101
|
Chiu YM, Chen DY. Infection risk in patients undergoing treatment for inflammatory arthritis: non-biologics versus biologics. Expert Rev Clin Immunol 2020; 16:207-228. [PMID: 31852268 DOI: 10.1080/1744666x.2019.1705785] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Introduction: Despite the therapeutic effectiveness of biologics targeting immune cells or cytokines in patients with inflammatory arthritis, which reflects their pathogenic roles, an increased infection risk is observed in those undergoing biological treatment. However, there are limited data regarding the comparison of infection risks in inflammatory arthritis patients treated with non-biologics (csDMARDs), biologics (bDMARDs), including tumor necrosis factor (TNF) inhibitors and non-TNF inhibitors, or targeted synthetic (ts)DMARDs.Areas covered: Through a review of English-language literature as of 30 June 2019, we focus on the existing evidence on the risk of infections caused by bacteria, Mycobacterium tuberculosis, and hepatitis virus in inflammatory arthritis patients undergoing treatment with csDMARDs, bDMARDs, or tsDMARDs.Expert opinion: While the risks of bacterial and mycobacterial infection are increased in arthritis patients treated with csDMARDs, the risks are further higher in those receiving bDMARDs therapy, particularly TNF inhibitors. Regarding HBV infection, antiviral therapy may effectively prevent HBV reactivation in patients receiving bDMARDs, especially rituximab. However, more data are needed to establish effective preventive strategies for HBsAg-negative/HBcAb-positive patients. It seems safe to use cyclosporine and TNF inhibitors in patients with HCV infection, while those undergoing rituximab therapies should be frequently monitored for HCV activity.Abbreviations: ABT: abatacept; ADA: adalimumab; AS: ankylosing spondylitis; bDMARDs: biologic disease-modifying anti-rheumatic drugs; CKD: chronic kidney disease; COPD: chronic obstructive pulmonary disease; CS: corticosteroids; CsA: cyclosporine A; csDMARDs: conventional synthetic disease-modifying anti-rheumatic drugs; CZP: certolizumab; DAAs: direct-acting antiviral agents; DM: diabetes mellitus; DOT: directly observed therapy; EIN: Emerging Infections Network; ETN: etanercept; GOL: golimumab; GPRD: General Practice Research Database; HBV: hepatitis B virus; HBVr: HBV reactivation; HBsAg+: HBsAg-positive; HBsAg-/anti-HBc+: HBsAg-negative anti-HBc antibodies-positive; HCV: hepatitis C virus; HCQ: hydroxychloroquine: IFX: infliximab; IL-6: interleukin-6; JAK: Janus kinase; LEF: leflunomide; LTBI: latent tuberculosis infection; mAb: monoclonal antibody; MTX: methotrexate; OR: odds ratio; PsA: psoriatic arthritis; PMS: post-marketing surveillance; RA: rheumatoid arthritis; TNF: tumor necrosis factor; TNFi: tumor necrosis factor inhibitor; SCK: secukinumab; SSZ: sulfasalazine; TOZ: tocilizumab; RCT: randomized controlled trial; RR: relative risk; RTX: rituximab; 3HP: 3-month once-weekly isoniazid plus rifapentine; TB: tuberculosis; tsDMARDs: targeted synthetic disease-modifying anti-rheumatic drugs; UTK: ustekinumab; WHO: World Health Organization.
Collapse
Affiliation(s)
- Ying-Ming Chiu
- Rheumatology and Immunology Center, China Medical University Hospital, Taichung, Taiwan.,College of Medicine, China Medical University, Taichung, Taiwan
| | - Der-Yuan Chen
- Rheumatology and Immunology Center, China Medical University Hospital, Taichung, Taiwan.,College of Medicine, China Medical University, Taichung, Taiwan.,Translational Medicine Laboratory, Rheumatic Diseases Research Center, China Medical University Hospital, Taichung, Taiwan.,Program in Translational Medicine and Rong Hsing Research Center for Translational Medicine, National Chung Hsing University, Taichung, Taiwan.,Institute of Biochemistry, Microbiology and Immunology, Chung Shan Medical University, Taichung, Taiwan
| |
Collapse
|
102
|
Van Coillie S, Wiernicki B, Xu J. Molecular and Cellular Functions of CTLA-4. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1248:7-32. [PMID: 32185705 DOI: 10.1007/978-981-15-3266-5_2] [Citation(s) in RCA: 120] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) is an inhibitory receptor belonging to the CD28 immunoglobulin subfamily, expressed primarily by T-cells. Its ligands, CD80 and CD86, are typically found on the surface of antigen-presenting cells and can either bind CD28 or CTLA-4, resulting in a costimulatory or a co-inhibitory response, respectively. Because of its dampening effect, CTLA-4 is a crucial regulator of T-cell homeostasis and self-tolerance. The mechanisms by which CTLA-4 exerts its inhibitory function can be categorized as either cell-intrinsic (affects the CTLA-4 expressing T-cell) or cell-extrinsic (affects secondary cells). Research from the last decade has shown that CTLA-4 mainly acts in a cell-extrinsic manner via its competition with CD28, CTLA-4-mediated trans-endocytosis of CD80 and CD86, and its direct tolerogenic effects on the interacting cell. Nonetheless, intrinsic CTLA-4 signaling has been implicated in T-cell motility and the regulation of CTLA-4 its subcellular localization amongst others. CTLA-4 is well recognized as a key immune checkpoint and has gained significant momentum as a therapeutic target in the field of autoimmunity and cancer. In this chapter, we describe the role of costimulation in immune response induction as well as the main mechanisms by which CTLA-4 can inhibit this process.
Collapse
Affiliation(s)
- Samya Van Coillie
- Molecular Signaling and Cell Death Unit, VIB-UGent Center for Inflammation Research, Zwijnaarde, 9052, Ghent, Belgium.
| | - Bartosz Wiernicki
- Molecular Signaling and Cell Death Unit, VIB-UGent Center for Inflammation Research, Zwijnaarde, 9052, Ghent, Belgium
| | - Jie Xu
- Institutes of Biomedical Sciences, Zhongshan-Xuhui Hospital, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
103
|
Gerussi A, D'Amato D, Cristoferi L, O'Donnell SE, Carbone M, Invernizzi P. Multiple therapeutic targets in rare cholestatic liver diseases: Time to redefine treatment strategies. Ann Hepatol 2020; 19:5-16. [PMID: 31771820 DOI: 10.1016/j.aohep.2019.09.009] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Revised: 09/27/2019] [Accepted: 09/27/2019] [Indexed: 02/04/2023]
Abstract
Primary biliary cholangitis and primary sclerosing cholangitis are rare diseases affecting the bile ducts and the liver. The limited knowledge of their pathogenesis leads to limited therapeutic options. Nevertheless, the landscape of novel therapies for these cholangiopathies is now rapidly changing, providing new treatment opportunities for patients and clinicians involved in their care. The aim of this review is to summarize the evidence of novel molecules under investigation for primary biliary cholangitis and primary sclerosing cholangitis and to discuss how they can potentially change current treatment paradigms.
Collapse
Affiliation(s)
- Alessio Gerussi
- Division of Gastroenterology and Center for Autoimmune Liver Diseases, Department of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy; European Reference Network on Hepatological Diseases (ERN RARE-LIVER), San Gerardo Hospital, Monza, Italy
| | - Daphne D'Amato
- Division of Gastroenterology and Center for Autoimmune Liver Diseases, Department of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy; European Reference Network on Hepatological Diseases (ERN RARE-LIVER), San Gerardo Hospital, Monza, Italy
| | - Laura Cristoferi
- Division of Gastroenterology and Center for Autoimmune Liver Diseases, Department of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy; European Reference Network on Hepatological Diseases (ERN RARE-LIVER), San Gerardo Hospital, Monza, Italy
| | - Sarah Elizabeth O'Donnell
- Division of Gastroenterology and Center for Autoimmune Liver Diseases, Department of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy; European Reference Network on Hepatological Diseases (ERN RARE-LIVER), San Gerardo Hospital, Monza, Italy
| | - Marco Carbone
- Division of Gastroenterology and Center for Autoimmune Liver Diseases, Department of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy; European Reference Network on Hepatological Diseases (ERN RARE-LIVER), San Gerardo Hospital, Monza, Italy
| | - Pietro Invernizzi
- Division of Gastroenterology and Center for Autoimmune Liver Diseases, Department of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy; European Reference Network on Hepatological Diseases (ERN RARE-LIVER), San Gerardo Hospital, Monza, Italy.
| |
Collapse
|
104
|
|
105
|
Drug repurposing to improve treatment of rheumatic autoimmune inflammatory diseases. Nat Rev Rheumatol 2019; 16:32-52. [PMID: 31831878 DOI: 10.1038/s41584-019-0337-0] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/01/2019] [Indexed: 02/08/2023]
Abstract
The past century has been characterized by intensive efforts, within both academia and the pharmaceutical industry, to introduce new treatments to individuals with rheumatic autoimmune inflammatory diseases (RAIDs), often by 'borrowing' treatments already employed in one RAID or previously used in an entirely different disease, a concept known as drug repurposing. However, despite sharing some clinical manifestations and immune dysregulation, disease pathogenesis and phenotype vary greatly among RAIDs, and limited understanding of their aetiology has made repurposing drugs for RAIDs challenging. Nevertheless, the past century has been characterized by different 'waves' of repurposing. Early drug repurposing occurred in academia and was based on serendipitous observations or perceived disease similarity, often driven by the availability and popularity of drug classes. Since the 1990s, most biologic therapies have been developed for one or several RAIDs and then tested among the others, with varying levels of success. The past two decades have seen data-driven repurposing characterized by signature-based approaches that rely on molecular biology and genomics. Additionally, many data-driven strategies employ computational modelling and machine learning to integrate multiple sources of data. Together, these repurposing periods have led to advances in the treatment for many RAIDs.
Collapse
|
106
|
Wu M, Shen J. From Super-Enhancer Non-coding RNA to Immune Checkpoint: Frameworks to Functions. Front Oncol 2019; 9:1307. [PMID: 31824865 PMCID: PMC6883490 DOI: 10.3389/fonc.2019.01307] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2019] [Accepted: 11/11/2019] [Indexed: 12/13/2022] Open
Abstract
Super-enhancers (SEs) are clusters of enhancers that play a key role in regulating genes that determine cell identity. Enhancer RNAs (eRNAs) are non-coding RNAs transcribed from enhancers that function to promote the enhancer's functions via multiple mechanisms, such as recruiting transcription factors to specific enhancers, promoting enhancer-promoter looping, directing chromatin accessibility, interacting with RNA polymerase II and facilitating histone acetylation. Understanding how super-enhancer RNAs (seRNAs) contribute to specific gene regulation has thus become an area of active interest. Immune checkpoint deregulation is one of the key characteristics of tumors and autoimmune diseases, and is also closely related to cell identity. Recent studies revealed a potential pathway for seRNA's involvement in regulating the expression of immune checkpoints. The present study reviews the current knowledge of eRNA function, immune checkpoint blockage mechanism, and its effect. In addition, for the first time, we explore the direct and indirect roles of seRNAs in regulating immune checkpoint expression in cancer and autoimmune diseases.
Collapse
Affiliation(s)
- Manqing Wu
- State Key Laboratory for Oncogenes and Related Genes, Key Laboratory of Gastroenterology & Hepatology, Division of Gastroenterology and Hepatology, Ministry of Health, School of Medicine, Shanghai Cancer Institute, Shanghai Institute of Digestive Disease, Ren Ji Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Jun Shen
- State Key Laboratory for Oncogenes and Related Genes, Key Laboratory of Gastroenterology & Hepatology, Division of Gastroenterology and Hepatology, Ministry of Health, School of Medicine, Shanghai Cancer Institute, Shanghai Institute of Digestive Disease, Ren Ji Hospital, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
107
|
Gratacós J, Galíndez E, Otón T. Is obesity a predictor for lack of response to treatment in psoriatic arthritis? A systematic review. ACTA ACUST UNITED AC 2019; 17:268-278. [PMID: 31708450 DOI: 10.1016/j.reuma.2019.06.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Revised: 04/23/2019] [Accepted: 06/05/2019] [Indexed: 10/25/2022]
Abstract
OBJECTIVES To update the study of the association between obesity and treatment response in psoriatic arthritis. METHODS Updating a systematic review of clinical trials, prospective or retrospective longitudinal studies and case-control studies in psoriatic arthritis in which obesity was assessed as a predictor of efficacy or toxicity. Risks of bias were assessed with validated scales. A meta-analysis of the results of studies with similar outcome variables and weight measurements was performed. RESULTS Twenty-one studies were included (6 review of clinical trials, 6 longitudinal studies, 7 registers and one case-control studie), with moderate quality. The risk of achieving an ACR20 response if weight≥100kg was estimated at OR=1.42 (1-2.08) and that of withdrawing treatment in an OR of 1.60 (95% CI: 1.34 - 1.92). CONCLUSIONS There seems to be a greater risk of withdrawal of treatment due to inefficacy and difficulty in achieving remission in patients with psoriatic arthritis if they are obese.
Collapse
Affiliation(s)
- Jordi Gratacós
- Servicio de Reumatología, Hospital Universitari Parc Taulí, Universitat Autònoma de Barcelona, Sabadell (Barcelona), España
| | - Eva Galíndez
- Servicio de Reumatología, Hospital Universitario Basurto, Bilbao, Vizcaya, España
| | - Teresa Otón
- Instituto de Salud Musculoesquelética, Madrid, España.
| |
Collapse
|
108
|
Lubrano E, Scriffignano S, Perrotta FM. TNF-alpha inhibitors for the six treatment targets of psoriatic arthritis. Expert Rev Clin Immunol 2019; 15:1303-1312. [PMID: 31652079 DOI: 10.1080/1744666x.2020.1685382] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Introduction: Psoriatic Arthritis (PsA) is a chronic inflammatory disease characterized by psoriasis, synovitis, enthesitis, spondylitis, and association with extra-articular manifestations and comorbidities. Treatment of PsA dramatically changed since the introduction of anti-TNF drugs which have shown to reduce the symptoms and signs of the disease and slow the radiographic progression. Despite the introduction of new molecules with different mechanisms of action, the role of anti-TNF in the treatment of all disease manifestations of this intriguing disease is still central.Areas covered: The aim of this paper is to review the role of anti-TNF drugs in the treatment of different disease domains in PsA (peripheral and axial joints, skin, enthesis, patient's reported outcomes, extra-articular manifestations), reporting data from randomized clinical trials and observational studies. An extensive literature search was performed on PubMed, with no limits or filters. The following search terms were used: 'anti-TNF', 'Psoriatic Arthritis'.Expert opinion: Despite the emergence of different new treatments, anti-TNF therapy remains central in the management of all disease domains in PsA patients.
Collapse
Affiliation(s)
- Ennio Lubrano
- Academic Rheumatology Unit. Dipartimento di Medicina e Scienze della salute "Vincenzo Tiberio", Università degli Studi del Molise, Campobasso, Italy
| | - Silvia Scriffignano
- Academic Rheumatology Unit. Dipartimento di Medicina e Scienze della salute "Vincenzo Tiberio", Università degli Studi del Molise, Campobasso, Italy
| | - Fabio Massimo Perrotta
- Academic Rheumatology Unit. Dipartimento di Medicina e Scienze della salute "Vincenzo Tiberio", Università degli Studi del Molise, Campobasso, Italy
| |
Collapse
|
109
|
Silvagni E, Bortoluzzi A, Ciancio G, Govoni M. Biological and synthetic target DMARDs in psoriatic arthritis. Pharmacol Res 2019; 149:104473. [PMID: 31585178 DOI: 10.1016/j.phrs.2019.104473] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Revised: 08/28/2019] [Accepted: 09/30/2019] [Indexed: 12/29/2022]
Abstract
Psoriatic arthritis (PsA) is a chronic multi-faceted immune-mediated systemic disorder, characterized by articular, cutaneous, enthesis, nail and spine involvement. Articular manifestations of PsA are particularly common and highly disabling for patients, while the heterogeneous clinical subsets of the disease are challenging for clinicians. In recent years, research has made many advances in understanding the pathogenesis of the disease from genetic, epigenetic and molecular points of view. New drugs are now available for the treatment of this condition, and, in particular, TNF-alfa inhibitors, historically the first biologicals approved in PsA, are now juxtaposed by new biological disease modifying anti-rheumatic drugs (bDMARDs) with different modes of action. Targeting IL-12/IL-23 p40 common subunit with ustekinumab, IL-17A with secukinumab and ixekizumab, T cells co-stimulation with abatacept, is now possible, safe and effective. Moreover, targeted synthetic molecules with oral administration are available, with the possibility to interfere with phosphodiesterase-4 and JAK/STAT pathways. Indeed, new drugs are under development, with the possibility to target selectively IL-17 receptor, IL-23, and other key molecular targets in the pathogenesis of this condition. In this narrative review, we provide an up-to-date overview of the current application of biological and targeted synthetic DMARDs in the field of PsA, with particular regard to the clinical significance of this possibility to target a higher number of distinct immune-pathways.
Collapse
Affiliation(s)
- Ettore Silvagni
- Department of Medical Sciences, Section of Rheumatology, University of Ferrara and Azienda Ospedaliero-Universitaria Sant'Anna, Cona, Ferrara, Italy
| | - Alessandra Bortoluzzi
- Department of Medical Sciences, Section of Rheumatology, University of Ferrara and Azienda Ospedaliero-Universitaria Sant'Anna, Cona, Ferrara, Italy
| | - Giovanni Ciancio
- Department of Medical Sciences, Section of Rheumatology, University of Ferrara and Azienda Ospedaliero-Universitaria Sant'Anna, Cona, Ferrara, Italy.
| | - Marcello Govoni
- Department of Medical Sciences, Section of Rheumatology, University of Ferrara and Azienda Ospedaliero-Universitaria Sant'Anna, Cona, Ferrara, Italy
| |
Collapse
|
110
|
Abstract
Psoriasis is a multisystemic, inflammatory skin condition that can affect many areas of the body, but most commonly the extensor surfaces of the elbows and knees, and sometimes the intergluteal and umbilical area. It has a prevalence of 2–4% in western adults, and 20–30% of psoriasis patients will develop psoriatic arthritis (PsA). PsA is an inflammatory musculoskeletal disease associated with cutaneous psoriasis. It affects men and women almost equally with a peak age at onset of 40 and 50 years. It is a diverse disease that affects multiple organ systems includes peripheral and axial joints, entheses, skin, and nails. PsA is associated with comorbidities such as osteoporosis, uveitis, subclinical bowel inflammation, and cardiovascular disease. Given this heterogeneity, its diagnosis has been difficult. Here we present an updated review of its classification criteria CASPAR (classification criteria for PsA), use of screening tools to aid in early diagnosis, recent findings on pathogenesis, and new therapeutic approaches including new biologic medications.
Collapse
Affiliation(s)
- Vanessa Ocampo D
- University of Toronto, Psoriatic Arthritis Program, University Health Network, Toronto Western Hospital, Toronto, Ontario, Canada
| | - Dafna Gladman
- University of Toronto, Psoriatic Arthritis Program, University Health Network, Toronto Western Hospital, Toronto, Ontario, Canada.,Krembil Research Institute, Toronto Western Hospital, Toronto, Ontario, Canada
| |
Collapse
|
111
|
Risk of major autoimmune diseases in female breast cancer patients: A nationwide, population-based cohort study. PLoS One 2019; 14:e0222860. [PMID: 31536611 PMCID: PMC6752851 DOI: 10.1371/journal.pone.0222860] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Accepted: 09/09/2019] [Indexed: 01/17/2023] Open
Abstract
BACKGROUND Breast cancer is one of the most common malignancies among women. However, there remains no consensus in current literature on the incidence of autoimmune diseases among breast cancer patients. The purpose of this study was to evaluate the risks of major autoimmune diseases (MAD) including systemic lupus erythematosus (SLE), rheumatoid arthritis (RA), Sjögren's syndrome (SS) and dermatomyositis (DMtis)/polymyositis (PM) in female breast cancer patients. METHODS Using the Taiwanese National Health Insurance Research Database (NHIRD) records from 2003 to 2013, we identified newly-diagnosed female breast cancer patients and randomly selected females without breast cancer in the period 2007 to 2013 into a control group. We matched the two cohorts using a 1:4 ratio based on age, and the year of index date for comparison of the risk of major autoimmune diseases. We estimated and compared the relative risks of autoimmune diseases in female breast cancer patients and females without breast cancer. RESULTS A total of 54,311 females with breast cancer and 217,244 matched females without breast cancer were included in this study. For SLE, the incidence rates were 2.3 (breast cancer group) vs. 10.0 (control group) per 100,000 women years; for RA rates were 19.3 (breast cancer group) vs. 42.7 (control group) per 100,000 women years; and for SS rates were 20.5 (breast cancer group) vs. 38.2 (control group) per 100,000 women years. After adjusting for potential confounders, the hazard ratios (95% confidence intervals) for female breast cancer patients vs. control group were 0.04 (0.01-0.24) for SLE; 0.03 (0.02-0.04) for RA; and 0.21 (0.09-0.48) for SS. CONCLUSION Female breast cancer patients had lower risks of SLE, RA and SS when compared to female individuals without breast cancer. However, there was no significant difference in the risk of developing DMtis/PM between both groups.
Collapse
|
112
|
Coates LC, Strand V, Wilson H, Revicki D, Stolshek B, Samad A, Chung JB, Gladman D, Mease PJ. Measurement properties of the minimal disease activity criteria for psoriatic arthritis. RMD Open 2019; 5:e001002. [PMID: 31565243 PMCID: PMC6744081 DOI: 10.1136/rmdopen-2019-001002] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2019] [Revised: 07/24/2019] [Accepted: 08/11/2019] [Indexed: 11/19/2022] Open
Abstract
Objective To comprehensively assess evidence on the measurement properties of the minimal disease activity (MDA) criteria, a composite measure of the state of disease activity in psoriatic arthritis (PsA). Methods A targeted literature review was conducted to identify studies that informed the validity and/or ability of the MDA to detect change among patients known to have experienced a change in clinical status. The search was conducted using MEDLINE and Embase databases (published as of October 2017). Pertinent articles provided by investigators and identified from select conference proceedings were also evaluated. Results A total of 20 publications met the inclusion criteria. The MDA criteria were consistently associated with other indicators of disease activity/severity. The ability of the MDA criteria to detect change was supported in randomised controlled trials (n=10), with a greater percentage of patients randomised to active treatments achieving MDA relative to patients in comparator arms. Long-term observational studies (n=2) provided additional support for the ability of the MDA to detect within-subject change in the real-world settings. Conclusion Evidence supports the MDA as a valid measure of disease activity in PsA that can detect between-group and within-subject change. The MDA is a comprehensive measure and clinically meaningful endpoint to assess the impact of interventions on PsA disease activity.
Collapse
Affiliation(s)
- Laura C Coates
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, UK
| | - Vibeke Strand
- Immunology/Rheumatology, Stanford University, Stanford, California, USA.,Biopharmaceutical Consultant, Portola Valley, California, USA
| | | | | | | | - Ahmed Samad
- Pharmaceutical Product Development, San Diego, California, USA
| | | | - Dafna Gladman
- Medicine/Rheumatology, Krembil Research Institute, Toronto, Ontario, Canada
| | - Philip J Mease
- School of Medicine, Swedish Medical Center and University of Washington, Seattle, Washington, USA
| |
Collapse
|
113
|
Bravo A, Kavanaugh A. Bedside to bench: defining the immunopathogenesis of psoriatic arthritis. Nat Rev Rheumatol 2019; 15:645-656. [PMID: 31485004 DOI: 10.1038/s41584-019-0285-8] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/23/2019] [Indexed: 02/07/2023]
Abstract
Psoriatic arthritis (PsA) is an immune-mediated, systemic inflammatory disorder. PsA can present with heterogeneous clinical features. Advances in understanding the immunopathogenesis of PsA have helped to facilitate the development of agents targeting specific components of the dysregulated inflammatory and immune responses relevant to PsA. Interestingly, agents with distinct mechanisms of action have shown differential responses across the various disease domains of PsA, counter to what might have been expected from basic science investigations. Here, we review data utilizing various novel targeted therapies for PsA, focusing on biologic and targeted synthetic therapies. These data might support the idea of a 'bedside to bench' concept, whereby results from clinical trials of specific targeted therapies inform our understanding of the immunopathogenesis of PsA. For example, TNF inhibition confers substantial and comparable benefit for all domains of PsA, supporting the view that TNF is a central pro-inflammatory cytokine across diverse areas of disease involvement. On the other hand, inhibition of IL-12-IL-23, as compared with inhibition of TNF, has greater efficacy for psoriasis, comparable efficacy for peripheral arthritis, but was ineffective in studies of axial spondyloarthritis. Data from studies of agents with distinct mechanisms of action will help to further refine our understanding of the immunopathogenesis of PsA.
Collapse
Affiliation(s)
- Arlene Bravo
- Division of Rheumatology, Allergy & Immunology, University of California San Diego, San Diego, CA, USA
| | - Arthur Kavanaugh
- Division of Rheumatology, Allergy & Immunology, University of California San Diego, San Diego, CA, USA.
| |
Collapse
|
114
|
Tateiwa D, Yoshikawa H, Kaito T. Cartilage and Bone Destruction in Arthritis: Pathogenesis and Treatment Strategy: A Literature Review. Cells 2019; 8:cells8080818. [PMID: 31382539 PMCID: PMC6721572 DOI: 10.3390/cells8080818] [Citation(s) in RCA: 102] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2019] [Revised: 07/30/2019] [Accepted: 08/01/2019] [Indexed: 02/07/2023] Open
Abstract
Arthritis is inflammation of the joints accompanied by osteochondral destruction. It can take many forms, including osteoarthritis, rheumatoid arthritis, and psoriatic arthritis. These diseases share one commonality—osteochondral destruction based on inflammation. The background includes a close interaction between osseous tissues and immune cells through various inflammatory cytokines. However, the tissues and cytokines that play major roles are different in each disease, and as a result, the mechanism of osteochondral destruction also differs. In recent years, there have been many findings regarding not only extracellular signaling pathways but also intracellular signaling pathways. In particular, we anticipate that the intracellular signals of osteoclasts, which play a central role in bone destruction, will become novel therapeutic targets. In this review, we have summarized the pathology of arthritis and the latest findings on the mechanism of osteochondral destruction, as well as present and future therapeutic strategies for these targets.
Collapse
Affiliation(s)
- Daisuke Tateiwa
- Department of Orthopedic Surgery, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Hideki Yoshikawa
- Department of Orthopedic Surgery, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Takashi Kaito
- Department of Orthopedic Surgery, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan.
| |
Collapse
|
115
|
Badell IR, Karadkhele GM, Vasanth P, Farris AB, Robertson JM, Larsen CP. Abatacept as rescue immunosuppression after calcineurin inhibitor treatment failure in renal transplantation. Am J Transplant 2019; 19:2342-2349. [PMID: 30768841 DOI: 10.1111/ajt.15319] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2018] [Revised: 01/23/2019] [Accepted: 02/11/2019] [Indexed: 01/25/2023]
Abstract
A majority of kidney transplant recipients receive calcineurin inhibitor-based immunosuppression. However, some do not tolerate calcineurin inhibitors and require other immunosuppressive strategies. Until recently, alternative approaches have been associated with inferior outcomes, but recent methods have effectively utilized belatacept in calcineurin inhibitor-intolerant patients. Though promising, belatacept uptake has been limited by higher acute rejection rates, unavailability due to production shortages, and logistical challenges as a result of intravenous infusion requirements. Interestingly, its predecessor abatacept is clinically available in subcutaneous formulation to treat autoimmune disorders but has not been used in clinical transplantation. Here we report on a series of 9 calcineurin inhibitor-intolerant transplant recipients converted to abatacept early after transplant as rescue immunosuppression during periods of belatacept unavailability. Retrospective review revealed successful allograft salvage and 100% patient and graft survival (median 115 months) after conversion to abatacept. Patients received abatacept for a median duration of 82 months with stable, long-term renal allograft function, a single cellular rejection episode, and no clinically apparent protective immunity concerns. Hence our findings suggest that future clinical studies utilizing abatacept either de novo or as conversion therapy in transplant recipients should be considered.
Collapse
Affiliation(s)
| | | | | | - Alton Brad Farris
- Department of Pathology & Laboratory Medicine, Emory University School of Medicine, Atlanta, Georgia
| | | | | |
Collapse
|
116
|
Bowlus CL, Yang GX, Liu CH, Johnson CR, Dhaliwal SS, Frank D, Levy C, Peters MG, Vierling JM, Gershwin ME. Therapeutic trials of biologics in primary biliary cholangitis: An open label study of abatacept and review of the literature. J Autoimmun 2019; 101:26-34. [DOI: 10.1016/j.jaut.2019.04.005] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Revised: 04/01/2019] [Accepted: 04/06/2019] [Indexed: 12/12/2022]
|
117
|
Chimenti MS, Triggianese P, De Martino E, Conigliaro P, Fonti GL, Sunzini F, Caso F, Perricone C, Costa L, Perricone R. An update on pathogenesis of psoriatic arthritis and potential therapeutic targets. Expert Rev Clin Immunol 2019; 15:823-836. [PMID: 31177868 DOI: 10.1080/1744666x.2019.1627876] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Introduction: Innate immune response and bone remodeling are key factors contributing to the pathogenesis of psoriatic arthritis (PsA). Moreover, the evidence of autoantibodies in patients' sera suggests an autoimmune side in PsA. Besides the immune pathways, studies strongly support the role of genetic risk alleles in affecting the clinical heterogeneity of PsA as well as the response to therapy. A good clinical response to treatment, indeed, represents a challenge in PsA patients and the identification of patient-targeted therapies is still a critical issue. Areas covered: We performed a systematic review aiming at describing new evidence on PsA pathogenesis and treatments. Reported items for systematic reviews (PRISMA checklist) were analyzed. Studies included from the PubMed database addressed the following items: innate immunity, autoimmunity, bone remodeling, and therapeutic targets in PsA; time frame of research 1970-2019. Specifically, we reviewed data on IL-17 inhibitors, abatacept, JAK inhibitors, ABT 122, and A (3) adenosine receptors agonist, CF101. Expert opinion: In PsA an intriguing pathogenetic network has been documented. Several biological and synthetic drugs are promising in terms of efficacy and safety profile.
Collapse
Affiliation(s)
- Maria Sole Chimenti
- a Rheumatology, allergology and clinical immunology, Department of Systems Medicine, University of Rome Tor Vergata , Rome , Italy
| | - Paola Triggianese
- a Rheumatology, allergology and clinical immunology, Department of Systems Medicine, University of Rome Tor Vergata , Rome , Italy
| | - Erica De Martino
- a Rheumatology, allergology and clinical immunology, Department of Systems Medicine, University of Rome Tor Vergata , Rome , Italy
| | - Paola Conigliaro
- a Rheumatology, allergology and clinical immunology, Department of Systems Medicine, University of Rome Tor Vergata , Rome , Italy
| | - Giulia Lavinia Fonti
- a Rheumatology, allergology and clinical immunology, Department of Systems Medicine, University of Rome Tor Vergata , Rome , Italy
| | - Flavia Sunzini
- a Rheumatology, allergology and clinical immunology, Department of Systems Medicine, University of Rome Tor Vergata , Rome , Italy
| | - Francesco Caso
- b Rheumatology Unit, Department of Clinical Medicine and Surgery, School of Medicine and Surgery, University Federico II , Naples , Italy
| | - Carlo Perricone
- c Arthritis Center, Department of Internal Medicine and Medical Specialties, Sapienza University of Rome , Rome , Italy
| | - Luisa Costa
- b Rheumatology Unit, Department of Clinical Medicine and Surgery, School of Medicine and Surgery, University Federico II , Naples , Italy
| | - Roberto Perricone
- a Rheumatology, allergology and clinical immunology, Department of Systems Medicine, University of Rome Tor Vergata , Rome , Italy
| |
Collapse
|
118
|
FitzGerald O, Ritchlin C. Opportunities and challenges in the treatment of psoriatic arthritis. Best Pract Res Clin Rheumatol 2019; 32:440-452. [PMID: 31171314 DOI: 10.1016/j.berh.2019.03.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
In this chapter, we review the opportunities and challenges posed by the treatment options currently available in the treatment of psoriatic arthritis. Both established and new or emerging treatment options are discussed using a domain-based approach. Finally, approaches to how treatment can be optimized together with some clinical pearls are presented and discussed. With the increasing treatment options available, we need a better way of deciding which treatment should be considered for which patient. On the basis of current knowledge, some guidance is provided on how these choices might best be made.
Collapse
Affiliation(s)
- Oliver FitzGerald
- St. Vincent's University Hospital, Conway Institute for Biomolecular Research, University College Dublin, Ireland.
| | - Christopher Ritchlin
- Chief, Allergy, Immunology & Rheumatology Division, Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, USA
| |
Collapse
|
119
|
McInnes IB, Ferraccioli G, D'Agostino MA, Le Bars M, Banerjee S, Ahmad HA, Elbez Y, Mease PJ. Body mass index and treatment response to subcutaneous abatacept in patients with psoriatic arthritis: a post hoc analysis of a phase III trial. RMD Open 2019; 5:e000934. [PMID: 31245054 PMCID: PMC6560664 DOI: 10.1136/rmdopen-2019-000934] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Revised: 04/30/2019] [Accepted: 05/03/2019] [Indexed: 12/23/2022] Open
Abstract
Objective This post hoc analysis of the phase III Active PSoriaTic Arthritis RAndomizEd TriAl (ASTRAEA) evaluated the effect of baseline body mass index (BMI) on subsequent response to subcutaneous (SC) abatacept in patients with psoriatic arthritis (PsA). Methods In ASTRAEA, patients with active PsA were randomised (1:1) to receive blinded weekly SC abatacept 125 mg or placebo for 24 weeks. Treatment response at week 24 was assessed by the proportions of patients achieving American College of Rheumatology 20% improvement response, Disease Activity Score in 28 joints (DAS28 (C reactive protein (CRP))) ≤3.6 and <2.6, Health Assessment Questionnaire-Disability Index reduction from baseline ≥0.35 and radiographic non-progression (defined as change from baseline ≤0 in PsA-modified total Sharp/van der Heijde score). Responses were stratified by baseline BMI (underweight/normal, <25 kg/m2; overweight, 25–30 kg/m2; obese, >30 kg/m2) and compared in univariate and multivariate models. Results Of 212/213 and 210/211 patients with baseline BMI data in the abatacept and placebo groups, respectively, 15% and 19% were underweight/normal, 36% and 27% were overweight, and 49% and 54% were obese. After adjusting for baseline characteristics, there were no significant differences for any outcome measure at week 24 with abatacept in the overweight or obese versus underweight/normal subgroup. In the placebo group, patients in the obese versus underweight/normal subgroup were significantly less likely to achieve DAS28 (CRP) <2.6 at week 24 (OR 0.26; 95% CI 0.08 to 0.87; p=0.03). Conclusion BMI does not impact clinical or radiographic response to SC abatacept in patients with PsA. Trial registration number NCT01860976.
Collapse
Affiliation(s)
- Iain B McInnes
- Institute of Infection, Immunity & Inflammation, University of Glasgow, Glasgow, UK
| | - Gianfranco Ferraccioli
- Division of Rheumatology, IRCCS-Fondazione Policlinico Universitario A. Gemelli-Catholic University of the Sacred Heart, Rome, Italy
| | - Maria-Antonietta D'Agostino
- Rheumatology, Université Versailles Saint-Quentin en Yvelines, Ambroise Paré Hospital, APHP, Boulogne-Billancourt, France
| | | | | | | | | | - Philip J Mease
- Swedish Medical Center and University of Washington, Seattle, Washington, USA
| |
Collapse
|
120
|
Cantini F, Niccoli L, Capone A, Petrone L, Goletti D. Risk of tuberculosis reactivation associated with traditional disease modifying anti-rheumatic drugs and non-anti-tumor necrosis factor biologics in patients with rheumatic disorders and suggestion for clinical practice. Expert Opin Drug Saf 2019; 18:415-425. [PMID: 31066297 DOI: 10.1080/14740338.2019.1612872] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
INTRODUCTION Two classes of biologics, anti-tumor necrosis factor (TNF) and non-anti-TNF targeted, are currently available for the treatment of rheumatic diseases. AREAS COVERED Discussion on the need for LTBI diagnosis in rheumatic patients treated csDMARDs and non-anti-TNFs through a review of the literature. The literature, updated to 15 April 2019, on tuberculosis (TB) reactivation risk in patients exposed to conventional synthetic disease-modifying antirheumatic drugs (csDMARDs) and non-anti-TNF biologics was reviewed. EXPERT OPINION An increased risk of TB reactivation in patients receiving csDMARDs (except sulphasalazine) resulted, while a review of clinical trials, and Periodic Safety Update Reports from pharmaceutical Companies evidenced a very low or absent risk for non-anti-TNF biologics. Hence, a contradiction emerges considering that latent TB infection (LTBI) screening is recommended for non-anti-TNF candidates but not for csDMARDs. Concerning the low TB incidence countries, several actions could be undertaken, including to screen all patients independently on the treatment, to omit the procedure in non-anti-TNF candidates, or to perform the LTBI investigations only in high-risk patients. According to WHO guidelines, LTBI screening in low TB risk countries seems unnecessary, except in high TB risk subjects.
Collapse
Affiliation(s)
- Fabrizio Cantini
- a Department of Rheumatology , Hospital of Prato , Prato , Italy
| | - Laura Niccoli
- a Department of Rheumatology , Hospital of Prato , Prato , Italy
| | - Alessandro Capone
- b Clinical Department , National Institute for Infectious Diseases L. Spallanzani-IRCCS , Rome , Italy
| | - Linda Petrone
- c Translational Research Unit, Department of Epidemiology and Preclinical Research , "L. Spallanzani" National Institute for Infectious Diseases (INMI), IRCCS , Rome , Italy
| | - Delia Goletti
- c Translational Research Unit, Department of Epidemiology and Preclinical Research , "L. Spallanzani" National Institute for Infectious Diseases (INMI), IRCCS , Rome , Italy
| |
Collapse
|
121
|
Abstract
Psoriatic arthritis (PsA) affects up to one-third of patients with psoriasis. It is the major comorbidity of psoriasis because of the likelihood that loss of function and permanent disability will develop if initiation of treatment is delayed. Dermatologists are uniquely positioned to recognize early signs of PsA and be the first-line healthcare practitioners to detect PsA in patients with psoriasis. PsA can affect six clinical domains: peripheral arthritis, dactylitis, enthesitis, psoriasis, psoriatic nail disease, and axial disease. However, not every patient will have involvement of all domains and the domains affected can change over time. Complicating the diagnosis is the condition's similarity with other arthritic diseases and potential heterogeneity. In this article, we provide practical guidance for dermatologists for detecting PsA in patients with psoriasis. We also review the available treatment options by each clinical domain of PsA and give advice on how to interpret the results of PsA clinical trials. Through early recognition of PsA in patients with psoriasis and initiation of proper treatment, dermatologists can help to prevent PsA disease progression, irreversible joint damage, and resultant permanent disability, and improve quality of life.
Collapse
Affiliation(s)
- Alice Gottlieb
- Department of Dermatology, Icahn School of Medicine at Mt Sinai, New York, NY, USA
| | - Joseph F Merola
- Department of Medicine, Division of Rheumatology and Department of Dermatology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
122
|
Furst DE, Belasco J, Louie JS. Genetic and inflammatory factors associated with psoriatic arthritis: Relevance to diagnosis and management. Clin Immunol 2019; 202:59-75. [DOI: 10.1016/j.clim.2019.02.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Revised: 01/21/2019] [Accepted: 02/04/2019] [Indexed: 12/22/2022]
|
123
|
Wallace K, Bean C, Bowles T, Spencer SK, Randle W, Kyle PB, Shaffery J. Hypertension, Anxiety, and Blood-Brain Barrier Permeability Are Increased in Postpartum Severe Preeclampsia/Hemolysis, Elevated Liver Enzymes, and Low Platelet Count Syndrome Rats. Hypertension 2019; 72:946-954. [PMID: 30354708 DOI: 10.1161/hypertensionaha.118.11770] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Hypertension and inflammation during pregnancy are suggested to contribute to the development of postpartum depression and anxiety. Using a rat model of severe preeclampsia and hemolysis, elevated liver enzymes, and low platelet count syndrome, which displays both hypertension and inflammation during pregnancy, we evaluated whether rats were prone to develop depression or anxiety in the postpartum period. On gestational day 12, miniosmotic pumps infusing sFlt-1 (soluble fms-like tyrosine kinase-1) and sEng (soluble endoglin) were placed into rats, a subset of these rats was infused with 2 mg/kg of Orencia (abatacept) the following day to determine whether immune suppression via T-cell depletion prevented any changes in maternal depression or anxiety-like behavior. All rats, including normal pregnant (NP) controls, delivered between gestational days 21 and 22. Postpartum severe preeclamptic rats buried significantly more marbles compared with NP rats ( P=0.002) and Orencia-treated rats ( P=0.05). Severe preeclamptic rats spent significantly more time in closed arms of the elevated plus maze compared with NP rats ( P=0.009) and Orencia-treated rats ( P=0.05). Severe preeclamptic rats were hypertensive compared with NP ( P=0.03) and Orencia-treated rats ( P=0.01). Finally, severe preeclamptic rats had increased blood-brain barrier permeability compared with NP rats ( P=0.03), which was reversed in Orencia-treated rats ( P=0.008). These results suggest that severe preeclampsia/hemolysis, elevated liver enzymes, and low platelet count syndrome during pregnancy contributes to an increase in anxiety-like behavior, blood-brain barrier permeability, and hypertension in the postpartum. The current results suggest that T-cell suppression during pregnancy can also help prevent chronic hypertension and increased anxiety in the postpartum period.
Collapse
Affiliation(s)
- Kedra Wallace
- From the Department of Obstetrics and Gynecology (K.W., C.B., T.B., S.-K.S., W.R.), University of Mississippi Medical Center, Jackson
| | - Cynthia Bean
- From the Department of Obstetrics and Gynecology (K.W., C.B., T.B., S.-K.S., W.R.), University of Mississippi Medical Center, Jackson
| | - Teylor Bowles
- From the Department of Obstetrics and Gynecology (K.W., C.B., T.B., S.-K.S., W.R.), University of Mississippi Medical Center, Jackson
| | - Shauna-Kay Spencer
- From the Department of Obstetrics and Gynecology (K.W., C.B., T.B., S.-K.S., W.R.), University of Mississippi Medical Center, Jackson
| | - Wisdom Randle
- From the Department of Obstetrics and Gynecology (K.W., C.B., T.B., S.-K.S., W.R.), University of Mississippi Medical Center, Jackson
| | - Patrick B Kyle
- Department of Pathology (P.B.K.), University of Mississippi Medical Center, Jackson
| | - James Shaffery
- Department of Psychiatry, Center of Psychiatric Neuroscience (J.S.), University of Mississippi Medical Center, Jackson
| |
Collapse
|
124
|
Oelke KR, Chambenoit O, Majjhoo AQ, Gray S, Higgins K, Hur P. Persistence and adherence of biologics in US patients with psoriatic arthritis: analyses from a claims database. J Comp Eff Res 2019; 8:607-621. [PMID: 30912454 DOI: 10.2217/cer-2019-0023] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Aim: To evaluate the persistence and adherence of subcutaneous biologics in patients with psoriatic arthritis (PsA). Patients & methods: Psoriatic arthritis patients who initiated adalimumab, certolizumab pegol, etanercept, golimumab or secukinumab between 15 January 2016 and 31 July 2017 were identified in the Truven Databases. Outcomes included discontinuation rate, persistence and adherence over 12 months. Results: Of 1558 patients included, the 12-month discontinuation rate was lowest with secukinumab (36.5%), followed by adalimumab, golimumab, etanercept and certolizumab pegol (42.6-51.6%). Mean persistence ranged from 240.7 (certolizumab pegol) to 282.8 days (secukinumab). The mean proportion of days covered was highest with secukinumab (0.67) and lowest with certolizumab pegol (0.49). Conclusion: Patients who initiated secukinumab had the lowest discontinuation rate and highest persistence and adherence over 12 months.
Collapse
Affiliation(s)
- Kurt R Oelke
- Rheumatic Disease Center, Glendale, WI 53217, USA
| | | | | | | | - Kate Higgins
- Truven Health Analytics, Cambridge, MA 02142, USA
| | - Peter Hur
- Novartis Pharmaceuticals Corporation, East Hanover, NJ 07936, USA
| |
Collapse
|
125
|
So A, Inman RD. An overview of biologic disease-modifying antirheumatic drugs in axial spondyloarthritis and psoriatic arthritis. Best Pract Res Clin Rheumatol 2019; 32:453-471. [PMID: 31171315 DOI: 10.1016/j.berh.2018.12.002] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2018] [Revised: 07/23/2018] [Accepted: 12/21/2018] [Indexed: 12/12/2022]
Abstract
Biologic disease-modifying antirheumatic drugs (bDMARDs) are engineered proteins with high affinity for various proinflammatory immune mediators to reduce inflammation and its sequelae in various rheumatic diseases. These medications, introduced at the advent of the 21st century, have revolutionized the treatment of axial spondyloarthritis (including ankylosing spondylitis) and psoriatic arthritis. Currently approved bDMARDs for axial spondyloarthritis are etanercept, infliximab, adalimumab, golimumab, certolizumab pegol, and secukinumab. For psoriatic arthritis, all of these drugs are approved in addition to ixekizumab, ustekinumab, abatacept, and tofacitinib. Selection of the optimal bDMARD should consider patient comorbidity including uveitis, psoriasis, and inflammatory bowel disease.
Collapse
Affiliation(s)
- Anthony So
- Toronto Western Hospital, Suite 1E - 423, 399 Bathurst Street, Toronto, ON, M5T 2S8, Canada.
| | - Robert Davies Inman
- Toronto Western Hospital, Suite 1E - 423, 399 Bathurst Street, Toronto, ON, M5T 2S8, Canada.
| |
Collapse
|
126
|
Wade SM, Canavan M, McGarry T, Low C, Wade SC, Mullan RH, Veale DJ, Fearon U. Association of synovial tissue polyfunctional T-cells with DAPSA in psoriatic arthritis. Ann Rheum Dis 2019; 78:350-354. [PMID: 30626658 PMCID: PMC6390025 DOI: 10.1136/annrheumdis-2018-214138] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2018] [Revised: 11/29/2018] [Accepted: 12/18/2018] [Indexed: 01/23/2023]
Abstract
OBJECTIVE This study examines polyfunctional T-cells in psoriatic arthritis (PsA) synovial tissue and their associations with clinical disease and implications for therapy. METHODS PsA synovial tissue was enzymatically/mechanically digested to generate synovial tissue single cell suspensions. Frequencies of polyfunctional CD4, CD8, T-helper 1 (Th1), Th17 and exTh17 cells, using CD161 as a marker of Th17 plasticity, were determined by flow cytometry in matched PsA synovial tissue and peripheral blood. Synovial T-cell polyfunctionality was assessed in relation to Disease Activity in PSoriatic Arthritis (DAPSA) and in synovial cell suspensions cultured with a current mode of treatment, phosphodiesterase 4 (PDE4) inhibitor. RESULTS PsA synovial tissue infiltrating CD4+ T-cells expressed higher levels of interleukin (IL)-17A, interferon gamma (IFN-γ), GM-CSF and CD161, with parallel enrichment of Th1, Th17 and exTh17 T-helper subsets (all p<0.05). Interestingly, a significant proportion of synovial T-cell subsets were triple-positive for GM-CSF, tumour necrosis factor (-TNF), -IL-17 or IFN-γ compared with matched blood (all p<0.05). Importantly, frequencies of polyfunctional T-cells correlated with DAPSA: Th1-GM-CSF+/TNF+/IFN-γ+ (r=0.7, p<0.01), Th17-GM-CSF+/TNF+/IL-17+ (r=0.6, p<0.057) and exTh17-GM-CSF+/TNF+/IFN-γ+ (r=0.7, p=0.0096), with no associations observed for single cytokine-producing T-cells. Following ex vivo culture of PsA synovial tissue cell suspensions, polyfunctional GM-CSF+TNFα+IL-17A+ or/IFN-γ+-producing T-cells (p<0.05), but not single cytokine-producing T-cells, were inhibited with a PDE4 inhibitor. CONCLUSION These data demonstrate enrichment of polyfunctional T-cells in PsA synovial tissue which were strongly associated with DAPSA and ex vivo therapeutic response.
Collapse
Affiliation(s)
- Sarah M Wade
- Molecular Rheumatology, School of Medicine, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland
- St Vincent's University Hospital, Dublin Academic Health Care and University College Dublin, Dublin 4, Ireland
| | - Mary Canavan
- Molecular Rheumatology, School of Medicine, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland
| | - Trudy McGarry
- Molecular Rheumatology, School of Medicine, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland
- St Vincent's University Hospital, Dublin Academic Health Care and University College Dublin, Dublin 4, Ireland
| | - Candice Low
- St Vincent's University Hospital, Dublin Academic Health Care and University College Dublin, Dublin 4, Ireland
| | - Siobhan C Wade
- Molecular Rheumatology, School of Medicine, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland
| | - Ronan H Mullan
- Department of Rheumatology, Tallaght University Hospital, Dublin 24, Ireland
| | - Douglas J Veale
- St Vincent's University Hospital, Dublin Academic Health Care and University College Dublin, Dublin 4, Ireland
| | - Ursula Fearon
- Molecular Rheumatology, School of Medicine, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland
| |
Collapse
|
127
|
Alten R, Conaghan PG, Strand V, Sullivan E, Blackburn S, Tian H, Gandhi K, Jugl SM, Deodhar A. Unmet needs in psoriatic arthritis patients receiving immunomodulatory therapy: results from a large multinational real-world study. Clin Rheumatol 2019; 38:1615-1626. [DOI: 10.1007/s10067-019-04446-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Revised: 01/15/2019] [Accepted: 01/21/2019] [Indexed: 01/01/2023]
|
128
|
Glatigny S, Höllbacher B, Motley SJ, Tan C, Hundhausen C, Buckner JH, Smilek D, Khoury SJ, Ding L, Qin T, Pardo J, Nepom GT, Turka LA, Harris KM, Campbell DJ, Bettelli E. Abatacept Targets T Follicular Helper and Regulatory T Cells, Disrupting Molecular Pathways That Regulate Their Proliferation and Maintenance. THE JOURNAL OF IMMUNOLOGY 2019; 202:1373-1382. [PMID: 30683697 DOI: 10.4049/jimmunol.1801425] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/26/2018] [Accepted: 12/24/2018] [Indexed: 01/07/2023]
Abstract
Abatacept is a CTLA-4-Ig fusion protein that binds to the costimulatory ligands CD80 and CD86 and blocks their interaction with the CD28 and CTLA-4 receptors expressed by T cells, therefore inhibiting T cell activation and function. Abatacept has shown clinical efficacy in treating some autoimmune diseases but has failed to show clinical benefit in other autoimmune conditions. The reasons for these disparate results are not clear and warrant further investigation of abatacept's mode of action. Longitudinal specimens from the Immune Tolerance Network's A Cooperative Clinical Study of Abatacept in Multiple Sclerosis trial were used to examine the effects of abatacept treatment on the frequency and transcriptional profile of specific T cell populations in peripheral blood. We found that the relative abundance of CD4+ T follicular helper (Tfh) cells and regulatory T cells was selectively decreased in participants following abatacept treatment. Within both cell types, abatacept reduced the proportion of activated cells expressing CD38 and ICOS and was associated with decreased expression of genes that regulate cell-cycle and chromatin dynamics during cell proliferation, thereby linking changes in costimulatory signaling to impaired activation, proliferation, and decreased abundance. All cellular and molecular changes were reversed following termination of abatacept treatment. These data expand upon the mechanism of action of abatacept reported in other autoimmune diseases and identify new transcriptional targets of CD28-mediated costimulatory signaling in human regulatory T and Tfh cells, further informing on its potential use in diseases associated with dysregulated Tfh activity.
Collapse
Affiliation(s)
- Simon Glatigny
- Immunology Program, Benaroya Research Institute, Seattle, WA 98101.,Department of Immunology, University of Washington, Seattle, WA 98109
| | | | | | - Cathy Tan
- Immunology Program, Benaroya Research Institute, Seattle, WA 98101
| | | | - Jane H Buckner
- Immunology Program, Benaroya Research Institute, Seattle, WA 98101.,Department of Immunology, University of Washington, Seattle, WA 98109
| | - Dawn Smilek
- Immune Tolerance Network, University of California San Francisco, San Francisco, CA 94107
| | - Samia J Khoury
- Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115.,Abu Haidar Neuroscience Institute, American University of Beirut, Beirut 1107 2020, Lebanon
| | - Linna Ding
- National Institute of Allergy and Infectious Diseases, Division of Allergy, Immunology, and Transplantation, Rockville, MD 20852
| | - Tielin Qin
- Immune Tolerance Network, Bethesda, MD 20814; and
| | - Jorge Pardo
- Immune Tolerance Network, Bethesda, MD 20814; and
| | - Gerald T Nepom
- Immunology Program, Benaroya Research Institute, Seattle, WA 98101.,Immune Tolerance Network, Bethesda, MD 20814; and
| | - Laurence A Turka
- Immune Tolerance Network, Bethesda, MD 20814; and.,Center for Transplantation Sciences, Massachusetts General Hospital, Boston, MA 02129
| | | | - Daniel J Campbell
- Immunology Program, Benaroya Research Institute, Seattle, WA 98101; .,Department of Immunology, University of Washington, Seattle, WA 98109
| | - Estelle Bettelli
- Immunology Program, Benaroya Research Institute, Seattle, WA 98101; .,Department of Immunology, University of Washington, Seattle, WA 98109
| |
Collapse
|
129
|
Sin CZ, Wang TS, Chiu HY, Tsai TF. Human leukocyte antigen and demographic characteristics in Chinese patients with active peripheral type psoriatic arthritis who had inadequate response to conventional disease-modifying antirheumatic drugs in a single dermatologic clinic. PLoS One 2019; 14:e0210076. [PMID: 30650098 PMCID: PMC6334904 DOI: 10.1371/journal.pone.0210076] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2018] [Accepted: 11/10/2018] [Indexed: 11/19/2022] Open
Abstract
Background Correlation between severity of psoriasis and psoriatic arthritis (PsA) is inconsistent. Also, human leukocyte antigen (HLA)-Cw6 was found to be underrepresented in severe psoriasis who failed conventional systemic therapies, but the effect of HLA polymorphism on PsA severity needs to be confirmed. Objectives To describe the severity of psoriasis, demographic features and HLA polymorphism among Chinese patients with active peripheral type PsA who had inadequate response to conventional disease-modifying antirheumatic drugs. Methods We included all patients with PsA who had at least 3 tender and swollen peripheral joints despite at least two conventional non-biologic treatments in our clinic. Demographic results were compared with global pivotal studies of biologics for PsA. HLA-Cw and HLA-DRB1 genotyping was also analyzed. Results We identified 60 patients who met our inclusion criteria. The male to female ratio was 1.31:1. The majority of patients presented with psoriasis first (81.7%). The mean interval between psoriasis and PsA was 7.2 ± 8.1 years (mean ± SD). The baseline number of tender and swollen joints was 14.9 ± 10.7 and 11.3 ±10.2, respectively. In total, 41.7% subjects had more than 3% body surface area involvement of psoriasis. Genotyping of HLA-Cw and HLA-DRB1 was performed in 47 subjects. HLA-Cw*0702 was the most frequent allele (29.8%), followed by HLA-Cw*01 (26.6%). The frequency of HLA-Cw*0602 allele was similar to normal population. The most frequent HLA-DRB1 allele was HLA-DRB1*04 (20.2%), followed by HLA-DRB1*08 (16.0%). No cases carrying HLA-DRB1*13 were detected. Conclusions Compared with Western population, our patients had less psoriasis and PsA burden. The frequencies of HLA-Cw*06, HLA-Cw*12, and HLA-DRB1*07 were not increased. In contrast, HLA-Cw*0702 and HLA-DRB1*08 allele frequencies were increased compared with psoriasis patients and normal population in Taiwan. Future studies are still needed to characterize the demographic and genetic features of high need PsA patients.
Collapse
Affiliation(s)
- Chi-Zai Sin
- Department of Dermatology, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan
| | - Ting-Shun Wang
- Department of Dermatology, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan
- Division of Dermatology, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Hsien-Yi Chiu
- Department of Dermatology, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan
- Department of Dermatology, Hsin-Chu Branch, National Taiwan University Hospital, Hsin-Chu, Taiwan
| | - Tsen-Fang Tsai
- Department of Dermatology, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan
- * E-mail:
| |
Collapse
|
130
|
Characteristics of Patients with Psoriatic Arthritis Receiving Secukinumab and Reasons for Initiation: A US Retrospective Medical Chart Review. Rheumatol Ther 2019; 6:89-100. [PMID: 30612321 PMCID: PMC6393268 DOI: 10.1007/s40744-018-0137-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Indexed: 01/06/2023] Open
Abstract
Introduction Secukinumab is a fully human anti-interleukin 17A monoclonal antibody approved for the treatment of psoriatic arthritis (PsA) in the United States. Few studies have investigated prescribing patterns among rheumatologists who have initiated secukinumab for the treatment of patients with PsA in real-world settings. This US medical chart review describes clinical and treatment characteristics of patients with psoriatic arthritis (PsA) who were prescribed secukinumab and rheumatologist-reported reasons for prescribing secukinumab in clinical practice. Methods This US medical chart review included patients with physician-diagnosed PsA aged ≥ 18 years initiating secukinumab after January 15, 2016. Eligible rheumatologists used online forms to collect patient demographics, disease characteristics, comorbidity profiles, and treatment histories before or on the date of the first secukinumab prescription recorded in the medical chart. Information on reasons for secukinumab prescription and dosing was also collected. Results Medical charts from 153 patients with PsA who initiated secukinumab were reviewed by 46 rheumatologists between July 7, 2017, and August 11, 2017. Overall, 53.6% of patients were male, mean (standard deviation) age was 47.3 (11.5) years, and 24.8% were biologic naive. The most common reasons for secukinumab prescription among biologic-naive and biologic-experienced patients, respectively, were efficacy/effectiveness of secukinumab (84.2%) and failure of other prior biologics (80.9%). Nearly all patients (94.1%) received a loading regimen, including 150 mg every week (32.7%) and 300 mg every week (61.4%). Overall, 145 patients (94.8%) received ≥ 1 maintenance dose, of whom 49.7% received 150 mg every 4 weeks and 50.3% received 300 mg every 4 weeks. Conclusions At the time of the chart review, most patients with PsA who initiated secukinumab were biologic experienced, although one-quarter received secukinumab as first-line biologic therapy. Efficacy/effectiveness of secukinumab and failure of other biologics were the most common reasons for initiating secukinumab. Funding Novartis Pharmaceuticals Corporation, East Hanover, NJ. Plain Language Summary Plain language summary available for this article.
Collapse
|
131
|
A Review for Physician Assistants and Nurse Practitioners on the Considerations for Diagnosing and Treating Psoriatic Arthritis. Rheumatol Ther 2018; 6:5-21. [PMID: 30570707 PMCID: PMC6393269 DOI: 10.1007/s40744-018-0133-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2018] [Indexed: 02/08/2023] Open
Abstract
Psoriatic arthritis (PsA) is a clinically heterogeneous form of progressive inflammatory arthritis that affects up to 30% of patients with psoriasis. The rapid rate of progression associated with PsA makes early-disease diagnosis and treatment crucial to patients' quality of life and long-term health. With the aim of providing clinical guidance to physician assistants and nurse practitioners, this article gives an overview of the different PsA clinical domains, including peripheral arthritis, axial disease, enthesitis, dactylitis, skin disease, and nail dystrophy, which should be considered as part of diagnosis and treatment strategies. The efficacy of different therapies across these PsA domains is reviewed within the context of current PsA treatment guidelines while considering more recent data on newly approved therapies for PsA.Funding: Novartis Pharmaceuticals Corp., East Hanover, NJ, USA.
Collapse
|
132
|
Strand V, Alemao E, Lehman T, Johnsen A, Banerjee S, Ahmad HA, Mease PJ. Improved patient-reported outcomes in patients with psoriatic arthritis treated with abatacept: results from a phase 3 trial. Arthritis Res Ther 2018; 20:269. [PMID: 30522501 PMCID: PMC6282264 DOI: 10.1186/s13075-018-1769-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Accepted: 11/14/2018] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND To explore the effect of abatacept treatment on patient-reported outcomes (PROs) in psoriatic arthritis (PsA). METHODS Patients with PsA were randomised (1:1) to subcutaneous abatacept 125 mg weekly/placebo for 24 weeks with early escape (EE) to open-label abatacept (week 16). Adjusted mean changes from baseline to weeks 16 (all patients) and 24 (non-EE responders) in Health Assessment Questionnaire-Disability Index (HAQ-DI), Short Form-36 (SF-36; physical and mental component summary and domains), Dermatology Life Quality Index and Functional Assessment of Chronic Illness Therapy-Fatigue (FACIT-F) were evaluated. Subpopulations were analysed by baseline C-reactive protein (CRP) level (> vs ≤ upper limit of normal [ULN]) and prior tumour necrosis factor inhibitor (TNFi) exposure. Proportions of patients reporting improvements ≥ minimal clinically important differences (MCIDs) and ≥ normative values (NVs) in HAQ-DI, SF-36 and FACIT-F (week 16 before EE) were analysed. RESULTS In total population, numerically higher improvements in most PROs were reported with abatacept (n = 213) versus placebo (n = 211) at both time points (P > 0.05). Higher proportions of abatacept versus placebo patients reported PRO improvements ≥ MCID and ≥ NV at week 16. At week 16, all PRO improvements were numerically greater (P > 0.05) in patients with baseline CRP > ULN versus CRP ≤ ULN (all significant [95% confidence interval] for abatacept vs placebo); improvements in SF-36 component summaries and FACIT-F were greater in TNFi-naïve versus TNFi-exposed patients (abatacept > placebo). Week 24 subgroup data were difficult to interpret due to low patient numbers. CONCLUSIONS Abatacept treatment improved PROs in patients with PsA versus placebo, with better results in elevated baseline CRP and TNFi-naïve subpopulations. TRIAL REGISTRATION ClinicalTrials.gov number, NCT01860976 (funded by Bristol-Myers Squibb); date of registration: 23 May 2013.
Collapse
Affiliation(s)
- Vibeke Strand
- Division of Immunology/Rheumatology, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Evo Alemao
- Bristol-Myers Squibb, Princeton, NJ, USA.
| | | | | | | | | | - Philip J Mease
- Swedish Medical Center and University of Washington, Seattle, WA, USA
| |
Collapse
|
133
|
Abstract
Psoriatic arthritis (PsA) is an inflammatory arthritis that is estimated to affect approximately 30% of patients with psoriasis. Enthesitis and dactylitis, two hallmarks of PsA, are associated with radiographic peripheral/axial joint damage and severe disease. Clinical symptoms of enthesitis include tenderness, soreness, and pain at entheses on palpation, whereas dactylitis is recognized by swelling of an entire digit that is different from adjacent digits. Both ultrasound and magnetic resonance imaging can be used to diagnose enthesitis and dactylitis, especially in patients in whom symptoms may be difficult to discern. Delayed treatment of PsA can result in irreversible joint damage and reduced quality of life. Thus, it is recommended that dermatologists monitor patients with psoriasis for these two early and important manifestations of PsA.
Collapse
Affiliation(s)
- Jerry Bagel
- Psoriasis Treatment Center of Central New Jersey, 59 One Mile Road Ext. Suite G, East Windsor, NJ, 08520, USA.
| | | |
Collapse
|
134
|
Mease P, Strand V, Gladman D. Functional impairment measurement in psoriatic arthritis: Importance and challenges. Semin Arthritis Rheum 2018; 48:436-448. [DOI: 10.1016/j.semarthrit.2018.05.010] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2017] [Revised: 04/27/2018] [Accepted: 05/21/2018] [Indexed: 12/21/2022]
|
135
|
Singh JA, Guyatt G, Ogdie A, Gladman DD, Deal C, Deodhar A, Dubreuil M, Dunham J, Husni ME, Kenny S, Kwan-Morley J, Lin J, Marchetta P, Mease PJ, Merola JF, Miner J, Ritchlin CT, Siaton B, Smith BJ, Van Voorhees AS, Jonsson AH, Shah AA, Sullivan N, Turgunbaev M, Coates LC, Gottlieb A, Magrey M, Nowell WB, Orbai AM, Reddy SM, Scher JU, Siegel E, Siegel M, Walsh JA, Turner AS, Reston J. Special Article: 2018 American College of Rheumatology/National Psoriasis Foundation Guideline for the Treatment of Psoriatic Arthritis. Arthritis Rheumatol 2018; 71:5-32. [PMID: 30499246 DOI: 10.1002/art.40726] [Citation(s) in RCA: 309] [Impact Index Per Article: 44.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2017] [Accepted: 09/11/2018] [Indexed: 12/13/2022]
Abstract
OBJECTIVE To develop an evidence-based guideline for the pharmacologic and nonpharmacologic treatment of psoriatic arthritis (PsA), as a collaboration between the American College of Rheumatology (ACR) and the National Psoriasis Foundation (NPF). METHODS We identified critical outcomes in PsA and clinically relevant PICO (population/intervention/comparator/outcomes) questions. A Literature Review Team performed a systematic literature review to summarize evidence supporting the benefits and harms of available pharmacologic and nonpharmacologic therapies for PsA. GRADE (Grading of Recommendations Assessment, Development and Evaluation) methodology was used to rate the quality of the evidence. A voting panel, including rheumatologists, dermatologists, other health professionals, and patients, achieved consensus on the direction and the strength of the recommendations. RESULTS The guideline covers the management of active PsA in patients who are treatment-naive and those who continue to have active PsA despite treatment, and addresses the use of oral small molecules, tumor necrosis factor inhibitors, interleukin-12/23 inhibitors (IL-12/23i), IL-17 inhibitors, CTLA4-Ig (abatacept), and a JAK inhibitor (tofacitinib). We also developed recommendations for psoriatic spondylitis, predominant enthesitis, and treatment in the presence of concomitant inflammatory bowel disease, diabetes, or serious infections. We formulated recommendations for a treat-to-target strategy, vaccinations, and nonpharmacologic therapies. Six percent of the recommendations were strong and 94% conditional, indicating the importance of active discussion between the health care provider and the patient to choose the optimal treatment. CONCLUSION The 2018 ACR/NPF PsA guideline serves as a tool for health care providers and patients in the selection of appropriate therapy in common clinical scenarios. Best treatment decisions consider each individual patient situation. The guideline is not meant to be proscriptive and should not be used to limit treatment options for patients with PsA.
Collapse
Affiliation(s)
- Jasvinder A Singh
- University of Alabama at Birmingham and Birmingham Veterans Affairs Medical Center, Birmingham, Alabama
| | | | | | - Dafna D Gladman
- University of Toronto and Toronto Western Hospital, Toronto, Ontario, Canada
| | | | | | | | | | | | | | | | - Janice Lin
- Stanford University, Stanford, California
| | | | - Philip J Mease
- Swedish-Providence Health Systems and University of Washington, Seattle, Washington
| | - Joseph F Merola
- Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Julie Miner
- Comprehensive Therapy Consultants and Therapy Steps, Roswell, Georgia
| | | | | | - Benjamin J Smith
- Florida State University College of Medicine School of Physician Assistant Practice, Tallahassee
| | | | - Anna Helena Jonsson
- Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | | | | | | | | | - Alice Gottlieb
- New York Medical College at Metropolitan Hospital, New York, New York
| | | | | | | | - Soumya M Reddy
- New York University School of Medicine, New York, New York
| | - Jose U Scher
- New York University School of Medicine, New York, New York
| | - Evan Siegel
- Arthritis & Rheumatism Associates, Rockville, Maryland
| | | | - Jessica A Walsh
- University of Utah and George E. Wahlen VeteranS Affairs Medical Center, Salt Lake City, Utah
| | - Amy S Turner
- American College of Rheumatology, Atlanta, Georgia
| | | |
Collapse
|
136
|
Singh JA, Guyatt G, Ogdie A, Gladman DD, Deal C, Deodhar A, Dubreuil M, Dunham J, Husni ME, Kenny S, Kwan-Morley J, Lin J, Marchetta P, Mease PJ, Merola JF, Miner J, Ritchlin CT, Siaton B, Smith BJ, Van Voorhees AS, Jonsson AH, Shah AA, Sullivan N, Turgunbaev M, Coates LC, Gottlieb A, Magrey M, Nowell WB, Orbai AM, Reddy SM, Scher JU, Siegel E, Siegel M, Walsh JA, Turner AS, Reston J. 2018 American College of Rheumatology/National Psoriasis Foundation Guideline for the Treatment of Psoriatic Arthritis. ACTA ACUST UNITED AC 2018. [DOI: 10.1177/2475530318812244] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Objective: To develop an evidence-based guideline for the pharmacologic and nonpharmacologic treatment of psoriatic arthritis (PsA), as a collaboration between the American College of Rheumatology (ACR) and the National Psoriasis Foundation (NPF). Methods: We identified critical outcomes in PsA and clinically relevant PICO (population/intervention/comparator/outcomes) questions. A Literature Review Team performed a systematic literature review to summarize evidence supporting the benefits and harms of available pharmacologic and nonpharmacologic therapies for PsA. GRADE (Grading of Recommendations Assessment, Development and Evaluation) methodology was used to rate the quality of the evidence. A voting panel, including rheumatologists, dermatologists, other health professionals, and patients, achieved consensus on the direction and the strength of the recommendations. Results: The guideline covers the management of active PsA in patients who are treatment-naive and those who continue to have active PsA despite treatment, and addresses the use of oral small molecules, tumor necrosis factor inhibitors, interleukin-12/23 inhibitors (IL-12/23i), IL-17 inhibitors, CTLA4-Ig (abatacept), and a JAK inhibitor (tofacitinib). We also developed recommendations for psoriatic spondylitis, predominant enthesitis, and treatment in the presence of concomitant inflammatory bowel disease, diabetes, or serious infections. We formulated recommendations for a treat-to-target strategy, vaccinations, and nonpharmacologic therapies. Six percent of the recommendations were strong and 94% conditional, indicating the importance of active discussion between the health care provider and the patient to choose the optimal treatment. Conclusion: The 2018 ACR/NPF PsA guideline serves as a tool for health care providers and patients in the selection of appropriate therapy in common clinical scenarios. Best treatment decisions consider each individual patient situation. The guideline is not meant to be proscriptive and should not be used to limit treatment options for patients with PsA.
Collapse
Affiliation(s)
- Jasvinder A. Singh
- University of Alabama at Birmingham and Birmingham Veterans Affairs Medical Center, Birmingham, Alabama
| | | | - Alexis Ogdie
- University of Pennsylvania, Philadelphia, PA, USA
| | - Dafna D. Gladman
- University of Toronto and Toronto Western Hospital, Toronto, Ontario, Canada
| | - Chad Deal
- Cleveland Clinic, Cleveland, OH, USA
| | - Atul Deodhar
- Oregon Health & Science University, Portland, OR, USA
| | | | | | | | | | | | | | | | - Philip J. Mease
- Swedish-Providence Health Systems and University of Washington, Seattle, WA, USA
| | - Joseph F. Merola
- Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, USA
| | - Julie Miner
- Comprehensive Therapy Consultants and Therapy Steps, Roswell, GA, USA
| | | | | | - Benjamin J. Smith
- Florida State University College of Medicine School of Physician Assistant Practice, Tallahassee, FL, USA
| | | | | | | | | | | | | | - Alice Gottlieb
- New York Medical College at Metropolitan Hospital, New York, NY, USA
| | | | | | | | | | - Jose U. Scher
- New York University School of Medicine, New York, NY, USA
| | - Evan Siegel
- Arthritis & Rheumatism Associates, Rockville, MA, USA
| | | | - Jessica A. Walsh
- University of Utah and George E. Wahlen Veterans Affairs Medical Center, Salt Lake City, UT, USA
| | | | | |
Collapse
|
137
|
Singh JA, Guyatt G, Ogdie A, Gladman DD, Deal C, Deodhar A, Dubreuil M, Dunham J, Husni ME, Kenny S, Kwan-Morley J, Lin J, Marchetta P, Mease PJ, Merola JF, Miner J, Ritchlin CT, Siaton B, Smith BJ, Van Voorhees AS, Jonsson AH, Shah AA, Sullivan N, Turgunbaev M, Coates LC, Gottlieb A, Magrey M, Nowell WB, Orbai AM, Reddy SM, Scher JU, Siegel E, Siegel M, Walsh JA, Turner AS, Reston J. Special Article: 2018 American College of Rheumatology/National Psoriasis Foundation Guideline for the Treatment of Psoriatic Arthritis. Arthritis Care Res (Hoboken) 2018; 71:2-29. [PMID: 30499259 DOI: 10.1002/acr.23789] [Citation(s) in RCA: 168] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2017] [Accepted: 09/11/2018] [Indexed: 12/13/2022]
Abstract
OBJECTIVE To develop an evidence-based guideline for the pharmacologic and nonpharmacologic treatment of psoriatic arthritis (PsA), as a collaboration between the American College of Rheumatology (ACR) and the National Psoriasis Foundation (NPF). METHODS We identified critical outcomes in PsA and clinically relevant PICO (population/intervention/comparator/outcomes) questions. A Literature Review Team performed a systematic literature review to summarize evidence supporting the benefits and harms of available pharmacologic and nonpharmacologic therapies for PsA. GRADE (Grading of Recommendations Assessment, Development and Evaluation) methodology was used to rate the quality of the evidence. A voting panel, including rheumatologists, dermatologists, other health professionals, and patients, achieved consensus on the direction and the strength of the recommendations. RESULTS The guideline covers the management of active PsA in patients who are treatment-naive and those who continue to have active PsA despite treatment, and addresses the use of oral small molecules, tumor necrosis factor inhibitors, interleukin-12/23 inhibitors (IL-12/23i), IL-17 inhibitors, CTLA4-Ig (abatacept), and a JAK inhibitor (tofacitinib). We also developed recommendations for psoriatic spondylitis, predominant enthesitis, and treatment in the presence of concomitant inflammatory bowel disease, diabetes, or serious infections. We formulated recommendations for a treat-to-target strategy, vaccinations, and nonpharmacologic therapies. Six percent of the recommendations were strong and 94% conditional, indicating the importance of active discussion between the health care provider and the patient to choose the optimal treatment. CONCLUSION The 2018 ACR/NPF PsA guideline serves as a tool for health care providers and patients in the selection of appropriate therapy in common clinical scenarios. Best treatment decisions consider each individual patient situation. The guideline is not meant to be proscriptive and should not be used to limit treatment options for patients with PsA.
Collapse
Affiliation(s)
- Jasvinder A Singh
- University of Alabama at Birmingham and Birmingham Veterans Affairs Medical Center, Birmingham, Alabama
| | | | | | - Dafna D Gladman
- University of Toronto and Toronto Western Hospital, Toronto, Ontario, Canada
| | | | | | | | | | | | | | | | - Janice Lin
- Stanford University, Stanford, California
| | | | - Philip J Mease
- Swedish-Providence Health Systems and University of Washington, Seattle, Washington
| | - Joseph F Merola
- Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Julie Miner
- Comprehensive Therapy Consultants and Therapy Steps, Roswell, Georgia
| | | | | | - Benjamin J Smith
- Florida State University College of Medicine School of Physician Assistant Practice, Tallahassee
| | | | - Anna Helena Jonsson
- Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | | | | | | | | | - Alice Gottlieb
- New York Medical College at Metropolitan Hospital, New York, New York
| | | | | | | | - Soumya M Reddy
- New York University School of Medicine, New York, New York
| | - Jose U Scher
- New York University School of Medicine, New York, New York
| | - Evan Siegel
- Arthritis & Rheumatism Associates, Rockville, Maryland
| | | | - Jessica A Walsh
- University of Utah and George E. Wahlen Veterans Affairs Medical Center, Salt Lake City, Utah
| | - Amy S Turner
- American College of Rheumatology, Atlanta, Georgia
| | | |
Collapse
|
138
|
Li SJ, Perez-Chada LM, Merola JF. TNF Inhibitor-Induced Psoriasis: Proposed Algorithm for Treatment and Management. ACTA ACUST UNITED AC 2018; 4:70-80. [PMID: 31093599 PMCID: PMC6513344 DOI: 10.1177/2475530318810851] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Tumor necrosis factor a (TNF-α)-targeted therapies have expanded the therapeutic options for patients with inflammatory bowel disease (IBD), rheumatoid arthritis (RA), psoriasis, and psoriatic arthritis (PsA) and have significantly improved patients' quality of life. Paradoxically, anti-TNF-α agents may induce psoriatic eruptions or worsen preexisting psoriatic skin disease. Currently, there is no standard approach for the management of TNF inhibitor-induced psoriasis. Here, we conduct a literature review on TNF inhibitor-induced psoriasis and introduce a novel treatment algorithm for maintaining otherwise effective anti-TNF therapy versus switching to a different class as appropriate in the management of patients with IBD, RA, psoriasis, or PsA.
Collapse
Affiliation(s)
- Sara Jiayang Li
- Department of Dermatology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Lourdes M Perez-Chada
- Department of Dermatology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Joseph F Merola
- Department of Dermatology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
139
|
Efficacy of Tofacitinib for the Treatment of Psoriatic Arthritis: Pooled Analysis of Two Phase 3 Studies. Rheumatol Ther 2018; 5:567-582. [PMID: 30414064 PMCID: PMC6251851 DOI: 10.1007/s40744-018-0131-5] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2018] [Indexed: 01/06/2023] Open
Abstract
Introduction Tofacitinib is an oral Janus kinase inhibitor for the treatment of psoriatic arthritis (PsA). This post hoc analysis assessed the efficacy of tofacitinib using pooled data from two phase 3 studies of patients with active PsA. Methods Data were pooled from OPAL Broaden (NCT01877668) and OPAL Beyond (NCT01882439). Patients had active PsA and either an inadequate response (IR) to ≥ 1 conventional synthetic disease-modifying antirheumatic drug (csDMARD) and were tumor necrosis factor inhibitor (TNFi)-naïve (OPAL Broaden), or had IR to ≥ 1 TNFi (OPAL Beyond). Pooled data included tofacitinib 5 or 10 mg twice daily (BID; to month 6) and placebo (to month 3; patients then switched to tofacitinib 5 or 10 mg BID). Patients also received one background csDMARD. Endpoints included American College of Rheumatology (ACR)20 response and change from baseline in Health Assessment Questionnaire-Disability Index (HAQ-DI) at month 3 (primary endpoints), ACR50/70 response, HAQ-DI response (decrease from baseline ≥ 0.35) and improvements in painful and swollen joint counts, psoriasis, enthesitis and dactylitis to month 6. Results A total of 710 patients were included (tofacitinib 5 mg BID: 238; tofacitinib 10 mg BID: 236; placebo: 236). Primary endpoints showed significant improvements at month 3 in patients receiving tofacitinib 5 or 10 mg BID vs. placebo. Significant improvements in HAQ-DI response, painful and swollen joints, psoriasis, enthesitis and dactylitis vs. placebo were observed for both tofacitinib doses at month 3. Efficacy was maintained to month 6 (final pooled time point). Conclusions In a pooled analysis of csDMARD-IR/TNFi-naïve and TNFi-IR patients, tofacitinib was superior to placebo at month 3 across four PsA domains: peripheral arthritis, psoriasis, enthesitis and dactylitis. Trial Registration OPAL Broaden (NCT01877668); OPAL Beyond (NCT01882439). Funding Pfizer Inc. Electronic supplementary material The online version of this article (10.1007/s40744-018-0131-5) contains supplementary material, which is available to authorized users.
Collapse
|
140
|
Gottlieb AB, Strand V, Kishimoto M, Mease P, Thaçi D, Birt J, Lee CH, Shuler CL, Lin CY, Gladman DD. Ixekizumab improves patient-reported outcomes up to 52 weeks in bDMARD-naïve patients with active psoriatic arthritis (SPIRIT-P1). Rheumatology (Oxford) 2018; 57:1777-1788. [PMID: 29945203 PMCID: PMC6152421 DOI: 10.1093/rheumatology/key161] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Indexed: 01/13/2023] Open
Abstract
Objective To report patient-reported outcomes of patients with PsA treated with ixekizumab up to 52 weeks. Methods In SPIRIT-P1, biologic-naïve patients with active PsA were randomized to ixekizumab 80 mg every 4 weeks (IXEQ4W; N = 107) or every 2 weeks (IXEQ2W; N = 103) following a 160 mg starting dose, adalimumab 40 mg every 2 weeks (ADA; N = 101) or placebo (PBO; N = 106) during the initial 24-week double-blind treatment period. At week 24 (week 16 for inadequate responders), ADA (8-week washout before starting ixekizumab) and PBO patients were re-randomized to IXEQ2W or IXEQ4W. Patients receiving ixekizumab at week 24 received the same dose during the extension period (EP) to week 52. Patients completed measures including the Dermatology Life Quality Index (DLQI), Itch Numeric Rating Scale, 36-Item Short Form Health Survey version 2, European Quality of Life 5 Dimensions Visual Analogue Scale and Work Productivity and Activity Impairment Questionnaire-Specific Health Problem. Results The IXEQ4W, IXEQ2W and ADA groups reported significant improvements in DLQI at week 24; 22% (PBO), 53% (IXEQ4W), 63% (IXEQ2W) and 54% (ADA) of patients reported DLQI scores of 0/1. The IXEQ4W, IXEQ2W and ADA groups reported significant improvements in Itch Numeric Rating Scale, 36-Item Short Form Health Survey version 2 physical component summary and some domain scores, and European Quality of Life 5 Dimensions Visual Analogue Scale at weeks 12 and 24; and in three of four Work Productivity and Activity Impairment Questionnaire-Specific Health Problem domains at week 24. Results are also presented through week 52 for the EP. Conclusion In biologic-naïve patients with active PsA, ixekizumab significantly improved skin symptoms, health-related quality of life and work productivity. Trial Registration ClinicalTrials.gov, http://clinicaltrials.gov, NCT01695239; EU Clinical Trials Register, https://www.clinicaltrialsregister.eu, EudraCT2011-002326-49
Collapse
Affiliation(s)
- Alice B Gottlieb
- Department of Dermatology, New York Medical College, Metropolitan Hospital, New York, NY
| | - Vibeke Strand
- Division of Immunology/Rheumatology, Stanford University, Palo Alto, CA, USA
| | | | - Philip Mease
- Department of Rheumatology, Swedish Medical Centre, Seattle, WA, USA.,Division of Rheumatology, University of Washington, Seattle, WA, USA
| | - Diamant Thaçi
- Comprehensive Centre for Inflammation Medicine, University Hospital Schleswig-Holstein, Campus Lübeck, Lübeck, Germany
| | - Julie Birt
- Eli Lilly and Company, Indianapolis, IN, USA
| | - Chin H Lee
- Eli Lilly and Company, Indianapolis, IN, USA
| | | | | | - Dafna D Gladman
- Division of Rheumatology, Department of Medicine, University of Toronto, Krembil Research Institute, Toronto Western Hospital, Toronto, Ontario, Canada
| |
Collapse
|
141
|
Walsh JA, Adejoro O, Chastek B, Palmer JB, Hur P. Treatment Patterns Among Patients with Psoriatic Arthritis Treated with a Biologic in the United States: Descriptive Analyses from an Administrative Claims Database. J Manag Care Spec Pharm 2018; 24:623-631. [PMID: 29952704 PMCID: PMC10397599 DOI: 10.18553/jmcp.2018.24.7.623] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
BACKGROUND In patients with psoriatic arthritis (PsA), limited data exist regarding patterns of biologic therapy use. OBJECTIVE To examine treatment patterns and therapy modifications in U.S. patients with PsA receiving a tumor necrosis factor inhibitor (TNFi) or an anti-interleukin (IL)-12/23 inhibitor. METHODS Adults with PsA who newly initiated a biologic therapy (index biologic) between January 1, 2013, and January 31, 2015, were included from the Optum Research Database. Biologic therapies comprised those that were approved by the FDA for the treatment of PsA at the time of the study initiation (adalimumab, certolizumab pegol, etanercept, golimumab, infliximab, or ustekinumab). Outcomes included adherence, persistence, and discontinuation of the index biologic; initiation of adjunctive medications (nonbiologics, including those commonly used for pain and/or inflammation); and dose escalation of the index biologic during the 12-month follow-up period. RESULTS Of the 1,235 patients included, 52.5% were female, and mean (SD) age was 50.3 (12.1) years. The mean (SD) duration of persistence with a newly initiated index biologic (etanercept [48.1%], adalimumab [24.0%], infliximab [10.4%], golimumab [8.3%], ustekinumab [7.2%], or certolizumab pegol [2.0%]) was 246 (128) days; 44.5% of patients persisted with the index biologic for ≥ 12 months. During the 12-month follow-up period, 22.9% of patients switched to a different biologic, 26.8% discontinued without switching or restarting, and 5.8% discontinued and restarted the index biologic. Of the 1,010 patients who persisted with the index biologic for > 90 days, 45.6% received ≥ 1 adjunctive medication during the period from 90 days after the index date to the end of persistence or 12 months. The most commonly initiated adjunctive medications were corticosteroids (22.0%), opioids (17.1%), and nonsteroidal anti-inflammatory drugs (12.9%). Overall, 9.6% of patients had a dose escalation of the index biologic in the immediate 12-month post-index period. CONCLUSIONS This real-world study of treatment patterns for PsA, which used a large U.S. claims database, demonstrated that the majority of patients with PsA discontinued their index biologic (TNFi or anti-IL-12/23 inhibitor) before 12 months. Nearly half of patients initiated an adjunctive medication, many of which were pain and conventional anti-inflammatory medications. DISCLOSURES This study was sponsored by Novartis Pharmaceuticals. Optum was commissioned by Novartis to conduct this study, but employment was not contingent on results of the study. Walsh is a paid consultant for Novartis. Adejoro was an employee of Optum at the time of the study and writing of the manuscript. Chastek is an employee of Optum. Palmer and Hur are employees of Novartis. Results of this study were presented as an abstract and poster at the Academy of Managed Care Pharmacy Nexus 2017; October 16-19, 2017; Dallas, TX; and the EULAR 2017 Annual European Congress of Rheumatology; June 14-17, 2017; Madrid, Spain.
Collapse
Affiliation(s)
- Jessica A Walsh
- 1 University of Utah School of Medicine and Salt Lake City Veterans Affairs Medical Center, Salt Lake City, Utah
| | | | | | | | - Peter Hur
- 3 Novartis Pharmaceuticals, East Hanover, New Jersey
| |
Collapse
|
142
|
Efficacy of new treatments for dactylitis of psoriatic arthritis: update of literature review. Clin Rheumatol 2018; 38:591-596. [PMID: 30328022 DOI: 10.1007/s10067-018-4328-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2018] [Revised: 09/14/2018] [Accepted: 10/08/2018] [Indexed: 01/05/2023]
Abstract
Dactylitis is a frequent disabling feature of psoriatic arthritis (PsA). Therapeutic strategy on dactylitis is not really codified. We performed a complementary literature review (since a previous one in 2014) of efficacy of new treatments recently used in PsA on this specific clinical manifestation. Eleven publications were retained (4697 patients). In the randomized double-blind placebo-controlled trials analyzed, authors declared ustekinumab, ixekizumab, adalimumab, and apremilast efficient. Secukinumab, clazakizumab, abatacept, and tofacitinib were promising. Brodalumab was ineffective. Calculations of odds ratios for residual dactylitis were significant for clazakizumab 100 mg and secukinumab in anti-TNF-naïve population. Homogenization of dactylitis assessment and use of this criterion as primary outcome are necessary to have better data on treatment efficacy in the future.
Collapse
|
143
|
Lubrano E, Scriffignano S, Perrotta FM. Abatacept for the treatment of psoriatic arthritis. Expert Rev Clin Immunol 2018; 14:899-905. [DOI: 10.1080/1744666x.2018.1527218] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Affiliation(s)
- Ennio Lubrano
- Academic Rheumatology Unit, Dipartimento di Medicina e Scienze della Salute “Vincenzo Tiberio”, Università degli Studi del Molise, Campobasso, Italy
| | - Silvia Scriffignano
- Academic Rheumatology Unit, Dipartimento di Medicina e Scienze della Salute “Vincenzo Tiberio”, Università degli Studi del Molise, Campobasso, Italy
| | - Fabio Massimo Perrotta
- Academic Rheumatology Unit, Dipartimento di Medicina e Scienze della Salute “Vincenzo Tiberio”, Università degli Studi del Molise, Campobasso, Italy
| |
Collapse
|
144
|
Dactylitis: A hallmark of psoriatic arthritis. Semin Arthritis Rheum 2018; 48:263-273. [DOI: 10.1016/j.semarthrit.2018.02.002] [Citation(s) in RCA: 69] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2017] [Revised: 01/26/2018] [Accepted: 02/12/2018] [Indexed: 01/07/2023]
|
145
|
Aggarwal D, Arumalla N, Jethwa H, Abraham S. The use of biomarkers as a tool for novel psoriatic disease drug discovery. Expert Opin Drug Discov 2018; 13:875-887. [PMID: 30124339 DOI: 10.1080/17460441.2018.1508206] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
INTRODUCTION Psoriatic disease is a relatively new term which encompasses psoriatic arthritis, psoriasis, and associated comorbidities. In this heterogeneous condition, the study of biomarkers is necessary to direct best therapy. Resulting in significant disability and socioeconomic burden, recent recommendations stress the need for tight control in psoriatic disease. Areas covered: The authors outline recent advances in the understanding of psoriatic disease pathogenesis which has highlighted multiple biomarkers that have been pursued as drug targets with varying degrees of success. Current drugs targeting biomarkers and therapies in development are evaluated. The methods of biomarker discovery through genomics, transcriptomics, proteomics, metabolomics, and study of the microbiome are also discussed. Expert opinion: Targeting biomarkers for therapeutic benefit appears to a promising field with multiple success stories, notably those associated with signaling through T-helper-17 cells. The use of genomics, transcriptomics, proteomics, and more recently metabolomics will help individualize targeted biomarker therapies, assist in monitoring therapeutic success, and ultimately yield novel therapeutic targets. Advances in the development of novel biologic molecules targeting more than one cytokine may offer additional gains in therapeutic response.
Collapse
Affiliation(s)
- Dinesh Aggarwal
- a Department of Infectious Diseases , Chelsea and Westminster Hospital , London , UK
| | | | - Hannah Jethwa
- c Department of Rheumatology , Ealing Hospital , Southall , UK
| | - Sonya Abraham
- d Department of Rheumatology , Hammersmith Hospital , London , UK.,e Department of Rheumatology , Imperial College Healthcare NHS Trust , London , UK
| |
Collapse
|
146
|
Targeted drugs in spondyloarthritis during pregnancy and lactation. Pharmacol Res 2018; 136:21-28. [PMID: 30125669 DOI: 10.1016/j.phrs.2018.08.009] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2018] [Revised: 07/29/2018] [Accepted: 08/13/2018] [Indexed: 12/16/2022]
Abstract
Spondyloarthritis (SpA) are a heterogeneous group of chronic inflammatory joint diseases that includes several clinical subgroups. SpA can affect women in the reproductive stage so pregnancy can influence the course of the disease and SpA can affect the maternal-fetal outcome. The treatment of SpA has changed dramatically in recent years and the use of targeted drugs is part of therapeutic armamentarium. The use of targeted drugs during pregnancy is controversial because the information available on safety during this period is still limited. Several cytokines have an important role in the normal development of pregnancy or other cytokines may play a role in certain maternal-fetal complications. Potentially targeted drugs can affect the function of these cytokines during pregnancy. The aim of this study is to review the interrelationship between SpA during pregnancy and lactation, the role of some cytokines during normal pregnancy and the development of maternal-fetal complications as well as to review recent information on targeted drugs during pregnancy and breastfeeding in these patients in order to maximize their use in these critical periods of life.
Collapse
|
147
|
The History of Psoriatic Arthritis (PsA): From Moll and Wright to Pathway-Specific Therapy. Curr Rheumatol Rep 2018; 20:58. [DOI: 10.1007/s11926-018-0771-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
|
148
|
Noisette A, Hochberg MC. Abatacept for the treatment of adults with psoriatic arthritis: patient selection and perspectives. PSORIASIS-TARGETS AND THERAPY 2018; 8:31-39. [PMID: 30035098 PMCID: PMC6047621 DOI: 10.2147/ptt.s146076] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Psoriatic arthritis (PsA) is a heterogeneous disease with several clinical subtypes including peripheral arthritis, dactylitis, enthesitis, nail disease, and axial arthritis. Nonsteroidal anti-inflammatory drugs, glucocorticoids, and conventional disease-modifying agents are used as first line in the treatment of active PsA. For moderate-to-severe PsA failing conventional therapy, antitumor necrosis factor inhibitors have historically been the drugs of choice. In recent years, novel interleukin-23/interleukin-17 pathway targets such as ustekinumab and secukinumab, and phosphodiesterase-4 inhibitor apremilast have been approved for use in the United States and Europe. Two sets of recommendations for the management of PsA were published in 2016 with consideration for these newer therapies. Since then, the results from a Phase III randomized controlled trial demonstrated that abatacept has efficacy in the treatment of PsA. Abatacept, a cytotoxic-T-lymphocyte-associated antigen 4 (CTLA-4)–Ig human fusion protein, acts to prevent naïve T-cell activation through the inhibition of the critical CD28 co-stimulatory signal. In the 2017 Active Psoriatic Arthritis Randomized Trial (ASTRAEA), 424 participants were randomized 1:1 to receive subcutaneous abatacept 125 mg weekly versus placebo. At week 24, 39.4% of those who received abatacept achieved a minimum of 20% improvement in the American College of Rheumatology (ACR) response compared to 22.3% in the placebo arm, a statistically significant finding (P<0.001). The 2011 Phase II study published by Mease et al demonstrated statistically significant improvements in the ACR20 response by week 169 in participants treated with intravenous abatacept 10 mg/kg (48%) and 30/10 mg/kg (42%) when compared with placebo (19%). This article reviews the data supporting the efficacy of abatacept in the management of PsA and attempts to place this agent in the context of other biologic disease-modifying antirheumatic drugs and targeted small molecules used in the treatment of patients with PsA.
Collapse
Affiliation(s)
- Ashley Noisette
- Division of Rheumatology and Clinical Immunology, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, USA,
| | - Marc C Hochberg
- Division of Rheumatology and Clinical Immunology, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, USA,
| |
Collapse
|
149
|
Evolution of clinical trials for rheumatoid arthritis and spondyloarthritis. Curr Opin Rheumatol 2018; 30:340-346. [DOI: 10.1097/bor.0000000000000516] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
|
150
|
Ursini F, Russo E, De Giorgio R, De Sarro G, D'Angelo S. Current treatment options for psoriatic arthritis: spotlight on abatacept. Ther Clin Risk Manag 2018; 14:1053-1059. [PMID: 29922065 PMCID: PMC5995419 DOI: 10.2147/tcrm.s148586] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Psoriatic arthritis (PsA) is a chronic inflammatory disease of joints, tendon sheaths, and entheses affecting patients with established skin psoriasis, or, less frequently, patients without a personal history of psoriasis with a positive familial history. Many treatment options are now available to deal with the different aspects of the disease, including traditional and biological disease-modifying antirheumatic drugs and the recently released targeted synthetic disease-modifying antirheumatic drugs. However, ~40% of patients still fail to achieve a meaningful clinical response to first-line biologic therapy advocating the development of novel medications. It is now well accepted that T-cells participate in the immunopathogenesis of several autoimmune diseases. For this reason, the potential intervention on T-cells represented an attractive therapeutic target for a long time, becoming a clinical reality with the development of abatacept. Abatacept is a biologic agent selectively targeting the T-cell costimulatory signal delivered through the CD80/86-CD28 pathway and was approved in December 2005 by the US Food and Drug Administration and in May 2007 by European Medicines Agency for the treatment of patients with rheumatoid arthritis in combination with methotrexate. Based on the relevant role of T-cells in PsA pathogenesis and following the positive results obtained in a phase III clinical trial, abatacept recently received approval for treatment of patients with PsA. In this review, we will focus on the current knowledge about the emerging role of abatacept in treatment of PsA.
Collapse
Affiliation(s)
- Francesco Ursini
- Department of Health Sciences, University of Catanzaro "Magna Graecia", Catanzaro, Italy.,Associazione Calabrese per la Ricerca in Reumatologia, Catanzaro, Italy
| | - Emilio Russo
- Department of Health Sciences, University of Catanzaro "Magna Graecia", Catanzaro, Italy
| | | | | | - Salvatore D'Angelo
- Rheumatology Department of Lucania, Rheumatology Institute of Lucania (IReL), Potenza Italy.,Basilicata Ricerca Biomedica (BRB), Potenza, Italy
| |
Collapse
|