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
|
Ren Y, Qi F, Li J, Liu X, Zhang Y, Shen Y, Liu H, Wang Y, Zhang YR, Wang G, Li N. Expression and correlation analysis of VISTA in peripheral blood mononuclear cells of myasthenia gravis patients. Int Immunopharmacol 2025; 148:114096. [PMID: 39862630 DOI: 10.1016/j.intimp.2025.114096] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Revised: 01/13/2025] [Accepted: 01/13/2025] [Indexed: 01/27/2025]
Abstract
Myasthenia gravis (MG) is a T cell-dependent, B cell-mediated disorder strongly associated with antigen presentation by dendritic cells (DCs). In MG, mucosal tolerance is linked to increased expression of TGF-β mRNA in monocytes. Additionally, monocytic myeloid-derived suppressor cells (M-MDSCs) exhibit negative immunomodulatory effects by suppressing autoreactive T and B cells. In short, all these cells play an important role in the occurrence of MG. V domain-containing Ig suppressor of T-cell activation (VISTA) is an immune checkpoint molecule constitutively expressed on dendritic cells (DCs), CD4+, CD8+ T cells, monocytes and M-MDSCs. Similar to CTLA-4 and PD-1, VISTA controls peripheral tolerance and autoimmune disorder. VISTA expressed on APCs acts as a ligand to suppress the proliferation and cytokine production of both CD4+ and CD8+ T cells. VISTA expressed on CD4+ T cells also suppresses T cell activation in a T-cell autonomous manner. Thus, VISTA may also play an important role in the immune regulation of MG disease. To investigate the role of VISTA in MG pathogenesis, we collected peripheral blood mononuclear cells (PBMCs) from MG patients and detected VISTA expression in different immune cell populations by FACs. VISTA expression was increased in CD4+ T cells, CD8+ T cells, B cells, monocytes, DCs, and M-MDSCs of PBMCs from MG patients. Correlation analysis further demonstrated that VISTA expression in monocytes was correlated with anti-AChR-IgG levels and MG-ADL scores. VISTA expression was positively associated with IL-4, TGF-β, Foxp3, IL-10, and TNF-α mRNA and negatively associated with IL-1β and IL-6 mRNA in MG PBMCs. VISTA can inhibit expression of IL-6 and TNF-α in vitro. These results suggest that VISTA may modulate MG disease progression and indicate the potential utility of VISTA agonists in MG treatment.
Collapse
Affiliation(s)
- Yan Ren
- Health Science Center, Ningbo University, Ningbo, China.
| | - Feiteng Qi
- Department of Neurology, Ningbo Medical Center Lihuili Hospital, Ningbo University, Ningbo, China.
| | - Jianing Li
- Health Science Center, Ningbo University, Ningbo, China.
| | - Xinyue Liu
- Health Science Center, Ningbo University, Ningbo, China.
| | - Yuanting Zhang
- Health Science Center, Ningbo University, Ningbo, China.
| | - Yaoyang Shen
- Health Science Center, Ningbo University, Ningbo, China.
| | - Hua Liu
- Health Science Center, Ningbo University, Ningbo, China.
| | - Yixuan Wang
- Health Science Center, Ningbo University, Ningbo, China.
| | - Yan Ru Zhang
- Health Science Center, Ningbo University, Ningbo, China.
| | - Geng Wang
- Health Science Center, Ningbo University, Ningbo, China.
| | - Na Li
- Health Science Center, Ningbo University, Ningbo, China.
| |
Collapse
|
3
|
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
|
4
|
Miyagawa I, Tanaka Y. Precision medicine using molecular-target drugs in psoriatic arthritis. Ther Adv Musculoskelet Dis 2025; 17:1759720X241311462. [PMID: 39776828 PMCID: PMC11701904 DOI: 10.1177/1759720x241311462] [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: 05/08/2024] [Accepted: 12/18/2024] [Indexed: 01/11/2025] Open
Abstract
Psoriatic arthritis (PsA) presents various clinical manifestations, including skin lesions, peripheral arthritis, axial involvement, enthesitis, nail involvement, dactylitis, and uveitis. In addition, it causes a high incidence of lifestyle-related diseases and an increase in cerebrovascular and cardiovascular events. As the pathology of PsA has been clarified, molecular-targeted drugs targeting tumor necrosis factor-α, interleukin (IL)-17A, IL-17A/F, IL-17 receptor, IL-12/23(p40), IL-23p19, Cytotoxic T-lymphocyte Antigen-4 (CTLA-4), Janus kinase, and phosphodiesterase-4 have been developed and are widely used in clinical practice. PsA is clinically and molecularly heterogeneous, and it is necessary to improve various clinical symptoms with limited treatment options simultaneously; therefore, rheumatologists sometimes encounter difficult situations in clinical practice. Hence, the development of precision medicine may improve treatment outcomes. Recently, the strategic use of molecular-targeted drugs based on the stratification of patients with PsA by peripheral blood lymphocyte phenotyping and serum cytokine concentrations has been reported to possibly lead to a higher therapeutic response. A randomized controlled trial was initiated to verify the efficacy of this treatment strategy. However, to make precision medicine in PsA feasible, shifting from conventional clinical trials to clinical trials based on biomarker profiles and accumulating further data are necessary.
Collapse
Affiliation(s)
- Ippei Miyagawa
- The First Department of Internal Medicine, School of Medicine, University of Occupational and Environmental Health, Japan, Kitakyushu, Japan
| | - Yoshiya Tanaka
- The First Department of Internal Medicine, School of Medicine, University of Occupational and Environmental Health, Japan, 1-1 Iseigaoka, Yahata-nishi, Kitakyushu 807-8555, Japan
| |
Collapse
|
5
|
Kim GR, Nam KH, Choi JM. Belatacept and regulatory T cells in transplantation: synergistic strategies for immune tolerance and graft survival. CLINICAL TRANSPLANTATION AND RESEARCH 2024; 38:326-340. [PMID: 39690903 PMCID: PMC11732762 DOI: 10.4285/ctr.24.0057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Revised: 11/22/2024] [Accepted: 11/28/2024] [Indexed: 12/19/2024]
Abstract
Calcineurin inhibitors (CNIs) have been a cornerstone in solid organ transplantation for many years; however, their prolonged use is linked to significant adverse effects, most notably nephrotoxicity. Belatacept, a modified version of cytotoxic T lymphocyte antigen-4 immunoglobulin with increased binding affinity for its ligand, has emerged as a viable alternative to traditional CNIs due to its lower toxicity profile. Despite these benefits, belatacept is associated with a higher rate of acute rejection, which presents a challenge for long-term graft survival. This review reevaluates the limitations of belatacept in achieving long-term acceptance of transplants and highlights the importance of regulatory T (Treg) cells in maintaining immune tolerance and preventing graft rejection. Additionally, it discusses the potential benefits of combining therapies that boost Treg cells with belatacept to increase the effectiveness of immunosuppression and improve graft outcomes.
Collapse
Affiliation(s)
- Gil-Ran Kim
- Department of Life Science, College of Natural Sciences, Hanyang University, Seoul, Korea
- Research Institute for Natural Sciences, Hanyang University, Seoul, Korea
| | - Kyung-Ho Nam
- Department of Life Science, College of Natural Sciences, Hanyang University, Seoul, Korea
| | - Je-Min Choi
- Department of Life Science, College of Natural Sciences, Hanyang University, Seoul, Korea
- Research Institute for Natural Sciences, Hanyang University, Seoul, Korea
- Hanyang Institute of Bioscience and Biotechnology, Hanyang University, Seoul, Korea
- Research Institute for Convergence of Basic Sciences, Hanyang University, Seoul, Korea
| |
Collapse
|
6
|
Boboryko D, Olejnik-Wojciechowska J, Baranowska M, Bratborska AW, Skórka P, Pawlik A. Biological therapy for psoriatic arthritis: current state and future perspectives. Rheumatol Int 2024; 44:2711-2725. [PMID: 39311915 DOI: 10.1007/s00296-024-05722-1] [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: 07/23/2024] [Accepted: 09/10/2024] [Indexed: 12/14/2024]
Abstract
Psoriatic arthritis is a medical condition that lies at the intersection of various fields of medicine, and its therapy always requires a comprehensive, holistic approach. Biological disease-modifying antirheumatic drugs (bDMARDs) constitute an extremely effective treatment method for PsA, provided that appropriate principles for patient qualification for the drug are followed, along with subsequent monitoring of the response to treatment. Based on their mechanisms of action, four main groups of bDMARDs used in PsA can be distinguished (TNF inhibitors, IL-12/23 and IL-23 inhibitors, IL-17 inhibitors, CTLA4 agonists). Clinical trials are ongoing in search of registration for additional bDMARDs, and the tasks for doctors and scientists worldwide include patient education, increasing treatment accessibility, and optimizing its costs.
Collapse
Affiliation(s)
- Dominika Boboryko
- Department of Physiology, Pomeranian Medical University, Szczecin, 70-111, Poland
| | | | - Magdalena Baranowska
- Department of Physiology, Pomeranian Medical University, Szczecin, 70-111, Poland
| | | | - Patryk Skórka
- Department of Physiology, Pomeranian Medical University, Szczecin, 70-111, Poland
| | - Andrzej Pawlik
- Department of Physiology, Pomeranian Medical University, Szczecin, 70-111, Poland.
| |
Collapse
|
7
|
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
|
8
|
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
|
9
|
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
|
10
|
Srinivasalu H, Simpson J, Stoll ML. Drug therapy in juvenile spondyloarthritis. Curr Opin Rheumatol 2024; 36:295-301. [PMID: 38639758 DOI: 10.1097/bor.0000000000001016] [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: 04/20/2024]
Abstract
PURPOSE OF REVIEW This review summarizes latest developments in treatment of juvenile spondyloarthritis (JSpA), specifically enthesitis-related arthritis (ERA) and juvenile psoriatic arthritis (JPsA). RECENT FINDINGS There has been addition of biologic disease modifying antirheumatic drugs (bDMARDs) beyond tumor necrosis factor inhibitors (TNFi) for JSpA such as IL-17 blockers, IL-23 blockers, and janus activating kinase inhibitors with favorable safety profile. Conducting robust clinical trials for this subpopulation of JIA remains a challenge; extrapolation studies are being used to obtain approval from regulatory agencies. SUMMARY Newer drug therapies have expanded the scope of treatment for patients with JSpA. bDMARDs such as adalimumab, etanercept, infliximab, and secukinumab have demonstrated clinically significant treatment efficacy in ERA and JPsA. Based on extrapolation studies, intravenous golimumab, etanercept, abatacept, and ustekinumab have gained Food and Drug Administration (FDA) approval for JPsA. Long-term follow-up studies continue to demonstrate acceptable safety profiles. There is need for more real-world data on drug efficacy from Registry studies and research on effective de-escalation strategies.
Collapse
Affiliation(s)
- Hemalatha Srinivasalu
- GW University School of Medicine
- Division of Rheumatology, Children's National Hospital, Washington, DC
| | - Jessica Simpson
- Division of Rheumatology, Children's National Hospital, Washington, DC
| | - Matthew L Stoll
- Division of Rheumatology, Department of Pediatrics, University of Alabama at Birmingham, Birmingham, Alabama, USA
| |
Collapse
|
11
|
Gossec L, Kerschbaumer A, Ferreira RJO, Aletaha D, Baraliakos X, Bertheussen H, Boehncke WH, Esbensen BA, McInnes IB, McGonagle D, Winthrop KL, Balanescu A, Balint PV, Burmester GR, Cañete JD, Claudepierre P, Eder L, Hetland ML, Iagnocco A, Kristensen LE, Lories R, Queiro R, Mauro D, Marzo-Ortega H, Mease PJ, Nash P, Wagenaar W, Savage L, Schett G, Shoop-Worrall SJW, Tanaka Y, Van den Bosch FE, van der Helm-van Mil A, Zabotti A, van der Heijde D, Smolen JS. EULAR recommendations for the management of psoriatic arthritis with pharmacological therapies: 2023 update. Ann Rheum Dis 2024; 83:706-719. [PMID: 38499325 PMCID: PMC11103320 DOI: 10.1136/ard-2024-225531] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Accepted: 02/26/2024] [Indexed: 03/20/2024]
Abstract
OBJECTIVE New modes of action and more data on the efficacy and safety of existing drugs in psoriatic arthritis (PsA) required an update of the EULAR 2019 recommendations for the pharmacological treatment of PsA. METHODS Following EULAR standardised operating procedures, the process included a systematic literature review and a consensus meeting of 36 international experts in April 2023. Levels of evidence and grades of recommendations were determined. RESULTS The updated recommendations comprise 7 overarching principles and 11 recommendations, and provide a treatment strategy for pharmacological therapies. Non-steroidal anti-inflammatory drugs should be used in monotherapy only for mild PsA and in the short term; oral glucocorticoids are not recommended. In patients with peripheral arthritis, rapid initiation of conventional synthetic disease-modifying antirheumatic drugs is recommended and methotrexate preferred. If the treatment target is not achieved with this strategy, a biological disease-modifying antirheumatic drug (bDMARD) should be initiated, without preference among modes of action. Relevant skin psoriasis should orient towards bDMARDs targeting interleukin (IL)-23p40, IL-23p19, IL-17A and IL-17A/F inhibitors. In case of predominant axial or entheseal disease, an algorithm is also proposed. Use of Janus kinase inhibitors is proposed primarily after bDMARD failure, taking relevant risk factors into account, or in case bDMARDs are not an appropriate choice. Inflammatory bowel disease and uveitis, if present, should influence drug choices, with monoclonal tumour necrosis factor inhibitors proposed. Drug switches and tapering in sustained remission are also addressed. CONCLUSION These updated recommendations integrate all currently available drugs in a practical and progressive approach, which will be helpful in the pharmacological management of PsA.
Collapse
Affiliation(s)
- Laure Gossec
- INSERM, Institut Pierre Louis d'Epidémiologie et de Santé Publique, Sorbonne Universite, Paris, France
- APHP, Rheumatology Department, Hopital Universitaire Pitie Salpetriere, Paris, France
| | - Andreas Kerschbaumer
- Division of Rheumatology, Department of Medicine 3, Medical University of Vienna, Vienna, Austria
| | - Ricardo J O Ferreira
- Nursing Research, Innovation and Development Centre of Lisbon (CIDNUR), Higher School of Nursing of Lisbon, Lisbon, Portugal
- Rheumatology Department, Centro Hospitalar e Universitário de Coimbra EPE, Coimbra, Portugal
| | - Daniel Aletaha
- Division of Rheumatology, Department of Medicine 3, Medical University of Vienna, Vienna, Austria
| | | | | | | | - Bente Appel Esbensen
- Copenhagen Center for Arthritis Research, Center for Rheumatology and Spine Diseases, Centre for Head and Orthopaedics, Rigshospitalet, Glostrup, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Iain B McInnes
- College of Medical Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Dennis McGonagle
- LTHT, NIHR Leeds Biomedical Research Centre, Leeds, UK
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Leeds, UK
| | - Kevin L Winthrop
- Division of Infectious Diseases, School of Medicine, School of Public Health, Oregon Health & Science University, Portland, Oregon, USA
| | - Andra Balanescu
- Sf Maria Hospital, University of Medicine and Pharmacy Carol Davila Bucharest, Bucharest, Romania
| | - Peter V Balint
- Medical Imaging Centre, Semmelweis University, 3rd Rheumatology Department, National Institute of Musculoskeletal Diseases, Budapest, Hungary
| | - Gerd R Burmester
- Department of Rheumatology and Clinical Immunology, Freie Universität Berlin and Humboldt-Universität zu Berlin, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Juan D Cañete
- Arthritis Unit, Department of Rheumatology, Hospital Clínic Barcelona, Barcelona, Spain
- FCRB, IDIBAPS, Barcelona, Spain
| | - Pascal Claudepierre
- Rheumatology, AP-HP, Henri Mondor University Hospital, Creteil, France
- EA Epiderme, UPEC, Creteil, France
| | - Lihi Eder
- Department of Medicine, University of Toronto, Women's College Hospital, Toronto, Toronto, Canada
| | - Merete Lund Hetland
- The Copenhagen Center for Arthritis Research, Center for Rheumatology and Spine Diseases, Centre of Head and Orthopedics, Rigshospitalet Glostrup, Glostrup, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Annamaria Iagnocco
- Academic Rheumatology Centre, Dipartimento Scienze Cliniche Biologiche, Università di Torino - AO Mauriziano Torino, Turin, Italy
| | - Lars Erik Kristensen
- The Parker Institute, Bispebjerg, Denmark
- Frederiksberg Hospital, Copenhagen University, Copenhagen, Denmark
| | - Rik Lories
- Laboratory of Tissue Homeostasis and Disease, Skeletal Biology and Engineering Research Center, KU Leuven, Leuven, Belgium
- Division of Rheumatology, University Hospitals Leuven, Leuven, Belgium
| | - Rubén Queiro
- Rheumatology, Hospital Universitario Central de Asturias, Oviedo, Spain
- Translational Immunology Division, Biohealth Research Institute of the Principality of Asturias, Oviedo University School of Medicine, Oviedo, Spain
| | - Daniele Mauro
- Department of Precision Medicine, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Helena Marzo-Ortega
- LTHT, NIHR Leeds Biomedical Research Centre, Leeds, UK
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Leeds, UK
| | - Philip J Mease
- Rheumatology Research, Providence Swedish, Seattle, Washington, USA
- University of Washington School of Medicine, Seattle, Washington, USA
| | - Peter Nash
- School of Medicine, Griffith University, Brisbane, Queensland, Australia
| | - Wendy Wagenaar
- Tranzo, Tilburg School of Social and Behavioral Sciences, Tilburg University, Tilburg, The Netherlands
- Young PARE Patient Research Partner, EULAR, Zurich, Switzerland
| | - Laura Savage
- School of Medicine and Dermatology, Leeds Teaching Hospitals NHS Trust, University of Leeds, Leeds, UK
| | - Georg Schett
- Department of Internal Medicine 3, Rheumatology and Immunology and Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Stephanie J W Shoop-Worrall
- Children and Young Person's Rheumatology Research Programme, Centre for Musculoskeletal Research, The University of Manchester, Manchester, UK
| | - Yoshiya Tanaka
- First Department of Internal Medicine, University of Occupational and Environmental Health, Japan, Kitakyushu, Japan
| | - Filip E Van den Bosch
- Department of Internal Medicine and Pediatrics, VIB Center for Inflammation Research, Ghent University, Gent, Belgium
| | | | - Alen Zabotti
- Department of Medical and Biological Sciences, Azienda sanitaria universitaria Friuli Centrale, Udine, Italy
| | | | - Josef S Smolen
- Division of Rheumatology, Department of Medicine 3, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
12
|
De Stefano L, Bugatti S, Mazzucchelli I, Rossi S, Xoxi B, Bozzalla Cassione E, Luvaro T, Montecucco C, Manzo A. Synovial and serum B cell signature of autoantibody-negative rheumatoid arthritis vs autoantibody-positive rheumatoid arthritis and psoriatic arthritis. Rheumatology (Oxford) 2024; 63:1322-1331. [PMID: 37481716 DOI: 10.1093/rheumatology/kead378] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Accepted: 05/10/2023] [Indexed: 07/24/2023] Open
Abstract
OBJECTIVES Autoantibody-negative RA differs from autoantibody-positive RA in several clinical aspects, possibly underpinned by pathogenetic differences. At present, the role of adaptive immune responses in autoantibody-negative RA remains unclear. Here, we investigated the synovial and serum immunophenotype indicative of B lymphocyte involvement across the spectrum of autoantibody-positive and -negative chronic arthritides. METHODS Ultrasound-guided synovial biopsies were retrieved from 131 patients: 43 autoantibody-positive RA, 35 autoantibody-negative RA, 25 polyarticular PsA and 28 oligoarticular PsA. Samples were analysed for the degree of histological inflammation, B lymphocyte infiltration and the distribution of different pathotypes (lympho-myeloid, myeloid, pauci-immune). Serum levels of the B cell chemoattractant CXCL13 were compared among groups. RESULTS Synovitis scores and CD68+ sublining macrophage infiltration were comparable irrespective of clinical diagnosis and disease subtype. In contrast, the degree of B lymphocyte infiltration and the frequency of lympho-myeloid synovitis in autoantibody-negative RA were lower than those of autoantibody-positive RA (mean [s.d.] 1.8 [1] vs 2.4 [0.6], P = 0.03, and 38.2% vs 62.9%, P = 0.07, respectively), and similar to polyarticular PsA. Oligoarticular PsA had the lowest B cell scores. Serum CXCL13 was associated with lympho-myeloid synovitis and followed a similar gradient, with the highest levels in autoantibody-positive RA, intermediate and comparable levels in autoantibody-negative RA and polyarticular PsA, and low levels in oligoarticular PsA. CONCLUSIONS The synovial and serum immunophenotype indicative of B lymphocyte involvement in autoantibody-negative RA differs from that of autoantibody-positive RA and more closely resembles that observed in polyarticular PsA. The pathobiological stratification of chronic inflammatory arthritides beyond clinical diagnosis may fuel personalized treatment strategies.
Collapse
Affiliation(s)
- Ludovico De Stefano
- Rheumatology and Translational Immunology Research Laboratories (LaRIT), Department of Internal Medicine and Therapeutics, Università di Pavia, Pavia, Italy
- Division of Rheumatology, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Serena Bugatti
- Rheumatology and Translational Immunology Research Laboratories (LaRIT), Department of Internal Medicine and Therapeutics, Università di Pavia, Pavia, Italy
- Division of Rheumatology, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Iolanda Mazzucchelli
- Rheumatology and Translational Immunology Research Laboratories (LaRIT), Department of Internal Medicine and Therapeutics, Università di Pavia, Pavia, Italy
| | - Silvia Rossi
- Division of Rheumatology, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Blerina Xoxi
- Division of Rheumatology, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Emanuele Bozzalla Cassione
- Rheumatology and Translational Immunology Research Laboratories (LaRIT), Department of Internal Medicine and Therapeutics, Università di Pavia, Pavia, Italy
- Division of Rheumatology, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Terenzj Luvaro
- Rheumatology and Translational Immunology Research Laboratories (LaRIT), Department of Internal Medicine and Therapeutics, Università di Pavia, Pavia, Italy
| | - Carlomaurizio Montecucco
- Rheumatology and Translational Immunology Research Laboratories (LaRIT), Department of Internal Medicine and Therapeutics, Università di Pavia, Pavia, Italy
- Division of Rheumatology, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Antonio Manzo
- Rheumatology and Translational Immunology Research Laboratories (LaRIT), Department of Internal Medicine and Therapeutics, Università di Pavia, Pavia, Italy
- Division of Rheumatology, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| |
Collapse
|
13
|
Gladman DD, Chandran V, Rosen CF, Rohekar S, Boyd T, Eder L, Rahman P, Dutz J, Chan J, Haydey RP, Barac S, Laliberté MC, Girard T, Fournier PA, Sutton M, Pereira D, Chim T, Coupal L, Choquette D. Residual Disease Activity in Canadian Patients With Psoriatic Arthritis Treated With Advanced Therapies: Results From a Multiregistry Analysis (UNISON-PsA). J Rheumatol 2024; 51:479-487. [PMID: 38359937 DOI: 10.3899/jrheum.2023-0716] [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: 01/10/2024] [Indexed: 02/17/2024]
Abstract
OBJECTIVE Although patient outcomes in psoriatic arthritis (PsA) have improved with the advent of advanced therapies, there remains a high unmet need to treat residual disease activity. The objective of the current study was to quantify residual disease activity and burden of disease in Canadian patients with PsA. METHODS This was a multiregion, observational, retrospective analysis of patient data extracted from the Rhumadata and the International Psoriasis and Arthritis Research Team (IPART) registries, analyzing deidentified data from patients who had initiated advanced therapy for the treatment of PsA between January 2010 and December 2019. The primary endpoint was the proportion of patients failing to achieve minimal disease activity (MDA) within 6 months; secondary endpoints included clinical and patient-reported burden of disease. Descriptive statistics included summaries by region, treatment class, and number of prior advanced therapies. RESULTS One thousand five hundred ninety-six patients were included. The proportions of patients who failed to achieve MDA within 6 months of an advanced therapy were 64.8% in Ontario, 68.3% in Western Canada, 74.8% in Quebec, and 75% in the Atlantic/East region. Failure to achieve MDA was higher among patients receiving an IL-17i compared with a TNFi in all regions except the Atlantic/East. Between 73.2% and 78.6% of patients reported pain at 6 months, and continuing functional impairment varied from 24% in the West to 83.3% in the Atlantic/East. CONCLUSION There is substantial burden and unmet need for improved therapies for Canadians with PsA. There is a wide regional variation in outcomes that requires further assessment.
Collapse
Affiliation(s)
- Dafna D Gladman
- D.D. Gladman, MD, University of Toronto, Department of Medicine, Division of Rheumatology, and Schroeder Arthritis Institute, Krembil Research Institute, University Health Network, Toronto, Ontario;
| | - Vinod Chandran
- V. Chandran, MBBS, MD, DM, PhD, University of Toronto, Department of Medicine, Division of Rheumatology, Schroeder Arthritis Institute, Krembil Research Institute, University Health Network, and Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario
| | - Cheryl F Rosen
- C.F. Rosen, MD, Division of Dermatology, Department of Medicine, University of Toronto, and Division of Dermatology, Toronto Western Hospital, Toronto, Ontario
| | - Sherry Rohekar
- S. Rohekar, MD, Division of Rheumatology, Western University, London, Ontario
| | - Tristan Boyd
- T. Boyd, MD, Division of Rheumatology, Western University, and Division of Rheumatology, St. Joseph's Hospital, London, Ontario
| | - Lihi Eder
- L. Eder, MD, PhD, Division of Rheumatology, Women's College Hospital, and Department of Medicine, University of Toronto, Toronto, Ontario
| | - Proton Rahman
- P. Rahman, MD, Department of Medicine, Memorial University, St. John's, Newfoundland
| | - Jan Dutz
- J. Dutz, MD, Department of Dermatology and Skin Science, University of British Columbia, Vancouver, British Columbia
| | - Jonathan Chan
- J. Chan, MD, Department of Medicine, Division of Rheumatology, University of British Columbia, and Arthritis Research Canada, Vancouver, British Columbia
| | - Richard P Haydey
- R.P. Haydey, MD, S. Barac, MD, Winnipeg Clinic, Winnipeg, Manitoba
| | - Snezana Barac
- R.P. Haydey, MD, S. Barac, MD, Winnipeg Clinic, Winnipeg, Manitoba
| | - Marie-Claude Laliberté
- M.C. Laliberté, PhD, T. Girard, PhD, P.A. Fournier, MSc, MBA, AbbVie Corp., St. Laurent, Quebec
| | - Tanya Girard
- M.C. Laliberté, PhD, T. Girard, PhD, P.A. Fournier, MSc, MBA, AbbVie Corp., St. Laurent, Quebec
| | - Pierre-André Fournier
- M.C. Laliberté, PhD, T. Girard, PhD, P.A. Fournier, MSc, MBA, AbbVie Corp., St. Laurent, Quebec
| | - Mitchell Sutton
- M. Sutton, MSc, D. Pereira, BSc, T. Chim, MSc, Center for Prognosis Studies in the Rheumatic Diseases, Psoriatic Disease Program, Toronto Western Hospital, University Health Network, Toronto, Ontario
| | - Daniel Pereira
- M. Sutton, MSc, D. Pereira, BSc, T. Chim, MSc, Center for Prognosis Studies in the Rheumatic Diseases, Psoriatic Disease Program, Toronto Western Hospital, University Health Network, Toronto, Ontario
| | - Tina Chim
- M. Sutton, MSc, D. Pereira, BSc, T. Chim, MSc, Center for Prognosis Studies in the Rheumatic Diseases, Psoriatic Disease Program, Toronto Western Hospital, University Health Network, Toronto, Ontario
| | - Louis Coupal
- L. Coupal, MSc, D. Choquette, MD, Institut de Rhumatologie de Montréal, CHUM, University of Montreal, Montreal, Quebec, Canada
| | - Denis Choquette
- L. Coupal, MSc, D. Choquette, MD, Institut de Rhumatologie de Montréal, CHUM, University of Montreal, Montreal, Quebec, Canada
| |
Collapse
|
14
|
Séauve M, Auréal M, Laplane S, Lega JC, Cabrera N, Coury F. Risk of infections in psoriatic arthritis or axial spondyloarthritis patients treated with targeted therapies: A meta-analysis of randomized controlled trials. Joint Bone Spine 2024; 91:105673. [PMID: 38042364 DOI: 10.1016/j.jbspin.2023.105673] [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: 04/19/2023] [Revised: 11/01/2023] [Accepted: 11/14/2023] [Indexed: 12/04/2023]
Abstract
OBJECTIVE To evaluate the risk of global infections in patients with psoriatic arthritis (PsA) and axial spondyloarthritis encompassing ankylosing spondylitis (AS) and non-radiographic axial spondyloarthritis (nr-axSpA) treated with targeted therapies. METHODS Medline and Cochrane databases were systematically searched up to March 2021 for randomized controlled trials (RCTs) performed in patients with PsA or axial spondyloarthritis treated with biologic or targeted synthetic disease-modifying anti-rheumatic drugs (b/tsDMARDs). Global infections (any infections reported, including bacterial, viral and fungal infections, except serious infections) were the primary outcome. Secondary outcomes included serious infections defined as life-threatening infections or any infection requiring intravenous antibiotics or hospitalization. The relative risk of infections was determined by meta-analysis of RCTs. RESULTS A total of 60 RCTs were included (20,418 patients), encompassing 17 b/tsDMARDs, compared with placebo, conventional synthetic drugs (csDMARDs) or non-steroidal anti-inflammatory drugs (NSAIDs). An increased risk of any infection for patients exposed to these drugs was found (RR 1.15, 95% CI [1.06-1.25]), mainly with high doses and longer duration of treatment. Most infections were respiratory tract or ear, nose, and throat (ENT) infections. Subgroup analyses showed a statistically significant increased risk of infections for axial spondyloarthritis patients (RR 1.32, 95% CI [1.14-1.52]), but not for PsA patients (RR 1.05, 95% CI [0.97-1.14]). Infection risk was highest with TNF inhibitors (RR 1.23, 95% CI [1.11-1.37]) and IL-17 inhibitors (RR 1.30, 95% CI [1.07-1.59]). No increased risk of serious infections was shown. CONCLUSION In contrast to serious infections, the risk of global infections is moderately increased with b/tsDMARDs in spondyloarthritis, and is associated in particular with use of TNF and IL-17 inhibitors.
Collapse
Affiliation(s)
- Milène Séauve
- University of Lyon, University Lyon 1, 69100 Lyon, France; Department of Rheumatology, Lyon Sud Hospital, Hospices Civils de Lyon, 69495 Pierre-Bénite, France
| | - Mélanie Auréal
- University of Lyon, University Lyon 1, 69100 Lyon, France; Department of Rheumatology, Lyon Sud Hospital, Hospices Civils de Lyon, 69495 Pierre-Bénite, France
| | - Soline Laplane
- University of Lyon, University Lyon 1, 69100 Lyon, France; Department of Rheumatology, Lyon Sud Hospital, Hospices Civils de Lyon, 69495 Pierre-Bénite, France
| | - Jean-Christophe Lega
- University of Lyon, University Lyon 1, 69100 Lyon, France; Department of Internal and Vascular Medicine, Lyon Sud Hospital, Hospices Civils de Lyon, Pierre-Bénite, France; University of Lyon, UMR - CNRS 5558, Laboratoire de Biométrie et Biologie Évolutive, 69100 Lyon, France; Lyon Immunopathology Federation, Lyon, France
| | - Natalia Cabrera
- University of Lyon, UMR - CNRS 5558, Laboratoire de Biométrie et Biologie Évolutive, 69100 Lyon, France
| | - Fabienne Coury
- University of Lyon, University Lyon 1, 69100 Lyon, France; Department of Rheumatology, Lyon Sud Hospital, Hospices Civils de Lyon, 69495 Pierre-Bénite, France; Lyon Immunopathology Federation, Lyon, France; University of Lyon, Inserm UMR 1033, 69100 Lyon, France.
| |
Collapse
|
15
|
Yao Y, Zeng L, Huang X, Zhang J, Zhang G, Wang L. Role of co‑inhibitory molecules in the treatment of psoriasis (Review). Exp Ther Med 2024; 27:209. [PMID: 38590557 PMCID: PMC11000047 DOI: 10.3892/etm.2024.12497] [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: 06/18/2023] [Accepted: 01/24/2024] [Indexed: 04/10/2024] Open
Abstract
Psoriasis is a common chronic inflammatory skin disease characterized by abnormal activation and infiltration of T-cells and excessive proliferation of keratinocytes (KCs). Its pathogenesis is complex and frequently accompanied by the imbalance of T-cell subpopulations, contributing to its development and further exacerbation. Therefore, the immune system, especially T-cells, is mainly involved in the pathogenesis of psoriasis. While T-cell activation not only requires the first recognition of T-cell receptor and major histocompatibility complex peptide, co-stimulatory and co-inhibitory pathways are reported to promote or dampen T-cell responses through a variety of mechanisms. In recent years, immuno-related agents have been applied in the treatment of numerous clinical diseases, including psoriasis, and are starting to show promising and potential therapy prospects in autoimmune skin diseases. The present review outlined the role of co-inhibitory molecules in the pathogenesis of psoriasis and their application in the treatment of psoriasis.
Collapse
Affiliation(s)
- Yue Yao
- Department of Dermatology, The First Hospital of Hebei Medical University, Shijiazhuang, Hebei 050031, P.R. China
- Candidate Branch of National Clinical Research Center for Skin Diseases, Shijiazhuang, Hebei 050031, P.R. China
| | - Linxi Zeng
- Department of Dermatology, The First Hospital of Hebei Medical University, Shijiazhuang, Hebei 050031, P.R. China
- Candidate Branch of National Clinical Research Center for Skin Diseases, Shijiazhuang, Hebei 050031, P.R. China
| | - Xin Huang
- Department of Dermatology, The First Hospital of Hebei Medical University, Shijiazhuang, Hebei 050031, P.R. China
- Candidate Branch of National Clinical Research Center for Skin Diseases, Shijiazhuang, Hebei 050031, P.R. China
| | - Jinfang Zhang
- Department of Dermatology, The First Hospital of Hebei Medical University, Shijiazhuang, Hebei 050031, P.R. China
- Candidate Branch of National Clinical Research Center for Skin Diseases, Shijiazhuang, Hebei 050031, P.R. China
| | - Guoqiang Zhang
- Department of Dermatology, The First Hospital of Hebei Medical University, Shijiazhuang, Hebei 050031, P.R. China
- Candidate Branch of National Clinical Research Center for Skin Diseases, Shijiazhuang, Hebei 050031, P.R. China
| | - Ling Wang
- Department of Orthopedic Oncology, The Third Hospital of Hebei Medical University, Shijiazhuang, Hebei 050011, P.R. China
| |
Collapse
|
16
|
Vyas J, Johns JR, Ali FM, Singh RK, Ingram JR, Salek S, Finlay AY. A systematic review of 454 randomized controlled trials using the Dermatology Life Quality Index: experience in 69 diseases and 43 countries. Br J Dermatol 2024; 190:315-339. [PMID: 36971254 DOI: 10.1093/bjd/ljad079] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 01/31/2023] [Accepted: 03/14/2023] [Indexed: 02/18/2024]
Abstract
BACKGROUND Over 29 years of clinical application, the Dermatology Life Quality Index (DLQI) has remained the most used patient-reported outcome (PRO) in dermatology due to its robustness, simplicity and ease of use. OBJECTIVES To generate further evidence of the DLQI's utility in randomized controlled trials (RCTs) and to cover all diseases and interventions. METHODS The methodology followed PRISMA guidelines and included seven bibliographical databases, searching articles published from 1 January 1994 until 16 November 2021. Articles were reviewed independently by two assessors, and an adjudicator resolved any opinion differences. RESULTS Of 3220 screened publications, 454 articles meeting the eligibility criteria for inclusion, describing research on 198 190 patients, were analysed. DLQI scores were primary endpoints in 24 (5.3%) of studies. Most studies were of psoriasis (54.1%), although 69 different diseases were studied. Most study drugs were systemic (85.1%), with biologics comprising 55.9% of all pharmacological interventions. Topical treatments comprised 17.0% of total pharmacological interventions. Nonpharmacological interventions, mainly laser therapy and ultraviolet radiation treatment, comprised 12.2% of the total number of interventions. The majority of studies (63.7%) were multicentric, with trials conducted in at least 42 different countries; 40.2% were conducted in multiple countries. The minimal clinically importance difference (MCID) was reported in the analysis of 15.0% of studies, but only 1.3% considered full score meaning banding of the DLQI. Forty-seven (10.4%) of the studies investigated statistical correlation of the DLQI with clinical severity assessment or other PRO/quality of life tools; and 61-86% of studies had within-group scores differences greater than the MCID in 'active treatment arms'. The Jadad risk-of-bias scale showed that bias was generally low, as 91.8% of the studies had Jadad scores of ≥ 3; only 0.4% of studies showed a high risk of bias from randomization. Thirteen per cent had a high risk of bias from blinding and 10.1% had a high risk of bias from unknown outcomes of all participants in the studies. In 18.5% of the studies the authors declared that they followed an intention-to-treat protocol; imputation for missing DLQI data was used in 34.4% of studies. CONCLUSIONS This systematic review provides a wealth of evidence of the use of the DLQI in clinical trials to inform researchers' and -clinicians' decisions for its further use. Recommendations are also made for improving the reporting of data from future RCTs using the DLQI.
Collapse
Affiliation(s)
| | - Jeffrey R Johns
- Division of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, UK
| | - Faraz M Ali
- Division of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, UK
| | - Ravinder K Singh
- Division of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, UK
| | - John R Ingram
- Division of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, UK
| | - Sam Salek
- School of Life and Medical Sciences, University of Hertfordshire, Hatfield, UK
| | - Andrew Y Finlay
- Division of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, UK
| |
Collapse
|
17
|
Tanaka Y, Shaw S. Bimekizumab for the treatment of psoriatic arthritis. Expert Rev Clin Immunol 2024; 20:155-168. [PMID: 37909894 DOI: 10.1080/1744666x.2023.2277266] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Accepted: 10/26/2023] [Indexed: 11/03/2023]
Abstract
INTRODUCTION Interleukin (IL)-17A and IL-17F have overlapping roles in pro-inflammatory signaling and have been implicated in the pathogenesis of psoriatic disease. Bimekizumab is the first human monoclonal antibody to selectively inhibit IL-17F in addition to IL-17A. Bimekizumab has been studied in several phase II/III trials and has been approved for the treatment of patients with psoriatic arthritis (PsA) in the EU and UK. AREAS COVERED A literature search identified clinical trials examining the efficacy and safety of bimekizumab for PsA, which were critically appraised. EXPERT OPINION Clinical trials of bimekizumab in PsA have demonstrated rapid and sustained treatment responses and depth of response across the multiple disease domains. High levels of efficacy were sustained to 152 weeks in phase IIb trials, and to 52 weeks in phase III trials. Bimekizumab was generally well tolerated. As expected, due to the role of IL-17 in the immune response to fungal pathogens, there was an increase in mild-to-moderate, localized fungal infections with bimekizumab treatment, very few of which led to discontinuation. Studies over longer time periods, with relevant comparators from the IL-17A inhibitor class, and real-world data will be important to further define the role of bimekizumab among currently available treatments for PsA. [Figure: see text].
Collapse
Affiliation(s)
- Yoshiya Tanaka
- The First Department of Internal Medicine, University of Occupational and Environmental Health, Japan, Kitakyushu, Fukuoka, Japan
| | | |
Collapse
|
18
|
Simon TA, Dong L, Suissa S, Michaud K, Pedro S, Hochberg M, Boers M, Askling J, Frisell T, Strangfeld A, Meissner Y, Khaychuk V, Dominique A, Maldonado MA. Abatacept and non-melanoma skin cancer in patients with rheumatoid arthritis: a comprehensive evaluation of randomised controlled trials and observational studies. Ann Rheum Dis 2024; 83:177-183. [PMID: 37932010 PMCID: PMC10850629 DOI: 10.1136/ard-2023-224356] [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: 04/25/2023] [Accepted: 09/29/2023] [Indexed: 11/08/2023]
Abstract
OBJECTIVES This study aims to evaluate non-melanoma skin cancer (NMSC) risk associated with abatacept treatment for rheumatoid arthritis (RA). METHODS This evaluation included 16 abatacept RA clinical trials and 6 observational studies. NMSC incidence rates (IRs)/1000 patient-years (p-y) of exposure were compared between patients treated with abatacept versus placebo, conventional synthetic (cs) disease-modifying antirheumatic drugs (DMARDs) and other biological/targeted synthetic (b/ts)DMARDs. For observational studies, a random-effects model was used to pool rate ratios (RRs). RESULTS ~49 000 patients receiving abatacept were analysed from clinical trials (~7000) and observational studies (~42 000). In randomised trials (n=4138; median abatacept exposure, 12 (range 2-30) months), NMSC IRs (95% CIs) were not significantly different for abatacept (6.0 (3.3 to 10.0)) and placebo (4.0 (1.3 to 9.3)) and remained stable throughout the long-term, open-label period (median cumulative exposure, 28 (range 2-130 months); 21 335 p-y of exposure (7044 patients over 3 years)). For registry databases, NMSC IRs/1000 p-y were 5-12 (abatacept), 1.6-10 (csDMARDs) and 3-8 (other b/tsDMARDs). Claims database IRs were 19-22 (abatacept), 15-18 (csDMARDs) and 14-17 (other b/tsDMARDs). Pooled RRs (95% CIs) from observational studies for NMSC in patients receiving abatacept were 1.84 (1.00 to 3.37) vs csDMARDs and 1.11 (0.98 to 1.26) vs other b/tsDMARDs. CONCLUSIONS Consistent with the warnings and precautions of the abatacept label, this analysis suggests a potential increase in NMSC risk with abatacept use compared with csDMARDs. No significant increase was observed compared with b/tsDMARDs, but the lower limit of the 95% CI was close to unity.
Collapse
Affiliation(s)
- Teresa A Simon
- Global Pharmacovigilance and Epidemiology, Bristol Myers Squibb, Princeton, New Jersey, USA
| | - Lixian Dong
- Global Pharmacovigilance and Epidemiology, Bristol Myers Squibb, Princeton, New Jersey, USA
| | - Samy Suissa
- Department of Epidemiology and Biostatistics, McGill University, Montreal, Québec, Canada
- Department of Medicine, McGill University, Montreal, Québec, Canada
| | - Kaleb Michaud
- Internal Medicine, Division of Rheumatology, University of Nebraska Medical Center, Omaha, Nebraska, USA
- FORWARD, The National Databank for Rheumatic Diseases, Wichita, Kansas, USA
| | - Sofia Pedro
- FORWARD, The National Databank for Rheumatic Diseases, Wichita, Kansas, USA
| | - Marc Hochberg
- Department of Medicine, University of Maryland, School of Medicine, Baltimore, Maryland, USA
| | - Maarten Boers
- Department of Epidemiology and Data Science, Amsterdam UMC - Vrije Universiteit, Amsterdam, The Netherlands
| | - Johan Askling
- Rheumatology, Theme Inflammation and Ageing, Karolinska University Hospital, Stockholm, Sweden
- Clinical Epidemiology Division, Karolinska Institutet, Stockholm, Sweden
| | - Thomas Frisell
- Clinical Epidemiology Division, Karolinska Institutet, Stockholm, Sweden
| | - Anja Strangfeld
- Epidemiology and Health Services Research, German Rheumatism Research Center, Berlin, Germany
- Pharmakoepidemiologie, Charité University Medicine, Berlin, Germany
| | - Yvette Meissner
- Epidemiology and Health Services Research, German Rheumatism Research Center, Berlin, Germany
| | - Vadim Khaychuk
- US Medical Immunology and Fibrosis, Bristol Myers Squibb, Princeton, New Jersey, USA
| | - Alyssa Dominique
- Global Pharmacovigilance and Epidemiology, Bristol Myers Squibb, Princeton, New Jersey, USA
| | | |
Collapse
|
19
|
da Rosa LC, Scales HE, Benson RA, Brewer JM, McInnes IB, Garside P. The effect of abatacept on T-cell activation is not long-lived in vivo. DISCOVERY IMMUNOLOGY 2024; 3:kyad029. [PMID: 38567291 PMCID: PMC10917171 DOI: 10.1093/discim/kyad029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Revised: 12/06/2023] [Accepted: 01/03/2024] [Indexed: 04/04/2024]
Abstract
Abatacept, a co-stimulatory blocker comprising the extracellular portion of human CTLA-4 linked to the Fc region of IgG1, is approved for the treatment of rheumatoid arthritis. By impairing the interaction between CD28 on T cells and CD80/CD86 on APCs, its mechanisms of action include the suppression of follicular T helper cells (preventing the breach of self-tolerance in B cells), inhibition of cell cycle progression holding T cells in a state described as 'induced naïve' and reduction in DC conditioning. However, less is known about how long these inhibitory effects might last, which is a critical question for therapeutic use in patients. Herein, employing a murine model of OVA-induced DTH, we demonstrate that the effect of abatacept is short-lived in vivo and that the inhibitory effects diminish markedly when treatment is ceased.
Collapse
Affiliation(s)
- Larissa C da Rosa
- School of Infection & Immunity, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8TA, UK
| | - Hannah E Scales
- School of Infection & Immunity, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8TA, UK
| | - Robert A Benson
- School of Infection & Immunity, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8TA, UK
| | - James M Brewer
- School of Infection & Immunity, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8TA, UK
| | - Iain B McInnes
- School of Infection & Immunity, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8TA, UK
| | - Paul Garside
- School of Infection & Immunity, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8TA, UK
| |
Collapse
|
20
|
Eder L, Mylvaganam S, Pardo Pardo J, Petkovic J, Strand V, Mease P, Colaco K. Sex-related differences in patient characteristics, and efficacy and safety of advanced therapies in randomised clinical trials in psoriatic arthritis: a systematic literature review and meta-analysis. THE LANCET. RHEUMATOLOGY 2023; 5:e716-e727. [PMID: 38251562 DOI: 10.1016/s2665-9913(23)00264-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Revised: 09/19/2023] [Accepted: 09/20/2023] [Indexed: 01/23/2024]
Abstract
BACKGROUND Sex-related differences in clinical manifestations and disease outcomes exist in psoriatic arthritis, however, there is limited information on sex-related differences in randomised controlled trials of psoriatic arthritis. We aimed to compare patient characteristics and efficacy and safety of advanced therapies (including biological and targeted synthetic therapies) between male and female patients with psoriatic arthritis participating in randomised controlled trials. METHODS In this systematic review and meta-analysis, we searched Medline, Embase, and Central databases, and conference abstract archives, from their inception to June 10, 2022, for randomised controlled trials that assessed the efficacy of advanced therapies in psoriatic arthritis. Two reviewers extracted information on participants' characteristics and rates of American College of Rheumatology (ACR) 20 and ACR50 response and minimal disease activity (MDA) by sex. Random-effects models were used to calculate pooled effects of ACR20, ACR50, and MDA in male versus female patients by drug class. FINDINGS We included 54 trials (11 514 [50·9%] of 22 621 participants were female and 11 107 [49·1%] were male). Sex-disaggregated results were reported in a minority of studies (nine [17%] of 54 reported baseline characteristics by sex, 18 [33%] reported efficacy by sex, and two [4%] reported safety endpoints by sex). At baseline, male patients had lower baseline tender joint count (mean difference -3·01 [95% CI -3·83 to -2·18], health assessment questionnaire scores (-0·28 [-0·33 to -0·24]), pain scores (-4·58 [-6·86 to -2·30]), patient global assessment (-3·22 [-5·27 to -1·17]), and physician global assessment (-1·34 [-2·08 to -0·08]) than did female patients. Male patients had higher baseline psoriasis area and severity index scores (mean difference 1·95 [95% CI 0·78 to 3·11]) and C-reactive protein concentrations (2·57 [0·40 to 4·74]) than did female patients. ACR20 response by sex varied across drug classes, with higher rates in males than females with interleukin (IL)-17 inhibitors (odds ratio [OR] 1·70 [95% CI 1·38-2·11]), IL-23 inhibitor (1·46 [1·20-1·78]), IL-12 and IL-23 inhibitor (2·67 [1·39-5·09]), and tumour necrosis factor (TNF) inhibitors (1·55 [1·11-2·18]), but no difference with JAK and TYK2 inhibitors (1·10 [0·87-1·38]). Similarly, ACR50 response rates were higher in male patients versus female patients in all drug classes, with exception of JAK and TYK2 inhibitors (TNF inhibitors, OR 2·17 [95% CI 1·62-2·90]; IL-17 inhibitors, 1·93 [1·56-2·38]; IL-23 inhibitor, 1·71 [1·25-2·34]; IL-12 and 23 inhibitor, 2·43 [1·14-5·20]; and JAK and TYK2 inhibitors, 1·09 [0·73-1·62]). Male patients were more likely to reach MDA with most drug classes, including IL-17 inhibitors (OR 1·99 [95% CI 1·50-2·63]), IL-23 inhibitors (1·79 [1·29-2·50]), TNF inhibitors (2·62 [1·54-4·44]), and JAK and TYK2 inhibitors (1·77 [1·15-2·73]). Risk of bias was low for most studies. INTERPRETATION Biological sex of patients with psoriatic arthritis influences their response to advanced therapies, but the effect varies by drug class. Selective reporting might have influenced these results. Future trials should report baseline characteristics and endpoint results by sex. FUNDING Canadian Rheumatology Association.
Collapse
Affiliation(s)
- Lihi Eder
- Women's College Research Institute and Department of Medicine, University of Toronto, Toronto, ON, Canada.
| | | | - Jordi Pardo Pardo
- Cochrane Equity Thematic Network, University of Ottawa, Ottawa, ON, Canada
| | - Jennifer Petkovic
- Cochrane Equity Thematic Network, University of Ottawa, Ottawa, ON, Canada
| | - Vibeke Strand
- Division of Immunology and Rheumatology, Stanford University, Stanford, CA, USA
| | - Philip Mease
- Swedish Medical Center, Seattle, WA, USA; Providence St Joseph Hospital, Seattle, WA, USA; University of Washington School of Medicine, Seattle, WA, USA
| | - Keith Colaco
- Women's College Research Institute and Department of Medicine, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
21
|
Nash P, Dutz JP, Peterson S, Patel BP, Eaton K, Shawi M, Zazzetti F, Wei JCC. Systematic literature review and network meta-analysis of therapies for psoriatic arthritis on patient-reported outcomes. BMJ Open 2023; 13:e062306. [PMID: 37940157 PMCID: PMC10632897 DOI: 10.1136/bmjopen-2022-062306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Accepted: 06/26/2023] [Indexed: 11/10/2023] Open
Abstract
OBJECTIVES Head-to-head clinical trials are common in psoriasis, but scarce in psoriatic arthritis (PsA), making treatment comparisons between therapeutic classes difficult. This study describes the relative effectiveness of targeted synthetic (ts) and biologic (b) disease-modifying antirheumatic drugs (DMARDs) on patient-reported outcomes (PROs) through network meta-analysis (NMA). DESIGN A systematic literature review (SLR) was conducted in January 2020. Bayesian NMAs were conducted to compare treatments on Health Assessment Questionnaire Disability Index (HAQ-DI) and 36-item Short Form (SF-36) Health Survey including Mental Component Summary (MCS) and Physical Component Summary (PCS) scores. DATA SOURCES Ovid MEDLINE (including Epub Ahead of Print, In-Process & Other Non-Indexed Citations and Daily),Embase and Cochrane Central Register of Controlled Trials. ELIGIBILITY CRITERIA Phase III randomised controlled trials (RCTs) evaluating patients with PsA receiving tsDMARDS, bDMARDs or placebo were included in the SLR; there was no restriction on outcomes. DATA EXTRACTION AND SYNTHESIS Two independent researchers reviewed all citations. Data for studies meeting all inclusion criteria were extracted into a standardised Excel-based form by one reviewer and validated by a second reviewer. A third reviewer was consulted to resolve any discrepancies, as necessary. Risk of bias was assessed using the The National Institute for Health and Care Excellence clinical effectiveness quality assessment checklist. RESULTS In total, 26 RCTs were included. For HAQ-DI, SF-36 PCS and SF-36 MCS scores, intravenous tumour necrosis factor (TNF) alpha inhibitors generally ranked higher than most other classes of therapies available to treat patients with PsA. For almost all outcomes, several interleukin (IL)-23, IL-17A, subcutaneous TNF and IL-12/23 agents offered comparable improvement, while cytotoxic T-lymphocyte-associated antigen 4, phosphodiesterase-4 and Janus kinase inhibitors often had the lowest efficacy. CONCLUSIONS While intravenous TNFs may provide some improvements in PROs relative to several other tsDMARDs and bDMARDs for the treatment of patients with PsA, differences between classes of therapies across outcomes were small.
Collapse
Affiliation(s)
- Peter Nash
- School of Medicine, Griffith University, Brisbane, Queensland, Australia
| | - Jan P Dutz
- Department of Dermatology and Skin Sciences, The University of British Columbia, Vancouver, British Columbia, Canada
| | - Steve Peterson
- Immunology Global Commercial Strategy Organization, Janssen Global Services LLC, Horsham, Pennsylvania, USA
| | | | | | - May Shawi
- Immunology Medical Affairs, Janssen Global Services LLC, Horsham, Pennsylvania, USA
| | - Federico Zazzetti
- Immunology Medical Affairs, Janssen Latin America, LLC, Buenos Aires, Argentina
| | - James Cheng-Chung Wei
- Department of Allergy Immunology & Rheumatology, Chung Shan Medical University Hospital, Taichung, Taiwan
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan
- Graduate Institute of Integrated Medicine, China Medical University, Taichung, Taiwan
| |
Collapse
|
22
|
Lee WS, Nam KH, Kim JH, Kim WJ, Kim JE, Shin EC, Kim GR, Choi JM. Alleviating psoriatic skin inflammation through augmentation of Treg cells via CTLA-4 signaling peptide. Front Immunol 2023; 14:1233514. [PMID: 37818377 PMCID: PMC10560854 DOI: 10.3389/fimmu.2023.1233514] [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: 06/02/2023] [Accepted: 09/11/2023] [Indexed: 10/12/2023] Open
Abstract
Psoriasis is a chronic inflammatory skin disease characterized by hyperplasia of keratinocytes and immune cell infiltration. The IL-17-producing T cells play a key role in psoriasis pathogenesis, while regulatory T (Treg) cells are diminished during psoriatic inflammation. Current psoriasis treatments largely focus on IL-17 and IL-23, however, few studies have explored therapeutic drugs targeting an increase of Treg cells to control immune homeostasis. In this study, we investigated the effects of a cytotoxic T lymphocyte antigen-4 (CTLA-4) signaling peptide (dNP2-ctCTLA-4) in Th17, Tc17, γδ T cells, Treg cells in vitro and a mouse model of psoriasis. Treatment with dNP2-ctCTLA-4 peptide showed a significant reduction of psoriatic skin inflammation with increased Treg cell proportion and reduced IL-17 production by T cells, indicating a potential role in modulating psoriatic skin disease. We compared dNP2-ctCTLA-4 with CTLA-4-Ig and found that only dNP2-ctCTLA-4 ameliorated the psoriasis progression, with increased Treg cells and inhibited IL-17 production from γδ T cells. In vitro experiments using a T cell-antigen presenting cell co-culture system demonstrated the distinct mechanisms of dNP2-ctCTLA-4 compared to CTLA-4-Ig in the induction of Treg cells. These findings highlight the therapeutic potential of dNP2-ctCTLA-4 peptide in psoriasis by augmenting Treg/Teff ratio, offering a new approach to modulating the disease.
Collapse
Affiliation(s)
- Woo-Sung Lee
- Department of Life Science, College of Natural Sciences, Hanyang University, Seoul, Republic of Korea
| | - Kyung-Ho Nam
- Department of Life Science, College of Natural Sciences, Hanyang University, Seoul, Republic of Korea
| | - Jong Hoon Kim
- Department of Dermatology and Cutaneous Biology Research Institute, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Won-Ju Kim
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, United States
| | - Jeong Eun Kim
- Department of Dermatology, Hanyang University College of Medicine, Seoul, Republic of Korea
- Hanyang Institute of Bioscience and Biotechnology, Hanyang University, Seoul, Republic of Korea
| | - Eui-Cheol Shin
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
- The Center for Viral Immunology, Korea Virus Research Institute, Institute for Basic Science (IBS), Daejeon, Republic of Korea
| | - Gil-Ran Kim
- Department of Life Science, College of Natural Sciences, Hanyang University, Seoul, Republic of Korea
- Research Institute for Natural Sciences, Hanyang University, Seoul, Republic of Korea
| | - Je-Min Choi
- Department of Life Science, College of Natural Sciences, Hanyang University, Seoul, Republic of Korea
- Hanyang Institute of Bioscience and Biotechnology, Hanyang University, Seoul, Republic of Korea
- Research Institute for Natural Sciences, Hanyang University, Seoul, Republic of Korea
- Research Institute for Convergence of Basic Sciences, Hanyang University, Seoul, Republic of Korea
| |
Collapse
|
23
|
Kumthekar A, Ashrafi M, Deodhar A. Difficult to treat psoriatic arthritis - how should we manage? Clin Rheumatol 2023; 42:2251-2265. [PMID: 37097525 DOI: 10.1007/s10067-023-06605-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 03/18/2023] [Accepted: 04/16/2023] [Indexed: 04/26/2023]
Abstract
Psoriatic arthritis (PsA) is a chronic, multi-domain immune-mediated inflammatory arthritis with a high disease burden. PsA patients have significant co-morbidities like obesity, depression, fibromyalgia which can impact disease activity assessment. The management of PsA has undergone a paradigm shift over the last decade due to the availability of multiple biologic and targeted synthetic disease modifying anti-rheumatic drugs. Despite the availability of multiple therapeutic agents, it is not uncommon to find patients not responding adequately and continuing to have active disease and/or high disease burden. In our review, we propose what is "difficult to treat PsA", discuss differential diagnosis, commonly overlooked factors, co-morbidities that affect treatment responses, and suggest a stepwise algorithm to manage these patients.
Collapse
Affiliation(s)
- Anand Kumthekar
- Division of Rheumatology, Department of Medicine, Albert Einstein College of Medicine, Montefiore Medical Center, New York, NY, USA.
| | - Maedeh Ashrafi
- Department of Medicine, Jacobi Medical Center / Albert Einstein College of Medicine, New York, NY, USA
| | - Atul Deodhar
- Division of Arthritis and Rheumatic Diseases, Oregon Health & Science University, Portland, OR, USA
| |
Collapse
|
24
|
Ayan G, Ribeiro A, Macit B, Proft F. Pharmacologic Treatment Strategies in Psoriatic Arthritis. Clin Ther 2023; 45:826-840. [PMID: 37455227 DOI: 10.1016/j.clinthera.2023.05.010] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Revised: 05/30/2023] [Accepted: 05/31/2023] [Indexed: 07/18/2023]
Abstract
PURPOSE The goal of this narrative review was to provide current data on psoriatic arthritis (PsA) therapeutic strategies, supporting treatment decisions with a domain-based approach. METHODS This narrative review of treatment strategies for PsA focused on several disease domains (ie, peripheral arthritis, enthesitis, axial disease, dactylitis, skin and nail disease), as well as the so-called "related conditions" of uveitis, Crohn's disease, and ulcerative colitis. We searched PubMed, EMBASE, international guidelines, and recent congress abstracts. FINDINGS Currently, multiple approved treatment options offer a wide range of options, such as tumor necrosis factor (TNF) inhibitors; inhibitors of interleukin-17 (IL-17), IL-12/23 (IL-12/23), IL-23 (IL-23), and Janus kinase; the phosphodiesterase 4 inhibitor apremilast; and the T-cell modulator abatacept. However, no treatment option shows clear superiority concerning efficacy on peripheral arthritis and dactylitis over the others, whereas limited evidence suggests that the IL-17 inhibitor ixekizumab and the IL-12/23 inhibitor ustekinumab may be superior to TNF inhibitors in treating enthesitis. Recent data on enthesitis have also shown promising results for methotrexate. Treatment of axial PsA is mostly derived from axial spondyloarthritis, and more data are needed focusing on this specific subgroup of PsA patients. Thus far, the most important finding from the only randomized controlled trial in this specific population is that the IL-17 inhibitor secukinumab was superior to placebo in terms of clinical and radiologic end-points in axial PsA. Regarding psoriatic skin involvement, head-to-head trials in PsA as well as skin psoriasis showed the superiority of IL-17, IL-23, and IL-12/23 inhibitors over TNF inhibitors. When treating PsA with concurrent uveitis, according to the existing data, monoclonal TNF inhibitor antibodies should be preferred. In PsA and concomitant inflammatory bowel disease, treatment decisions must include the consideration of which specific type of inflammatory bowel disease (Crohn's disease or ulcerative colitis) is present, as some of the agents either lack data or are ineffective in treating these 2 conditions. In both types, IL-17 inhibitors should be avoided. When determining treatment strategy, comorbidities should be carefully assessed, and the corresponding risk profile of the respective treatment modalities should be taken into consideration. IMPLICATIONS There are many approved therapeutic options for treating patients with PsA, and additional emerging treatment options are in the pipeline. Individualized treatment decisions for each patient, depending on the leading disease phenotype, underlying comorbidities, and patient preferences, should be made based on shared decision-making.
Collapse
Affiliation(s)
- G Ayan
- Hacettepe University, Department of Internal Medicine, Division of Rheumatology, Ankara, Turkey
| | - A Ribeiro
- Hospital de Clínicas de Porto Alegre, Department of Rheumatology, Porto Alegre, Brazil
| | - Betul Macit
- Department of Dermatology, The Warren Alpert Medical School of Brown University, Providence, Rhode Island, USA
| | - Fabian Proft
- Department of Gastroenterology, Infectiology and Rheumatology (including Nutrition Medicine), Charité-Universitätsmedizin Berlin, Berlin, Germany.
| |
Collapse
|
25
|
Xue T, Kong X, Ma L. Trends in the Epidemiology of Pneumocystis Pneumonia in Immunocompromised Patients without HIV Infection. J Fungi (Basel) 2023; 9:812. [PMID: 37623583 PMCID: PMC10455156 DOI: 10.3390/jof9080812] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 06/19/2023] [Accepted: 07/19/2023] [Indexed: 08/26/2023] Open
Abstract
The increasing morbidity and mortality of life-threatening Pneumocystis pneumonia (PCP) in immunocompromised people poses a global concern, prompting the World Health Organization to list it as one of the 19 priority invasive fungal diseases, calling for increased research and public health action. In response to this initiative, we provide this review on the epidemiology of PCP in non-HIV patients with various immunodeficient conditions, including the use of immunosuppressive agents, cancer therapies, solid organ and stem cell transplantation, autoimmune and inflammatory diseases, inherited or primary immunodeficiencies, and COVID-19. Special attention is given to the molecular epidemiology of PCP outbreaks in solid organ transplant recipients; the risk of PCP associated with the increasing use of immunodepleting monoclonal antibodies and a wide range of genetic defects causing primary immunodeficiency; the trend of concurrent infection of PCP in COVID-19; the prevalence of colonization; and the rising evidence supporting de novo infection rather than reactivation of latent infection in the pathogenesis of PCP. Additionally, we provide a concise discussion of the varying effects of different immunodeficient conditions on distinct components of the immune system. The objective of this review is to increase awareness and knowledge of PCP in non-HIV patients, thereby improving the early identification and treatment of patients susceptible to PCP.
Collapse
Affiliation(s)
- Ting Xue
- NHC Key Laboratory of Pneumoconiosis, Key Laboratory of Prophylaxis and Treatment and Basic Research of Respiratory Diseases of Shanxi Province, Shanxi Province Key Laboratory of Respiratory, Department of Respiratory and Critical Care Medicine, First Hospital of Shanxi Medical University, Taiyuan 030001, China
| | - Xiaomei Kong
- NHC Key Laboratory of Pneumoconiosis, Key Laboratory of Prophylaxis and Treatment and Basic Research of Respiratory Diseases of Shanxi Province, Shanxi Province Key Laboratory of Respiratory, Department of Respiratory and Critical Care Medicine, First Hospital of Shanxi Medical University, Taiyuan 030001, China
| | - Liang Ma
- Critical Care Medicine Department, NIH Clinical Center, Bethesda, MD 20892, USA
| |
Collapse
|
26
|
Denis A, Sztejkowski C, Arnaud L, Becker G, Felten R. The 2023 pipeline of disease-modifying antirheumatic drugs (DMARDs) in clinical development for spondyloarthritis (including psoriatic arthritis): a systematic review of trials. RMD Open 2023; 9:e003279. [PMID: 37507210 PMCID: PMC10387652 DOI: 10.1136/rmdopen-2023-003279] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Accepted: 07/15/2023] [Indexed: 07/30/2023] Open
Abstract
OBJECTIVES The objective of this systematic review was to provide an overview of current developments and potentially available therapeutic options for spondyloarthritis (SpA) in the coming years. METHODS We conducted a systematic review of 17 national and international clinical trial databases for all disease-modifying antirheumatic drugs (DMARDs) for SpA that are already marketed, in clinical development or withdrawn. The search was performed on February 2023 with the keywords "spondyloarthritis", "ankylosing spondylitis" and "psoriatic arthritis". For each molecule, we only considered the study at the most advanced stage of clinical development. RESULTS Concerning axial SpA (axSpA), a total of 44 DMARDs were identified: 6 conventional synthetic DMARDs (csDMARDs), 27 biological DMARDs (bDMARDs) and 11 targeted synthetic DMARDs (tsDMARDs). Among the 18 targeted treatments (b+tsDMARDs) in current development, corresponding trials reached phase I (n=1), II (n=10) and III (n=7). Ten molecules are IL-17 inhibitors, two Janus kinase (JAK) inhibitors and two granulocyte-macrophage colony-stimulating factor inhibitors; four have another mode of action. Concerning psoriatic arthritis (PsA), 44 DMARDs were identified: 5 csDMARDs, 27 bDMARDs and 12 tsDMARDs. Among the 15 molecules in current development, corresponding trials reached phase II (n=8) and III (n=7). Six molecules are JAK inhibitors, six IL-17 inhibitors and one an IL-23 inhibitor; two have another mode of action. CONCLUSION This systematic review identified 18 and 15 molecules in clinical development for axSpA and PsA, respectively, which suggests a strengthening of the therapeutic arsenal in the coming years. However, with so many DMARDs but low target diversity, we will need to develop strategies or biomarkers to help clinicians make informed treatment decisions.
Collapse
Affiliation(s)
- Agathe Denis
- Service de Rhumatologie de Hautepierre, RESO, Centre de Référence des Maladies Autoimmunes Systémiques Rares Est Sud-Ouest, Hôpitaux Universitaires de Strasbourg, Strasbourg, France
| | - Cédric Sztejkowski
- Service de Rhumatologie de Hautepierre, RESO, Centre de Référence des Maladies Autoimmunes Systémiques Rares Est Sud-Ouest, Hôpitaux Universitaires de Strasbourg, Strasbourg, France
| | - Laurent Arnaud
- Service de Rhumatologie de Hautepierre, RESO, Centre de Référence des Maladies Autoimmunes Systémiques Rares Est Sud-Ouest, Hôpitaux Universitaires de Strasbourg, Strasbourg, France
| | - Guillaume Becker
- Pôle Pharmacie-Pharmacologie, Hôpitaux universitaires de Strasbourg, Strasbourg, France
| | - Renaud Felten
- Service de Rhumatologie de Hautepierre, RESO, Centre de Référence des Maladies Autoimmunes Systémiques Rares Est Sud-Ouest, Hôpitaux Universitaires de Strasbourg, Strasbourg, France
- Centre d'Investigation Clinique, Inserm 1434, INSERM, Strasbourg, France
| |
Collapse
|
27
|
Edwards A, Chandran V, Rahman P. Investigational monoclonal antibodies in early development for psoriatic arthritis: beyond the biosimilars. Expert Opin Investig Drugs 2023; 32:741-753. [PMID: 37655430 DOI: 10.1080/13543784.2023.2254684] [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: 05/22/2023] [Revised: 08/08/2023] [Accepted: 08/30/2023] [Indexed: 09/02/2023]
Abstract
INTRODUCTION Psoriatic Arthritis (PsA) is an inflammatory arthritis that is present in approximately 25% of psoriasis patients. Currently, several targeted therapies are available to manage PsA; however, many patients fail these therapies. Several new therapeutic options, with differing mechanisms of action, are currently being evaluated. AREAS COVERED This article reviews available results from phase I to phase III trials of several investigational monoclonal antibodies that the FDA has not yet approved for PsA. The proposed mechanisms of the new therapeutic agents and their relevance to the pathogenesis of PsA will be discussed. The investigational agents' efficacy and safety will be summarized, and their potential clinical applications for managing PsA will be contemplated. EXPERT OPINION Due to recent advances in understanding psoriatic arthritis, therapeutic agents are increasingly focused on inhibiting interleukin-17 and interleukin-23 pathways. Various strategies have been used to inhibit these cytokines, demonstrating favorable efficacy and acceptable safety profile.
Collapse
Affiliation(s)
- Anna Edwards
- Faculty of Pharmacy, Memorial University of Newfoundland, St. John's, Newfoundland, Canada
| | - Vinod Chandran
- Department of Medicine, Division of Rheumatology, University of Toronto, Toronto, Ontario, Canada
| | - Proton Rahman
- Department of Medicine, Division of Rheumatology, Memorial University of Newfoundland, St. John's, Newfoundland, Canada
| |
Collapse
|
28
|
Tian X, Li M, Liu S, Leng X, Wang Q, Zhao J, Liu Y, Zhao Y, Zhang Y, Xu H, Gu J, Zeng X. Consensus on targeted drug therapy for spondyloarthritis. RHEUMATOLOGY AND IMMUNOLOGY RESEARCH 2023; 4:47-59. [PMID: 37485474 PMCID: PMC10362604 DOI: 10.2478/rir-2023-0009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Accepted: 06/26/2023] [Indexed: 07/25/2023]
Abstract
Spondyloarthritis (SpA) is a group of chronic inflammatory diseases that predominantly involve the spine and/or peripheral joints. The clinical manifestations of SpA are highly heterogenous and complicated with various comorbidities. SpA is a disabling disease and adversely affects the quality of life of patients. Many new medications that target cytokines or pathways specific for the pathogenesis of SpA have been developed and they are becoming increasingly important in the treatment of SpA. However, identifying the target patient population and standardizing the usage of these drugs are critical issues in the clinical application of these "targeted therapeutic drugs". Under the leadership of National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), managed by Peking Union Medical College Hospital, the "Consensus on targeted drug therapy for spondyloarthritis" has been developed in collaboration with the Rheumatology and Immunology Physicians Committee, Chinese Medical Doctors Association, Rheumatology and Immunology Professional Committee, Chinese Association of Rehabilitation Medicine, and Chinese Research Hospital Association Rheumatology and Immunology Professional Committee. This consensus has been developed with evidence-based methodology and has followed the international standard for consensus development.
Collapse
Affiliation(s)
- Xinping Tian
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, National Clinical Research Center for Dermatologic and Immunologic Diseases, Ministry of Science & Technology, State Key Laboratory of Complex Severe and Rare Diseases, Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Beijing100730, China
| | - Mengtao Li
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, National Clinical Research Center for Dermatologic and Immunologic Diseases, Ministry of Science & Technology, State Key Laboratory of Complex Severe and Rare Diseases, Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Beijing100730, China
| | - Shengyun Liu
- Department of Rheumatology and Immunology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou450052, Henan Province, China
| | - Xiaomei Leng
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, National Clinical Research Center for Dermatologic and Immunologic Diseases, Ministry of Science & Technology, State Key Laboratory of Complex Severe and Rare Diseases, Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Beijing100730, China
| | - Qian Wang
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, National Clinical Research Center for Dermatologic and Immunologic Diseases, Ministry of Science & Technology, State Key Laboratory of Complex Severe and Rare Diseases, Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Beijing100730, China
| | - Jiuliang Zhao
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, National Clinical Research Center for Dermatologic and Immunologic Diseases, Ministry of Science & Technology, State Key Laboratory of Complex Severe and Rare Diseases, Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Beijing100730, China
| | - Yi Liu
- Department of Rheumatology and Immunology, West China Hospital Sichuan University, Chengdu610041, Sichuan Province, China
| | - Yan Zhao
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, National Clinical Research Center for Dermatologic and Immunologic Diseases, Ministry of Science & Technology, State Key Laboratory of Complex Severe and Rare Diseases, Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Beijing100730, China
| | - Yizhi Zhang
- Department of Rheumatology and Immunology, the First Affiliated Hospital of Harbin Medical University, Haerbin150001, Heilongjiang Province, China
| | - Huji Xu
- Department of Rheumatology and Immunology, Shanghai Changzheng Hopital, Shanghai200003, China
| | - Jieruo Gu
- Department of Rheumatology and Immunology, the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou510630, Guangdong Province, China
| | - Xiaofeng Zeng
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, National Clinical Research Center for Dermatologic and Immunologic Diseases, Ministry of Science & Technology, State Key Laboratory of Complex Severe and Rare Diseases, Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Beijing100730, China
| |
Collapse
|
29
|
Torosian K, Lal E, Kavanaugh A, Loomba R, Ajmera V, Guma M. Psoriatic disease and non-alcoholic fatty liver disease shared pathogenesis review. Semin Arthritis Rheum 2023; 59:152165. [PMID: 36716599 PMCID: PMC9992353 DOI: 10.1016/j.semarthrit.2023.152165] [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: 09/30/2022] [Revised: 12/03/2022] [Accepted: 01/04/2023] [Indexed: 01/20/2023]
Abstract
Psoriatic disease (PD) and non-alcoholic fatty liver disease (NAFLD) potentially share disease pathways given the numerous inflammatory pathways involved in both diseases and a higher prevalence of NAFLD in PD patients. Metabolic syndrome and obesity are a key link between the two diseases, but even when controlling for this, associations between both diseases are still seen. Therapeutics that impact metabolic or inflammatory pathways may be impactful in both PD and NAFLD. In this review, we describe common inflammatory pathways contributing to both PD and NAFLD and critically review the potential impact of treatments for and on both diseases.
Collapse
Affiliation(s)
- Kelly Torosian
- Department of Medicine, School of Medicine, University of California, San Diego, 9500 Gilman Drive, San Diego, CA 92093, USA
| | - Esha Lal
- Department of Medicine, School of Medicine, University of California, San Diego, 9500 Gilman Drive, San Diego, CA 92093, USA
| | - Arthur Kavanaugh
- Department of Rheumatology, University of California, San Diego, 9500 Gilman Drive, San Diego, CA 92093, USA
| | - Rohit Loomba
- Division of Gastroenterology and Hepatology, University of California, San Diego, 9500 Gilman Drive, San Diego, CA 92093, USA; NAFLD Research Center, Department of Medicine, University of California at San Diego, La Jolla, USA; Division of Epidemiology, Department of Family and Preventative Medicine, University of California at San Diego, La Jolla, USA
| | - Veeral Ajmera
- Division of Gastroenterology and Hepatology, University of California, San Diego, 9500 Gilman Drive, San Diego, CA 92093, USA; NAFLD Research Center, Department of Medicine, University of California at San Diego, La Jolla, USA.
| | - Monica Guma
- Department of Rheumatology, University of California, San Diego, 9500 Gilman Drive, San Diego, CA 92093, USA; Department of Medicine, Autonomous University of Barcelona, Plaça Cívica, 08193 Bellaterra, Barcelona, Spain; San Diego VA Healthcare Service, San Diego, CA, 92161, USA.
| |
Collapse
|
30
|
Sundanum S, Orr C, Veale D. Targeted Therapies in Psoriatic Arthritis-An Update. Int J Mol Sci 2023; 24:6384. [PMID: 37047357 PMCID: PMC10094037 DOI: 10.3390/ijms24076384] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 03/19/2023] [Accepted: 03/21/2023] [Indexed: 03/31/2023] Open
Abstract
Psoriatic arthritis (PsA) is a systemic inflammatory condition characterised by multiple clinical manifestations. Over the last decade, significant progress has been made in understanding the pathobiology of the disease. An expanded set of targeted therapies have emerged and have shown efficacy in PsA. Nevertheless, there is still a substantial subset of patients who experience no response or only a partial response to currently licensed therapies. The heterogeneous nature of the disease, together with a varying level of severity at presentation and disease activity during follow-up, brings tremendous challenges to devising management strategies. While there are certain pathophysiological similarities between PsA and rheumatoid arthritis (RA), it has become clear that there are discriminating features between these two conditions at the clinical, cellular, and molecular levels. However, there is a degree of overlap in the clinical approach when treating both PsA and RA, given that many biological and targeted therapies have proven efficacy for both pathologies. With an increasing understanding of the relevance of the IL-23/IL-17 axis in PsA, pharmacological agents blocking this pathway have provided promising possibilities for patients with PsA.
Collapse
Affiliation(s)
| | | | - Douglas Veale
- Centre for Arthritis and Rheumatic Diseases, Dublin Academic Medical Centre, University College Dublin, D04 V1W8 Dublin, Ireland
| |
Collapse
|
31
|
Konen FF, Möhn N, Witte T, Schefzyk M, Wiestler M, Lovric S, Hufendiek K, Schwenkenbecher P, Sühs KW, Friese MA, Klotz L, Pul R, Pawlitzki M, Hagin D, Kleinschnitz C, Meuth SG, Skripuletz T. Treatment of autoimmunity: The impact of disease-modifying therapies in multiple sclerosis and comorbid autoimmune disorders. Autoimmun Rev 2023; 22:103312. [PMID: 36924922 DOI: 10.1016/j.autrev.2023.103312] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Accepted: 03/09/2023] [Indexed: 03/17/2023]
Abstract
More than 10 disease-modifying therapies (DMT) are approved by the European Medicines Agency (EMA) and the US Food and Drug Administration (FDA) for the treatment of multiple sclerosis (MS) and new therapeutic options are on the horizon. Due to different underlying therapeutic mechanisms, a more individualized selection of DMTs in MS is possible, taking into account the patient's current situation. Therefore, concomitant treatment of various comorbid conditions, including autoimmune mediated disorders such as rheumatoid arthritis, should be considered in MS patients. Because the pathomechanisms of autoimmunity partially overlap, DMT could also treat concomitant inflammatory diseases and simplify the patient's treatment. In contrast, the exacerbation and even new occurrence of several autoimmune diseases have been reported as a result of immunomodulatory treatment of MS. To simplify treatment and avoid disease exacerbation, knowledge of the beneficial and adverse effects of DMT in other autoimmune disorders is critical. Therefore, we conducted a literature search and described the beneficial and adverse effects of approved and currently studied DMT in a large number of comorbid autoimmune diseases, including rheumatoid arthritis, ankylosing spondylitis, inflammatory bowel diseases, cutaneous disorders including psoriasis, Sjögren´s syndrome, systemic lupus erythematosus, systemic vasculitis, autoimmune hepatitis, and ocular autoimmune disorders. Our review aims to facilitate the selection of an appropriate DMT in patients with MS and comorbid autoimmune diseases.
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 and Venerology, Hannover Medical School, 30625 Hannover, Germany..
| | - Miriam Wiestler
- Department of Internal Medicine, Division of Gastroenterology, Hepatology 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.
| | | | - 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, Essen 45147, 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, Essen 45147, 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
|
32
|
Richette P, Vis M, Ohrndorf S, Tillett W, Ramírez J, Neuhold M, van Speybroeck M, Theander E, Noel W, Zimmermann M, Shawi M, Kollmeier A, Zabotti A. Identification of PsA phenotypes with machine learning analytics using data from two phase III clinical trials of guselkumab in a bio-naïve population of patients with PsA. RMD Open 2023; 9:e002934. [PMID: 37001920 PMCID: PMC10069583 DOI: 10.1136/rmdopen-2022-002934] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Accepted: 02/21/2023] [Indexed: 04/03/2023] Open
Abstract
OBJECTIVES Psoriatic arthritis (PsA) phenotypes are typically defined by their clinical components, which may not reflect patients' overlapping symptoms. This post hoc analysis aimed to identify hypothesis-free PsA phenotype clusters using machine learning to analyse data from the phase III DISCOVER-1/DISCOVER-2 clinical trials. METHODS Pooled data from bio-naïve patients with active PsA receiving guselkumab 100 mg every 8/4 weeks were retrospectively analysed. Non-negative matrix factorisation was applied as an unsupervised machine learning technique to identify PsA phenotype clusters; baseline patient characteristics and clinical observations were input features. Minimal disease activity (MDA), disease activity index for psoriatic arthritis (DAPSA) low disease activity (LDA) and DAPSA remission at weeks 24 and 52 were evaluated. RESULTS Eight clusters (n=661) were identified: cluster 1 (feet dominant), cluster 2 (male, overweight, psoriasis dominant), cluster 3 (hand dominant), cluster 4 (dactylitis dominant), cluster 5 (enthesitis, large joints), cluster 6 (enthesitis, small joints), cluster 7 (axial dominant) and cluster 8 (female, obese, large joints). At week 24, MDA response was highest in cluster 2 and lowest in clusters 3, 5 and 6; at week 52, it was highest in cluster 2 and lowest in cluster 5. At weeks 24 and 52, DAPSA LDA and remission were highest in cluster 2 and lowest in clusters 4 and 6, respectively. All clusters improved with guselkumab treatment over 52 weeks. CONCLUSIONS Unsupervised machine learning identified eight PsA phenotype clusters with significant differences in demographics, clinical features and treatment responses. In the future, such data could help support individualised treatment decisions.
Collapse
Affiliation(s)
- Pascal Richette
- Rheumatology Department, AP-HP, Lariboisière Hospital, INSERM U1132, Paris, France
- Université Paris Cité, Paris, France
| | - Marijn Vis
- Rheumatology, Erasmus MC Universitair Medisch Centrum, Rotterdam, The Netherlands
| | - Sarah Ohrndorf
- Department of Rheumatology and Clinical Immunology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - William Tillett
- Department of Rheumatology, Royal National Hospital for Rheumatic Diseases, Bath, UK
- Department of Life Sciences, Centre for Therapeutic Innovation, University of Bath, Bath, UK
| | - Julio Ramírez
- Rheumatology Department, Hospital Universitari Clínic and IDIBAPS, Universidad de Barcelona, Barcelona, Spain
| | - Marlies Neuhold
- EMEA Medical Affairs Immunology, Janssen-Cilag, Zug, Switzerland
| | | | - Elke Theander
- EMEA Medical Affairs Immunology, Janssen-Cilag, Solna, Sweden
| | - Wim Noel
- EMEA Medical Affairs Immunology, Janssen-Cilag BV, Breda, The Netherlands
| | | | - May Shawi
- Medical Affairs, Janssen Global Services LLC, Horsham, Pennsylvania, USA
| | - Alexa Kollmeier
- Research and Development, Immunology, Janssen Research and Development LLC, San Diego, California, USA
| | - Alen Zabotti
- Department of Medical and Biological Sciences, Rheumatology Institute, University of Udine, Udine, Italy
| |
Collapse
|
33
|
Dauth S, Klippstein M, Köhm M. [Psoriatic arthritis : Clinical challenges and pharmaceutical management]. Z Rheumatol 2023; 82:220-232. [PMID: 36856805 DOI: 10.1007/s00393-023-01326-5] [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: 01/16/2023] [Indexed: 03/02/2023]
Abstract
Psoriatic arthritis (PsA) is a systemic immune-mediated inflammatory disease of the musculoskeletal system, which is accompanied by a chronic and progressive course. It is characterized by different clinical manifestations and can severely impair the quality of life and function of patients due to the existing heterogeneity of the manifestations. The (early) diagnosis of PsA and individualized therapeutic management in routine clinical practice are difficult due to the enormous clinical variability. In addition to the appearance of arthritis of the peripheral joints, there can be involvement of the axial skeleton, skin psoriasis, nail psoriasis, enthesitis and dactylitis. The clinical appearance, course of the disease, risk factors and pathophysiological mechanisms of PsA have been extensively researched in recent decades. With the associated better understanding of the disease, new treatment options and goals for effective treatment have also been established.
Collapse
Affiliation(s)
- Stephanie Dauth
- Fraunhofer-Institut für Translationale Medizin und Pharmakologie (ITMP), Frankfurt/Main, Deutschland
- Fraunhofer Exzellenzcluster für Immunmediierte Erkrankungen (CIMD), Frankfurt/Main, Deutschland
| | - Maximilian Klippstein
- Fraunhofer-Institut für Translationale Medizin und Pharmakologie (ITMP), Frankfurt/Main, Deutschland
- Fraunhofer Exzellenzcluster für Immunmediierte Erkrankungen (CIMD), Frankfurt/Main, Deutschland
| | - Michaela Köhm
- Fraunhofer-Institut für Translationale Medizin und Pharmakologie (ITMP), Frankfurt/Main, Deutschland.
- Fraunhofer Exzellenzcluster für Immunmediierte Erkrankungen (CIMD), Frankfurt/Main, Deutschland.
- Abteilung Rheumatologie, Goethe-Universität Frankfurt/Main, Theodor-Stern-Kai 7, 60596, Frankfurt/Main, Deutschland.
| |
Collapse
|
34
|
Zhu W, Ayoub S, Morand E, Tillett W, Antony A. The evolving demographics of participants in psoriatic arthritis phase III randomised controlled trials of b/tsDMARDs: A systematic review. Semin Arthritis Rheum 2023; 60:152175. [PMID: 36803867 DOI: 10.1016/j.semarthrit.2023.152175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Revised: 01/21/2023] [Accepted: 02/06/2023] [Indexed: 02/16/2023]
Abstract
OBJECTIVES To characterize the evolving demographics of participants recruited to phase III randomised controlled trials (RCTs) of biologic/targeted synthetic disease-modifying anti-rheumatic drugs (b/tsDMARDs) in peripheral psoriatic arthritis (PsA). METHODS We conducted a systematic review of EMBASE, MEDLINE, and the Cochrane Database of Clinical Trials (CENTRAL) to identify all placebo-controlled phase III RCTs of b/tsDMARDs in peripheral PsA published up to 1 June 2022. Data extracted included inclusion criteria, date of initiation, countries in which studies were conducted, age, sex, race, disease duration, swollen joint count, tender joint count, Health Assessment Questionnaire - Disability Index, Psoriasis Area and Severity Index, and radiographic damage scores. Trends over time were evaluated using descriptive statistics. RESULTS 34 eligible RCTs from 33 reports were included. The proportion of female participants increased over time with females representing 29.0-43.7% of participants in studies initiated in 2000-2004 which increased to 46.0-58.8% in 2015-2019. While the number of countries included in RCTs increased significantly from 1-8 countries (2000-2004) to 2-46 (2015-2019), the proportion of white participants changed marginally from 90.0-98.0% (2000-2004) to 80.9-97.3% (2015-2019). The SJC and TJC decreased from 13.9 to 24.6 respectively (2000-2004), to 7.0-13.9 and 12.9-24.9 (2015-2019). Baseline CRP and HAQ-DI remained stable. CONCLUSION Despite the expansion of countries from which PsA RCT participants were recruited from, non-white participants continue to be under-represented. Improving diversity in patient representation is imperative to further our understanding of PsA phenotypes, proteogenomics, socioeconomic determinants, and treatment effects, to advance the care of all patients with psoriatic disease.
Collapse
Affiliation(s)
- Wendy Zhu
- Department of Rheumatology, Monash Health, 246 Clayton Road, Clayton, VIC 3168, Australia.
| | - Sally Ayoub
- Department of Rheumatology, Monash Health, 246 Clayton Road, Clayton, VIC 3168, Australia; School of Clinical Sciences, Monash University, 246 Clayton Road, Clayton, VIC 3168, Australia
| | - Eric Morand
- Department of Rheumatology, Monash Health, 246 Clayton Road, Clayton, VIC 3168, Australia; School of Clinical Sciences, Monash University, 246 Clayton Road, Clayton, VIC 3168, Australia
| | - William Tillett
- Department of Rheumatology, Royal United Hospital, Bath, United Kingdom; Department of Pharmacology, University of Bath, Bath, United Kingdom
| | - Anna Antony
- Department of Rheumatology, Monash Health, 246 Clayton Road, Clayton, VIC 3168, Australia; School of Clinical Sciences, Monash University, 246 Clayton Road, Clayton, VIC 3168, Australia
| |
Collapse
|
35
|
Palominos PE, Fernández-Ávila DG, Coates LC, Adebajo A, Toukap AN, Abogamal A, Polachek A, van Kuijk AWR, Caso F, de Marco G, Kaeley GS, Steinkoenig I, Chau J, Feletar M, Vis M, Elkayam O, Sewerin P, d'Angelo S, Aydin SZ, AlShehhi W, Helliwell PS. Management of Dactylitis in Patients With Psoriatic Arthritis: An Updated Literature Review Informing the 2021 GRAPPA Treatment Recommendations. J Rheumatol 2023; 50:265-278. [PMID: 36319013 DOI: 10.3899/jrheum.220311] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/06/2022] [Indexed: 01/07/2023]
Abstract
OBJECTIVE This literature review aimed to identify the most efficacious current interventions for dactylitis and provide up-to-date scientific evidence to support the 2021 Group for Research and Assessment of Psoriasis and Psoriatic Arthritis (GRAPPA) recommendations on the management of psoriatic arthritis. METHODS Original articles published from 2013 to 2020, registered in MEDLINE, Embase, and Cochrane Library, describing interventional trials and reporting dactylitis-related outcomes were included. The 20 members of the GRAPPA dactylitis group were divided into 9 subgroups according to treatment, and members of each group independently extracted data from articles/abstracts corresponding to their group by using a standardized data extraction form. RESULTS Forty-nine publications were analyzed, representing 40 randomized clinical trials (RCTs) and including 16,752 patients. Dactylitis was assessed as a secondary outcome in 97.5% of these trials and more than 40% of RCTs did not employ a specific dactylitis measure or instrument. CONCLUSION The emergence of agents with novel mechanisms of action in recent years, such as interleukin 17 (IL-17), IL-12/23, IL-23, and Janus kinase inhibitors, has significantly expanded the available treatment options for dactylitis. This article points out the lack of consensus regarding dactylitis assessment and the paucity of data concerning the effect of local steroid injections, nonsteroidal antiinflammatory drugs, and conventional disease-modifying antirheumatic drugs. Clinical trials evaluating the effect of these traditional and low-cost medications used to treat dactylitis should be encouraged.
Collapse
Affiliation(s)
- Penélope Esther Palominos
- P.E. Palominos, MD, PhD, Rheumatology Unit, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil;
| | - Daniel G Fernández-Ávila
- D.G. Fernández-Ávila, MD, PhD, Rheumatology Unit, Pontificia Universidad Javeriana, Hospital Universitario San Ignacio, Bogotá, Colombia
| | - Laura C Coates
- L.C. Coates, MD, PhD, Department of Orthopaedics, Rheumatology, and Musculoskeletal Sciences, University of Oxford, Oxford, UK
| | - Adewale Adebajo
- A. Adebajo, MD, MBE, Faculty of Medicine, Dentistry and Health, University of Sheffield, Sheffield, UK
| | - Adrien Nzeusseu Toukap
- A. Nzeusseu Toukap, MD, Rheumatology Department, Saint-Luc University Hospitals, and Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
| | - Ahmed Abogamal
- A. Abogamal, MD, PhD, Al-Azhar Faculty of Medicine, Cairo, Egypt
| | - Ari Polachek
- A. Polachek, MD, Department of Rheumatology, Sourasky Medical Center, affiliated with Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Arno W R van Kuijk
- A.W.R. van Kuijk, MD, PhD, Department of Rheumatology, Amsterdam Rheumatology and Immunology Center, Reade, Amsterdam, the Netherlands
| | - Francesco Caso
- F. Caso, MD, PhD, Department of Clinical Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Gabriele de Marco
- G. de Marco, MD, Leeds Biomedical Research Center at Leeds Teaching Hospitals NHS Trust, and University of Leeds, Leeds, UK
| | - Gurjit S Kaeley
- G.S. Kaeley, MD, University of Florida College of Medicine, Jacksonville, Florida, USA
| | - Ingrid Steinkoenig
- I. Steinkoenig, BA, GRAPPA Patient Research Partner, Cleveland, Ohio, USA
| | - Jeffrey Chau
- J. Chau, MCS, GRAPPA Patient Research Partner, Hong Kong SAR, China
| | - Marie Feletar
- M. Feletar, MD, Dandenong Rheumatology, Melbourne, Australia
| | - Marijn Vis
- M. Vis, MD, PhD, Department of Rheumatology, Erasmus MC, Rotterdam, the Netherlands
| | - Ori Elkayam
- O. Elkayam, MD, Department of Rheumatology, Sourasky Medical Center, Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Philipp Sewerin
- P. Sewerin, MD, PhD, Heinrich-Heine-Universität Duesseldorf, University Hospital Duesseldorf, Department and Hiller Research-Unit for Rheumatology, Duesseldorf, Germany
| | - Salvatore d'Angelo
- S. d'Angelo, MD, PhD, Rheumatology Institute of Lucania, and Rheumatology Department of Lucania, San Carlo Hospital of Potenza and Madonna delle Grazie Hospital of Matera, Potenza, Italy
| | - Sibel Zehra Aydin
- S.Z. Aydin, MD, University of Ottawa, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
| | | | | |
Collapse
|
36
|
Eder L, Mathew AJ, Carron P, Bertheussen H, Cañete JD, Azem M, Delle Sedie A, Salvarani C, Ranza R, Elliott A, Turkiewicz A, de Toledo RA, Bukulmez H, Stoenoiu MS, Mandelin AM, Koehm M, Lindsay CA, Siegel E, Mease PJ. Management of Enthesitis in Patients With Psoriatic Arthritis: An Updated Literature Review Informing the 2021 GRAPPA Treatment Recommendations. J Rheumatol 2023; 50:258-264. [PMID: 36319005 DOI: 10.3899/jrheum.220312] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/05/2022] [Indexed: 11/06/2022]
Abstract
OBJECTIVE Enthesitis is a key pathological and clinical feature of psoriatic arthritis (PsA) in children and adults. Enthesitis is typically assessed clinically using several validated enthesitis scoring systems that have been used in clinical trials. Enthesitis treatment response has been reported as change in the total enthesitis score or the proportion of patients who achieved complete resolution. The majority of trials in PsA did not require patients to have enthesitis at study entry since enthesitis was evaluated only as a secondary outcome. Despite the inherent limitations of the clinical assessment of enthesitis, imaging of the entheses using ultrasound or magnetic resonance imaging has rarely been used in clinical trials to assess response to treatment of enthesitis. This systematic review summarizes existing evidence regarding pharmaceutical and nonpharmaceutical interventions for enthesitis in patients with PsA to facilitate an evidence-based update of the Group for Research and Assessment in Psoriasis and Psoriatic Arthritis (GRAPPA) treatment recommendations for PsA. METHODS We performed a systematic literature review to identify 41 randomized clinical trials that reported enthesitis treatment response in patients with PsA. For each intervention, the response effect size was summarized and the quality of evidence was graded. Recommendations were then formulated for the various pharmacological and nonpharmacological therapies. RESULTS We included 41 randomized clinical trials in our review and graded each intervention. CONCLUSION Several classes of systemic conventional and advanced therapies and local measures were recommended for active enthesitis in patients with PsA.
Collapse
Affiliation(s)
- Lihi Eder
- L. Eder, MD, PhD, Women's College Research Institute, University of Toronto, Toronto, Ontario, Canada;
| | - Ashish J Mathew
- A.J. Mathew, MBBS, DNB, DM, The Copenhagen Center for Arthritis Research (COPECARE), Center for Rheumatology and Spine Diseases, Rigshospitalet Glostrup, University of Copenhagen, Copenhagen, Denmark, and Department of Clinical Immunology and Rheumatology, Christian Medical College, Vellore, India
| | - Philippe Carron
- P. Carron, MD, PhD, Department of Internal Medicine and Pediatrics, Ghent University, Ghent University Hospital, and VIB Center for Inflammation Research, Ghent, Belgium
| | | | - Juan D Cañete
- J.D. Cañete, DMD, PhD, Department of Rheumatology, Hospital Clinic and IDIBAPS, Barcelona, Spain
| | - May Azem
- M. Azem, MD, Solo Private Practice, and Department of Internal Medicine, Lake Hospital, University Hospital Systems, Cleveland, Ohio, USA
| | - Andrea Delle Sedie
- A. Delle Sedie, MD, Rheumatology Unit, Azienda Ospedaliero-Universitaria Pisana, Pisa, Italy
| | - Carlo Salvarani
- C. Salvarani, MD, Dipartimento Specialità Mediche, Azienda Unità Sanitaria Locale-IRCCS di Reggio Emilia, Reggio Emilia and Dipartimento Chirurgico, Medico, Odontoiatrico e di Scienze Morfologiche con interesse Trapiantologico, Oncologico e di Medicina Rigenerativa, Università di Modena e Reggio Emilia, Modena, Italy
| | - Roberto Ranza
- R. Ranza, MD, PhD, Rheumatology Unit, Hospital de Clinicas, EBSERH, Universidade Federal de Uberlandia, Uberlandia, Minas Gerais, Brazil
| | - Ashley Elliott
- A. Elliott, MB Bch BAO, MSc, MRCP, Centre for Experimental Medicine, School of Medicine, Dentistry and Biomedical Sciences, Queen's University Belfast, Belfast, UK
| | | | - Ricardo Acayaba de Toledo
- R.A. de Toledo, MD, MSc, Regional School of Medicine Foundation of São José do Rio Preto, São José do Rio Preto, São Paulo, Brazil
| | - Hulya Bukulmez
- H. Bukulmez, MD, Department of Pediatrics, Division of Pediatric Rheumatology, MetroHealth Medical Center, Case Western Reserve University, Cleveland, Ohio, USA
| | - Maria S Stoenoiu
- M.S. Stoenoiu, MD, PhD, Department of Rheumatology, Cliniques Universitaires Saint-Luc, Institut de Recherche Expérimentale et Clinique, Université catholique de Louvain, Brussels, Belgium
| | - Arthur M Mandelin
- A.M. Mandelin II, MD, PhD, Northwestern University, Department of Medicine, Feinberg School of Medicine, Chicago, Illinois, USA
| | - Michaela Koehm
- M. Koehm, MD, Rheumatology Division, University Hospital Frankfurt and Fraunhofer-Institute for Translational Medicine and Pharmacology ITMP, Frankfurt, Germany
| | - Chris A Lindsay
- C.A. Lindsay, PharmD, GRAPPA Patient Research Partner, Aurinia Pharmaceuticals Inc., Rockville, Maryland, USA
| | - Evan Siegel
- E. Siegel, MD, Arthritis and Rheumatism Associates, Rockville, Maryland, and Georgetown University School of Medicine, Washington, DC, USA
| | - Philip J Mease
- P.J. Mease MD, MACR, Rheumatology Research, Swedish Medical Center/Providence St. Joseph Health and University of Washington School of Medicine, Seattle, Washington, USA
| |
Collapse
|
37
|
Duffin KC, Mazzuoccolo LD, Cura MJ, Esposito M, Fernandez AP, Gisondi P, Giunta A, Hillary T, Piaserico S, Solomon JA, Merola JF. Treatment of Psoriasis in Patients With Psoriatic Arthritis: An Updated Literature Review Informing the 2021 GRAPPA Treatment Recommendations. J Rheumatol Suppl 2023; 50:131-143. [PMID: 36319014 DOI: 10.3899/jrheum.220316] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/12/2022] [Indexed: 11/05/2022]
Abstract
OBJECTIVE Our aim was to summarize and evaluate the current quality of evidence regarding the efficacy of therapies for cutaneous psoriasis (PsO) in patients with psoriatic arthritis (PsA). METHODS A literature search of MEDLINE, Embase, Cochrane Library databases, and conference abstracts was conducted to identify interventional randomized controlled trials in patients with PsA between February 2013 and December 2021. Studies were included if PsO outcomes included achieving at least 75% improvement in the Psoriasis Area and Severity Index and the blinded comparison period was ≥ 10 weeks. The Grading of Recommendations, Assessment, Development, and Evaluation (GRADE) methodology was employed to assess quality of the evidence to inform and update the 2021 Group for Research and Assessment of Psoriasis and Psoriatic Arthritis (GRAPPA) treatment recommendations. RESULTS A total of 116 studies and 36 abstracts identified in the initial search were screened. A total of 37 studies (40 treatment arms) met the criteria for final inclusion. Phosphodiesterase 4 inhibitors, Janus kinase inhibitors, and tyrosine kinase 2 inhibitors, interleukin 17 inhibitors (IL-17i), IL-12/23i, IL-23i, and tumor necrosis factor inhibitors (TNFi) had high-quality data broadly supporting the efficacy of each class for plaque PsO over placebo. Head-to-head studies with high-quality data supported both IL-17i and IL-23i over TNFi. CONCLUSION Several pharmacologic therapeutic classes have high-quality evidence demonstrating efficacy for cutaneous PsO in the PsA population. The findings will be integrated into the 2021 GRAPPA treatment recommendations, intended to guide selection of a therapeutic class where efficacy in 1 or more cutaneous or musculoskeletal domains is required.
Collapse
Affiliation(s)
- Kristina Callis Duffin
- K. Callis Duffin, MD, Department of Dermatology, Spencer Fox Eccles School of Medicine, University of Utah, Salt Lake City, Utah, USA;
| | - Luis Daniel Mazzuoccolo
- L.D. Mazzuoccolo, MD, MPH, M.J. Cura, MD, Department of Dermatology, Hospital Italiano de Buenos Aires, Buenos Aires, Argentina
| | - María Julia Cura
- L.D. Mazzuoccolo, MD, MPH, M.J. Cura, MD, Department of Dermatology, Hospital Italiano de Buenos Aires, Buenos Aires, Argentina
| | - Maria Esposito
- M. Esposito, MD, Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| | - Anthony P Fernandez
- A.P. Fernandez, MD, PhD, Departments of Dermatology and Pathology, Cleveland Clinic, Cleveland, Ohio, USA
| | - Paolo Gisondi
- P. Gisondi, MD, Section of Dermatology and Venereology, University of Verona, Verona, Italy
| | - Alessandro Giunta
- A. Giunta, MD, Department of Dermatology, University of Rome Tor Vergata, Rome, Italy
| | - Tom Hillary
- T. Hillary, MD, Department of Dermatology, University Hospitals Leuven, Leuven, Belgium
| | - Stefano Piaserico
- S. Piaserico, MD, PhD, Unit of Dermatology, Department of Medicine, University of Padua, Padova, Italy
| | - James A Solomon
- J.A. Solomon, MD, PhD, University Central Florida College of Medicine, and Florida State University College of Medicine, Orlando, Florida, and Carle-Illinois College of Medicine, Urbana, Illinois, USA
| | - Joseph F Merola
- J.F. Merola, MD, MMSc, Division of Rheumatology and Immunology, Department of Dermatology and Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
38
|
Chow TG, Oppenheimer J, Joshi SR. A Review of Adverse Reactions to Biologics Used in Allergy-Immunology Practice. THE JOURNAL OF ALLERGY AND CLINICAL IMMUNOLOGY. IN PRACTICE 2022; 10:3075-3086. [PMID: 36162800 DOI: 10.1016/j.jaip.2022.09.020] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Revised: 09/12/2022] [Accepted: 09/13/2022] [Indexed: 02/06/2023]
Abstract
Biologic agents have become an integral therapeutic option for practicing allergists-immunologists for the management of asthma, atopic dermatitis, chronic rhinosinusitis with nasal polyps, and various immunologic conditions. As these agents vary considerably from traditional small-molecule drugs, various adverse reactions have been noted. A different approach must be used to classify these reactions beyond the classic Gell-Coombs classification system as it does not capture many of the adverse events seen with biologic therapy. This article addresses the available literature on proposed classification systems and diagnostic modalities for adverse events associated with biologics and reviews each approved agent used frequently in allergy-immunology practice.
Collapse
Affiliation(s)
- Timothy G Chow
- Division of Allergy and Immunology, Department of Internal Medicine, University of Texas Southwestern, Dallas, Texas
| | - John Oppenheimer
- Department of Internal Medicine, UMDMJ Rutgers University School of Medicine, Newark, NJ; Pulmonary and Allergy Associates, Morristown, NJ
| | - Shyam R Joshi
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, Oregon Health and Science University, Portland, Ore.
| |
Collapse
|
39
|
Lin CP, Merola JF, Wallace EB. Current and emerging biologic and small molecule systemic treatment options for psoriasis and psoriatic arthritis. Curr Opin Pharmacol 2022; 67:102292. [PMID: 36228472 DOI: 10.1016/j.coph.2022.102292] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Revised: 08/31/2022] [Accepted: 09/01/2022] [Indexed: 01/25/2023]
Abstract
Psoriasis and psoriatic arthritis are chronic inflammatory diseases affecting the skin and joints, respectively. Psoriasis and psoriatic arthritis are associated with a high comorbidity burden as well as negative impact on quality of life. Impact on health-related quality of life is optimized when both skin and joint manifestations are effectively treated. The identification of key cytokines involved in disease pathogenesis has led to the development of several therapeutic options for psoriatic disease. When selecting a therapy, it is important to consider disease severity, psoriasis disease subtypes or domains of psoriatic arthritis, comorbidities, patient preference for treatment, among other factors. This review summarizes current biologic and small molecule treatment options as well as emerging therapies for moderate-to-severe adult plaque psoriasis and psoriatic arthritis.
Collapse
Affiliation(s)
- Christine P Lin
- Department of Internal Medicine, University of Colorado School of Medicine, Aurora, CO, USA
| | - Joseph F Merola
- Department of Dermatology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA; Department of Medicine, Division of Rheumatology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Elizabeth B Wallace
- Department of Dermatology, University of Colorado School of Medicine, Aurora, CO, USA.
| |
Collapse
|
40
|
Therapeutic Utility and Adverse Effects of Biologic Disease-Modifying Anti-Rheumatic Drugs in Inflammatory Arthritis. Int J Mol Sci 2022; 23:ijms232213913. [PMID: 36430392 PMCID: PMC9692587 DOI: 10.3390/ijms232213913] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 11/03/2022] [Accepted: 11/09/2022] [Indexed: 11/16/2022] Open
Abstract
Targeting specific pathologic pro-inflammatory cytokines or related molecules leads to excellent therapeutic effects in inflammatory arthritis, including rheumatoid arthritis, ankylosing spondylitis, and psoriatic arthritis. Most of these agents, known as biologic disease-modifying anti-rheumatic drugs (bDMARDs), are produced in live cell lines and are usually monoclonal antibodies. Several types of monoclonal antibodies target different pro-inflammatory cytokines, such as tumor necrosis factor-α, interleukin (IL)-17A, IL-6, and IL-23/12. Some bDMARDs, such as rituximab and abatacept, target specific cell-surface molecules to control the inflammatory response. The therapeutic effects of these bDMARDs differ in different forms of inflammatory arthritis and are associated with different adverse events. In this article, we summarize the therapeutic utility and adverse effects of bDMARDs and suggest future research directions for developing bDMARDs.
Collapse
|
41
|
Østergaard M, Bird P, Pachai C, Du S, Wu C, Landis J, Fuerst T, Ahmad HA, Connolly SE, Conaghan PG. Implementation of the OMERACT Psoriatic Arthritis Magnetic Resonance Imaging Scoring System in a randomized phase IIb study of abatacept in psoriatic arthritis. Rheumatology (Oxford) 2022; 61:4305-4313. [PMID: 35137002 PMCID: PMC9629349 DOI: 10.1093/rheumatology/keac073] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 01/22/2022] [Indexed: 08/27/2023] Open
Abstract
OBJECTIVES To investigate if the OMERACT PsA MRI Scoring System (PsAMRIS), including a novel total inflammation score, shows sensitivity to change with an agent (abatacept) known to impact clinical outcomes in PsA. METHODS We performed a post hoc analysis of a randomized phase IIb study of abatacept in patients with PsA and inadequate DMARD response. Participants received one of three abatacept dosing regimens [ABA3, ABA10 or ABA30/10 mg/kg (30 mg/kg switched to 10 mg/kg after two doses)] or placebo until day 169, then ABA10 through day 365. MRIs at baseline and days 85, 169 and 365 were centrally evaluated by two readers blinded to chronological order and treatment arm. Synovitis, osteitis, tenosynovitis, periarticular inflammation, bone erosions, joint space narrowing and bone proliferation were assessed using the PsAMRIS. A novel total inflammation score was tested. RESULTS MRIs for 123 patients were included. On day 169, ABA10 and ABA30/10 significantly reduced MRI synovitis and tenosynovitis, respectively, vs placebo [differences -0.966 (P = 0.039) and -1.652 (P = 0.014), respectively]. Synovitis in the placebo group increased non-significantly from baseline to day 169, total inflammation and tenosynovitis decreased non-significantly and all measures improved significantly after a switch to ABA10 [-1.019, -0.940, -2.275 (P < 0.05), respectively, day 365 vs day 169]. Structural outcomes changed minimally across groups. CONCLUSION Adults with PsA receiving ABA10 and ABA30/10 demonstrated significant resolution of inflammatory components of disease, confirmed by MRI, with synovitis and tenosynovitis improvements consistent with previously reported clinical responses for these doses. Results indicate that a reduction in OMERACT PsAMRIS inflammation scores may provide proof of tissue-level efficacy in PsA clinical trials. REGISTRATION ClinicalTrials.gov (https://clinicaltrials.gov), NCT00534313.
Collapse
Affiliation(s)
- Mikkel Østergaard
- Copenhagen Center for Arthritis Research, Center for Rheumatology and Spine Diseases, Copenhagen University Hospital Rigshospitalet, Glostrup
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Paul Bird
- Division of Medicine, University of New South Wales, Sydney, NSW, Australia
| | | | | | - Chun Wu
- Translational Bioinformatics, Bristol Myers Squibb, Princeton, NJ
| | - Jessica Landis
- Translational Bioinformatics, Bristol Myers Squibb, Princeton, NJ
| | - Thomas Fuerst
- Medical and Scientific Affairs, Bioclinica Inc., Newark, CA
| | | | - Sean E Connolly
- Global Drug Development, Bristol Myers Squibb, Princeton, NJ, USA
| | - Philip G Conaghan
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds
- NIHR Leeds Biomedical Research Centre, Leeds, UK
| |
Collapse
|
42
|
Wang SH, Yu CL, Wang TY, Yang CH, Chi CC. Biologic Disease-Modifying Antirheumatic Drugs for Preventing Radiographic Progression in Psoriatic Arthritis: A Systematic Review and Network Meta-Analysis. Pharmaceutics 2022; 14:2140. [PMID: 36297574 PMCID: PMC9608970 DOI: 10.3390/pharmaceutics14102140] [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: 08/30/2022] [Revised: 09/30/2022] [Accepted: 10/05/2022] [Indexed: 11/06/2022] Open
Abstract
The prevention of joint deformity is among the most important treatment goals of psoriatic arthritis. Some biologics disease-modifying antirheumatic drugs (bDMARDs) have been demonstrated to be effective for both the skin and joints, as well as for slowing radiographic progression. However, there has been a lack of direct comparisons of bDMARDs. To evaluate the comparative effects of bDMARDs in preventing radiographic progression in psoriatic arthritis, we conducted a systematic review and network meta-analysis. On March 7 2022, a search for relevant randomized trials was conducted on MEDLINE, Embase, and the Cochrane Central Register of Controlled Trials. Our outcomes included radiographic non-progression, a mean change in the total radiographic score, and adverse events leading to discontinuation (DAE) at week 24. We included 11 trials on 10 bDMARDs, involving 4010 participants. Most bDMARDs were more effective than placebos in achieving radiographic non-progression, including adalimumab (odds ratio (OR) 4.7, 95% confidence interval (CI) 2.66-8.29), etanercept (OR 4.19, 95% CI 1.65-10.61), certolizumab pegol (OR 2.83, 95% CI 1.55-5.2), secukinumab 300 mg (OR 2.63, CI 1.62-4.27), infliximab (OR 2.54, CI 1.13-5.69), ixekizumab (OR 2.22, 95% CI 1.06-4.65), golimumab (OR 2.21, 95% CI 1.24-3.93), and abatacept (OR 1.54, 95% CI 1.03-2.28). A significant reduction in the total radiographic score was found in infliximab (standardized mean difference (SMD) -0.59, 95% CI -0.87, -0.3), etanercept (SMD -0.51, 95% CI -0.78, -0.23), adalimumab (SMD -0.45, 95% CI -0.64, -0.26), ixekizumab (SMD -0.37, 95% CI -0.62, -0.12), secukinumab 300 mg (SMD -0.33, 95% CI -0.50, -0.15), golimumab (SMD -0.33, 95% CI -0.58, -0.09), secukinumab 150 mg (SMD -0.25, 95% CI -0.43, -0.07), certolizumab pegol (SMD -0.23, 95% CI -0.44, -0.03), and ustekinumab (SMD -0.19, 95% CI -0.35, -0.33). No significant differences in DAE were detected between bDMARDs. In conclusion, anti-tumor necrosis factor agents (adalimumab, infliximab, and etanercept) may be preferred for treating psoriatic arthritis for their superiority in preventing radiographic progression.
Collapse
Affiliation(s)
- Szu-Hsuan Wang
- Department of Pharmacy, Chang Gung Memorial Hospital, Linkou, Taoyuan 33305, Taiwan
| | - Chia-Ling Yu
- Department of Pharmacy, Chang Gung Memorial Hospital, Linkou, Taoyuan 33305, Taiwan
| | - Tzu-Yu Wang
- Department of Applied Cosmetology, Lee-Ming Institute of Technology, New Taipei 24346, Taiwan
| | - Chung-Han Yang
- Division of Rheumatology, Allergy and Immunology, Chang Gung Memorial Hospital, Linkou, Taoyuan 33305, Taiwan
| | - Ching-Chi Chi
- Department of Dermatology, Chang Gung Memorial Hospital, Linkou, Taoyuan 33305, Taiwan
- School of Medicine, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan
| |
Collapse
|
43
|
Vassilopoulos A, Shehadeh F, Benitez G, Kalligeros M, Cunha JS, Cunha CB, Mylonakis E. The incidence of opportunistic infections in patients with psoriatic arthritis treated with biologic and targeted synthetic agents: A systematic review and meta-analysis. Front Pharmacol 2022; 13:992713. [PMID: 36278224 PMCID: PMC9579334 DOI: 10.3389/fphar.2022.992713] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Accepted: 09/20/2022] [Indexed: 11/13/2022] Open
Abstract
Background: Biologic (bDMARD) and targeted synthetic (tsDMARD) disease-modifying anti-rheumatic drugs have broadened the treatment options and are increasingly used for patients with psoriatic arthritis (PsA). These agents block different pro-inflammatory cytokines or specific intracellular signaling pathways that promote inflammation and can place patients at risk of serious infections. We aimed to review the incidence of opportunistic infections (OIs) in patients with PsA who were treated with these agents.Methods: We searched PubMed and EMBASE through 14 April 2022 for randomized clinical trials evaluating bDMARD or tsDMARD in the treatment of PsA. Trials were eligible if they compared the effect of a bDMARD or tsDMARD with placebo and provided safety data. We used the Revised Cochrane risk-of-bias tool to assess the risk of bias among trials, and stratified the studies by mechanism of action (MOA) of the agents studied.Results: We included 47 studies in this analysis. A total of 17,197 patients received at least one dose of an agent of interest. The cumulative incidence of OIs by MOA was as follows: 1) JAK inhibitors: 2.72% (95% CI: 1.05%–5.04%), 2) anti-IL-17: 1.18% (95% CI: 0.60%–1.9%), 3) anti-IL-23: 0.24% (95% CI: 0.04%–0.54%), and 4) anti-TNFs: 0.01% (95% CI: 0.00%–0.21%). Based on their MOA, these agents are known to increase the risk of certain serious infections. The cumulative incidence of herpes zoster infection following treatment with JAK inhibitors (JAKi) was 2.53% (95% CI: 1.03%–4.57%) and the cumulative incidence of opportunistic Candida spp. infections following treatment with anti-IL-17, was 0.97% (95% CI: 0.51%–1.56%).Conclusion: The overall incidence of OIs among patients with PsA who were treated with biologic and targeted synthetic agents is low. However, careful monitoring is warranted for specific OIs such as herpes zoster infection following JAKi treatment, mucocutaneous candidiasis following anti-IL-17 treatment, and Mycobacterium tuberculosis infection following anti-TNF treatment.
Collapse
Affiliation(s)
- Athanasios Vassilopoulos
- Infectious Diseases Division, Rhode Island Hospital, Providence, RI, United States
- Warren Alpert Medical School of Brown University, Providence, RI, United States
| | - Fadi Shehadeh
- Infectious Diseases Division, Rhode Island Hospital, Providence, RI, United States
- Warren Alpert Medical School of Brown University, Providence, RI, United States
- School of Electrical and Computer Engineering, National Technical University of Athens, Athens, Greece
| | - Gregorio Benitez
- Infectious Diseases Division, Rhode Island Hospital, Providence, RI, United States
- Warren Alpert Medical School of Brown University, Providence, RI, United States
| | - Markos Kalligeros
- Infectious Diseases Division, Rhode Island Hospital, Providence, RI, United States
- Warren Alpert Medical School of Brown University, Providence, RI, United States
| | - Joanne S. Cunha
- Warren Alpert Medical School of Brown University, Providence, RI, United States
| | - Cheston B. Cunha
- Infectious Diseases Division, Rhode Island Hospital, Providence, RI, United States
- Warren Alpert Medical School of Brown University, Providence, RI, United States
| | - Eleftherios Mylonakis
- Infectious Diseases Division, Rhode Island Hospital, Providence, RI, United States
- Warren Alpert Medical School of Brown University, Providence, RI, United States
- *Correspondence: Eleftherios Mylonakis,
| |
Collapse
|
44
|
Alnaqbi KA, Hannawi S, Namas R, Alshehhi W, Badsha H, Al‐Saleh J. Consensus statements for pharmacological management, monitoring of therapies, and comorbidity management of psoriatic arthritis in the United Arab Emirates. Int J Rheum Dis 2022; 25:1107-1122. [PMID: 35916205 PMCID: PMC9804226 DOI: 10.1111/1756-185x.14406] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Revised: 06/28/2022] [Accepted: 07/17/2022] [Indexed: 01/05/2023]
Abstract
OBJECTIVE Psoriatic arthritis (PsA), a chronic inflammatory disease characterized by heterogeneous clinical manifestations, substantially impacts the quality of life of affected individuals. This article aims at developing consensus recommendations for the management of PsA and associated comorbidities and screening and monitoring requirements of PsA therapies in the United Arab Emirates (UAE) population. METHODS An extensive review of present international and regional guidelines and publications on the pharmacological management, monitoring of therapies in the context of PsA was performed. Key findings from guidelines and literature were reviewed by a panel of experts from the UAE at several meetings to align with current clinical practices. Consensus statements were formulated based on collective agreement of the experts and members of Emirates Society for Rheumatology. RESULTS The consensus recommendations were developed to aid practitioners in clinical decision-making with respect to dosage recommendations for pharmacological therapies for PsA, including conventional drugs, non-biologic, and biologic therapies. Consensus recommendations for therapeutic options for the treatment of PsA domains, including peripheral arthritis, axial disease, enthesitis, dactylitis, psoriasis, and nail disease, were developed. The panel emphasized the importance of monitoring PsA therapies and arrived at a consensus on monitoring requirements for PsA therapies. The expert panel proposed recommendations for the management of common comorbidities associated with PsA. CONCLUSION These consensus recommendations can guide physicians and healthcare professionals in the UAE in making proper treatment decisions, as well as efficiently managing comorbidities and monitoring therapies in patients with PsA.
Collapse
Affiliation(s)
- Khalid A. Alnaqbi
- Department of RheumatologyTawam HospitalAl AinUAE
- College of Medicine and Health SciencesUAE UniversityAl AinUAE
| | - Suad Hannawi
- Emirates Health Services (EHS)DubaiUAE
- Ministry of Health and PreventionDubaiUAE
| | - Rajaie Namas
- Division of Rheumatology, Department of Internal MedicineCleveland Clinic Abu DhabiUAE
| | | | | | | |
Collapse
|
45
|
Kim GR, Choi JM. Current Understanding of Cytotoxic T Lymphocyte Antigen-4 (CTLA-4) Signaling in T-Cell Biology and Disease Therapy. Mol Cells 2022; 45:513-521. [PMID: 35950451 PMCID: PMC9385567 DOI: 10.14348/molcells.2022.2056] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 04/26/2022] [Accepted: 05/02/2022] [Indexed: 12/21/2022] Open
Abstract
Cytotoxic T lymphocyte antigen-4 (CTLA-4) is an immune checkpoint molecule that is mainly expressed on activated T cells and regulatory T (Treg) cells that inhibits T-cell activation and regulates immune homeostasis. Due to the crucial functions of CTLA-4 in T-cell biology, CTLA-4-targeted immunotherapies have been developed for autoimmune disease as well as cancers. CTLA-4 is known to compete with CD28 to interact with B7, but some studies have revealed that its downstream signaling is independent of its ligand interaction. As a signaling domain of CTLA-4, the tyrosine motif plays a role in inhibiting T-cell activation. Recently, the lysine motif has been shown to be required for the function of Treg cells, emphasizing the importance of CTLA-4 signaling. In this review, we summarize the current understanding of CTLA-4 biology and molecular signaling events and discuss strategies to target CTLA-4 signaling for immune modulation and disease therapy.
Collapse
Affiliation(s)
- Gil-Ran Kim
- Department of Life Science, College of Natural Sciences, Hanyang University, Seoul 04763, Korea
- Research Institute for Natural Sciences, Hanyang University, Seoul 04763, Korea
| | - Je-Min Choi
- Department of Life Science, College of Natural Sciences, Hanyang University, Seoul 04763, Korea
- Research Institute for Natural Sciences, Hanyang University, Seoul 04763, Korea
- Research Institute for Convergence of Basic Sciences, Hanyang University, Seoul 04763, Korea
- Institute for Rheumatology Research, Hanyang University, Seoul 04763, Korea
- Hanyang Institute of Bioscience and Biotechnology, Hanyang University, Seoul 04763, Korea
| |
Collapse
|
46
|
Coates LC, Soriano ER, Corp N, Bertheussen H, Callis Duffin K, Campanholo CB, Chau J, Eder L, Fernández-Ávila DG, FitzGerald O, Garg A, Gladman DD, Goel N, Helliwell PS, Husni ME, Jadon DR, Katz A, Laheru D, Latella J, Leung YY, Lindsay C, Lubrano E, Mazzuoccolo LD, Mease PJ, O'Sullivan D, Ogdie A, Olsder W, Palominos PE, Schick L, Steinkoenig I, de Wit M, van der Windt DA, Kavanaugh A. Group for Research and Assessment of Psoriasis and Psoriatic Arthritis (GRAPPA): updated treatment recommendations for psoriatic arthritis 2021. Nat Rev Rheumatol 2022; 18:465-479. [PMID: 35761070 PMCID: PMC9244095 DOI: 10.1038/s41584-022-00798-0] [Citation(s) in RCA: 291] [Impact Index Per Article: 97.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/23/2022] [Indexed: 02/07/2023]
Abstract
Since the second version of the Group for Research and Assessment of Psoriasis and Psoriatic Arthritis (GRAPPA) treatment recommendations were published in 2015, therapeutic options for psoriatic arthritis (PsA) have advanced considerably. This work reviews the literature since the previous recommendations (data published 2013-2020, including conference presentations between 2017 and 2020) and reports high-quality, evidence-based, domain-focused recommendations for medication selection in PsA developed by GRAPPA clinicians and patient research partners. The overarching principles for the management of adults with PsA were updated by consensus. Principles considering biosimilars and tapering of therapy were added, and the research agenda was revised. Literature searches covered treatments for the key domains of PsA: peripheral arthritis, axial disease, enthesitis, dactylitis, and skin and nail psoriasis; additional searches were performed for PsA-related conditions (uveitis and inflammatory bowel disease) and comorbidities. Individual subcommittees used a GRADE-informed approach, taking into account the quality of evidence for therapies, to generate recommendations for each of these domains, which were incorporated into an overall schema. Choice of therapy for an individual should ideally address all disease domains active in that patient, supporting shared decision-making. As safety issues often affect potential therapeutic choices, additional consideration was given to relevant comorbidities. These GRAPPA treatment recommendations provide up-to-date, evidence-based guidance on PsA management for clinicians and people with PsA.
Collapse
Affiliation(s)
- Laura C Coates
- Department of Orthopaedics, Rheumatology, and Musculoskeletal Sciences, University of Oxford, Oxford, UK.
| | - Enrique R Soriano
- University Institute and Rheumatology Unit, Hospital Italiano de Buenos Aires, Buenos Aires, Argentina
| | - Nadia Corp
- Primary Care Centre Versus Arthritis, School of Medicine, Keele University, Staffordshire, UK
| | | | | | | | | | - Lihi Eder
- Women's College Research Institute, University of Toronto, Toronto, ON, Canada
| | | | - Oliver FitzGerald
- School of Medicine, Conway Institute for Biomolecular Research, University College Dublin, Dublin, Ireland
| | - Amit Garg
- Donald and Barbara Zucker School of Medicine at Hofstra Northwell, Hempstead, NY, USA
| | | | - Niti Goel
- Duke University School of Medicine, Durham, NC, USA
| | | | | | | | | | | | | | - Ying-Ying Leung
- Singapore General Hospital, Duke-NUS Medical School, Singapore, Singapore
| | | | - Ennio Lubrano
- Academic Rheumatology Unit, University of Molise, Campobasso, Italy
| | | | - Philip J Mease
- Swedish Medical Center/Providence St. Joseph Health and University of Washington, Seattle, WA, USA
| | | | - Alexis Ogdie
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | | | | | | | | | - Maarten de Wit
- Department of Medical Humanities, VU University Medical Centre, Amsterdam, Netherlands
| | - D A van der Windt
- Primary Care Centre Versus Arthritis, School of Medicine, Keele University, Staffordshire, UK
| | - Arthur Kavanaugh
- Division of Rheumatology Allergy and Immunology, University of California, San Diego, La Jolla, CA, USA
| |
Collapse
|
47
|
El Miedany Y, El Gaafary M, GadAllah N, Mansour M, Fathy N, Hassan W, Mortada M, Galal S, Eissa M, Tabra SA, Foad N, Ali R, Medhat B, El Olemy G, Adel Y, Ghaleb R, El-Latif EA, Saber S, Elkaraly N, Abu-Zaid MH. Psoriatic arthritis treatment to the target: a consensus, evidence-based clinical practice recommendations for the management of psoriatic arthritis and its concomitant clinical manifestations. EGYPTIAN RHEUMATOLOGY AND REHABILITATION 2022. [DOI: 10.1186/s43166-022-00128-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Abstract
Background
We aimed to provide up-to-date, evidence-based and consensus-based recommendations for Treat-to-Target management of psoriatic arthritis (PsA) and associated clinical manifestations.
In this recommendations, 14 key clinical questions were identified by scientific committee according to the Patient/Population, Intervention, Comparison, Outcomes and Timing (PICOT) approach. Literature Review team performed a systematic review to summarize evidence advocating the benefits and harms of available pharmacologic and non-pharmacologic therapies for psoriatic arthritis. Subsequently, recommendations were formulated. The level of evidence was determined for each section using the Oxford Centre for Evidence-based Medicine (CEBM) system. A 3-round Delphi process was conducted with 19 experts whom were drawn from different governorates and health centers across Egypt with diverse in their experiences, including private, governmental workplace, tertiary university hospitals, and insurance hospitals. All rounds were conducted online. A consensus was achieved on the direction and the strength of the recommendations.
Results
An online questionnaire was sent to an expert panel who participated in the three rounds (response rate 100%). At the end of round 3, a total of 51 recommendation items, categorized into 6 sections to address the main 6 psoriatic arthritis categories, were obtained. Agreement with the recommendations (rank 7–9) ranged from 89.5 to 100%. Consensus was reached (i.e., ≥ 75%of respondents strongly agreed or agreed) on the wording of all the 51 clinical standards identified by the scientific committee. Algorithms for the management of psoriatic arthritis have been suggested.
Conclusion
These recommendations provide an updated consensus on the pharmacological treatment of psoriatic arthritis and strategies to reach optimal treat-to-target outcomes in in common clinical scenarios, based on a combination of evidence and expert opinion. Best treatment decisions should be tailored to each individual patient situation.
Collapse
|
48
|
Gootjes C, Zwaginga JJ, Roep BO, Nikolic T. Functional Impact of Risk Gene Variants on the Autoimmune Responses in Type 1 Diabetes. Front Immunol 2022; 13:886736. [PMID: 35603161 PMCID: PMC9114814 DOI: 10.3389/fimmu.2022.886736] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Accepted: 04/08/2022] [Indexed: 11/17/2022] Open
Abstract
Type 1 diabetes (T1D) is an autoimmune disease that develops in the interplay between genetic and environmental factors. A majority of individuals who develop T1D have a HLA make up, that accounts for 50% of the genetic risk of disease. Besides these HLA haplotypes and the insulin region that importantly contribute to the heritable component, genome-wide association studies have identified many polymorphisms in over 60 non-HLA gene regions that also contribute to T1D susceptibility. Combining the risk genes in a score (T1D-GRS), significantly improved the prediction of disease progression in autoantibody positive individuals. Many of these minor-risk SNPs are associated with immune genes but how they influence the gene and protein expression and whether they cause functional changes on a cellular level remains a subject of investigation. A positive correlation between the genetic risk and the intensity of the peripheral autoimmune response was demonstrated both for HLA and non-HLA genetic risk variants. We also observed epigenetic and genetic modulation of several of these T1D susceptibility genes in dendritic cells (DCs) treated with vitamin D3 and dexamethasone to acquire tolerogenic properties as compared to immune activating DCs (mDC) illustrating the interaction between genes and environment that collectively determines risk for T1D. A notion that targeting such genes for therapeutic modulation could be compatible with correction of the impaired immune response, inspired us to review the current knowledge on the immune-related minor risk genes, their expression and function in immune cells, and how they may contribute to activation of autoreactive T cells, Treg function or β-cell apoptosis, thus contributing to development of the autoimmune disease.
Collapse
Affiliation(s)
- Chelsea Gootjes
- Laboratory of Immunomodulation and Regenerative Cell Therapy, Department of Internal Medicine, Leiden University Medical Center, Leiden, Netherlands
| | - Jaap Jan Zwaginga
- Laboratory of Immunomodulation and Regenerative Cell Therapy, Department of Internal Medicine, Leiden University Medical Center, Leiden, Netherlands
| | - Bart O Roep
- Laboratory of Immunomodulation and Regenerative Cell Therapy, Department of Internal Medicine, Leiden University Medical Center, Leiden, Netherlands
| | - Tatjana Nikolic
- Laboratory of Immunomodulation and Regenerative Cell Therapy, Department of Internal Medicine, Leiden University Medical Center, Leiden, Netherlands
| |
Collapse
|
49
|
Miyagawa I, Tanaka Y. Dawn of Precision Medicine in Psoriatic Arthritis. Front Med (Lausanne) 2022; 9:851892. [PMID: 35372404 PMCID: PMC8973395 DOI: 10.3389/fmed.2022.851892] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Accepted: 02/22/2022] [Indexed: 01/01/2023] Open
Abstract
The establishment of precision medicine is considered particularly important in heterogeneous autoimmune diseases (e.g., psoriatic arthritis, systemic lupus erythematosus), which reveal clinical and molecular heterogeneity. The selection of optimal treatment strategies for individual patients may be more important and complex in autoimmune diseases than in other diseases. Two factors are important in precision medicine: patient stratification and use of targeted. When both factors work, patients are likely to have good outcomes. However, research into precision medicine and its practice in systemic autoimmune diseases is lacking. In contrast, the usefulness of peripheral immune cell phenotyping in the evaluation of immunological characteristics and stratification into subgroups of individual patients with systemic autoimmune diseases such as immunoglobulin 4-related disease, systemic lupus erythematosus, and anti-neutrophil cytoplasmic antibody-related vasculitis was reported. Furthermore, the potential of precision medicine using biological disease-modifying antirheumatic drugs based on peripheral immune cell phenotyping was recently demonstrated for psoriatic arthritis in the clinical setting. Precision medicine has not yet been sufficiently investigated in real world clinical settings. However, a dawn of precision medicine has emerged. We should shed further light on precision medicine in PsA and other autoimmune diseases. Here, we first review the usefulness of peripheral immune cell phenotyping in systemic autoimmune diseases and the potential of precision medicine in PsA based on this method.
Collapse
|
50
|
McInnes IB, Sawyer LM, Markus K, LeReun C, Sabry-Grant C, Helliwell PS. Targeted systemic therapies for psoriatic arthritis: a systematic review and comparative synthesis of short-term articular, dermatological, enthesitis and dactylitis outcomes. RMD Open 2022; 8:e002074. [PMID: 35321874 PMCID: PMC8943739 DOI: 10.1136/rmdopen-2021-002074] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Accepted: 03/02/2022] [Indexed: 01/11/2023] Open
Abstract
INTRODUCTION Randomised controlled trials (RCTs) have compared biological and targeted systemic disease-modifying antirheumatic drugs (DMARDS) against placebo in psoriatic arthritis (PsA); few have compared them head to head. OBJECTIVES To compare the efficacy and safety of all evaluated DMARDs for active PsA, with a special focus on biological DMARDs (bDMARDs) licensed for PsA or psoriasis. METHODS A systematic review identified RCTs and Bayesian network meta-analysis (NMA) compared treatments on efficacy (American College of Rheumatology (ACR) response, Psoriasis Area and Severity Index (PASI) response, resolution of enthesitis and dactylitis) and safety (patients discontinuing due to adverse events (DAE)) outcomes. Subgroup analyses explored ACR response among patients with and without prior biological therapy exposure. RESULTS The NMA included 46 studies. Results indicate that some tumour necrosis factor inhibitors (anti-TNFs) may perform numerically, but not significantly, better than interleukin (IL) inhibitors on ACR response but perform worse on PASI response. Few significant differences between bDMARDs on ACR response were observed after subgrouping for prior bDMARD exposure. Guselkumab and IL-17A or IL-17RA inhibitors-brodalumab, ixekizumab, secukinumab-were best on PASI response. These IL-inhibitors and adalimumab were similarly efficacious on resolution of enthesitis and dactylitis. Infliximab with and without methotrexate, certolizumab 400 mg every 4 weeks and tildrakizumab showed the highest rates of DAE; abatacept, golimumab and the IL-inhibitors, the lowest. CONCLUSIONS Despite similar efficacy for ACR response, IL-17A and IL-17RA inhibitors and guselkumab offered preferential efficacy to anti-TNFs in skin manifestations, and for enthesitis and dactylitis, thereby supporting drug selection based on predominant clinical phenotype.
Collapse
Affiliation(s)
- Iain B McInnes
- College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | | | | | - Corinne LeReun
- Independent Senior Biostatistician, Sainte-Anne, Guadeloupe
| | | | - Philip S Helliwell
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Leeds, UK
| |
Collapse
|