijingChina
| | - Wenliang Dong
- Department of Pharmacy Administration and Clinical Pharmacy, School of Pharmaceutical SciencesPeking UniversityBeijingChina
| | - Xiangxing Liu
- Clinical Trial InstitutionPeking University People's HospitalBeijingChina
- Department of Clinical PharmacyXuzhou Medical UniversityXuzhouChina
| | - Yanting Li
- Clinical Trial InstitutionPeking University People's HospitalBeijingChina
| | - Diqin Yan
- Clinical Trial InstitutionPeking University People's HospitalBeijingChina
- Department of Pharmacy Administration and Clinical Pharmacy, School of Pharmaceutical SciencesPeking UniversityBeijingChina
| | - Rui Ding
- Clinical Trial InstitutionPeking University People's HospitalBeijingChina
| | - Suping Niu
- Clinical Trial InstitutionPeking University People's HospitalBeijingChina
| | - Liming Chen
- Clinical Trial InstitutionPeking University People's HospitalBeijingChina
| | - Xiaoyan Nie
- Department of Pharmacy Administration and Clinical Pharmacy, School of Pharmaceutical SciencesPeking UniversityBeijingChina
| | - Yi Fang
- Clinical Trial InstitutionPeking University People's HospitalBeijingChina
| |
Collapse
|
10
|
Yamamoto Y, Shirai Y, Edahiro R, Kumanogoh A, Okada Y. Large-scale cross-trait genetic analysis highlights shared genetic backgrounds of autoimmune diseases. Immunol Med 2025; 48:1-10. [PMID: 39171621 DOI: 10.1080/25785826.2024.2394258] [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: 06/25/2024] [Accepted: 08/15/2024] [Indexed: 08/23/2024] Open
Abstract
Disorders associated with the immune system burden multiple organs, although the shared biology exists across the diseases. Preceding family-based studies reveal that immune diseases are heritable to varying degrees, providing the basis for immunogenomics. The recent cost reduction in genetic analysis intensively promotes biobank-scale studies and the development of frameworks for statistical genetics. The accumulating multi-layer omics data, including genome-wide association studies (GWAS) and RNA-sequencing at single-cell resolution, enable us to dissect the genetic backgrounds of immune-related disorders. Although autoimmune and allergic diseases are generally categorized into different disease categories, epidemiological studies reveal the high incidence of autoimmune and allergic disease complications, suggesting the shared genetics and biology between the disease categories. Biobank resources and consortia cover multiple immune-related disorders to accumulate phenome-wide associations of genetic variants and enhance researchers to analyze the shared and heterogeneous genetic backgrounds. The emerging post-GWAS and integrative multi-omics analyses provide genetic and biological insights into the multicategorical disease associations.
Collapse
Affiliation(s)
- Yuji Yamamoto
- Department of Statistical Genetics, Osaka University Graduate School of Medicine, Suita, Japan
- Department of Respiratory Medicine and Clinical Immunology, Osaka University Graduate School of Medicine, Suita, Japan
| | - Yuya Shirai
- Department of Statistical Genetics, Osaka University Graduate School of Medicine, Suita, Japan
- Department of Respiratory Medicine and Clinical Immunology, Osaka University Graduate School of Medicine, Suita, Japan
- Laboratory of Statistical Immunology, Immunology Frontier Research Center (WPI-IFReC), Osaka University, Suita, Japan
- RIKEN Center for Integrative Medical Sciences, Laboratory for Systems Genetics, Yokohama, Japan
| | - Ryuya Edahiro
- Department of Statistical Genetics, Osaka University Graduate School of Medicine, Suita, Japan
- Department of Respiratory Medicine and Clinical Immunology, Osaka University Graduate School of Medicine, Suita, Japan
- RIKEN Center for Integrative Medical Sciences, Laboratory for Systems Genetics, Yokohama, Japan
| | - Atsushi Kumanogoh
- Department of Respiratory Medicine and Clinical Immunology, Osaka University Graduate School of Medicine, Suita, Japan
- Department of Immunopathology, Immunology Frontier Research Center (WPI-IFReC), Osaka University, Suita, Japan
- Integrated Frontier Research for Medical Science Division, Institute for Open and Transdisciplinary Research Initiatives (OTRI), Osaka University, Suita, Japan
- Center for Infectious Diseases for Education and Research (CiDER), Osaka University, Suita, Japan
- Japan Agency for Medical Research and Development, Core Research for Evolutional Science and Technology (AMED-CREST), Tokyo, Japan
- Center for Advanced Modalities and DDS (CAMaD), Osaka University, Suita, Japan
| | - Yukinori Okada
- Department of Statistical Genetics, Osaka University Graduate School of Medicine, Suita, Japan
- Laboratory of Statistical Immunology, Immunology Frontier Research Center (WPI-IFReC), Osaka University, Suita, Japan
- RIKEN Center for Integrative Medical Sciences, Laboratory for Systems Genetics, Yokohama, Japan
- Center for Infectious Diseases for Education and Research (CiDER), Osaka University, Suita, Japan
- Center for Advanced Modalities and DDS (CAMaD), Osaka University, Suita, Japan
- Department of Genome Informatics, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
- Premium Research Institute for Human Metaverse Medicine (WPI-PRIMe), Osaka University, Suita, Japan
| |
Collapse
|
11
|
Currie GR, Storek J, MacDonald KV, Hazlewood G, Durand C, Bridges JFP, Mosher D, Marshall DA. Measuring Patient Preferences to Inform Clinical Trial Design: An Example in Rheumatoid Arthritis. THE PATIENT 2025; 18:161-171. [PMID: 39666176 DOI: 10.1007/s40271-024-00724-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 11/21/2024] [Indexed: 12/13/2024]
Abstract
BACKGROUND Allogeneic bone marrow transplantation (BMT) may be a curative treatment for patients with rheumatoid arthritis (RA), but it has serious risks, including death. It is uncertain whether patients would accept the risks and benefits of BMT and participate in clinical trials. We conducted a discrete choice experiment (DCE) to quantify risk tolerance and benefit-risk trade-offs to inform the design of a clinical trial for BMT. METHODS We conducted a DCE with three attributes (three levels each): chance of stopping disease progression (50-90%), increased chance of death in year after transplant (3-15%), and chance of chronic graft-versus-host disease (cGVHD) (3-15%). An orthogonal main effects design of nine binary choice tasks were presented for two scenarios: one considering their current situation and a second scenario where the patient has failed seven anti-rheumatic drugs. Participants were recruited from the Rheum4U inflammatory arthritis registry. Choice data were analyzed using a logit model accounting for multiple responses per participant. RESULTS Sixty patients participated. Most (82%) had severe disease, and the median number of anti-rheumatic drugs previously taken was 6 (range 0-18). As expected, an increased chance of stopping disease progression increases the probability of choosing BMT, while increased chance of both risks decreases the probability. Patients were willing to accept a 3% increase in risk of death or 6% increase in chance of chronic GVHD for a 10% increase in the chance of stopping disease progression. For the most clinically likely BMT risk-benefit profiles, and the likely initial target population of patients who have failed multiple biologics, between 72% and 91% of patients would choose BMT. CONCLUSIONS Patients with RA are willing to accept substantial risks for a chance to stop disease progression with BMT, suggesting that a pilot trial of BMT for RA could successfully recruit patients. Preference studies have an important role in informing patient-centered clinical trial planning and design.
Collapse
Affiliation(s)
- Gillian R Currie
- Department of Community Health Sciences, University of Calgary, Calgary, AB, Canada.
| | - Jan Storek
- Department of Hematology, University of Calgary, Calgary, AB, Canada
| | - Karen V MacDonald
- Department of Community Health Sciences, University of Calgary, Calgary, AB, Canada
| | - Glen Hazlewood
- Department of Medicine, University of Calgary, Calgary, AB, Canada
| | - Caylib Durand
- Department of Medicine, University of Calgary, Calgary, AB, Canada
| | - John F P Bridges
- Department of Biomedical Informatics, The Ohio State University College of Medicine, Columbus, OH, USA
| | - Dianne Mosher
- Department of Medicine, University of Calgary, Calgary, AB, Canada
| | - Deborah A Marshall
- Department of Community Health Sciences, University of Calgary, Calgary, AB, Canada
| |
Collapse
|
12
|
Wang J, Shi H, Yang Y, Gong X. Crosstalk between ferroptosis and innate immune in diabetic kidney disease: mechanisms and therapeutic implications. Front Immunol 2025; 16:1505794. [PMID: 40092979 PMCID: PMC11906378 DOI: 10.3389/fimmu.2025.1505794] [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: 10/03/2024] [Accepted: 02/10/2025] [Indexed: 03/19/2025] Open
Abstract
Diabetic kidney disease (DKD) is a prevalent complication of diabetes mellitus (DM), and its incidence is increasing alongside the number of diabetes cases. Effective treatment and long-term management of DKD present significant challenges; thus, a deeper understanding of its pathogenesis is essential to address this issue. Chronic inflammation and abnormal cell death in the kidney closely associate with DKD development. Recently, there has been considerable attention focused on immune cell infiltration into renal tissues and its inflammatory response's role in disease progression. Concurrently, ferroptosis-a novel form of cell death-has emerged as a critical factor in DKD pathogenesis, leading to increased glomerular filtration permeability, proteinuria, tubular injury, interstitial fibrosis, and other pathological processes. The cardiorenal benefits of SGLT2 inhibitors (SGLT2-i) in DKD patients have been demonstrated through numerous large clinical trials. Moreover, further exploratory experiments indicate these drugs may ameliorate serum and urinary markers of inflammation, such as TNF-α, and inhibit ferroptosis in DKD models. Consequently, investigating the interplay between ferroptosis and innate immune and inflammatory responses in DKD is essential for guiding future drug development. This review presents an overview of ferroptosis within the context of DKD, beginning with its core mechanisms and delving into its potential roles in DKD progression. We will also analyze how aberrant innate immune cells, molecules, and signaling pathways contribute to disease progression. Finally, we discuss the interactions between ferroptosis and immune responses, as well as targeted therapeutic agents, based on current evidence. By analyzing the interplay between ferroptosis and innate immunity alongside its inflammatory responses in DKD, we aim to provide insights for clinical management and drug development in this area.
Collapse
Affiliation(s)
- Jinyang Wang
- Department of Geriatric Integrative, Second Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China
| | - Haonan Shi
- School of Medicine, Shanghai University, Shanghai, China
| | - Ye Yang
- Department of Geriatric Integrative, Second Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China
| | - Xueli Gong
- Department of Pathophysiology, School of Basic Medical Science, Xinjiang Medical University, Urumqi, Xinjiang, China
| |
Collapse
|
13
|
Konzett V, Smolen JS, Nash P, Winthrop K, Aletaha D, Dörner T, Fleischmann R, Tanaka Y, Primdahl J, Baraliakos X, McInnes IB, Trauner M, Sattar N, de Wit M, Schoones JW, Kerschbaumer A. Safety of Janus kinase inhibitors in immune-mediated inflammatory diseases-a systematic literature review informing the 2024 update of an international expert consensus statement. Ann Rheum Dis 2025:S0003-4967(25)00080-9. [PMID: 39934016 DOI: 10.1016/j.ard.2025.01.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2024] [Revised: 11/19/2024] [Accepted: 11/25/2024] [Indexed: 02/13/2025]
Abstract
OBJECTIVES This systematic literature review (SLR) on safety outcomes was performed to inform the 2024 update of the expert consensus statement on the treatment of immune-mediated inflammatory diseases (IMIDs) with Janus kinase inhibitors (JAKi). METHODS An update of the 2019 SLR was performed in MEDLINE, Embase, and the Cochrane Library. For safety, randomised, placebo-controlled or active-controlled trials on all JAKi investigated in IMIDs, long-term extension (LTE) studies, pooled trial data analyses, and cohort and claims studies were included. RESULTS We screened 13,905 records, of which 209 were finally included. Three safety trials and 13 post hoc analyses, 83 efficacy randomised controlled trials (RCTs) with adequate safety reporting, 56 integrated safety analyses and LTE of RCTs, 20 additional conference abstracts on RCT data, as well as 37 real-world cohort studies were presented to the task force. Safety profiles of JAKi were overall consistent across compounds and indications, but impacts of patient profiles, treatment dosing, and other cofactors like background medications on drug safety could be observed. Furthermore, differential effects of variously selective JAKi on distinct adverse events of special interest (AESI) and laboratory outcomes were discerned. CONCLUSION A substantial amount of literature was published on JAKi safety since 2019. A comprehensive overview of these data supports the optimal use of JAKi in patients with IMIDs, by consideration and balance of their benefits as well as risks in every patient.
Collapse
Affiliation(s)
- Victoria Konzett
- Department of Medicine III, Division of Rheumatology, Medical University of Vienna, Vienna, Austria.
| | - Josef S Smolen
- Department of Medicine III, Division of Rheumatology, Medical University of Vienna, Vienna, Austria
| | - Peter Nash
- Griffith University School of Medicine, Gold Coast, QLD, Australia
| | | | - Daniel Aletaha
- Department of Medicine III, Division of Rheumatology, Medical University of Vienna, Vienna, Austria
| | - Thomas Dörner
- Rheumatology, Charite Medical Faculty Berlin, Berlin, Germany
| | - Roy Fleischmann
- Metroplex Clinical Research Center, University of Texas Southwestern Medical Center at Dallas, Dallas, TX, USA
| | - Yoshiya Tanaka
- The First Department of Internal Medicine, University of Occupational and Environmental Health, Japan, Kitakyushu, Japan
| | - Jette Primdahl
- Danish Hospital for Rheumatic Diseases, University Hospital of Southern Denmark, Sønderborg, Denmark
| | | | - Iain B McInnes
- College of Medical Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Michael Trauner
- Department of Medicine III, Division of Gastroenterology and Hepatology, Medical University of Vienna, Vienna, Austria
| | - Naveed Sattar
- Cardiovascular and Metabolic Health, University of Glasgow, Glasgow, UK
| | - Maarten de Wit
- Stichting Tools, Patient Research Partner, Amsterdam, the Netherlands
| | - Jan W Schoones
- Directorate of Research Policy, Leiden University Medical Center, Leiden, the Netherlands
| | - Andreas Kerschbaumer
- Department of Medicine III, Division of Rheumatology, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
14
|
Bala V, Hafström I, Svensson B, Ajeganova S. Persistently different patterns of patient's global assessment of health in rheumatoid arthritis are associated with pain and impaired function more than with inflammation: an inception cohort study over 15 years. RMD Open 2025; 11:e004744. [PMID: 39842872 PMCID: PMC11784202 DOI: 10.1136/rmdopen-2024-004744] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Accepted: 11/13/2024] [Indexed: 01/24/2025] Open
Abstract
OBJECTIVE To assess patient's self-reported global assessment of health (PGA) in relation to inflammation, pain and disability in patients with rheumatoid arthritis (RA). METHODS Data were obtained from the Better Anti-Rheumatic PharmacOTherapy early RA cohort, in which patients were scheduled for follow-up of 15 years. Longitudinal PGA trajectories were identified using hierarchical agglomerative clustering procedure with Wards methods, based on PGA assessments during the first 2 years. Multivariate linear regression and mixed models were used to evaluate associations between PGA, inflammation, pain and disability. RESULTS Totally 2238 patients were included in 1991-2006. Three PGA trajectories were identified, low, medium and high, including 34%, 48% and 18% of the patients, respectively. The similar PGA patterns were shown for the inclusion periods before and after 1999. The patients in the low PGA group were less often women, had lower body mass index, lower levels of inflammatory variables, visual analogue scale (VAS) pain and Health Assessment Questionnaire (HAQ) scores at baseline than patients in the higher PGA groups. After adjustments, smoking and antibodies to cyclic citrullinated peptide status differentiated between being in low and high PGA groups. During the first year, all PGA groups showed improved outcomes, most pronounced in low PGA group. Fewer patients in high PGA group achieved remission, independent of remission criteria used. The outcomes were consistently different between the PGA groups from 6 months and onwards. The PGA levels from baseline over 15 years were best explained by VAS pain, followed by HAQ score, Disease Activity Score on 28 joints-3 and tender joint count, adjusted R2 up to 77%, 41%, 27% and 26%, respectively. CONCLUSION Persistently higher PGA levels in RA were mostly related to pain and disability.
Collapse
Affiliation(s)
- Valentina Bala
- Department of Health Sciences, Faculty of Medicine, Lund University, Lund, Sweden
- Department of Medicine, Section of Rheumatology, Helsingborg Hospital, Helsingborg, Sweden
| | - Ingiäld Hafström
- Department of Medicine, Huddinge, Karolinska Institutet, Stockholm, Sweden
- Karolinska University Hospital, Stockholm, Sweden
| | | | - Sofia Ajeganova
- Department of Medicine, Huddinge, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
15
|
Zhang J, Wang C, Wu X, Shen Q, Du Y. Nanozyme-based therapeutic strategies for rheumatoid arthritis. J Control Release 2025; 377:716-734. [PMID: 39617172 DOI: 10.1016/j.jconrel.2024.11.072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 11/13/2024] [Accepted: 11/26/2024] [Indexed: 12/06/2024]
Abstract
Rheumatoid arthritis (RA) is a prevalent chronic autoimmune disease that leads to severe joint damage and disability. Conventional treatment options are limited by their efficacy and side effect profiles. Nanozymes, nanomaterials with enzyme-like activities, offer a novel therapeutic approach for RA. This review summarizes recent advances in nanozyme-based treatments, focusing on their antioxidant and immunomodulatory roles in mitigating RA. We discuss various nanozymes, including those based on cerium, iron, manganese, silver, copper, platinum, rhodium, and multi-metallic nanozymes, which mimic natural enzymes such as superoxide dismutase, catalase, and peroxidase to reduce oxidative stress. Additionally, we explore nanozyme-based combination therapies that integrate with other strategies, such as vesicles and phototherapy, to achieve synergistic effects and enhance efficacy. This review highlights the significant potential of nanozymes in improving RA treatment, offering a new perspective for future research and clinical applications.
Collapse
Affiliation(s)
- Jucong Zhang
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Chenxi Wang
- School of Pharmacy, Hangzhou Normal University, Hangzhou 311121, China
| | - Xiaochuan Wu
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Qiying Shen
- School of Pharmacy, Hangzhou Normal University, Hangzhou 311121, China
| | - Yongzhong Du
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China; Jinhua Institute of Zhejiang University, Jinhua, Zhejiang 321299, China.
| |
Collapse
|
16
|
Wu C, Bi C, Kim GS, Yang Z, Li S, Dai T, Wu X, Tan J, He N, Li S. Oral colon-targeted responsive chitosan/pectin-based nanoparticles propels the application of tofacitinib in colitis therapy. Sci Rep 2025; 15:1569. [PMID: 39794457 PMCID: PMC11723933 DOI: 10.1038/s41598-024-84322-2] [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: 07/24/2024] [Accepted: 12/23/2024] [Indexed: 01/13/2025] Open
Abstract
Tofacitinib (Tof), a commercially available pan-Janus kinases inhibitor, is approved for the treatment of moderate to severe ulcerative colitis. However, its clinical application is limited due to dose-dependent systemic side effects. The present study aims to develop an efficient oral colon-targeted drug delivery systems using prebiotic pectin (Pcn) and chitosan (Csn) polysaccharides as a shell, with Tof loaded into a Bovine Serum Albumin (BSA) core, and improving it with chondroitin sulfate (Chs), thus constructing Tof@BSA-Chs-CP nanoparticles (NPs). Our results suggest that the pH-sensitive characteristics of the Pcn/Csn shell contribute to its capacity for attenuating absorption and systemic diffusion in the gastrointestinal tract, and exhibiting targeted localization at inflamed colonic sites in mice. Additionally, the gut microbiota-secreted polysaccharide-degrading enzyme acts as the triggering agent for Pcn/Csn shell degradation. In mice colitis models, we demonstrated that oral administration of Tof@BSA-Chs-CP NPs effectively ameliorated colitis and expedited its resolution by modulating the expression of pro-inflammatory cytokines and immune regulatory factors. Collectively, our synthetic NPs demonstrate the promising potential of Tof for the therapy of UC.
Collapse
Affiliation(s)
- Chunfei Wu
- Medical School, Qingdao Huanghai University, Qingdao, China
- School of Basic Medicine, Qingdao Medical College, Qingdao University, Qingdao, China
| | - Chuanlin Bi
- Qingdao Institute for Food and Drug Control, Qingdao, China
- NMPA Key Laboratory for Quality Research and Evaluation of Traditional Marine Chinese Medicine, Qingdao, China
| | - Geun-Soo Kim
- School of Basic Medicine, Qingdao Medical College, Qingdao University, Qingdao, China
| | - Zizhen Yang
- School of Basic Medicine, Qingdao Medical College, Qingdao University, Qingdao, China
| | - Shuao Li
- School of Basic Medicine, Qingdao Medical College, Qingdao University, Qingdao, China
- Department of Ultrasound, the Affiliated Hospital of Qingdao University, Qingdao, 266003, China
| | - Tong Dai
- School of Basic Medicine, Qingdao Medical College, Qingdao University, Qingdao, China
| | - Xiaoyu Wu
- School of Basic Medicine, Qingdao Medical College, Qingdao University, Qingdao, China
| | - Jiaojiao Tan
- Medical School, Qingdao Huanghai University, Qingdao, China
| | - Ningning He
- School of Basic Medicine, Qingdao Medical College, Qingdao University, Qingdao, China.
| | - Shangyong Li
- School of Basic Medicine, Qingdao Medical College, Qingdao University, Qingdao, China.
- Department of Ultrasound, the Affiliated Hospital of Qingdao University, Qingdao, 266003, China.
| |
Collapse
|
17
|
Zavoriti A, Miossec P. Understanding Cardiovascular Events With JAK Inhibitors: Tofacitinib Reduces Synovial and Vascular Inflammation but not the Prothrombotic Effects of Inflammatory Cytokines on Endothelium. ACR Open Rheumatol 2025; 7:e11790. [PMID: 39800890 PMCID: PMC11725533 DOI: 10.1002/acr2.11790] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Revised: 10/30/2024] [Accepted: 12/09/2024] [Indexed: 01/16/2025] Open
Abstract
OBJECTIVE Inflammation drives cardiovascular disease in rheumatoid arthritis (RA). Treatment with tofacitinib, a JAK1/JAK3 inhibitor, is associated with increased cardiovascular events in patients with RA. Here, we determined its effects on cytokine production during interactions between immune cells at the synovial and vascular levels and its impact on endothelial activation and coagulation during inflammation. METHODS Activated human peripheral blood mononuclear cells (PBMCs) were cocultured with RA synoviocytes or endothelial cells (ECs) mimicking the cellular interactions in synovium and vessels responsible for cytokine production. A dose-response of tofacitinib was tested on interferon γ, interleukin (IL) 17A, IL-10, IL-6, and IL-1β, and cytokine production was measured by enzyme-linked immunosorbent assay at 48 hours. Endothelial activation was induced with IL-17A and tumor necrosis factor (TNF) or on contact with PBMCs. Shortly after tofacitinib treatment, the expression of EC activation markers, specifically IL-6, IL-8, vascular cell adhesion molecule 1 (VCAM-1), and E-selectin, and of coagulation, including tissue factor and thrombomodulin, was assessed by real-time polymerase chain reaction. RESULTS Tofacitinib differentially inhibited production of IFNɣ, IL-17A, and IL-10 from PBMC cocultures with RA synoviocytes or ECs (all P < 0.001). In cocultures with ECs, tofacitinib reduced further IL-6 and IL-8 production (both P < 0.001). In ECs activated by TNF/IL-17A or indirectly via contact with activated PBMCs, tofacitinib decreased IL-6 upregulation but not that of IL-8, E-selectin, or tissue factor. Thrombomodulin was significantly decreased. VCAM-1 was greatly induced with a higher dose of tofacitinib in ECs incubated directly with added inflammatory cytokines (P < 0.05) or released by interaction with activated PBMCs (P < 0.001). CONCLUSION Tofacitinib inhibits synovium and vascular inflammation but fails to prevent the prothrombotic effects of inflammatory cytokines on ECs.
Collapse
Affiliation(s)
- Aliki Zavoriti
- Hôpital Edouard HerriotHospices Civils de LyonLyonFrance
| | - Pierre Miossec
- Hôpital Edouard HerriotHospices Civils de LyonLyonFrance
| |
Collapse
|
18
|
Misselwitz B, Zeißig S, Schreiber S, Dignass A. [Application of advanced treatment in chronic inflammatory bowel diseases]. INNERE MEDIZIN (HEIDELBERG, GERMANY) 2025; 66:3-14. [PMID: 39747696 PMCID: PMC11761996 DOI: 10.1007/s00108-024-01833-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 12/09/2024] [Indexed: 01/04/2025]
Abstract
BACKGROUND The treatment options for chronic inflammatory bowel diseases (IBD) have been greatly expanded due to a better understanding of the underlying pathogenesis. A total of five classes of advanced treatment are available. OBJECTIVE A practical overview of advanced treatment of IBD. METHODS Narrative review. RESULTS AND DISCUSSION Advanced treatments are indicated for moderate to severe IBD. A timely use is recommended to achieve better response rates and to avoid irreversible bowel damage. Tumor necrosis factor (TNF) inhibitors and Janus kinase (JAK) inhibitors have a broad efficacy, also for extraintestinal disease manifestations. The risk of reactivation of varicella zoster virus is increased with JAK inhibitors. In high-risk patients and an age >65 years there is possibly a moderately elevated cardiovascular risk and neoplastic side effects. The integrin alpha4beta7 inhibitor vedolizumab and the interleukin (IL) 12 and 23 inhibitor ustekinumab have very good safety profiles. Selective IL-23 inhibitors are sometimes superior to ustekinumab with comparable safety profiles with respect to efficacy. The sphingosine-1-phosphate receptor modulators ozanimod and etrasimod are approved for oral treatment of ulcerative colitis. The treatment success of the medications remains still limited and a minority of patients will not respond to every individual treatment. Thus, sequential administration of several treatments is often needed. Due to the lack of comparative studies, the personalized choice, sequence and decision for treatments are usually based on personal experience and should take patient preferences, efficacy, safety and individual patient profiles into consideration.
Collapse
Affiliation(s)
| | - Sebastian Zeißig
- Klinik für Innere Medizin A, Universitätsmedizin Greifswald, Greifswald, Deutschland
| | - Stefan Schreiber
- Klinik für Innere Medizin I, Universitätsklinikum Schleswig-Holstein, Kiel, Deutschland
| | - Axel Dignass
- Medizinischen Klinik I, Agaplesion Markus Krankenhaus, Wilhelm-Epstein-Str. 4, 60431, Frankfurt/Main, Deutschland.
| |
Collapse
|
19
|
Li N, Xu T, Wu Z, Zhao Y, Ruan M, Xu H, Chen W, Wang H, Wang S, Wang Y, Liang Q. Arabinogalactan from Cynanchum atratum induces tolerogenic dendritic cells in gut to restrain autoimmune response and alleviate collagen-induced arthritis in mice. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 136:156269. [PMID: 39586124 DOI: 10.1016/j.phymed.2024.156269] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/28/2024] [Revised: 11/08/2024] [Accepted: 11/15/2024] [Indexed: 11/27/2024]
Abstract
BACKGROUND Rheumatoid arthritis (RA) is an autoimmune disease characterized by multiple joints lesions. Tolerogenic dendritic cells (tolDCs) play crucial roles in maintaining immune homeostasis. The immunomodulatory activity of plant-derived arabinogalactan (AGs) has been well investigated, however, whether AGs could suppress autoimmune responses by inducing tolDCs is remain unclear. DESIGN Collagen-induced arthritis (CIA, a mouse model of RA) mice were utilized to ascertain the role of AGs (obtained from Cynanchum atratum) in autoimmune responses. An antibiotic cocktail was administered to eliminate gut microbiota. Germ-free (GF) and Toll-like receptor 2 (TLR2) knockout mice were used to determine the function of AGs in intestinal immune cells. RESULTS The oral administration of dietary AGs substantially reduced the severity of CIA and rebalanced the ratio of regulatory T cells (Tregs) to T helper 17 (Th17) cells. Although the antibiotic cocktail depleted the mice's gut microbiota, AGs had a therapeutic effect on their CIA. AGs restored Treg/Th17 homeostasis by inducing CD103+ tolDCs, regardless of the gut microbiota of the GF mice. Coculture experiments confirmed that AGs induced tolDCs and transforming growth factor β (TGF-β) secretion, leading to Treg amplification. RNA sequencing and TLR2 knockout experiments revealed that AGs induced tolDCs through a TLR2-mediated mechanism. Preventive interventions with AGs established a tolerogenic intestinal immune microenvironment, which delayed the onset and progression of CIA. AGs functioned synergistically with tofacitinib, a JAK inhibitor, to effectively restore Treg/Th17 balance and alleviate CIA. CONCLUSION This study introduces a novel microbiota-independent mechanism through which soluble dietary AGs inhibit systemic autoimmune responses. Our findings provide insights into the supplementation of dietary AGs in patients with preclinical or progressive RA.
Collapse
Affiliation(s)
- Ning Li
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, 725 Wan-Ping South Road, Shanghai 200032, PR China; Spine Institute, Shanghai University of Traditional Chinese Medicine, 725 Wan-Ping South Road, Shanghai 200032, PR China
| | - Tianhao Xu
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, 725 Wan-Ping South Road, Shanghai 200032, PR China; Spine Institute, Shanghai University of Traditional Chinese Medicine, 725 Wan-Ping South Road, Shanghai 200032, PR China
| | - Zhaoshun Wu
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, 725 Wan-Ping South Road, Shanghai 200032, PR China; Spine Institute, Shanghai University of Traditional Chinese Medicine, 725 Wan-Ping South Road, Shanghai 200032, PR China
| | - Yuchen Zhao
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, 725 Wan-Ping South Road, Shanghai 200032, PR China; Spine Institute, Shanghai University of Traditional Chinese Medicine, 725 Wan-Ping South Road, Shanghai 200032, PR China
| | - Ming Ruan
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, 725 Wan-Ping South Road, Shanghai 200032, PR China; Spine Institute, Shanghai University of Traditional Chinese Medicine, 725 Wan-Ping South Road, Shanghai 200032, PR China
| | - Hao Xu
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, 725 Wan-Ping South Road, Shanghai 200032, PR China; Spine Institute, Shanghai University of Traditional Chinese Medicine, 725 Wan-Ping South Road, Shanghai 200032, PR China
| | - Weihao Chen
- The MOE Key Laboratory for Standardization of Chinese Medicines and the SATCM Key Laboratory for New Resources and Quality Evaluation of Chinese Medicines, Shanghai Key Laboratory of Compound Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, 1200 Cailun Road, Shanghai 201203, PR China
| | - Huijun Wang
- The MOE Key Laboratory for Standardization of Chinese Medicines and the SATCM Key Laboratory for New Resources and Quality Evaluation of Chinese Medicines, Shanghai Key Laboratory of Compound Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, 1200 Cailun Road, Shanghai 201203, PR China
| | - Shunchun Wang
- The MOE Key Laboratory for Standardization of Chinese Medicines and the SATCM Key Laboratory for New Resources and Quality Evaluation of Chinese Medicines, Shanghai Key Laboratory of Compound Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, 1200 Cailun Road, Shanghai 201203, PR China.
| | - Yongjun Wang
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, 725 Wan-Ping South Road, Shanghai 200032, PR China; Spine Institute, Shanghai University of Traditional Chinese Medicine, 725 Wan-Ping South Road, Shanghai 200032, PR China.
| | - Qianqian Liang
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, 725 Wan-Ping South Road, Shanghai 200032, PR China; Spine Institute, Shanghai University of Traditional Chinese Medicine, 725 Wan-Ping South Road, Shanghai 200032, PR China.
| |
Collapse
|
20
|
Dawudi Y, Benarroch S, Helfer H, Smadja DM, Mahé I. Janus kinase inhibitor treatment for inflammatory diseases: excess or no excess risk of venous thromboembolism? Res Pract Thromb Haemost 2025; 9:102667. [PMID: 39980606 PMCID: PMC11840193 DOI: 10.1016/j.rpth.2024.102667] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Revised: 12/04/2024] [Accepted: 12/24/2024] [Indexed: 02/22/2025] Open
Abstract
Janus kinase inhibitors (JAKis) have revolutionized the treatment landscape for various inflammatory and autoimmune diseases since their introduction in 2012. The expanded indications of JAKis have raised concerns about the associated risk of thrombosis, venous thromboembolic events (VTEs), and arterial thrombosis. This literature review examines studies reporting the risk of VTEs associated with JAKis in patients with inflammatory diseases. Phase I to III trials showed no increased risk of VTEs. However, these studies were not designed to detect adverse events such as VTEs. The pharmacovigilance data indicated that the frequency of VTE reports was higher than that of other adverse events. An increased risk of VTEs was also observed in the ORAL Surveillance study, a randomized, noninferiority, postmarketing phase IV safety study comparing tofacitinib with anti-tumor necrosis factor in patients with rheumatoid arthritis. However, limitations have to be acknowledged: pharmacovigilance data are declarative and subject to bias, VTE was a secondary outcome in the ORAL study, with noncomparable VTE risk factors between groups and increased thrombosis risks only at high doses of tofacitinib. Nevertheless, these data have led regulatory organizations such as the Food and Drug Administration and the European Medicines Agency to issue precautionary measures regarding the use of JAKis in inflammatory diseases. Most well-conducted real-life studies are in rheumatoid arthritis and do not confirm an excess of VTE risk associated with JAKis. Considering those conflicting results and limitations, future research should focus on specific indications and patient profiles, taking into account the complex interaction between drug treatment and underlying disease activity, to be able to draw definite conclusion about the VTE risk associated with JAKis.
Collapse
Affiliation(s)
- Yachar Dawudi
- Internal Medicine Department, Hôpital Louis-Mourier, Assistance Publique - Hôpitaux de Paris, Colombes, France
| | - Samuel Benarroch
- Internal Medicine Department, Hôpital Louis-Mourier, Assistance Publique - Hôpitaux de Paris, Colombes, France
| | - Hélène Helfer
- Internal Medicine Department, Hôpital Louis-Mourier, Assistance Publique - Hôpitaux de Paris, Colombes, France
| | - David M. Smadja
- Hematology Department, European Georges Pompidou Hospital, Assistance Publique - Hôpitaux de Paris, Paris, France
- Université Paris Cité, Paris, France
- INSERM Cardiovascular Research Center, Team « Endotheliopathy and Hemostasis Disorders », Paris, France
- Investigation Network On Venous Thrombo-Embolism (INNOVTE) - French Clinical Research Infrastructure Network, Saint-Etienne, France
| | - Isabelle Mahé
- Internal Medicine Department, Hôpital Louis-Mourier, Assistance Publique - Hôpitaux de Paris, Colombes, France
- Université Paris Cité, Paris, France
- INSERM Cardiovascular Research Center, Team « Endotheliopathy and Hemostasis Disorders », Paris, France
- Investigation Network On Venous Thrombo-Embolism (INNOVTE) - French Clinical Research Infrastructure Network, Saint-Etienne, France
| |
Collapse
|
21
|
Priora M, Becciolini A, Celletti E, Di Penta M, Lo Gullo A, Paroli M, Bravi E, Andracco R, Nucera V, Ometto F, Lumetti F, Farina A, Del Medico P, Colina M, Ravagnani V, Scolieri P, Larosa M, Visalli E, Addimanda O, Vitetta R, Volpe A, Bezzi A, Girelli F, Molica Colella AB, Caccavale R, Di Donato E, Adorni G, Santilli D, Lucchini G, Arrigoni E, Sabatini E, Platè I, Mansueto N, Ianniello A, Fusaro E, Ditto MC, Bruzzese V, Camellino D, Bianchi G, Serale F, Foti R, Amato G, De Lucia F, Bosco YD, Foti R, Reta M, Fiorenza A, Rovera G, Marchetta A, Focherini MC, Mascella F, Bernardi S, Sandri G, Giuggioli D, Salvarani C, Franchina V, Molica Colella F, Ferrero G, Ariani A, Parisi S. Effectiveness and Predictors of Long-Term Treatment Response to Tofacitinib in Rheumatoid Arthritis Cohort: General Analysis and Focus on High-Cardiovascular-Risk Subgroup-A Multicenter Study. MEDICINA (KAUNAS, LITHUANIA) 2024; 60:1982. [PMID: 39768862 PMCID: PMC11677110 DOI: 10.3390/medicina60121982] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/28/2024] [Revised: 11/17/2024] [Accepted: 11/25/2024] [Indexed: 01/11/2025]
Abstract
Background and Objectives: The treatment landscape for Rheumatoid Arthritis (RA) has evolved significantly with the introduction of Janus kinase inhibitors (JAKi), such as Tofacitinib (TOFA), which offer a new therapeutic option for patients who have failed or are intolerant to conventional synthetic disease-modifying antirheumatic drugs (csDMARDs). Safety concerns, particularly related to cardiovascular and cancer risks, prompted a need for additional investigation in real-world clinical settings. This study aimed to evaluate the long-term effectiveness and predictors of response to TOFA in two subpopulations of RA patients, categorized by differing cardiovascular risk profiles. Materials and Methods: This was a retrospective, multicenter observational study conducted as part of the BIRRA project, involving 23 Italian rheumatological referral centers. A total of 213 patients diagnosed with RA and treated with TOFA were included, with data collected on baseline demographics, clinical history, disease activity, and comorbidities. Patients were divided into high-risk and low-risk cardiovascular groups based on age (≥65 years) and the presence of at least one cardiovascular risk factor. Disease activity was assessed at baseline, 6 months, and 12 months using DAS28-ESR and DAS28-CRP. Treatment response was evaluated using intention-to-treat (ITT) and per-protocol (PP) approaches. Predictors of low disease activity (LDA) and remission were assessed through logistic regression, and clustering analyses were used to identify subgroups of patients with different therapeutic responses. Results: The study included 213 patients, with 129 classified as high-risk. For the overall cohort, patients achieving LDA and remission at 6 months were 20% and 12%, respectively, for the ITT analysis, and 29% and 14% for the PP analysis. At 12 months, 26% of patients reached LDA, and 17% achieved remission according to ITT, while for the PP analysis, these rates were 30% and 19%, respectively. No significant differences in remission or LDA rates were observed between the high-risk and low-risk groups. In the high-risk subgroup, 17% of patients reached LDA and 9% achieved remission at 6 months (ITT analysis), while these rates increased to 22% and 13%, respectively, in the PP analysis. At 12 months, 22% achieved LDA and 13% achieved remission in the ITT analysis, while 28% and 17% did so in the PP analysis. The reduction in DAS28-ESR and DAS28-CRP scores was significant (p < 0.001) across all time points for both high-risk and low-risk patients. Logistic regression analyses revealed that none of the baseline characteristics-including age, sex, comorbidities, rheumatoid factor, anti-citrullinated protein antibody (ACPA) positivity, initial disease severity, or treatment history-were significant predictors of remission or LDA at 6 or 12 months. The clustering analysis suggested that older patients, particularly those with worse baseline DAS28 scores, tended to show a less favorable response to treatment, potentially indicating impacts of age-related factors such as immunosenescence on therapeutic outcomes. Conclusions: Tofacitinib demonstrated similar effectiveness in both high- and low-risk cardiovascular subgroups of RA patients, with significant reductions in disease activity observed at both 6 and 12 months. Despite safety concerns related to cardiovascular risk, TOFA remained an effective treatment option across patient subgroups, with no significant differences in remission or LDA rates based on cardiovascular risk profiles. Age appeared to negatively impact treatment response, highlighting the role of immunosenescence in RA management. These findings support the use of TOFA as a personalized therapeutic option for RA, emphasizing the need for careful evaluation of cardiovascular and age-related risks in clinical decision-making.
Collapse
Affiliation(s)
- Marta Priora
- Rheumatology Day Hospital and Outpatient Clinic, ASL CN1, 12100 Cuneo, Italy; (M.P.); (F.S.)
| | - Andrea Becciolini
- Internal Medicine and Rheumatology Unit, University Hospital of Parma, 43126 Parma, Italy; (A.B.); (E.D.D.); (G.A.); (D.S.); (G.L.); (A.A.)
| | - Eleonora Celletti
- Rheumatology Unit, Clinica Medica Institute, SS Annunziata Hospital, 66100 Chieti, Italy; (M.D.P.); (E.S.)
| | - Myriam Di Penta
- Rheumatology Unit, Clinica Medica Institute, SS Annunziata Hospital, 66100 Chieti, Italy; (M.D.P.); (E.S.)
| | | | - Marino Paroli
- Department of Clinical, Anesthesiological and Cardiovascular Sciences, Sapienza University of Rome, 00185 Rome, Italy; (M.P.); (R.C.)
| | - Elena Bravi
- Reumathology Unit, Guglielmo da Saliceto Hospital, 29121 Piacenza, Italy; (E.B.); (E.A.); (I.P.)
| | - Romina Andracco
- Rheumatology Unit, Imperia Hospital, 18100 Imperia, Italy; (R.A.); (N.M.)
| | - Valeria Nucera
- Rheumatology Outpatient Unit, ASL Novara, 28100 Novara, Italy; (V.N.); (A.I.)
| | - Francesca Ometto
- Rheumatology Outpatient Clinic, Azienda ULSS 6 Euganea, 35131 Padova, Italy;
| | - Federica Lumetti
- Rheumatology Unit, Azienda USL of Modena and AOU Policlinico of Modena, 41100 Modena, Italy;
| | - Antonella Farina
- Internal Medicine Unit, Rheumatology Outpatient Clinic, Augusto Murri Hospital, 63900 Fermo, Italy;
| | - Patrizia Del Medico
- Rheumatology Outpatient Clinic, Internal Medicine Unit, Civitanova Marche Hospital, 62012 Civitanova Marche, Italy;
| | - Matteo Colina
- Rheumatology Service, Internal Medicine Section, Department of Medicine and Oncology, Santa Maria della Scaletta Hospital, 40026 Imola, Italy;
- Alma Mater Studiorum, Department of Biomedical and Neuromotor Sciences, University of Bologna, 40126 Bologna, Italy
| | - Viviana Ravagnani
- Rheumatology Unit, Santa Chiara Hospital APSS—Trento, 38122 Trento, Italy;
| | - Palma Scolieri
- Rheumatology Unit, Department of Medical Specialties, Nuovo Regina Margherita Hospital, 00154 Roma, Italy; (P.S.); (V.B.)
| | - Maddalena Larosa
- Division of Rheumatology, Department of Medical Specialties, La Colletta Hospital, ASL 3 Genova, 16132 Genova, Italy; (M.L.); (D.C.); (G.B.)
| | - Elisa Visalli
- Rheumatology Unit, Policlinico San Marco Hospital, 95121 Catania, Italy; (E.V.); (R.F.); (G.A.); (F.D.L.); (Y.D.B.); (R.F.)
| | - Olga Addimanda
- Rheumatology Unit, AUSL of Bologna—Policlinico Sant’Orsola—AOU—IRCCS of Bologna, 40138 Bologna, Italy; (O.A.); (M.R.)
| | - Rosetta Vitetta
- Rheumatology Unit, ASL VC Sant’Andrea Hospital, 13100 Vercelli, Italy; (R.V.); (A.F.); (G.R.)
| | - Alessandro Volpe
- Rheumatology Unit, IRCCS Sacro Cuore Don Calabria Hospital, 37024 Negrar di Valpolicella, Italy; (A.V.); (A.M.)
| | - Alessandra Bezzi
- Internal Medicine and Rheumatology Unit, ASL Romagna—Rimini, 47924 Rimini, Italy (M.C.F.); (F.M.)
| | - Francesco Girelli
- Rheumatology Unit, G.B. Morgagni—L. Pierantoni Hospital, 47121 Forli, Italy; (F.G.); (S.B.)
| | | | - Rosalba Caccavale
- Department of Clinical, Anesthesiological and Cardiovascular Sciences, Sapienza University of Rome, 00185 Rome, Italy; (M.P.); (R.C.)
| | - Eleonora Di Donato
- Internal Medicine and Rheumatology Unit, University Hospital of Parma, 43126 Parma, Italy; (A.B.); (E.D.D.); (G.A.); (D.S.); (G.L.); (A.A.)
| | - Giuditta Adorni
- Internal Medicine and Rheumatology Unit, University Hospital of Parma, 43126 Parma, Italy; (A.B.); (E.D.D.); (G.A.); (D.S.); (G.L.); (A.A.)
| | - Daniele Santilli
- Internal Medicine and Rheumatology Unit, University Hospital of Parma, 43126 Parma, Italy; (A.B.); (E.D.D.); (G.A.); (D.S.); (G.L.); (A.A.)
| | - Gianluca Lucchini
- Internal Medicine and Rheumatology Unit, University Hospital of Parma, 43126 Parma, Italy; (A.B.); (E.D.D.); (G.A.); (D.S.); (G.L.); (A.A.)
| | - Eugenio Arrigoni
- Reumathology Unit, Guglielmo da Saliceto Hospital, 29121 Piacenza, Italy; (E.B.); (E.A.); (I.P.)
| | - Emanuela Sabatini
- Rheumatology Unit, Clinica Medica Institute, SS Annunziata Hospital, 66100 Chieti, Italy; (M.D.P.); (E.S.)
| | - Ilaria Platè
- Reumathology Unit, Guglielmo da Saliceto Hospital, 29121 Piacenza, Italy; (E.B.); (E.A.); (I.P.)
| | - Natalia Mansueto
- Rheumatology Unit, Imperia Hospital, 18100 Imperia, Italy; (R.A.); (N.M.)
| | - Aurora Ianniello
- Rheumatology Outpatient Unit, ASL Novara, 28100 Novara, Italy; (V.N.); (A.I.)
| | - Enrico Fusaro
- Rheumatology Department, Azienda Ospedaliero-Universitaria Città della Salute e della Scienza di Torino, 10126 Torino, Italy; (E.F.); (M.C.D.); (S.P.)
| | - Maria Chiara Ditto
- Rheumatology Department, Azienda Ospedaliero-Universitaria Città della Salute e della Scienza di Torino, 10126 Torino, Italy; (E.F.); (M.C.D.); (S.P.)
| | - Vincenzo Bruzzese
- Rheumatology Unit, Department of Medical Specialties, Nuovo Regina Margherita Hospital, 00154 Roma, Italy; (P.S.); (V.B.)
| | - Dario Camellino
- Division of Rheumatology, Department of Medical Specialties, La Colletta Hospital, ASL 3 Genova, 16132 Genova, Italy; (M.L.); (D.C.); (G.B.)
| | - Gerolamo Bianchi
- Division of Rheumatology, Department of Medical Specialties, La Colletta Hospital, ASL 3 Genova, 16132 Genova, Italy; (M.L.); (D.C.); (G.B.)
| | - Francesca Serale
- Rheumatology Day Hospital and Outpatient Clinic, ASL CN1, 12100 Cuneo, Italy; (M.P.); (F.S.)
| | - Rosario Foti
- Rheumatology Unit, Policlinico San Marco Hospital, 95121 Catania, Italy; (E.V.); (R.F.); (G.A.); (F.D.L.); (Y.D.B.); (R.F.)
| | - Giorgio Amato
- Rheumatology Unit, Policlinico San Marco Hospital, 95121 Catania, Italy; (E.V.); (R.F.); (G.A.); (F.D.L.); (Y.D.B.); (R.F.)
| | - Francesco De Lucia
- Rheumatology Unit, Policlinico San Marco Hospital, 95121 Catania, Italy; (E.V.); (R.F.); (G.A.); (F.D.L.); (Y.D.B.); (R.F.)
| | - Ylenia Dal Bosco
- Rheumatology Unit, Policlinico San Marco Hospital, 95121 Catania, Italy; (E.V.); (R.F.); (G.A.); (F.D.L.); (Y.D.B.); (R.F.)
| | - Roberta Foti
- Rheumatology Unit, Policlinico San Marco Hospital, 95121 Catania, Italy; (E.V.); (R.F.); (G.A.); (F.D.L.); (Y.D.B.); (R.F.)
| | - Massimo Reta
- Rheumatology Unit, AUSL of Bologna—Policlinico Sant’Orsola—AOU—IRCCS of Bologna, 40138 Bologna, Italy; (O.A.); (M.R.)
| | - Alessia Fiorenza
- Rheumatology Unit, ASL VC Sant’Andrea Hospital, 13100 Vercelli, Italy; (R.V.); (A.F.); (G.R.)
| | - Guido Rovera
- Rheumatology Unit, ASL VC Sant’Andrea Hospital, 13100 Vercelli, Italy; (R.V.); (A.F.); (G.R.)
| | - Antonio Marchetta
- Rheumatology Unit, IRCCS Sacro Cuore Don Calabria Hospital, 37024 Negrar di Valpolicella, Italy; (A.V.); (A.M.)
| | - Maria Cristina Focherini
- Internal Medicine and Rheumatology Unit, ASL Romagna—Rimini, 47924 Rimini, Italy (M.C.F.); (F.M.)
| | - Fabio Mascella
- Internal Medicine and Rheumatology Unit, ASL Romagna—Rimini, 47924 Rimini, Italy (M.C.F.); (F.M.)
| | - Simone Bernardi
- Rheumatology Unit, G.B. Morgagni—L. Pierantoni Hospital, 47121 Forli, Italy; (F.G.); (S.B.)
| | - Gilda Sandri
- Rheumatology Unit, University of Modena and Reggio Emilia, 41125 Modena, Italy; (G.S.); (D.G.); (C.S.)
| | - Dilia Giuggioli
- Rheumatology Unit, University of Modena and Reggio Emilia, 41125 Modena, Italy; (G.S.); (D.G.); (C.S.)
| | - Carlo Salvarani
- Rheumatology Unit, University of Modena and Reggio Emilia, 41125 Modena, Italy; (G.S.); (D.G.); (C.S.)
| | | | | | - Giulio Ferrero
- Unit of Diagnostic and Interventional Radiology, Santa Corona Hospital, 17027 Pietra Ligure, Italy;
| | - Alarico Ariani
- Internal Medicine and Rheumatology Unit, University Hospital of Parma, 43126 Parma, Italy; (A.B.); (E.D.D.); (G.A.); (D.S.); (G.L.); (A.A.)
| | - Simone Parisi
- Rheumatology Department, Azienda Ospedaliero-Universitaria Città della Salute e della Scienza di Torino, 10126 Torino, Italy; (E.F.); (M.C.D.); (S.P.)
| |
Collapse
|
22
|
Yu X, Duan R, Jiang L, Wang T, Li Z, Zhang B, Su W, Lin Y. Interleukin-6 in non-infectious uveitis: Biology, experimentalevidence and treatment strategies. Biochem Pharmacol 2024; 230:116605. [PMID: 39491564 DOI: 10.1016/j.bcp.2024.116605] [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: 06/22/2024] [Revised: 10/10/2024] [Accepted: 10/31/2024] [Indexed: 11/05/2024]
Abstract
Uveitis is the leading cause of visual impairment worldwide. Interleukin-6 (IL-6), which is upregulated in response to inflammation, is one of the most important inflammatory cytokines associated with uveitis. Two major IL-6 receptors (IL-6R) mediate the pro-inflammatory and anti-inflammatory biological effects of IL-6. This review summarized multiple perspectives on the mechanism of IL-6-mediated uveitis, based on experimental evidence from clinical and animal models. It includes discussions on the roles of the downstream IL-6 signaling pathway, immunocytes, and the blood-retinal barrier. Therapeutic strategies aimed at blocking the action of IL-6 have progressed in clinical practice. However, due to the adverse events associated with existing biologics including infections, drugs that selectively inhibit intraocular IL-6 still require further development. The novel concept of converting the pro-inflammatory effects of IL-6 into protective effects also requires further research. In addition, the relationship between the trans-presentation of IL-6R and T-helper17 cells in uveitis remains unexplored. This review aims to consolidate our current understanding of the biology, signaling pathways, experimental models, and immune pathogenesis related to IL-6 and uveitis. We also discuss clinical strategies focused on blocking IL-6 as a treatment for uveitis. Targeting IL-6 provides unlimited potential for improving the diagnosis, treatment, and prognosis of uveitis.
Collapse
Affiliation(s)
- Xiaoyang Yu
- Guangzhou University of Chinese Medicine, Guangzhou 510060, China
| | - Runping Duan
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou 510060, China
| | - Loujing Jiang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou 510060, China
| | - Tianfu Wang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou 510060, China
| | - Zhaohuai Li
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou 510060, China
| | - Bowen Zhang
- Sun Yat-sen University Zhongshan School of Medicine, Guangzhou 510060, China
| | - Wenru Su
- Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai 200000, China.
| | - Ying Lin
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou 510060, China.
| |
Collapse
|
23
|
Neri B, Mancone R, Fiorillo M, Schiavone SC, Migliozzi S, Biancone L. Efficacy and Safety of Janus Kinase-Inhibitors in Ulcerative Colitis. J Clin Med 2024; 13:7186. [PMID: 39685645 DOI: 10.3390/jcm13237186] [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: 10/24/2024] [Revised: 11/17/2024] [Accepted: 11/19/2024] [Indexed: 12/18/2024] Open
Abstract
Background: Janus kinase-inhibitors (JAK-i) have recently been approved for treating patients with Ulcerative Colitis (UC); therefore, further information is needed, particularly regarding efficacy and safety. Objectives: To provide a comprehensive review regarding the efficacy and safety of currently available JAK-i in UC. Methods: The PubMed and Scopus databases were considered, searching for 'JAK', 'JAK-inhibitor', 'Janus Kinases', 'Tofacitinib', 'Filgotinib', 'Upadacitinib', individually or in combination with 'IBD', 'Ulcerative Colitis', 'safety', 'efficacy', 'study' and 'trial'. The search was focused on full-text papers published in English, with no publication date restrictions. Results: The efficacy and safety of JAK-i approved for treating patients with UC have been summarized. These included Tofacitinib, Filgotinib and Upadacitinib. Findings from both clinical trials and real-life studies in UC were reported, with particular regard to their efficacy in inducing clinical response and remission, steroid-free remission and endoscopic and histological healing. Overall, JAK-i proved to be effective and safe in selected subgroups of patients with UC. The rapid onset of action and the oral route of administration represent the most relevant characteristics of these drugs. Safety concerns using Tofacitinib in subgroups of patients (infections, hypercholesterolemia, venous thromboembolism and cardiovascular events) were initially raised. More recently, all JAK-i for UC showed an overall satisfactory safety profile. However, indication should be carefully given. Conclusions: The use of JAK-i UC is promising, although no predictive markers of response are currently available. Optimizing their use, as monotherapy or combined with other immunomodulators, may increase their efficacy in appropriately selected subgroups of patients with UC.
Collapse
Affiliation(s)
- Benedetto Neri
- Gastroenterology Unit, Department of Systems Medicine, University "Tor Vergata" of Rome, 00133 Rome, Italy
| | - Roberto Mancone
- Gastroenterology Unit, Department of Systems Medicine, University "Tor Vergata" of Rome, 00133 Rome, Italy
| | - Mariasofia Fiorillo
- Gastroenterology Unit, Department of Systems Medicine, University "Tor Vergata" of Rome, 00133 Rome, Italy
| | - Sara Concetta Schiavone
- Gastroenterology Unit, Department of Systems Medicine, University "Tor Vergata" of Rome, 00133 Rome, Italy
| | - Stefano Migliozzi
- Gastroenterology Unit, Department of Systems Medicine, University "Tor Vergata" of Rome, 00133 Rome, Italy
| | - Livia Biancone
- Gastroenterology Unit, Department of Systems Medicine, University "Tor Vergata" of Rome, 00133 Rome, Italy
| |
Collapse
|
24
|
Takanashi S, Ohmura K, Misaki K, Ihata A, Matsui T, Tohma S, Saegusa J, Sato S, Matsubara T, Yamaoka K, Amano K, Miyamoto T, Mori Y, Kaneko Y. Optimal timing of recombinant herpes zoster virus vaccination for a JAK inhibitor treatment in rheumatoid arthritis: a multicentre, open-label, randomised comparative study (STOP-HZ study): study protocol. BMJ Open 2024; 14:e090668. [PMID: 39551586 PMCID: PMC11574477 DOI: 10.1136/bmjopen-2024-090668] [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] [Indexed: 11/19/2024] Open
Abstract
INTRODUCTION Janus kinase (JAK) inhibitors are an important therapeutic option in the treatment of rheumatoid arthritis, but increase the risk of developing herpes zoster. Although a dry recombinant zoster vaccine (RZV) that can be used under immunosuppressive conditions has recently been developed, its optimal use and appropriate timing in patients scheduled to start JAK inhibitors is still unclear. The present study is designed to clarify the appropriate timing of JAK inhibitor initiation to measure varicella zoster virus (VZV)-specific IgG titers and VZV-specific T cell response in patients with rheumatoid arthritis who start tofacitinib at the first RZV vaccination or at the second one. METHODS AND ANALYSIS STOP HZ (Effectiveness and S afe T y O f P rophylactic Recombinant H erpes Z oster Virus Vaccination for Rheumatoid Arthritis Patients with Tofacitinib Treatment) study is a multicentre, open-label, randomised, comparative study in patients with rheumatoid arthritis who are scheduled to start tofacitinib. This study enrols 60 study subjects in 12 sites. Enrolled subjects receive RZV two times on day 1 and week 8 and initiate tofacitinib 5 mg two times a day at the time of their first RZV (day 1, group A) or second RZV (week 8, group B) based on randomisation. The random assignment is performed centrally in a 1:1 ratio. Patients in Group B continue the same treatment until the start of tofacitinib treatment. Primary endpoint is VZV-specific IgG antibody titers at week 12 compared with those at baseline in each group. Secondary endpoints include comparison of VZV-specific IgG antibody between the groups, changes in disease activity of rheumatoid arthritis, VZV-specific T cell response and adverse events. ETHICS AND DISSEMINATION The study has been approved by the Certified Review Board of Keio (No. 2022008), and conforms to the Declaration of Helsinki and good clinical practice guidelines. Written informed consent is obtained from participants prior to enrolment. The results of this study are planned to be submitted for publishment in relevant peer-review journals. TRIAL REGISTRATION NUMBER jRCTs031230329.
Collapse
Affiliation(s)
- Satoshi Takanashi
- Division of Rheumatology, Department of Internal Medicine, Keio University School of Medicine, Shinjuku-ku, Tokyo, Japan
| | - Koichiro Ohmura
- Department of Rheumatology, Kobe City Medical Center General Hospital, Kobe, Hyogo, Japan
| | - Kenta Misaki
- Department of Rheumatology, Kita-Harima Medical Center, Kita-harima, Hyogo, Japan
| | - Atsushi Ihata
- Department of Rheumatology, NHO Yokohama Medical Center, Yokohama, Kanagawa, Japan
| | - Toshihiro Matsui
- Department of Rheumatology Research, Clinical Research Center for Allergy and Rheumatology, National Hospital Organization Sagamihara National Hospital, Sagamihara, Kanagawa, Japan
| | - Shigeto Tohma
- Department of Rheumatology, National Hospital Organization Tokyo National Hospital, Kiyose, Tokyo, Japan
| | - Jun Saegusa
- Department of Rheumatology and Clinical Immunology, Kobe University Graduate School of Medicine, Kobe, Hyogo, Japan
| | - Shinji Sato
- Division of Rheumatology, Department of Internal Medicine, Tokai University School of Medicine, Isehara, Kanagawa, Japan
| | | | - Kunihiro Yamaoka
- Department of Rheumatology and Infectious Diseases, Kitasato University School of Medicine, Sagamihara, Kanagawa, Japan
| | - Koichi Amano
- Department of Rheumatology and Clinical Immunology, Saitama Medical Center, Saitama Medical University, Kawagoe, Saitama, Japan
| | - Toshiaki Miyamoto
- Department of Rheumatology, Seirei Hamamatsu General Hospital, Hamamatsu, Shizuoka, Japan
| | - Yasuko Mori
- Division of Clinical Virology, Center for Infectious Diseases, Kobe University Graduate School of Medicine, Kobe, Hyogo, Japan
| | - Yuko Kaneko
- Division of Rheumatology, Department of Internal Medicine, Keio University School of Medicine, Shinjuku-ku, Tokyo, Japan
| |
Collapse
|
25
|
Xie W, Huang H, Geng Y, Fan Y, Zhang Z. Current practice, trends and attitudes of rheumatologists towards glucocorticoids use for rheumatoid arthritis (GURANTEE): a national cross-sectional survey across China. Rheumatol Int 2024; 44:2473-2482. [PMID: 39261371 DOI: 10.1007/s00296-024-05713-2] [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: 07/02/2024] [Accepted: 08/23/2024] [Indexed: 09/13/2024]
Abstract
INTRODUCTION To investigate current practices, changes, and perceptions of rheumatologists regarding GC use in RA patients. METHODS A cross-sectional survey was conducted using a structured questionnaire between April and August 2023. Rheumatologists from 31 province-level regions of Mainland China were invited to participate. Chi-squared tests were adopted to investigate the differences by sociodemographic characteristics. RESULTS 1,717 rheumatologists from 598 hospitals completed the survey with a response rate of 92%. Up to 60% of participants expressed currently infrequent initiation of GC co-therapy with csDMARDs (hardly ever 7.0%; occasionally 24.6%; sometimes 29.1%), accompanied by a decline of frequency over time reported in 64.2%. Regarding attitudes towards bridging therapy with GC, 604 (35.2%) participants supported this approach, 468 (27.3%) opposed it, and 645 (37.6%) remained inconclusive. Time to GC discontinuation in context of csDMARDs was commonly reported within 6 months in current practice which has been narrowed over time. Reasons for chronic GC use were mostly reported due to suboptimal disease control, followed by the need of RA complications, and pre-existing comorbidities. After failure of GC cessation, majority of respondents (84.4%) would escalate RA therapy (commonly by addition of JAK inhibitors, TNF inhibitors), which usually or often facilitated the GC cessation. The most frequently reported advantages and weaknesses of GC were rapid and strong efficacy, adverse events, respectively. Regarding long-term low-dose GC use for RA, the percentage of respondents who supported, opposed, or depended on the situation were 15.9%, 17.2%, and 66.9%, respectively. CONCLUSIONS The current data demonstrate that GC initiation for RA treatment is not as frequent as before and the awareness of GC discontinuation is growing in current practice. Attitudes towards GC co-therapy with csDMARDs vary considerably and long-term low-dose GC use remain situation dependent.
Collapse
Affiliation(s)
- Wenhui Xie
- Department of Rheumatology and Clinical Immunology, Peking University First Hospital, No.8, Xishiku Street, West District, Beijing, 100034, China
| | - Hong Huang
- Department of Rheumatology and Clinical Immunology, Peking University First Hospital, No.8, Xishiku Street, West District, Beijing, 100034, China
| | - Yan Geng
- Department of Rheumatology and Clinical Immunology, Peking University First Hospital, No.8, Xishiku Street, West District, Beijing, 100034, China
| | - Yong Fan
- Department of Rheumatology and Clinical Immunology, Peking University First Hospital, No.8, Xishiku Street, West District, Beijing, 100034, China
| | - Zhuoli Zhang
- Department of Rheumatology and Clinical Immunology, Peking University First Hospital, No.8, Xishiku Street, West District, Beijing, 100034, China.
| |
Collapse
|
26
|
Fang YF, Chang SH, Kuo CF, See LC. Safety outcomes of tocilizumab and tofacitinib treatment for rheumatoid arthritis: Target trial emulation. Int J Rheum Dis 2024; 27:e15406. [PMID: 39523495 DOI: 10.1111/1756-185x.15406] [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: 05/28/2024] [Revised: 10/04/2024] [Accepted: 10/25/2024] [Indexed: 11/16/2024]
Abstract
OBJECTIVES Biological disease-modifying antirheumatic drugs have been the primary treatment option for moderate to severe rheumatoid arthritis (RA) in Taiwan since 2010. Tocilizumab is an interleukin-6 receptor inhibitor, whereas tofacitinib is a Janus kinase inhibitor. The two medications were indicated to treat RA by direct and indirect inhibition of interleukin-6 cytokine. We compared the safety outcomes of tocilizumab and tofacitinib in patients with RA in real-world clinical settings, following the 7 key components of target trial emulation (TTE). METHODS The data source was the Taiwan National Health Insurance Research Database. Patients with RA between 2010 and 2018 were eligible and assigned to either tocilizumab or tofacitinib based on their first prescription of these two medications. The index date was set as the first prescription date of either study medication. Propensity score stabilized weighting (PSSW) was used to balance the characteristics between the two medication groups. The incidences of safety outcomes were all-cause mortality, cancer, coronary heart disease, stroke, venous thrombosis events, tuberculosis, joint replacement events, and herpes zoster infection. The intention-to-treat (ITT) effect was commenced from the index date until the study outcomes, independently, 3 years, withdrawal, or December 31, 2021, whichever occurred first. For the per-protocol (PP) effect, patients were required to maintain the same medical group during the entire follow-up period. RESULTS A total of 2125 patients with RA who were prescribed tocilizumab (n = 844) or tofacitinib (n = 1281) were included in this study. The mean follow-up duration was 2.78 years in the tocilizumab group and 2.83 years in the tofacitinib group. For ITT, the sample sizes were 721 and 1196 for the tocilizumab and tofacitinib, respectively, after PSSW. A substantially lower incidence rate of herpes zoster infection (per 100 patient-years) was observed in the tocilizumab group (3.67) than in the tofacitinib group (7.61), with a hazard ratio of 0.48 (95%CI =0.37-0.63; p < .0001). All-cause mortality, cancer, coronary heart disease, stroke, total hip replacement, and tuberculosis were similar between the two medication groups. For PP, the sample sizes were 619 and 1085 for the tocilizumab and tofacitinib, respectively, after PSSW. The results of the PP analysis were similar to those of the ITT analysis. CONCLUSIONS Tocilizumab and tofacitinib act along the same inflammatory pathway. A lower herpes zoster incident rate was observed in the tocilizumab group than in the tofacitinib group. The incidence rates of other safety concerns and mortality rates were comparable in both groups of patients with RA treated in real-world settings.
Collapse
Affiliation(s)
- Yao-Fan Fang
- Division of Rheumatology, Allergy and Immunology, Department of Internal Medicine, Chang Gung Memorial Hospital at Linkou, Taoyuan City, Taiwan
| | - Shu-Hao Chang
- Department of Public Health, College of Medicine, Chang Gung University, Taoyuan City, Taiwan
| | - Chang-Fu Kuo
- Division of Rheumatology, Allergy and Immunology, Department of Internal Medicine, Chang Gung Memorial Hospital at Linkou, Taoyuan City, Taiwan
| | - Lai-Chu See
- Division of Rheumatology, Allergy and Immunology, Department of Internal Medicine, Chang Gung Memorial Hospital at Linkou, Taoyuan City, Taiwan
- Department of Public Health, College of Medicine, Chang Gung University, Taoyuan City, Taiwan
- Biostatistics Core Laboratory, Molecular Medicine Research Centre, Chang Gung University, Taoyuan City, Taiwan
| |
Collapse
|
27
|
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
|
28
|
Ray S, Palui R. Immunotherapy in type 1 diabetes: Novel pathway to the future ahead. World J Diabetes 2024; 15:2022-2035. [PMID: 39493558 PMCID: PMC11525730 DOI: 10.4239/wjd.v15.i10.2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2024] [Revised: 07/23/2024] [Accepted: 07/26/2024] [Indexed: 09/26/2024] Open
Abstract
Since the discovery of insulin over 100 years ago, the focus of research in the management of type 1 diabetes (T1D) has centered around glycemic control and management of complications rather than the prevention of autoimmune destruction of pancreatic β cells. Fortunately, in recent years, there has been significant advancement in immune-targeted pharmacotherapy to halt the natural progression of T1D. The immune-targeted intervention aims to alter the underlying pathogenesis of T1D by targeting different aspects of the immune system. The immunotherapy can either antagonize the immune mediators like T cells, B cells or cytokines (antibody-based therapy), or reinduce self-tolerance to pancreatic β cells (antigen-based therapy) or stem-cell treatment. Recently, the US Food and Drug Administration approved the first immunotherapy teplizumab to be used only in stage 2 of T1D. However, the window of opportunity to practically implement this approved molecule in the selected target population is limited. In this Editorial, we briefly discuss the various promising recent developments in the field of immunotherapy research in T1D. However, further studies of these newer therapeutic agents are needed to explore their true potential for prevention or cure of T1D.
Collapse
Affiliation(s)
- Sayantan Ray
- Department of Endocrinology, All India Institute of Medical Sciences, Bhubaneswar 751019, India
| | - Rajan Palui
- Department of Endocrinology, The Mission Hospital, Durgapur 713212, India
| |
Collapse
|
29
|
Buch MH, Mallat Z, Dweck MR, Tarkin JM, O'Regan DP, Ferreira V, Youngstein T, Plein S. Current understanding and management of cardiovascular involvement in rheumatic immune-mediated inflammatory diseases. Nat Rev Rheumatol 2024; 20:614-634. [PMID: 39232242 DOI: 10.1038/s41584-024-01149-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/22/2024] [Indexed: 09/06/2024]
Abstract
Immune-mediated inflammatory diseases (IMIDs) are a spectrum of disorders of overlapping immunopathogenesis, with a prevalence of up to 10% in Western populations and increasing incidence in developing countries. Although targeted treatments have revolutionized the management of rheumatic IMIDs, cardiovascular involvement confers an increased risk of mortality and remains clinically under-recognized. Cardiovascular pathology is diverse across rheumatic IMIDs, ranging from premature atherosclerotic cardiovascular disease (ASCVD) to inflammatory cardiomyopathy, which comprises myocardial microvascular dysfunction, vasculitis, myocarditis and pericarditis, and heart failure. Epidemiological and clinical data imply that rheumatic IMIDs and associated cardiovascular disease share common inflammatory mechanisms. This concept is strengthened by emergent trials that indicate improved cardiovascular outcomes with immune modulators in the general population with ASCVD. However, not all disease-modifying therapies that reduce inflammation in IMIDs such as rheumatoid arthritis demonstrate equally beneficial cardiovascular effects, and the evidence base for treatment of inflammatory cardiomyopathy in patients with rheumatic IMIDs is lacking. Specific diagnostic protocols for the early detection and monitoring of cardiovascular involvement in patients with IMIDs are emerging but are in need of ongoing development. This Review summarizes current concepts on the potentially targetable inflammatory mechanisms of cardiovascular pathology in rheumatic IMIDs and discusses how these concepts can be considered for the diagnosis and management of cardiovascular involvement across rheumatic IMIDs, with an emphasis on the potential of cardiovascular imaging for risk stratification, early detection and prognostication.
Collapse
Affiliation(s)
- Maya H Buch
- Centre for Musculoskeletal Research, Division of Musculoskeletal & Dermatological Sciences, Faculty of Biology, Medicine & Health, University of Manchester, Manchester, UK.
- NIHR Manchester Biomedical Research Centre, Manchester University NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester, UK.
| | - Ziad Mallat
- Section of Cardiorespiratory Medicine, Victor Phillip Dahdaleh Heart & Lung Research Institute, University of Cambridge, Cambridge, UK
| | - Marc R Dweck
- Centre for Cardiovascular Science, Chancellors Building, Little France Crescent, University of Edinburgh, Edinburgh, UK
| | - Jason M Tarkin
- Section of Cardiorespiratory Medicine, Victor Phillip Dahdaleh Heart & Lung Research Institute, University of Cambridge, Cambridge, UK
| | - Declan P O'Regan
- MRC Laboratory of Medical Sciences, Imperial College London, London, UK
| | - Vanessa Ferreira
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Taryn Youngstein
- National Heart & Lung Institute, Imperial College London, London, UK
- Department of Rheumatology, Hammersmith Hospital, Imperial College Healthcare NHS Trust, London, UK
| | - Sven Plein
- Biomedical Imaging Science Department, Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, UK
- School of Biomedical Engineering and Imaging Sciences, Kings College London, London, UK
| |
Collapse
|
30
|
Shen X, Liu X, Guo X, Hou X, Huang H, Feng Z. Systematic review of Janus kinases inhibitors for rheumatoid arthritis: methodology, reporting, and quality of evidence evaluation. Front Pharmacol 2024; 15:1459511. [PMID: 39386036 PMCID: PMC11461343 DOI: 10.3389/fphar.2024.1459511] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Accepted: 09/10/2024] [Indexed: 10/12/2024] Open
Abstract
Objective To evaluate the methodological, reporting and evidence quality of systematic reviews or meta-analyses of Janus kinases (JAK) inhibitors for the treatment of rheumatoid arthritis (RA). Methods Our study systematically retrieved reviews from various databases, spanning from inception to June 2024. Two evaluators independently assessed the methodological, reporting, and evidence quality of each review using the AMSTAR-2 and PRIAMA2020 tools. The evidence quality was evaluated according to GRADE criteria. Six aspects were evaluated: publication year, study type, homogeneity, risk of publication bias, AMSTAR-2 methodology, and PRIAMA2020 reporting quality. Excel 2016 facilitated conversion of scores into radar plots. Results Following stringent selection criteria, a total of 18 relevant studies were identified. The AMSTAR-2 scores ranged from 4 to 13 points, with five studies rated as low quality and the remaining 13 as critically low quality. All studies encompassed populations, interventions, controls, and outcome measures, demonstrating commendable integrity. However, there is room for improvement in study protocol development and registration, comprehensive search strategies, inclusion and exclusion criteria, conflict of interest disclosure, and discussion of heterogeneity. PRIAMA2020 assessments ranged from 14.5 to 21 points, with two studies scoring below 15 points due to increased bias risk from data transformation and sensitivity analysis. Notably, all reviews (100%) adhered to PRIAMA2020 guidelines for certain items but none met all criteria. GRADE evaluation included 446 outcome measures, with 158 of moderate, 156 of low, and 132 of very low quality, indicating JAK inhibitors is effective in improving RA. According to radar chart, the average rank score was 13.13. One study achieved a balanced score across all dimensions, while 11 exceeded the average, five showed significant differences in PRIAMA2020 scores, and four in AMSTAR two scores. Conclusion Despite summarizing the efficacy and safety of JAK inhibitors in treating RA, the included studies exhibited poor methodological and reporting quality, along with low-quality evidence overall. Therefore, caution is warranted among decision-makers regarding the use of JAK inhibitors in RA treatment. Urgent requirements include high-quality, multicenter studies investigating JAK inhibitors for RA. Systematic Review registration https://www.crd.york.ac.uk/PROSPERO, identifier 413415.
Collapse
Affiliation(s)
- Xiaolan Shen
- College of Medicine and Health Sciences, China Three Gorges University, Yichang, China
- The First College of Clinical Medical Sciences, Institute of Rheumatology, China Three Gorges University, Yichang, China
| | - Xiaoman Liu
- College of Medicine and Health Sciences, China Three Gorges University, Yichang, China
- The First College of Clinical Medical Sciences, Institute of Rheumatology, China Three Gorges University, Yichang, China
| | - Xiang Guo
- The Second Clinical Medical School, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xiaoqiang Hou
- The First College of Clinical Medical Sciences, Institute of Rheumatology, China Three Gorges University, Yichang, China
| | - Huiliang Huang
- The Second People’s Hospital of Yichang, The Second Clinical Hospital of Three Gorges University, Yichang, Hubei, China
| | - Zhitao Feng
- College of Medicine and Health Sciences, China Three Gorges University, Yichang, China
- The First College of Clinical Medical Sciences, Institute of Rheumatology, China Three Gorges University, Yichang, China
| |
Collapse
|
31
|
Chang J, Wang G. A retrospective study of efficacy of tofacitinib combined with bDMARDs in the treatment of rheumatoid arthritis patients with inadequate response to bDMARDs. Int J Rheum Dis 2024; 27:e15311. [PMID: 39198040 DOI: 10.1111/1756-185x.15311] [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: 05/10/2024] [Revised: 07/12/2024] [Accepted: 08/15/2024] [Indexed: 09/01/2024]
Abstract
INTRODUCTION Rheumatoid arthritis (RA) is a chronic systemic autoimmune disease characterized by synovial inflammation, joint swelling, and pain involving multiple joints. While biologic disease-modifying antirheumatic drugs (bDMARDs) and targeted synthetic DMARDs (tsDMARDs) are popular treatments for RA, there is limited research on their combined use. This study examined a cohort of RA patients who demonstrated inadequate response to bDMARDs and subsequently initiated combination therapy with tofacitinib and bDMARDs, assessing both the efficacy and safety profile of this therapeutic approach. METHODS In this study, we retrospectively collected the electronic medical records (EMR) of 62 adult patients with RA who were admitted to the Fourth Affiliated Hospital Zhejiang University School of Medicine between August 2018 and December 2022. All patients had received at least one bDMARD treatment for more than 3 months and still exhibited moderate-to-high disease activity. Tofacitinib 5 mg bid was added to their original biological treatment in 28 cases, and other 34 cases switched to another bDMARD or tsDMARD as control group. Treatment was continued for 24 weeks following the initiation of combination therapy. Changes in DAS28-ESR and ACR20, 50, 70 response rates at week 24 were collected and analyzed from baseline, while changes in C-reactive protein (CRP) and erythrocyte sedimentation rate (ESR) at weeks 4, 8, 12, 24 were also collected and analyzed. RESULTS After 24 weeks of treatment, the DAS28-ESR score in combined treatment group decreased significantly from a baseline of 5.26 ± 0.90 (3.87-8.31) to 2.67 ± 0.86 (1.41-5.11), with remission achieved by 19 patients (67.9%) and low disease activity achieved by five patients (17.9%). The DAS28-ESR in the control group exhibited a decrease from 5.20 ± 0.77 (3.87-7.23) at baseline to 3.25 ± 1.29 (1.54-5.69). In all, 13 patients (38.2%) achieved remission, while another 11 patients (32.4%) achieved low disease activity. The ACR20, 50, 70 response rates were 85.71%, 75%, and 39.29% in the combined treatment group, whereas it were 75.0%, 53.57%, 21.43% in the control group. Additionally, both ESR and CRP levels decreased significantly during the course of treatment without any reported adverse events leading to discontinuation. CONCLUSION Our findings offer some evidence, supporting the effectiveness and safety of combining bDMARD with JAKi tofacitinib in RA patients who have an inadequate response to bDMARD monotherapy. This combination effectively manages disease activity while maintaining a relatively low and manageable incidence of adverse events. Further prospective randomized controlled trials with large sample sizes are anticipated to provide evidence-based medical support.
Collapse
Affiliation(s)
- Jie Chang
- Department of Rheumatology, The Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, China
| | - Gang Wang
- Department of Rheumatology, The Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, China
| |
Collapse
|
32
|
Neurath L, Sticherling M, Schett G, Fagni F. Targeting cytokines in psoriatic arthritis. Cytokine Growth Factor Rev 2024; 78:1-13. [PMID: 39068140 DOI: 10.1016/j.cytogfr.2024.06.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Revised: 06/26/2024] [Accepted: 06/27/2024] [Indexed: 07/30/2024]
Abstract
Psoriatic arthritis (PsA) is part of the psoriatic disease spectrum and is characterized by a chronic inflammatory process that affects entheses, tendons and joints. Cytokines produced by immune and non-immune cells play a central role in the pathogenesis of PsA by orchestrating key aspects of the inflammatory response. Pro-inflammatory cytokines such as TNF, IL-23 and IL-17 have been shown to regulate the initiation and progression of PsA, ultimately leading to the destruction of the architecture of the local tissues such as soft tissue, cartilage and bone. The important role of cytokines in PsA has been underscored by the clinical success of antibodies that neutralize their function. In addition to biologic agents targeting individual pro-inflammatory cytokines, signaling inhibitors that block multiple cytokines simultaneously such as JAK inhibitors have been approved for PsA therapy. In this review, we will focus on our current understanding of the role of cytokines in the disease process of PsA and discuss potential new treatment options based on modulation of cytokine function.
Collapse
Affiliation(s)
- Laura Neurath
- Department of Internal Medicine 3, Friedrich-Alexander Universität (FAU) Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany; Deutsches Zentrum Immuntherapie DZI, Friedrich-Alexander Universität (FAU) Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Michael Sticherling
- Deutsches Zentrum Immuntherapie DZI, Friedrich-Alexander Universität (FAU) Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany; Department of Dermatology, Friedrich-Alexander Universität (FAU) Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Georg Schett
- Department of Internal Medicine 3, Friedrich-Alexander Universität (FAU) Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany; Deutsches Zentrum Immuntherapie DZI, Friedrich-Alexander Universität (FAU) Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Filippo Fagni
- Department of Internal Medicine 3, Friedrich-Alexander Universität (FAU) Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany; Deutsches Zentrum Immuntherapie DZI, Friedrich-Alexander Universität (FAU) Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany.
| |
Collapse
|
33
|
Tang B, Xie X, Lu J, Huang W, Yang J, Tian J, Lei L. Designing biomaterials for the treatment of autoimmune diseases. APPLIED MATERIALS TODAY 2024; 39:102278. [DOI: 10.1016/j.apmt.2024.102278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/04/2024]
|
34
|
Saito K, Yoshida S, Ebina H, Miyata M, Suzuki E, Kanno T, Sumichika Y, Matsumoto H, Temmoku J, Fujita Y, Matsuoka N, Asano T, Sato S, Migita K. Real-world comparative study of drug retention of Janus kinase inhibitors in patients with rheumatoid arthritis. PLoS One 2024; 19:e0306714. [PMID: 38990897 PMCID: PMC11239012 DOI: 10.1371/journal.pone.0306714] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Accepted: 06/22/2024] [Indexed: 07/13/2024] Open
Abstract
BACKGROUND Janus kinase (JAK) inhibitors (JAKis) are effective therapeutic agents against rheumatoid arthritis (RA). However, patients having RA with particular risk factors may have a higher incidence of adverse effects (AEs), including major cardiovascular events (MACE) and infections. In this multicenter cohort study, we aimed to clarify the risk factors affecting the drug retention of JAKis in patients with RA. METHODS We retrospectively evaluated patients with RA who received their first JAKi (tofacitinib, baricitinib, upadacitinib, or filgotinib) at our institute. The clinical outcomes, including AEs, were recorded, particularly MACE and serious infections. The drug retention rates were analyzed using the Kaplan-Meier method, and risk factors affecting drug retention rates were determined using a multivariable Cox regression hazards model. RESULTS Overall 184 patients with RA receiving their first use of baricitinib (57.6%), tofacitinib (23.9%), upadacitinib (12.0%), or filgotinib (6.5%) were included in this study. Fifty-six (30.4%) patients discontinued JAKi treatment owing to ineffectiveness (9.2%) or AEs, including infections (21.2%). The overall drug retention rates were significantly lower in patients treated with pan-JAKi than in those treated with JAK1 inhibitors (p = 0.03). In the Cox regression model, the presence of baseline high RA disease activity, use of glucocorticoid and treatments with pan-JAKis were associated with reduced drug retention rates of JAKis (p < 0.001, p = 0.01 and 0.04, respectively). Pan-JAKi treated patients with high disease activity had significantly lower drug retention rates (p < 0.001). CONCLUSIONS In a real-world setting, the drug retention rates of JAKis were reduced mainly by treatment discontinuation owing to AEs. Treatment with pan-JAKis and high baseline RA disease activity were identified as predictive factors for the discontinuation of JAKis. Lower drug retention rates were found in patients receiving pan-JAKis with high disease activity than in those without high disease activity.
Collapse
Affiliation(s)
- Kenji Saito
- Department of Rheumatology, Fukushima Medical University School of Medicine, Fukushima, Japan
| | - Shuhei Yoshida
- Department of Rheumatology, Fukushima Medical University School of Medicine, Fukushima, Japan
| | - Honoka Ebina
- Department of Rheumatology, Fukushima Medical University School of Medicine, Fukushima, Japan
| | - Masayuki Miyata
- Department of Rheumatology, Japanese Red Cross Fukushima Hospital, Fukushima, Japan
| | - Eiji Suzuki
- Department of Rheumatology, Ohta Nishinouchi General Hospital Foundation, Koriyama, Fukushima, Japan
| | - Takashi Kanno
- Department of Rheumatology, Ohta Nishinouchi General Hospital Foundation, Koriyama, Fukushima, Japan
| | - Yuya Sumichika
- Department of Rheumatology, Fukushima Medical University School of Medicine, Fukushima, Japan
| | - Haruki Matsumoto
- Department of Rheumatology, Fukushima Medical University School of Medicine, Fukushima, Japan
| | - Jumpei Temmoku
- Department of Rheumatology, Fukushima Medical University School of Medicine, Fukushima, Japan
| | - Yuya Fujita
- Department of Rheumatology, Fukushima Medical University School of Medicine, Fukushima, Japan
| | - Naoki Matsuoka
- Department of Rheumatology, Fukushima Medical University School of Medicine, Fukushima, Japan
| | - Tomoyuki Asano
- Department of Rheumatology, Fukushima Medical University School of Medicine, Fukushima, Japan
| | - Shuzo Sato
- Department of Rheumatology, Fukushima Medical University School of Medicine, Fukushima, Japan
| | - Kiyoshi Migita
- Department of Rheumatology, Fukushima Medical University School of Medicine, Fukushima, Japan
- Department of Rheumatology, St. Francis Hospital, Nagasaki, Japan
| |
Collapse
|
35
|
Garweg JG, Straessle KA. Janus Kinase Inhibitors as a Third-Line Therapy for Refractory Endogenous Noninfectious Uveitis. Ocul Immunol Inflamm 2024:1-8. [PMID: 38709218 DOI: 10.1080/09273948.2024.2348125] [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: 10/26/2023] [Accepted: 04/23/2024] [Indexed: 05/07/2024]
Abstract
PURPOSE Janus kinase (JAK) inhibitors have recently been used to treat patients with biologic refractory noninfectious uveitis (NIU). This narrative review updates the current evidence relevant for their application in patients with refractory NIU. METHODS A literature search was performed for articles published until October 2023 in the PubMed, Scopus, and CENTRAL databases using the key terms "noninfectious uveitis" and "Janus kinase inhibitor" or "JAK inhibitor" without any exclusion criteria. Published articles were selected based on their clinical focus, relevance for ocular disease, time since publication and study design reflecting their scientific soundness with a critical appraisal of drug safety aspects. RESULTS Janus kinases are transmembrane signaling proteins. Their inhibition has shown therapeutic potential experimentally and in patients with multiple immune-mediated diseases, including NIU. JAK inhibitors differ from biological agents in that they inhibit not one specific but multiple cytokines. These agents can be ingested orally and seem superior to adalimumab for most indications. While there is no doubt regarding their efficacy in treating immune-mediated inflammatory diseases, reports regarding their safety are increasing, and the findings are generally confusing and contradictory. Since substantiated information about their specific safety profiles in patients with inflammatory eye disease is lacking, their position in the therapeutic algorithm for uveitis has yet to be determined. CONCLUSIONS In the absence of evidence from controlled clinical trials, JAK inhibitor therapy is still rendered experimental and currently considered only for sight-threatening uveitis. JAK inhibitors may be considered for specific NIU entities for which there is insufficient response or secondary loss of response to conventional or biologic disease-modifying drugs.
Collapse
Affiliation(s)
- Justus G Garweg
- Swiss Eye Institute and Clinic for Vitreoretinal Diseases, Berner Augenklinik, Bern, Switzerland
- Department of Ophthalmology, Inselspital, Medical Faculty, University of Bern, Bern, Switzerland
| | - Kim A Straessle
- Swiss Eye Institute and Clinic for Vitreoretinal Diseases, Berner Augenklinik, Bern, Switzerland
| |
Collapse
|
36
|
Cai W, Tong R, Sun Y, Yao Y, Zhang J. Comparative efficacy of five approved Janus kinase inhibitors as monotherapy and combination therapy in patients with moderate-to-severe active rheumatoid arthritis: a systematic review and network meta-analysis of randomized controlled trials. Front Pharmacol 2024; 15:1387585. [PMID: 38725657 PMCID: PMC11080655 DOI: 10.3389/fphar.2024.1387585] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2024] [Accepted: 04/08/2024] [Indexed: 05/12/2024] Open
Abstract
Background The European League of Rheumatology(EULAR)guidelines recommend Janus kinase (JAK) inhibitors for patients with moderate to severe rheumatoid arthritis (RA) who are insensitive or under-responsive to conventional synthetic disease-modifying anti-rheumatic drugs (csDMARDs). But there was no recommendation for which one was preferred in five currently approved JAK inhibitors. The objective of this network meta-analysis study was to evaluate the efficacy of five JAK inhibitors as monotherapy and combination therapy in patients with moderate-to-severe active rheumatoid arthritis. Methods The randomized controlled trials (RCTs) of tofacitinib, baricitinib, upadacitinib, filgotinib and peficitinib as monotherapy or combined with csDMARD in the treatment of active RA were searched in database of PubMed, Embase, Web of Science and Cochrane Library, up to December 2023. The control group included placebo or csDMARD. Outcome indicators included American College of Rheumatology 20% response (ACR20), ACR50, ACR70 and the percentage of patients achieving 28-joint disease activity score using C-reactive protein (DAS28(CRP))<2.6 at 12 weeks and 24 weeks. The statistical analysis was performed by Stata14 and RevMan5.4. Data processing, network evidence plots, surface under the cumulative ranking curve (SUCRA) ranking, league plots and funnel plots were generated. Risk ratio (RR) and 95% confidence interval (95%CI) as effect sizes to analyze the statistics. Results This study included thirty-six RCTs with 16,713 patients. All JAK inhibitors were more effective than placebo in ACR20 (RRs ranging between 1.74 and 3.08), ACR50 (RRs ranging between 2.02 and 7.47), ACR70 (RRs ranging between 2.68 and 18.13), DAS28(CRP) < 2.6 (RRs ranging between 2.70 and 7.09) at 12 weeks. Upadacitinib 30 mg and upadacitinib 15 mg showed relatively good efficacy according to their relative SUCRA ranking. All JAK inhibitors were more effective than csDMARD or placebo in ACR20 (RRs ranging between 1.16 and 1.86), ACR50 (RRs ranging between 1.69 and 2.84), ACR70 (RRs ranging between 1.50 and 4.47), DAS28(CRP) < 2.6 (RRs ranging between 2.28 and 7.56) at 24 weeks. Upadacitinib 15 mg + csDMARD and baricitinib 4 mg + csDMARD showed relatively good efficacy according to their relative SUCRA ranking. The safety analysis results such as serious infection, malignancy, major adverse cardiovascular event (MACE), and venous thromboembolic events (VTE) showed no statistical difference. Conclusion This NMA study indicated that all JAK inhibitors performed better than placebo. Based on the results of this study, upadacitinib 30 mg, upadacitinib 15 mg, upadacitinib 15 mg + csDMARD and baricitinib 4 mg + csDMARD were recommended treatment options with relatively good efficacy and safety. However, attention should be paid to monitoring the occurrence of adverse events in high-risk RA patients with medication. Combination therapy with csDMARD might be more suitable for the maintenance of long-term efficacy. However, in clinical practice, it is still necessary to select the appropriate therapeutic regimen based on the actual clinical situation.
Collapse
Affiliation(s)
- Wenting Cai
- Department of Pharmacy, Nanjing Drum Tower Hospital, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
- Department of Pharmacy, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Rui Tong
- Department of Pharmacy, Nanjing Drum Tower Hospital, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
- Department of Pharmacy, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Yue Sun
- Department of Pharmacy, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Yao Yao
- Department of Pharmacy, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Jinping Zhang
- Department of Pharmacy, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| |
Collapse
|
37
|
Anyfanti P, Angeloudi E, Dara A, Pagkopoulou E, Moysidou GS, Deuteraiou K, Boutel M, Bekiari E, Doumas M, Kitas GD, Dimitroulas T. Non-Invasive Assessment of Micro- and Macrovascular Function after Initiation of JAK Inhibitors in Patients with Rheumatoid Arthritis. Diagnostics (Basel) 2024; 14:834. [PMID: 38667479 PMCID: PMC11048865 DOI: 10.3390/diagnostics14080834] [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: 02/27/2024] [Revised: 04/13/2024] [Accepted: 04/17/2024] [Indexed: 04/28/2024] Open
Abstract
BACKGROUND Janus kinase (JAK) inhibitors constitute a novel class of oral biologic disease-modifying antirheumatic drugs for patients with rheumatoid arthritis (RA). However, their use has been associated with increased risk of major cardiovascular events. We investigated whether treatment with JAK inhibitors exerts significant alterations in the micro- and microvasculature in RA patients. METHODS Thirteen patients with RA initiating treatment with JAK inhibitors were prospectively studied. Eventually, data from 11 patients who completed the study were analyzed. Procedures were performed at baseline and 3 months after treatment. Nailfold videocapillaroscopy was applied to detect alterations of the dermal capillary network. Participants underwent 24 h ambulatory blood pressure monitoring (Mobil-O-Graph device) for the assessment of blood pressure (both brachial and aortic) and markers of large artery stiffening [pulse wave velocity (PWV), augmentation index] throughout the whole 24 h and the respective day- and nighttime periods. Carotid intima-media thickness was assessed with ultrasound. RESULTS Three-month treatment with JAK inhibitors was not associated with any differences in brachial and aortic blood pressure, arterial stiffness, and carotid atherosclerosis, with the only exception of nighttime PWV, which was significantly elevated at follow-up. However, three-month treatment with JAK inhibitors induced significant microvascular alterations and increased the total number of capillaroscopic abnormalities. CONCLUSIONS Three-month treatment with JAK inhibitors may exert significant effects on microcirculation as assessed with nailfold videocapillaroscopy, whereas macrovascular structure and function appears largely unaffected. Further research toward this direction may add substantial information to the available literature regarding cardiovascular aspects of JAK inhibitors in RA.
Collapse
Affiliation(s)
- Panagiota Anyfanti
- Second Medical Department, Hippokration Hospital, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece; (P.A.); (E.A.); (E.B.)
| | - Elena Angeloudi
- Second Medical Department, Hippokration Hospital, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece; (P.A.); (E.A.); (E.B.)
| | - Athanasia Dara
- Fourth Department of Internal Medicine, Hippokration Hospital, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece; (A.D.); (E.P.); (K.D.); (M.B.)
| | - Eleni Pagkopoulou
- Fourth Department of Internal Medicine, Hippokration Hospital, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece; (A.D.); (E.P.); (K.D.); (M.B.)
| | - Georgia-Savina Moysidou
- Rheumatology-Clinical Immunology Unit, 4th Department of Internal Medicine, Attikon University Hospital, National and Kapodistrian University of Athens Medical School, 11527 Athens, Greece;
| | - Kleopatra Deuteraiou
- Fourth Department of Internal Medicine, Hippokration Hospital, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece; (A.D.); (E.P.); (K.D.); (M.B.)
| | - Maria Boutel
- Fourth Department of Internal Medicine, Hippokration Hospital, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece; (A.D.); (E.P.); (K.D.); (M.B.)
| | - Eleni Bekiari
- Second Medical Department, Hippokration Hospital, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece; (P.A.); (E.A.); (E.B.)
| | - Michael Doumas
- 2nd Propedeutic Department of Internal Medicine, Hippokration Hospital, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece;
| | - George D. Kitas
- Department of Rheumatology, Russells Hall Hospital, Dudley Group NHS Foundation Trust, Dudley DY1 2HQ, UK;
- School of Sport, Exercise and Rehabilitation Sciences, University of Birmingham, Birmingham B15 2TT, UK
| | - Theodoros Dimitroulas
- Fourth Department of Internal Medicine, Hippokration Hospital, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece; (A.D.); (E.P.); (K.D.); (M.B.)
| |
Collapse
|
38
|
Affiliation(s)
- Francesco Ciccia
- University della Campania, Department of Precision Medicine, Naples, Italy.
| | | |
Collapse
|