1
|
Renkli NÖ, Kleinrensink NJ, Spierings J, Mastbergen S, Vonkeman HE, Mooij SC, Schipper LG, Herman A, ten Katen I, Nap FJ, Hol ME, de Jong PA, Jansen MP, Foppen W. Multimodal imaging of structural damage and inflammation in psoriatic arthritis: a comparison of DMARD-naive and DMARD-failure patients. Rheumatology (Oxford) 2025; 64:1760-1769. [PMID: 39153007 PMCID: PMC11962931 DOI: 10.1093/rheumatology/keae450] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Revised: 07/15/2024] [Accepted: 07/23/2024] [Indexed: 08/19/2024] Open
Abstract
OBJECTIVES To compare inflammatory and structural differences in active PsA between DMARD-naive and DMARD-failure patients using diverse imaging approaches for future analyses. Additionally, to explore the influence of patient characteristics (clinical and demographic variables) on imaging findings. METHODS Of the 80 patients included from the first cohort of the ongoing multicentre TOFA-PREDICT trial, 40 were DMARD-naive and 40 were DMARD-failure (csDMARD failure; one prior bDMARD excluding etanercept was allowed), all meeting classification criteria for PsA with a minimum disease duration of eight weeks. Baseline conventional radiographs of hands and feet, MRIs of both ankles, and whole-body [18F]-fluorodeoxyglucose PET/CT (18F-FDG PET/CT) were evaluated for inflammatory and structural imaging parameters, including Sharp-van der Heijde (SHS), Heel Enthesitis Magnetic Resonance Imaging Scoring System (HEMRIS) and Deauville synovitis scoring. Differences between groups and the influence of patient characteristics were examined with multiple linear regression. RESULTS At baseline, patient characteristics were similar between groups. Imaging parameters showed limited inflammation and structural damage. Inflammatory imaging parameters were not significantly different (P > 0.200). Among structural parameters, only HEMRIS Achilles tendon structural damage was significantly different (P = 0.024, R2 = 0.071) and SHS Joint Space Narrowing was not statistically significant (P = 0.050, R2 = 0.048) with higher values for both in DMARD failures. After correction of patient characteristics, these differences in imaging disappeared (both P > 0.600). CONCLUSION At baseline, PsA patient groups were comparable concerning structural and inflammatory imaging parameters, especially after correcting for patient characteristics. Thus, DMARD-naive and DMARD-failure patient groups may be combined in future PsA progression and treatment decision studies. TRIAL REGISTRATION www.clinicaltrialsregister.eu. EudraCT: 2017-003900-28.
Collapse
Affiliation(s)
- Nağme Ö Renkli
- Department of Rheumatology and Clinical Immunology, UMC Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Nienke J Kleinrensink
- Department of Rheumatology and Clinical Immunology, UMC Utrecht, Utrecht University, Utrecht, The Netherlands
- Department of Radiology and Nuclear Medicine, UMC Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Julia Spierings
- Department of Rheumatology and Clinical Immunology, UMC Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Simon Mastbergen
- Department of Rheumatology and Clinical Immunology, UMC Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Harald E Vonkeman
- Department of Rheumatology, Medisch Spectrum Twente, Enschede, The Netherlands
- Department of Psychology, Health and Technology, University of Twente, Enschede, The Netherlands
| | - Shasti C Mooij
- Department of Rheumatology, Medisch Spectrum Twente, Enschede, The Netherlands
| | - Lydia G Schipper
- Department of Rheumatology, Elisabeth- TweeSteden Hospital, Tilburg, The Netherlands
| | - Amin Herman
- Department of Rheumatology, Antonius Hospital, Utrecht, The Netherlands
| | - Iris ten Katen
- Department of Radiology and Nuclear Medicine, UMC Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Frank J Nap
- Department of Radiology and Nuclear Medicine, UMC Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Marjolein E Hol
- Department of Radiology and Nuclear Medicine, UMC Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Pim A de Jong
- Department of Radiology and Nuclear Medicine, UMC Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Mylène P Jansen
- Department of Rheumatology and Clinical Immunology, UMC Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Wouter Foppen
- Department of Radiology and Nuclear Medicine, UMC Utrecht, Utrecht University, Utrecht, The Netherlands
| |
Collapse
|
2
|
Cagnotto G, Bruschettini M, Stróżyk A, Scirè CA, Compagno M. Tumor necrosis factor (TNF) inhibitors for psoriatic arthritis. Cochrane Database Syst Rev 2025; 2:CD013614. [PMID: 39945386 PMCID: PMC11822884 DOI: 10.1002/14651858.cd013614.pub2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/16/2025]
Abstract
BACKGROUND Psoriatic arthritis (PsA) is a chronic arthritis affecting people with psoriasis. If untreated, it may lead to disability. Recommended drugs are non-steroidal anti-inflammatory drugs (NSAIDs), corticosteroids, conventional synthetic disease-modifying anti-rheumatic drugs (csDMARDs), biologic DMARDs (bDMARDs), and targeted synthetic DMARDs (tsDMARDs). Tumour necrosis factor inhibitors (TNFi) are the first choice bDMARDs. OBJECTIVES To assess the benefits and harms of TNFi in adults with psoriatic arthritis. SEARCH METHODS We searched CENTRAL, MEDLINE, Embase, ClinicalTrials.gov and the World Health Organization trials portal up to 28 March 2024. SELECTION CRITERIA We included randomized controlled trials (RCTs) in adults with PsA, comparing TNFi to placebo, physiotherapy, NSAIDs, corticosteroids, and cs/b/tsDMARDs. Major outcomes included clinical improvement, minimal disease activity, physical function, health-related quality of life, radiographic progression, serious adverse events, and withdrawals due to adverse events. DATA COLLECTION AND ANALYSIS We used standard Cochrane methods. The primary comparison was TNFi versus placebo. The primary time point was 12 weeks for clinical improvement; 24 weeks for minimal disease activity, function, quality of life, and radiographic progression; and the end of the trial period for serious adverse events and withdrawals due to adverse events. MAIN RESULTS We included 25 RCTs randomizing 7857 participants. Four studies compared TNFi to methotrexate and one to ustekinumab in DMARD-naïve participants. In csDMARD-inadequate responders, 11 studies compared TNFi to placebo; four studies compared TNFi to placebo and ixekizumab, bimekizumab, tofacitinib, or upadacitinib; and three studies compared TNFi to ixekizumab, secukinumab, and ustekinumab. Two studies compared different TNFi. We found no studies with b/tsDMARD-inadequate responders (b/tsDMARD-IR). No studies compared TNFi to NSAIDs, corticosteroids, or physiotherapy. Performance (32%), detection (56%) and reporting (80%) biases were at high or unclear risk across studies. Only one study had a low risk of bias in all domains. We limit reporting to the primary comparison, TNFi versus placebo. DMARD-naïve We found no studies comparing TNFi with placebo in DMARD-naïve participants. csDMARD-inadequate responders TNFi probably result in a large clinical improvement compared to placebo. At 12 weeks, 149/1926 (8%) participants in the placebo group showed a clinical improvement (ACR50) compared to 784/2141 (37%) participants in the TNFi group (risk ratio (RR) 5.63, 95% confidence interval (CI) 3.98 to 7.96; I2 = 65%; 14 studies, 4067 participants; moderate-certainty evidence). TNFi probably result in a higher proportion of participants in minimal disease activity. At 24 weeks, 95/1017 (9%) participants in the placebo group were in minimal disease activity compared to 428/1336 (32%) participants in the TNFi group (RR 3.76, 95% CI 2.39 to 5.92; I2 = 72%; 5 studies, 2353 participants; moderate-certainty evidence). At 24 weeks, TNFi may improve function compared to placebo. The mean change in function from baseline (assessed with the Health Assessment Questionnaire; score from 0 to 3, 0 = no disability; minimal clinically important difference (MCID) = 0.35) was -0.14 points with placebo and 0.33 points lower (0.41 lower to 0.25 lower) with TNFi (I2 = 72%; 8 studies, 2949 participants; low-certainty evidence). TNFi probably result in a clinically important improvement in health-related quality of life. The mean change in quality of life from baseline (assessed with the Short Form 36-item Mental Component Summary questionnaire; score from 0 to 100, 100 = best score; MCID = 1.7) was 2.4 points with placebo and 3.29 points higher (2.18 points higher to 4.40 points higher) with TNFi (I2 = 52%; 8 studies, 2928 participants; moderate-certainty evidence). TNFi probably slightly reduce radiographic progression. The mean change in radiographic progression (assessed with the Sharp/Van der Heijde-PsA score; scale from 0 to 528, 0 = no damage) was 0.25 points with placebo and 0.37 points lower with TNFi (0.48 lower to 0.25 lower) (I2 = 32%; 7 studies, 2478 participants; moderate-certainty evidence) at 24 weeks. We downgraded the evidence to moderate certainty for clinical improvement, minimal disease activity, quality of life, and radiographic progression due to risk of bias. For function, we downgraded the evidence to low certainty for risk of bias and imprecision. TNFi may result in little to no difference in serious adverse events, but may slightly increase withdrawals due to adverse events, compared to placebo. At the end of follow-up: 56/1826 participants (3%) given placebo and 69/1900 (4%) participants given TNFi experienced serious adverse events (RR 1.00, 95% CI 0.70 to 1.42; I2 = 0%; 13 studies, 3866 participants; low-certainty evidence); and 35/1926 (2%) participants given placebo and 65/2140 (3%) given TNFi withdrew due to adverse events (RR 1.53, 95% CI 1.01 to 2.33; I2 = 0%; 14 studies, 4066 participants; low-certainty evidence). We downgraded the evidence to low certainty for risk of bias and imprecision. AUTHORS' CONCLUSIONS In csDMARD-inadequate responders, moderate-certainty evidence showed that TNFi probably result in a large clinical improvement, lower disease activity, small decrease in radiographic progression, and better quality of life compared to placebo. Low-certainty evidence showed that TNFi may lead to a slight improvement in physical function compared to placebo. Low-certainty evidence suggested that TNFi may lead to a slight increase in withdrawals due to adverse events, whereas they may result in little to no difference in serious adverse events compared to placebo. No trials assessed TNFi compared to placebo in DMARD-naïve participants or in b/tsDMARD-IR.
Collapse
Affiliation(s)
- Giovanni Cagnotto
- Clinical Sciences Malmö, Lund University, Malmö, Sweden
- Rheumatology, Skåne University Hospital, Malmö, Sweden
| | - Matteo Bruschettini
- Department of Clinical Sciences Lund, Paediatrics, Lund University, Skåne University Hospital, Lund, Sweden
- Cochrane Sweden, Department of Research, Development, Education and Innovation, Lund University, Skåne University Hospital, Lund, Sweden
| | - Agata Stróżyk
- Department of Paediatrics, Medical University of Warsaw, Warsaw, Poland
| | - Carlo Alberto Scirè
- School of Medicine, University of Milano-Bicocca, Milan, Italy
- Rheumatology Unit, IRCCS San Gerardo dei Tintori Foundation, Monza, Italy
| | - Michele Compagno
- Clinical Sciences Malmö, Lund University, Malmö, Sweden
- Rheumatology, Skåne University Hospital, Malmö, Sweden
| |
Collapse
|
3
|
Xiao A, Roy A, Dennett L, Yacyshyn E, Li MD. Imaging in clinical trials for psoriatic arthritis: a scoping review. Skeletal Radiol 2025:10.1007/s00256-025-04884-8. [PMID: 39912887 DOI: 10.1007/s00256-025-04884-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2024] [Revised: 01/09/2025] [Accepted: 01/26/2025] [Indexed: 02/07/2025]
Abstract
Psoriatic arthritis (PsA) is a chronic inflammatory condition principally affecting the skin and musculoskeletal system, associated with comorbidities and decreased quality of life. Imaging is crucial for diagnosis, monitoring progression, and evaluating treatment efficacy; therefore, it plays an important role in PsA clinical trials. In this review, we aimed to characterize how imaging modalities and advances in imaging techniques are being used specifically in the PsA clinical trial literature. Following the Arksey and O'Malley framework for scoping reviews, we conducted a comprehensive search of multiple literature databases, from January 1, 2000, to June 27, 2024. We captured 1961 articles and after deduplication, title and abstract screening, and full-text review, 53 studies were included. Radiographs were the most used imaging modality (n = 32/53, 60%), and radiographic progression was frequently measured using the PsA-modified Sharp/van der Heijde score. MRI (n = 16/53, 30%) was used in evaluating peripheral and axial disease, with more recent adoption of validated scoring systems (PsAMRIS and SPARCC). Ultrasound (n = 11/53, 21%), including power Doppler, was used to assess soft tissue inflammation. Standardized scoring systems (e.g. GLOESS) were used in a minority of ultrasound-based studies. Multimodality imaging (n = 7/53, 13%) and CT (n = 2/53, 4%) was uncommon. The development of PsA-specific scoring systems for radiographs and MRI has been instrumental in advancing imaging assessment in PsA. However, their application remains limited, particularly in ultrasound, where further standardization is needed. Future clinical trials should focus on increasing the adoption of PsA-specific scoring systems, exploring advanced techniques (e.g. DCE-MRI), and multi-modal imaging to improve PsA disease monitoring.
Collapse
Affiliation(s)
- Andrew Xiao
- Department of Medicine, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Ainge Roy
- Faculty of Science, University of Alberta, Edmonton, AB, Canada
| | - Liz Dennett
- Geoffrey and Robyn Sperber Health Sciences Library, University of Alberta, Edmonton, AB, Canada
| | - Elaine Yacyshyn
- Department of Medicine, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Matthew D Li
- Department of Radiology and Diagnostic Imaging, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, AB, Canada.
| |
Collapse
|
4
|
Kharouf F, Gladman DD. Treatment controversies in spondyloarthritis and psoriatic arthritis: focus on biologics and targeted therapies. Expert Rev Clin Immunol 2024; 20:1381-1400. [PMID: 39072530 DOI: 10.1080/1744666x.2024.2384705] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2024] [Accepted: 07/22/2024] [Indexed: 07/30/2024]
Abstract
INTRODUCTION There are several treatment controversies that have emerged in spondyloarthritis and psoriatic arthritis. These are related to the nature of the conditions as well as to the use of medications. AREAS COVERED This review, which included a search of PubMed database as well as the references within the articles provides an overview of the nature of spondyloarthritis, controversy over the inclusion of psoriatic arthritis (PsA) as a peripheral spondyloarthritis, and a summary of current treatments for both PsA and axial spondyloarthritis (axSpA), with special emphasis on targeted therapy. The review highlights the differences in response to certain medications, particularly biologic therapy and summarizes the randomized controlled trials in psoriatic arthritis and axial spondyloarthritis providing data about the responses in table format. EXPERT OPINION There is a need for better outcome measures in axSpA. Currently, the measures are subjective. Imaging may be more appropriate but there is a need for research into the reliability and responsiveness of imaging techniques. In PsA, there may also be better response measures and research into the reliability and responsiveness of available measures is underway. There is also a need for novel therapies as well as biomarkers for response in both diseases.
Collapse
Affiliation(s)
- Fadi Kharouf
- Division of Rheumatology, University Health Network, University of Toronto, Toronto, Ontario, Canada
- Gladman-Krembil Psoriatic Disease Program, Schroeder Arthritis Institute, Krembil Research Institute, Toronto Western Hospital, Toronto, Ontario, Canada
| | - Dafna D Gladman
- Division of Rheumatology, University Health Network, University of Toronto, Toronto, Ontario, Canada
- Gladman-Krembil Psoriatic Disease Program, Schroeder Arthritis Institute, Krembil Research Institute, Toronto Western Hospital, Toronto, Ontario, Canada
| |
Collapse
|
5
|
Jin Y, Cheng IT, Wu D, Yan X, Lau SL, Wong NS, Hung VW, Qin L, Lee RKL, Griffith JF, Szeto CC, So H, Tam LS. Imaging in psoriatic arthritis: established methods and emerging techniques. Ther Adv Musculoskelet Dis 2024; 16:1759720X241288060. [PMID: 39421802 PMCID: PMC11483715 DOI: 10.1177/1759720x241288060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Accepted: 09/13/2024] [Indexed: 10/19/2024] Open
Abstract
Psoriatic arthritis (PsA) is a heterogeneous, chronic, inflammatory musculoskeletal disease that can lead to peripheral and axial damage and loss of function. A clear difference between PsA and other forms of inflammatory arthritis is the different forms of bone remodeling seen in PSA which incorporates not only increased bone resorption with bone erosions, osteolysis, and loss of bone mineral density but also increased bone formation with periostitis, syndesmophytes, enthesiophytes, and ankylosis. PsA, if diagnosed late, will lead to significant structural damage, the most severe form of which is known as arthritis mutilans, and loss of physical function. Imaging plays a crucial role in diagnosing and monitoring both peripheral and axial conditions associated with PsA. Radiography is currently the main modality used to monitor structural damage in PsA though commonly used scoring systems do not include bony proliferation as a criterion. Besides, radiography is limited in determining the presence and cause of periarticular soft tissue thickening, which may arise from tendinosis, tenosynovitis, synovial proliferation, bursitis, or enthesitis. Recently, much more attention has been paid to determining the imaging characteristics of PsA, which enables more precise identification of disease and severity assessment. Newer imaging technologies also enable variations in normal bone microstructure to be distinguished from disease-related abnormality. This review discusses the current state of innovative imaging modalities in PsA, specifically concentrating on their roles in PsA diagnosis and treatment, improving the early detection of PsA, and identifying patients with skin psoriasis at risk of developing psoriatic arthritis.
Collapse
Affiliation(s)
- Yingzhao Jin
- Department of Medicine and Therapeutics, The Prince of Wales Hospital, The Chinese University of Hong Kong, Shatin, Hong Kong
- Li Ka Shing Institute of Health Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Isaac T Cheng
- Department of Medicine and Therapeutics, The Prince of Wales Hospital, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Dongze Wu
- Department of Medicine and Therapeutics, The Prince of Wales Hospital, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Xianfeng Yan
- Department of Medicine and Therapeutics, The Prince of Wales Hospital, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Sze-Lok Lau
- Department of Medicine and Therapeutics, The Prince of Wales Hospital, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Nga Sze Wong
- Department of Medicine and Therapeutics, The Prince of Wales Hospital, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Vivian W Hung
- Bone Quality and Health Centre, Department of Orthopaedics and Traumatology, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Ling Qin
- Bone Quality and Health Centre, Department of Orthopaedics and Traumatology, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Ryan Ka Lok Lee
- Department of Imaging and Interventional Radiology, The Prince of Wales Hospital, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - James F Griffith
- Department of Imaging and Interventional Radiology, The Prince of Wales Hospital, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Cheuk-Chun Szeto
- Li Ka Shing Institute of Health Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Ho So
- Department of Medicine and Therapeutics, The Prince of Wales Hospital, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Lai-Shan Tam
- Department of Medicine and Therapeutics, The Prince of Wales Hospital, The Chinese University of Hong Kong, 9F, LCW Clinical Sciences Building, Shatin, Hong Kong
| |
Collapse
|
6
|
Kristensen LE, Keiserman M, Papp K, McCasland L, White D, Carter K, Lippe R, Photowala H, Drogaris L, Soliman AM, Chen M, Padilla B, Behrens F. Efficacy and Safety of Risankizumab for Active Psoriatic Arthritis: 100-Week Results from the Phase 3 KEEPsAKE 1 Randomized Clinical Trial. Rheumatol Ther 2024; 11:617-632. [PMID: 38498141 PMCID: PMC11111619 DOI: 10.1007/s40744-024-00654-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Accepted: 02/13/2024] [Indexed: 03/20/2024] Open
Abstract
INTRODUCTION Patients with psoriatic arthritis (PsA) require treatment providing durable long-term efficacy in different disease domains as well as safety. We present 100-week efficacy and safety results of risankizumab in patients with active PsA and previous inadequate response/intolerance to ≥ 1 conventional synthetic disease-modifying antirheumatic drug (csDMARD-IR). METHODS KEEPsAKE 1 (NCT03675308) is a global phase 3 study, including a 24-week, double-blind, placebo-controlled and ongoing open-label extension periods. Patients were randomized 1:1 to receive risankizumab 150 mg or placebo at baseline and weeks 4 and 16. After week 24, all patients received open-label risankizumab every 12 weeks thereafter. Patients were evaluated through 100 weeks. Endpoints included achieving ≥ 20% reduction in American College of Rheumatology criteria for symptoms of rheumatoid arthritis (ACR20), minimal disease activity (MDA; defined as ≥ 5/7 criteria of low disease activity and extent), and other measures. RESULTS Overall, 828/964 (85.9%) patients completed week 100. For patients receiving continuous risankizumab, 57.3%, 70.6%, and 64.3% achieved ACR20 at weeks 24, 52, and 100, respectively. For the placebo/risankizumab cohort, 33.5% achieved ACR20 at week 24 but increased after switching to active treatment at weeks 52 (63.7%) and 100 (62.1%). In ACR20 responders at week 52, 81.2% of both treatment cohorts maintained response at week 100. MDA was achieved by 25.0%, 38.3%, and 38.2% of the continuous risankizumab cohort at weeks 24, 52, and 100. In the placebo/risankizumab cohort, 10.2% achieved MDA at week 24, increasing at weeks 52 (28.0%) and 100 (35.2%). MDA response was maintained at week 100 in week 52 responders in the continuous risankizumab (75.5%) and placebo/risankizumab cohorts (78.2%). Similar trends were observed for other efficacy measures. Risankizumab was generally well tolerated through 100 weeks. CONCLUSIONS For patients with active PsA who are csDMARD-IR, risankizumab demonstrated durable long-term efficacy and was generally well tolerated, with a consistent long-term safety profile. TRIAL REGISTRATION ClinicalTrials.gov identifier, NCT03675308.
Collapse
Affiliation(s)
- Lars Erik Kristensen
- The Parker Institute, Copenhagen University Hospital, Bispebjerg and Frederiksberg, Nordre Fasanvej 57 Road 8, Entrance 19, 2000, Frederiksberg, Copenhagen, Denmark.
| | - Mauro Keiserman
- Rheumatology Section, Pontifical Catholic University, School of Medicine, Porto Alegre, Brazil
| | - Kim Papp
- Probity Medical Research and Alliance Clinical Trials, Waterloo, ON, Canada
- Department of Medicine, University of Toronto, Toronto, ON, Canada
| | | | - Douglas White
- Rheumatology Department, Waikato Hospital, Hamilton, New Zealand
- Waikato Clinical School, University of Auckland, Auckland, New Zealand
| | | | | | | | | | | | | | | | - Frank Behrens
- Rheumatology, Immunology, Inflammation Medicine, University Hospital and Fraunhofer Institute for Translational Medicine & Pharmacology ITMP, Fraunhofer Cluster of Excellence Immune-Mediated Diseases CIMD, Goethe University, Frankfurt Am Main, Germany
| |
Collapse
|
7
|
Watson M, Tillett W, Jadon D, Massa MS, Francis A, Gullick N, Rombach I, Sinomati Y, Tucker L, Coates LC. The protocol of a clinical effectiveness trial comparing standard step-up care, early combination DMARD therapy and early use of TNF inhibitors for the treatment of moderate to severe psoriatic arthritis: the 3-arm parallel group SPEED randomized controlled trial. Ther Adv Musculoskelet Dis 2024; 16:1759720X241240913. [PMID: 38826570 PMCID: PMC11143811 DOI: 10.1177/1759720x241240913] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Accepted: 03/04/2024] [Indexed: 06/04/2024] Open
Abstract
Objectives The aim of the Severe Psoriatic arthritis - Early intervEntion to control Disease trial is to compare outcomes in psoriatic arthritis (PsA) patients with poor prognostic factors treated with standard step-up conventional synthetic disease-modifying anti-rheumatic drugs (csDMARDs), combination csDMARDs or a course of early biologics. Design This multicentre UK trial was embedded within the MONITOR-PsA cohort, which uses a trial within cohort design. Methods and analysis Patients with newly diagnosed PsA and at least one poor prognostic factor (polyarthritis, C-reactive protein >5 mg/dL, health assessment questionnaire >1, radiographic erosions) were randomized equally and open-label to either standard care with 'step-up' csDMARD therapy, initial therapy with combination csDMARDs (methotrexate with either sulfasalazine or leflunomide) or to early biologics induction therapy (adalimumab plus methotrexate). The primary outcome is the PsA disease activity score at week 24. Ethics Ethical approval for the study was granted by the South Central Research Ethics Committee (ref 18/SC/0107). Discussion Treatment recommendations for PsA suggest more intensive therapy for those with poor prognostic factors but there are no studies that have previously used prognostic factors to guide therapy. Applying initial intensive therapy has shown improved outcomes in other inflammatory arthritides but has never been tried in PsA. Combination csDMARDs have shown some superiority over single therapies but there are limited data and concerns about side effects. Early use of biologics has also been shown to be superior to methotrexate but these drugs are costly and not usually funded first line. However, if a short course of biologics can rapidly suppress inflammation allowing treatment to be withdrawn and response maintained on methotrexate, this may be a cost-effective model for early use. Trial registration ClinicalTrials.gov (NCT03739853) and EudraCT (2017-004542-24).
Collapse
Affiliation(s)
- Marion Watson
- Oxford Clinical Trials Research Unit, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, UK
| | - William Tillett
- Royal National Hospital for Rheumatic Diseases, Bath, Somerset, UK
- Department of Life Sciences, University of Bath, Bath, UK
| | - Deepak Jadon
- Department of Medicine, University of Cambridge, Cambridge, UK
| | - M. Sofia Massa
- Oxford Clinical Trials Research Unit, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, UK
| | - Anne Francis
- Oxford Clinical Trials Research Unit, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, UK
| | - Nicola Gullick
- University Hospitals Coventry and Warwickshire, Coventry, UK
| | - Ines Rombach
- Oxford Clinical Trials Research Unit, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, UK
- Sheffield Clinical Trials Research Unit, Sheffield Centre for Health and Related Research, University of Sheffield, Sheffield, UK
| | - Yvonne Sinomati
- Oxford Clinical Trials Research Unit, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, UK
| | - Laura Tucker
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, UK
| | - Laura C. Coates
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, Botnar Research Centre, University of Oxford, Windmill Road, Oxford OX3 7LD, UK
| |
Collapse
|
8
|
Mease PJ, Gottlieb AB, Ogdie A, McInnes IB, Chakravarty SD, Rampakakis E, Kollmeier A, Xu XL, Shawi M, Lavie F, Kishimoto M, Rahman P. Earlier clinical response predicts low rates of radiographic progression in biologic-naïve patients with active psoriatic arthritis receiving guselkumab treatment. Clin Rheumatol 2024; 43:241-249. [PMID: 37787903 PMCID: PMC10774160 DOI: 10.1007/s10067-023-06745-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 08/17/2023] [Accepted: 08/18/2023] [Indexed: 10/04/2023]
Abstract
OBJECTIVE Assess relationship between earlier clinical improvement and radiographic progression (RP) over 2 years in guselkumab-treated patients with active psoriatic arthritis (PsA). METHOD Post hoc analyses combined data from DISCOVER-2 biologic-naïve adults with active PsA randomized to either guselkumab 100 mg every 4 weeks (Q4W) or guselkumab at W0, W4, then Q8W. Correlations (Spearman's coefficient) between baseline disease parameters and total PsA-modified van der Heijde-Sharp (vdH-S) score were examined. Repeated-measures mixed models, adjusted for known RP risk factors, assessed the relationship between Disease Activity Index in PsA (DAPSA) improvement, DAPSA improvement exceeding the median or the minimal clinically important difference (MCID), or DAPSA low disease activity (LDA) at W8 and RP rate, assessed by change from baseline in vdH-S score through W100. RESULTS Baseline age, PsA duration, CRP level, and swollen joint count, but not psoriasis duration/severity, weakly correlated with baseline vdH-S score. Elevated baseline CRP (parameter estimate [β] = 0.17-0.18, p < 0.03) and vdH-S score (β = 0.02, p < 0.0001) significantly associated with greater RP through W100. Greater improvement in DAPSA (β = -0.03, p = 0.0096), achievement of DAPSA improvement > median (least squares mean [LSM] difference: -0.66, p = 0.0405) or > MCID (-0.67, p = 0.0610), or DAPSA LDA (-1.44, p = 0.0151) by W8 with guselkumab significantly associated with less RP through W100. The effect of W8 DAPSA LDA on future RP was strengthened over time among achievers vs. non-achievers (LSM difference enhanced from -1.05 [p = 0.0267] at W52 to -1.84 [p = 0.0154] at W100). CONCLUSIONS In guselkumab-treated patients with active PsA, earlier improvement in joint symptoms significantly associated with lower RP rates through 2 years, indicating blockade of the IL-23 pathway may modify long-term disease course and prevent further joint damage. Key Points • Greater improvement in DAPSA at Week 8 of guselkumab treatment was significantly associated with less progression of structural joint damage at 2 years in patients with active psoriatic arthritis (PsA). • Early control of peripheral joint disease activity with blockade of the IL-23 pathway may modify long-term PsA trajectory and prevent further joint damage.
Collapse
Affiliation(s)
- Philip J Mease
- Swedish Medical Center/Providence St. Joseph Health and University of Washington, Seattle, WA, USA.
| | - Alice B Gottlieb
- Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Alexis Ogdie
- Division of Rheumatology, University of Pennsylvania School of Medicine, Philadelphia, PA, USA
| | - Iain B McInnes
- College of Medical Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Soumya D Chakravarty
- Janssen Scientific Affairs, LLC, Horsham, PA, USA
- Drexel University College of Medicine, Philadelphia, PA, USA
| | - Emmanouil Rampakakis
- Department of Pediatrics, McGill University, Montreal, QC, Canada
- JSS Medical Research, Montreal, Canada
| | - Alexa Kollmeier
- Immunology, Janssen Research & Development, LLC, San Diego, CA, USA
| | - Xie L Xu
- Immunology, Janssen Research & Development, LLC, San Diego, CA, USA
| | - May Shawi
- Immunology, Janssen Research & Development, LLC, Titusville, NJ, USA
| | - Frederic Lavie
- Janssen Cilag Global Medial Affairs, Immunology Global Medical Affairs, Issy Les Moulineaux, France
| | - Mitsumasa Kishimoto
- Department of Nephrology and Rheumatology, Kyorin University School of Medicine, Mitaka, Tokyo, Japan
| | - Proton Rahman
- Discipline of Medicine, Division of Rheumatology, Memorial University of Newfoundland, St. John's, NF, Canada
| |
Collapse
|
9
|
Kristensen LE, Keiserman M, Papp K, McCasland L, White D, Lu W, Soliman AM, Eldred A, Barcomb L, Behrens F. Efficacy and safety of risankizumab for active psoriatic arthritis: 52-week results from the KEEPsAKE 1 study. Rheumatology (Oxford) 2023; 62:2113-2121. [PMID: 36282530 PMCID: PMC10234205 DOI: 10.1093/rheumatology/keac607] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Accepted: 10/15/2022] [Indexed: 09/30/2023] Open
Abstract
OBJECTIVE PsA is a chronic disease with heterogeneous clinical manifestations requiring treatment options with long-term efficacy and safety. In this follow-up analysis, the 52-week efficacy and safety of risankizumab 150 mg in patients with active PsA who had previous inadequate response/intolerance to one or more conventional synthetic DMARDs (csDMARD-IR) were evaluated. METHODS KEEPsAKE 1 is an ongoing, global, phase 3 study with a 24-week, double-blind, placebo-controlled period (period 1) and an open-label extension period (period 2). In period 1, eligible patients were randomized 1:1 to receive subcutaneous risankizumab 150 mg or placebo at weeks 0, 4 and 16. At week 24 (period 2), all continuing patients received open-label risankizumab 150 mg every 12 weeks through week 208. RESULTS At week 24, 57.3% of risankizumab-treated patients (n = 483) achieved ≥20% improvement in ACR criteria (ACR20) vs 33.5% of placebo-treated patients (n = 481; P < 0.001). At week 52, 70.0% of patients who were randomized to receive continuous risankizumab therapy and 63.0% of patients who were randomized to receive placebo in period 1 and then receive risankizumab at week 24 achieved ACR20. Similar result trends were observed for other efficacy measures. Risankizumab was well tolerated through 52 weeks of treatment with a consistent safety profile from week 24 through week 52. CONCLUSION In patients with active PsA who were csDMARD-IR, continuous risankizumab treatment demonstrated robust long-term efficacy and was well tolerated through 52 weeks of treatment. TRIAL REGISTRATION ClinicalTrials.gov, http://clinicaltrials.gov, KEEPsAKE1, NCT03675308.
Collapse
Affiliation(s)
- Lars Erik Kristensen
- The Parker Institute, Copenhagen University Hospital, Bispebjerg and Frederiksberg Hospital, Copenhagen, Denmark
| | - Mauro Keiserman
- Rheumatology Section, Pontifical Catholic University, School of Medicine, Porto Alegre, Brazil
| | - Kim Papp
- Probity Medical Research–K Papp Clinical Research, Waterloo, ON, Canada
| | - Leslie McCasland
- Department of Rheumatology, Loyola University Medical Center, Maywood, IL, USA
- Department of Veterans Affairs, Hines VA Hospital, Hines, IL, USA
| | - Douglas White
- Rheumatology Department, Waikato Hospital, Hamilton, New Zealand
- Waikato Clinical School, University of Auckland, Auckland, New Zealand
| | | | | | | | | | - Frank Behrens
- Rheumatology & Fraunhofer Institute for Translational Medicine and Pharmacology (ITMP), Fraunhofer Cluster of Excellence for Immune-Mediated Disease (CIMD), Goethe University, Frankfurt, Germany
| |
Collapse
|
10
|
The relationship between structural analysis of the hand and clinical characteristics in psoriatic arthritis. Sci Rep 2022; 12:18913. [PMID: 36344592 PMCID: PMC9640661 DOI: 10.1038/s41598-022-23555-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2022] [Accepted: 11/02/2022] [Indexed: 11/09/2022] Open
Abstract
Up to now, there is only limited information available on a possible relationship between clinical characteristics and the mineralization of metacarpal bones and finger joint space distance (JSD) in patients with psoriatic arthritis (PsA). Computerized digital imaging techniques like digital X-ray radiogrammetry (DXR) and computer-aided joint space analysis (CAJSA) have significantly improved the structural analysis of hand radiographs and facilitate the recognition of radiographic damage. The objective of this study was to evaluate clinical features which potentially influence periarticular mineralization of the metacarpal bones and finger JSD in PsA-patients. 201 patients with PsA underwent computerized measurements of the metacarpal bone mineral density (BMD) with DXR and JSD of all finger joints by CAJSA. DXR-BMD and JSD were compared with clinical features such as age and sex, disease duration, C-reactive protein (CRP) as well as treatment with prednisone and disease-modifying antirheumatic drugs (DMARDs). A longer disease duration and an elevated CRP value were associated with a significant reduction of DXR-BMD, whereas JSD-parameters were not affected by both parameters. DXR-BMD was significantly reduced in the prednisone group (-0.0383 g/cm²), but prednisone showed no impact on finger JSD. Patients under the treatment with bDMARDs presented significant lower DXR-BMD (-0.380 g/cm²), JSDMCP (-0.0179 cm), and JSDPIP (-0.0121 cm) values. Metacarpal BMD was influenced by inflammatory activity, prednisone use, and DMARDs. In contrast, finger JSD showed only a change compared to baseline therapy. Therefore, metacarpal BMD as well as finger JSD represent radiographic destruction under different aspects.
Collapse
|
11
|
Kleinrensink NJ, Perton FT, Pouw JN, Vincken NLA, Hartgring SAY, Jansen MP, Arbabi S, Foppen W, de Jong PA, Tekstra J, Leijten EFA, Spierings J, Lafeber FPJG, Welsing PMJ, Heijstek MW. TOFA-PREDICT study protocol: a stratification trial to determine key immunological factors predicting tofacitinib efficacy and drug-free remission in psoriatic arthritis (PsA). BMJ Open 2022; 12:e064338. [PMID: 36216430 PMCID: PMC9557317 DOI: 10.1136/bmjopen-2022-064338] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
INTRODUCTION Psoriatic arthritis (PsA) is a chronic, inflammatory, musculoskeletal disease that affects up to 30% of patients with psoriasis. Current challenges in clinical care and research include personalised treatment, understanding the divergence of therapy response and unravelling the multifactorial pathophysiology of this complex disease. Moreover, there is an urgent clinical need to predict, assess and understand the cellular and molecular pathways underlying the response to disease-modifying antirheumatic drugs (DMARDs). The TOFA-PREDICT clinical trial addresses this need. Our primary objective is to determine key immunological factors predicting tofacitinib efficacy and drug-free remission in PsA. METHODS AND ANALYSIS In this investigator-initiated, phase III, multicentre, open-label, four-arm randomised controlled trial, we plan to integrate clinical, molecular and imaging parameters of 160 patients with PsA. DMARD-naïve patients are randomised to methotrexate or tofacitinib. Additionally, patients who are non-responsive to conventional synthetic (cs)DMARDs continue their current csDMARD and are randomised to etanercept or tofacitinib. This results in four arms each with 40 patients. Patients are followed for 1 year. Treatment response is defined as minimal disease activity at week 16. Clinical data, biosamples and images are collected at baseline, 4 weeks and 16 weeks; at treatment failure (treatment switch) and 52 weeks. For the first 80 patients, we will use a systems medicine approach to assess multiomics biomarkers and develop a prediction model for treatment response. Subsequently, data from the second 80 patients will be used for validation. ETHICS AND DISSEMINATION The study was approved by the Medical Research Ethics Committee in Utrecht, Netherlands, is registered in the European Clinical Trials Database and is carried out in accordance with the Declaration of Helsinki. The study's progress is monitored by Julius Clinical, a science-driven contract research organisation. TRIAL REGISTRATION NUMBER EudraCT: 2017-003900-28.
Collapse
Affiliation(s)
- Nienke J Kleinrensink
- Department of Rheumatology and Clinical Immunology, University Medical Center Utrecht, Utrecht, The Netherlands
- Radiology, UMC Utrecht, Utrecht, The Netherlands
| | - Frank T Perton
- Department of Rheumatology and Clinical Immunology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Juliëtte N Pouw
- Department of Rheumatology and Clinical Immunology, University Medical Center Utrecht, Utrecht, The Netherlands
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Nanette L A Vincken
- Department of Rheumatology and Clinical Immunology, University Medical Center Utrecht, Utrecht, The Netherlands
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Sarita A Y Hartgring
- Department of Rheumatology and Clinical Immunology, University Medical Center Utrecht, Utrecht, The Netherlands
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Mylène P Jansen
- Department of Rheumatology and Clinical Immunology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Saeed Arbabi
- Image Sciences Institute, University Medical Center Utrecht, Utrecht, The Netherlands
- Orthopedics, University Medical Center Utrecht, Utrecht, The Netherlands
| | | | | | - Janneke Tekstra
- Department of Rheumatology and Clinical Immunology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Emmerik F A Leijten
- Department of Rheumatology and Clinical Immunology, University Medical Center Utrecht, Utrecht, The Netherlands
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Julia Spierings
- Department of Rheumatology and Clinical Immunology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Floris P J G Lafeber
- Department of Rheumatology and Clinical Immunology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Paco M J Welsing
- Department of Rheumatology and Clinical Immunology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Marloes W Heijstek
- Department of Rheumatology and Clinical Immunology, University Medical Center Utrecht, Utrecht, The Netherlands
| |
Collapse
|
12
|
Wang SH, Yu CL, Wang TY, Yang CH, Chi CC. Biologic Disease-Modifying Antirheumatic Drugs for Preventing Radiographic Progression in Psoriatic Arthritis: A Systematic Review and Network Meta-Analysis. Pharmaceutics 2022; 14:2140. [PMID: 36297574 PMCID: PMC9608970 DOI: 10.3390/pharmaceutics14102140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 09/30/2022] [Accepted: 10/05/2022] [Indexed: 11/06/2022] Open
Abstract
The prevention of joint deformity is among the most important treatment goals of psoriatic arthritis. Some biologics disease-modifying antirheumatic drugs (bDMARDs) have been demonstrated to be effective for both the skin and joints, as well as for slowing radiographic progression. However, there has been a lack of direct comparisons of bDMARDs. To evaluate the comparative effects of bDMARDs in preventing radiographic progression in psoriatic arthritis, we conducted a systematic review and network meta-analysis. On March 7 2022, a search for relevant randomized trials was conducted on MEDLINE, Embase, and the Cochrane Central Register of Controlled Trials. Our outcomes included radiographic non-progression, a mean change in the total radiographic score, and adverse events leading to discontinuation (DAE) at week 24. We included 11 trials on 10 bDMARDs, involving 4010 participants. Most bDMARDs were more effective than placebos in achieving radiographic non-progression, including adalimumab (odds ratio (OR) 4.7, 95% confidence interval (CI) 2.66-8.29), etanercept (OR 4.19, 95% CI 1.65-10.61), certolizumab pegol (OR 2.83, 95% CI 1.55-5.2), secukinumab 300 mg (OR 2.63, CI 1.62-4.27), infliximab (OR 2.54, CI 1.13-5.69), ixekizumab (OR 2.22, 95% CI 1.06-4.65), golimumab (OR 2.21, 95% CI 1.24-3.93), and abatacept (OR 1.54, 95% CI 1.03-2.28). A significant reduction in the total radiographic score was found in infliximab (standardized mean difference (SMD) -0.59, 95% CI -0.87, -0.3), etanercept (SMD -0.51, 95% CI -0.78, -0.23), adalimumab (SMD -0.45, 95% CI -0.64, -0.26), ixekizumab (SMD -0.37, 95% CI -0.62, -0.12), secukinumab 300 mg (SMD -0.33, 95% CI -0.50, -0.15), golimumab (SMD -0.33, 95% CI -0.58, -0.09), secukinumab 150 mg (SMD -0.25, 95% CI -0.43, -0.07), certolizumab pegol (SMD -0.23, 95% CI -0.44, -0.03), and ustekinumab (SMD -0.19, 95% CI -0.35, -0.33). No significant differences in DAE were detected between bDMARDs. In conclusion, anti-tumor necrosis factor agents (adalimumab, infliximab, and etanercept) may be preferred for treating psoriatic arthritis for their superiority in preventing radiographic progression.
Collapse
Affiliation(s)
- Szu-Hsuan Wang
- Department of Pharmacy, Chang Gung Memorial Hospital, Linkou, Taoyuan 33305, Taiwan
| | - Chia-Ling Yu
- Department of Pharmacy, Chang Gung Memorial Hospital, Linkou, Taoyuan 33305, Taiwan
| | - Tzu-Yu Wang
- Department of Applied Cosmetology, Lee-Ming Institute of Technology, New Taipei 24346, Taiwan
| | - Chung-Han Yang
- Division of Rheumatology, Allergy and Immunology, Chang Gung Memorial Hospital, Linkou, Taoyuan 33305, Taiwan
| | - Ching-Chi Chi
- Department of Dermatology, Chang Gung Memorial Hospital, Linkou, Taoyuan 33305, Taiwan
- School of Medicine, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan
| |
Collapse
|
13
|
Long-Term Efficacy and Safety of Risankizumab in Patients with Active Psoriatic Arthritis: Results from a 76-Week Phase 2 Randomized Trial. Rheumatol Ther 2022; 9:1361-1375. [PMID: 35931879 PMCID: PMC9510089 DOI: 10.1007/s40744-022-00474-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Accepted: 06/21/2022] [Indexed: 10/28/2022] Open
Abstract
INTRODUCTION The objective of this work was to assess the efficacy and safety of risankizumab in psoriatic arthritis (PsA) over 76 weeks. METHODS In this double-blind, dose-ranging phase 2 study, adults with active PsA were randomized 2:2:2:1:2 to risankizumab 150 mg at weeks 0, 4, 8, 12, and 16 (arm 1), 150 mg at weeks 0, 4, and 16 (arm 2), 150 mg at weeks 0 and 12 (arm 3), 75 mg at week 0 (arm 4), or placebo (arm 5). Patients completing week 24 could receive risankizumab 150 mg in a 52-week open-label extension study. Efficacy assessments included American College of Rheumatology (ACR) responses, Psoriasis Area Severity Index (PASI) responses, minimal disease activity (MDA), and 28-joint Disease Activity Score based on C-reactive protein (DAS28[CRP]). RESULTS Of 185 randomized patients, 173 (93.5%) completed week 16 and 145 (78.4%) entered the open-label extension. Significantly more patients in each risankizumab arm achieved ACR20 at week 16 versus placebo (primary endpoint: pooled arms 1 + 2 [59.5%] versus placebo [35.7%]; treatment difference [90% CI] 24.0 [9.3, 38.7]; P = 0.007). Similarly, significantly more patients in most risankizumab arms achieved ACR20/50/70, PASI75/90/100, MDA, and greater improvements in DAS28(CRP) versus placebo at week 16. These benefits of risankizumab were maintained long term. Treatment-emergent adverse events were comparable across treatment arms. Risankizumab 150 mg was well tolerated over 76 weeks. CONCLUSIONS Risankizumab improved joint and skin symptoms versus placebo in patients with active PsA over 16 weeks; improvements were sustained long term. Risankizumab was well tolerated over the long term with no new safety findings. TRIAL REGISTRATION NUMBERS NCT02719171 and NCT02986373.
Collapse
|
14
|
Distal Interphalangeal Joint Involvement May Be Associated with Disease Activity and Affected Joint Distribution in Rheumatoid Arthritis. J Clin Med 2022; 11:jcm11051405. [PMID: 35268496 PMCID: PMC8911492 DOI: 10.3390/jcm11051405] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Revised: 03/01/2022] [Accepted: 03/02/2022] [Indexed: 11/17/2022] Open
Abstract
We investigated the relationship between distal interphalangeal (DIP) joint involvement and disease activity in 10,038 patients with adult-onset rheumatoid arthritis (RA). The affected joint distribution was investigated using the joint indices (JI) x, y, and z, corresponding to the upper and lower joints, and the predominance of large-joint involvement, respectively. DIP joint involvement (defined by the presence of tenderness and/or swelling in DIP joints) was present in 206 (2.1%) of 10,038 patients with RA. Patients with RA exhibiting DIP joint involvement were significantly younger, and more frequently women. DIP joint involvement was positively associated with Disease Activity Score-28 using C-reactive protein, and clinical variables related to high RA disease activity, including JIs x and y, and was negatively associated with JI z. JI x was significantly higher than JI y in RA patients with DIP joint involvement. An odds ratio analysis revealed that small-to-medium sized and upper-extremity joints ranked first, second, and fourth among the eight variables significantly associated with DIP joint involvement. The correlation coefficients revealed that small-sized and upper-extremity joints ranked first and second among the five significant variables. DIP joint involvement, albeit rare, is significantly associated with high RA disease activity with predominance of small-sized and upper-extremity joints.
Collapse
|
15
|
Sarbu MI, Sarbu N, Cristea Ene D, Corche D, Baz R, Negru D, Nechita A, Fotea S, Anghel L, Tatu AL. New Perspectives on Diagnosing Psoriatic Arthritis by Imaging Techniques. Open Access Rheumatol 2021; 13:343-352. [PMID: 35221735 PMCID: PMC8866993 DOI: 10.2147/oarrr.s331859] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Accepted: 11/22/2021] [Indexed: 11/23/2022] Open
Abstract
Psoriatic arthritis is a chronic inflammatory condition that can lead to severe functional impairment and irreversible damage. The diagnosis can be difficult in early cases where the clinical exam is often scarce. The lack of a serological biomarker can lead to a considerable delay in diagnosis. In this review, we discuss the existent imaging methods that have improved the diagnosis of psoriatic arthritis (PsA). The degree and type of musculoskeletal involvement cannot be assessed by only one imaging method. We think that a combination of methods is the best approach to evaluate both structural damage and inflammatory lesions and that ultrasound (US) could be the best tool to screen a patient when considering the diagnosis of PsA. US is an accessible, non-ionizing technique that offers information regarding active inflammation in joints, entheses, and soft tissues.
Collapse
Affiliation(s)
| | - Nicolae Sarbu
- Faculty of Medicine and Pharmacy, “Dunarea de Jos” University, Al. I. Cuza No 35, Galati, Romania
- Correspondence: Nicolae Sarbu Faculty of Medicine and Pharmacy, “Dunarea de Jos” University, Al. I. Cuza No 35, Galati, RomaniaTel +40728301044 Email
| | | | - Daniela Corche
- Sf Apostol Andrei Clinical County Emergency Hospital, Galati, Romania
| | - Radu Baz
- Department of Radiology and Medical Imaging, Clinical County Emergency Hospital Constanta, “Ovidius” University, Constanta, Romania
| | - Dragos Negru
- Department of Radiology – Imaging, University Hospital “Sf. Spiridon”, University of Medicine and Pharmacy “Grigore T. Popa”, Iasi, Romania
| | - Aurel Nechita
- Faculty of Medicine and Pharmacy, “Dunarea de Jos” University, Galati, Romania
- Department of Pediatrics, “Sf. Ioan” Clinical Hospital for Children, Galati, Romania
| | - Silvia Fotea
- Faculty of Medicine and Pharmacy, “Dunarea de Jos” University, Galati, Romania
- Department of Pediatrics, “Sf. Ioan” Clinical Hospital for Children, Galati, Romania
| | - Lucretia Anghel
- Faculty of Medicine and Pharmacy, “Dunarea de Jos” University, Galati, Romania
- Sf Apostol Andrei Clinical County Emergency Hospital, Galati, Romania
| | - Alin Laurentiu Tatu
- Faculty of Medicine and Pharmacy, Clinical Department, Medical and Pharmaceutical Research Unit/Competitive, Interdisciplinary Research Integrated Platform’, ReForm-UDJG, “Dunarea de Jos” University, Galati, Romania
- Clinical Hospital St Parascheva of Infectious Diseases, Dermatology Department, Galati, Romania
| |
Collapse
|
16
|
McInnes IB, Rahman P, Gottlieb AB, Hsia EC, Kollmeier AP, Xu XL, Jiang Y, Sheng S, Shawi M, Chakravarty SD, van der Heijde D, Mease PJ. Long-term Efficacy and Safety of Guselkumab, a Monoclonal Antibody Specific to the p19 Subunit of Interleukin-23, Through 2 Years: Results from a Phase 3, Randomized, Double-blind, Placebo-controlled Study Conducted in Biologic-naïve Patients with Active Psoriatic Arthritis. Arthritis Rheumatol 2021; 74:475-485. [PMID: 34719872 PMCID: PMC9305108 DOI: 10.1002/art.42010] [Citation(s) in RCA: 63] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Revised: 09/14/2021] [Accepted: 10/19/2021] [Indexed: 11/10/2022]
Abstract
OBJECTIVE Assess long-term efficacy and safety of guselkumab, an IL-23p19-subunit inhibitor, in patients with active psoriatic arthritis (PsA) from the Phase-3 DISCOVER-2 trial. METHODS In DISCOVER-2, patients with active PsA (≥5 swollen and ≥5 tender joints; CRP ≥0.6 mg/dL) despite prior nonbiologic therapy were randomized to: guselkumab 100mg every-4-weeks (Q4W); at Week0, Week4, and Q8W; or placebo➔guselkumab Q4W at Week24. Efficacy assessments included ≥20%/50%/70% improvement in ACR components (ACR20/50/70), Investigator's Global Assessment of psoriasis score=0 (IGA=0; indicating complete skin clearance), enthesitis (Leeds Enthesitis Index) and dactylitis (Dactylitis Severity Score) resolution, and changes in PsA-modified van der Heijde-Sharp (vdH-S) radiographic scores. Clinical data (imputed as no response/no change from baseline if missing) and observed radiographic data were summarized through Week100; safety assessments continued through Week112. RESULTS Of 739 randomized and treated patients, 652 (88%) completed treatment through Week100. Across groups of guselkumab-treated patients (including placebo➔Q4W) ACR20 (68%-76%), ACR50 (48%-56%), ACR70 (30%-36%), and IGA=0 (55%-67%) responses and enthesitis (62%-70%) and dactylitis (72%-83%) resolution rates at Week100 indicated amelioration of arthritis signs/symptoms and extra-articular manifestations was durable through 2years. Mean changes in PsA-modified vdH-S scores from Week52-100 (0.13-0.75) indicated the low rates of radiographic progression observed among guselkumab-treated patients at earlier timepoints extended through Week100. Through Week112, 8% (5.8/100 patient-years) and 3% (1.9/100 patient-years) of 731 guselkumab-treated patients had a serious adverse event or serious infection, respectively; one death occurred (road traffic accident). CONCLUSION In biologic-naïve PsA patients, guselkumab provided durable improvements in multiple disease domains with no unexpected safety findings through 2years.
Collapse
Affiliation(s)
| | - Proton Rahman
- Memorial University of Newfoundland, St. Johns, NL, Canada
| | | | - Elizabeth C Hsia
- Janssen Research & Development, LLC, Spring House, PA, USA.,University of Pennsylvania, Philadelphia, PA, USA
| | | | - Xie L Xu
- Janssen Research & Development, LLC, Spring House, PA, USA
| | - Yusang Jiang
- Janssen Research & Development, LLC, Spring House, PA, USA
| | - Shihong Sheng
- Janssen Research & Development, LLC, Spring House, PA, USA
| | - May Shawi
- Immunology Global Medical Affairs, Janssen Pharmaceutical Companies of Johnson & Johnson, Horsham, PA, USA
| | - Soumya D Chakravarty
- Janssen Scientific Affairs, LLC, Horsham, PA, USA.,Drexel University College of Medicine, Philadelphia, PA, USA
| | | | - Philip J Mease
- Swedish Medical Center/Providence St. Joseph Health, University of Washington, Seattle, WA, USA
| |
Collapse
|
17
|
McInnes IB, Rahman P, Gottlieb AB, Hsia EC, Kollmeier AP, Chakravarty SD, Xu XL, Subramanian RA, Agarwal P, Sheng S, Jiang Y, Zhou B, Zhuang Y, van der Heijde D, Mease PJ. Efficacy and Safety of Guselkumab, an Interleukin-23p19-Specific Monoclonal Antibody, Through One Year in Biologic-Naive Patients With Psoriatic Arthritis. Arthritis Rheumatol 2021; 73:604-616. [PMID: 33043600 PMCID: PMC9291746 DOI: 10.1002/art.41553] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Accepted: 10/06/2020] [Indexed: 12/28/2022]
Abstract
Objective Guselkumab, a human monoclonal antibody specific to interleukin‐23p19, demonstrated efficacy and safety versus placebo through week 24 of the phase III DISCOVER‐2 trial in biologic‐naive patients with psoriatic arthritis (PsA). Here we report 1‐year DISCOVER‐2 findings. Methods Adults with active PsA (≥5 swollen and ≥5 tender joints; C‐reactive protein level ≥0.6 mg/dl) despite standard nonbiologic treatment were randomized to receive subcutaneous injections of guselkumab 100 mg every 4 weeks, guselkumab 100 mg at week 0, week 4 and every 8 weeks thereafter, or placebo with crossover to guselkumab 100 mg every 4 weeks at week 24. We primarily evaluated clinical efficacy through week 52 by imputing missing data (nonresponse for categorical end points; no change/using multiple imputation for continuous end points). Observed radiographic scores and adverse events (AEs) were summarized. Results Of 739 randomized, treated patients, 93% completed week 52. The proportions of patients in whom a ≥20% improvement from baseline in American College of Rheumatology criteria (ACR20) was achieved were maintained after week 24, reaching 71% (173 of 245) and 75% (185 of 248) for patients randomized to receive treatment every 4 weeks or every 8 weeks, respectively, by week 52. The proportions of patients in whom ACR50/ACR70 and skin responses, minimal or very low disease activity, and dactylitis or enthesitis resolution were achieved at week 24 were also maintained through week 52. Further, low levels of radiographic progression, along with improvements in physical function and health‐related quality of life, were sustained through week 52 with continued guselkumab treatment. Few patients experienced serious infections through week 52, with no evidence of a dosing regimen response or increase from weeks 0–24 (4 of 493 [0.8%]) to weeks 24–52 (3 of 493 [0.6%]) among guselkumab‐randomized patients. No patient developed an opportunistic infection or died. Conclusion In biologic‐naive PsA patients, guselkumab provided sustained improvements across diverse manifestations and maintained a favorable risk–benefit profile through week 52.
Collapse
Affiliation(s)
| | - Proton Rahman
- Memorial University of NewfoundlandSt. JohnsNewfoundlandCanada
| | | | - Elizabeth C. Hsia
- Janssen Research and Development, LLCSpring House, Pennsylvania, and University of Pennsylvania Perelman School of MedicinePhiladelphia
| | | | - Soumya D. Chakravarty
- Janssen Scientific Affairs, LLC, Horsham, Pennsylvania, and Drexel University College of MedicinePhiladelphiaPennsylvania
| | - Xie L. Xu
- Janssen Research and Development, LLCSan DiegoCalifornia
| | | | | | - Shihong Sheng
- Janssen Research & Development, LLCSpring HousePennsylvania
| | | | - Bei Zhou
- Janssen Research & Development, LLCSpring HousePennsylvania
| | - Yanli Zhuang
- Janssen Research & Development, LLCSpring HousePennsylvania
| | | | - Philip J. Mease
- Swedish Medical Center/Providence St. Joseph Health and University of WashingtonSeattleWashington
| |
Collapse
|
18
|
Does current evidence on disease-modifying antirheumatic drugs for psoriatic arthritis reinforce an effect on radiographic progression? Results from a systematic review and meta-analysis. Clin Rheumatol 2021; 40:3499-3510. [PMID: 33576924 DOI: 10.1007/s10067-021-05622-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Revised: 01/09/2021] [Accepted: 01/31/2021] [Indexed: 10/22/2022]
Abstract
This study aims to estimate the effect of synthetic and biologic disease-modifying antirheumatic drugs (DMARDs) on radiographic progression and quality of life in adult patients with psoriatic arthritis. A comprehensive search was performed using MEDLINE, Embase, Web of Science, Scopus, and Cochrane Central Register of Controlled Trials (CCRCT). Clinical trials comparing DMARDs with placebo for ≥ 12 weeks were included. The meta-analysis was conducted with a random-effects model using mean differences (MD). A total of 16 trials with overall moderate quality of evidence were included. Exposure to a biologic agent reduced radiographic progression at 24 weeks of treatment (MD: - 0.66; [95% CI - 0.97 to - 0.34]; P < .00001; I2 = 100%). The reduction of the baseline score was more than two times higher for TNF blockers compared with IL-17 and IL-12/IL-23 inhibitors (MD: - 0.94 vs - 0.41). Improvement in health-related quality of life scores was observed in biologic-treated populations (MD: - 0.21; [95% CI - 0.25 to - 0.18]; P < .00001; I2 = 97%). No sufficient data were available regarding conventional synthetic agents. Our data analyses suggest a better control of radiological damage with bDMARDs, as compared to placebo, after 24 weeks of treatment. However, the accuracy of these results in real life are jeopardized by the exceedingly high level of heterogeneity exhibited within and across included studies, and the true intervention effect cannot be determined with confidence. Further research is required to assess long-term outcomes and to control heterogeneity in the evaluation of treatments for psoriatic arthritis. PROSPERO registration number: CRD42019122223. Key Points • Radiographic progression is not the primary outcome for most efficacy studies in psoriatic arthritis; hence, baseline data are substantially diverse in major clinical trials. • The best available evidence on this particular outcome is currently at a moderate risk of bias. • Existing reports of the effect of DMARDs on structural damage must be taken with caution. • Further research is required to assess long-term outcomes and to control heterogeneity between studies.
Collapse
|
19
|
Chandran V, van der Heijde D, Fleischmann RM, Lespessailles E, Helliwell PS, Kameda H, Burgos-Vargas R, Erickson JS, Rathmann SS, Sprabery AT, Birt JA, Shuler CL, Gallo G. Ixekizumab treatment of biologic-naïve patients with active psoriatic arthritis: 3-year results from a phase III clinical trial (SPIRIT-P1). Rheumatology (Oxford) 2021; 59:2774-2784. [PMID: 32031665 PMCID: PMC7516094 DOI: 10.1093/rheumatology/kez684] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Revised: 12/16/2019] [Indexed: 01/20/2023] Open
Abstract
OBJECTIVE The aim was to assess the safety and efficacy of up to 156 weeks of ixekizumab (an IL-17A antagonist) treatment in PsA patients. METHODS In a phase III study, patients naïve to biologic treatment were randomized to placebo, adalimumab 40 mg every 2 weeks (ADA; active reference) or ixekizumab 80 mg every 2 weeks (IXEQ2W) or every 4 weeks (IXEQ4W) after an initial dose of 160 mg. At week 24 (week 16 for inadequate responders), ADA (after 8-week washout) and placebo patients were re-randomized to IXEQ2W or IXEQ4W. Outcomes were evaluated using a modified non-responder imputation [linear extrapolation for radiographic progression (modified total Sharp score = 0)] during extended treatment until week 156. RESULTS Of 417 patients, 381 entered the extension, and 243 of 381 (63.8%) completed the 156-week study. Incidence rates of treatment-emergent and serious adverse events, respectively, were 38.0 and 5.2 with IXEQ2W (n = 189) and 38.1 and 8.0 with IXEQ4W (n = 197). One death occurred (IXEQ4W). With IXEQ2W and IXEQ4W, respectively, the response rates persisted to week 156 as measured by the ACR response ≥20% (62.5 and 69.8%), ≥50% (56.1 and 51.8%) and ≥70% (43.8 and 33.4%), psoriasis area and severity index (PASI) 75 (69.1 and 63.5%), PASI 90 (64.5 and 51.2%) and PASI 100 (60.5 and 43.6%). Inhibition of radiographic progression also persisted to week 156 in 61% of IXEQ2W and 71% of IXEQ4W patients. CONCLUSION In this 156-week study of ixekizumab, the safety profile remained consistent with previous reports, and improvements in signs and symptoms of PsA were observed, including persistent low rates of radiographic progression. TRIAL REGISTRATION ClinicalTrials.gov, http://clinicaltrials.gov, NCT01695239, EudraCT 2011-002326-49.
Collapse
Affiliation(s)
- Vinod Chandran
- Division of Rheumatology, Department of Medicine, University of Toronto, ON, Canada.,Institute of Medical Science, University of Toronto, ON, Canada.,Krembil Research Institute, University Health Network, Toronto, ON, Canada
| | | | - Roy M Fleischmann
- Metroplex Clinical Research Center, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Eric Lespessailles
- Department of Rheumatology CHR Orléans, University of Orléans, Orléans, France
| | | | - Hideto Kameda
- Department of Internal Medicine, Toho University, Tokyo, Japan
| | | | | | | | | | | | | | - Gaia Gallo
- Eli Lilly and Company, Indianapolis, IN, USA
| |
Collapse
|
20
|
Vecellio M, Hake VX, Davidson C, Carena MC, Wordsworth BP, Selmi C. The IL-17/IL-23 Axis and Its Genetic Contribution to Psoriatic Arthritis. Front Immunol 2021; 11:596086. [PMID: 33574815 PMCID: PMC7871349 DOI: 10.3389/fimmu.2020.596086] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Accepted: 11/25/2020] [Indexed: 12/14/2022] Open
Abstract
Psoriatic arthritis (PsA) is a chronic inflammatory disease belonging to the family of spondyloarthropathies (SpA). PsA commonly aggravates psoriasis of the skin and frequently manifests as an oligoarthritis with axial skeletal involvement and extraarticular manifestations including dactylitis, enthesitis, and uveitis. The weight of genetic predisposition to psoriasis and PsA is illustrated by the concordance rates in monozygotic twins which clearly demonstrate that genomics is insufficient to induce the clinical phenotype. The association of PsA with several single nucleotide polymorphisms (SNPs) at the IL23R locus and the involvement of Th17 cells in the immunopathogenesis of PsA clearly put the IL-23/IL-17 axis in the spotlight. The IL-23 and IL-17 cytokines have a pivotal role in the chronic inflammation of the synovium in PsA and are also prominent in the skin lesions of those with PsA. In this review, we focus on the genetic association of the IL-23/IL-17 axis with PsA and the contribution of these master cytokines in the pathophysiology of the disease, highlighting the main cell types incriminated in PsA and their specific role in the peripheral blood, lesional skin and joints of patients. We then provide an overview of the approved biologic drugs targeting the IL-23/IL-17 axis and discuss the advantages of genetic stratification to enhance personalized therapies in PsA.
Collapse
Affiliation(s)
- Matteo Vecellio
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, United Kingdom.,Division of Rheumatology and Clinical Immunology, Humanitas Clinical and Research Center, IRCCS, Milan, Italy
| | - Vivien Xanath Hake
- Division of Rheumatology and Clinical Immunology, Humanitas Clinical and Research Center, IRCCS, Milan, Italy
| | - Connor Davidson
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, United Kingdom
| | | | - B Paul Wordsworth
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, United Kingdom
| | - Carlo Selmi
- Division of Rheumatology and Clinical Immunology, Humanitas Clinical and Research Center, IRCCS, Milan, Italy
| |
Collapse
|
21
|
Applying precision medicine to unmet clinical needs in psoriatic disease. Nat Rev Rheumatol 2020; 16:609-627. [PMID: 33024296 DOI: 10.1038/s41584-020-00507-9] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/07/2020] [Indexed: 02/08/2023]
Abstract
Psoriatic disease (PsD) is a heterogeneous condition that can affect peripheral and axial joints (arthritis), entheses, skin (psoriasis) and other structures. Over the past decade, considerable advances have been made both in our understanding of the pathogenesis of PsD and in the treatment of its diverse manifestations. However, several major areas of continued unmet need in the care of patients with PsD have been identified. One of these areas is the prediction of poor outcome, notably radiographic outcome in patients with psoriatic arthritis, so that stratified medicine approaches can be taken; another is predicting response to the numerous current and emerging therapies for PsD, so that precision medicine can be applied to rapidly improve clinical outcome and reduce the risk of toxicity. In order to address these needs, novel approaches, including imaging, tissue analysis and the application of proteogenomic technologies, are proposed as methodological solutions that will assist the dissection of the critical immune-metabolic pathways in this complex disease. Learning from advances made in other inflammatory diseases, it is time to address these unmet needs in a multi-centre partnership aimed at improving short-term and long-term outcomes for patients with PsD.
Collapse
|
22
|
Husni ME, Kavanaugh A, Murphy F, Rekalov D, Harrison DD, Kim L, Lo KH, Leu JH, Hsia EC. Efficacy and Safety of Intravenous Golimumab Through One Year in Patients With Active Psoriatic Arthritis. Arthritis Care Res (Hoboken) 2020; 72:806-813. [PMID: 30980514 PMCID: PMC7318581 DOI: 10.1002/acr.23905] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Accepted: 04/09/2019] [Indexed: 12/13/2022]
Abstract
Objective The present study was undertaken to evaluate the safety and efficacy of intravenous (IV) golimumab in patients with active psoriatic arthritis (PsA) through 1 year. Methods GO‐VIBRANT was a phase III, randomized, placebo‐controlled trial of 480 adults with active PsA. Patients were randomized to receive IV placebo (n = 239) or golimumab 2 mg/kg (n = 241) at weeks 0, 4, and every 8 weeks, with placebo crossover to golimumab at weeks 24, 28, and every 8 weeks thereafter. Efficacy through week 52 was assessed using the American College of Rheumatology (ACR) ≥20%, 50%, or 70% improvement criteria (ACR20/50/70), and the Psoriasis Area and Severity Index ≥75% improvement criteria (PASI75). Radiographic progression was measured using the PsA‐modified Sharp/van der Heijde score (SHS). Adverse events (AEs) were monitored through week 60. Results The primary and major secondary end points through week 24 were achieved. At week 52, 76.8% of patients in the golimumab group and 77.0% in the placebo‐crossover group achieved an ACR20 response, 58.1% and 53.6%, respectively, achieved an ACR50 response, and 38.6% and 33.9%, respectively, achieved an ACR70 response. Among patients with ≥3% body surface area affected, 71.9% in the golimumab group and 60.6% in the placebo‐crossover group achieved a PASI75 response at week 52. Mean change from baseline in total SHS at week 52 was –0.5 in the golimumab group and 0.8 in the placebo‐crossover group. Through week 60, 50.9% of all golimumab‐treated patients had ≥1 AE, and 5.2% had ≥1 serious AE. There were no opportunistic infections, 2 malignancies, and 1 death in patients treated with golimumab. Conclusion Sustained improvements in joint and skin disease in patients with PsA were maintained through 1 year in the GO‐VIBRANT study. No new safety signals for IV golimumab were identified.
Collapse
Affiliation(s)
| | | | - Frederick Murphy
- Altoona Center for Clinical Research, Duncansville, Pennsylvania
| | | | | | - Lilianne Kim
- Janssen Research & Development, Spring House, Pennsylvania
| | - Kim Hung Lo
- Janssen Research & Development, Spring House, Pennsylvania
| | - Jocelyn H Leu
- Janssen Research & Development, Spring House, Pennsylvania
| | - Elizabeth C Hsia
- Janssen Research & Development, Spring House, Pennsylvania, and University of Pennsylvania, Philadelphia
| |
Collapse
|
23
|
Laasonen L, Lindqvist U, Iversen L, Ejstrup L, Jonmundsson T, Ståhle M, Gudbjornsson B. Radiographic scoring systems for psoriatic arthritis are insufficient for psoriatic arthritis mutilans: results from the Nordic PAM Study. Acta Radiol Open 2020; 9:2058460120920797. [PMID: 32426163 PMCID: PMC7218277 DOI: 10.1177/2058460120920797] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2019] [Accepted: 03/29/2020] [Indexed: 01/21/2023] Open
Abstract
Background Psoriatic arthritis mutilans (PAM) is the most severe phenotype of psoriatic arthritis (PsA). Purpose To describe the radiological features in PAM and explore whether existing scoring systems for radiological damage in psoriatic arthritis are applicable for PAM. Material and Methods Radiographs were scored according to the modified Sharp-van der Heijde (mSvdH) and the Psoriatic Arthritis Ratingen Score (PARS) systems for PsA. Results At inclusion, 55 PAM patients (49% women, mean age 58 ± 12 years) had conventional radiographs of both hands and feet. A total of 869 PAM joints were detected and 193 joints with ankylosis. The mean total mSvdH score was 213.7 ± 137.8 (41% of maximum) with a higher score for hands than for feet: 136.6 ± 90.1 vs. 79.1 ± 60.9. However, the total score was relatively higher in the feet than in the hands when compared to the highest possible scoring (47% vs. 38% of max). The mean total PARS score was 126.3 ± 79.6 (35% of max). Scoring for joint destruction was higher than for proliferation (22% vs. 11% of max). Strong correlation was found between mSvdH and PARS (r2 = 0.913). A significant correlation was found between scoring and duration of arthritis and the Health Assessment Questionnaire. History of smoking, BMI, and gender did not influence the scoring values. Conclusions The two scoring systems studied may not be ideal to indicate progression of PAM in advanced disease since they reach ceiling effects rather early. Therefore, reporting early signs suggestive of PAM, e.g. signs of pencil-in-cup deformities or osteolysis, is crucial. This would reveal the presence of PAM and might lead to improved treatment in order to minimize joint damage.
Collapse
Affiliation(s)
- Leena Laasonen
- Helsinki Medical Imaging Center, Helsinki University Central Hospital, Helsinki, Finland
| | - Ulla Lindqvist
- Department of Medical Sciences, Rheumatology, Uppsala University, Uppsala, Sweden
| | - Lars Iversen
- Department of Dermatology, Aarhus University Hospital, Aarhus, Denmark
| | - Leif Ejstrup
- Department of Rheumatology, Odense University Hospital, Odense, Denmark
| | - Thorarinn Jonmundsson
- Centre for Rheumatology Research, University Hospital and Faculty of Medicine, University of Iceland, Reykjavik, Iceland
| | - Mona Ståhle
- Dermatology Division, Department of Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Bjorn Gudbjornsson
- Centre for Rheumatology Research, University Hospital and Faculty of Medicine, University of Iceland, Reykjavik, Iceland
| |
Collapse
|
24
|
Mease P, Husni ME, Kafka S, Chakravarty SD, Harrison DD, Lo KH, Xu S, Hsia EC, Kavanaugh A. Inhibition of radiographic progression across levels of composite index-defined disease activity in patients with active psoriatic arthritis treated with intravenous golimumab: results from a phase-3, double-blind, placebo-controlled trial. Arthritis Res Ther 2020; 22:43. [PMID: 32143685 PMCID: PMC7059340 DOI: 10.1186/s13075-020-2126-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Accepted: 02/06/2020] [Indexed: 11/10/2022] Open
Abstract
Background In the GO-VIBRANT trial of intravenous golimumab in psoriatic arthritis (PsA), golimumab significantly inhibited radiographic progression. In post hoc analyses, we evaluated changes in total PsA-modified Sharp/van der Heijde scores (SHS) across levels of composite index-defined disease activity following treatment. Methods In this phase-3, double-blind, placebo-controlled trial, 480 bio-naïve patients with active PsA randomly received intravenous golimumab 2 mg/kg (N = 241; week 0, week 4, every 8 weeks [q8w]) or placebo (N = 239; week 0, week 4, week 12, week 20) followed by golimumab (week 24, week 28, q8w) through week 52. Week 24 and week 52 SHS changes in patient subgroups, defined by levels of disease activity as assessed by several composite measures (minimal disease activity [MDA], very low disease activity [VLDA], Psoriatic ArthritiS Disease Activity Score [PASDAS], Disease Activity in Psoriatic Arthritis [DAPsA], Clinical Disease Activity Index [CDAI]), were evaluated post hoc in 474 patients with evaluable radiographic data. Partially (last-observation-carried-forward methodology) and completely (nonresponder methodology) missing data were imputed. Results Across indices, golimumab-treated patients demonstrated less radiographic progression than placebo-treated patients, regardless of disease activity state achieved via golimumab, from week 0 to 24 (e.g., mean changes in PsA-modified SHS were − 0.83 vs. 0.91, respectively, in patients achieving MDA and − 0.05 vs. 1.49, respectively, in those not achieving MDA). Treatment differences observed at week 24 persisted through week 52, despite placebo-randomized patients crossing over to golimumab at week 24 (e.g., mean changes in PsA-modified SHS from week 0 to 52 for golimumab- vs. placebo→golimumab-treated patients achieving MDA were − 1.16 vs. 1.19, respectively) and regardless of whether low disease activity was achieved (0.03 vs. 1.50, respectively, in those not achieving MDA). Consistent patterns were observed for disease activity assessed using VLDA, PASDAS, DAPsA, and CDAI composite endpoints. Conclusions The extent of structural damage inhibition afforded by up to 1 year of intravenous golimumab treatment paralleled levels of PsA activity, with greater progression of structural damage observed in patients with sustained higher disease activity. Among patients not achieving low levels of disease activity across several composite indices, golimumab-randomized patients appeared to exhibit far less progression of structural damage than placebo-randomized PsA patients, illustrating a potential disconnect between responses, wherein golimumab can inhibit structural damage independent of clinical effect. Trial registration ClinicalTrials.gov. NCT02181673. Registered 04 July 2014.
Collapse
Affiliation(s)
- Philip Mease
- Seattle Rheumatology Associates, Swedish Medical Center/Providence St. Joseph Health and University of Washington School of Medicine, 601 Broadway, Suite 600, Seattle, WA, 98122, USA.
| | | | - Shelly Kafka
- Janssen Scientific Affairs, LLC, Horsham, PA, USA
| | - Soumya D Chakravarty
- Janssen Scientific Affairs, LLC, Horsham, PA, USA.,Drexel University College of Medicine, Philadelphia, PA, USA
| | | | - Kim Hung Lo
- Janssen Research & Development, LLC, Spring House, PA, USA
| | - Stephen Xu
- Janssen Research & Development, LLC, Spring House, PA, USA
| | - Elizabeth C Hsia
- Janssen Research & Development, LLC, Spring House, PA, USA.,University of Pennsylvania, Philadelphia, PA, USA
| | | |
Collapse
|
25
|
van der Heijde D, Gladman DD, Kavanaugh A, Mease PJ. Assessing structural damage progression in psoriatic arthritis and its role as an outcome in research. Arthritis Res Ther 2020; 22:18. [PMID: 32014044 PMCID: PMC6998345 DOI: 10.1186/s13075-020-2103-8] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Accepted: 01/12/2020] [Indexed: 12/16/2022] Open
Abstract
Psoriatic arthritis (PsA) is an immune-mediated, clinically heterogeneous disease characterized by arthritis, enthesitis, dactylitis, spondylitis, and psoriasis of the skin and nails. Persistent articular inflammation in patients with PsA can lead to structural damage, which can result in reduced physical function and quality of life. Structural damage can occur rapidly, and irreversible joint damage may be observed if patients are not treated promptly and appropriately. Therefore, evaluating therapeutic agents for their ability to inhibit structural progression has become increasingly important, with radiographic progression becoming a key efficacy outcome in clinical trials in PsA. Here, we review how structural damage and progression are assessed in clinical trials and the use of radiographic progression as a study outcome. We also discuss possible limitations in the current assessment of radiographic progression as well as areas of research that may improve the assessment of structural damage in clinical trials of PsA.
Collapse
Affiliation(s)
- Désirée van der Heijde
- Department of Rheumatology, Leiden University Medical Center, Albinusdreef 2, 2333, ZA, Leiden, the Netherlands.
| | - Dafna D Gladman
- Krembil Research Institute, Toronto Western Hospital and University of Toronto, 60 Leonard Ave, Toronto, ON, M5T 0S8, Canada
| | - Arthur Kavanaugh
- Division of Rheumatology, Allergy, and Immunology, UC San Diego School of Medicine, 9500 Gilman Drive, La Jolla, CA, 92093, USA
| | - Philip J Mease
- Swedish Medical Center/Providence St. Joseph Health and University of Washington, 601 Broadway Suite 600, Seattle, WA, 98122, USA
| |
Collapse
|
26
|
Wu D, Griffith JF, Lam SHM, Wong PCH, Shi L, Li EK, Cheng IT, Li TK, Hung VW, Qin L, Tam LS. Progressive structural bone changes and their relationship with treatment in patients with psoriatic arthritis: a longitudinal HR-pQCT study. Arthritis Res Ther 2019; 21:265. [PMID: 31801610 PMCID: PMC6894233 DOI: 10.1186/s13075-019-2043-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Accepted: 10/28/2019] [Indexed: 12/17/2022] Open
Abstract
Background Although the short-term effects of tumor necrosis factor alpha (TNF-α) and interleukin-17A (IL-17A) inhibition on the structural changes in psoriatic arthritis (PsA) using high-resolution peripheral quantitative computed tomography (HR-pQCT) have been reported, no studies have investigated the long-term structural changes in PsA patients receiving routine care. We reported longitudinal changes of erosions and enthesiophytes using HR-pQCT and their relationship with treatments in PsA patients over a 5-year period. Methods HR-pQCT examination at the second and third metacarpal heads (MCH2 and MCH3) was performed in 60 PsA patients at baseline and after 5 years. The size of each individual lesion was quantified. Erosion and enthesiophyte progression were defined as change exceeding the smallest detectable change (SDC). Results A total of 108 bone erosions and 99 enthesiophytes were detected at baseline. Three new bone erosions but no new enthesiophytes were evident at 5 years. A significant increase in mean (±SD) erosion (0.58 ± 1.50 mm3, P < 0.001) and enthesiophyte (0.47 ± 0.76 mm3, P < 0.001) volume was observed. Erosion and enthesiophyte progression were found in 37/111 (33.3%) and 50/99 (50.5%) lesions, respectively. During this 5-year period, 26 (43%) out of the 60 patients achieved sustained Disease Activity index for PSoriatic Arthritis (DAPSA) low disease activity (LDA) (SDL group, defined as achieving DAPSA-LDA at both baseline and 5 years). Fourteen (23%) out of 60 patients received a TNF inhibitor throughout the 5-year period (TNFi group). Fewer erosions progressed (12/51 [23.5%] vs 25/60 [41.7%], P = 0.047) and the increased in enthesiophyte volume was significantly less (0.28 ± 0.67 vs 0.61 ± 0.80 mm3, P = 0.048) in the SDL group than in the non-SDL group. However, no significant difference between the TNFi and non-TNFi groups was detected in terms of the change in volume or progression of bone erosion and enthesiophyte. Conclusion Damage accrual in terms of bone erosion and enthesiophyte was observed in PsA patients over a period of 5 years despite receiving routine clinical care. Nonetheless, sustained control of disease activity may be able to prevent these bony damages.
Collapse
Affiliation(s)
- Dongze Wu
- Department of Medicine & Therapeutics, The Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China
| | - James F Griffith
- Department of Imaging and Interventional Radiology, The Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China
| | - Steven H M Lam
- Department of Medicine & Therapeutics, The Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China
| | - Priscilla C H Wong
- Department of Medicine & Therapeutics, The Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China
| | - Lin Shi
- Research Centre for Medical Image Computing, Department of Imaging and Interventional Radiology, The Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China
| | - Edmund K Li
- Department of Medicine & Therapeutics, The Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China
| | - Isaac T Cheng
- Department of Medicine & Therapeutics, The Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China
| | - Tena K Li
- Department of Medicine & Therapeutics, The Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China
| | - Vivian W Hung
- Bone Quality and Health Centre, Department of Orthopaedics and Traumatology, The Chinese University of Hong Kong, Hong Kong, China
| | - Ling Qin
- Bone Quality and Health Centre, Department of Orthopaedics and Traumatology, The Chinese University of Hong Kong, Hong Kong, China
| | - Lai-Shan Tam
- Department of Medicine & Therapeutics, The Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China.
| |
Collapse
|
27
|
McInnes IB, Chakravarty SD, Apaolaza I, Kafka S, Hsia EC, You Y, Kavanaugh A. Efficacy of ustekinumab in biologic-naïve patients with psoriatic arthritis by prior treatment exposure and disease duration: data from PSUMMIT 1 and PSUMMIT 2. RMD Open 2019; 5:e000990. [PMID: 31565242 PMCID: PMC6744084 DOI: 10.1136/rmdopen-2019-000990] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Revised: 07/15/2019] [Accepted: 07/22/2019] [Indexed: 11/03/2022] Open
Abstract
Objective To evaluate the efficacy of ustekinumab by prior treatment exposure and disease duration in tumour necrosis factor inhibitor (TNF)-naïve patients with psoriatic arthritis (PsA) in the PSUMMIT 1 and PSUMMIT 2 studies. Methods In the phase 3, randomised, placebo-controlled PSUMMIT 1 and PSUMMIT 2 studies, adults with active PsA for ≥6 months despite conventional synthetic disease-modifying antirheumatic drugs (csDMARDs) and/or non-steroidal anti-inflammatory drugs (NSAIDs) (PSUMMIT 1) or csDMARDs, NSAIDs and/or anti-TNF agents (PSUMMIT 2) were enrolled. Patients were randomised to subcutaneous injections of placebo, ustekinumab 45 mg or ustekinumab 90 mg at weeks 0 and 4 and every 12 weeks. Efficacy was assessed at week 24 using the American College of Rheumatology criteria and 28-joint count disease activity score using C reactive protein (DAS28-CRP); radiographical progression, enthesitis, and dactylitis were also assessed in this post hoc analysis. Results A total of 747 patients were included; all 747 were TNF-naïve, of which, 179 were methotrexate-naïve and TNF-naïve, and 146 were all csDMARD-naïve and TNF-naïve. At week 24, greater proportions of ustekinumab-treated patients had ≥20%/50%/70% improvement in American College of Rheumatology criteria (ACR20/ACR50/ACR70) responses, DAS28-CRP response and DAS28-CRP remission versus placebo in all three prior-treatment populations, with similar differences between treatment groups. Greater proportions of ustekinumab-treated patients also had complete resolution of enthesitis and dactylitis at week 24 across the three prior-treatment populations. Mean changes from baseline in total van der Heijde-Sharp Score at week 24 were generally smaller for ustekinumab-treated patients versus placebo but were statistically significant only in the full TNF-naïve population. Response rates for ACR20/ACR50/ACR70 were similar for TNF-naïve patients with PsA durations of <1 year, ≥1 to <3 years, and ≥3 years. Conclusion Ustekinumab-treated patients demonstrated greater clinical response at week 24 compared with placebo regardless of prior treatment exposure and PsA disease duration.
Collapse
Affiliation(s)
- Iain B McInnes
- Institute of Infection, Immunity & Inflammation, University of Glasgow, Glasgow, UK
| | - Soumya D Chakravarty
- Immunology Medical Affairs, Janssen Scientific Affairs, LLC, Horsham, Pennsylvania, USA.,Drexel University, College of Medicine, Philadelphia, Pennsylvania, USA
| | - Isabel Apaolaza
- Medical Affairs, Janssen Biologics BV, Leiden, The Netherlands
| | - Shelly Kafka
- Immunology Medical Affairs, Janssen Scientific Affairs, LLC, Horsham, Pennsylvania, USA
| | - Elizabeth C Hsia
- Immunology, Janssen Research & Development, LLC, Spring House, Pennsylvania, USA.,Division of Rheumatology, University of Pennslyvania, Philadelphia, Pennsylvania, USA
| | - Yin You
- Biostatistics, Janssen Research & Development, LLC, Spring House, Pennsylvania, USA
| | - Arthur Kavanaugh
- Department of Medicine, University of California, San Diego, School of Medicine, La Jolla, California, USA
| |
Collapse
|
28
|
Mease PJ, Gladman DD, Collier DH, Ritchlin CT, Helliwell PS, Liu L, Kricorian G, Chung JB. Etanercept and Methotrexate as Monotherapy or in Combination for Psoriatic Arthritis: Primary Results From a Randomized, Controlled Phase III Trial. Arthritis Rheumatol 2019; 71:1112-1124. [PMID: 30747501 PMCID: PMC6618246 DOI: 10.1002/art.40851] [Citation(s) in RCA: 157] [Impact Index Per Article: 26.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2018] [Accepted: 02/05/2019] [Indexed: 12/15/2022]
Abstract
Objective To examine the efficacy of methotrexate monotherapy relative to etanercept monotherapy and the value of combining methotrexate and etanercept for the treatment of patients with psoriatic arthritis (PsA). Methods In this double‐blind study, 851 patients with PsA were randomized to 1 of 3 treatment arms, as follows: oral methotrexate (20 mg) plus subcutaneous placebo given weekly (n = 284), subcutaneous etanercept (50 mg) plus oral placebo given weekly (n = 284), or subcutaneous etanercept (50 mg) plus oral methotrexate (20 mg) given weekly (combination therapy; n = 283). The American College of Rheumatology 20% improvement (ACR20) response and Minimal Disease Activity (MDA) response at week 24 were the primary end point and key secondary end point, respectively. Other measures of inflammatory arthritis, radiographic progression, and nonarticular disease manifestations were also assessed. Results Patients with PsA had a mean ± SD age of 48.4 ± 13.1 years, and the mean ± SD duration of PsA was 3.2 ± 6.3 years (median 0.6 years). ACR20 and MDA response rates at week 24 were significantly greater in patients who received etanercept monotherapy compared with those who received methotrexate monotherapy (ACR20, 60.9% versus 50.7% of patients [P = 0.029]; MDA, 35.9% versus 22.9% of patients [P = 0.005]), and both were significantly greater in the combination therapy group compared with the methotrexate monotherapy group at week 24 (ACR20, 65.0% versus 50.7% of patients [P = 0.005]; MDA, 35.7% versus 22.9% of patients [P = 0.005]). Other secondary outcomes (ACR50 and ACR70 response rates, proportions of patients achieving a Very Low Disease Activity score, and PsA disease activity scores) showed between‐group differences that were consistent with the primary and key secondary end point results. Furthermore, patients in both etanercept treatment arms showed less radiographic progression at week 48 compared with patients who received methotrexate monotherapy. Outcomes were similar in the combination therapy and etanercept monotherapy groups, except for some skin end points. No new safety signals were seen. Conclusion Etanercept monotherapy and combination therapy with etanercept and methotrexate showed greater efficacy than methotrexate monotherapy in patients with PsA, according to the ACR and MDA response rates and extent of radiographic progression at follow‐up. Overall, combining methotrexate and etanercept did not improve the efficacy of etanercept.
Collapse
Affiliation(s)
- Philip J Mease
- Swedish Medical Center/Providence St. Joseph Health and the University of Washington, Seattle, Washington
| | | | | | | | | | | | | | | |
Collapse
|
29
|
van der Heijde D, Gladman DD, FitzGerald O, Kavanaugh A, Graham D, Wang C, Fallon L. Radiographic Progression According to Baseline C-reactive Protein Levels and Other Risk Factors in Psoriatic Arthritis Treated with Tofacitinib or Adalimumab. J Rheumatol 2019; 46:1089-1096. [DOI: 10.3899/jrheum.180971] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/21/2018] [Indexed: 01/30/2023]
Abstract
Objective.To evaluate the effect of baseline risk factors on radiographic progression in patients with active psoriatic arthritis (PsA) who had an inadequate response to conventional synthetic disease-modifying antirheumatic drugs (csDMARD) and were treated with tofacitinib or adalimumab (ADA).Methods.Tofacitinib is an oral Janus kinase inhibitor for the treatment of PsA. OPAL Broaden was a 12-month, double-blind phase III trial. Patients received tofacitinib 5 mg twice daily (BID; n = 107), tofacitinib 10 mg BID (n = 104), or ADA 40 mg once every 2 weeks (n = 106), all with 1 background csDMARD. Radiographs (baseline and Month 12) were scored using the van der Heijde-modified total Sharp score (mTSS) for PsA. Radiographic nonprogression was defined as an increase from baseline in mTSS ≤ 0.5, ≤ 0, or ≤ 0.66. Changes from baseline in mTSS and nonprogression (≤ 0.5 increase from baseline in mTSS) were analyzed by baseline C-reactive protein (CRP) > 2.87 or ≤ 2.87 mg/l. Baseline predictors of radiographic progression were analyzed.Results.At Month 12, > 90% of patients receiving tofacitinib or ADA met all radiographic nonprogression criteria. Mean changes from baseline through Month 12 in mTSS, erosion, and joint space narrowing scores were close to 0. Changes in radiographic outcomes were minimal, irrespective of baseline CRP levels > 2.87 or ≤ 2.87 mg/l, with a small numerical difference observed for tofacitinib 5 mg BID. A significant relationship was observed between baseline CRP level and increases from baseline in mTSS > 0.5 at Month 12.Conclusion.Elevated CRP levels at baseline were associated with greater structural progression. Changes in radiographic outcomes were minimal regardless of CRP levels. [Clinical trial registration number (www.ClinicalTrials.gov): NCT01877668]
Collapse
|
30
|
Kavanaugh A, Husni ME, Harrison DD, Kim L, Lo KH, Noonan L, Hsia EC. Radiographic Progression Inhibition with Intravenous Golimumab in Psoriatic Arthritis: Week 24 Results of a Phase III, Randomized, Double-blind, Placebo-controlled Trial. J Rheumatol 2019; 46:595-602. [PMID: 30770519 DOI: 10.3899/jrheum.180681] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/18/2018] [Indexed: 01/08/2023]
Abstract
OBJECTIVE Evaluate effects of intravenous (IV) golimumab (GOL) on radiographic progression in psoriatic arthritis (PsA). METHODS This phase III, randomized, double-blind, placebo-controlled trial (GO-VIBRANT) randomized patients with active PsA to receive IV placebo (n = 239) or IV GOL 2 mg/kg (n = 241) at weeks 0, 4, 12, and 20. Radiographic progression (controlled secondary endpoint) was evaluated as change from baseline at Week 24 in PsA-modified total Sharp/van der Heijde scores (SvdH). The proportions of patients with a change from baseline at Week 24 in the total PsA-modified SvdH exceeding the smallest detectable change (SDC) or > 0 or 0.5 also were determined. RESULTS Overall, 474 patients (237/arm) contributed radiographic data. Results obtained from the 2 blinded, independent radiographic readers demonstrated good agreement (total score intraclass correlation coefficients: baseline = 0.93, Week 24 = 0.92, Week 24 change score = 0.73). GOL demonstrated significant inhibition of radiographic progression relative to placebo from baseline to Week 24 (mean changes in PsA-modified total SvdH: -0.36 vs 1.95; treatment difference: -2.32; p < 0.001). At Week 24, smaller proportions of GOL- versus placebo-treated patients demonstrated an increase in the total PsA-modified SvdH score exceeding the SDC (8.0% vs 27.0%, respectively; difference: -19.0%; p < 0.001), > 0 (28.3% vs 57.0%, respectively; difference: -28.7%; p < 0.001), or > 0.5 (18.6% vs 41.8%, respectively; difference: -23.2%; p < 0.001). Results were consistent for erosion and joint space narrowing scores, in hands and feet, and in patients with/without baseline concomitant methotrexate use. Prevention of radiographic progression by GOL was independent of clinical response. CONCLUSION IV GOL is significantly better than placebo in inhibiting radiographic progression of structural damage in active PsA. [Clinical trial registration number (www.ClinicalTrials.gov): NCT02181673].
Collapse
Affiliation(s)
- Arthur Kavanaugh
- From Internal Medicine - Rheumatology, University of California at San Diego, La Jolla, California; Department of Internal Medicine - Rheumatology, Cleveland Clinic, Cleveland, Ohio; Immunology, Janssen Research & Development LLC, Spring House, Pennsylvania; Rheumatology, University of Pennsylvania, Philadelphia, Pennsylvania, USA.,Drs. Harrison and Hsia, and L. Kim, K.H. Lo, and L. Noonan are employees of Janssen Research & Development LLC and own stock or stock options in Johnson & Johnson, of which Janssen Research & Development LLC is a wholly owned subsidiary.,A. Kavanaugh, MD, Internal Medicine - Rheumatology, University of California at San Diego; M.E. Husni, MD, MPH, Internal Medicine - Rheumatology, Cleveland Clinic; D.D. Harrison, MD, MPH, Immunology, Janssen Research & Development LLC; L. Kim, PhD, Immunology, Janssen Research & Development LLC; K.H. Lo, PhD, Immunology, Janssen Research & Development LLC; L. Noonan, RT(MR), Immunology, Janssen Research & Development LLC; E.C. Hsia, MD, MSCE, Immunology, Janssen Research & Development LLC, and Rheumatology, University of Pennsylvania
| | - M Elaine Husni
- From Internal Medicine - Rheumatology, University of California at San Diego, La Jolla, California; Department of Internal Medicine - Rheumatology, Cleveland Clinic, Cleveland, Ohio; Immunology, Janssen Research & Development LLC, Spring House, Pennsylvania; Rheumatology, University of Pennsylvania, Philadelphia, Pennsylvania, USA.,Drs. Harrison and Hsia, and L. Kim, K.H. Lo, and L. Noonan are employees of Janssen Research & Development LLC and own stock or stock options in Johnson & Johnson, of which Janssen Research & Development LLC is a wholly owned subsidiary.,A. Kavanaugh, MD, Internal Medicine - Rheumatology, University of California at San Diego; M.E. Husni, MD, MPH, Internal Medicine - Rheumatology, Cleveland Clinic; D.D. Harrison, MD, MPH, Immunology, Janssen Research & Development LLC; L. Kim, PhD, Immunology, Janssen Research & Development LLC; K.H. Lo, PhD, Immunology, Janssen Research & Development LLC; L. Noonan, RT(MR), Immunology, Janssen Research & Development LLC; E.C. Hsia, MD, MSCE, Immunology, Janssen Research & Development LLC, and Rheumatology, University of Pennsylvania
| | - Diane D Harrison
- From Internal Medicine - Rheumatology, University of California at San Diego, La Jolla, California; Department of Internal Medicine - Rheumatology, Cleveland Clinic, Cleveland, Ohio; Immunology, Janssen Research & Development LLC, Spring House, Pennsylvania; Rheumatology, University of Pennsylvania, Philadelphia, Pennsylvania, USA.,Drs. Harrison and Hsia, and L. Kim, K.H. Lo, and L. Noonan are employees of Janssen Research & Development LLC and own stock or stock options in Johnson & Johnson, of which Janssen Research & Development LLC is a wholly owned subsidiary.,A. Kavanaugh, MD, Internal Medicine - Rheumatology, University of California at San Diego; M.E. Husni, MD, MPH, Internal Medicine - Rheumatology, Cleveland Clinic; D.D. Harrison, MD, MPH, Immunology, Janssen Research & Development LLC; L. Kim, PhD, Immunology, Janssen Research & Development LLC; K.H. Lo, PhD, Immunology, Janssen Research & Development LLC; L. Noonan, RT(MR), Immunology, Janssen Research & Development LLC; E.C. Hsia, MD, MSCE, Immunology, Janssen Research & Development LLC, and Rheumatology, University of Pennsylvania
| | - Lilianne Kim
- From Internal Medicine - Rheumatology, University of California at San Diego, La Jolla, California; Department of Internal Medicine - Rheumatology, Cleveland Clinic, Cleveland, Ohio; Immunology, Janssen Research & Development LLC, Spring House, Pennsylvania; Rheumatology, University of Pennsylvania, Philadelphia, Pennsylvania, USA.,Drs. Harrison and Hsia, and L. Kim, K.H. Lo, and L. Noonan are employees of Janssen Research & Development LLC and own stock or stock options in Johnson & Johnson, of which Janssen Research & Development LLC is a wholly owned subsidiary.,A. Kavanaugh, MD, Internal Medicine - Rheumatology, University of California at San Diego; M.E. Husni, MD, MPH, Internal Medicine - Rheumatology, Cleveland Clinic; D.D. Harrison, MD, MPH, Immunology, Janssen Research & Development LLC; L. Kim, PhD, Immunology, Janssen Research & Development LLC; K.H. Lo, PhD, Immunology, Janssen Research & Development LLC; L. Noonan, RT(MR), Immunology, Janssen Research & Development LLC; E.C. Hsia, MD, MSCE, Immunology, Janssen Research & Development LLC, and Rheumatology, University of Pennsylvania
| | - Kim Hung Lo
- From Internal Medicine - Rheumatology, University of California at San Diego, La Jolla, California; Department of Internal Medicine - Rheumatology, Cleveland Clinic, Cleveland, Ohio; Immunology, Janssen Research & Development LLC, Spring House, Pennsylvania; Rheumatology, University of Pennsylvania, Philadelphia, Pennsylvania, USA.,Drs. Harrison and Hsia, and L. Kim, K.H. Lo, and L. Noonan are employees of Janssen Research & Development LLC and own stock or stock options in Johnson & Johnson, of which Janssen Research & Development LLC is a wholly owned subsidiary.,A. Kavanaugh, MD, Internal Medicine - Rheumatology, University of California at San Diego; M.E. Husni, MD, MPH, Internal Medicine - Rheumatology, Cleveland Clinic; D.D. Harrison, MD, MPH, Immunology, Janssen Research & Development LLC; L. Kim, PhD, Immunology, Janssen Research & Development LLC; K.H. Lo, PhD, Immunology, Janssen Research & Development LLC; L. Noonan, RT(MR), Immunology, Janssen Research & Development LLC; E.C. Hsia, MD, MSCE, Immunology, Janssen Research & Development LLC, and Rheumatology, University of Pennsylvania
| | - Lenore Noonan
- From Internal Medicine - Rheumatology, University of California at San Diego, La Jolla, California; Department of Internal Medicine - Rheumatology, Cleveland Clinic, Cleveland, Ohio; Immunology, Janssen Research & Development LLC, Spring House, Pennsylvania; Rheumatology, University of Pennsylvania, Philadelphia, Pennsylvania, USA.,Drs. Harrison and Hsia, and L. Kim, K.H. Lo, and L. Noonan are employees of Janssen Research & Development LLC and own stock or stock options in Johnson & Johnson, of which Janssen Research & Development LLC is a wholly owned subsidiary.,A. Kavanaugh, MD, Internal Medicine - Rheumatology, University of California at San Diego; M.E. Husni, MD, MPH, Internal Medicine - Rheumatology, Cleveland Clinic; D.D. Harrison, MD, MPH, Immunology, Janssen Research & Development LLC; L. Kim, PhD, Immunology, Janssen Research & Development LLC; K.H. Lo, PhD, Immunology, Janssen Research & Development LLC; L. Noonan, RT(MR), Immunology, Janssen Research & Development LLC; E.C. Hsia, MD, MSCE, Immunology, Janssen Research & Development LLC, and Rheumatology, University of Pennsylvania
| | - Elizabeth C Hsia
- From Internal Medicine - Rheumatology, University of California at San Diego, La Jolla, California; Department of Internal Medicine - Rheumatology, Cleveland Clinic, Cleveland, Ohio; Immunology, Janssen Research & Development LLC, Spring House, Pennsylvania; Rheumatology, University of Pennsylvania, Philadelphia, Pennsylvania, USA. .,Drs. Harrison and Hsia, and L. Kim, K.H. Lo, and L. Noonan are employees of Janssen Research & Development LLC and own stock or stock options in Johnson & Johnson, of which Janssen Research & Development LLC is a wholly owned subsidiary. .,A. Kavanaugh, MD, Internal Medicine - Rheumatology, University of California at San Diego; M.E. Husni, MD, MPH, Internal Medicine - Rheumatology, Cleveland Clinic; D.D. Harrison, MD, MPH, Immunology, Janssen Research & Development LLC; L. Kim, PhD, Immunology, Janssen Research & Development LLC; K.H. Lo, PhD, Immunology, Janssen Research & Development LLC; L. Noonan, RT(MR), Immunology, Janssen Research & Development LLC; E.C. Hsia, MD, MSCE, Immunology, Janssen Research & Development LLC, and Rheumatology, University of Pennsylvania.
| |
Collapse
|
31
|
Wervers K, Luime JJ, Tchetverikov I, Gerards AH, Kok MR, Appels CWY, van der Graaff WL, van Groenendael JHLM, Korswagen LA, Veris-van Dieren JJ, Hazes JMW, Vis M. Time to minimal disease activity in relation to quality of life, productivity, and radiographic damage 1 year after diagnosis in psoriatic arthritis. Arthritis Res Ther 2019; 21:25. [PMID: 30651121 PMCID: PMC6335829 DOI: 10.1186/s13075-019-1811-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Accepted: 01/02/2019] [Indexed: 11/16/2022] Open
Abstract
Background In a cohort of patients with newly diagnosed psoriatic arthritis (PsA) who received usual care, we investigated the impact of time elapsed to minimal disease activity (MDA) on health-related quality of life (HRQoL), work productivity, and radiographic damage throughout the first year after diagnosis. Methods Data collected in the Dutch southwest early PsA cohort (DEPAR) study were analyzed. These three-monthly data encompassed disease activity, HRQOL was measured with the Short Form 36 (SF36) Physical Component Scale (SF36-PCS) and Mental Component Scale, and productivity was measured with the Productivity Cost Questionnaire. Radiographic damage was scored at baseline and at 12 months with the PsA-modified Sharp/van der Heijde score. Patients were classified by time to MDA as in early (within 3 months), late (at 6–12 months), and never MDA in the first year. Results We included 296 patients who had had their 1-year outpatient visit (mean age 51 years, 53% male). Ninety-six (32%) were classified as early MDA, 78 (26%) as late MDA, and 98 (33%) as never MDA. Data of 24 patients (8%) were missing. SF36-PCS and productivity scores improved after gaining MDA, but remained low in never MDA patients. At 1 year, SF36-PCS and productivity scores were similar in early and late MDA patients. Radiographic progression rate was low and similar in all groups. Conclusion Gaining MDA was associated with considerable improvement in HRQoL and functioning, irrespective of time to first MDA. In the one third of patients not in MDA in the first year, the disease had a substantial health impact.
Collapse
Affiliation(s)
- Kim Wervers
- Department of Rheumatology, Erasmus University Medical Center, Wytemaweg 80, 3015 CN, Rotterdam, The Netherlands
| | - Jolanda J Luime
- Department of Rheumatology, Erasmus University Medical Center, Wytemaweg 80, 3015 CN, Rotterdam, The Netherlands
| | - Ilja Tchetverikov
- Department of Rheumatology, Albert Schweitzer Hospital, Dordrecht, The Netherlands
| | - Andreas H Gerards
- Department of Rheumatology, Vlietland Hospital, Schiedam, The Netherlands
| | - Marc R Kok
- Department of Rheumatology, Maasstad Hospital, Rotterdam, The Netherlands
| | | | | | | | - Lindy-Anne Korswagen
- Department of Rheumatology, Sint Franciscus Gasthuis, Rotterdam, The Netherlands
| | | | - Johanna M W Hazes
- Department of Rheumatology, Erasmus University Medical Center, Wytemaweg 80, 3015 CN, Rotterdam, The Netherlands
| | - Marijn Vis
- Department of Rheumatology, Erasmus University Medical Center, Wytemaweg 80, 3015 CN, Rotterdam, The Netherlands.
| | | |
Collapse
|
32
|
Efficacy of certolizumab pegol with and without concomitant use of disease-modifying anti-rheumatic drugs over 4 years in psoriatic arthritis patients: results from the RAPID-PsA randomized controlled trial. Clin Rheumatol 2018; 37:3285-3296. [PMID: 30191421 DOI: 10.1007/s10067-018-4227-7] [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: 05/24/2018] [Accepted: 07/20/2018] [Indexed: 12/23/2022]
Abstract
To report long-term efficacy of certolizumab pegol (CZP) treatment with and without concomitant DMARDs in patients with psoriatic arthritis (PsA). RAPID-PsA (NCT01087788) was double-blind and placebo-controlled to week 24, dose-blind to week 48, and open-label to week 216. Patients had active PsA with ≥ 1 failed DMARD. At baseline, patients were randomized 1:1:1 to CZP 200 mg every 2 weeks: CZP 400 mg every 4 weeks: placebo. CZP-randomized patients continued their dose into open-label. Observed case efficacy data are reported to week 216 for week 0 CZP-randomized patients (dose combined) with and without baseline DMARD use (DMARD+/DMARD-). Dactylitis (tenderness and ≥ 10% difference in swelling between affected and opposite digits) and enthesitis were measured using Leeds Dactylitis Index (LDI) and Leeds Enthesitis Index (LEI). 273/409 randomized patients received CZP from baseline: 199/273 (72.9%) DMARD+ and 74/273 (27.1%) DMARD- patients. 141/199 (70.9%) DMARD+ and 42/74 (56.8%) DMARD- patients completed Week 216. DMARD+ (79.7%) and 83.3% of DMARD- patients achieved ACR20 response at week 216; 79.2 and 78.1% achieved 75% improvement from baseline in Psoriasis Area and Severity Index (PASI75). High proportions of DMARD+/DMARD- patients with extra-articular manifestations at baseline reported total resolution at week 216; dactylitis 91.4% of DMARD+ and 93.3% of DMARD- patients, enthesitis 74.4% of DMARD+ and 87.5% of DMARD- patients. Long-term improvements in PsA symptoms were observed with CZP monotherapy or concomitant DMARDs, across important psoriatic disease domains, including joint disease, psoriasis, nail disease, dactylitis, and enthesitis.Trial registration: NCT01087788.
Collapse
|
33
|
Mease PJ, Kavanaugh A, Reimold A, Tahir H, Rech J, Hall S, Geusens P, Pellet P, Delicha EM, Mpofu S, Pricop L. Secukinumab in the treatment of psoriatic arthritis: efficacy and safety results through 3 years from the year 1 extension of the randomised phase III FUTURE 1 trial. RMD Open 2018; 4:e000723. [PMID: 30167329 PMCID: PMC6109799 DOI: 10.1136/rmdopen-2018-000723] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Revised: 06/28/2018] [Accepted: 07/02/2018] [Indexed: 12/14/2022] Open
Abstract
OBJECTIVE To assess the long-term (3 year) efficacy and safety of secukinumab in patients with active psoriatic arthritis (PsA) in the extension phase of the FUTURE 1 study (NCT01892436). METHODS Following the 2-year core trial, eligible patients receiving subcutaneous secukinumab 150 or 75 mg entered a 3-year extension phase. Results are presented for key efficacy and safety endpoints at week 156. RESULTS In total, 460 patients entered the extension study; 308 patients originally randomised to secukinumab were assessed for efficacy. Sustained improvements in all efficacy endpoints were achieved with secukinumab through week 156. Overall, 76.8%/54.9% (secukinumab 150 mg) and 65.2%/39.0% (secukinumab 75 mg) of patients achieved an American College of Rheumatology (ACR) 20/50 response (multiple imputation data); ACR20 responses were sustained irrespective of previous anti-tumour necrosis factor exposure. Improvements in quality of life and physical function were also sustained through week 156. Radiographic results (observed data; van der Heijde modified total Sharp score (mTSS)) showed that 78.1% (secukinumab 150 mg) and 74.8% (secukinumab 75 mg) of patients had no radiographic progression (≤0.5 increase in mTSS) through week 156. Exposure-adjusted incidence rates for selected adverse events per 100 patient-years (secukinumab 150/75 mg) were serious infections (1.7/1.6), Candida infections (1.4/0.7), Crohn's disease (0/0.3), ulcerative colitis (0/0.3) and major adverse cardiac events (0.3/0.8). CONCLUSION Subcutaneous secukinumab provided sustained improvements in the signs and symptoms, quality of life and physical function of patients with active PsA with low rate of radiographic disease progression through 3 years. Secukinumab was well tolerated with no new safety signals.
Collapse
Affiliation(s)
- Philip J Mease
- Swedish Medical Centre and University of Washington, Seattle, Washington, USA
| | - Arthur Kavanaugh
- University of California San Diego School of Medicine, La Jolla, California, USA
| | - Andreas Reimold
- Dallas VAMC and University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | | | - Jürgen Rech
- University of Erlangen-Nuremberg, Erlangen, Germany
| | - Stephen Hall
- Monash University, Melbourne, Victoria, Australia
| | - Piet Geusens
- University of Hasselt, Hasselt, Belgium
- Maastricht University Hospital, Maastricht, The Netherlands
| | | | | | | | - Luminita Pricop
- Novartis Pharmaceuticals Corporation, East Hanover, New Jersey, USA
| |
Collapse
|
34
|
van der Heijde D, Deodhar A, FitzGerald O, Fleischmann R, Gladman D, Gottlieb AB, Hoepken B, Bauer L, Irvin-Sellers O, Khraishi M, Peterson L, Turkiewicz A, Wollenhaupt J, Mease PJ. 4-year results from the RAPID-PsA phase 3 randomised placebo-controlled trial of certolizumab pegol in psoriatic arthritis. RMD Open 2018; 4:e000582. [PMID: 29556416 PMCID: PMC5856919 DOI: 10.1136/rmdopen-2017-000582] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2017] [Revised: 01/31/2018] [Accepted: 02/15/2018] [Indexed: 11/30/2022] Open
Abstract
Objective To report the efficacy, patient-reported, radiographic and safety outcomes of 4 years’ certolizumab pegol (CZP) treatment in patients with psoriatic arthritis (PsA). Methods RAPID-PsA (NCT01087788) was double-blind and placebo-controlled to Week 24, dose-blind to Week 48 and open-label (OL) to Week 216. Patients were randomised 1:1:1 to either placebo or CZP 200 mg every 2 weeks (Q2W) or 400 mg every 4 weeks (Q4W) (following 400 mg at Weeks 0/2/4). Patients randomised to CZP continued their assigned dose in the OL period. Patients randomised to placebo were re-randomised to CZP 200 mg Q2W or 400 mg Q4W (post-loading dose) at Week 16 (early escape) or after the double-blind phase. We present observed and imputed data; missing values were imputed using non-responder imputation (NRI) for categorical and last observation carried forward (LOCF) for continuous measures. Results 409 patients were randomised; 20% (54/273) of Week 0 patients randomised to CZP had prior anti-tumour necrosis factor (TNF) exposure; 67% (183/273) completed 216 weeks. By Week 48, 60.4% of patients achieved Disease Activity Index for Psoriatic Arthritis low disease activity or remission, which was maintained; 66.3% achieved these outcomes at Week 216 (NRI). At Weeks 48 and 216, 39.2% of patients achieved minimal disease activity (NRI). 75% reduction in Psoriasis Area and Severity Index responses were 65% and 52% at Weeks 48 and 216 (NRI). Total resolution rates for enthesitis, dactylitis and nail psoriasis, at 4 years, were 71%, 81% and 65%, respectively (LOCF). Structural damage progression was low over 4 years’ treatment. No new safety signals were identified after Week 96. Conclusions CZP efficacy in treating PsA was maintained over 4 years, in patients both with and without prior anti-TNF exposure, with no new safety signals identified.
Collapse
Affiliation(s)
| | - Atul Deodhar
- Oregon Health and Science University, Portland, Oregon, USA
| | - Oliver FitzGerald
- Department of Rheumatology, St Vincent's University Hospital and Conway Institute for Biomolecular Research, University College Dublin, Dublin, Ireland
| | - Roy Fleischmann
- Metroplex Clinical Research Center, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Dafna Gladman
- Centre for Prognosis Studies in the Rheumatic Diseases and Krembil Research Institute, Toronto Western Hospital, Toronto, Ontario, Canada
| | - Alice B Gottlieb
- Department of Dermatology, New York Medical College, Metropolitan Hospital, New York City, New York, USA
| | | | | | | | - Majed Khraishi
- Department of Medicine, Memorial University of Newfoundland, St John's, Canada
| | | | - Anthony Turkiewicz
- Rheumatology Associates Clinical Research Unit, Birmingham, Alabama, USA
| | | | - Philip J Mease
- Swedish Medical Center and University of Washington, Seattle, Washington, USA
| |
Collapse
|
35
|
Coates LC, Kishimoto M, Gottlieb A, Shuler CL, Lin CY, Lee CH, Mease PJ. Ixekizumab efficacy and safety with and without concomitant conventional disease-modifying antirheumatic drugs (cDMARDs) in biologic DMARD (bDMARD)-naïve patients with active psoriatic arthritis (PsA): results from SPIRIT-P1. RMD Open 2017; 3:e000567. [PMID: 29299340 PMCID: PMC5743900 DOI: 10.1136/rmdopen-2017-000567] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2017] [Revised: 10/16/2017] [Accepted: 11/05/2017] [Indexed: 11/09/2022] Open
Abstract
Objective To evaluate the efficacy and safety of ixekizumab alone or with concomitant conventional disease-modifying antirheumatic drugs (cDMARDs) versus placebo in patients with active psoriatic arthritis (PsA) as part of a SPIRIT-P1 subgroup analysis (NCT01695239). Methods Patients were stratified by cDMARD use (concomitant cDMARDs use (including methotrexate) or none (past or naïve use)) and randomly assigned to treatment groups (ixekizumab 80 mg every 4 weeks (IXEQ4W) or every 2 weeks (IXEQ2W) or placebo). Efficacy was evaluated versus placebo at week 24 by the American College of Rheumatology criteria (ACR20/50/70), modified total Sharp score and Health Assessment Questionnaire-Disability Index (HAQ-DI). Safety was assessed according to cDMARD status. Results Regardless of concomitant cDMARD usage, ACR20, ACR50 and ACR70 response rates were significantly higher versus placebo with IXEQ4W and IXEQ2W. The proportion of patients achieving HAQ-DI minimal clinically important difference was significantly higher versus placebo with IXEQ4W with concomitant cDMARD use and IXEQ2W, regardless of concomitant cDMARD use. Treatment-emergent adverse events (AE) were more frequent versus placebo for either ixekizumab-dosing regimen, regardless of concomitant cDMARD use. Serious AEs were not higher versus placebo, regardless of concomitant cDMARD use. Conclusion Ixekizumab treatment improved measures of disease activity and physical function in patients with active PsA relative to placebo, when used with or without concomitant cDMARD therapy.
Collapse
Affiliation(s)
- Laura C Coates
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Leeds, UK.,Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, UK
| | | | - Alice Gottlieb
- Department of Dermatology, New York Medical College at Metropolitan Hospital, New York, New York, USA
| | | | - Chen-Yen Lin
- Eli Lilly and Company, Indianapolis, Indiana, USA
| | | | - Philip J Mease
- Department of Rheumatology, Swedish Medical Center and University of Washington, Seattle, Washington, USA
| |
Collapse
|
36
|
van der Heijde D, Gladman DD, Kishimoto M, Okada M, Rathmann SS, Moriarty SR, Shuler CL, Carlier H, Benichou O, Mease PJ. Efficacy and Safety of Ixekizumab in Patients with Active Psoriatic Arthritis: 52-week Results from a Phase III Study (SPIRIT-P1). J Rheumatol 2017; 45:367-377. [DOI: 10.3899/jrheum.170429] [Citation(s) in RCA: 76] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/20/2017] [Indexed: 01/30/2023]
Abstract
Objective.To evaluate the efficacy and safety of ixekizumab (IXE), an interleukin 17A antagonist, in patients with psoriatic arthritis (PsA) after 52 weeks in a phase III study.Methods.Patients were initially randomly assigned to IXE 80 mg every 2 weeks (IXEQ2W) or every 4 weeks (IXEQ4W) after a 160-mg starting dose, placebo (PBO), or adalimumab (ADA) 40 mg Q2W. At Week 24 (Week 16 for inadequate responders), ADA (8-week washout before starting IXE) and PBO patients were rerandomized to IXEQ2W or IXEQ4W. Six treatment groups were evaluated in the extension period (weeks 24–52): IXEQ2W/IXEQ2W, IXEQ4W/IXEQ4W, ADA/IXEQ2W, ADA/IXEQ4W, PBO/IXEQ2W, and PBO/IXEQ4W. The extension period population (EPP) included patients who received ≥ 1 dose of study medication during the extension period.Results.There were 381/417 (91.4%) patients who entered the extension period. In the IXEQ4W/IXEQ4W and IXEQ2W/IXEQ2W groups (EPP), respectively, American College of Rheumatology (ACR)20 (69.1% and 68.8%), ACR50 (54.6% and 53.1%), and ACR70 (39.2% and 39.6%) response rates were sustained at Week 52. Patients rerandomized to IXE also demonstrated efficacy measured by ACR response rates at Week 52. A similar pattern was observed for Psoriasis Area and Severity Index outcomes. Radiographic progression in all 6 groups was minimal. The most frequently reported treatment-emergent adverse events (≥ 4%) were nasopharyngitis, injection site reaction, injection site erythema, upper respiratory tract infection, and back pain. No deaths were reported, and serious adverse event frequency was 0–4% with IXE.Conclusion.During the extension period, IXEQ4W or IXEQ2W treatment demonstrated sustained efficacy in key PsA domains with a safety profile consistent with other studies investigating IXE. Clinical trial number: NCT01695239; EudraCT 2011-002326-49.
Collapse
|
37
|
Kerschbaumer A, Baker D, Smolen JS, Aletaha D. The effects of structural damage on functional disability in psoriatic arthritis. Ann Rheum Dis 2017; 76:2038-2045. [PMID: 28835465 DOI: 10.1136/annrheumdis-2017-211433] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2017] [Revised: 07/28/2017] [Accepted: 07/31/2017] [Indexed: 11/04/2022]
Abstract
BACKGROUND Functional outcomes are central in patients with chronic inflammatory musculoskeletal diseases. In a secondary data analysis of the GO-REVEAL trial (NCT00265096), we investigated wether structural damage is linked to functional impairment in patients with psoriatic arthritis (PsA), a link that is still elusive in this disease. METHODS We analysed 363 patients enrolled in the GO-REVEAL study and obtained modified Sharp/van der Heijde Scores (mSvdHS) from X-rays performed at baseline, after 24, 52 and 104 weeks. Using longitudinal analyses, we assessed the effect of total mSvdHS (and its subscores, joint space narrowing (JSN) and erosions (ERO)) on functional status (measured by the Health Assessment Questionnaire) in all patients and in those attaining remission (n=117). Furthermore, we analysed wether structural damage reduces the responsiveness of functional limitations to treatment in a subgroup of responders who had functional impairment at baseline (n=67). Additionally, internal and external validation analyses were performed. RESULTS The effect of damage on function was seen in the disease activity-adjusted models using total mSvdHS (p=0.005), JSN (p=0.019) and ERO (p=0.001) as well as in the remission analyses for mSvdHS (p=0.029) and JSN (p=0.010), respectively. Functional responsiveness was limited by increasing total mSvdHS (p=0.010), JSN (p=0.002) and ERO (p=0.040). The results were validated using other functional outcomes and in an independent clinical cohort. CONCLUSIONS In PsA, structural damage, particularly JSN, has implications for physical function. Functional outcomes have an irreversible component that is strongly related to the extent of joint destruction. This needs to be considered when targeting functional outcomes in clinical practice.
Collapse
Affiliation(s)
- Andreas Kerschbaumer
- Department of Internal Medicine III, Division of Rheumatology, Medical University of Vienna, Vienna, Austria
| | - Daniel Baker
- Janssen Research & Development, LLC, Spring House, Pennsylvania, USA
| | - Josef S Smolen
- Department of Internal Medicine III, Division of Rheumatology, Medical University of Vienna, Vienna, Austria
| | - Daniel Aletaha
- Department of Internal Medicine III, Division of Rheumatology, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
38
|
Kavanaugh A, Husni ME, Harrison DD, Kim L, Lo KH, Leu JH, Hsia EC. Safety and Efficacy of Intravenous Golimumab in Patients With Active Psoriatic Arthritis: Results Through Week Twenty-Four of the GO-VIBRANT Study. Arthritis Rheumatol 2017; 69:2151-2161. [PMID: 28805045 PMCID: PMC5765449 DOI: 10.1002/art.40226] [Citation(s) in RCA: 84] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2017] [Accepted: 08/03/2017] [Indexed: 01/12/2023]
Abstract
OBJECTIVE To evaluate the safety and efficacy of intravenous (IV) golimumab treatment in psoriatic arthritis (PsA). METHODS In this phase III, randomized, double-blind, placebo-controlled trial, patients were randomly assigned to receive IV placebo (n = 239) or golimumab at 2 mg/kg (n = 241) at weeks 0, 4, 12, and 20. The primary end point was the proportion of patients meeting the American College of Rheumatology 20% improvement criteria (achieving an ACR20 response) at week 14. Controlled secondary end points included change from baseline in Health Assessment Questionnaire disability index (HAQ DI) score at week 14, proportions of patients with ACR50 and ACR70 responses and ≥75% improvement on the Psoriasis Area and Severity Index (a PASI75 response) at week 14, and change from baseline at week 24 in the total modified Sharp/van der Heijde score (SHS) with modifications for patients with PsA. RESULTS At week 14, an ACR20 response was achieved by 75.1% of patients in the golimumab group compared with 21.8% of patients in the placebo group (P < 0.001). Greater proportions of golimumab-treated patients had an ACR50 response (43.6% versus 6.3%), an ACR70 response (24.5% versus 2.1%), and a PASI75 response (59.2% versus 13.6%) at week 14 (P < 0.001 for all). Patients in the golimumab group had greater mean changes at week 14 in HAQ DI score (-0.60 versus -0.12; P < 0.001). At week 24, the mean change in total PsA-modified SHS was -0.4 in the golimumab group and 2.0 in the placebo group (P < 0.001). Through week 24, 40.6% of patients in the placebo group and 46.3% of patients in the golimumab group had ≥1 adverse event (AE); infections were the most common type. CONCLUSION Patients receiving IV golimumab at 2 mg/kg had significantly greater improvements in the signs and symptoms of PsA and less radiographic progression through week 24. AEs were consistent with those seen with other anti-tumor necrosis factor agents.
Collapse
Affiliation(s)
| | | | | | - Lilianne Kim
- Janssen Research & Development, LLCSpring HousePennsylvania
| | - Kim Hung Lo
- Janssen Research & Development, LLCSpring HousePennsylvania
| | - Jocelyn H. Leu
- Janssen Research & Development, LLCSpring HousePennsylvania
| | - Elizabeth C. Hsia
- Janssen Research & Development, LLC, Spring House, Pennsylvania, and University of PennsylvaniaPhiladelphia
| |
Collapse
|
39
|
Mease P, Hall S, FitzGerald O, van der Heijde D, Merola JF, Avila-Zapata F, Cieślak D, Graham D, Wang C, Menon S, Hendrikx T, Kanik KS. Tofacitinib or Adalimumab versus Placebo for Psoriatic Arthritis. N Engl J Med 2017; 377:1537-1550. [PMID: 29045212 DOI: 10.1056/nejmoa1615975] [Citation(s) in RCA: 400] [Impact Index Per Article: 50.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
BACKGROUND Tofacitinib is an oral Janus kinase inhibitor that is under investigation for the treatment of psoriatic arthritis. We evaluated tofacitinib in patients with active psoriatic arthritis who previously had an inadequate response to conventional synthetic disease-modifying antirheumatic drugs (DMARDs). METHODS In this 12-month, double-blind, active-controlled and placebo-controlled, phase 3 trial, we randomly assigned patients in a 2:2:2:1:1 ratio to receive one of the following regimens: tofacitinib at a 5-mg dose taken orally twice daily (107 patients), tofacitinib at a 10-mg dose taken orally twice daily (104), adalimumab at a 40-mg dose administered subcutaneously once every 2 weeks (106), placebo with a blinded switch to the 5-mg tofacitinib dose at 3 months (52), or placebo with a blinded switch to the 10-mg tofacitinib dose at 3 months (53). Placebo groups were pooled for analyses up to month 3. Primary end points were the proportion of patients who had an American College of Rheumatology 20 (ACR20) response (≥20% improvement from baseline in the number of tender and swollen joints and at least three of five other important domains) at month 3 and the change from baseline in the Health Assessment Questionnaire-Disability Index (HAQ-DI) score (scores range from 0 to 3, with higher scores indicating greater disability) at month 3. RESULTS ACR20 response rates at month 3 were 50% in the 5-mg tofacitinib group and 61% in the 10-mg tofacitinib group, as compared with 33% in the placebo group (P=0.01 for the comparison of the 5-mg dose with placebo; P<0.001 for the comparison of the 10-mg dose with placebo); the rate was 52% in the adalimumab group. The mean change in the HAQ-DI score was -0.35 in the 5-mg tofacitinib group and -0.40 in the 10-mg tofacitinib group, as compared with -0.18 in the placebo group (P=0.006 for the comparison of the 5-mg dose with placebo; P<0.001 for the comparison of the 10-mg dose with placebo); the score change was -0.38 in the adalimumab group. The rate of adverse events through month 12 was 66% in the 5-mg tofacitinib group, 71% in the 10-mg tofacitinib group, 72% in the adalimumab group, 69% in the placebo group that switched to the 5-mg tofacitinib dose, and 64% in the placebo group that switched to the 10-mg tofacitinib dose. There were four cases of cancer, three serious infections, and four cases of herpes zoster in patients who received tofacitinib during the trial. CONCLUSIONS The efficacy of tofacitinib was superior to that of placebo at month 3 in patients with psoriatic arthritis who had previously had an inadequate response to conventional synthetic DMARDs. Adverse events were more frequent with tofacitinib than with placebo. (Funded by Pfizer; OPAL Broaden ClinicalTrials.gov number, NCT01877668 .).
Collapse
Affiliation(s)
- Philip Mease
- From the Swedish Medical Center and University of Washington, Seattle (P.M.); Cabrini Health and Monash University, Melbourne, VIC, Australia (S.H.); the Department of Rheumatology, St. Vincent's University Hospital and Conway Institute of Biomolecular and Biomedical Research, University College, Dublin (O.F.); Leiden University Medical Center, Leiden, the Netherlands (D.H.); Brigham and Women's Hospital, Harvard Medical School, Boston (J.F.M.); Centro Multidisciplinario para el Desarrollo Especializado de la Investigación Clínica en Yucatán Sociedad Civil Privada, Merida, Mexico (F.A.-Z.); the Department of Rheumatology and Clinical Immunology, Medical University of Poznan, Poznan, Poland (D.C.); Pfizer, Groton, CT (D.G., C.W., S.M., K.S.K.); and Pfizer, Collegeville, PA (T.H.)
| | - Stephen Hall
- From the Swedish Medical Center and University of Washington, Seattle (P.M.); Cabrini Health and Monash University, Melbourne, VIC, Australia (S.H.); the Department of Rheumatology, St. Vincent's University Hospital and Conway Institute of Biomolecular and Biomedical Research, University College, Dublin (O.F.); Leiden University Medical Center, Leiden, the Netherlands (D.H.); Brigham and Women's Hospital, Harvard Medical School, Boston (J.F.M.); Centro Multidisciplinario para el Desarrollo Especializado de la Investigación Clínica en Yucatán Sociedad Civil Privada, Merida, Mexico (F.A.-Z.); the Department of Rheumatology and Clinical Immunology, Medical University of Poznan, Poznan, Poland (D.C.); Pfizer, Groton, CT (D.G., C.W., S.M., K.S.K.); and Pfizer, Collegeville, PA (T.H.)
| | - Oliver FitzGerald
- From the Swedish Medical Center and University of Washington, Seattle (P.M.); Cabrini Health and Monash University, Melbourne, VIC, Australia (S.H.); the Department of Rheumatology, St. Vincent's University Hospital and Conway Institute of Biomolecular and Biomedical Research, University College, Dublin (O.F.); Leiden University Medical Center, Leiden, the Netherlands (D.H.); Brigham and Women's Hospital, Harvard Medical School, Boston (J.F.M.); Centro Multidisciplinario para el Desarrollo Especializado de la Investigación Clínica en Yucatán Sociedad Civil Privada, Merida, Mexico (F.A.-Z.); the Department of Rheumatology and Clinical Immunology, Medical University of Poznan, Poznan, Poland (D.C.); Pfizer, Groton, CT (D.G., C.W., S.M., K.S.K.); and Pfizer, Collegeville, PA (T.H.)
| | - Désirée van der Heijde
- From the Swedish Medical Center and University of Washington, Seattle (P.M.); Cabrini Health and Monash University, Melbourne, VIC, Australia (S.H.); the Department of Rheumatology, St. Vincent's University Hospital and Conway Institute of Biomolecular and Biomedical Research, University College, Dublin (O.F.); Leiden University Medical Center, Leiden, the Netherlands (D.H.); Brigham and Women's Hospital, Harvard Medical School, Boston (J.F.M.); Centro Multidisciplinario para el Desarrollo Especializado de la Investigación Clínica en Yucatán Sociedad Civil Privada, Merida, Mexico (F.A.-Z.); the Department of Rheumatology and Clinical Immunology, Medical University of Poznan, Poznan, Poland (D.C.); Pfizer, Groton, CT (D.G., C.W., S.M., K.S.K.); and Pfizer, Collegeville, PA (T.H.)
| | - Joseph F Merola
- From the Swedish Medical Center and University of Washington, Seattle (P.M.); Cabrini Health and Monash University, Melbourne, VIC, Australia (S.H.); the Department of Rheumatology, St. Vincent's University Hospital and Conway Institute of Biomolecular and Biomedical Research, University College, Dublin (O.F.); Leiden University Medical Center, Leiden, the Netherlands (D.H.); Brigham and Women's Hospital, Harvard Medical School, Boston (J.F.M.); Centro Multidisciplinario para el Desarrollo Especializado de la Investigación Clínica en Yucatán Sociedad Civil Privada, Merida, Mexico (F.A.-Z.); the Department of Rheumatology and Clinical Immunology, Medical University of Poznan, Poznan, Poland (D.C.); Pfizer, Groton, CT (D.G., C.W., S.M., K.S.K.); and Pfizer, Collegeville, PA (T.H.)
| | - Francisco Avila-Zapata
- From the Swedish Medical Center and University of Washington, Seattle (P.M.); Cabrini Health and Monash University, Melbourne, VIC, Australia (S.H.); the Department of Rheumatology, St. Vincent's University Hospital and Conway Institute of Biomolecular and Biomedical Research, University College, Dublin (O.F.); Leiden University Medical Center, Leiden, the Netherlands (D.H.); Brigham and Women's Hospital, Harvard Medical School, Boston (J.F.M.); Centro Multidisciplinario para el Desarrollo Especializado de la Investigación Clínica en Yucatán Sociedad Civil Privada, Merida, Mexico (F.A.-Z.); the Department of Rheumatology and Clinical Immunology, Medical University of Poznan, Poznan, Poland (D.C.); Pfizer, Groton, CT (D.G., C.W., S.M., K.S.K.); and Pfizer, Collegeville, PA (T.H.)
| | - Dorota Cieślak
- From the Swedish Medical Center and University of Washington, Seattle (P.M.); Cabrini Health and Monash University, Melbourne, VIC, Australia (S.H.); the Department of Rheumatology, St. Vincent's University Hospital and Conway Institute of Biomolecular and Biomedical Research, University College, Dublin (O.F.); Leiden University Medical Center, Leiden, the Netherlands (D.H.); Brigham and Women's Hospital, Harvard Medical School, Boston (J.F.M.); Centro Multidisciplinario para el Desarrollo Especializado de la Investigación Clínica en Yucatán Sociedad Civil Privada, Merida, Mexico (F.A.-Z.); the Department of Rheumatology and Clinical Immunology, Medical University of Poznan, Poznan, Poland (D.C.); Pfizer, Groton, CT (D.G., C.W., S.M., K.S.K.); and Pfizer, Collegeville, PA (T.H.)
| | - Daniela Graham
- From the Swedish Medical Center and University of Washington, Seattle (P.M.); Cabrini Health and Monash University, Melbourne, VIC, Australia (S.H.); the Department of Rheumatology, St. Vincent's University Hospital and Conway Institute of Biomolecular and Biomedical Research, University College, Dublin (O.F.); Leiden University Medical Center, Leiden, the Netherlands (D.H.); Brigham and Women's Hospital, Harvard Medical School, Boston (J.F.M.); Centro Multidisciplinario para el Desarrollo Especializado de la Investigación Clínica en Yucatán Sociedad Civil Privada, Merida, Mexico (F.A.-Z.); the Department of Rheumatology and Clinical Immunology, Medical University of Poznan, Poznan, Poland (D.C.); Pfizer, Groton, CT (D.G., C.W., S.M., K.S.K.); and Pfizer, Collegeville, PA (T.H.)
| | - Cunshan Wang
- From the Swedish Medical Center and University of Washington, Seattle (P.M.); Cabrini Health and Monash University, Melbourne, VIC, Australia (S.H.); the Department of Rheumatology, St. Vincent's University Hospital and Conway Institute of Biomolecular and Biomedical Research, University College, Dublin (O.F.); Leiden University Medical Center, Leiden, the Netherlands (D.H.); Brigham and Women's Hospital, Harvard Medical School, Boston (J.F.M.); Centro Multidisciplinario para el Desarrollo Especializado de la Investigación Clínica en Yucatán Sociedad Civil Privada, Merida, Mexico (F.A.-Z.); the Department of Rheumatology and Clinical Immunology, Medical University of Poznan, Poznan, Poland (D.C.); Pfizer, Groton, CT (D.G., C.W., S.M., K.S.K.); and Pfizer, Collegeville, PA (T.H.)
| | - Sujatha Menon
- From the Swedish Medical Center and University of Washington, Seattle (P.M.); Cabrini Health and Monash University, Melbourne, VIC, Australia (S.H.); the Department of Rheumatology, St. Vincent's University Hospital and Conway Institute of Biomolecular and Biomedical Research, University College, Dublin (O.F.); Leiden University Medical Center, Leiden, the Netherlands (D.H.); Brigham and Women's Hospital, Harvard Medical School, Boston (J.F.M.); Centro Multidisciplinario para el Desarrollo Especializado de la Investigación Clínica en Yucatán Sociedad Civil Privada, Merida, Mexico (F.A.-Z.); the Department of Rheumatology and Clinical Immunology, Medical University of Poznan, Poznan, Poland (D.C.); Pfizer, Groton, CT (D.G., C.W., S.M., K.S.K.); and Pfizer, Collegeville, PA (T.H.)
| | - Thijs Hendrikx
- From the Swedish Medical Center and University of Washington, Seattle (P.M.); Cabrini Health and Monash University, Melbourne, VIC, Australia (S.H.); the Department of Rheumatology, St. Vincent's University Hospital and Conway Institute of Biomolecular and Biomedical Research, University College, Dublin (O.F.); Leiden University Medical Center, Leiden, the Netherlands (D.H.); Brigham and Women's Hospital, Harvard Medical School, Boston (J.F.M.); Centro Multidisciplinario para el Desarrollo Especializado de la Investigación Clínica en Yucatán Sociedad Civil Privada, Merida, Mexico (F.A.-Z.); the Department of Rheumatology and Clinical Immunology, Medical University of Poznan, Poznan, Poland (D.C.); Pfizer, Groton, CT (D.G., C.W., S.M., K.S.K.); and Pfizer, Collegeville, PA (T.H.)
| | - Keith S Kanik
- From the Swedish Medical Center and University of Washington, Seattle (P.M.); Cabrini Health and Monash University, Melbourne, VIC, Australia (S.H.); the Department of Rheumatology, St. Vincent's University Hospital and Conway Institute of Biomolecular and Biomedical Research, University College, Dublin (O.F.); Leiden University Medical Center, Leiden, the Netherlands (D.H.); Brigham and Women's Hospital, Harvard Medical School, Boston (J.F.M.); Centro Multidisciplinario para el Desarrollo Especializado de la Investigación Clínica en Yucatán Sociedad Civil Privada, Merida, Mexico (F.A.-Z.); the Department of Rheumatology and Clinical Immunology, Medical University of Poznan, Poznan, Poland (D.C.); Pfizer, Groton, CT (D.G., C.W., S.M., K.S.K.); and Pfizer, Collegeville, PA (T.H.)
| |
Collapse
|
40
|
Mease PJ, Gottlieb AB, van der Heijde D, FitzGerald O, Johnsen A, Nys M, Banerjee S, Gladman DD. Efficacy and safety of abatacept, a T-cell modulator, in a randomised, double-blind, placebo-controlled, phase III study in psoriatic arthritis. Ann Rheum Dis 2017; 76:1550-1558. [PMID: 28473423 PMCID: PMC5561378 DOI: 10.1136/annrheumdis-2016-210724] [Citation(s) in RCA: 166] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2016] [Revised: 03/16/2017] [Accepted: 03/29/2017] [Indexed: 12/19/2022]
Abstract
OBJECTIVES To assess the efficacy and safety of abatacept, a selective T-cell costimulation modulator, in a phase III study in psoriatic arthritis (PsA). METHODS This study randomised patients (1:1) with active PsA (~60% with prior exposure to a tumour necrosis factor inhibitor) to blinded weekly subcutaneous abatacept 125 mg (n=213) or placebo (n=211) for 24 weeks, followed by open-label subcutaneous abatacept. Patients without ≥20% improvement in joint counts at week 16 were switched to open-label abatacept. The primary end point was the proportion of patients with ≥20% improvement in the American College of Rheumatology (ACR20) criteria at week 24. RESULTS Abatacept significantly increased ACR20 response versus placebo at week 24 (39.4% vs 22.3%; p<0.001). Although abatacept numerically increased Health Assessment Questionnaire-Disability Index response rates (reduction from baseline ≥0.35) at week 24, this was not statistically significant (31.0% vs 23.7%; p=0.097). The benefits of abatacept were seen in ACR20 responses regardless of tumour necrosis factor inhibitor exposure and in other musculoskeletal manifestations, but significance could not be attributed due to ranking below Health Assessment Questionnaire-Disability Index response in hierarchical testing. However, the benefit on psoriasis lesions was modest. Efficacy was maintained or improved up to week 52. Abatacept was well tolerated with no new safety signals. CONCLUSIONS Abatacept treatment of PsA in this phase III study achieved its primary end point, ACR20 response, showed beneficial trends overall in musculoskeletal manifestations and was well tolerated. There was only a modest impact on psoriasis lesions. TRIAL REGISTRATION NUMBER ClinicalTrials.gov number, NCT01860976 (funded by Bristol-Myers Squibb).
Collapse
Affiliation(s)
- Philip J Mease
- Swedish Medical Center and University of Washington, Seattle, Washington, USA
| | | | | | - Oliver FitzGerald
- St Vincent’s University Hospital and University College Dublin, Dublin, Ireland
| | | | - Marleen Nys
- Bristol-Myers Squibb, Braine-l’Alleud, Belgium
| | | | - Dafna D Gladman
- University of Toronto and Toronto Western Hospital, Toronto, Canada
| |
Collapse
|
41
|
van der Heijde D, Landewé RB, Mease PJ, McInnes IB, Conaghan PG, Pricop L, Ligozio G, Richards HB, Mpofu S. Brief Report: Secukinumab Provides Significant and Sustained Inhibition of Joint Structural Damage in a Phase III Study of Active Psoriatic Arthritis. Arthritis Rheumatol 2017; 68:1914-21. [PMID: 27014997 PMCID: PMC5129532 DOI: 10.1002/art.39685] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2015] [Accepted: 03/15/2016] [Indexed: 01/12/2023]
Abstract
OBJECTIVE To assess whether secukinumab treatment in patients with active psoriatic arthritis (PsA) is associated with sustained inhibition of radiographic progression. METHODS In this phase III, double-blind, placebo-controlled study, 606 patients with PsA were randomized to receive intravenous (IV) secukinumab at a dose of 10 mg/kg (weeks 0, 2, 4) followed by subcutaneous secukinumab at a dose of 150 mg or 75 mg (the IV→150 mg and IV→75 mg groups, respectively) or placebo. Patients were stratified according to prior anti-tumor necrosis factor (anti-TNF) exposure (71% were anti-TNF naive). At week 16, placebo-treated patients who had at least a 20% reduction in the tender and swollen joint counts (responders) continued to receive placebo until week 24; nonresponders were re-randomized to receive secukinumab at a dose of 150 mg or 75 mg. The modified total Sharp/van der Heijde score (SHS) was determined at baseline, week 16 or 24, and week 52. RESULTS In the overall population, radiographic progression was inhibited through 52 weeks; efficacy was demonstrated for both erosion and joint space narrowing scores and in patients who switched from placebo to secukinumab at week 24. Subgroup analyses showed that secukinumab reduced radiographic progression at week 24, regardless of previous anti-TNF treatment. Among anti-TNF-naive patients, the mean changes from baseline to week 24 in the modified total SHS were 0.05 in the pooled secukinumab group and 0.57 in the placebo group; among patients with an inadequate response or intolerance to anti-TNF treatment, the mean changes were 0.16 and 0.58, respectively. Anti-TNF-naive patients showed negligible progression through week 52. Inhibition of structural damage was observed through week 52 irrespective of concomitant methotrexate use. A high proportion of patients receiving secukinumab showed no progression (change in SHS of ≤ 0.5) from baseline to week 24 (82.3% of the IV→150 mg group and 92.3% of the IV→75 mg group) and from week 24 to week 52 (85.7% of the IV→150 mg group and 85.8% of the IV→75 mg group). CONCLUSION Secukinumab inhibited radiographic progression over 52 weeks of treatment in patients with active PsA.
Collapse
Affiliation(s)
| | - Robert B Landewé
- Robert B. Landewé, MD: Academic Medical Centre, University of Amsterdam, Amsterdam, The Netherlands
| | - Philip J Mease
- Swedish Medical Center and University of Washington, Seattle
| | - Iain B McInnes
- Institute of Infection, Immunity, and Inflammation, University of Glasgow, Glasgow, UK
| | - Philip G Conaghan
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, and NIHR Leeds Musculoskeletal Biomedical Research Unit, Leeds, UK
| | | | - Greg Ligozio
- Novartis Pharmaceuticals, East Hanover, New Jersey
| | | | | |
Collapse
|
42
|
Izquierdo-Ramírez YE, Calvo-Páramo E, Castañeda-Castillo LM, Gómez Correa SV, Zambrano FS. Cambios radiográficos en el penacho de la falange distal de las manos en pacientes con psoriasis o artritis psoriásica. Revisión sistemática. REVISTA DE LA FACULTAD DE MEDICINA 2017. [DOI: 10.15446/revfacmed.v65n2.59228] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Introducción. El penacho de la falange distal de las manos (PFDM) es una estructura ósea que sufre cambios sutiles en diversas enfermedades musculoesqueléticas. Hasta el momento, se desconoce el tipo y frecuencia de las lesiones del PFDM en psoriasis y artritis psoriásica (APs).Objetivo. Determinar en radiografía simple las anormalidades del PFDM en adultos con psoriasis y APs.Materiales y métodos. Se realizó una revisión sistemática buscando alteraciones radiográficas del PFDM en adultos con psoriasis y APs. La prevalencia de los hallazgos fue sintetizada usando un modelo de efectos fijos. Las asociaciones se expresaron como odds ratio (OR) con intervalos de confianza (IC) y valores p.Resultados. Se incluyeron 11 estudios observacionales. La alteración de mayor frecuencia en APs fue la resorción del PFDM con una prevalencia del 16%. El compromiso del PFDM en sujetos con onicopatía psoriásica fue mayor que en sujetos sin compromiso ungueal (OR=2.91; IC95%: 1.04–8.13; p=0.04), siendo las erosiones y la esclerosis los hallazgos de mayor importancia.Conclusión. La evidencia actual acerca del compromiso del PFDM en APs y psoriasis es limitada. Existe relación entre las alteraciones del PFDM y la patología ungueal en donde las erosiones y la esclerosis son las anormalidades más significativas.
Collapse
|
43
|
Mease PJ, van der Heijde D, Ritchlin CT, Okada M, Cuchacovich RS, Shuler CL, Lin CY, Braun DK, Lee CH, Gladman DD. Ixekizumab, an interleukin-17A specific monoclonal antibody, for the treatment of biologic-naive patients with active psoriatic arthritis: results from the 24-week randomised, double-blind, placebo-controlled and active (adalimumab)-controlled period of the phase III trial SPIRIT-P1. Ann Rheum Dis 2017; 76:79-87. [PMID: 27553214 PMCID: PMC5264219 DOI: 10.1136/annrheumdis-2016-209709] [Citation(s) in RCA: 413] [Impact Index Per Article: 51.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2016] [Revised: 07/21/2016] [Accepted: 07/25/2016] [Indexed: 01/29/2023]
Abstract
OBJECTIVE To assess the safety and efficacy of ixekizumab, a monoclonal antibody that inhibits interleukin-17A, in a double-blind phase III trial enrolling patients with active psoriatic arthritis (PsA). METHODS Patients naive to biologic therapy with active PsA were randomised to subcutaneous injections of placebo (N=106), adalimumab 40 mg once every 2 weeks (active reference; N=101), ixekizumab 80 mg once every 2 weeks (IXEQ2W) (N=103), or ixekizumab 80 mg once every 4 weeks (IXEQ4W) (N=107). Both ixekizumab regimens included a 160-mg starting dose. The primary objective was to assess the superiority of IXEQ2W or IXEQ4W versus placebo as measured by the proportion of patients achieving an American College of Rheumatology 20 (ACR20) response at week 24. RESULTS Significantly more patients treated with ixekizumab achieved an ACR20 response with IXEQ2W (62.1%) or IXEQ4W (57.9%) than placebo (30.2%) (p≤0.001; non-responder imputation method). Disease activity and functional disability were significantly improved with both ixekizumab doses versus placebo at weeks 12 and 24, and there was significantly less progression of structural damage at week 24 (p≤0.01). Clearance of plaque psoriasis was greater with ixekizumab than placebo (p≤0.001). Efficacy results with adalimumab, the active reference arm, showed significant improvements versus placebo. Treatment-emergent adverse events were more frequent with ixekizumab (65.7-66.4%) and adalimumab (64.4%) than placebo (47.2%) (p<0.05). CONCLUSIONS In biologic-naive patients with active PsA, ixekizumab treatment resulted in improvements in disease activity and physical function, as well as in the inhibition of structural damage progression. Overall, adverse events were more frequent in all active groups compared with placebo. TRIAL REGISTRATION NUMBER NCT01695239; EudraCT2011-002326-49; Results.
Collapse
Affiliation(s)
- Philip J Mease
- Department of Rheumatology, Swedish Medical Center, and University of Washington, Seattle, Washington, USA
| | | | - Christopher T Ritchlin
- Allergy, Immunology, & Rheumatology Division, University of Rochester Medical Center, Rochester, New York, USA
| | - Masato Okada
- Immuno-Rheumatology Center, St. Luke's International Hospital, Tokyo, Japan
| | - Raquel S Cuchacovich
- Eli Lilly and Company, Indianapolis, Indiana, USA
- Rheumatology Division, Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | | | - Chen-Yen Lin
- Eli Lilly and Company, Indianapolis, Indiana, USA
| | | | - Chin H Lee
- Eli Lilly and Company, Indianapolis, Indiana, USA
| | - Dafna D Gladman
- Division of Rheumatology, Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
44
|
Haque N, Lories RJ, de Vlam K. Orthopaedic interventions in patients with psoriatic arthritis: a descriptive report from the SPAR cohort. RMD Open 2016; 2:e000293. [PMID: 27651927 PMCID: PMC5013510 DOI: 10.1136/rmdopen-2016-000293] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2016] [Accepted: 07/25/2016] [Indexed: 11/08/2022] Open
Abstract
Objectives To evaluate the current needs for joint surgery in patients with psoriatic arthritis (PsA). Methods The patient database at the Rheumatology Department of the University Hospitals Leuven, was cross-sectionally analysed using demographic, medical, laboratory, radiological and surgical data of 269 patients with PsA. Patients were grouped by the presence or absence of orthopaedic surgery and compared for gender, age, mean health assessment questionnaire (HAQ) score, current medication and disease duration. The data were assessed using descriptive statistics and Student's t-tests. Results Overall 48.33% of the patients underwent 1 or more orthopaedic surgeries at some point of time. A total of 280 surgical interventions were flagged in the database, including both joint sacrificing and non-joint sacrificing procedures. Mean disease duration±SD at the time of surgery was 1.58 years±12.05. Age of the patients with surgeries was 54.13 years±11.03 SD and not different from those without surgeries (53.73 years±12.81 SD; p=0.78). 41.54% of the patients underwent a single surgery while 58.46% had multiple surgeries. A significant difference in the mean HAQ score was observed among the patients with and without surgeries (p<0.001). Of all the surgeries 63.92% were performed after diagnosis whereas 36.07% were performed before a diagnosis of PsA was made. Among the surgeries performed before diagnosis 40.59% were arthroscopies including 9.90% of diagnostic arthroscopies. Conclusions The number of surgical interventions has significantly increased in patients with PsA compared with historical cohorts even with a relatively shorter disease duration. There was a significant difference in HAQ score between the patients with or without surgeries.
Collapse
Affiliation(s)
- Naba Haque
- Department of Development and Regeneration , Skeletal Biology and Engineering Research Center , Leuven , Belgium
| | - Rik J Lories
- Department of Development and Regeneration, Skeletal Biology and Engineering Research Center, Leuven, Belgium; Division of Rheumatology, University Hospitals Leuven, Leuven, Belgium
| | - Kurt de Vlam
- Department of Development and Regeneration, Skeletal Biology and Engineering Research Center, Leuven, Belgium; Division of Rheumatology, University Hospitals Leuven, Leuven, Belgium
| |
Collapse
|
45
|
Kavanaugh A, van der Heijde D, Beutler A, Gladman D, Mease P, Krueger GG, McInnes IB, Helliwell P, Coates LC, Xu S. Radiographic Progression of Patients With Psoriatic Arthritis Who Achieve Minimal Disease Activity in Response to Golimumab Therapy: Results Through 5 Years of a Randomized, Placebo-Controlled Study. Arthritis Care Res (Hoboken) 2016; 68:267-74. [PMID: 25779603 PMCID: PMC5066795 DOI: 10.1002/acr.22576] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2014] [Revised: 01/22/2015] [Accepted: 03/03/2015] [Indexed: 11/25/2022]
Abstract
Objective To evaluate long‐term outcomes in psoriatic arthritis (PsA) patients who achieved or did not achieve minimal disease activity (MDA) through 5 years of golimumab treatment in the GO‐REVEAL trial. Methods The GO‐REVEAL trial was a phase III, randomized, double‐blind trial with placebo‐control through week 24 followed by an open‐label extension of golimumab 50/100 mg treatment up to 5 years. In these post‐hoc analyses, MDA was defined by the presence of ≥5 of 7 PsA outcome measures (≤1 swollen joint, ≤1 tender joint, Psoriasis Area and Severity Index [PASI] ≤1, patient pain score ≤15, patient global disease activity score ≤20 [range 0–100], Health Assessment Questionnaire disability index [HAQ DI] ≤0.5, and ≤1 tender enthesis point). Results Treatment with golimumab yielded significantly higher MDA response rates versus patients randomized to placebo at week 14 (23.5% versus 1.0%; P < 0.0001), week 24 (28.1% versus 7.7%; P < 0.0001), and week 52 (42.4% versus 30.2%; P = 0.037). MDA was achieved at least once by ∼50% of golimumab‐treated patients overall. Irrespective of treatment randomization, achievement of MDA at ≥3 and ≥4 consecutive visits was associated with significantly less radiographic progression and more improvement in MDA components allowing specific assessment of physical function (HAQ DI) and overall disease activity (patient global assessment of disease activity) at week 256 versus patients not achieving MDA. Logistic regression analyses indicated that a 1‐unit higher baseline HAQ DI score yielded a significantly lower likelihood of achieving MDA at ≥3 (odds ratio 0.514 [95% confidence interval 0.321–0.824]; P = 0.006) and ≥4 (odds ratio 0.480 [95% confidence interval 0.290–0.795]; P = 0.004) consecutive visits. Conclusion Among golimumab‐treated PsA patients, better long‐term functional improvement, patient global assessment, and radiographic outcomes were observed when patients achieved persistent MDA.
Collapse
Affiliation(s)
| | | | - Anna Beutler
- Janssen Research & Development, Spring House, Pennsylvania
| | | | - Philip Mease
- Swedish Medical Center, Seattle, Washington, and University of Washington, Seattle
| | | | | | - Philip Helliwell
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, Leeds, UK
| | - Laura C Coates
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, Leeds, UK
| | - Stephen Xu
- Janssen Research & Development, Spring House, Pennsylvania
| |
Collapse
|
46
|
Kavanaugh A, Puig L, Gottlieb AB, Ritchlin C, Li S, Wang Y, Mendelsohn AM, Song M, Zhu Y, Rahman P, McInnes IB. Maintenance of Clinical Efficacy and Radiographic Benefit Through Two Years of Ustekinumab Therapy in Patients With Active Psoriatic Arthritis: Results From a Randomized, Placebo-Controlled Phase III Trial. Arthritis Care Res (Hoboken) 2016; 67:1739-49. [PMID: 26097039 PMCID: PMC5063124 DOI: 10.1002/acr.22645] [Citation(s) in RCA: 79] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2014] [Revised: 06/05/2015] [Accepted: 06/16/2015] [Indexed: 01/01/2023]
Abstract
Objective To evaluate the efficacy and safety of ustekinumab through 2 years in adult patients with active psoriatic arthritis (PsA). Methods A total of 615 adult patients with active PsA were randomized to placebo, ustekinumab 45 mg, or ustekinumab 90 mg, at weeks 0, 4, and every 12 weeks through week 88 (last dose). At week 16, patients with <5% improvement in both tender and swollen joint counts entered blinded early escape (placebo to 45 mg, 45 mg to 90 mg, and 90 mg to 90 mg). All remaining placebo patients crossed over to ustekinumab 45 mg at week 24. Clinical efficacy measures included American College of Rheumatology criteria for 20% improvement (ACR20), Disease Activity Score in 28 joints using the C‐reactive protein level (DAS28‐CRP), and ≥75% improvement in the Psoriasis Area and Severity Index (PASI75). Radiographic progression was evaluated using the modified Sharp/van der Heijde score (SHS). Results At week 100, ACR20, DAS28‐CRP moderate/good response, and PASI75 rates ranged from 56.7–63.6%, 71.9–76.7%, and 63.9–72.5%, respectively, across the 3 treatment groups. In both ustekinumab groups, the median percent improvement in dactylitis and enthesitis was 100% at week 100. The mean changes in SHS score from week 52 to week 100 were similar to those observed from week 0 to week 52 in the ustekinumab groups. Through week 108, 70.7% and 9.7% of patients had an adverse event (AE) or serious AE, respectively. The rates and type of AEs were similar between the dose groups. Conclusion Clinical and radiographic benefits from ustekinumab treatment were maintained through week 100 in the PSUMMIT 1 study. No unexpected safety events were observed; the safety profile of ustekinumab in this population was similar to that previously observed in psoriasis patients treated with ustekinumab.
Collapse
Affiliation(s)
| | - Lluís Puig
- Universitat Autònoma de Barcelona, Barcelona, Spain
| | | | | | - Shu Li
- Janssen Research & Development, Spring House, Pennsylvania
| | - Yuhua Wang
- Janssen Research & Development, Spring House, Pennsylvania
| | | | - Michael Song
- Janssen Research & Development, Spring House, Pennsylvania
| | - Yaowei Zhu
- Janssen Research & Development, Spring House, Pennsylvania
| | - Proton Rahman
- Memorial University, St. John's, Newfoundland and Labrador, Canada
| | | | | |
Collapse
|
47
|
Szentpetery A, Heffernan E, Haroon M, Kilbane M, Gallagher P, McKenna MJ, FitzGerald O. Striking difference of periarticular bone density change in early psoriatic arthritis and rheumatoid arthritis following anti-rheumatic treatment as measured by digital X-ray radiogrammetry. Rheumatology (Oxford) 2016; 55:891-6. [PMID: 26850991 DOI: 10.1093/rheumatology/kev443] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2015] [Indexed: 01/01/2023] Open
Abstract
OBJECTIVES To examine changes in hand BMD as measured by digital X-ray radiogrammetry (DXR-BMD) in early PsA compared with RA patients prior to and 3 and 12 months after introducing an antirheumatic treatment. Further, to identify predictors for hand bone loss at the time of disease presentation. METHODS Recent-onset, active, treatment-naïve patients were recruited. Clinical assessment, hand X-rays and DXR were obtained at 0, 3 and 12 months. Mean DXR-BMD for both hands and changes in DXR-BMD (mg/cm(2)/month) were compared between the two groups. We compared baseline disease characteristics of patients with normal hand DXR-BMD with those with bone loss. Logistic regression analyses were performed to identify predictors of hand BMD loss. RESULTS A total of 64 patients were included. Hand DXR-BMD decreased in RA throughout the study (P = 0.043). Changes in periarticular bone density over 12 months differed between PsA and RA (P = 0.001). Hand bone loss at 3 months was associated with elevated BMI [odds ratio (OR) = 3.59, P = 0.041] and heavier alcohol intake (OR = 1.13, P = 0.035). Diagnosis of RA (OR = 57.48, P = 0.008), heavier alcohol intake (OR = 1.27, P = 0.012) and higher swollen joint count (SJC28) (OR = 1.5, P = 0.036) were independent predictors for hand bone loss in the first year. CONCLUSION Following treatment, we found ongoing hand bone loss in RA and unchanged periarticular bone density in PsA, supporting the hypothesis that different pathomechanisms are involved in hand bone remodelling in PsA. Presence of RA, heavier alcohol intake and higher SJC were identified as independent predictors for hand bone loss over 1 year.
Collapse
Affiliation(s)
| | | | | | - Mark Kilbane
- Metabolism Laboratory, St. Vincent's University Hospital, Dublin, Ireland
| | | | - Malachi J McKenna
- Metabolism Laboratory, St. Vincent's University Hospital, Dublin, Ireland
| | | |
Collapse
|
48
|
Laasonen L, Gudbjornsson B, Ejstrup L, Iversen L, Ternowitz T, Ståhle M, Lindqvist U. Radiographic development during three decades in a patient with psoriatic arthritis mutilans. Acta Radiol Open 2015; 4:2058460115588098. [PMID: 26346445 PMCID: PMC4548743 DOI: 10.1177/2058460115588098] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2015] [Accepted: 04/25/2015] [Indexed: 11/29/2022] Open
Abstract
Psoriatic arthritis mutilans (PAM) is the most severe and rare form of psoriatic arthritis (PsA). We describe radiological development in a typical case of PAM covering three decades in order to elucidate the need for early diagnosis of PAM. Radiographs of hands and feet, taken from 1981 to 2010, were evaluated using the Psoriatic Arthritis Ratingen Score (PARS). When PsA was diagnosed, in 1981, gross deformity was observed in the second PIP joint of the left foot. Several pencil-in-cup deformities and gross osteolysis were present in the feet in the first decade of the disease. Over 10 years, many joints had reached maximum scores. During the follow-up, other joints became involved and the disease developed clinically. Reporting early signs suggestive of PAM, e.g. pencil-in cup deformities and gross osteolysis in any joint, should be mandatory and crucial. This would heighten our awareness of PAM, accelerate the diagnosis, and lead to improved effective treatment in order to minimize joint damages resulting in PAM.
Collapse
Affiliation(s)
- Leena Laasonen
- Helsinki Medical Imaging Center, Helsinki University Central Hospital, Helsinki, Finland
| | - Björn Gudbjornsson
- Center for Rheumatology Research, University of Iceland, Reykjavik, and Faculty of Medicine, University of Iceland, Iceland
| | - Leif Ejstrup
- Department of Rheumatology, Odense University Hospital, Odense, Denmark
| | - Lars Iversen
- Department of Dermatology, Aarhus University Hospital, Aarhus, Denmark
| | - Thomas Ternowitz
- Department of Dermatology, Stavanger University Hospital, Stavanger, Norway
| | - Mona Ståhle
- Dermatology Unit, Department of Medicine, Karolinska Institute, Stockholm, Sweden
| | - Ulla Lindqvist
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| |
Collapse
|
49
|
Mease P, Deodhar A, Fleischmann R, Wollenhaupt J, Gladman D, Leszczyński P, Vitek P, Turkiewicz A, Khraishi M, FitzGerald O, Landewé R, de Longueville M, Hoepken B, Peterson L, van der Heijde D. Effect of certolizumab pegol over 96 weeks in patients with psoriatic arthritis with and without prior antitumour necrosis factor exposure. RMD Open 2015; 1:e000119. [PMID: 26509074 PMCID: PMC4612702 DOI: 10.1136/rmdopen-2015-000119] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2015] [Revised: 06/01/2015] [Accepted: 06/07/2015] [Indexed: 12/16/2022] Open
Abstract
Objective Previous reports of RAPID-PsA (NCT01087788) demonstrated efficacy and safety of certolizumab pegol (CZP) over 24 weeks in patients with psoriatic arthritis (PsA), including patients with prior antitumour necrosis factor (TNF) therapy. We report efficacy and safety data from a 96-week data cut of RAPID-PsA. Methods RAPID-PsA was placebo-controlled to week 24, dose-blind to week 48 and open-label to week 216. We present efficacy data including American College of Rheumatology (ACR)/Psoriasis Area and Severity Index (PASI) responses, HAQ-DI, pain, minimal disease activity (MDA), modified total Sharp score (mTSS) and ACR responses in patients with/without prior anti-TNF exposure, in addition to safety data. Results Of 409 patients randomised, 273 received CZP from week 0. 54 (19.8%) CZP patients had prior anti-TNF exposure. Of patients randomised to CZP, 91% completed week 24, 87% week 48 and 80% week 96. ACR responses were maintained to week 96: 60% of patients achieved ACR20 at week 24, and 64% at week 96. Improvements were observed with both CZP dose regimens. ACR20 responses were similar in patients with (week 24: 59%; week 96: 63%) and without (week 24: 60%; week 96: 64%) prior anti-TNF exposure. Placebo patients switching to CZP displayed rapid clinical improvements, maintained to week 96. In patients with ≥3% baseline skin involvement (60.8% week 0 CZP patients), PASI responses were maintained to week 96. No progression of structural damage was observed over the 96-week period. In the Safety Set (n=393), adverse events occurred in 345 patients (87.8%) and serious adverse events in 67 (17.0%), including 6 fatal events. Conclusions CZP efficacy was maintained to week 96 with both dose regimens and in patients with/without prior anti-TNF exposure. The safety profile was in line with that previously reported from RAPID-PsA, with no new safety signals observed with increased exposure. Trial registration number NCT01087788.
Collapse
Affiliation(s)
- P Mease
- Swedish Medical Center and University of Washington , Seattle, Washington , USA
| | - A Deodhar
- Oregon Health & Science University , Portland, Oregon , USA
| | - R Fleischmann
- University of Texas SW Medical Center , Dallas, Texas , USA
| | | | - D Gladman
- Toronto Western Research Institute , Toronto , Canada
| | - P Leszczyński
- Poznan Medical University , Poznan , Poland ; Division of Rheumatology and Osteoporosis , Jozef Strus Hospital , Poznan , Poland
| | - P Vitek
- PV-MEDICAL, Revmavita Centre , Zlin , Czech Republic
| | - A Turkiewicz
- Rheumatology Associates Clinical Research Unit , Birmingham, Alabama , USA
| | - M Khraishi
- Department of Medicine , Memorial University of Newfoundland , St. John's , Canada
| | - O FitzGerald
- Department of Rheumatology , St. Vincent's University Hospital and Conway Institute for Biomolecular Research, University College , Dublin , Ireland
| | - R Landewé
- Academic Medical Center, Amsterdam and Atrium Medical Center , Heerlen , The Netherlands
| | | | | | - L Peterson
- UCB Pharma , Raleigh, North Carolina , USA
| | | |
Collapse
|
50
|
Jadon DR, Nightingale AL, McHugh NJ, Lindsay MA, Korendowych E, Sengupta R. Serum soluble bone turnover biomarkers in psoriatic arthritis and psoriatic spondyloarthropathy. J Rheumatol 2014; 42:21-30. [PMID: 25362660 DOI: 10.3899/jrheum.140223] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Because psoriatic arthritis (PsA) is an inflammatory disease of joints, serum soluble biomarkers specific for chronic joint and bone inflammation may predict future disease severity and response to therapy, thereby informing stratified medicine approaches. The objectives of our systematic review were to determine whether serum soluble bone and cartilage turnover biomarkers are (1) associated with PsA or psoriatic spondyloarthropathy; and (2) associated with disease activity, disease severity, or clinical phenotype. Ten studies met eligibility criteria. Matrix metalloproteinase (MMP)-3, Dickkopf (DKK)-1, macrophage colony-stimulating factor (M-CSF), crosslinked telopeptide of collagen-1, and tumor necrosis factor-related apoptosis-inducing ligand were associated with PsA, with equivocal results for osteoprotegerin (OPG) and bone alkaline phosphatase (ALP). MMP-3, DKK-1, M-CSF, CPII:C2C (ratio of cartilage degradation vs byproduct formation), and possibly OPG were associated with PsA independently of psoriasis. C1-2C (a neoepitope released when type 2 cartilage is degraded by collagenases) was associated with both tender and swollen joint counts, and bone morphogenetic protein-4 with patient global assessment of disease, pain score, and the Bath Ankylosing Spondylitis Disease Activity Index. Bone ALP was associated with disease activity. M-CSF and receptor activator of nuclear factor-κB ligand were associated with several plain radiographic features. No studies have investigated biomarker associations specifically with axial PsA.
Collapse
Affiliation(s)
- Deepak R Jadon
- From the Royal National Hospital for Rheumatic Diseases, and the University of Bath, Bath, UK.D.R. Jadon, MRCP, Research Fellow, Rheumatology; E. Korendowych, FRCP, Consultant Rheumatologist; R. Sengupta, FRCP, Consultant Rheumatologist, Royal National Hospital for Rheumatic Diseases; A.L. Nightingale, PhD, Research Fellow; M.A. Lindsay, PhD, Professor, Pharmacy and Pharmacology, University of Bath; N.J. McHugh, FRCP, Consultant Rheumatologist, Royal National Hospital for Rheumatic Diseases, and the University of Bath.
| | - Alison L Nightingale
- From the Royal National Hospital for Rheumatic Diseases, and the University of Bath, Bath, UK.D.R. Jadon, MRCP, Research Fellow, Rheumatology; E. Korendowych, FRCP, Consultant Rheumatologist; R. Sengupta, FRCP, Consultant Rheumatologist, Royal National Hospital for Rheumatic Diseases; A.L. Nightingale, PhD, Research Fellow; M.A. Lindsay, PhD, Professor, Pharmacy and Pharmacology, University of Bath; N.J. McHugh, FRCP, Consultant Rheumatologist, Royal National Hospital for Rheumatic Diseases, and the University of Bath
| | - Neil J McHugh
- From the Royal National Hospital for Rheumatic Diseases, and the University of Bath, Bath, UK.D.R. Jadon, MRCP, Research Fellow, Rheumatology; E. Korendowych, FRCP, Consultant Rheumatologist; R. Sengupta, FRCP, Consultant Rheumatologist, Royal National Hospital for Rheumatic Diseases; A.L. Nightingale, PhD, Research Fellow; M.A. Lindsay, PhD, Professor, Pharmacy and Pharmacology, University of Bath; N.J. McHugh, FRCP, Consultant Rheumatologist, Royal National Hospital for Rheumatic Diseases, and the University of Bath
| | - Mark A Lindsay
- From the Royal National Hospital for Rheumatic Diseases, and the University of Bath, Bath, UK.D.R. Jadon, MRCP, Research Fellow, Rheumatology; E. Korendowych, FRCP, Consultant Rheumatologist; R. Sengupta, FRCP, Consultant Rheumatologist, Royal National Hospital for Rheumatic Diseases; A.L. Nightingale, PhD, Research Fellow; M.A. Lindsay, PhD, Professor, Pharmacy and Pharmacology, University of Bath; N.J. McHugh, FRCP, Consultant Rheumatologist, Royal National Hospital for Rheumatic Diseases, and the University of Bath
| | - Eleanor Korendowych
- From the Royal National Hospital for Rheumatic Diseases, and the University of Bath, Bath, UK.D.R. Jadon, MRCP, Research Fellow, Rheumatology; E. Korendowych, FRCP, Consultant Rheumatologist; R. Sengupta, FRCP, Consultant Rheumatologist, Royal National Hospital for Rheumatic Diseases; A.L. Nightingale, PhD, Research Fellow; M.A. Lindsay, PhD, Professor, Pharmacy and Pharmacology, University of Bath; N.J. McHugh, FRCP, Consultant Rheumatologist, Royal National Hospital for Rheumatic Diseases, and the University of Bath
| | - Raj Sengupta
- From the Royal National Hospital for Rheumatic Diseases, and the University of Bath, Bath, UK.D.R. Jadon, MRCP, Research Fellow, Rheumatology; E. Korendowych, FRCP, Consultant Rheumatologist; R. Sengupta, FRCP, Consultant Rheumatologist, Royal National Hospital for Rheumatic Diseases; A.L. Nightingale, PhD, Research Fellow; M.A. Lindsay, PhD, Professor, Pharmacy and Pharmacology, University of Bath; N.J. McHugh, FRCP, Consultant Rheumatologist, Royal National Hospital for Rheumatic Diseases, and the University of Bath
| |
Collapse
|