1
|
Elman SA, Perez-Chada LM, Armstrong A, Gottlieb AB, Merola JF. Psoriatic arthritis: A comprehensive review for the dermatologist-Part II: Screening and management. J Am Acad Dermatol 2025; 92:985-998. [PMID: 38857766 DOI: 10.1016/j.jaad.2024.03.059] [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: 10/13/2023] [Revised: 02/16/2024] [Accepted: 03/08/2024] [Indexed: 06/12/2024]
Abstract
Psoriatic arthritis (PsA) is a common comorbidity of psoriasis occurring in up to one-third of patients. Dermatologists hold an essential role in screening patients with psoriasis for PsA, since as many as 85% of patients develop psoriasis before PsA. Early detection and treatment of PsA are important for both short- and long-term patient outcomes and quality of life. Many factors must be weighed when selecting the appropriate therapy for PsA. One must consider the 'domains of disease' that are manifested, the disease severity, patient comorbidities, patient preferences (routes of dosing or frequency, as examples) as well as factors often outside of patient-physician control, such as access to medications based on insurance coverage and formularies. As many patients will have involvement of multiple domains of psoriatic disease, selecting the therapy that best captures the patient's disease is required. In this review, we will address PsA screening, diagnosis, therapeutic approach to psoriatic disease, comorbidity considerations, and comanagement.
Collapse
Affiliation(s)
- Scott A Elman
- Dr. Phillip Frost Department of Dermatology & Cutaneous Surgery, Leonard M. Miller School of Medicine, University of Miami, Miami, Florida
| | - Lourdes M Perez-Chada
- Department of Dermatology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - April Armstrong
- Department of Dermatology, University of California, Los Angeles, Los Angeles, California
| | - Alice B Gottlieb
- Department of Dermatology, Icahn School of Medicine at Mt. Sinai, New York, New York
| | - Joseph F Merola
- Division of Rheumatology, Departments of Dermatology and Medicine, O'Donnell School of Public Health, UT Southwestern Medical Center, Dallas, Texas.
| |
Collapse
|
2
|
Iorizzo M, Sechi A, Neubauer ZJK, Zhou M, Lipner SR. JAK Inhibitors and Inflammatory Nail Disorders: A Systematic Review of Clinical Outcomes and Therapeutic Potential. Am J Clin Dermatol 2025:10.1007/s40257-025-00946-8. [PMID: 40268819 DOI: 10.1007/s40257-025-00946-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/31/2025] [Indexed: 04/25/2025]
Abstract
BACKGROUND Inflammatory nail disorders can have a significant impact on patients' quality of life owing to aesthetic and functional concerns. They are also challenging to treat because the therapeutic armamentarium is quite limited. This systematic review aims to report the efficacy and safety of Janus kinase and Tyrosine kinase 2 inhibitors in treating these conditions. METHODS We conducted a comprehensive search on PubMed, Cochrane, and Embase Library to find eligible case reports, case series, single-arm clinical trials, and randomized controlled trials. We used the following search terms from inception until 15 December, 2024: "nail" AND "jak inhibitors" OR "tofacitinib" OR "baricitinib" OR "abrocitinib" OR "ruxolitinib" OR "deuruxolitinib" OR "upadacitinib" OR "ritlecitinib" OR "deucravacitinib" (nine searches in total). RESULTS Of 441 articles found, 31 were included in this study. The most extensively studied drug was tofacitinib, followed by baricitinib, deucravacitinib, upadacitinib, and abrocitinib. Janus kinase/Tyrosine kinase 2 inhibitors demonstrated improvements in inflammatory nail conditions, with generally mild adverse events (nasopharyngitis and transient laboratory abnormalities being most common). The topical formulation of tofacitinib, the only one studied in these nail diseases, also demonstrated promising results with minimal systemic absorption and no side effects. CONCLUSIONS This review highlights Janus kinase/Tyrosine kinase 2 inhibitors as a valuable addition to the therapeutic arsenal for inflammatory nail disorders while emphasizing the importance of safety assessments and tailored treatment approaches. The long-term safety of Janus kinase/Tyrosine kinase 2 inhibitors still needs further investigation and the potential for adverse events emphasizes the need for tailored therapeutic strategies, including more studies on topical formulations.
Collapse
Affiliation(s)
- Matilde Iorizzo
- Private Dermatology Practice, Lugano/Bellinzona, Switzerland
| | - Andrea Sechi
- Dermatology Unit, Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, Milan, Italy.
| | - Zachary J K Neubauer
- Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, USA
| | - Maggie Zhou
- Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, USA
| | - Shari R Lipner
- Department of Dermatology, Weill Cornell Medicine, New York, NY, USA
| |
Collapse
|
3
|
Tsiogkas SG, Gkouvi A, Kontouli KM, Simopoulou T, Grammatikopoulou MG, Bogdanos DP. Janus kinase inhibitors for psoriatic arthritis: Evidence from a systematic review and network meta-analysis. Autoimmun Rev 2025:103819. [PMID: 40268128 DOI: 10.1016/j.autrev.2025.103819] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2025] [Revised: 04/16/2025] [Accepted: 04/18/2025] [Indexed: 04/25/2025]
Abstract
Psoriatic arthritis (PsA) constitutes a heterogeneous disease. Diagnosis is commonly made by identifying joint inflammation, dactylitis, enthesitis, or axial spine involvement in cutaneous or nail psoriasis. Janus kinases (JAKs) are intracellular kinases that mediate cytokine signaling. The present network meta-analysis aimed to provide a comprehensive summary regarding the use of JAK inhibitors (JAKis) in PsA. We systematically searched MEDLINE, CENTRAL, Web of Science databases, and the clinicaltrials.gov registry until October 2023 and included randomized controlled trials (RCTs). Examined outcomes consisted of the proportion of participants achieving ACR20, ACR50, ACR70, PASI75, resolution of enthesitis, resolution of dactylitis, and experiencing serious adverse events (SAEs). Changes in the Health Assessment Questionnaire Disability Index (HAQ-DI) from the baseline were also recorded. The revised Cochrane Risk of Bias 2.0 tool determined the risk of bias. Networks were constructed, and random-effects frequentist analyses were employed utilizing the placebo arms from each study. Most JAKis were superior to placebo for all examined outcomes. Statistical advantages of one JAKi over another were recorded, and all concerned upadacitinib. However, the safety profile of upadacitinib did not differ significantly from that of other JAKis. Our network meta-analysis is the first to concentrate on comparable, current outcome data of JAKis in PsA on clinical efficacy. It reveals statistically significant variations in the advantages and disadvantages among JAKis in PsA, which require further meticulous assessment.
Collapse
Affiliation(s)
- Sotirios G Tsiogkas
- Department of Rheumatology and Clinical Immunology, Faculty of Medicine, University of Thessaly, Larissa, Greece
| | - Arriana Gkouvi
- Department of Rheumatology and Clinical Immunology, Faculty of Medicine, University of Thessaly, Larissa, Greece
| | - Katerina Maria Kontouli
- Laboratory of Hygiene and Epidemiology, Medical School, University of Thessaly, Larissa, Greece; Department of Primary Education, School of Education University of Ioannina, Ioannina, Greece
| | - Theodora Simopoulou
- Department of Rheumatology and Clinical Immunology, Faculty of Medicine, University of Thessaly, Larissa, Greece
| | - Maria G Grammatikopoulou
- Department of Rheumatology and Clinical Immunology, Faculty of Medicine, University of Thessaly, Larissa, Greece
| | - Dimitrios P Bogdanos
- Department of Rheumatology and Clinical Immunology, Faculty of Medicine, University of Thessaly, Larissa, Greece.
| |
Collapse
|
4
|
Wan H, Zhou X, Su J, Xia T, Zhang D. Comparative Efficacy and Safety of Different Regimens of Current JAK Inhibitors in Psoriatic Arthritis: A Network Meta-analysis. J Clin Rheumatol 2025:00124743-990000000-00336. [PMID: 40184480 DOI: 10.1097/rhu.0000000000002232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/06/2025]
Abstract
BACKGROUND Janus kinase (JAK) inhibitors have been approved for treating psoriatic arthritis (PsA); however, the comparative efficacy of different JAK inhibitors remains unclear. This study aimed to investigate the comparative efficacy and safety of different JAK inhibitors in treating PsA. METHODS This network meta-analysis was conducted in strict accordance with the Preferred Reporting Items for Systematic reviews and Meta-Analyses for Network Meta-Analyses and Cochrane methods. RESULTS Five studies involving 2757 patients were included. Pairwise meta-analysis revealed that JAK inhibitors significantly increased the American College of Rheumatology 20 score and Psoriasis Area and Severity Index 75 responses, which were confirmed by the network meta-analysis. The network meta-analysis further suggested that filgotinib 200 mg once daily (OD) (odds ratio [OR] = 3.17, 95% credible interval [CrI] = 1.07-9.88) and upadacitinib 30 mg OD (OR = 2.34, 95% CrI = 1.13-4.78) had higher American College of Rheumatology 20 score responses compared with tofacitinib 5 mg twice a day. However, upadacitinib 30 mg OD was associated with a higher risk of adverse events (placebo: OR = 1.80, 95% CrI = 1.14-2.87) and serious adverse events compared with filgotinib 200 mg OD (OR = 0.05, 95% CrI = 0.00-0.82). Upadacitinib 15 mg OD, the currently recommended therapy, is comparable in both efficacy and safety to other treatment regimens. CONCLUSIONS Filgotinib 200 mg OD is the safest and most effective JAK inhibitor for PsA, followed by upadacitinib 30 mg OD. However, upadacitinib 30 mg OD carries the highest risk of adverse events. Upadacitinib 15 mg OD, the currently recommended therapy, is not superior in efficacy and safety compared with other treatment options. More high-quality studies are needed to confirm these findings due to the limited number of included studies.
Collapse
Affiliation(s)
- Huiying Wan
- From the Department of Dermatology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China
| | - Xiyuan Zhou
- From the Department of Dermatology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China
| | - Jia Su
- From the Department of Dermatology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China
| | - Tian Xia
- Department of Pathology, Western Theater Command Air Force Hospital
| | - Dingding Zhang
- Sichuan Provincial Key Laboratory for Genetic Disease, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| |
Collapse
|
5
|
Konen FF, Möhn N, Witte T, Schefzyk M, Wiestler M, Lovric S, Hufendiek K, Jendretzky KF, Gingele S, Schwenkenbecher P, Sühs KW, Friese MA, Klotz L, Pul R, Pawlitzki M, Hagin D, Kleinschnitz C, Meuth SG, Skripuletz T. Disease-modifying strategies: Targeting protein kinases in multiple sclerosis and other autoimmune disorders. Autoimmun Rev 2025; 24:103754. [PMID: 39842533 DOI: 10.1016/j.autrev.2025.103754] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Revised: 01/04/2025] [Accepted: 01/15/2025] [Indexed: 01/24/2025]
Abstract
A wide variety of immunomodulatory therapies are already available for the treatment of multiple sclerosis (MS). Through fundamental insights from basic research with a gain of knowledge in the pathological processes underlying MS, the exploration of additional medical compounds within clinical trials has been ignited. Emerging novel medications with innovative mechanisms of action are being introduced. Those mechanisms of action include a broad therapeutic spectrum of substances targeting various protein kinases, some of which could also be used for the treatment of other autoimmune-mediated diseases. The advancement of new compounds could therefore enable a more personalized approach in treating MS, taking into consideration patients' co-existing autoimmune-mediated diseases. In this review, we discuss potential compounds targeting protein kinases, currently under investigation in clinical trials for various autoimmune diseases that could become viable treatment options for MS and comorbid autoimmune conditions in the future.
Collapse
Affiliation(s)
- Franz Felix Konen
- Department of Neurology, Hannover Medical School, 30625 Hannover, Germany.
| | - Nora Möhn
- Department of Neurology, Hannover Medical School, 30625 Hannover, Germany.
| | - Torsten Witte
- Department of Rheumatology and Clinical Immunology, Hannover Medical School, 30625 Hannover, Germany.
| | - Matthias Schefzyk
- Department of Dermatology, Allergology und Venerology, Hannover Medical School, 30625 Hannover, Germany.
| | - Miriam Wiestler
- Department of Gastroenterology, Hepatology, Infectious Diseases and Endocrinology, Hannover Medical School, 30625 Hannover, Germany.
| | - Svjetlana Lovric
- Department of Nephrology and Hypertension, Hannover Medical School, 30625 Hannover, Germany.
| | - Karsten Hufendiek
- University Eye Hospital, Hannover Medical School, 30625 Hannover, Germany.
| | | | - Stefan Gingele
- Department of Neurology, Hannover Medical School, 30625 Hannover, Germany.
| | | | - Kurt-Wolfram Sühs
- Department of Neurology, Hannover Medical School, 30625 Hannover, Germany.
| | - Manuel A Friese
- Institute of Neuroimmunology and Multiple Sclerosis, Center for Molecular Neurobiology Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg 20251, Germany.
| | - Luisa Klotz
- Department of Neurology with Institute of Translational Neurology, University Hospital Muenster, 48149, Muenster, Germany.
| | - Refik Pul
- Department of Neurology, University Medicine Essen, Essen, Germany; Center for Translational Neuro- and Behavioral Sciences, University Hospital Essen, 45147 Essen, Germany.
| | - Marc Pawlitzki
- Department of Neurology, Medical Faculty, Heinrich Heine University Dusseldorf, 40225 Dusseldorf, Germany.
| | - David Hagin
- Allergy and Clinical Immunology Unit, Department of Medicine, Tel-Aviv Sourasky Medical Center and Sackler Faculty of Medicine, University of Tel Aviv, 6 Weizmann St., Tel-Aviv 6423906, Israel.
| | - Christoph Kleinschnitz
- Department of Neurology, University Medicine Essen, Essen, Germany; Center for Translational Neuro- and Behavioral Sciences, University Hospital Essen, 45147 Essen, Germany.
| | - Sven G Meuth
- Department of Neurology, Medical Faculty, Heinrich Heine University Dusseldorf, 40225 Dusseldorf, Germany.
| | - Thomas Skripuletz
- Department of Neurology, Hannover Medical School, 30625 Hannover, Germany.
| |
Collapse
|
6
|
Li W, Dong P, Zhang G, Hu J, Yang S. Emerging Therapeutic Innovations for Vitiligo Treatment. Curr Issues Mol Biol 2025; 47:191. [PMID: 40136446 PMCID: PMC11940846 DOI: 10.3390/cimb47030191] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2025] [Revised: 03/11/2025] [Accepted: 03/13/2025] [Indexed: 03/27/2025] Open
Abstract
Vitiligo is a chronic autoimmune disorder with a multifactorial etiology, typically manifesting as localized or generalized hypopigmentation or depigmentation of the skin and mucous membranes. The pathogenesis of vitiligo is complex and significantly impacts patients' quality of life. Although traditional treatments such as hormone therapy, topical medications, and laser therapy can help control the disease to some extent, their outcomes remain unsatisfactory. Therefore, ongoing research is crucial to explore and develop novel treatment strategies while assessing their efficacy and safety. This review aims to classify and summarize various new candidate drugs for vitiligo currently undergoing clinical trials, providing a reference for clinical practice. Recent advancements in the understanding of the pathogenesis of vitiligo have facilitated the development of potential treatment strategies, such as Janus kinase inhibitors, cytokine blockers, and agents targeting tissue-resident memory or regulatory T cells. These emerging therapies offer hope to patients with vitiligo, though further investigation is needed to confirm their safety, efficacy, and optimal treatment regimens.
Collapse
Affiliation(s)
- Weiran Li
- Institute of Dermatology and Department of Dermatology, The First Affiliated Hospital, Anhui Medical University, Hefei 230000, China; (P.D.); (G.Z.); (J.H.)
- Department of Dermatology, Dushu Lake Hospital Affiliated to Soochow University, Suzhou 215128, China
- Key Laboratory of Dermatology, Ministry of Education of the People’s Republic of China, Hefei 230000, China
| | - Penghao Dong
- Institute of Dermatology and Department of Dermatology, The First Affiliated Hospital, Anhui Medical University, Hefei 230000, China; (P.D.); (G.Z.); (J.H.)
- Key Laboratory of Dermatology, Ministry of Education of the People’s Republic of China, Hefei 230000, China
| | - Guiyuan Zhang
- Institute of Dermatology and Department of Dermatology, The First Affiliated Hospital, Anhui Medical University, Hefei 230000, China; (P.D.); (G.Z.); (J.H.)
- Key Laboratory of Dermatology, Ministry of Education of the People’s Republic of China, Hefei 230000, China
| | - Junjie Hu
- Institute of Dermatology and Department of Dermatology, The First Affiliated Hospital, Anhui Medical University, Hefei 230000, China; (P.D.); (G.Z.); (J.H.)
- Key Laboratory of Dermatology, Ministry of Education of the People’s Republic of China, Hefei 230000, China
| | - Sen Yang
- Institute of Dermatology and Department of Dermatology, The First Affiliated Hospital, Anhui Medical University, Hefei 230000, China; (P.D.); (G.Z.); (J.H.)
- Department of Dermatology, Dushu Lake Hospital Affiliated to Soochow University, Suzhou 215128, China
- Key Laboratory of Dermatology, Ministry of Education of the People’s Republic of China, Hefei 230000, China
| |
Collapse
|
7
|
Rios Rodriguez V, Sánchez-Riera L, Haibel H, Höppner C, Torgutalp M, Proft F, Rademacher J, Binder E, Diehl A, Vranic I, Zhao Y, Mundayat R, Yndestad A, Poddubnyy D. Tofacitinib in early active axial spondyloarthritis: protocol of a randomized double-blind, placebo-controlled, multicenter phase IV study, FASTLANE. Ther Adv Musculoskelet Dis 2025; 17:1759720X251324429. [PMID: 40078463 PMCID: PMC11898075 DOI: 10.1177/1759720x251324429] [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: 11/27/2024] [Accepted: 02/13/2025] [Indexed: 03/14/2025] Open
Abstract
Background Early treatment initiation is one of the strongest predictors of good treatment response in axial spondyloarthritis (axSpA). Recently, the Assessment in SpondyloArthritis International Society (ASAS) defined early axSpA as a diagnosis of axSpA with a duration of axial symptoms equal to or less than 2 years. Tofacitinib is a Janus kinase (JAK) inhibitor for the treatment of ankylosing spondylitis. Objectives Compare the efficacy and safety of tofacitinib versus placebo (both on non-steroidal anti-inflammatory drug (NSAID) background) in patients with active early axSpA and inadequate response to at least one NSAID. Design This is a phase IV, randomized, double-blind, placebo-controlled, multicenter clinical trial. Methods and analysis The study will recruit 104 patients aged ⩾18 and ⩽45 years with active early axSpA (chronic back pain ⩽2 years), inadequate response to at least one NSAID, and objective signs of active inflammation (on magnetic resonance imaging (MRI) of sacroiliac joints (SIJs) or elevated C-reactive protein). Patients will be randomized 1:1 to receive tofacitinib 5 mg twice daily or placebo, with background naproxen 500 mg twice daily for 16 weeks. Patients not meeting early treatment response criteria at week 4 will receive open-label tofacitinib until week 16. Primary and key secondary endpoints at week 16 will be the proportion of patients achieving disease remission (Axial Spondyloarthritis Disease Activity Score <1.3) and change from baseline in MRI SIJ Spondyloarthritis Research Consortium of Canada osteitis score, respectively. Safety will be monitored up to 4 weeks after the last study drug dose. Ethics The study will be performed according to the ethical principles of the Declaration of Helsinki and will be approved by independent ethics committees of each center. Discussion This is one of the first randomized clinical trials designed to evaluate the efficacy and safety of a JAK inhibitor in the recently ASAS-defined "early" axSpA population. Trial registration: ClinicalTrials.gov: NCT06112665; CTIS: 2023-505050-18-00.
Collapse
Affiliation(s)
- Valeria Rios Rodriguez
- Department of Gastroenterology, Infectious Diseases and Rheumatology (including Nutrition Medicine), Charité—Universitätsmedizin Berlin, Hindenburgdamm 30, Berlin 12203, Germany
| | | | - Hildrun Haibel
- Department of Gastroenterology, Infectiology and Rheumatology (including Nutrition Medicine), Charité—Universitätsmedizin Berlin, Berlin, Germany
| | - Caroline Höppner
- Department of Gastroenterology, Infectiology and Rheumatology (including Nutrition Medicine), Charité—Universitätsmedizin Berlin, Berlin, Germany
| | - Murat Torgutalp
- Department of Gastroenterology, Infectiology and Rheumatology (including Nutrition Medicine), Charité—Universitätsmedizin Berlin, Berlin, Germany
| | - Fabian Proft
- Department of Gastroenterology, Infectiology and Rheumatology (including Nutrition Medicine), Charité—Universitätsmedizin Berlin, Berlin, Germany
| | - Judith Rademacher
- Department of Gastroenterology, Infectiology and Rheumatology (including Nutrition Medicine), Charité—Universitätsmedizin Berlin, Berlin, Germany
| | | | | | | | | | | | | | - Denis Poddubnyy
- Department of Gastroenterology, Infectiology and Rheumatology (including Nutrition Medicine), Charité—Universitätsmedizin Berlin, Berlin, Germany
- Toronto University Hospital, Toronto, ON, Canada
| |
Collapse
|
8
|
Ciccia F, McGonagle D, Thomas R, Marzo-Ortega H, Martin DA, Yndestad A, Volkov M. JAK inhibition and axial spondyloarthritis: new steps on the path to understanding pathophysiology. Front Immunol 2025; 16:1488357. [PMID: 40103808 PMCID: PMC11913702 DOI: 10.3389/fimmu.2025.1488357] [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: 08/29/2024] [Accepted: 02/13/2025] [Indexed: 03/20/2025] Open
Abstract
Axial spondyloarthritis (axSpA) is a chronic inflammatory disease that predominantly affects the sacroiliac joints and spine. Tumor necrosis factor (TNF) and interleukin (IL)-17A are key cytokines in disease pathogenesis and are established axSpA treatment targets. Recently, axSpA treatment options have been complemented by Janus kinase inhibitors (JAKi), which inhibit various cytokines without directly impacting TNF or IL-17 signaling. The effect of JAKi on axSpA remains under investigation: besides a JAK2-mediated (and potentially tyrosine kinase 2 [TYK2]-mediated) effect on the IL-23/IL-17 axis, emerging evidence suggests γδ T cells, type 3 innate lymphoid cells, and mucosa-associated invariant T cells, which are dependent on IL-7 and/or IL-15 and thus on JAK1, are strongly inhibited by JAKi used to treat axSpA. This review summarizes potential effects of JAKi on axSpA and shows evidence from pre-clinical/clinical studies. Greater understanding of the mechanisms of action of available treatments may improve knowledge of axSpA and pave the road for future therapies.
Collapse
Affiliation(s)
- Francesco Ciccia
- Dipartimento di Medicina di Precisione, Università della Campania L. Vanvitelli, Naples, Italy
| | - Dennis McGonagle
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds and NIHR Leeds Biomedical Research Centre, The Leeds Teaching Hospitals NHS Trust, Leeds, United Kingdom
| | - Ranjeny Thomas
- Frazer Institute, The University of Queensland, Woolloongabba, QLD, Australia
| | - Helena Marzo-Ortega
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds and NIHR Leeds Biomedical Research Centre, The Leeds Teaching Hospitals NHS Trust, Leeds, United Kingdom
| | | | | | | |
Collapse
|
9
|
Wang T, Wu W, Zhang X, Gan B, Zhou Y, Cheng X. Tofacitinib treatment for plaque psoriasis and psoriatic arthritis: A meta-analysis of randomised controlled trials. Indian J Dermatol Venereol Leprol 2025; 91:172-179. [PMID: 39361844 DOI: 10.25259/ijdvl_14_2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Accepted: 06/26/2024] [Indexed: 10/05/2024]
Abstract
Objectives Tofacitinib is used as an oral Janus-associated kinase (JAK) inhibitor acting on JAK1 and JAK3, in treating psoriatic disease. However, there is still no consensus on the optimal dosage and duration of tofacitinib. In this study, we aimed to evaluate the effects of tofacitinib in treating psoriatic disease. Methods A literature search was done utilising Cochrane library, Medline, EMBASE, Wiley Online library, Web of Science and BIOSIS Previews through December 18, 2022. We performed a meta-analysis of published original studies to assess the impact of tofacitinib in plaque psoriasis or psoriatic arthritis therapy based on seven randomised controlled trials (RCTs) involving 2,672 patients (receiving tofacitinib) and 853 controls (receiving placebo). Results Compared with placebo, the treatment of 5 mg twice-daily (BID) tofacitinib for 12 weeks is sufficient to significantly alleviate the main clinical manifestations of psoriasis [≥75% decrease in Psoriasis Area and Severity Index score (PASI 75): Risk ratio (RR)=4.38 (95% Confidence interval (CI) 2.51 to 7.64); ≥90% decrease in PASI score (PASI 90): RR=21.68 (95% CI 4.20 to 111.85); Physician's Global Assessment of 'clear' or 'almost clear' (PGA 0/1): RR=3.93 (95%CI 3.03 to 5.09)]. Interestingly, there was no significant difference in improvement in PGA 0/1 with 5 mg BID tofacitinib given for 16 weeks when compared with 5 mg BID tofacitinib for 12 weeks [RR=1.11 (95%CI 0.98 to 1.25)]. Additionally, the 5 mg BID tofacitinib for 16 weeks treatment schedule significantly increased the incidence of upper respiratory tract infection (URTI) [RR=1.89 (95%CI 1.06 to 3.38)] as compared to 5 mg BID tofacitinib for 12 weeks treatment schedule [RR=1.15 (95%CI 0.60 to 2.20)]. Conclusion The 5 mg BID tofacitinib for 12 weeks treatment significantly improved psoriasis without causing too many specific adverse events. This indicated that tofacitinib is an effective treatment plan for psoriatic disease by reasonably controlling dosage and dosing time.
Collapse
Affiliation(s)
- Tao Wang
- Department of Sexual Medicine/Traditional Chinese Medicine, Fifth People's Hospital of Shunde District (Longjiang Hospital of Shunde District), Foshan, Guangdong, China
| | - Wei Wu
- Department of Medical Science, Shunde Polytechnic, Foshan, Guangdong, China
| | - Xiaoqing Zhang
- Department of Basic Medical Sciences, Guangdong Medical University, Dongguan, Guangdong, China
| | - Bin Gan
- Department of Respiratory Medicine, Fifth People's Hospital of Shunde District (Longjiang Hospital of Shunde District), Foshan, Guangdong, China
| | - Yanfang Zhou
- Department of Basic Medical Sciences, Guangdong Medical University, Dongguan, Guangdong, China
| | - Xiaoyan Cheng
- Department of Traditional Chinese Medicine, The Second Clinical Medical College, Guangdong Medical University, Dongguan, Guangdong, China
| |
Collapse
|
10
|
Menon S, Shoji S, Tsuchiwata S, Fallon L, Kanik K. Exposure-Response Analysis of Tofacitinib in Active Psoriatic Arthritis: Results from Two Phase 3 Studies. J Clin Pharmacol 2025; 65:369-377. [PMID: 39453735 PMCID: PMC11867917 DOI: 10.1002/jcph.6147] [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: 07/10/2024] [Accepted: 09/23/2024] [Indexed: 10/27/2024]
Abstract
Tofacitinib is an oral Janus kinase inhibitor for the treatment of psoriatic arthritis (PsA). These post hoc exposure-response (E-R) analyses of pooled data from two Phase 3 studies (NCT01877668 and NCT01882439) characterized the relationships between tofacitinib exposure and efficacy (American College of Rheumatology [ACR] criteria), and changes in hemoglobin (Hgb) in patients with PsA. Efficacy data for the proportion of patients receiving tofacitinib 5 or 10 mg twice daily, or placebo, achieving ACR ≥20%, ≥50%, or ≥70% response criteria (ACR20, ACR50, and ACR70, respectively) at Month 3, were modeled jointly using a four-category ordered categorical exposure-response model (ACR20 non-responder, ACR20 responder but not ACR50 responder, ACR50 responder but not ACR70 responder, and ACR70 responder). A maximum drug effect (Emax) model (using average concentrations of tofacitinib at steady state [Cavg]) adequately described the exposure-ACR response rate relationship. Model-predicted response rates for tofacitinib 5 and 10 mg twice daily were 51% and 58%, respectively, for ACR20; 29% and 36% for ACR50; and 15% and 20% for ACR70. The E-R relationship between tofacitinib exposure and changes in Hgb was assessed using an indirect response model, which generally predicted Hgb concentration-time profiles across treatments well. The proportions of patients experiencing a decrease in Hgb of >2 g/dL were similar with tofacitinib 5 mg twice daily or placebo. These results were generally consistent with previous analyses in rheumatoid arthritis and psoriasis, and support the use of tofacitinib 5 mg twice daily for active PsA.
Collapse
|
11
|
Cagnotto G, Bruschettini M, Stróżyk A, Scirè CA, Compagno M. Tumor necrosis factor (TNF) inhibitors for psoriatic arthritis. Cochrane Database Syst Rev 2025; 2:CD013614. [PMID: 39945386 PMCID: PMC11822884 DOI: 10.1002/14651858.cd013614.pub2] [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: 02/16/2025]
Abstract
BACKGROUND Psoriatic arthritis (PsA) is a chronic arthritis affecting people with psoriasis. If untreated, it may lead to disability. Recommended drugs are non-steroidal anti-inflammatory drugs (NSAIDs), corticosteroids, conventional synthetic disease-modifying anti-rheumatic drugs (csDMARDs), biologic DMARDs (bDMARDs), and targeted synthetic DMARDs (tsDMARDs). Tumour necrosis factor inhibitors (TNFi) are the first choice bDMARDs. OBJECTIVES To assess the benefits and harms of TNFi in adults with psoriatic arthritis. SEARCH METHODS We searched CENTRAL, MEDLINE, Embase, ClinicalTrials.gov and the World Health Organization trials portal up to 28 March 2024. SELECTION CRITERIA We included randomized controlled trials (RCTs) in adults with PsA, comparing TNFi to placebo, physiotherapy, NSAIDs, corticosteroids, and cs/b/tsDMARDs. Major outcomes included clinical improvement, minimal disease activity, physical function, health-related quality of life, radiographic progression, serious adverse events, and withdrawals due to adverse events. DATA COLLECTION AND ANALYSIS We used standard Cochrane methods. The primary comparison was TNFi versus placebo. The primary time point was 12 weeks for clinical improvement; 24 weeks for minimal disease activity, function, quality of life, and radiographic progression; and the end of the trial period for serious adverse events and withdrawals due to adverse events. MAIN RESULTS We included 25 RCTs randomizing 7857 participants. Four studies compared TNFi to methotrexate and one to ustekinumab in DMARD-naïve participants. In csDMARD-inadequate responders, 11 studies compared TNFi to placebo; four studies compared TNFi to placebo and ixekizumab, bimekizumab, tofacitinib, or upadacitinib; and three studies compared TNFi to ixekizumab, secukinumab, and ustekinumab. Two studies compared different TNFi. We found no studies with b/tsDMARD-inadequate responders (b/tsDMARD-IR). No studies compared TNFi to NSAIDs, corticosteroids, or physiotherapy. Performance (32%), detection (56%) and reporting (80%) biases were at high or unclear risk across studies. Only one study had a low risk of bias in all domains. We limit reporting to the primary comparison, TNFi versus placebo. DMARD-naïve We found no studies comparing TNFi with placebo in DMARD-naïve participants. csDMARD-inadequate responders TNFi probably result in a large clinical improvement compared to placebo. At 12 weeks, 149/1926 (8%) participants in the placebo group showed a clinical improvement (ACR50) compared to 784/2141 (37%) participants in the TNFi group (risk ratio (RR) 5.63, 95% confidence interval (CI) 3.98 to 7.96; I2 = 65%; 14 studies, 4067 participants; moderate-certainty evidence). TNFi probably result in a higher proportion of participants in minimal disease activity. At 24 weeks, 95/1017 (9%) participants in the placebo group were in minimal disease activity compared to 428/1336 (32%) participants in the TNFi group (RR 3.76, 95% CI 2.39 to 5.92; I2 = 72%; 5 studies, 2353 participants; moderate-certainty evidence). At 24 weeks, TNFi may improve function compared to placebo. The mean change in function from baseline (assessed with the Health Assessment Questionnaire; score from 0 to 3, 0 = no disability; minimal clinically important difference (MCID) = 0.35) was -0.14 points with placebo and 0.33 points lower (0.41 lower to 0.25 lower) with TNFi (I2 = 72%; 8 studies, 2949 participants; low-certainty evidence). TNFi probably result in a clinically important improvement in health-related quality of life. The mean change in quality of life from baseline (assessed with the Short Form 36-item Mental Component Summary questionnaire; score from 0 to 100, 100 = best score; MCID = 1.7) was 2.4 points with placebo and 3.29 points higher (2.18 points higher to 4.40 points higher) with TNFi (I2 = 52%; 8 studies, 2928 participants; moderate-certainty evidence). TNFi probably slightly reduce radiographic progression. The mean change in radiographic progression (assessed with the Sharp/Van der Heijde-PsA score; scale from 0 to 528, 0 = no damage) was 0.25 points with placebo and 0.37 points lower with TNFi (0.48 lower to 0.25 lower) (I2 = 32%; 7 studies, 2478 participants; moderate-certainty evidence) at 24 weeks. We downgraded the evidence to moderate certainty for clinical improvement, minimal disease activity, quality of life, and radiographic progression due to risk of bias. For function, we downgraded the evidence to low certainty for risk of bias and imprecision. TNFi may result in little to no difference in serious adverse events, but may slightly increase withdrawals due to adverse events, compared to placebo. At the end of follow-up: 56/1826 participants (3%) given placebo and 69/1900 (4%) participants given TNFi experienced serious adverse events (RR 1.00, 95% CI 0.70 to 1.42; I2 = 0%; 13 studies, 3866 participants; low-certainty evidence); and 35/1926 (2%) participants given placebo and 65/2140 (3%) given TNFi withdrew due to adverse events (RR 1.53, 95% CI 1.01 to 2.33; I2 = 0%; 14 studies, 4066 participants; low-certainty evidence). We downgraded the evidence to low certainty for risk of bias and imprecision. AUTHORS' CONCLUSIONS In csDMARD-inadequate responders, moderate-certainty evidence showed that TNFi probably result in a large clinical improvement, lower disease activity, small decrease in radiographic progression, and better quality of life compared to placebo. Low-certainty evidence showed that TNFi may lead to a slight improvement in physical function compared to placebo. Low-certainty evidence suggested that TNFi may lead to a slight increase in withdrawals due to adverse events, whereas they may result in little to no difference in serious adverse events compared to placebo. No trials assessed TNFi compared to placebo in DMARD-naïve participants or in b/tsDMARD-IR.
Collapse
Affiliation(s)
- Giovanni Cagnotto
- Clinical Sciences Malmö, Lund University, Malmö, Sweden
- Rheumatology, Skåne University Hospital, Malmö, Sweden
| | - Matteo Bruschettini
- Department of Clinical Sciences Lund, Paediatrics, Lund University, Skåne University Hospital, Lund, Sweden
- Cochrane Sweden, Department of Research, Development, Education and Innovation, Lund University, Skåne University Hospital, Lund, Sweden
| | - Agata Stróżyk
- Department of Paediatrics, Medical University of Warsaw, Warsaw, Poland
| | - Carlo Alberto Scirè
- School of Medicine, University of Milano-Bicocca, Milan, Italy
- Rheumatology Unit, IRCCS San Gerardo dei Tintori Foundation, Monza, Italy
| | - Michele Compagno
- Clinical Sciences Malmö, Lund University, Malmö, Sweden
- Rheumatology, Skåne University Hospital, Malmö, Sweden
| |
Collapse
|
12
|
Xiao A, Roy A, Dennett L, Yacyshyn E, Li MD. Imaging in clinical trials for psoriatic arthritis: a scoping review. Skeletal Radiol 2025:10.1007/s00256-025-04884-8. [PMID: 39912887 DOI: 10.1007/s00256-025-04884-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2024] [Revised: 01/09/2025] [Accepted: 01/26/2025] [Indexed: 02/07/2025]
Abstract
Psoriatic arthritis (PsA) is a chronic inflammatory condition principally affecting the skin and musculoskeletal system, associated with comorbidities and decreased quality of life. Imaging is crucial for diagnosis, monitoring progression, and evaluating treatment efficacy; therefore, it plays an important role in PsA clinical trials. In this review, we aimed to characterize how imaging modalities and advances in imaging techniques are being used specifically in the PsA clinical trial literature. Following the Arksey and O'Malley framework for scoping reviews, we conducted a comprehensive search of multiple literature databases, from January 1, 2000, to June 27, 2024. We captured 1961 articles and after deduplication, title and abstract screening, and full-text review, 53 studies were included. Radiographs were the most used imaging modality (n = 32/53, 60%), and radiographic progression was frequently measured using the PsA-modified Sharp/van der Heijde score. MRI (n = 16/53, 30%) was used in evaluating peripheral and axial disease, with more recent adoption of validated scoring systems (PsAMRIS and SPARCC). Ultrasound (n = 11/53, 21%), including power Doppler, was used to assess soft tissue inflammation. Standardized scoring systems (e.g. GLOESS) were used in a minority of ultrasound-based studies. Multimodality imaging (n = 7/53, 13%) and CT (n = 2/53, 4%) was uncommon. The development of PsA-specific scoring systems for radiographs and MRI has been instrumental in advancing imaging assessment in PsA. However, their application remains limited, particularly in ultrasound, where further standardization is needed. Future clinical trials should focus on increasing the adoption of PsA-specific scoring systems, exploring advanced techniques (e.g. DCE-MRI), and multi-modal imaging to improve PsA disease monitoring.
Collapse
Affiliation(s)
- Andrew Xiao
- Department of Medicine, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Ainge Roy
- Faculty of Science, University of Alberta, Edmonton, AB, Canada
| | - Liz Dennett
- Geoffrey and Robyn Sperber Health Sciences Library, University of Alberta, Edmonton, AB, Canada
| | - Elaine Yacyshyn
- Department of Medicine, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Matthew D Li
- Department of Radiology and Diagnostic Imaging, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, AB, Canada.
| |
Collapse
|
13
|
Chikhoune L, Poggi C, Moreau J, Dubucquoi S, Hachulla E, Collet A, Launay D. JAK inhibitors (JAKi): Mechanisms of action and perspectives in systemic and autoimmune diseases. Rev Med Interne 2025; 46:89-106. [PMID: 39550233 DOI: 10.1016/j.revmed.2024.10.452] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Revised: 10/07/2024] [Accepted: 10/27/2024] [Indexed: 11/18/2024]
Abstract
Janus kinase (JAK) molecules are involved in important cellular activation pathways. Over the past decade, many targeted therapies have emerged, including the increasingly promising role of JAK inhibitors (JAKi) in the treatment of inflammatory and autoimmune diseases. The spectrum of use of these small molecules is increasingly broader. JAKi have been approved in several autoimmune diseases. Currently, four molecules (tofacitinib, baricitinib, upadacitinib and filgotinib) have been labeled for moderate to severe rheumatoid arthritis (RA) with failure or poor tolerance of one or more conventional disease-modifying antirheumatic drug (csDMARDS), or biologics (bDMARDS). JAKi are now also commonly used in other diseases such as psoriatic arthritis, ankylosing spondylitis, and ulcerative colitis. They have also shown promising results in clinical trials for the treatment of other autoimmune conditions. We present here their mechanisms of action, and the main data about JAKi use on systemic and autoimmune diseases.
Collapse
Affiliation(s)
- Liticia Chikhoune
- CHU de Lille, Service de Médecine Interne et Immunologie Clinique, Centre de référence des Maladies Auto-Immunes et Auto-inflammatoires Systémiques rares de l'Adulte du Nord, Nord-Ouest, Méditerranée et Guadeloupe (CeRAINOM), 59000 Lille, France
| | - Claire Poggi
- CHU de Lille, Service de Médecine Interne et Immunologie Clinique, Centre de référence des Maladies Auto-Immunes et Auto-inflammatoires Systémiques rares de l'Adulte du Nord, Nord-Ouest, Méditerranée et Guadeloupe (CeRAINOM), 59000 Lille, France
| | - Julie Moreau
- CHU de Lille, Service de Médecine Interne et Immunologie Clinique, Centre de référence des Maladies Auto-Immunes et Auto-inflammatoires Systémiques rares de l'Adulte du Nord, Nord-Ouest, Méditerranée et Guadeloupe (CeRAINOM), 59000 Lille, France
| | - Sylvain Dubucquoi
- Institut d'Immunologie, Pôle de Biologie Pathologie Génétique Médicale, CHU de Lille, 59000 Lille, France; U1286-INFINITE-Institute for Translational Research in Inflammation, Université de Lille, 59000 Lille, France
| | - Eric Hachulla
- CHU de Lille, Service de Médecine Interne et Immunologie Clinique, Centre de référence des Maladies Auto-Immunes et Auto-inflammatoires Systémiques rares de l'Adulte du Nord, Nord-Ouest, Méditerranée et Guadeloupe (CeRAINOM), 59000 Lille, France; U1286-INFINITE-Institute for Translational Research in Inflammation, Université de Lille, 59000 Lille, France; Inserm, 59000 Lille, France
| | - Aurore Collet
- Institut d'Immunologie, Pôle de Biologie Pathologie Génétique Médicale, CHU de Lille, 59000 Lille, France; U1286-INFINITE-Institute for Translational Research in Inflammation, Université de Lille, 59000 Lille, France
| | - David Launay
- CHU de Lille, Service de Médecine Interne et Immunologie Clinique, Centre de référence des Maladies Auto-Immunes et Auto-inflammatoires Systémiques rares de l'Adulte du Nord, Nord-Ouest, Méditerranée et Guadeloupe (CeRAINOM), 59000 Lille, France; U1286-INFINITE-Institute for Translational Research in Inflammation, Université de Lille, 59000 Lille, France; Inserm, 59000 Lille, France.
| |
Collapse
|
14
|
Horbal N, Maksymowych WP. Nociplastic pain in axial spondyloarthritis and psoriatic arthritis: role of JAK kinases in immunopathology and therapeutic impact of JAK inhibitors. Expert Rev Clin Immunol 2025; 21:137-152. [PMID: 39225245 DOI: 10.1080/1744666x.2024.2400294] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Revised: 07/22/2024] [Accepted: 08/30/2024] [Indexed: 09/04/2024]
Abstract
INTRODUCTION Pain in both peripheral and axial joints is a major symptom in patients with psoriatic arthritis (PsA) and axial spondyloarthritis (axSpA). Emerging evidence demonstrates pain mechanisms, beyond those related to inflammation or joint damage, based on aberrant processing of nociceptive stimuli peripherally as well as centrally. The Janus kinase/signal transducers and activators of transcription (JAK-STAT) signaling pathway has been implicated in the processing of pain beyond its role in mediating inflammation and inhibitors of this pathway approved for the treatment of axSpA and PsA have been shown to alleviate a broad array of pain outcomes in both axial and peripheral joints. AREAS COVERED We review recent definitions and standardization of the nomenclature for categorizing chronic pain according to causality, assessment tools to evaluate nociplastic pain, the pathophysiologic role of JAK-STAT signaling in nociplastic pain, evidence for the presence of nociplastic pain in axSpA and PsA, and the impact of JAK inhibitors (JAKi) on pain outcomes in clinical trials (PubMed: 01/01/2019-04/01-2024). EXPERT OPINION Nociplastic pain assessment has been confined almost entirely to the use of a limited number of questionnaires in cross-sectional studies of these diseases. Though effective for alleviating pain, it is unclear if JAKi specifically impact nociplastic pain.
Collapse
Affiliation(s)
- Natalya Horbal
- Department of Medicine, Danylo Halytsky Lviv National Medical University, Lviv, Ukraine
| | | |
Collapse
|
15
|
Braun J. Fast, Present and Future of the Concept of Spondyloarthritis. Curr Rheumatol Rep 2025; 27:15. [PMID: 39869233 DOI: 10.1007/s11926-024-01179-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/24/2024] [Indexed: 01/28/2025]
Abstract
PURPOSE OF REVIEW Axial spondyloarthritis (axSpA) is a rather prevalent chronic inflammatory rheumatic disease that affects already relatively young patients. It has been known better since the end of the nineteenth century but quite a lot has been learned since the early 60ies when the first classification (diagnostic) criteria for ankylosing spondylitis (AS) were agreed on. I have been part of many developments in the last 30 years, and I'm happy to have been able to contribute to the scientific progress in terms of diagnosis, imaging, pathophysiology and therapy. When I was asked to write a manuscript about the SpA concept I felt honored. Thus, the purpose of this extensive review was, on the one hand, to describe the history of AS and axSpA, and on the other hand, to reason about the concept and the gestalt of axSpA, and finally to deliver some ideas what future researchers could possibly do to further study the disease. RECENT FINDINGS The last 3 decades were full of innovations for both, classification and treatment of axSpA which also helped us to learn about the pathophysiology. Thus, TNFa, IL-17, IL-23 and Janus kinase are established targets to reduce inflammation. IL-17 and IL-23 are very special in that regard because they both work for psoriasis but only anti-IL-17 agents which don't work in IBD are approved for axSpA, while IL 23 inhibitors are approved for both, psoriasis and IBD, but they don't work in axSpA. New imaging techniques such as low dose CT and synthetic MRI are likely to improve the detection of both active and structural lesions of axSpA. This manuscript tries to describe the most important findings about axSpA. The main aim of research remains to discover the pathophysiology and to further improve treatment options in order to reduce and abolish inflammation and prevent new bone formation to increase the quality of life of our patients. The differences between male and female disease and the role of the immune system in axSpA are now the main challenges, and the role of special T-cell receptors seem to deserve special interest.
Collapse
Affiliation(s)
- J Braun
- Rheumatologisches Versorgungszentrum Steglitz, Ruhr Universität Bochum, Schloßstr.110, 12163, Berlin, Germany.
| |
Collapse
|
16
|
Gladman DD, Dougados M, Marzo-Ortega H, Cadatal MJ, Agarwal E, Kinch CD, Nash P. Long-term tofacitinib efficacy and safety in psoriatic arthritis with or without prior biologic DMARD exposure: a post hoc analysis. Rheumatol Adv Pract 2025; 9:rkaf008. [PMID: 40241921 PMCID: PMC12002910 DOI: 10.1093/rap/rkaf008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Accepted: 11/25/2024] [Indexed: 04/18/2025] Open
Abstract
Objective To assess long-term tofacitinib efficacy and safety in patients with PsA with or without prior biologic DMARD (bDMARD) exposure. Methods Data were pooled post hoc from three phase 3 and one long-term extension (LTE) PsA studies and stratified by TNF inhibitor-inadequate responder (TNFi-IR) or bDMARD-naïve patient status at the phase 3 study baseline. Data were reported as all tofacitinib (patients receiving one or more tofacitinib doses) or average tofacitinib 5 and 10 mg twice daily (patients receiving an average total daily dose <15 and ≥15 mg, respectively). Drug survival to month 51, efficacy to month 42 and safety were assessed descriptively. Results A total of 408 TNFi-IR patients (including 29 TNFi-experienced with unknown IR status) and 562 bDMARD-naïve patients were included. At baseline, TNFi-IR patients were more likely to be ≥65 years old, have cardiovascular/venous thromboembolism risk and have longer disease duration vs bDMARD-naïve patients. Drug survival was numerically shorter in TNFi-IR vs bDMARD-naïve patients. Tofacitinib efficacy was generally sustained to month 42, regardless of prior bDMARD treatment. Minimal disease activity/ PsA Disease Activity Score ≤3.2/>75% Psoriasis Area and Severity Index improvement response rates were numerically lower in TNFi-IR vs bDMARD-naïve patients to month 42, but rates of achieving an HAQ Disability Index ≤0.5 and enthesitis/dactylitis resolution were similar. In TNFi-IR vs bDMARD-naïve patients, treatment-emergent adverse event incidence rates were higher and serious adverse event, serious infection and herpes zoster incidence rates were numerically higher (CI overlapped). Conclusion These findings support long-term tofacitinib efficacy and safety in TNFi-IR and bDMARD-naïve patients. However, the benefit-risk profile appeared more favourable in bDMARD-naïve patients, likely due to differences in baseline characteristics and risk factors between subgroups. Trial registration ClinicalTrials.gov: NCT01877668, NCT01882439, NCT03486457, NCT01976364.
Collapse
Affiliation(s)
- Dafna D Gladman
- Schroeder Arthritis Institute, Krembil Research Institute, Department of Medicine, Toronto Western Hospital, Toronto, ON, Canada
| | - Maxime Dougados
- Department of Rheumatology, Hôpital Cochin, Assistance Publique-Hôpitaux de Paris, INSERM (U1153): Clinical Epidemiology and Biostatistics, PRES Sorbonne Paris-Cité, Université de Paris Cité, Paris, France
| | - Helena Marzo-Ortega
- NIHR Leeds Biomedical Research Centre, Leeds Teaching Hospital Trusts, Leeds Institute for Rheumatic and Musculoskeletal Medicine, University of Leeds, Leeds, UK
| | | | | | | | - Peter Nash
- School of Medicine, Griffith University, Brisbane, QLD, Australia
| |
Collapse
|
17
|
Gladman DD, Nash P, Mease PJ, FitzGerald O, Duench S, Cadatal MJ, Masri KR. Efficacy and safety of tofacitinib in an open-label, long-term extension study in patients with psoriatic arthritis who received adalimumab or tofacitinib in a Phase 3 randomized controlled study: a post hoc analysis. Arthritis Res Ther 2024; 26:218. [PMID: 39702318 DOI: 10.1186/s13075-024-03442-2] [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: 07/03/2024] [Accepted: 11/24/2024] [Indexed: 12/21/2024] Open
Abstract
BACKGROUND Data on treatment switching directly from tumor necrosis factor inhibitors to tofacitinib in psoriatic arthritis (PsA) are limited. This post hoc analysis assessed efficacy and safety outcomes in patients with PsA who directly switched to tofacitinib in a long-term extension (LTE) study after receiving adalimumab (ADA) in a Phase 3 study, compared with those who continued to receive tofacitinib. METHODS Patients with active PsA received tofacitinib 5 mg twice daily (BID) or ADA 40 mg once every 2 weeks in a 12-month, randomized, double-blind study (OPAL Broaden) and then continued or switched to tofacitinib 5 mg BID and maintained this dose in an open-label LTE study (OPAL Balance). Efficacy was assessed 3 months before the last visit and at the last visit in the Phase 3 study, and at month 3 (or month 6 for select outcomes) in the LTE study and included rates of ≥ 20/50/70% improvement in American College of Rheumatology response criteria, Psoriasis Area and Severity Index ≥ 75% improvement, Health Assessment Questionnaire-Disability Index (HAQ-DI) response (decrease from baseline ≥ 0.35 for patients with baseline HAQ-DI ≥ 0.35), Psoriatic Arthritis Disease Activity Score ≤ 3.2, and minimal disease activity; and change from baseline in Functional Assessment of Chronic Illness Therapy-Fatigue score. Safety was assessed at months 3 and 12 in both studies via incidence rates (patients with first events/100 patient-years). RESULTS Overall, 180 patients were included (ADA→tofacitinib 5 mg BID: n = 91; continuing tofacitinib 5 mg BID: n = 89). At Phase 3 baseline, patients in the ADA→tofacitinib 5 mg BID group tended to be younger and have less active disease compared with those continuing tofacitinib. Efficacy was similar between groups in the Phase 3 study, and was maintained to month 3 or 6 in the LTE study. Treatment-emergent adverse events (AEs), serious AEs, and serious infections were generally similar in the Phase 3 and LTE studies, and between groups within each study. CONCLUSION Tofacitinib efficacy and safety were similar in patients with PsA who directly switched from ADA to tofacitinib and those who continued tofacitinib, suggesting that patients can be directly switched from ADA to tofacitinib without any washout period. TRIAL REGISTRATION NCT01877668; NCT01976364.
Collapse
Affiliation(s)
- Dafna D Gladman
- Department of Medicine, University of Toronto, and Schroeder Arthritis Institute, Krembil Research Institute, Toronto Western Hospital, 399 Bathurst St. 1E-411, Toronto, ON, M5T 2S8, Canada.
| | - Peter Nash
- School of Medicine, Griffith University, Brisbane, QLD, Australia
| | - Philip J Mease
- Rheumatology Research, Swedish Medical Center/Providence St. Joseph Health, and University of Washington School of Medicine, Seattle, WA, USA
| | - Oliver FitzGerald
- Conway Institute for Biomolecular Research, University College Dublin, Dublin, Ireland
| | | | | | | |
Collapse
|
18
|
Lamberg O, Pandher K, Troost JP, Lim HW. Long-term adverse event risks of oral JAK inhibitors versus immunomodulators: a literature review. Arch Dermatol Res 2024; 317:109. [PMID: 39666160 DOI: 10.1007/s00403-024-03578-w] [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: 09/24/2024] [Revised: 09/27/2024] [Accepted: 11/26/2024] [Indexed: 12/13/2024]
Abstract
The long-term adverse event risks associated with oral Janus kinase (JAK) inhibitors compared to broader immunomodulators are poorly understood, with limited comparative studies available. This study aims to assess the long-term adverse event risks of oral JAK inhibitors compared to broader immunomodulators in dermatology. A PubMed search included terms such as specific drug names and "adverse events," "long-term safety," "malignancy," "cardiovascular events," and/or "infections." Included were studies with over one year of medication exposure reporting adverse events. A total of 25 studies were included, comprising 19 clinical trials and 6 cohort studies, representing greater than 100,000 patient years of data. We found comparable long-term incidence rates of malignancy (excluding non-melanoma skin cancer (NMSC)), venous thromboembolism (VTE), and serious infections between oral JAK inhibitors and broader immunomodulators. Oral JAK inhibitors demonstrated lower incidence rates of NMSC and major adverse cardiovascular events (MACE), but higher rates of herpes zoster virus (HZV) infections compared to non-JAK medications. Study limitations include variations in underlying diseases, age, and comorbidities between studies introducing patient population heterogeneity. Overall, oral JAK inhibitors may be viable long-term treatment options given their comparable safety profile to broader immunomodulators currently used in dermatology. However, monitoring for HZV infections may be warranted. These findings aid clinicians in making informed treatment decisions and prioritizing patient safety.
Collapse
Affiliation(s)
- Olivia Lamberg
- University of Michigan Medical School, 1301 Catherine St, Ann Arbor, MI, 48109, USA.
| | - Karan Pandher
- Department of Dermatology, Henry Ford Health, Detroit, MI, USA
| | - Jonathon P Troost
- Michigan Institute for Clinical and Health Research, University of Michigan, Ann Arbor, MI, USA
| | - Henry W Lim
- Department of Dermatology, Henry Ford Health, Detroit, MI, USA
| |
Collapse
|
19
|
Ogdie A, Kristensen LE, Soriano ER, Akar S, Sun Y, Gruben D, Fallon L, Kinch CD, Gladman DD. Efficacy and Safety of Tofacitinib in Patients with Psoriatic Arthritis or Ankylosing Spondylitis by Cigarette Smoking Status. Rheumatol Ther 2024; 11:1649-1664. [PMID: 39320582 PMCID: PMC11557744 DOI: 10.1007/s40744-024-00711-z] [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: 05/16/2024] [Accepted: 07/30/2024] [Indexed: 09/26/2024] Open
Abstract
INTRODUCTION Routine care studies of psoriatic arthritis (PsA) and ankylosing spondylitis (AS) demonstrated attenuated responses to tumor necrosis factor inhibitors in current/past versus never smokers. This post hoc analysis assessed tofacitinib efficacy and safety in patients with PsA or AS by cigarette smoking status at trial screening. METHODS Pooled data from phase 3 and long-term extension (safety only) PsA trials and phase 2 and 3 AS trials were assessed by current/past versus never smoker status. Analysis included efficacy and safety data for tofacitinib 5 (PsA/AS) and 10 (PsA only) mg twice daily (BID) or placebo, and safety data in AS for tofacitinib 2 and 10 mg BID. Efficacy outcomes included American College of Rheumatology ≥ 50% responses (ACR50) and minimal disease activity (MDA) responses to month (M)6/M3 (tofacitinib/placebo) in PsA; and ≥ 40% improvement in Assessment of SpondyloArthritis international Society responses (ASAS40) and AS Disease Activity Score (ASDAS) < 2.1 responses to week (W)16 in AS. Safety was assessed to M48/W48 (PsA/AS), adjusted for treatment/smoking status/median body mass index (BMI) status/sex/trial/treatment-smoking status interaction. RESULTS PsA/AS cohorts included 342/178 current/past and 572/194 never smokers. Tofacitinib efficacy was generally greater versus placebo to M3/W6 (PsA/AS), and comparable in current/past and never smokers to M6/W16 (PsA/AS). In patients receiving ≥ 1 tofacitinib dose, adjusted treatment-emergent adverse event (TEAE)/serious AE (SAE)/discontinuation due to AE incidence rates (IRs) to M48 in PsA were higher in current/past versus never smokers; adjusted IRs to W48 in AS were higher in current/past versus never smokers for TEAEs, but similar for SAEs/discontinuation due to AEs. CONCLUSIONS In both patients with PsA and AS, tofacitinib efficacy was greater versus placebo, and comparable across smoking categories. Adjusted IRs were higher in current/past versus never smokers for TEAEs, SAEs, discontinuation due to AEs in PsA, and for TEAEs in AS, complementing reports of associations between smoking and comorbidities in spondyloarthritis. Findings support increased surveillance/caution for patients with PsA or AS with smoking history. TRIAL REGISTRATION ClinicalTrials.gov: NCT01877668/NCT01882439/NCT03486457/NCT01976364/NCT01786668/NCT03502616.
Collapse
Affiliation(s)
- Alexis Ogdie
- Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Lars E Kristensen
- The Parker Institute, Copenhagen University Hospital, Bispebjerg and Frederiksberg, Copenhagen, Denmark
| | - Enrique R Soriano
- Rheumatology Unit, Hospital Italiano de Buenos Aires, University Institute Hospital Italiano de Buenos Aires, Buenos Aires, Argentina
| | - Servet Akar
- Division of Rheumatology, Department of Internal Medicine, Izmir Kâtip Çelebi University Faculty of Medicine, Izmir, Turkey
| | | | | | | | | | - Dafna D Gladman
- Schroeder Arthritis Institute, Krembil Research Institute, Department of Medicine, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
20
|
Nash P, Sumpton D, Tellus M, Feletar M, Bird P, Hall S. A review and recommendations on the management of psoriatic arthritis in Australia 2024. Intern Med J 2024. [PMID: 39587898 DOI: 10.1111/imj.16557] [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: 01/22/2024] [Accepted: 09/29/2024] [Indexed: 11/27/2024]
Abstract
Psoriatic arthritis (PsA) is a progressive, systemic inflammatory disease. It can lead to serious joint damage and disability, increased cardiovascular risk and reduced quality of life. Six experts met to develop the recommendations for the management of PsA in Australia. The final recommendations are approved by all panel members. Management and treatment recommendations have been made under six subheadings: Recommendations for non-steroidal anti-inflammatory drugs and glucocorticoids; Disease-modifying treatment; Screening and monitoring; Family planning; Symptom treatment and extra-articular manifestations; Comorbidities and lifestyle considerations. Our recommendations for the management of PsA in Australia draw heavily on the established global guidelines. These recommendations aim to assist clinicians to make informed, patient-centric choices when delivering treatment to people with PsA.
Collapse
Affiliation(s)
- Peter Nash
- School of Medicine, Griffith University, Brisbane, Queensland, Australia
- Rheumatology Research Unit, Sunshine Coast, Queensland, Australia
- Department of Medicine, University of Queensland, Brisbane, Queensland, Australia
| | - Daniel Sumpton
- Concord Repatriation General Hospital, Sydney, New South Wales, Australia
| | - Michelle Tellus
- St Vincent's Private Hospital, Melbourne, Victoria, Australia
| | | | - Paul Bird
- School of Clinical Medicine, University of New South Wales, Sydney, New South Wales, Australia
| | - Stephen Hall
- Melbourne Rheumatology, Cabrini Hospital, Melbourne, Victoria, Australia
- Monash University, Monash, Victoria, Australia
| |
Collapse
|
21
|
Picchianti Diamanti A, Panebianco C, Di Gioia V, Bellofatto IA, Salemi S, Di Rosa R, Sesti G, Nalli G, Salerno G, Finocchiaro E, Laganà B. A Case Series Report on the Effect of Tofacitinib on Joint Inflammation and Gut Microbiota Composition in Psoriatic Arthritis Patients Naive to Biologic Agents. Microorganisms 2024; 12:2387. [PMID: 39770590 PMCID: PMC11676042 DOI: 10.3390/microorganisms12122387] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2024] [Revised: 11/18/2024] [Accepted: 11/19/2024] [Indexed: 01/11/2025] Open
Abstract
INTRODUCTION Psoriatic arthritis (PsA) is a complex condition within the Spondyloarthritis (SpA) group. Recent studies have focused on the important role of the intestinal microbiota in maintaining immunological homeostasis, highlighting how intestinal dysbiosis may act as a trigger for autoimmune diseases. Tofacitinib is a Janus kinase inhibitor (JAK-i) with proven efficacy for the treatment of both rheumatoid arthritis and PsA. However, there is a lack of data on its ability to reduce joint remission through ultrasonography (US) and the effects it might have on the composition of the gut microbiota. METHODS Here, we present a case series of seven bio-naïve PsA patients who received tofacitinib treatment and were followed up for 12 months. The clinical response was assessed using validated scores (DAPSA, ASDAS, and BASDAI), laboratory tests, and US assessment of the target joint and enthesis. Finally, we evaluated changes in the composition of the intestinal microbiota using next-generation sequencing analysis of fecal samples. RESULTS The patients in the study showed a significant improvement in all clinical scores used; this improvement was also confirmed by a significant reduction in the US synovitis scores. The data on the microbiota analysis suggested that the effectiveness of tofacitinib in ameliorating PsA activity was associated with a relevant modification of some gut bacterial lineages. No cases of severe adverse effects were reported. CONCLUSIONS Treatment with tofacitinib proved to be effective, safe and capable of varying the composition of the gut microbiota by selecting bacterial strains considered beneficial in immune modulation.
Collapse
Affiliation(s)
- Andrea Picchianti Diamanti
- Department of Clinical and Molecular Medicine, S. Andrea University Hospital, “Sapienza” University of Rome, 00189 Rome, Italy; (V.D.G.); (S.S.); (R.D.R.); (G.S.); (G.N.); (G.S.); (B.L.)
| | - Concetta Panebianco
- Division of Gastroenterology, Fondazione IRCCS Casa Sollievo della Sofferenza Hospital, 71013 San Giovanni Rotondo, Italy;
| | - Valeria Di Gioia
- Department of Clinical and Molecular Medicine, S. Andrea University Hospital, “Sapienza” University of Rome, 00189 Rome, Italy; (V.D.G.); (S.S.); (R.D.R.); (G.S.); (G.N.); (G.S.); (B.L.)
| | - Ilaria Anna Bellofatto
- Department of Internal Medicine, University of Genoa, 6 Viale Benedetto XV, 16132 Genoa, Italy;
| | - Simonetta Salemi
- Department of Clinical and Molecular Medicine, S. Andrea University Hospital, “Sapienza” University of Rome, 00189 Rome, Italy; (V.D.G.); (S.S.); (R.D.R.); (G.S.); (G.N.); (G.S.); (B.L.)
| | - Roberta Di Rosa
- Department of Clinical and Molecular Medicine, S. Andrea University Hospital, “Sapienza” University of Rome, 00189 Rome, Italy; (V.D.G.); (S.S.); (R.D.R.); (G.S.); (G.N.); (G.S.); (B.L.)
| | - Giorgio Sesti
- Department of Clinical and Molecular Medicine, S. Andrea University Hospital, “Sapienza” University of Rome, 00189 Rome, Italy; (V.D.G.); (S.S.); (R.D.R.); (G.S.); (G.N.); (G.S.); (B.L.)
| | - Gabriele Nalli
- Department of Clinical and Molecular Medicine, S. Andrea University Hospital, “Sapienza” University of Rome, 00189 Rome, Italy; (V.D.G.); (S.S.); (R.D.R.); (G.S.); (G.N.); (G.S.); (B.L.)
| | - Gerardo Salerno
- Department of Clinical and Molecular Medicine, S. Andrea University Hospital, “Sapienza” University of Rome, 00189 Rome, Italy; (V.D.G.); (S.S.); (R.D.R.); (G.S.); (G.N.); (G.S.); (B.L.)
| | - Etta Finocchiaro
- Dietetic and Clinical Nutrition Unit, City of Health and Science University Hospital, 10126 Turin, Italy;
| | - Bruno Laganà
- Department of Clinical and Molecular Medicine, S. Andrea University Hospital, “Sapienza” University of Rome, 00189 Rome, Italy; (V.D.G.); (S.S.); (R.D.R.); (G.S.); (G.N.); (G.S.); (B.L.)
| |
Collapse
|
22
|
Kharouf F, Gladman DD. Advances in the management of psoriatic arthritis in adults. BMJ 2024; 387:e081860. [PMID: 39572047 DOI: 10.1136/bmj-2024-081860] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2024]
Abstract
Psoriatic arthritis is an inflammatory arthritis that affects around 30% of patients with psoriasis. The disease spectrum includes peripheral arthritis, enthesitis, tenosynovitis, dactylitis, axial involvement, and skin and nail psoriasis in most patients. In addition to the cutaneous and musculoskeletal manifestations, several comorbidities can complicate the disease course, including cardiovascular disease, diabetes mellitus, metabolic syndrome, gout, anxiety, and depression. The management of patients with psoriatic arthritis begins with a careful assessment of the skin and joints and screening for comorbidities. This review describes the assessment tools and outcome measures used in the evaluation of patients with psoriatic arthritis. It summarizes the approach to therapy, including non-medicinal interventions such as education, lifestyle changes, physiotherapy, and occupational therapy. It discusses the evidence on pharmacologic treatments, including drugs used for symptomatic relief such as non-steroidal anti-inflammatory drugs, and those used to control the disease process; this last group comprises conventional synthetic disease modifying anti-rheumatic drugs (DMARDs), including methotrexate, leflunomide, and sulfasalazine, and biologic and targeted DMARDs, including anti-tumor necrosis factor (TNFα), anti-interleukin-17 (IL-17), anti-IL-12/23, and anti-IL-23 agents, as well as Janus kinase (JAK) inhibitors and phosphodiesterase 4 (PDE4) antagonists. Although these drugs are usually tailored to the clinical profile of the patient, biomarkers predictive of response to therapy are needed so that a more personalized approach can be followed.
Collapse
Affiliation(s)
- Fadi Kharouf
- University of Toronto, Psoriatic Arthritis Program, University Health Network, Toronto Western Hospital, Toronto, ON, Canada
- Gladman-Krembil Psoriatic Disease Program, Toronto Western Hospital, Toronto, ON, Canada
| | - Dafna D Gladman
- University of Toronto, Psoriatic Arthritis Program, University Health Network, Toronto Western Hospital, Toronto, ON, Canada
- Gladman-Krembil Psoriatic Disease Program, Toronto Western Hospital, Toronto, ON, Canada
| |
Collapse
|
23
|
Xue S, Lin Y, Chen H, Yang Z, Zha J, Jiang X, Han Z, Wang K. Mechanisms of autophagy and their implications in dermatological disorders. Front Immunol 2024; 15:1486627. [PMID: 39559368 PMCID: PMC11570406 DOI: 10.3389/fimmu.2024.1486627] [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: 08/26/2024] [Accepted: 10/18/2024] [Indexed: 11/20/2024] Open
Abstract
Autophagy is a highly conserved cellular self-digestive process that underlies the maintenance of cellular homeostasis. Autophagy is classified into three types: macrophage, chaperone-mediated autophagy (CMA) and microphagy, which maintain cellular homeostasis through different mechanisms. Altered autophagy regulation affects the progression of various skin diseases, including psoriasis (PA), systemic lupus erythematosus (SLE), vitiligo, atopic dermatitis (AD), alopecia areata (AA) and systemic sclerosis (SSc). In this review, we review the existing literature focusing on three mechanisms of autophagy, namely macrophage, chaperone-mediated autophagy and microphagy, as well as the roles of autophagy in the above six dermatological disorders in order to aid in further studies in the future.
Collapse
Affiliation(s)
- Shenghao Xue
- School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Deyang Hospital Affiliated Hospital of Chengdu University of Traditional Chinese Medicine, Deyang, China
| | - Yumeng Lin
- Health Management Center, Nanjing Tongren Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Haoran Chen
- Chengdu Xinhua Hospital Affiliated to North Sichuan Medical College, Chengdu, China
| | - Zhengyu Yang
- School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Deyang Hospital Affiliated Hospital of Chengdu University of Traditional Chinese Medicine, Deyang, China
| | - Junting Zha
- Health Management Center, Nanjing Tongren Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Xuan Jiang
- School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Deyang Hospital Affiliated Hospital of Chengdu University of Traditional Chinese Medicine, Deyang, China
| | - Zhongyu Han
- Chengdu Xinhua Hospital Affiliated to North Sichuan Medical College, Chengdu, China
| | - Ke Wang
- School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Deyang Hospital Affiliated Hospital of Chengdu University of Traditional Chinese Medicine, Deyang, China
| |
Collapse
|
24
|
Cantini F, Marchesoni A, Novelli L, Gualberti G, Marando F, McDearmon-Blondell EL, Gao T, McGonagle D, Salvarani C. Effects of upadacitinib on enthesitis in patients with psoriatic arthritis: a post hoc analysis of SELECT-PsA 1 and 2 trials. Rheumatology (Oxford) 2024; 63:3146-3154. [PMID: 38331400 PMCID: PMC11534117 DOI: 10.1093/rheumatology/keae057] [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: 07/04/2023] [Revised: 12/13/2023] [Accepted: 01/10/2024] [Indexed: 02/10/2024] Open
Abstract
OBJECTIVES To characterize the effect of upadacitinib 15 mg once daily (UPA15) on enthesitis in patients with PsA from the SELECT-PsA Phase 3 trials. METHODS Patients with an inadequate response/intolerance to one or more non-biologic DMARD (SELECT-PsA 1) or one or more biologic DMARD (SELECT-PsA 2) received UPA15, adalimumab 40 mg every other week or placebo (weeks 0-24) switched to UPA15 (week 24 onward). The Leeds Enthesitis Index (LEI) and Spondyloarthritis Research Consortium of Canada (SPARCC) index were used to assess improvement in enthesitis, enthesitis resolution, maintenance of enthesitis resolution and protection from enthesitis development through week 56. RESULTS Data from 639 patients receiving UPA15 and 635 patients receiving placebo (including 317 patients who switched from placebo to UPA15) were analysed. UPA15 led to higher rates of enthesitis resolution vs placebo at week 24 (LEI: 59.8% vs 38.0%; SPARCC index: 50.6% vs 31.5%, respectively) and greater improvements in the LEI (-1.7 vs -1.0) and SPARCC index (-3.4 vs -1.9); improvements were maintained through week 56. Improvements were observed after 12 weeks of UPA15 treatment. Over 90% of patients without enthesitis (LEI = 0) at baseline receiving UPA15 were enthesitis-free at week 56, and UPA15 prevented recurrence of enthesitis at week 56 in >80% of patients with enthesitis at baseline who achieved resolution (LEI = 0) at week 24. CONCLUSIONS UPA15 is associated with a comprehensive improvement in enthesitis, with improvements observed after 12 weeks of treatment. Additionally, treatment with UPA15 was associated with maintaining an enthesitis-free state after resolution and protection against new-onset enthesitis. TRIAL REGISTRATION ClinicalTrials.gov identifiers: NCT03104400 (SELECT-PsA 1) and NCT03104374 (SELECT-PsA 2).
Collapse
Affiliation(s)
- Fabrizio Cantini
- Department of Rheumatology, Azienda USL Toscana Centro, Hospital of Prato, Prato, Italy
| | | | | | | | | | | | | | - Dennis McGonagle
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Leeds, UK
- Leeds Teaching Hospitals NHS Trust, NIHR Leeds Biomedical Research Centre, Leeds, UK
| | - Carlo Salvarani
- Unità Operativa di Reumatologia, Azienda USL-IRCCS di Reggio Emilia, Reggio Emilia, Italy
- Università di Modena e Reggio Emilia, Reggio Emilia, Italy
| |
Collapse
|
25
|
Kharouf F, Gladman DD. Treatment controversies in spondyloarthritis and psoriatic arthritis: focus on biologics and targeted therapies. Expert Rev Clin Immunol 2024; 20:1381-1400. [PMID: 39072530 DOI: 10.1080/1744666x.2024.2384705] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2024] [Accepted: 07/22/2024] [Indexed: 07/30/2024]
Abstract
INTRODUCTION There are several treatment controversies that have emerged in spondyloarthritis and psoriatic arthritis. These are related to the nature of the conditions as well as to the use of medications. AREAS COVERED This review, which included a search of PubMed database as well as the references within the articles provides an overview of the nature of spondyloarthritis, controversy over the inclusion of psoriatic arthritis (PsA) as a peripheral spondyloarthritis, and a summary of current treatments for both PsA and axial spondyloarthritis (axSpA), with special emphasis on targeted therapy. The review highlights the differences in response to certain medications, particularly biologic therapy and summarizes the randomized controlled trials in psoriatic arthritis and axial spondyloarthritis providing data about the responses in table format. EXPERT OPINION There is a need for better outcome measures in axSpA. Currently, the measures are subjective. Imaging may be more appropriate but there is a need for research into the reliability and responsiveness of imaging techniques. In PsA, there may also be better response measures and research into the reliability and responsiveness of available measures is underway. There is also a need for novel therapies as well as biomarkers for response in both diseases.
Collapse
Affiliation(s)
- Fadi Kharouf
- Division of Rheumatology, University Health Network, University of Toronto, Toronto, Ontario, Canada
- Gladman-Krembil Psoriatic Disease Program, Schroeder Arthritis Institute, Krembil Research Institute, Toronto Western Hospital, Toronto, Ontario, Canada
| | - Dafna D Gladman
- Division of Rheumatology, University Health Network, University of Toronto, Toronto, Ontario, Canada
- Gladman-Krembil Psoriatic Disease Program, Schroeder Arthritis Institute, Krembil Research Institute, Toronto Western Hospital, Toronto, Ontario, Canada
| |
Collapse
|
26
|
Yi RC, Gantz HY, Shah SC, Moran SK, Klionsky YE, Feldman SR. Pharmacotherapeutic management of psoriatic disease: addressing psoriatic arthritis and cutaneous manifestations. Expert Opin Pharmacother 2024; 25:2203-2212. [PMID: 39422251 DOI: 10.1080/14656566.2024.2419563] [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: 08/09/2024] [Revised: 09/27/2024] [Accepted: 10/17/2024] [Indexed: 10/19/2024]
Abstract
INTRODUCTION Psoriasis and psoriatic arthritis (PsA) are interrelated autoimmune conditions sharing similar genetic and immunological pathways. PsA often develops within 10 years of psoriasis onset, though it may precede cutaneous symptoms in some patients. Effective management of these conditions requires a multidisciplinary approach to address skin, bone, joint, and vascular manifestations. AREAS COVERED The review summarizes the current pharmacotherapies and to provide treatment guidelines for managing cutaneous psoriasis and PsA in psoriatic disease. EXPERT OPINION The management for mild psoriasis and mild PsA flare-ups can be addressed with topical treatments and with NSAIDs or intra-articular glucocorticoid injections. For more persistent or severe cases, systemic treatments with oral small molecules (Methotrexate, Apremilast, Janus kinase inhibitors) or with biologics (TNF inhibitors, IL-17 inhibitors, IL-23 inhibitors, CTLA-4 Ig) are effective in managing both psoriasis and PsA. With many treatment options, providers can tailor management, which considers patient disease severity, preference, comorbidities, and other factors. Early detection and a multidisciplinary management strategy can optimize patient quality of life and improve health outcomes.
Collapse
Affiliation(s)
- Robin C Yi
- Center for Dermatology Research, Department of Dermatology, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Hannah Y Gantz
- Center for Dermatology Research, Department of Dermatology, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Shailey C Shah
- Center for Dermatology Research, Department of Dermatology, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Shannon K Moran
- Center for Dermatology Research, Department of Dermatology, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Yael E Klionsky
- Department of Internal Medicine, Section of Rheumatology, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Steven R Feldman
- Center for Dermatology Research, Department of Dermatology, Wake Forest University School of Medicine, Winston-Salem, NC, USA
- Department of Pathology, Wake Forest University School of Medicine, Winston-Salem, NC, USA
- Department of Social Sciences & Health Policy, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| |
Collapse
|
27
|
Shi JLH, Chuang WY, Tam LS. JAK Inhibitors and Cardiovascular Disease in Psoriatic Arthritis: Friends or Foe? Int J Rheum Dis 2024; 27:e15419. [PMID: 39555659 DOI: 10.1111/1756-185x.15419] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 09/23/2024] [Accepted: 11/05/2024] [Indexed: 11/19/2024]
Affiliation(s)
- Jenny Lin-Hong Shi
- JC School of Public Health and Primary Care, The Prince of Wales Hospital, The Chinese University of Hong Kong, New Territories, Hong Kong
| | - Wei-Yuan Chuang
- Department of Internal Medicine, Chung-Shan Medical University Hospital, School of Medicine, Chung-Shan Medical University, Taichung, Taiwan
| | - Lai-Shan Tam
- Department of Medicine & Therapeutics, The Prince of Wales Hospital, The Chinese University of Hong Kong, New Territories, Hong Kong
| |
Collapse
|
28
|
Vassilopoulos A, Thomas K, Vassilopoulos D. Infections in psoriatic arthritis: association with treatment. Ther Adv Musculoskelet Dis 2024; 16:1759720X241289201. [PMID: 39429971 PMCID: PMC11487508 DOI: 10.1177/1759720x241289201] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2024] [Accepted: 07/30/2024] [Indexed: 10/22/2024] Open
Abstract
Serious infections (SIs) remain one of the most significant comorbidities in patients with inflammatory arthritides including psoriatic arthritis (PsA). Apart from methotrexate (MTX) and biologics such as tumor necrosis factor (TNFi), interleukin-12/23 (IL-12/23i), and IL-17 inhibitors (IL-17i), traditionally used for the treatment of PsA, recently biologics such as IL-23i and targeted synthetic agents like JAK inhibitors (JAKi) have been introduced in the daily clinical practice for the treatment of this disease. Although overall the incidence of SIs in patients with PsA treated with these agents is lower compared to patients with rheumatoid arthritis, still a number of unresolved issues regarding their safety remain. Current evidence is reassuring regarding the safety profile of conventional synthetic disease-modifying anti-rheumatic drugs, such as MTX. The increased risk for reactivation of latent infections, such as tuberculosis and hepatitis B virus (HBV) with the use of TNFi, is well described; nevertheless, it is significantly ameliorated with the appropriate screening and prophylaxis. Regarding IL-12/23i and IL-17i, there are no significant safety signals, except from an increased incidence of usually mild Candida infections with the latter class. Newer biologics such as IL-23i and targeted synthetic agents like JAKi have been recently introduced in the daily clinical practice for the treatment of this disease. While IL-23i has not been shown to increase the risk for common or opportunistic infections, a well-established association of JAKi with herpes zoster warrants the attention of rheumatologists. In this narrative review, we summarize the infectious complications of available treatment options by drug class in patients with PsA.
Collapse
Affiliation(s)
- Athanasios Vassilopoulos
- Division of Internal Medicine, Rhode Island Hospital, Warren Alpert Medical School of Brown University, Providence, RI, USA
| | - Konstantinos Thomas
- Fourth Department of Medicine, National and Kapodistrian University of Athens School of Medicine, Attikon University General Hospital, Athens, Greece
| | - Dimitrios Vassilopoulos
- Joint Rheumatology Program, Clinical Immunology–Rheumatology Unit, Second Department of Medicine and Laboratory, National and Kapodistrian University of Athens School of Medicine, General Hospital of Athens Hippokration, 114 Vass. Sophias Avenue, Athens 115 27, Greece
| |
Collapse
|
29
|
Hu T, Liu CH, Lei M, Zeng Q, Li L, Tang H, Zhang N. Metabolic regulation of the immune system in health and diseases: mechanisms and interventions. Signal Transduct Target Ther 2024; 9:268. [PMID: 39379377 PMCID: PMC11461632 DOI: 10.1038/s41392-024-01954-6] [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: 03/27/2024] [Revised: 07/18/2024] [Accepted: 08/11/2024] [Indexed: 10/10/2024] Open
Abstract
Metabolism, including glycolysis, oxidative phosphorylation, fatty acid oxidation, and other metabolic pathways, impacts the phenotypes and functions of immune cells. The metabolic regulation of the immune system is important in the pathogenesis and progression of numerous diseases, such as cancers, autoimmune diseases and metabolic diseases. The concept of immunometabolism was introduced over a decade ago to elucidate the intricate interplay between metabolism and immunity. The definition of immunometabolism has expanded from chronic low-grade inflammation in metabolic diseases to metabolic reprogramming of immune cells in various diseases. With immunometabolism being proposed and developed, the metabolic regulation of the immune system can be gradually summarized and becomes more and more clearer. In the context of many diseases including cancer, autoimmune diseases, metabolic diseases, and many other disease, metabolic reprogramming occurs in immune cells inducing proinflammatory or anti-inflammatory effects. The phenotypic and functional changes of immune cells caused by metabolic regulation further affect and development of diseases. Based on experimental results, targeting cellular metabolism of immune cells becomes a promising therapy. In this review, we focus on immune cells to introduce their metabolic pathways and metabolic reprogramming, and summarize how these metabolic pathways affect immune effects in the context of diseases. We thoroughly explore targets and treatments based on immunometabolism in existing studies. The challenges of translating experimental results into clinical applications in the field of immunometabolism are also summarized. We believe that a better understanding of immune regulation in health and diseases will improve the management of most diseases.
Collapse
Affiliation(s)
- Tengyue Hu
- West China School of clinical medical, West China Second University Hospital, Sichuan University, Chengdu, China
- Center of Infectious Diseases, West China Hospital of Sichuan University, Chengdu, China
- Laboratory of Infectious and Liver Diseases, Institution of Infectious Diseases, West China Hospital, Sichuan University, Chengdu, China
| | - Chang-Hai Liu
- West China School of clinical medical, West China Second University Hospital, Sichuan University, Chengdu, China
- Center of Infectious Diseases, West China Hospital of Sichuan University, Chengdu, China
- Laboratory of Infectious and Liver Diseases, Institution of Infectious Diseases, West China Hospital, Sichuan University, Chengdu, China
| | - Min Lei
- West China School of clinical medical, West China Second University Hospital, Sichuan University, Chengdu, China
- National Center for Birth Defect Monitoring, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, China
- State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Qingmin Zeng
- West China School of clinical medical, West China Second University Hospital, Sichuan University, Chengdu, China
- Center of Infectious Diseases, West China Hospital of Sichuan University, Chengdu, China
- Laboratory of Infectious and Liver Diseases, Institution of Infectious Diseases, West China Hospital, Sichuan University, Chengdu, China
| | - Li Li
- Division of Renal and endocrinology, Qin Huang Hospital, Xi'an, China
| | - Hong Tang
- Center of Infectious Diseases, West China Hospital of Sichuan University, Chengdu, China.
- Laboratory of Infectious and Liver Diseases, Institution of Infectious Diseases, West China Hospital, Sichuan University, Chengdu, China.
- State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy, West China Second University Hospital, Sichuan University, Chengdu, China.
| | - Nannan Zhang
- West China School of clinical medical, West China Second University Hospital, Sichuan University, Chengdu, China.
- National Center for Birth Defect Monitoring, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, China.
- State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy, West China Second University Hospital, Sichuan University, Chengdu, China.
| |
Collapse
|
30
|
Mease PJ, Papademetriou E, Potluri R, Agarwal E, Cappelleri JC, Ling YL. Adherence, Persistence, Healthcare Resource Use, and Costs in Tofacitinib-Treated Patients with Psoriatic Arthritis: Data from Two United States Claims Databases. Adv Ther 2024; 41:3850-3867. [PMID: 39143312 PMCID: PMC11399223 DOI: 10.1007/s12325-024-02904-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Accepted: 05/15/2024] [Indexed: 08/16/2024]
Abstract
INTRODUCTION Associations between increased functional disability and higher healthcare resource utilization (HCRU) and costs were reported in patients with psoriatic arthritis (PsA). We assessed characteristics/outcomes of patients with PsA receiving tofacitinib monotherapy vs combination therapy with conventional synthetic disease-modifying antirheumatic drugs. METHODS Claims data from Optum® Clinformatics® Data Mart (OC) and Merative™ MarketScan® (MS) databases between December 2017 and February 2020 were analyzed. Outcomes assessed were adherence/persistence by therapy type (monotherapy/combination therapy); HCRU/costs (per patient per month) by periods on-treatment (sum time on tofacitinib) and off-treatment (sum time off tofacitinib [gap of > 60 days]) plus therapy type. RESULTS This analysis included 274 and 395 tofacitinib-treated patients in OC (70.4% female, mean age 54.4 years) and MS (68.9% female, mean age 51.4 years), respectively. Percentages of patients with a proportion of days covered ≥ 0.8 at 12 months for monotherapy vs combination therapy were OC, 44.5% vs 53.8%; MS, 36.4% vs 45.7%. Generally similar trends were seen over 24 months and for medication possession ratio ≥ 0.8. Median (95% confidence interval) times to treatment discontinuation for monotherapy vs combination therapy were OC, 10.1 (7.4-11.8) vs 16.7 (8.3-26.6) months; MS, 6.9 (5.6-9.4) vs 11.0 (6.1-13.9) months. During off-treatment vs on-treatment periods, numerical decreases were observed for all-cause (OC, $5383 vs $6149; MS, $4145 vs $5180) and PsA-related costs (OC, $3237 vs $4515; MS, $2703 vs $3907) regardless of therapy type. During off-treatment vs on-treatment periods, numerical increases in outpatient visits for all-cause (OC, 2.37 vs 2.05; MS, 2.15 vs 1.99) and PsA-related visits (OC, 0.60 vs 0.46; MS, 0.47 vs 0.44) were observed, and PsA-related medications numerically decreased (OC, 1.21 vs 1.53; MS, 1.05 vs 1.48). CONCLUSION In this USA-based claims analysis, tofacitinib adherence was numerically lower for patients with PsA receiving monotherapy vs combination therapy. Costs numerically decreased off-treatment vs on-treatment, irrespective of therapy type, driven by lower medication costs.
Collapse
Affiliation(s)
- Philip J Mease
- Swedish Medical Center/Providence St Joseph Health and University of Washington, Seattle, WA, USA.
- Seattle Rheumatology Associates, 601 Broadway, Suite 600, Seattle, WA, 98122, USA.
| | | | | | | | | | | |
Collapse
|
31
|
Goupille P, Carvajal Alegria G, Verhoeven F, Wendling D. Treatment with Targeted Therapy in Patients with Psoriatic Arthritis and Inadequate Response to Methotrexate: Proposal for a Rational Strategy. Rheumatol Ther 2024; 11:1065-1079. [PMID: 39134832 PMCID: PMC11422401 DOI: 10.1007/s40744-024-00704-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Accepted: 07/18/2024] [Indexed: 09/25/2024] Open
Abstract
INTRODUCTION The therapeutic arsenal for psoriatic arthritis (PsA) is gradually being expanded, but the use of these targeted treatments must be optimal. Our objective was to guide the choice of targeted therapy to use as first-line treatment in a patient with PsA in whom methotrexate (MTX) has failed. METHODS We searched for literature data in PubMed with the appropriate keywords for the six points of our argument: (1) the tolerance of MTX; (2) the efficacy of targeted therapies combined with MTX vs monotherapy; (3) immunogenicity of anti-tumor necrosis alpha (TNFα) monoclonal antibodies (mAbs); (4) immunogenicity of anti-interleukin (IL)-17, anti-IL-12/23, and anti-IL-23 mAbs; (5) the therapeutic maintenance of anti-TNFα mAbs when combined or not with MTX; (6) the therapeutic maintenance of anti-IL-17 vs anti-TNFα mAbs as first-line targeted therapy. RESULTS The proposed rational strategy is as follows: in case of initiation of an anti-TNFα agent, maintaining treatment with MTX seems preferable, even in the absence of evidence of the superior efficacy of the combination, to avoid immunization and reduced therapeutic maintenance; in case of initiation of anti-IL-17, anti-IL-12/23, anti-IL-23 agents, or Janus kinase (JAK) inhibitors, again in the absence of evidence of the superior efficacy of the combination, discontinuing MTX therapy may be possible, at least in two steps, after verifying the efficacy of the targeted therapy initiated on the joints and skin. CONCLUSION We have data from the literature to guide the choice of targeted therapy to use as first-line treatment in a patient with PsA in whom MTX has failed.
Collapse
Affiliation(s)
- Philippe Goupille
- Rheumatology Department, CHU de Tours, UPR CNRS 4301 CBM, NMNS, University of Tours, 37044, Tours Cedex 9, France.
| | - Guillermo Carvajal Alegria
- Rheumatology Department, CHU de Tours, UPR CNRS 4301 CBM, NMNS, University of Tours, 37044, Tours Cedex 9, France
| | - Frank Verhoeven
- Rheumatology Department, CHU de Besançon, University of Franche-Comté, Besançon, France
| | - Daniel Wendling
- Rheumatology Department, CHU de Besançon, University of Franche-Comté, Besançon, France
| |
Collapse
|
32
|
Proft F, Duran TI, Ghoreschi K, Pleyer U, Siegmund B, Poddubnyy D. Treatment strategies for Spondyloarthritis: Implementation of precision medicine - Or "one size fits all" concept? Autoimmun Rev 2024; 23:103638. [PMID: 39276959 DOI: 10.1016/j.autrev.2024.103638] [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: 07/02/2024] [Revised: 09/05/2024] [Accepted: 09/07/2024] [Indexed: 09/17/2024]
Abstract
Spondyloarthritis (SpA) is a term to describe a group of chronic inflammatory rheumatic diseases, which have common pathophysiological, genetic, and clinical features. Under the umbrella term SpA, two main groups are subsumed: axial SpA (radiographic axSpA and non-radiographic axSpA) and peripheral SpA (with the leading representative being psoriatic arthritis (PsA) but also arthritis associated with inflammatory bowel disease (IBD), reactive arthritis, and undifferentiated pSpA). The key clinical symptom in axSpA is chronic back pain, typically with inflammatory characteristics, which starts in early adulthood, while the leading clinical manifestations of peripheral SpA (pSpA) are arthritis, enthesitis, and/or dactylitis. Furthermore, extra-musculoskeletal manifestations (EMMs) (acute anterior uveitis, psoriasis, and IBD) can accompany axial or peripheral symptoms. All these factors need to be taken into account when making treatment decisions in SpA patients. Despite the major advances in the treatment landscape over the past two decades with the introduction of biological disease-modifying anti-rheumatic drugs (bDMARDs) and most recently targeted synthetic DMARDs (tsDMARDs), a relevant proportion of patients still does not achieve the desired state of remission (=absence of disease activity). With this implementation of new treatment modalities, clinicians now have more choices to make in the treatment algorithms. However, despite generalized treatment recommendations, all factors need to be carefully considered when deciding on the optimal treatment strategy for an individual patient in clinical practice, aiming at an important first step towards personalized treatment strategies in SpA. In this narrative review, we focus on the efficacy of approved and emerging treatment options in axSpA and PsA as the main representative of pSpA and discuss their selective effect on the different manifestations associated with SpA to provide guidance on drivers of treatment decisions in specific situations.
Collapse
Affiliation(s)
- Fabian Proft
- Department of Gastroenterology, Infectiology and Rheumatology (including Nutrition Medicine), Charité - Universitätsmedizin Berlin, Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany.
| | - Tugba Izci Duran
- Department of Gastroenterology, Infectiology and Rheumatology (including Nutrition Medicine), Charité - Universitätsmedizin Berlin, Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany; Clinic of Rheumatology, Denizli State Hospital, Denizli, Turkey
| | - Kamran Ghoreschi
- Department of Dermatology, Venereology and Allergology, Charité - Universitätsmedizin Berlin, Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Uwe Pleyer
- Department of Ophthalmology Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin; Berlin, Germany and (5)Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany
| | - Britta Siegmund
- Department of Gastroenterology, Infectiology and Rheumatology (including Nutrition Medicine), Charité - Universitätsmedizin Berlin, Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Denis Poddubnyy
- Department of Gastroenterology, Infectiology and Rheumatology (including Nutrition Medicine), Charité - Universitätsmedizin Berlin, Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany; Epidemiology unit, German Rheumatism Research Centre, Berlin, Germany; Division of Rheumatology, Department of Medicine, University Health Network and University of Toronto, Toronto, Canada
| |
Collapse
|
33
|
Solimani F, Ghoreschi K. [Janus kinase inhibitors for skin disorders]. DERMATOLOGIE (HEIDELBERG, GERMANY) 2024; 75:781-790. [PMID: 39212722 DOI: 10.1007/s00105-024-05406-8] [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: 07/22/2024] [Indexed: 09/04/2024]
Abstract
Immune factors such as interferon‑ɣ and interleukin 4 belong to the group of cytokines that are dependent on type I/II receptors for their signal transmission. Upon activation, these receptors transmit their signal to the cell nucleus and, thus, modulate gene transcription via a signaling cascade consisting of Janus kinases (JAK). This family of four kinases (JAK 1, JAK 2, JAK 3, and tyrosine kinase 2 (TYK2)) subsequently activate members of the signal transducer and activator of transcription (STAT). This finding turned the JAK/STAT signaling pathway into a pharmacological target for the treatment of inflammatory diseases in which cytokines using type I/II receptors play a pathogenic role. In 2018, the European Medicines Agency (EMA) approved tofacitinib for the treatment of psoriatic arthritis. This was the first approval of a JAK/STAT pathway inhibitor for patients treated by dermatologists and rheumatologists. Since then, several new JAK inhibitors have been approved for dermatologic diseases such as atopic dermatitis, alopecia areata, vitiligo, and plaque-type psoriasis. In addition, JAK inhibitors are being investigated for the treatment of many other skin diseases. Thus, systemic JAK inhibitors complete the spectrum of immunotherapeutics with a broader immunological approach compared to monoclonal antibodies. The low molecular weight of JAK inhibitors enables the preparation of these drugs for both systemic and topical administration. Their utilization could represent a valuable alternative to topical steroids. The safety profile of JAK inhibitors must be taken into account. Possible long-term effects may become apparent in the next few years. This article describes both approved JAK inhibitors and relevant new JAK inhibitors that are promising candidates for approval as therapeutics in dermatology.
Collapse
Affiliation(s)
- Farzan Solimani
- Department of Dermatology, Venereology and Allergology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Luisenstr. 2, 10117, Berlin, Deutschland.
- BIH Biomedical Innovation Academy, BIH Charité Clinician Scientist Program, Berlin Institute of Health at Charité-Universitätsmedizin Berlin, Berlin, Deutschland.
| | - Kamran Ghoreschi
- Department of Dermatology, Venereology and Allergology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Luisenstr. 2, 10117, Berlin, Deutschland
| |
Collapse
|
34
|
Liao W, Armstrong AW, Duffin KC. GRAPPA Debate: Targeted Small Molecules Versus Biologics as First-Line Systemic Therapy After Conventional Therapy for Moderate-to-Severe Psoriasis. J Rheumatol 2024; 51:89-92. [PMID: 39009402 DOI: 10.3899/jrheum.2024-0293] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/17/2024] [Indexed: 07/17/2024]
Abstract
In this debate at the Group for Research and Assessment of Psoriasis and Psoriatic Arthritis (GRAPPA) 2023 annual meeting, arguments were made contrasting the first-line use of oral targeted small-molecule drugs vs biologic therapy for the treatment of moderate-to-severe psoriasis (PsO) after failure of conventional therapy. Arguments in favor of small-molecule drugs included good efficacy and safety, patient preference, cost savings, global health equity, and environmental stewardship. Arguments in favor of biologics included superior efficacy, excellent safety, availability of long-term data, pediatric regulatory approvals, and potential benefit for comorbidities. By the end of the debate, there was recognition of significant pros and cons of each approach. Both small-molecule drugs and biologic therapy are valuable options for PsO treatment, and their use can be tailored toward specific individuals or healthcare systems.
Collapse
Affiliation(s)
- Wilson Liao
- W. Liao, MD, Department of Dermatology, University of California San Francisco, San Francisco, California;
| | - April W Armstrong
- A.W. Armstrong, MD, MPH, Division of Dermatology, University of California Los Angeles, Los Angeles, California
| | - Kristina Callis Duffin
- K. Callis Duffin, MD, MS, Department of Dermatology, Spencer Fox Eccles School of Medicine, University of Utah, Salt Lake City, Utah, USA
| |
Collapse
|
35
|
Skouvaklidou E, Avgerou P, Vassilakis KD, Fragoulis GE, Kougkas N. Monotherapy or combination therapy in PsA: current aspects. Ther Adv Musculoskelet Dis 2024; 16:1759720X241274055. [PMID: 39314822 PMCID: PMC11418363 DOI: 10.1177/1759720x241274055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Accepted: 07/19/2024] [Indexed: 09/25/2024] Open
Abstract
Psoriatic arthritis (PsA) is an immune-mediated inflammatory disease with heterogeneity regarding its clinical features, mainly affecting the skin and the musculoskeletal system; additionally, extra-musculoskeletal manifestations and comorbidities are common, adding complexity to its treatment. In the last decades, a plethora of therapeutic options have been available, including conventional synthetic disease-modifying anti-rheumatic drugs (DMARDs), biological DMARDs (bDMARDs), and targeted synthetic DMARDs (tsDMARDs), and many recommendations have been published regarding the proper use of them in patients with PsA. In rheumatoid arthritis, the combination of conventional with bDMARDs or tsDMARDs is a common and recommended practice, whereas in PsA there is scarce data about the benefit of this combination. This review summarizes all the available data from randomized clinical trials, observational studies, and registries about the value of this therapeutic strategy.
Collapse
Affiliation(s)
- Elpida Skouvaklidou
- Department of Rheumatology, Fourth Department of Internal Medicine, Hippokration Hospital, Aristoteleio Panepistemio Thessalonikes, Thessaloniki, Greece
| | - Paraskevi Avgerou
- Department of Rheumatology, Fourth Department of Internal Medicine, Hippokration Hospital, Aristoteleio Panepistemio Thessalonikes, Thessaloniki, Greece
| | - Konstantinos D. Vassilakis
- Joint Academic Rheumatology Program, First Department of Propaedeutic and Internal Medicine, Laiko General Hospital, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - George E. Fragoulis
- Joint Academic Rheumatology Program, First Department of Propaedeutic and Internal Medicine, Laiko General Hospital, School of Medicine, National and Kapodistrian University of Athens, 17 Agiou Thoma Street, Athens 10679, Greece
- Institute of Infection, Immunity & Inflammation, University of Glasgow Institute of Infection Immunity and Inflammation, Glasgow, UK
| | - Nikolaos Kougkas
- Department of Rheumatology, Fourth Department of Internal Medicine, Hippokration Hospital, Aristoteleio Panepistemio Thessalonikes, Thessaloniki, Greece
| |
Collapse
|
36
|
Dey M, Nikiphorou E. Cardiovascular comorbidities in psoriatic arthritis: state of the art. Ther Adv Musculoskelet Dis 2024; 16:1759720X241274537. [PMID: 39290781 PMCID: PMC11406594 DOI: 10.1177/1759720x241274537] [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: 02/07/2024] [Accepted: 07/10/2024] [Indexed: 09/19/2024] Open
Abstract
Psoriatic arthritis (PsA) is a complex multi-system immune-mediated condition, characterised by a high comorbidity burden, one of the most prevalent of which is cardiovascular disease (CVD), affecting up to 80% of patients. This narrative review explores the current understanding of cardiovascular comorbidities in PsA, focusing on mechanistic pathways, risk assessment, and the impact of treatment choices on cardiovascular health. Here, we outline the role of inflammatory cytokines, immune system dysregulation, and genetic predispositions in PsA, not only as drivers of musculoskeletal manifestations but also atherosclerosis and endothelial dysfunction, giving rise to cardiovascular pathology. Given these insights, accurately assessing and predicting cardiovascular risk in PsA patients is a critical challenge. This review evaluates traditional risk calculators as well as innovative biomarkers and imaging techniques, emphasising their utility and limitations in capturing the true cardiovascular risk profile of PsA patients. There are multiple complexities surrounding the treatment of PsA in the context of concurrent CVD, and therapeutic choices must carefully balance efficacy in managing PsA symptoms with the potential cardiovascular implications. A multidisciplinary approach, integrating dermatological, rheumatological, and cardiological perspectives, amongst others, to optimise patient outcomes, is key. Overall, a heightened clinical awareness and research focus on cardiovascular comorbidities in PsA is warranted, aiming to refine risk assessment strategies and therapeutic interventions that holistically address the multifaceted needs of patients with PsA.
Collapse
Affiliation(s)
- Mrinalini Dey
- Centre for Rheumatic Diseases, King's College London, Weston Education Centre, London, UK
| | - Elena Nikiphorou
- Centre for Rheumatic Diseases, King's College London, Weston Education Centre, Cutcombe Road, London SE5 9RJ, UK
- Rheumatology Department, King's College Hospital, London, UK
| |
Collapse
|
37
|
Hoisnard L, Meyer A, Dray-Spira R, Weill A, Zureik M, Sbidian E. Risk of Gastrointestinal Perforation in Patients With Rheumatic Diseases Exposed to Janus Kinase Inhibitors Versus Adalimumab: A Nationwide Cohort Study. Arthritis Rheumatol 2024; 76:1364-1376. [PMID: 38699822 DOI: 10.1002/art.42862] [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: 08/10/2023] [Revised: 03/20/2024] [Accepted: 04/11/2024] [Indexed: 05/05/2024]
Abstract
OBJECTIVE To compare the risk of gastrointestinal perforation (GIP), a rare but serious adverse event, in patients who a JAK inhibitor (JAKi; tofacitinib, baricitinib, upadacitinib, or filgotinib) versus adalimumab (tumor necrosis factor inhibitor) among a comprehensive real-world population of patients with rheumatic diseases. METHODS We conducted a nationwide population-based cohort study of the French national health data system, the exposed group that received a JAKi and the comparison group adalimumab. We included all individuals with a rheumatic disease who had their first dispensation of these treatments from July 2017 to December 2021. The primary endpoint was the occurrence of GIP (end of follow-up May 2022). Weighted hazard ratios (wHRs) were estimated with the inverse probability of treatment weighting method to account for confounding factors. Concomitant administration of systemic glucocorticoids, nonsteroidal anti-inflammatory drugs, and proton-pump inhibitors were time-varying variables. RESULTS The cohort included 39,758 patients: 12,335 and 27,423 in the groups that received a JAKi and adalimumab (mean age 58.2 and 47.3 years; female 76% and 58%; rheumatoid arthritis 85.3% and 27.3%, and psoriatic arthritis/axial spondyloarthritis 14.7% and 72.7%), respectively. During follow-up, 38 and 42 GIPs occurred in the groups that received a JAKi and adalimumab groups; incidence rates were 2.1 (95% confidence interval [CI] 1.5-2.8) and 1.1 (95% CI 0.8-1.5) per 1,000 person-years, respectively. Rates of GIP did not differ between the groups that received a JAKi and adalimumab: wHR 1.1 (95% CI 0.7-1.9; P = 0.65). Despite the lack of power in some subgroup analyses, results were consistent whatever the subgroup of a type of JAKi received or subgroup with a type of rheumatic disease. CONCLUSION In this nationwide cohort study, the rates of GIPs did not differ between groups of patients who received JAKi and adalimumab treatment. These results need to be confirmed in other observational studies.
Collapse
Affiliation(s)
- Lea Hoisnard
- Henri Mondor Hospital, INSERM, and Paris Est Créteil University, Créteil, France
| | - Antoine Meyer
- French National Agency for Medicines and Health Products Safety, Saint-Denis, France, and Bicêtre University Hospital, Paris-Saclay University, Le Kremlin Bicêtre, France
| | - Rosemary Dray-Spira
- French National Agency for Medicines and Health Products Safety, Saint-Denis, France
| | - Alain Weill
- French National Agency for Medicines and Health Products Safety, Saint-Denis, France
| | - Mahmoud Zureik
- French National Agency for Medicines and Health Products Safety, Saint-Denis, France
| | - Emilie Sbidian
- Henri Mondor Hospital, Créteil, France, INSERM, Créteil, France, Paris Est Créteil University, Créteil, France, French National Agency for Medicines and Health Products Safety, Saint-Denis, France, Le Kremlin Bicêtre, France
| |
Collapse
|
38
|
Taxonera C, Carpio López D, Cabez Manas A, Hinojosa Del Val JE. Clinical settings with tofacitinib in ulcerative colitis. REVISTA ESPANOLA DE ENFERMEDADES DIGESTIVAS 2024; 116:484-492. [PMID: 35373565 DOI: 10.17235/reed.2022.8660/2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
There are aspects of Janus kinase (JAK) inhibitors, specifically tofacitinib, that distinguish them from other drugs used in the treatment of ulcerative colitis (UC), such as their oral administration, their short half-life and their lack of immunogenicity. With the available evidence, we can highlight tofacitinib's quick action and flexibility of use, and its efficacy in patients, irrespective of whether or not they have previously been exposed to TNF inhibitors (anti-TNF drugs) and other biologic agents. Moreover, their safety profile is known and manageable, with certain considerations and precautions being factored in before and during treatment. In this review, we have defined various scenarios pertaining to this drug, e.g. its use in the event of failure or intolerance to previous treatment with biologics, when a quick response is required or in patients with other concurrent immune-mediated diseases.
Collapse
Affiliation(s)
- Carlos Taxonera
- Aparato Digestivo, Hospital Clínico Universitario San Carlos, España
| | - Daniel Carpio López
- Gastroenterología, Complexo Hospitalario Universitario de Pontevedra, España
| | | | | |
Collapse
|
39
|
Hen O, Harrison SR, De Marco G, Marzo-Ortega H. Early psoriatic arthritis: when is the right time to start advanced therapy? Ther Adv Musculoskelet Dis 2024; 16:1759720X241266727. [PMID: 39071239 PMCID: PMC11283661 DOI: 10.1177/1759720x241266727] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Accepted: 06/20/2024] [Indexed: 07/30/2024] Open
Abstract
Despite significant advances in the treatment of psoriatic arthritis (PsA) in the last two decades, remission remains elusive and there is no cure. Evidence from rheumatoid arthritis (RA) confirming enhanced response and outcome from earlier treatment intervention suggests the plausibility of the window of opportunity in the pathogenesis of RA. Yet, data are lacking in PsA. Although treatment response may be enhanced in shorter disease duration, it is unknown how this early intervention may impact long-term outcomes. Furthermore, it remains to be demonstrated whether there is a best treatment strategy and time of intervention. Crucially, the main hurdle when aiming for early treatment intervention is the ability to achieve a timely diagnosis that highlights the need to focus research efforts on characterizing the very early disease stages including the transition to PsA in the at-risk psoriasis population.
Collapse
Affiliation(s)
- Or Hen
- NIHR Leeds Biomedical Research Centre, The Leeds Teaching Hospitals NHS Trust, Leeds
- Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Leeds, UK
- Department of Medicine ‘C’, Sheba Medical Center, Tel HaShomer, Ramat Gan, Israel
| | - Stephanie R. Harrison
- NIHR Leeds Biomedical Research Centre, The Leeds Teaching Hospitals NHS Trust, Leeds
- Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Leeds, UK
- Stephanie R. Harrison is also affiliated to Leeds Institute of Data Analytics, University of Leeds, Leeds
| | - Gabriele De Marco
- NIHR Leeds Biomedical Research Centre, The Leeds Teaching Hospitals NHS Trust, Leeds
- Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Leeds, UK
- Stephanie R. Harrison is also affiliated to Leeds Institute of Data Analytics, University of Leeds, Leeds
| | - Helena Marzo-Ortega
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, Second Floor, Chapel Allerton Hospital, Chapeltown Road, Leeds LS7 4SA, UK
| |
Collapse
|
40
|
Ando T, Abe Y, Yamaji K, Nishikomori R, Tamura N. A case of cryopyrin-associated periodic syndrome due to somatic mosaic mutation complicated with recurrent circinate erythematous psoriasis. Mod Rheumatol Case Rep 2024; 8:368-372. [PMID: 38036300 DOI: 10.1093/mrcr/rxad067] [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: 10/03/2023] [Revised: 11/13/2023] [Accepted: 11/22/2023] [Indexed: 12/02/2023]
Abstract
Cryopyrin-associated periotic syndrome (CAPS) is a rare autoinflammatory disease caused by genetic variants in innate immunity genes. Autoinflammatory diseases, including CAPS, mediate proinflammatory cytokines such as interleukin (IL)-1 and IL-18 and result in severe systemic inflammation. A gain-of-function mutation in the NLR family pyrin domain-containing 3 (NLRP3) gene, which encodes the protein cryopyrin, was identified to be responsible for CAPS in 2001, and since then several additional pathogenic mutations have been found. Moreover, other phenotypes have been identified based on severity and symptomatology, including familial cold autoinflammatory syndrome, Muckle-Wells syndrome, and neonatal-onset multisystem inflammatory disease/chronic neurologic cutaneous articular syndrome. Prompt diagnosis of CAPS remains challenging, however, due to unspecific, extensive clinical signs, and delayed diagnosis and treatment targeting IL-1 lead to multiorgan damage. Another factor complicating diagnosis is the existence of somatic mosaic mutations in the NLRP3 gene in some cases, resulting in symptoms and clinical courses that are atypical. The frequency of somatic mosaic mutations in CAPS was estimated to be 19% in a systematic review. Psoriasis is a chronic inflammatory skin disease that affects ∼3% of the global population. Although no reports have shown complication between CAPS and psoriasis, these diseases have several similarities and potential relationships, for instance activation of T helper 17 cells in the dermis and increased NLRP3 gene expression in psoriatic skin compared with normal skin. Here, we report a case of CAPS due to a somatic mosaic mutation with recurrent circinate erythematous psoriasis.
Collapse
Affiliation(s)
- Taiki Ando
- Department of Internal Medicine and Rheumatology, Juntendo University School of Medicine, Tokyo, Japan
| | - Yoshiyuki Abe
- Department of Internal Medicine and Rheumatology, Juntendo University School of Medicine, Tokyo, Japan
| | - Ken Yamaji
- Department of Internal Medicine and Rheumatology, Juntendo University School of Medicine, Tokyo, Japan
| | - Ryuta Nishikomori
- The Department of Pediatrics and Child Health, Kurume University School of Medicine, Fukuoka, Japan
| | - Naoto Tamura
- Department of Internal Medicine and Rheumatology, Juntendo University School of Medicine, Tokyo, Japan
| |
Collapse
|
41
|
Braña I, Loredo M, Pardo E, Burger S, Fernández-Bretón E, Queiro R. Patients With Psoriatic Arthritis-Related Enthesitis and Persistence on Tofacitinib Under Real-World Conditions. J Rheumatol 2024; 51:682-686. [PMID: 38561189 DOI: 10.3899/jrheum.2024-0016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/19/2024] [Indexed: 04/04/2024]
Abstract
OBJECTIVE Information on the persistence of tofacitinib (TOF) in psoriatic arthritis (PsA) is scarce in real-world conditions. Our objective was to analyze the persistence and safety of TOF under these conditions. METHODS This was a single-center retrospective longitudinal observational study of all patients with PsA who received at least 1 dose of TOF. The main focus was on adverse events (AEs) and drug survival. Drug survival was analyzed by Kaplan-Meier curves and persistence explanatory factors by multivariate Cox regression models. The hazard ratio (HR) was used to measure association. RESULTS Seventy-two patients were included, 54 women and 18 men, mean age 51.9 (SD 11.1) years, mean disease duration of 10.4 (SD 6.99) years. TOF was ≥ third line of therapy in > 70% of cases. The median survival was 13.0 (IQR 5.3-29.0) months. One-year retention rate was 52.7% (95% CI 42.4-65.6). TOF survival was not influenced by sex, disease duration, comorbidities, or line of treatment. Younger patients (HR 0.96, P = 0.01) and those with enthesitis (HR 0.37, P = 0.03) showed lower odds of drug discontinuation. The overall rate of AEs was 52.9 (95% CI 38.5-70.6)/100 person-years. Most AEs occurred during the first 6 months of exposure. CONCLUSION In this real-world study, TOF showed a reasonably good retention rate in a PsA population that was mostly refractory to biologic and oral targeted synthetic disease-modifying antirheumatic drugs. There were no new causes for concern regarding safety. Patients with refractory PsA and enthesitis might be a specific target population for this drug.
Collapse
Affiliation(s)
- Ignacio Braña
- I. Braña, MD, M. Loredo, MD, E. Pardo, MD, S. Burger, MD, Rheumatology Division, Hospital Universitario Central de Asturias
| | - Marta Loredo
- I. Braña, MD, M. Loredo, MD, E. Pardo, MD, S. Burger, MD, Rheumatology Division, Hospital Universitario Central de Asturias
| | - Estefanía Pardo
- I. Braña, MD, M. Loredo, MD, E. Pardo, MD, S. Burger, MD, Rheumatology Division, Hospital Universitario Central de Asturias
| | - Stefanie Burger
- I. Braña, MD, M. Loredo, MD, E. Pardo, MD, S. Burger, MD, Rheumatology Division, Hospital Universitario Central de Asturias
| | - Eva Fernández-Bretón
- E. Fernández-Bretón, MD, Biostatistics and Epidemiology Platform, Health Research Institute of Asturias
| | - Rubén Queiro
- R. Queiro, MD, PhD, Rheumatology Division, Hospital Universitario Central de Asturias, and Oviedo University School of Medicine, and ISPA Translational Immunology Division, Oviedo-Asturias, Spain.
| |
Collapse
|
42
|
Ruscitti P, Allanore Y, Baldini C, Barilaro G, Bartoloni Bocci E, Bearzi P, Bellis E, Berardicurti O, Biaggi A, Bombardieri M, Cantarini L, Cantatore FP, Caporali R, Caso F, Cervera R, Ciccia F, Cipriani P, Chatzis L, Colafrancesco S, Conti F, Corberi E, Costa L, Currado D, Cutolo M, D'Angelo S, Del Galdo F, Di Cola I, Di Donato S, Distler O, D'Onofrio B, Doria A, Fautrel B, Fasano S, Feist E, Fisher BA, Gabini M, Gandolfo S, Gatto M, Genovali I, Gerli R, Grembiale RD, Guggino G, Hoffmann-Vold AM, Iagnocco A, Iaquinta FS, Liakouli V, Manoussakis MN, Marino A, Mauro D, Montecucco C, Mosca M, Naty S, Navarini L, Occhialini D, Orefice V, Perosa F, Perricone C, Pilato A, Pitzalis C, Pontarini E, Prete M, Priori R, Rivellese F, Sarzi-Puttini P, Scarpa R, Sebastiani G, Selmi C, Shoenfeld Y, Triolo G, Trunfio F, Yan Q, Tzioufas AG, Giacomelli R. Tailoring the treatment of inflammatory rheumatic diseases by a better stratification and characterization of the clinical patient heterogeneity. Findings from a systematic literature review and experts' consensus. Autoimmun Rev 2024; 23:103581. [PMID: 39069240 DOI: 10.1016/j.autrev.2024.103581] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2024] [Accepted: 07/23/2024] [Indexed: 07/30/2024]
Abstract
Inflammatory rheumatic diseases are different pathologic conditions associated with a deregulated immune response, codified along a spectrum of disorders, with autoinflammatory and autoimmune diseases as two-end phenotypes of this continuum. Despite pathogenic differences, inflammatory rheumatic diseases are commonly managed with a limited number of immunosuppressive drugs, sometimes with partial evidence or transferring physicians' knowledge in different patients. In addition, several randomized clinical trials, enrolling these patients, did not meet the primary pre-established outcomes and these findings could be linked to the underlying molecular diversities along the spectrum of inflammatory rheumatic disorders. In fact, the resulting patient heterogeneity may be driven by differences in underlying molecular pathology also resulting in variable responses to immunosuppressive drugs. Thus, the identification of different clinical subsets may possibly overcome the major obstacles that limit the development more effective therapeutic strategies for these patients with inflammatory rheumatic diseases. This clinical heterogeneity could require a diverse therapeutic management to improve patient outcomes and increase the frequency of clinical remission. Therefore, the importance of better patient stratification and characterization is increasingly pointed out according to the precision medicine principles, also suggesting a new approach for disease treatment. In fact, based on a better proposed patient profiling, clinicians could more appropriately balance the therapeutic management. On these bases, we synthetized and discussed the available literature about the patient profiling in regard to therapy in the context of inflammatory rheumatic diseases, mainly focusing on randomized clinical trials. We provided an overview of the importance of a better stratification and characterization of the clinical heterogeneity of patients with inflammatory rheumatic diseases identifying this point as crucial in improving the management of these patients.
Collapse
Affiliation(s)
- Piero Ruscitti
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy.
| | - Yannick Allanore
- Rheumatology Department, Cochin Hospital, APHP, INSERM U1016, Université Paris Cité, Paris, France
| | - Chiara Baldini
- Rheumatology Unit, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Giuseppe Barilaro
- Department of Autoimmune Diseases, Reference Centre for Systemic Autoimmune Diseases, Vasculitis and Autoinflammatory Diseases of the Catalan and Spanish Health Systems, Member of ERN-ReCONNET/RITA, Hospital Clínic, University of Barcelona, Barcelona, Catalonia, Spain
| | - Elena Bartoloni Bocci
- Section of Rheumatology, Department of Medicine and Surgery, University of Perugia, Italy
| | - Pietro Bearzi
- Rheumatology and Clinical Immunology, Department of Medicine, School of Medicine, University of Rome "Campus Bio-Medico", 00128 Rome, Italy; Clinical and Research Section of Rheumatology and Clinical Immunology, Fondazione Policlinico Universitario Campus Bio-Medico, 00128 Rome, Italy
| | - Elisa Bellis
- Academic Rheumatology Centre, Dipartimento di Scienze Cliniche e Biologiche Università di Torino - AO Mauriziano di Torino, Turin, Italy
| | - Onorina Berardicurti
- Rheumatology and Clinical Immunology, Department of Medicine, School of Medicine, University of Rome "Campus Bio-Medico", 00128 Rome, Italy; Clinical and Research Section of Rheumatology and Clinical Immunology, Fondazione Policlinico Universitario Campus Bio-Medico, 00128 Rome, Italy
| | - Alice Biaggi
- Rheumatology and Clinical Immunology, Department of Medicine, School of Medicine, University of Rome "Campus Bio-Medico", 00128 Rome, Italy; Clinical and Research Section of Rheumatology and Clinical Immunology, Fondazione Policlinico Universitario Campus Bio-Medico, 00128 Rome, Italy
| | - Michele Bombardieri
- Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, Queen Mary University of London and Barts NIHR BRC & NHS Trust & National Institute for Health and Care Research (NIHR) Barts Biomedical Research Centre (BRC), London, UK
| | - Luca Cantarini
- Department of Medical Sciences, Surgery and Neurosciences, Research Center of Systemic Autoinflammatory Diseases and Behçet's Disease Clinic, University of Siena, Siena, Italy; Azienda Ospedaliero-Universitaria Senese [European Reference Network (ERN) for Rare Immunodeficiency, Autoinflammatory and Autoimmune Diseases (RITA) Center] Siena, Italy
| | - Francesco Paolo Cantatore
- Rheumatology Clinic, Department of Medical and Surgical Sciences, University of Foggia, 71122 Foggia, Italy
| | - Roberto Caporali
- Department of Clinical Sciences and Community Health, University of Milan, Paediatric Rheumatology Unit, and Clinical Rheumatology Unit, ASST Pini-CTO, Milan, Italy
| | - Francesco Caso
- Rheumatology Research Unit, Department of Clinical Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Ricard Cervera
- Department of Autoimmune Diseases, Reference Centre for Systemic Autoimmune Diseases, Vasculitis and Autoinflammatory Diseases of the Catalan and Spanish Health Systems, Member of ERN-ReCONNET/RITA, Hospital Clínic, University of Barcelona, Barcelona, Catalonia, Spain
| | - Francesco Ciccia
- Rheumatology Unit, Department of Precision Medicine, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Paola Cipriani
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| | - Loukas Chatzis
- Department of Pathophysiology, School of Medicine, National and Kapodistrian University of Athens, Greece
| | - Serena Colafrancesco
- Department of Internal Medicine and Medical Specialties, Rheumatology Unit, Sapienza University of Rome, Viale del Policlinico 155, 00185 Rome, Italy
| | - Fabrizio Conti
- Department of Internal Medicine and Medical Specialties, Rheumatology Unit, Sapienza University of Rome, Viale del Policlinico 155, 00185 Rome, Italy
| | - Erika Corberi
- Rheumatology and Clinical Immunology, Department of Medicine, School of Medicine, University of Rome "Campus Bio-Medico", 00128 Rome, Italy; Clinical and Research Section of Rheumatology and Clinical Immunology, Fondazione Policlinico Universitario Campus Bio-Medico, 00128 Rome, Italy
| | - Luisa Costa
- Rheumatology Research Unit, Department of Clinical Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Damiano Currado
- Rheumatology and Clinical Immunology, Department of Medicine, School of Medicine, University of Rome "Campus Bio-Medico", 00128 Rome, Italy; Clinical and Research Section of Rheumatology and Clinical Immunology, Fondazione Policlinico Universitario Campus Bio-Medico, 00128 Rome, Italy
| | - Maurizio Cutolo
- Laboratory of Experimental Rheumatology and Academic Division of Rheumatology, Department of Internal Medicine and Specialties, University of Genova Italy, IRCCS Polyclinic Hospital, Genova, Italy
| | - Salvatore D'Angelo
- Rheumatology Depatment of Lucania, San Carlo Hospital of Potenza and Madonna delle Grazie Hospital of Matera, Potenza, Italy
| | - Francesco Del Galdo
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Leeds, UK; NIHR Leeds Biomedical Research Centre, Leeds Teaching Hospitals NHS Trust, Leeds, UK
| | - Ilenia Di Cola
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| | - Stefano Di Donato
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Leeds, UK; NIHR Leeds Biomedical Research Centre, Leeds Teaching Hospitals NHS Trust, Leeds, UK
| | - Oliver Distler
- Department of Rheumatology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Bernardo D'Onofrio
- Department of Internal Medicine and Therapeutics, Università di Pavia, Division of Rheumatology, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Andrea Doria
- Rheumatology Unit, Department of Medicine (DIMED), University of Padua, Padua, Italy
| | - Bruno Fautrel
- Sorbonne Université - Assistance Publique Hôpitaux de Paris, INSERM UMRS 1136, Hôpital de La Pitié Salpêtrière, Paris, France
| | - Serena Fasano
- Rheumatology Unit, Department of Precision Medicine, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Eugen Feist
- Department of Rheumatology, Helios Fachklinik, Sophie-von-Boetticher-Straße 1, 39245, Vogelsang-Gommern, Germany; Charité - Universitätsmedizin Berlin, Medizinische Klinik mit Schwerpunkt Rheumatologie und Klinische Immunologie, Berlin, Germany
| | - Benjamin A Fisher
- Institute of Inflammation and Ageing, University Hospitals Birmingham, Birmingham, UK; Department of Rheumatology, National Institute for Health Research (NIHR), Birmingham Biomedical Research Centre, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
| | - Marco Gabini
- Rheumatology Unit, Santo Spirito Hospital, Pescara, Italy
| | - Saviana Gandolfo
- Unit of Rheumatology, San Giovanni Bosco Hospital, Naples, Italy
| | - Mariele Gatto
- Academic Rheumatology Centre, Dipartimento di Scienze Cliniche e Biologiche Università di Torino - AO Mauriziano di Torino, Turin, Italy
| | - Irene Genovali
- Rheumatology and Clinical Immunology, Department of Medicine, School of Medicine, University of Rome "Campus Bio-Medico", 00128 Rome, Italy; Clinical and Research Section of Rheumatology and Clinical Immunology, Fondazione Policlinico Universitario Campus Bio-Medico, 00128 Rome, Italy
| | - Roberto Gerli
- Section of Rheumatology, Department of Medicine and Surgery, University of Perugia, Italy
| | - Rosa Daniela Grembiale
- Rheumatology Research Unit, Dipartimento di Scienze della Salute, Università degli studi "Magna Graecia" di Catanzaro, Catanzaro, Italy
| | - Giuliana Guggino
- Rheumatology Unit, Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, University of Palermo, Italy
| | - Anna Maria Hoffmann-Vold
- Department of Rheumatology, University Hospital Zurich, University of Zurich, Zurich, Switzerland; Department of Rheumatology, Oslo University Hospital-Rikshospitalet, Oslo, Norway
| | - Annamaria Iagnocco
- Academic Rheumatology Centre, Dipartimento di Scienze Cliniche e Biologiche Università di Torino - AO Mauriziano di Torino, Turin, Italy
| | - Francesco Salvatore Iaquinta
- Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, Queen Mary University of London and Barts NIHR BRC & NHS Trust & National Institute for Health and Care Research (NIHR) Barts Biomedical Research Centre (BRC), London, UK
| | - Vasiliki Liakouli
- Rheumatology Unit, Department of Precision Medicine, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Menelaos N Manoussakis
- Department of Pathophysiology, School of Medicine, National and Kapodistrian University of Athens, Greece
| | - Annalisa Marino
- Rheumatology and Clinical Immunology, Department of Medicine, School of Medicine, University of Rome "Campus Bio-Medico", 00128 Rome, Italy; Clinical and Research Section of Rheumatology and Clinical Immunology, Fondazione Policlinico Universitario Campus Bio-Medico, 00128 Rome, Italy
| | - Daniele Mauro
- Rheumatology Unit, Department of Precision Medicine, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Carlomaurizio Montecucco
- Department of Internal Medicine and Therapeutics, Università di Pavia, Division of Rheumatology, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Marta Mosca
- Rheumatology Unit, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Saverio Naty
- Department of Health Sciences, "Magna Græcia" University of Catanzaro, 88100 Catanzaro, Italy
| | - Luca Navarini
- Rheumatology and Clinical Immunology, Department of Medicine, School of Medicine, University of Rome "Campus Bio-Medico", 00128 Rome, Italy; Clinical and Research Section of Rheumatology and Clinical Immunology, Fondazione Policlinico Universitario Campus Bio-Medico, 00128 Rome, Italy
| | - Daniele Occhialini
- Rheumatic and Systemic Autoimmune Diseases Unit, Department of Interdisciplinary Medicine (DIM), University of Bari Medical School, Italy
| | - Valeria Orefice
- Rheumatology Unit, San Camillo-Forlanini Hospital, Rome, Italy
| | - Federico Perosa
- Rheumatic and Systemic Autoimmune Diseases Unit, Department of Interdisciplinary Medicine (DIM), University of Bari Medical School, Italy
| | - Carlo Perricone
- Section of Rheumatology, Department of Medicine and Surgery, University of Perugia, Italy
| | - Andrea Pilato
- Rheumatology and Clinical Immunology, Department of Medicine, School of Medicine, University of Rome "Campus Bio-Medico", 00128 Rome, Italy; Clinical and Research Section of Rheumatology and Clinical Immunology, Fondazione Policlinico Universitario Campus Bio-Medico, 00128 Rome, Italy
| | - Costantino Pitzalis
- Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, Queen Mary University of London and Barts NIHR BRC & NHS Trust & National Institute for Health and Care Research (NIHR) Barts Biomedical Research Centre (BRC), London, UK; Department of Biomedical Sciences, Humanitas University, Milan, Italy
| | - Elena Pontarini
- Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, Queen Mary University of London and Barts NIHR BRC & NHS Trust & National Institute for Health and Care Research (NIHR) Barts Biomedical Research Centre (BRC), London, UK
| | - Marcella Prete
- Rheumatic and Systemic Autoimmune Diseases Unit, Department of Interdisciplinary Medicine (DIM), University of Bari Medical School, Italy
| | - Roberta Priori
- Department of Internal Medicine and Medical Specialties, Rheumatology Unit, Sapienza University of Rome, Viale del Policlinico 155, 00185 Rome, Italy
| | - Felice Rivellese
- Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, Queen Mary University of London and Barts NIHR BRC & NHS Trust & National Institute for Health and Care Research (NIHR) Barts Biomedical Research Centre (BRC), London, UK
| | - Piercarlo Sarzi-Puttini
- Rheumatology Department, ASST Fatebenefratelli Luigi Sacco University Hospital, Milan, Italy; Department of Clinical Sciences and Community Health, Università degli Studi di Milano, Milan, Italy
| | - Raffaele Scarpa
- Rheumatology Research Unit, Department of Clinical Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | | | - Carlo Selmi
- Department of Biomedical Sciences, Humanitas University, Milan, Italy
| | - Yehuda Shoenfeld
- Zabludovwicz autoimmunity center, Sheba medical center, Tel Hashomer Israel, Reichman University, Herzeliya, Israel
| | - Giovanni Triolo
- Rheumatology Unit, Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, University of Palermo, Italy
| | - Francesca Trunfio
- Rheumatology and Clinical Immunology, Department of Medicine, School of Medicine, University of Rome "Campus Bio-Medico", 00128 Rome, Italy; Clinical and Research Section of Rheumatology and Clinical Immunology, Fondazione Policlinico Universitario Campus Bio-Medico, 00128 Rome, Italy
| | - Qingran Yan
- Department of Rheumatology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Athanasios G Tzioufas
- Department of Pathophysiology, School of Medicine, National and Kapodistrian University of Athens, Greece
| | - Roberto Giacomelli
- Rheumatology and Clinical Immunology, Department of Medicine, School of Medicine, University of Rome "Campus Bio-Medico", 00128 Rome, Italy; Clinical and Research Section of Rheumatology and Clinical Immunology, Fondazione Policlinico Universitario Campus Bio-Medico, 00128 Rome, Italy
| |
Collapse
|
43
|
Bachelez H, Griffiths CEM, Papp KA, Hall S, Merola JF, Feldman SR, Khraishi M, Tan H, Fallon L, Cappelleri JC, Bushmakin AG, Young P. Tofacitinib efficacy, patient-reported outcomes and safety in patients with psoriasis and a medical history of psoriatic arthritis: Pooled analysis of two Phase III studies. J Eur Acad Dermatol Venereol 2024; 38:e557-e561. [PMID: 38213065 DOI: 10.1111/jdv.19701] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Accepted: 11/22/2023] [Indexed: 01/13/2024]
Affiliation(s)
- H Bachelez
- Department of Dermatology, AP-HP Hôpital St. Louis, Paris, France
- Unité INSERM U1163, Imagine Institute, Université de Paris, Paris, France
| | - C E M Griffiths
- Dermatology Centre, Salford Royal Hospital, NIHR Manchester Biomedical Research Centre, University of Manchester, Manchester, UK
- Department of Dermatology, King's College Hospital, King's College London, London, UK
| | - K A Papp
- Probity Medical Research and Alliance Clinical Trials Inc, Waterloo, Ontario, Canada
- University of Toronto, Toronto, Ontario, Canada
| | - S Hall
- Emeritus Research, Malvern, Melbourne, Australia
| | - J F Merola
- Harvard Medical School, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - S R Feldman
- Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA
| | - M Khraishi
- Memorial University of Newfoundland, St John's, Newfoundland, Canada
| | - H Tan
- Pfizer Inc, Groton, Connecticut, USA
| | - L Fallon
- Pfizer Inc, Kirkland, Quebec, Canada
| | | | | | - P Young
- Pfizer Inc, Collegeville, Pennsylvania, USA
| |
Collapse
|
44
|
Singer D, Thompson-Leduc P, Ma S, Gupta D, Cheng WY, Sendhil SR, Sundar M, Hagopian E, Stempniewicz N, Duh MS, Poston S. Burden of Herpes Zoster Among Patients with Psoriatic Arthritis in the United States. PSORIASIS (AUCKLAND, N.Z.) 2024; 14:63-78. [PMID: 38939905 PMCID: PMC11208162 DOI: 10.2147/ptt.s430151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Accepted: 12/13/2023] [Indexed: 06/29/2024]
Abstract
Purpose Patients with psoriasis (PsO) and psoriatic arthritis (PsA) are at increased risk of herpes zoster (HZ), but healthcare resource use (HRU) and costs relating to HZ in adults with PsA are unknown. We aimed to estimate the incidence of HZ among adults with PsA vs without psoriatic disease and the additional HRU and costs among patients with PsA with vs without HZ. Patients and Methods This retrospective, longitudinal, cohort study estimated HZ incidence in PsA+ vs PsO-/PsA- cohorts and HRU and medical/pharmacy costs among PsA+/HZ+ vs PsA+/HZ- cohorts comprised of adults from Optum's de-identified Clinformatics Data Mart Database during 2015-2020. For the HRU/cost analyses, index was the date of first HZ diagnosis (PsA+/HZ+ cohort) or was randomly assigned (PsA+/HZ- cohort). Generalized linear models were used for adjusted comparisons between cohorts. Results HZ incidence was higher in the PsA+ (n = 57,126) vs PsO-/PsA- (n = 23,837,237) cohort (14.85 vs 7.67 per 1000 person-years; adjusted incidence rate ratio [aIRR]: 1.23; 95% confidence interval [CI]: 1.16-1.30). Numbers of outpatient visits, emergency department visits, and inpatient admissions were significantly higher in the PsA+/HZ+ (n = 1045) vs PsA+/HZ- (n = 36,091) cohorts during the first month after HZ diagnosis (outpatient: aIRR: 1.74; 95% CI: 1.63-1.86; emergency department: 3.14; 95% CI: 2.46-4.02; inpatient: aIRR: 2.61; 95% CI: 1.89-3.61). Mean all-cause per-patient costs were significantly higher in the PsA+/HZ+ vs PsA+/HZ- cohorts during the first month after index ($6493 vs $4521; adjusted cost difference: $2012; 95% CI: $1204-$3007). HRU and costs were numerically higher in the PsA+/HZ+ cohort during the first 3 and 12 months. Conclusion These findings, which provide evidence on the increased incidence and HRU and economic burden associated with HZ among adults with PsA, could be used to inform clinical practice and decision-making.
Collapse
Affiliation(s)
- David Singer
- GSK, US Health Economics and Outcomes Research, Philadelphia, PA, USA
| | | | - Siyu Ma
- GSK, US Health Economics and Outcomes Research, Philadelphia, PA, USA
- Tufts Medical Center, Boston, MA, USA
| | - Deepshekhar Gupta
- Analysis Group, Inc., Health Economics and Outcomes Research, Menlo Park, CA, USA
| | - Wendy Y Cheng
- Analysis Group, Inc., Health Economics and Outcomes Research, Boston, MA, USA
| | - Selvam R Sendhil
- Analysis Group, Inc., Health Economics and Outcomes Research, Los Angeles, CA, USA
| | - Manasvi Sundar
- Analysis Group, Inc., Health Economics and Outcomes Research, Los Angeles, CA, USA
| | - Ella Hagopian
- Analysis Group, Inc., Health Economics and Outcomes Research, Boston, MA, USA
| | | | - Mei Sheng Duh
- Analysis Group, Inc., Health Economics and Outcomes Research, Boston, MA, USA
| | - Sara Poston
- GSK, US Health Economics and Outcomes Research, Philadelphia, PA, USA
| |
Collapse
|
45
|
Goswami RP, Sinha D, Chatterjee M, Bhadu D, Das S. Comparative Effectiveness of Tofacitinib and Adalimumab in Axial Spondyloarthritis: A Real-World Clinical Context Multicenter Study. J Clin Rheumatol 2024; 30:e108-e114. [PMID: 38509045 DOI: 10.1097/rhu.0000000000002069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/22/2024]
Abstract
INTRODUCTION Tofacitinib, an oral Janus kinase inhibitor, is a putative choice in the treatment of axial spondyloarthritis (AxSpA). The objective of this study was to compare the effectiveness and tolerability of tofacitinib with adalimumab, in AxSpA, in a real-world clinical setting. METHODS In this multicentric medical records review study, adult patients with active AxSpA treated with either tofacitinib 5 mg twice daily or adalimumab 40 mg subcutaneously fortnightly were recruited. Effectiveness was measured with Ankylosing Spondylitis Disease Activity Score (ASDAS) and Bath Ankylosing Spondylitis Disease Activity Index (BASDAI). Drug-cost analysis was calculated with Incremental Cost-Effectiveness Ratio (ICER drug ). RESULTS Among the 266 patients, 135 were treated with tofacitinib and 131 with adalimumab (follow-up: 6.5 ± 1.6 months). Mean improvement of BASDAI (3.39 ± 0.09 vs. 3.14 ± 1.16, respectively) and that of ASDAS (1.78 ± 0.68 vs. 2.07 ± 2.08, respectively) were comparable between the adalimumab and tofacitinib groups. A higher proportion of patients achieved BASDAI50 response in the second (49.5% vs. 31.6%) and fourth month (83.9% vs. 62.8%) and ASDAS low disease activity in the fourth month (71.6% vs. 47.9%) in the adalimumab group. All disease activity measurements were similar by the sixth month in both groups. A higher proportion of patients in the tofacitinib group than in the adalimumab group required change in therapy (14.8% vs. 7.6%, respectively). ICER drug for adalimumab compared with tofacitinib was US $188.8 per patient in the adalimumab group for each person-month with BASDAI <4. CONCLUSIONS Tofacitinib showed comparable effectiveness with adalimumab in patients with AxSpA at the sixth month, despite lesser response in the initial months, with favorable ICER drug .
Collapse
Affiliation(s)
- Rudra Prosad Goswami
- From the Department of Rheumatology, All India Institute of Medical Sciences (AIIMS), New Delhi
| | - Debanjali Sinha
- Department of Rheumatology, Institute of Neurosciences Kolkata (I-NK)
| | | | - Danveer Bhadu
- From the Department of Rheumatology, All India Institute of Medical Sciences (AIIMS), New Delhi
| | - Shyamashis Das
- Department of Rheumatology, Institute of Neurosciences Kolkata (I-NK)
| |
Collapse
|
46
|
Kristensen LE, Deodhar A, Leung YY, Vranic I, Mortezavi M, Fallon L, Yndestad A, Kinch CD, Gladman DD. Risk Stratification of Patients with Psoriatic Arthritis and Ankylosing Spondylitis for Treatment with Tofacitinib: A Review of Current Clinical Data. Rheumatol Ther 2024; 11:487-499. [PMID: 38696034 PMCID: PMC11111604 DOI: 10.1007/s40744-024-00662-5] [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: 12/22/2023] [Accepted: 03/08/2024] [Indexed: 05/23/2024] Open
Abstract
In this commentary, we review clinical data which helps inform individualized benefit-risk assessment for tofacitinib in patients with psoriatic arthritis (PsA) and ankylosing spondylitis (AS). ORAL Surveillance, a safety trial of patients ≥ 50 years of age with rheumatoid arthritis (RA) and cardiovascular risk factors, found increased rates of safety outcomes (including major adverse cardiovascular events [MACE], malignancies excluding non-melanoma skin cancer, and venous thromboembolism) with tofacitinib versus tumor necrosis factor inhibitors (TNFi). Post hoc analyses of ORAL Surveillance have identified subpopulations with different relative risk versus TNFi; higher risk with tofacitinib was confined to patients ≥ 65 years of age and/or long-time current/past smokers, and specifically for MACE, patients with a history of atherosclerotic cardiovascular disease (ASCVD). In patients without these risk factors, risk differences between tofacitinib and TNFi could not be detected. Given differences in demographics, pathophysiology, and comorbidities, we sought to examine whether the risk stratification observed in RA is also appropriate for PsA and AS. Data from the PsA tofacitinib development program show low absolute risk of safety outcomes in patients < 65 years of age and never smokers, and low MACE risk in patients with no history of ASCVD, consistent with results from ORAL Surveillance. No MACE, malignancies, or venous thromboembolism were reported in the tofacitinib AS development program. The mechanism of the ORAL Surveillance safety findings is unknown, and there are no similar prospective studies of sufficient size and duration. Accordingly, it is appropriate to use a precautionary approach and extrapolate differentiating risk factors identified from ORAL Surveillance (age ≥ 65 years, long-time current/past smoking, and history of ASCVD) to PsA and AS. We recommend an individualized approach to treatment decisions based on these readily identifiable risk factors, in line with updated labeling for Janus kinase inhibitors and international guidelines for the treatment of PsA and AS.Trial Registration: NCT02092467, NCT01262118, NCT01484561, NCT00147498, NCT00413660, NCT00550446, NCT00603512, NCT00687193, NCT01164579, NCT00976599, NCT01059864, NCT01359150, NCT02147587, NCT00960440, NCT00847613, NCT00814307, NCT00856544, NCT00853385, NCT01039688, NCT02281552, NCT02187055, NCT02831855, NCT00413699, NCT00661661, NCT01877668, NCT01882439, NCT01976364, NCT00678210, NCT01710046, NCT01241591, NCT01186744, NCT01276639, NCT01309737, NCT01163253, NCT01786668, NCT03502616.
Collapse
Affiliation(s)
- Lars Erik Kristensen
- The Parker Institute, Bispebjerg and Frederiksberg, Copenhagen, Denmark.
- Copenhagen University, Copenhagen, Denmark.
| | - Atul Deodhar
- Division of Arthritis and Rheumatic Diseases, Oregon Health and Science University, Portland, OR, USA
| | - Ying-Ying Leung
- Department of Rheumatology and Immunology, Singapore General Hospital, Singapore, Singapore
| | | | | | | | | | | | - Dafna D Gladman
- Schroeder Arthritis Institute, Krembil Research Institute, Toronto Western Hospital, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
47
|
Dragotto M, D’Onghia M, Trovato E, Tognetti L, Rubegni P, Calabrese L. Therapeutic Potential of Targeting the JAK/STAT Pathway in Psoriasis: Focus on TYK2 Inhibition. J Clin Med 2024; 13:3091. [PMID: 38892802 PMCID: PMC11172692 DOI: 10.3390/jcm13113091] [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: 03/22/2024] [Revised: 05/17/2024] [Accepted: 05/23/2024] [Indexed: 06/21/2024] Open
Abstract
Psoriasis is an inflammatory skin disease with a chronic relapsing course and an often-detrimental impact on patients' quality of life. Thanks to incredible advances in research over the past few decades, the therapeutic armamentarium of psoriasis is now reasonably broad and structured, with several therapeutic agents that have demonstrated successful long-term control of this condition. However, there are still unfulfilled gaps resulting from the inherent limitations of existing therapies, which have paved the way for the identification of new therapeutic strategies or the improvement of existing ones. A great deal of attention has recently been paid to the JAK/STAT pathway, playing a crucial role in chronic inflammatory skin diseases, including psoriasis. Indeed, in a disease with such a complex pathogenesis, the possibility to antagonize multiple molecular pathways via JAK/STAT inhibition offers an undeniable therapeutic advantage. However, data from clinical trials evaluating the use of oral JAK inhibitors in immune-mediated disorders, such as RA, have arisen safety concerns, suggesting a potentially increased risk of class-specific AEs such as infections, venous thromboembolism, and malignancies. New molecules are currently under investigation for the treatment of psoriasis, such as deucravacitinib, an oral selective inhibitor that binds to the regulatory domain of TYK2, brepocitinib (PF-06700841) and PF-06826647 that bind to the active site in the catalytic domain. Due to the selective TYK2 blockade allowing the inhibition of key cytokine-mediated signals, such as those induced by IL-12 and IL-23, anti-TYK2 agents appear to be very promising as the safety profile seems to be superior compared with pan-JAK inhibitors. The aim of our review is to thoroughly explore the rationale behind the usage of JAK inhibitors in PsO, their efficacy and safety profiles, with a special focus on oral TYK2 inhibitors, as well as to provide a forward-looking update on novel therapeutic strategies targeting the TYK2 pathway in psoriasis.
Collapse
Affiliation(s)
- Martina Dragotto
- Dermatology Unit, Department of Medical, Surgical and Neurological Sciences, University of Siena, 53100 Siena, Italy (E.T.); (P.R.)
| | - Martina D’Onghia
- Dermatology Unit, Department of Medical, Surgical and Neurological Sciences, University of Siena, 53100 Siena, Italy (E.T.); (P.R.)
| | - Emanuele Trovato
- Dermatology Unit, Department of Medical, Surgical and Neurological Sciences, University of Siena, 53100 Siena, Italy (E.T.); (P.R.)
| | - Linda Tognetti
- Dermatology Unit, Department of Medical, Surgical and Neurological Sciences, University of Siena, 53100 Siena, Italy (E.T.); (P.R.)
| | - Pietro Rubegni
- Dermatology Unit, Department of Medical, Surgical and Neurological Sciences, University of Siena, 53100 Siena, Italy (E.T.); (P.R.)
| | - Laura Calabrese
- Dermatology Unit, Department of Medical, Surgical and Neurological Sciences, University of Siena, 53100 Siena, Italy (E.T.); (P.R.)
- Institute of Dermatology, Catholic University of the Sacred Heart, 00168 Rome, Italy
| |
Collapse
|
48
|
Bizzarri F, Ruiz-Villaverde R, Morales-Garrido P, Ruiz-Carrascosa JC, Cebolla-Verdugo M, Prados-Carmona A, Rodriguez-Troncoso M, Raya-Alvarez E. JAKinhibs in Psoriatic Disease: Analysis of the Efficacy/Safety Profile in Daily Clinical Practice. Diagnostics (Basel) 2024; 14:988. [PMID: 38786286 PMCID: PMC11119658 DOI: 10.3390/diagnostics14100988] [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: 03/03/2024] [Revised: 04/28/2024] [Accepted: 05/02/2024] [Indexed: 05/25/2024] Open
Abstract
Psoriatic disease (PsD) affects multiple clinical domains and causes a significant inflammatory burden in patients, requiring comprehensive evaluation and treatment. In recent years, new molecules such as JAK inhibitors (JAKinhibs) have been developed. These have very clear advantages: they act quickly, have a beneficial effect on pain, are well tolerated and the administration route is oral. Despite all this, there is still little scientific evidence in daily clinical practice. This observational, retrospective, single-center study was carried out in patients diagnosed with PsA in the last two years, who started treatment with Tofacitinib or Upadacitinib due to failure of a DMARD. The data of 32 patients were analyzed, and the majority of them (75%) started treatment with Tofacitinib. Most had moderate arthritis activity and mild psoriasis involvement according to activity indices. Both Tofacitinib and Upadacitinib demonstrated significant efficacy, with rapid and statistically significant improvement in joint and skin activity indices, C-reactive protein reduction, and objective measures of disease activity such as the number of painful and inflamed joints. Although there was some difference in the baseline characteristics of the cohort, treatment responses were comparable or even superior to those in the pivotal clinical trials. In addition, there was a low frequency of mild adverse events leading to treatment discontinuation and no serious adverse events. These findings emphasize the strong efficacy and tolerability of JAKinhibs in daily clinical practice, supporting their role as effective therapeutic options for patients with PsD.
Collapse
Affiliation(s)
- Francesco Bizzarri
- Servicio de Reumatologia, Instituto Biosanitario de Granada, Ibs, Hospital Universitario San Cecilio, 18012 Granada, Spain; (F.B.); (P.M.-G.); (E.R.-A.)
| | - Ricardo Ruiz-Villaverde
- Servicio de Dermatología, Instituto Biosanitario de Granada, Ibs, Hospital Universitario San Cecilio, 18012 Granada, Spain; (J.C.R.-C.); (M.C.-V.); (M.R.-T.)
| | - Pilar Morales-Garrido
- Servicio de Reumatologia, Instituto Biosanitario de Granada, Ibs, Hospital Universitario San Cecilio, 18012 Granada, Spain; (F.B.); (P.M.-G.); (E.R.-A.)
| | - Jose Carlos Ruiz-Carrascosa
- Servicio de Dermatología, Instituto Biosanitario de Granada, Ibs, Hospital Universitario San Cecilio, 18012 Granada, Spain; (J.C.R.-C.); (M.C.-V.); (M.R.-T.)
| | - Marta Cebolla-Verdugo
- Servicio de Dermatología, Instituto Biosanitario de Granada, Ibs, Hospital Universitario San Cecilio, 18012 Granada, Spain; (J.C.R.-C.); (M.C.-V.); (M.R.-T.)
| | - Alvaro Prados-Carmona
- Servicio de Dermatología, Instituto Biosanitario de Granada, Ibs, Hospital Universitario San Cecilio, 18012 Granada, Spain; (J.C.R.-C.); (M.C.-V.); (M.R.-T.)
| | - Mar Rodriguez-Troncoso
- Servicio de Dermatología, Instituto Biosanitario de Granada, Ibs, Hospital Universitario San Cecilio, 18012 Granada, Spain; (J.C.R.-C.); (M.C.-V.); (M.R.-T.)
| | - Enrique Raya-Alvarez
- Servicio de Reumatologia, Instituto Biosanitario de Granada, Ibs, Hospital Universitario San Cecilio, 18012 Granada, Spain; (F.B.); (P.M.-G.); (E.R.-A.)
| |
Collapse
|
49
|
Macaluso FS, Caprioli F, Benedan L, Bezzio C, Caporali R, Cauli A, Chimenti MS, Ciccia F, D'Angelo S, Fantini MC, Festa S, Iannone F, Lubrano E, Mariani P, Papi C, Provenzano G, Pugliese D, Rispo A, Saibeni S, Salvarani C, Variola A, Zenga M, Armuzzi A, Orlando A, Gerli R. The management of patients with inflammatory bowel disease-associated spondyloarthritis: Italian Group for the Study of Inflammatory Bowel Disease (IG-IBD) and Italian Society of Rheumatology (SIR) recommendations based on a pseudo-Delphi consensus. Autoimmun Rev 2024; 23:103533. [PMID: 38521214 DOI: 10.1016/j.autrev.2024.103533] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2023] [Revised: 03/20/2024] [Accepted: 03/20/2024] [Indexed: 03/25/2024]
Abstract
Spondyloarthritis (SpA) is the most frequent extraintestinal manifestation in patients with inflammatory bowel diseases (IBD). When IBD and spondyloarthritis coexist, musculoskeletal and intestinal disease features should be considered when planning a therapeutic strategy. Treatment options for IBD and SpA have expanded enormously over the last few years, but randomized controlled trials with specific endpoints focused on SpA are not available in the IBD setting. To address this important clinical topic, the Italian Group for the Study of Inflammatory Bowel Disease (IG-IBD) and the Italian Society of Rheumatology (SIR) jointly planned to draw updated therapeutic recommendations for IBD-associated SpA using a pseudo-Delphi method. This document presents the official recommendations of IG-IBD and SIR on the management of IBD-associated SpA in the form of 34 statements and 4 therapeutic algorithms. It is intended to be a reference guide for gastroenterologists and rheumatologists dealing with IBD-associated SpA.
Collapse
Affiliation(s)
| | - Flavio Caprioli
- Department of Pathophysiology and Transplantation, University of Milan, Italy; Gastroenterology and Endoscopy Unit, Fondazione IRCCS Ca Granda, Ospedale Maggiore Policlinico di Milano, Milan, Italy.
| | - Laura Benedan
- Bicocca-Applied Statistics Center, Department of Economics, Management and Statistics, University of Milano-Bicocca, Milano, Italy
| | - Cristina Bezzio
- IBD Center, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy; Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy
| | - Roberto Caporali
- Rheumatology Unit, Department of Clinical and Community Sciences, University of Milan, ASST Gaetano Pini-CTO, Milan, Italy
| | - Alberto Cauli
- Rheumatology Unit, Department of Medicine and Public Health, AOU and University of Cagliari, Cagliari, Italy
| | - Maria Sole Chimenti
- Rheumatology, Allergology and Clinical Immunology, Department of Systems Medicine, University of Rome Tor Vergata, Italy
| | - Francesco Ciccia
- Department of Precision Medicine, Division of Rheumatology, Università della Campania L. Vanvitelli, Naples, Italy
| | - Salvatore D'Angelo
- Rheumatology Department of Lucania, San Carlo Hospital of Potenza, Potenza, Italy
| | - Massimo Claudio Fantini
- Department of Medical Science and Public Health, University of Cagliari, Cagliari, Italy; Azienda Ospedaliero-Universitaria di Cagliari, Cagliari, Italy
| | | | | | - Ennio Lubrano
- Dipartimento di Medicina e Scienze della Salute, Università degli Studi del Molise, Campobasso, Italy
| | - Paolo Mariani
- Bicocca-Applied Statistics Center, Department of Economics, Management and Statistics, University of Milano-Bicocca, Milano, Italy
| | | | | | - Daniela Pugliese
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Rome, Italy; IBD Unit, CEMAD, Digestive Diseases Center, Medicina Interna e Gastroenterologia, Dipartimento di Scienze Mediche e Chirurgiche, Fondazione Policlinico Universitario "A. Gemelli" IRCCS, Rome, Italy
| | - Antonio Rispo
- Gastroenterology Unit, Department of Clinical Medicine and Surgery, University "Federico II" of Naples, Naples, Italy
| | - Simone Saibeni
- IBD Center, Gastroenterology Unit, Rho Hospital ASST Rhodense, Italy
| | - Carlo Salvarani
- Azienda USL-IRCCS di Reggio Emilia e Università di Modena e Reggio Emilia, Italy
| | | | - Mariangela Zenga
- Bicocca-Applied Statistics Center, Department of Economics, Management and Statistics, University of Milano-Bicocca, Milano, Italy
| | - Alessandro Armuzzi
- IBD Center, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy; Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy
| | | | - Roberto Gerli
- Rheumatology Unit, Department of Medicine & Surgery, University of Perugia, Italy
| |
Collapse
|
50
|
Potlabathini T, Pothacamuri MA, Bandi VV, Anjum M, Shah P, Molina M, Dutta N, Adzhymuratov O, Mathew M, Sadu V, Zahid SA, Lingamgunta H, Sahotra M, Nasiri SMZJ, Daguipa CDM. FDA-Approved Janus Kinase-Signal Transducer and Activator of Transcription (JAK-STAT) Inhibitors for Managing Rheumatoid Arthritis: A Narrative Review of the Literature. Cureus 2024; 16:e59978. [PMID: 38854342 PMCID: PMC11162266 DOI: 10.7759/cureus.59978] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/09/2024] [Indexed: 06/11/2024] Open
Abstract
Rheumatoid arthritis (RA) is a complex autoimmune disease causing chronic joint inflammation and, in more serious cases, organ involvement. RA typically affects people between the ages of 35 and 60; however, it can also afflict children younger than the age of 16 years and can also demonstrate a pattern of remission later in the disease course. Non-steroidal anti-inflammatory drugs, glucocorticoids, exercise, and patient education are all used in the management of RA, which is divided into symptomatic management and disease-modifying management (disease-modifying antirheumatic drugs) to reduce pain and inflammation, thereby preserving joint function. Janus kinase inhibitors (JAKis) have led to a substantial improvement in the management of RA. By specifically targeting the JAK-signal transducer and activator of transcription pathway, which is essential for immunological modulation, these inhibitors also demonstrate promise in treating various autoimmune illnesses, including inflammatory bowel diseases, giant cell arteritis, ankylosing spondylitis, and psoriatic arthritis. Tofacitinib, baricitinib, upadacitinib, peficitinib, delgocitinib, and filgotinib are examples of FDA-approved JAKis that have distinct properties and indications for treating a range of autoimmune illnesses. JAKis demonstrate a promising treatment approach for managing RA and other autoimmune diseases while enhancing patient outcomes and quality of life. However, due to major safety concerns and the need for long-term success, meticulous patient monitoring is essential.
Collapse
Affiliation(s)
| | | | | | - Mahnoor Anjum
- Medicine, King Edward Medical University, Lahore, PAK
| | | | - M Molina
- Internal Medicine, International Medical Graduates (IMG) Helping Hands, Newark, USA
| | - Nilashis Dutta
- General Medicine, North Bengal Medical College and Hospital, Siliguri, IND
| | | | - Midhun Mathew
- Internal Medicine, Pennsylvania Hospital, Philadelphia, USA
| | - Vatsalya Sadu
- Medicine and Surgery, Kamineni Academy of Medical Sciences and Research Centre, Hyderabad, IND
| | - Shiza A Zahid
- Medicine and Surgery, Jinnah Sindh Medical University, Karachi, PAK
| | - Harini Lingamgunta
- Medicine, All Saints University School of Medicine Dominica, Chicago, USA
| | - Monika Sahotra
- Internal Medicine, International Medical Graduates (IMG) Helping Hands, San Pablo, USA
| | | | | |
Collapse
|