1
|
Séauve M, Auréal M, Laplane S, Lega JC, Cabrera N, Coury F. Risk of infections in psoriatic arthritis or axial spondyloarthritis patients treated with targeted therapies: A meta-analysis of randomized controlled trials. Joint Bone Spine 2024; 91:105673. [PMID: 38042364 DOI: 10.1016/j.jbspin.2023.105673] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Revised: 11/01/2023] [Accepted: 11/14/2023] [Indexed: 12/04/2023]
Abstract
OBJECTIVE To evaluate the risk of global infections in patients with psoriatic arthritis (PsA) and axial spondyloarthritis encompassing ankylosing spondylitis (AS) and non-radiographic axial spondyloarthritis (nr-axSpA) treated with targeted therapies. METHODS Medline and Cochrane databases were systematically searched up to March 2021 for randomized controlled trials (RCTs) performed in patients with PsA or axial spondyloarthritis treated with biologic or targeted synthetic disease-modifying anti-rheumatic drugs (b/tsDMARDs). Global infections (any infections reported, including bacterial, viral and fungal infections, except serious infections) were the primary outcome. Secondary outcomes included serious infections defined as life-threatening infections or any infection requiring intravenous antibiotics or hospitalization. The relative risk of infections was determined by meta-analysis of RCTs. RESULTS A total of 60 RCTs were included (20,418 patients), encompassing 17 b/tsDMARDs, compared with placebo, conventional synthetic drugs (csDMARDs) or non-steroidal anti-inflammatory drugs (NSAIDs). An increased risk of any infection for patients exposed to these drugs was found (RR 1.15, 95% CI [1.06-1.25]), mainly with high doses and longer duration of treatment. Most infections were respiratory tract or ear, nose, and throat (ENT) infections. Subgroup analyses showed a statistically significant increased risk of infections for axial spondyloarthritis patients (RR 1.32, 95% CI [1.14-1.52]), but not for PsA patients (RR 1.05, 95% CI [0.97-1.14]). Infection risk was highest with TNF inhibitors (RR 1.23, 95% CI [1.11-1.37]) and IL-17 inhibitors (RR 1.30, 95% CI [1.07-1.59]). No increased risk of serious infections was shown. CONCLUSION In contrast to serious infections, the risk of global infections is moderately increased with b/tsDMARDs in spondyloarthritis, and is associated in particular with use of TNF and IL-17 inhibitors.
Collapse
Affiliation(s)
- Milène Séauve
- University of Lyon, University Lyon 1, 69100 Lyon, France; Department of Rheumatology, Lyon Sud Hospital, Hospices Civils de Lyon, 69495 Pierre-Bénite, France
| | - Mélanie Auréal
- University of Lyon, University Lyon 1, 69100 Lyon, France; Department of Rheumatology, Lyon Sud Hospital, Hospices Civils de Lyon, 69495 Pierre-Bénite, France
| | - Soline Laplane
- University of Lyon, University Lyon 1, 69100 Lyon, France; Department of Rheumatology, Lyon Sud Hospital, Hospices Civils de Lyon, 69495 Pierre-Bénite, France
| | - Jean-Christophe Lega
- University of Lyon, University Lyon 1, 69100 Lyon, France; Department of Internal and Vascular Medicine, Lyon Sud Hospital, Hospices Civils de Lyon, Pierre-Bénite, France; University of Lyon, UMR - CNRS 5558, Laboratoire de Biométrie et Biologie Évolutive, 69100 Lyon, France; Lyon Immunopathology Federation, Lyon, France
| | - Natalia Cabrera
- University of Lyon, UMR - CNRS 5558, Laboratoire de Biométrie et Biologie Évolutive, 69100 Lyon, France
| | - Fabienne Coury
- University of Lyon, University Lyon 1, 69100 Lyon, France; Department of Rheumatology, Lyon Sud Hospital, Hospices Civils de Lyon, 69495 Pierre-Bénite, France; Lyon Immunopathology Federation, Lyon, France; University of Lyon, Inserm UMR 1033, 69100 Lyon, France.
| |
Collapse
|
2
|
Alharthy RF, Alharthy JM, Bawazir RO, Katib RI, Alharthy FS. The Efficacy and Safety of Apremilast in the Management of Psoriatic Arthritis: A Systematic Review and Meta-Analysis. Cureus 2024; 16:e55773. [PMID: 38590459 PMCID: PMC11000044 DOI: 10.7759/cureus.55773] [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] [Accepted: 03/08/2024] [Indexed: 04/10/2024] Open
Abstract
Psoriasis is a chronic autoimmune inflammatory skin disease that is associated with other conditions, one of them being psoriatic arthritis (PsA). Apremilast, a phosphodiesterase-4 inhibitor, displayed promising results in multiple trials for patients with PsA. This systematic review and meta-analysis aims to showcase its efficacy and safety when compared to placebo. Preferred Reporting Items for Systematic Reviews and Meta-Analysis (PRISMA) was adopted after registration on the International Prospective Register of Systematic Reviews (PROSPERO: CRD42023476245). Four databases were systematically searched from their inception until October 25, 2023. As a result, five randomized controlled trials were included with 1,849 participants, after thorough screening. The primary efficacy endpoint evaluated in this meta-analysis was the American College of Rheumatology Response Criteria 20 (ACR20). The results significantly favored apremilast (risk ratio [RR] = 1.92, 95% confidence interval [CI] 1.66-2.21; P < 0.00001; I2= 0%) as opposed to placebo. Similarly, secondary efficacy endpoints, ACR50 (RR = 2.34, 95% CI 1.79-3.06; P < 0.00001; I2 = 0%), ACR70 (RR = 2.89, 95% CI 1.62-5.18; P = 0.0003; I2 = 0%), and the Health Assessment Questionnaire and Disability Index (HAQ-DI; standardized mean difference [SMD] = -0.26, 95% CI -0.34 to -0.17; P < 0.00001; I2 = 0%) were also in significant favor of apremilast. However, apremilast had a higher occurrence of gastrointestinal adverse events than placebo (RR = 1.21, 95% CI 1.12-1.30; P < 0.00001; I2 = 19%). To conclude, apremilast shows promising efficaciousness with some nonserious side effects when compared to placebo, but further trials are needed for comparison with other management lines.
Collapse
Affiliation(s)
- Renad F Alharthy
- College of Medicine, King Saud Bin Abdulaziz University for Health Sciences, Jeddah, SAU
- College of Medicine, King Abdullah International Medical Research Center, Jeddah, SAU
| | - Joud M Alharthy
- College of Medicine, King Saud Bin Abdulaziz University for Health Sciences, Jeddah, SAU
- College of Medicine, King Abdullah International Medical Research Center, Jeddah, SAU
| | - Razan O Bawazir
- College of Medicine, King Saud Bin Abdulaziz University for Health Sciences, Jeddah, SAU
- College of Medicine, King Abdullah International Medical Research Center, Jeddah, SAU
| | - Renad I Katib
- College of Medicine, King Saud Bin Abdulaziz University for Health Sciences, Jeddah, SAU
- College of Medicine, King Abdullah International Medical Research Center, Jeddah, Saudi Arabia, Jeddah, SAU
| | - Fayez S Alharthy
- Internal Medicine/Rheumatology, King Abdulaziz Medical City, Jeddah, SAU
- Internal Medicine/Rheumatology, College of Medicine, King Saud Bin Abdulaziz University for Health Sciences, Jeddah, SAU
| |
Collapse
|
3
|
Hsieh TS, Tsai TF. Combination of methotrexate with oral disease-modifying antirheumatic drugs in psoriatic arthritis: a systematic review. Immunotherapy 2024; 16:115-130. [PMID: 38112064 DOI: 10.2217/imt-2023-0139] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Accepted: 11/13/2023] [Indexed: 12/20/2023] Open
Abstract
Background: Oral conventional synthetic disease-modifying antirheumatic drugs (csDMARDs), especially methotrexate, are the cornerstone of treating psoriatic arthritis (PsA). The use of csDMARDs with biologics has increased their efficacy in psoriasis. However, the combination of two oral DMARDs in patients with PsA has not been adequately reviewed. In this study, we explore the combinational use of methotrexate with DMARDs in PsA patients. Materials & methods: A review was conducted using Medline (PubMed), Embase, Web of Science and the Cochrane Library, covering articles up to February 2023. Results & conclusion: Nine studies comprising 1993 participants were included. The evidence supporting combination therapy remains limited. Combinational therapy could be considered in patients with inadequate response to monotherapy or no access to biologics.
Collapse
Affiliation(s)
- Tyng-Shiuan Hsieh
- Department of Dermatology, National Taiwan University Hospital & National Taiwan University College of Medicine, Taipei, 10002, Taiwan
| | - Tsen-Fang Tsai
- Department of Dermatology, National Taiwan University Hospital & National Taiwan University College of Medicine, Taipei, 10002, Taiwan
| |
Collapse
|
4
|
Bruggemeyer C, Nepal D, Putman M. Unintentional unblinding in rheumatic disease trials. THE LANCET. RHEUMATOLOGY 2023; 5:e633-e636. [PMID: 38251487 DOI: 10.1016/s2665-9913(23)00191-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Revised: 06/19/2023] [Accepted: 07/12/2023] [Indexed: 01/23/2024]
Abstract
The practice of blinding treatment assignment in randomised controlled trials mitigates important biases in observational studies. Unblinding, whereby study participants or investigators become aware of treatment assignments, is an important threat to the validity of trial results. Rheumatology studies might be particularly susceptible to unblinding because rheumatic disease therapies often cause high rates of idiosyncratic side-effects and frequently rely on subjective endpoints. Despite this susceptibility, the degree to which unblinding occurs in randomised controlled trials in rheumatic diseases has rarely been assessed during trials or acknowledged as a limitation. Rheumatologists should be aware of this important threat to the validity of trial results, assessments of unblinding should be undertaken, and strategies to prevent unblinding should be deployed when feasible.
Collapse
Affiliation(s)
- Cody Bruggemeyer
- Hub for Collaborative Medicine, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Desh Nepal
- Hub for Collaborative Medicine, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Michael Putman
- Hub for Collaborative Medicine, Medical College of Wisconsin, Milwaukee, WI, USA.
| |
Collapse
|
5
|
Kang Q, Chen JS, Yang H. Efficacy and safety profile of phosphodiesterase 4 inhibitor in the treatment of psoriasis: A systematic review and meta-analysis of randomized controlled trials. Front Immunol 2022; 13:1021537. [PMID: 36300119 PMCID: PMC9589065 DOI: 10.3389/fimmu.2022.1021537] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Accepted: 09/22/2022] [Indexed: 11/18/2022] Open
Abstract
Background Systemic therapy is an important treatment for psoriasis. Phosphodiesterase 4 (PDE4) inhibitors are new candidates for psoriasis therapy. Objectives To evaluate the efficacy and safety of PDE4 inhibitors in psoriasis. Method Randomized clinical trials with PDE4 inhibitors vs placebos in patients with psoriasis were identified from MEDLINE, Embase, Cochrane Controlled Register of Trials, ClinicalTrials.gov, from inception to July 14, 2022. The study was registered in PROSPERO (CRD42022345700). Results 18 studies were identified, 9 of which included moderate-to-severe plaque psoriasis, 2 mild-to-moderate plaque psoriasis, and 7 psoriatic arthritis. A total of 6036 patients were included. Only one oral PDE4 inhibitor, apremilast, met the inclusion criteria. Overall, compared with the placebo, apremilast was associated with higher response rates in PASI-75 (RR, 3.22; 95% CI, 2.59-4.01), ScPGA of 0 or 1 (RR, 2.21; 95% CI, 1.69-2.91), PPPGA of 0 or 1 (RR 2.33; 95%CI, 1.16-4.66), and a significant decrease in NPASI (SMD, -0.46; 95% CI, -0.58 to -0.33). There were no significant differences in serious adverse events. Subgroup analyses showed that significantly more patients achieved PASI-75 after 16 weeks of therapy with apremilast of 20 mg bid (RR, 2.82; 95% CI, 2.01-3.95) and 30 mg bid (RR, 4.08; 95% CI, 3.12-5.33). Heterogeneity was not significant across studies. Conclusion Apremilast is a safe and effective treatment for plaque psoriasis and psoriatic arthritis, especially for difficult-to-treat sites. Systematic review registration https://www.crd.york.ac.uk/prospero, identifier (CRD42022345700).
Collapse
Affiliation(s)
- Qin Kang
- Department of Health Statistics and Information Management, School of Public Health, Chongqing Medical University, Chongqing, China
| | - Jing-si Chen
- Department of Dermatology, Children’s Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China
- China International Science and Technology Cooperation Base of Child development and Critical Disorders, Chongqing, China
| | - Huan Yang
- Department of Dermatology, Children’s Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China
- Chongqing Key Laboratory of Child Health and Nutrition, Chongqing, China
- *Correspondence: Huan Yang,
| |
Collapse
|
6
|
Vassilopoulos A, Shehadeh F, Benitez G, Kalligeros M, Cunha JS, Cunha CB, Mylonakis E. The incidence of opportunistic infections in patients with psoriatic arthritis treated with biologic and targeted synthetic agents: A systematic review and meta-analysis. Front Pharmacol 2022; 13:992713. [PMID: 36278224 PMCID: PMC9579334 DOI: 10.3389/fphar.2022.992713] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Accepted: 09/20/2022] [Indexed: 11/13/2022] Open
Abstract
Background: Biologic (bDMARD) and targeted synthetic (tsDMARD) disease-modifying anti-rheumatic drugs have broadened the treatment options and are increasingly used for patients with psoriatic arthritis (PsA). These agents block different pro-inflammatory cytokines or specific intracellular signaling pathways that promote inflammation and can place patients at risk of serious infections. We aimed to review the incidence of opportunistic infections (OIs) in patients with PsA who were treated with these agents.Methods: We searched PubMed and EMBASE through 14 April 2022 for randomized clinical trials evaluating bDMARD or tsDMARD in the treatment of PsA. Trials were eligible if they compared the effect of a bDMARD or tsDMARD with placebo and provided safety data. We used the Revised Cochrane risk-of-bias tool to assess the risk of bias among trials, and stratified the studies by mechanism of action (MOA) of the agents studied.Results: We included 47 studies in this analysis. A total of 17,197 patients received at least one dose of an agent of interest. The cumulative incidence of OIs by MOA was as follows: 1) JAK inhibitors: 2.72% (95% CI: 1.05%–5.04%), 2) anti-IL-17: 1.18% (95% CI: 0.60%–1.9%), 3) anti-IL-23: 0.24% (95% CI: 0.04%–0.54%), and 4) anti-TNFs: 0.01% (95% CI: 0.00%–0.21%). Based on their MOA, these agents are known to increase the risk of certain serious infections. The cumulative incidence of herpes zoster infection following treatment with JAK inhibitors (JAKi) was 2.53% (95% CI: 1.03%–4.57%) and the cumulative incidence of opportunistic Candida spp. infections following treatment with anti-IL-17, was 0.97% (95% CI: 0.51%–1.56%).Conclusion: The overall incidence of OIs among patients with PsA who were treated with biologic and targeted synthetic agents is low. However, careful monitoring is warranted for specific OIs such as herpes zoster infection following JAKi treatment, mucocutaneous candidiasis following anti-IL-17 treatment, and Mycobacterium tuberculosis infection following anti-TNF treatment.
Collapse
Affiliation(s)
- Athanasios Vassilopoulos
- Infectious Diseases Division, Rhode Island Hospital, Providence, RI, United States
- Warren Alpert Medical School of Brown University, Providence, RI, United States
| | - Fadi Shehadeh
- Infectious Diseases Division, Rhode Island Hospital, Providence, RI, United States
- Warren Alpert Medical School of Brown University, Providence, RI, United States
- School of Electrical and Computer Engineering, National Technical University of Athens, Athens, Greece
| | - Gregorio Benitez
- Infectious Diseases Division, Rhode Island Hospital, Providence, RI, United States
- Warren Alpert Medical School of Brown University, Providence, RI, United States
| | - Markos Kalligeros
- Infectious Diseases Division, Rhode Island Hospital, Providence, RI, United States
- Warren Alpert Medical School of Brown University, Providence, RI, United States
| | - Joanne S. Cunha
- Warren Alpert Medical School of Brown University, Providence, RI, United States
| | - Cheston B. Cunha
- Infectious Diseases Division, Rhode Island Hospital, Providence, RI, United States
- Warren Alpert Medical School of Brown University, Providence, RI, United States
| | - Eleftherios Mylonakis
- Infectious Diseases Division, Rhode Island Hospital, Providence, RI, United States
- Warren Alpert Medical School of Brown University, Providence, RI, United States
- *Correspondence: Eleftherios Mylonakis,
| |
Collapse
|
7
|
Apremilast retention rate in clinical practice: observations from an Italian multi-center study. Clin Rheumatol 2022; 41:3219-3225. [PMID: 35796847 DOI: 10.1007/s10067-022-06255-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Revised: 05/25/2022] [Accepted: 06/15/2022] [Indexed: 11/03/2022]
Abstract
OBJECTIVE There are few real-world setting studies focused on apremilast effectiveness (i.e., retention rate) in psoriatic arthritis (PsA). The main aim of this retrospective observational study is the assessment of apremilast 3-year retention rate in real-world PsA patients. Moreover, the secondary objective is to report the reasons of apremilast discontinuation and the factors related to treatment persistence. METHODS In fifteen Italian rheumatological referral centers, all PsA consecutive patients who received apremilast were enrolled. Anamnestic data, treatment history, and PsA disease activity (DAPSA) at baseline were recorded. The Kaplan-Meier curve and the Cox analysis computed the apremilast retention rate and treatment persistence-related risk factors. A p-value < 0.05 was considered statistically significant. RESULTS The 356 enrolled patients (median age 60 [interquartile range IQR 52-67] yrs; male prevalence 42.7%) median observation period was 17 [IQR 7-34] months (7218 patients-months). The apremilast retention rate at 12, 24, and 36 months was, respectively, 85.6%, 73.6%, and 61.8%. The main discontinuation reasons were secondary inefficacy (34% of interruptions), gastro-intestinal intolerance (24%), and primary inefficacy (19%). Age and oligo-articular phenotype were related to treatment persistence (respectively hazard ratio 0.98 IQR 0.96-0.99; p = 0.048 and 0.54 IQR 0.31-0.95; p = 0.03). CONCLUSION Almost three-fifths of PsA patients receiving apremilast were still in treatment after 3 years. This study confirmed its effectiveness and safety profile. Apremilast appears as a good treatment choice in all oligo-articular PsA patients and in those ones burdened by relevant comorbidities. Key Points • Apremilast retention rates in this real-life cohort and trials are comparable. • The oligo-articular phenotype is associated with long-lasting treatment (i.e., 3 years). • No different or more prevalent adverse events were observed.
Collapse
|
8
|
McInnes IB, Sawyer LM, Markus K, LeReun C, Sabry-Grant C, Helliwell PS. Targeted systemic therapies for psoriatic arthritis: a systematic review and comparative synthesis of short-term articular, dermatological, enthesitis and dactylitis outcomes. RMD Open 2022; 8:e002074. [PMID: 35321874 PMCID: PMC8943739 DOI: 10.1136/rmdopen-2021-002074] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Accepted: 03/02/2022] [Indexed: 01/11/2023] Open
Abstract
INTRODUCTION Randomised controlled trials (RCTs) have compared biological and targeted systemic disease-modifying antirheumatic drugs (DMARDS) against placebo in psoriatic arthritis (PsA); few have compared them head to head. OBJECTIVES To compare the efficacy and safety of all evaluated DMARDs for active PsA, with a special focus on biological DMARDs (bDMARDs) licensed for PsA or psoriasis. METHODS A systematic review identified RCTs and Bayesian network meta-analysis (NMA) compared treatments on efficacy (American College of Rheumatology (ACR) response, Psoriasis Area and Severity Index (PASI) response, resolution of enthesitis and dactylitis) and safety (patients discontinuing due to adverse events (DAE)) outcomes. Subgroup analyses explored ACR response among patients with and without prior biological therapy exposure. RESULTS The NMA included 46 studies. Results indicate that some tumour necrosis factor inhibitors (anti-TNFs) may perform numerically, but not significantly, better than interleukin (IL) inhibitors on ACR response but perform worse on PASI response. Few significant differences between bDMARDs on ACR response were observed after subgrouping for prior bDMARD exposure. Guselkumab and IL-17A or IL-17RA inhibitors-brodalumab, ixekizumab, secukinumab-were best on PASI response. These IL-inhibitors and adalimumab were similarly efficacious on resolution of enthesitis and dactylitis. Infliximab with and without methotrexate, certolizumab 400 mg every 4 weeks and tildrakizumab showed the highest rates of DAE; abatacept, golimumab and the IL-inhibitors, the lowest. CONCLUSIONS Despite similar efficacy for ACR response, IL-17A and IL-17RA inhibitors and guselkumab offered preferential efficacy to anti-TNFs in skin manifestations, and for enthesitis and dactylitis, thereby supporting drug selection based on predominant clinical phenotype.
Collapse
Affiliation(s)
- Iain B McInnes
- College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | | | | | - Corinne LeReun
- Independent Senior Biostatistician, Sainte-Anne, Guadeloupe
| | | | - Philip S Helliwell
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Leeds, UK
| |
Collapse
|
9
|
Motolese A, Ceccarelli M, Macca L, Li Pomi F, Ingrasciotta Y, Nunnari G, Guarneri C. Novel Therapeutic Approaches to Psoriasis and Risk of Infectious Disease. Biomedicines 2022; 10:biomedicines10020228. [PMID: 35203438 PMCID: PMC8869084 DOI: 10.3390/biomedicines10020228] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2022] [Revised: 01/12/2022] [Accepted: 01/18/2022] [Indexed: 01/22/2023] Open
Abstract
Psoriasis is a chronic immune-mediated skin and joint disease, with a plethora of comorbidities, characterized by a certain genetic predisposition, and a complex pathogenesis based on the IL-23/IL-17 pathway. There is no doubt that the patients affected by psoriasis are more susceptible to infections as well as that the risk of infection is higher in psoriatic subjects than in the general population. The advent of biotechnological agents on the therapeutic arsenal actually available for the treatment of moderate-to-severe patients, given the fact that the severity of the disease is a predictor of the level of infectious risk, has raised the question of whether these ‘new’ drugs could be considered a safer option and how they can be used in selected cases. Old and newer strategies in cases of chronic infectious conditions are reviewed under the light of clinical trials and other studies present in literature.
Collapse
Affiliation(s)
- Alfonso Motolese
- Department of Clinical and Experimental Medicine, Section of Dermatology, University of Messina, Messina, Italy C/O A.O.U.P. “Gaetano Martino”, via Consolare Valeria, 1, 98125 Messina, Italy; (A.M.); (L.M.); (F.L.P.)
| | - Manuela Ceccarelli
- Department of Clinical and Experimental Medicine, Unit of Infectious Diseases, University of Catania, Catania, Italy C/O ARNAS “Garibaldi”, “Nesima” Hospital, via Palermo 636, 95122 Catania, Italy;
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, Unit of Infectious Diseases, University of Messina, Messina, Italy C/O A.O.U.P. “Gaetano Martino”, via Consolare Valeria, 1, 98125 Messina, Italy
| | - Laura Macca
- Department of Clinical and Experimental Medicine, Section of Dermatology, University of Messina, Messina, Italy C/O A.O.U.P. “Gaetano Martino”, via Consolare Valeria, 1, 98125 Messina, Italy; (A.M.); (L.M.); (F.L.P.)
| | - Federica Li Pomi
- Department of Clinical and Experimental Medicine, Section of Dermatology, University of Messina, Messina, Italy C/O A.O.U.P. “Gaetano Martino”, via Consolare Valeria, 1, 98125 Messina, Italy; (A.M.); (L.M.); (F.L.P.)
| | - Ylenia Ingrasciotta
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, Section of Pharmacology, University of Messina, Messina, Italy C/O A.O.U.P. “Gaetano Martino”, via Consolare Valeria, 1, 98125 Messina, Italy;
| | - Giuseppe Nunnari
- Department of Clinical and Experimental Medicine, Unit of Infectious Diseases, University of Messina, Messina, Italy C/O A.O.U.P. “Gaetano Martino”, via Consolare Valeria, 1, 98124 Messina, Italy;
| | - Claudio Guarneri
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, Section of Dermatology, University of Messina, Messina, Italy C/O A.O.U.P. “Gaetano Martino”, via Consolare Valeria, 1, 98125 Messina, Italy
- Correspondence: ; Tel.: +39-090-2212-894; Fax: +39-09-029-27691
| |
Collapse
|
10
|
Erre GL, Mavridis D, Woodman RJ, Mangoni AA. Placebo response in psoriatic arthritis clinical trials: a systematic review and meta-analysis. Rheumatology (Oxford) 2021; 61:1328-1340. [PMID: 34664615 DOI: 10.1093/rheumatology/keab774] [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: 07/15/2021] [Revised: 09/17/2021] [Accepted: 10/11/2021] [Indexed: 11/14/2022] Open
Abstract
OBJECTIVE To determine the placebo response rate in psoriatic arthritis (PsA) randomised clinical trials (RCTs), its contributing factors, and impact on the effect size of active treatments. METHODS We searched multiple databases, from inception to December 20, 2020, for placebo-controlled RCTs in PsA. We used a random-effects meta-analysis to pool the response rates for the American College of Rheumatology 20 (ACR20) criteria in the placebo arm, determined the risk difference for treatment vs placebo, and used meta-regression to determine the factors associated with placebo response rates. The risk of bias was assessed in duplicate. PROSPERO: CRD42021226000. RESULTS We included 42 RCTs (5,050 patients receiving placebo) published between 2000 and 2020; The risk of bias was low in 28 trials, high in four, and with some concerns in ten. The pooled placebo response rate was 20.3% (95% CI, 18.6% to 22.1%; predicted intervals, 11.7%-29.0%), with significant between-trial heterogeneity (I2=56.8%, p< 0.005). The pooled risk difference for treatment vs placebo was 27% (95%CI, 24% to 31%). In the multivariable meta-regression, there was a 15% (95% CI, 2.9% to 29.8%) increase in the odds of achieving the placebo response for each five-year increment in publication year (p= 0.016). In addition, the active treatment risk difference decreased for every five-year increment in publication year (β = -0.053; 95% CI -0.099 to -0.007; p= 0.024) but was not associated with the placebo response. CONCLUSION Despite increasing over time, the placebo response for ACR20 in PsA RCTs was not associated with the active treatment effect size.
Collapse
Affiliation(s)
- Gian Luca Erre
- Dipartimento di Scienze Mediche, Chirurgiche e Sperimentali, Università degli Studi di Sassari, Sassari, Italy.,Dipartimento di Specialità Mediche, UOC Reumatologia, Azienda Ospedaliero-Universitaria, Sassari, Italy
| | | | - Richard John Woodman
- Flinders Centre for Epidemiology and Biostatistics, College of Medicine and Public Health, Flinders University, Adelaide, Australia
| | - Arduino Aleksander Mangoni
- Discipline of Clinical Pharmacology, College of Medicine and Public Health, Flinders University, Adelaide, Australia.,Department of Clinical Pharmacology, Flinders Medical Centre, Southern Adelaide Local Health Network, Adelaide, Australia
| |
Collapse
|
11
|
Saalfeld W, Mixon AM, Zelie J, Lydon EJ. Differentiating Psoriatic Arthritis from Osteoarthritis and Rheumatoid Arthritis: A Narrative Review and Guide for Advanced Practice Providers. Rheumatol Ther 2021; 8:1493-1517. [PMID: 34519965 PMCID: PMC8572231 DOI: 10.1007/s40744-021-00365-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Accepted: 08/24/2021] [Indexed: 12/18/2022] Open
Abstract
Psoriatic arthritis (PsA) is a chronic inflammatory disease that affects multiple organ systems and is characterized by skin and joint manifestations. PsA is frequently undiagnosed and/or misdiagnosed, especially because of the similarities in clinical presentation shared with other arthritic diseases, including rheumatoid arthritis (RA) and osteoarthritis (OA). An accurate and timely diagnosis of PsA is crucial to prevent delays in optimal treatment, which can lead to irreversible joint damage and increased functional disability. Patients are usually seen by a number of different healthcare providers on their path to a diagnosis of PsA, including advanced practice providers (APPs). This review provides a comprehensive overview of the characteristic features that can be used to facilitate the differentiation of PsA from RA and OA. Detailed information on clinical manifestations, biomarkers, radiologic features, and therapeutic recommendations for PsA included here can be applied in routine clinical settings to provide APPs with the confidence and knowledge to recognize and refer patients more accurately to rheumatologists for management of patients with PsA.
Collapse
Affiliation(s)
- William Saalfeld
- Arthritis Center of Nebraska, 3901 Pine Lake Road, Suite 120, Lincoln, NE, 68516, USA.
| | - Amanda M Mixon
- Arthritis and Rheumatology Clinic of Northern Colorado, Fort Collins, CO, USA
| | - Jonna Zelie
- URMC Division of Rheumatology, Rochester, NY, USA
| | | |
Collapse
|
12
|
Mohanakrishnan R, Beier S, Deodhar A. IL-23 inhibition for the treatment of psoriatic arthritis. Expert Opin Biol Ther 2021; 22:59-65. [PMID: 34092169 DOI: 10.1080/14712598.2021.1938538] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Introduction: Psoriatic arthritis (PsA) is a complex, polygenic immune-mediated disease with varying clinical presentations involving the skin, nails, entheses, and axial/peripheral skeleton.Areas covered: Pathophysiology of PsA with special focus on IL-23/IL-17 axis. Novel classes of targeted therapies for PsA. Pharmacologic properties, efficacy and safety of guselkumab, the only FDA approved agent from IL-23p19 inhibitor class. Data regarding other IL-23 inhibitors (Ustekinumab - an IL-12/IL-23p40 inhibitor, Risankizumab and Tildrakizumab - both IL23p19 inhibitors), in the treatment of PsA.Expert opinion: There are seven classes of FDA-approved therapies for the treatment of PsA. IL-23p19 inhibitors are the newest class of medications that has shown efficacy and reasonable safety profile in the treatment of PsA in phase 2 and phase 3 studies; Guselkumab is the only FDA-approved biologic for PsA within this class . While no head-to-head studies of IL-23p19 inhibitors and other PsA targeted therapies are available, the efficacy of these agents on musculoskeletal system appears to be comparable to TNF-inhibitors (TNFi), and the efficacy on the skin appears to be comparable, or modestly superior to the IL-17 inhibitors (IL-17i). With a superior safety profile compared to TNFi and IL-17i, IL-23p19 inhibitors have the potential to become a first-line biologic in the treatment of PsA.
Collapse
Affiliation(s)
- Raagav Mohanakrishnan
- Division of Arthritis and Rheumatic Diseases (OP09), Oregon Health and Science University, Portland, OR, USA
| | - Secia Beier
- Division of Arthritis and Rheumatic Diseases (OP09), Oregon Health and Science University, Portland, OR, USA
| | - Atul Deodhar
- Division of Arthritis and Rheumatic Diseases (OP09), Oregon Health and Science University, Portland, OR, USA
| |
Collapse
|
13
|
Narode H, Gayke M, Eppa G, Yadav JS. A Review on Synthetic Advances toward the Synthesis of Apremilast, an Anti-inflammatory Drug. Org Process Res Dev 2021. [DOI: 10.1021/acs.oprd.1c00107] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Affiliation(s)
- Hanuman Narode
- Department of Chemistry, School of Science, Indrashil University, Kadi, Mehsana 382715, India
| | - Manoj Gayke
- Department of Chemistry, School of Science, Indrashil University, Kadi, Mehsana 382715, India
| | - Gyanchander Eppa
- Department of Chemistry, School of Science, Indrashil University, Kadi, Mehsana 382715, India
| | - Jhillu Singh Yadav
- Department of Chemistry, School of Science, Indrashil University, Kadi, Mehsana 382715, India
| |
Collapse
|
14
|
Jones GT, Macfarlane GJ, Forrest Keenan K, McNamee P, Neilson AR, Siebert S, Burden AD, Kay L, Helliwell PS. The BSR-PsA: study protocol for the British Society for Rheumatology psoriatic arthritis register. BMC Rheumatol 2021; 5:19. [PMID: 33993880 PMCID: PMC8126428 DOI: 10.1186/s41927-021-00189-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Accepted: 03/10/2021] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND Psoriatic arthritis (PsA) presents a unique clinical challenge. Affecting joints, skin, nails, and other organs, it is associated with various comorbidities and has a significant impact on quality of life, social participation and working life. While biologic and other targeted synthetic disease modifying anti-rheumatic drugs (bDMARDs and tsDMARDs) have revolutionised therapy, questions remain about the long-term safety of these agents, and their effectiveness and cost-effectiveness in the real-world clinical setting. METHODS/DESIGN The British Society for Rheumatology Psoriatic Arthritis Register (BSR-PsA) is a prospective registry of patients with PsA, recruited from across Great Britain, who are (a) commencing a bDMARD/tsDMARD; or (b) naïve to all bDMARDs/tsDMARDs. Ethical approval was given by the NHS West of Scotland Research Ethics Committee 3 (reference: 18/WS/0126). Clinical data are extracted from participants' medical records, including symptom onset and diagnosis, joint, skin and nail symptoms, dactylitis and enthesitis. Physical measurements (height, weight and 66/68 joint counts) and a detailed drug history are taken. Participants are also asked to complete questionnaires comprising instruments relating to general health and quality of life, axial disease, sleep and fatigue, impact of disease, functional status, mental health, other symptoms, and occupational status. The study duration is 5 years in the first instance, and all participants are followed up annually until the end of the study. Participants commencing a bDMARD/tsDMARD are also followed up three and six months after the start of therapy. Disease activity, including C-reactive protein, is assessed at each visit; and participants from some centres are invited to donate blood and urine samples for the creation of a biobank. DISCUSSION Complementing data from randomised trials, results from this study will contribute to the evidence base underpinning the clinical management of psoriatic arthritis. Various analyses will determine the effectiveness and safety of bDMARDs/tsDMARDs in the real-world, will examine the clinical and biological predictors of treatment response, and will provide real-world data on the cost-effectiveness of these therapies, as well as providing informative data important to patients such as quality of life and occupational outcomes. TRIAL REGISTRATION The full study protocol is registered on the Open Science Framework ( https://osf.io/jzs8n ).
Collapse
Affiliation(s)
- Gareth T Jones
- Epidemiology Group, Aberdeen Centre for Arthritis and Musculoskeletal Health, University of Aberdeen, Health Sciences Building, 1st floor, Foresterhill, Aberdeen, AB25 2ZD, UK.
| | - Gary J Macfarlane
- Epidemiology Group, Aberdeen Centre for Arthritis and Musculoskeletal Health, University of Aberdeen, Health Sciences Building, 1st floor, Foresterhill, Aberdeen, AB25 2ZD, UK
| | - Karen Forrest Keenan
- Epidemiology Group, Aberdeen Centre for Arthritis and Musculoskeletal Health, University of Aberdeen, Health Sciences Building, 1st floor, Foresterhill, Aberdeen, AB25 2ZD, UK
| | - Paul McNamee
- Health Economics Research Unit, School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Aberdeen, UK
| | - Aileen R Neilson
- Usher Institute, Edinburgh Medical School: Molecular, Genetic and Population Health Sciences, University of Edinburgh, Edinburgh, UK
| | - Stefan Siebert
- Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, UK
| | - A David Burden
- Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, UK
| | - Lesley Kay
- Musculoskeletal Services Directorate, The Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle, UK
| | - Philip S Helliwell
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Leeds, UK
| |
Collapse
|
15
|
Management of particular clinical situations in psoriatic arthritis: an expert's recommendation document based on systematic literature review and extended Delphi process. Rheumatol Int 2021; 41:1549-1565. [PMID: 33934175 PMCID: PMC8316175 DOI: 10.1007/s00296-021-04877-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Accepted: 04/23/2021] [Indexed: 02/07/2023]
Abstract
To establish practical recommendations for the management of patients with psoriatic arthritis (PsA) with particular clinical situations that might lead to doubts in the pharmacological decision-making. A group of six expert rheumatologists on PsA identified particular clinical situations in PsA. Then, a systematic literature review (SLR) was performed to analyse the efficacy and safety of csDMARDs, b/tsDMARDs in PsA. In a nominal group meeting, the results of the SLR were discussed and a set of recommendations were proposed for a Delphi process. A total of 65 rheumatologists were invited to participate in the Delphi. Agreement was defined if ≥ 70% of the participants voted ≥ 7 (from 1, totally disagree to 10, totally agree). For each recommendation, the level of evidence and grade of recommendation was established based on the Oxford Evidence-Based Medicine categorisation. Particular clinical situations included monoarthritis, axial disease, or non-musculoskeletal manifestations. The SLR finally comprised 131 articles. A total of 16 recommendations were generated, all but 1 reached consensus. According to them, it is crucial to carefully analyse the impact of individual manifestations on patients (disability, quality of life, etc.), but also to recognise the impact of each drug singularities on selected clinical phenotypes to adopt the most appropriate treatment strategy. Early diagnosis and treatment to target approach, along with a close risk management, is also necessary. These recommendations are intended to complement gaps in national and international guidelines by helping health professionals address and manage particular clinical situations in PsA.
Collapse
|
16
|
Epstein PM, Basole C, Brocke S. The Role of PDE8 in T Cell Recruitment and Function in Inflammation. Front Cell Dev Biol 2021; 9:636778. [PMID: 33937235 PMCID: PMC8085600 DOI: 10.3389/fcell.2021.636778] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Accepted: 03/29/2021] [Indexed: 01/07/2023] Open
Abstract
Inhibitors targeting cyclic nucleotide phosphodiesterases (PDEs) expressed in leukocytes have entered clinical practice to treat inflammatory disorders, with three PDE4 inhibitors currently in clinical use as therapeutics for psoriasis, psoriatic arthritis, atopic dermatitis and chronic obstructive pulmonary disease. In contrast, the PDE8 family that is upregulated in pro-inflammatory T cells is a largely unexplored therapeutic target. It was shown that PDE8A plays a major role in controlling T cell and breast cancer cell motility, including adhesion to endothelial cells under physiological shear stress and chemotaxis. This is a unique function of PDE8 not shared by PDE4, another cAMP specific PDE, employed, as noted, as an anti-inflammatory therapeutic. Additionally, a regulatory role was shown for the PDE8A-rapidly accelerated fibrosarcoma (Raf)-1 kinase signaling complex in myelin antigen reactive CD4+ effector T cell adhesion and locomotion by a mechanism differing from that of PDE4. The PDE8A-Raf-1 kinase signaling complex affects T cell motility, at least in part, via regulating the LFA-1 integrin mediated adhesion to ICAM-1. The findings that PDE8A and its isoforms are expressed at higher levels in naive and myelin oligodendrocyte glycoprotein (MOG)35–55 activated effector T (Teff) cells compared to regulatory T (Treg) cells and that PDE8 inhibition specifically affects MOG35–55 activated Teff cell adhesion, indicates that PDE8A could represent a new beneficial target expressed in pathogenic Teff cells in CNS inflammation. The implications of this work for targeting PDE8 in inflammation will be discussed in this review.
Collapse
Affiliation(s)
- Paul M Epstein
- Department of Cell Biology, UConn Health, Farmington, CT, United States
| | - Chaitali Basole
- Department of Immunology, UConn Health, Farmington, CT, United States
| | - Stefan Brocke
- Department of Immunology, UConn Health, Farmington, CT, United States
| |
Collapse
|
17
|
Diel R, Schaberg T, Nienhaus A, Otto-Knapp R, Kneitz C, Krause A, Fabri M, Mrowietz U, Bauer T, Häcker B. Joint Statement (DZK, DGRh, DDG) on the Tuberculosis Risk with Treatment Using Novel Non-TNF-Alpha Biologicals. Pneumologie 2021; 75:293-303. [PMID: 33598901 DOI: 10.1055/a-1294-1580] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
BACKGROUND While the risk of tuberculosis (TB) reactivation is adequately documented in relation to TNF-alpha inhibitors (TNFi), the question of what the tuberculosis risk is for newer, non-TNF biologics (non-TNFi) has not been thoroughly addressed. METHODS We conducted a systematic review of randomized phase 2 and phase 3 studies, and long-term extensions of same, published through March 2019. Of interest was information pertaining to screening and treating of latent tuberculosis (LTBI) in association with the use of 12 particular non-TNFi. Only rituximab was excluded. We searched MEDLINE and the ClinicalTrial.gov database for any and all candidate studies meeting these criteria. RESULTS 677 citations were retrieved; 127 studies comprising a total of 34,293 patients who received non-TNFi were eligible for evaluation. Only 80 out of the 127 studies, or 63 %, captured active TB (or at least opportunistic diseases) as potential outcomes and 25 TB cases were reported. More than two thirds of publications (86/127, 68 %) mentioned LTBI screening prior to inclusion of study participants in the respective trial, whereas in only 4 studies LTBI screening was explicitly considered redundant. In 21 studies, patients with LTBI were generally excluded from the trials and in 42 out of the 127 trials, or 33 %, latently infected patients were reported to receive preventive therapy (PT) at least 3 weeks prior to non-TNFi treatment. CONCLUSIONS The lack of information in many non-TNFi studies on the number of patients with LTBI who were either excluded prior to participating or had been offered PT hampers assessment of the actual TB risk when applying the novel biologics. Therefore, in case of insufficient information about drugs or drug classes, the existing recommendations of the German Central Committee against Tuberculosis should be applied in the same way as is done prior to administering TNFi. Well designed, long-term "real world" register studies on TB progression risk in relation to individual substances for IGRA-positive cases without prior or concomitant PT may help to reduce selection bias and to achieve valid conclusions in the future.
Collapse
Affiliation(s)
- R Diel
- Institute for Epidemiology, University Medical Hospital Schleswig-Holstein, Campus Kiel, Germany. Member of the German Center for Lung Research (ARCN).,LungClinic Grosshansdorf, Germany. Airway Research Center North (ARCN), German Center for Lung Research (DZL).,German Central Committee against Tuberculosis, Berlin, Germany
| | - T Schaberg
- German Central Committee against Tuberculosis, Berlin, Germany
| | - A Nienhaus
- Institution for Statutory Accident Insurance and Prevention in the Health and Welfare Services (BGW), Hamburg, Germany.,Institute for Health Service Research in Dermatology and Nursing, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - R Otto-Knapp
- German Central Committee against Tuberculosis, Berlin, Germany
| | - C Kneitz
- Medicine, Rheumatology, rheumatological main practice Schwerin, Germany
| | - A Krause
- Department of Rheumatology, Clinical Immunology and Osteology, Immanuel Hospital Berlin, Germany
| | - M Fabri
- Department of Dermatology, University of Cologne, Germany
| | - U Mrowietz
- Psoriasis Center, Department of Dermatology, University Medical Center Schleswig-Holstein, Campus Kiel, Germany
| | - T Bauer
- German Central Committee against Tuberculosis, Berlin, Germany
| | - B Häcker
- German Central Committee against Tuberculosis, Berlin, Germany
| |
Collapse
|
18
|
Abstract
Psoriatic arthritis (PsA) is a seronegative inflammatory arthritis often observed in patients with skin psoriasis. Treatment of PsA, especially peripheral PsA, has typically relied on disease-modifying anti-rheumatic agents (DMARDs); however, these agents have limited efficacy and considerable associated toxicity. More recently, monoclonal antibodies (biologic agents) have revolutionized management of immune-mediated diseases; however, these therapies carry a high cost and require parenteral administration. Apremilast, a novel oral DMARD, was approved by the European Union for psoriatic arthritis in 2015. Apremilast inhibits the function of phosphodiesterase-4, a regulator of cyclic adenosine monophosphate, leading to a broad inhibition of proinflammatory mediators and subsequent reduction in tumour necrosis factor-alpha (TNF-α) response. The PALACE and ACTIVE trials, phase III randomized controlled trials for apremilast, showed that apremilast is effective at improving various clinical and patient-reported outcome measures for psoriatic arthritis in both DMARD-naïve and DMARD-experienced PsA patients. Efficacy was limited in patients with previous biologic DMARD failure and the overall efficacy of apremilast appears to be less than biologics agents, though no head-to-head trials exist comparing apremilast to biologic DMARDs. Apremilast is generally well tolerated, with short-lived gastrointestinal side effects being the most commonly reported adverse events. Guidelines suggest a trial of apremilast in patients who have failed traditional oral DMARDs and for whom, biologics are contraindicated. More studies directly comparing apremilast to conventional DMARDs and biologic DMARDs are needed and will be crucial in informing clinical and economic decisions about apremilast role in management of PsA.
Collapse
Affiliation(s)
- Vijay K Sandhu
- Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Lihi Eder
- Division of Rheumatology, Women's College Hospital, Toronto, ON, Canada.,Department of Medicine, University of Toronto, Toronto, ON, Canada
| | - Jensen Yeung
- Department of Medicine, University of Toronto, Toronto, ON, Canada - .,Division of Dermatology, Women's College Hospital, Toronto, ON, Canada.,Division of Dermatology, Sunnybrook Health Sciences Centre, Toronto, ON, Canada.,Probity Medical Research Inc., Waterloo, ON, Canada
| |
Collapse
|
19
|
Chimenti MS, D’Antonio A, Conigliaro P, Ferrigno S, Vendola A, Ferraioli M, Triggianese P, Costa L, Caso F, Perricone R. An Update for the Clinician on Biologics for the Treatment of Psoriatic Arthritis. Biologics 2020; 14:53-75. [PMID: 32903867 PMCID: PMC7445514 DOI: 10.2147/btt.s260754] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Accepted: 08/05/2020] [Indexed: 12/16/2022]
Abstract
Psoriatic arthritis (PsA) is a chronic inflammatory arthropathy typically associated with psoriasis (PsO). The pathogenesis is strictly related to the association among the presence of genetic risk alleles and innate and acquired immune response with dramatic consequences on bone remodeling. Clinically, PsA patients may present heterogenicity of articular and periarticular manifestations that may be associated with the presence of comorbidities making treatment decision challenging in patients management. The identification of patient-targeted therapies is still a critical issue. Actually, several biological and synthetic drugs are promising in terms of efficacy and safety profile. National and international treatment recommendations support clinicians in the decision of the best treatment, although they may have limits basically related to updates and different outcomes included in the clinical studies evaluated. The aim of this narrative review is therefore to give guidance for clinicians for PsA patients treatment. For this purpose, we evaluated evidence on biological therapies efficacy used for PsA treatment. Specifically, we reviewed data on biological therapies, Janus kinases (JAK) inhibitors, and drugs with a new mechanism of action that are part of the treatment pipeline. The concept of "switching" and "swapping" is also described, as well as data concerning special populations such as pregnant women and elderly patients.
Collapse
Affiliation(s)
- Maria Sole Chimenti
- Rheumatology, Allergology and Clinical Immunology, Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Arianna D’Antonio
- Rheumatology, Allergology and Clinical Immunology, Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Paola Conigliaro
- Rheumatology, Allergology and Clinical Immunology, Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Sara Ferrigno
- Rheumatology, Allergology and Clinical Immunology, Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Andrea Vendola
- Rheumatology, Allergology and Clinical Immunology, Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Mario Ferraioli
- Rheumatology, Allergology and Clinical Immunology, Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Paola Triggianese
- Rheumatology, Allergology and Clinical Immunology, Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Luisa Costa
- Rheumatology Unit, Department of Clinical Medicine and Surgery, School of Medicine and Surgery, University Federico II, Naples, Italy
| | - Francesco Caso
- Rheumatology Unit, Department of Clinical Medicine and Surgery, School of Medicine and Surgery, University Federico II, Naples, Italy
| | - Roberto Perricone
- Rheumatology, Allergology and Clinical Immunology, Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| |
Collapse
|
20
|
Qiu M, Xu Z, Gao W, Xiong M, Wen X, Zhu W, Zhou X, Yu M. Fourteen small molecule and biological agents for psoriatic arthritis: A network meta-analysis of randomized controlled trials. Medicine (Baltimore) 2020; 99:e21447. [PMID: 32756163 PMCID: PMC7402795 DOI: 10.1097/md.0000000000021447] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
BACKGROUND The comparative efficacy and safety of small molecule and biological agents in the treatment of psoriatic arthritis (PsA) remain unknown. OBJECTIVES To compare the efficacy and safety of 14 small molecule and biological agents by network meta-analysis (NMA). METHODS Relevant randomized controlled trials involving biological treatments for PsA were identified by searching PubMed, Cochrane Library, EMBASE, Web of Science, and Clinicaltrials.gov and by manual retrieval, up to June 2018. NMA was conducted with Stata 14.0 based on the frequentist method. Effect measures were odds ratios (ORs) with 95% confidence intervals (CIs). Intervention efficacy and safety were ranked according to the surface under the cumulative ranking curve (SUCRA). RESULTS A total of 30 studies involving 10,191 adult subjects were included. According to NMA, ≥ 20% improvement in modifed American College of Rheumatology response criteria (ACR20) response, Psoriasis Area and Severity Index 75 (PASI75) response, and serious adverse events rate (SAEs) were observed. In direct comparisons, most of the biologics performed better than placebo in terms of ACR20 response rate and PASI75 response rate. Additionally, all medicines were comparable to placebo in terms of SAEs except secukinumab. In terms of mixed comparisons, with regard to the ACR20 response, etanercept (ETN) and infliximab (IFX) were more effective than golimumab (GOL), with ORs of 3.33 (95% CI: 1.17-9.48) and 1.24 (95% CI: 0.61-2.52), respectively. For PASI75 response, IFX was superior to certolizumab pegol (OR = 10.08, 95% CI: 1.54-75.48). In addition, these medicines were comparable to each other in terms of SAEs. ETN and IFX were shown to have the most favorable SUCRA for achieving improved ACR20 and PASI75 responses, respectively, while ABT-122 exhibited the best safety according to the SUCRA for SAEs. Considering both the efficacy (ACR20, PASI75) and safety (SAEs), GOL, ETN, and IFX are the top 3 treatments. CONCLUSIONS AND IMPLICATIONS Direct and indirect comparisons and integrated results suggested that the 3 anti- tumor necrosis factor -α biologics (GOL, ETN, and IFX) can be considered the best treatments for PsA after comprehensive consideration of efficacy and safety.
Collapse
Affiliation(s)
- Mingliang Qiu
- Clinical Medical College, Jiangxi University of Traditional Chinese Medicine
- Department of Rheumatology
| | | | - Wenjuan Gao
- Clinical Medical College, Jiangxi University of Traditional Chinese Medicine
| | - Meizhen Xiong
- Clinical Medical College, Jiangxi University of Traditional Chinese Medicine
| | - Xianhua Wen
- Clinical Medical College, Jiangxi University of Traditional Chinese Medicine
| | - Weina Zhu
- Clinical Medical College, Jiangxi University of Traditional Chinese Medicine
- Department of Pediatrics, The Affiliated Hospital of Jiangxi University of Traditional Chinese Medicine
| | - Xu Zhou
- Evidence-based Medicine Research Center, Jiangxi University of Traditional Chinese, Medicine, Nanchang, Jiangxi
| | - Minfeng Yu
- Department of Pediatrics, The Affiliated Hospital of Jiangxi University of Traditional Chinese Medicine
- Department of Pediatrics, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, Guangdong, China
| |
Collapse
|
21
|
Caso F, Navarini L, Ruscitti P, Chimenti MS, Girolimetto N, Del Puente A, Giacomelli R, Scarpa R, Costa L. Targeted synthetic pharmacotherapy for psoriatic arthritis: state of the art. Expert Opin Pharmacother 2020; 21:785-796. [PMID: 32057269 DOI: 10.1080/14656566.2020.1726317] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
INTRODUCTION In recent years, different studies regarding psoriatic arthritis (PsA) have shown the pathogenetic role of dysfunction of signaling pathways involving the phosphodiesterase-4 enzyme and transcription factors or enzymes belonging to the kinase (JAK)-signal family pathway. These also represent the target of several drugs known as targeted synthetic disease-modifying antirheumatic drugs (tsDMARDs). AREAS COVERED The authors performed a systematic literature search using the PubMed database, as well as through retrieving data from randomized controlled trials, their post-hoc analysis, and pooled data analysis on the efficacy and safety profile of the PDE4 inhibitor (PDE4i), apremilast, and the inhibitors of JAK (JAKis), tofacitinib, filgotinib, baricitinib, and upadacitinib, in PsA. EXPERT OPINION In PsA, the PDE4i, apremilast, and the JAKi, tofacitinib, are effective across multiple clinical domains and have an acceptable tolerability profile, thus expanding the treatment options available for PsA patients. Apremilast and tofacitinib show several advantages mainly represented by their oral administration, a fast onset of action, and a short half-life. Data on tsDMARDs in PsA are still limited, and randomized trials and real-life studies are advocated.
Collapse
Affiliation(s)
- Francesco Caso
- Rheumatology Unit, Department of Clinical Medicine and Surgery, University Federico II , Naples, Italy
| | - Luca Navarini
- Unit of Allergology, Clinical Immunology and Rheumatology, Università Campus Bio-Medico Di Roma , Rome, Italy
| | - Piero Ruscitti
- Rheumatology Unit, Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila , L'Aquila, Italy
| | - Maria Sole Chimenti
- Rheumatology, Allergology and Clinical Immunology, Department of System Medicine, University of Rome Tor Vergata , Rome, Italy
| | - Nicolò Girolimetto
- Rheumatology Unit, Department of Clinical Medicine and Surgery, University Federico II , Naples, Italy.,Department of Rheumatology, Azienda USL-IRCCS Di Reggio Emilia , Reggio Emilia, Italy
| | - Antonio Del Puente
- Rheumatology Unit, Department of Clinical Medicine and Surgery, University Federico II , Naples, Italy
| | - Roberto Giacomelli
- Rheumatology Unit, Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila , L'Aquila, Italy
| | - Raffaele Scarpa
- Rheumatology Unit, Department of Clinical Medicine and Surgery, University Federico II , Naples, Italy
| | - Luisa Costa
- Rheumatology Unit, Department of Clinical Medicine and Surgery, University Federico II , Naples, Italy
| |
Collapse
|
22
|
Hagberg KW, Persson R, Vasilakis-Scaramozza C, Niemcryk S, Peng M, Paris M, Lindholm A, Jick S. Herpes Zoster, Hepatitis C, and Tuberculosis Risk with Apremilast Compared to Biologics, DMARDs and Corticosteroids to Treat Psoriasis and Psoriatic Arthritis. Clin Epidemiol 2020; 12:153-161. [PMID: 32104099 PMCID: PMC7024766 DOI: 10.2147/clep.s239511] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Accepted: 01/24/2020] [Indexed: 01/13/2023] Open
Abstract
Purpose Psoriasis and psoriatic arthritis (PsA) are associated with an increased infection risk. In this cohort study of patients with treated psoriasis or PsA, we used MarketScan (2014–2018) to estimate rates of herpes zoster, hepatitis C (HepC) and tuberculosis (TB) with apremilast compared to other systemic treatments. Materials and Methods Patients were exposed from first apremilast [APR], DMARD, TNF-inhibitor [TNF], IL-inhibitor [IL], or corticosteroids [CS] prescription after March 21, 2014. Study exposures were APR, DMARDs only, TNF-only, IL-only, CS-only, DMARDs+CS, TNF+DMARDs and/or CS, IL+DMARDs and/or CS. Cases had treated herpes zoster, HepC, or TB event. We calculated incidence rates (IRs) [95% confidence intervals] per 1000 patient-years. Results The study population included 131,604 patients. For herpes zoster (N=2271), IRs were highest for users of DMARDs+CS (12.5 [9.8–15.7]), CS-only (12.5 [10.4–14.1]), and TNF+DMARDs and/or CS (11.9 [10.6–13.4]), compared with DMARDs only (9.9 [8.7–11.2]). IRs were lowest for users of IL-only (6.7 [5.8–7.8]) and APR (7.0 [5.8–8.4]). IRs of HepC (N=150) and TB (N=81) were low and between-treatment differences were not significant. Conclusion Rates of herpes zoster varied by treatment: highest among those who received polytherapy, lowest in users of apremilast only. IRs for HepC and TB were low for all exposures.
Collapse
Affiliation(s)
| | - Rebecca Persson
- Boston Collaborative Drug Surveillance Program, Lexington, MA 02421, USA
| | | | | | | | | | | | - Susan Jick
- Boston Collaborative Drug Surveillance Program, Lexington, MA 02421, USA.,Boston University School of Public Health, Lexington, MA 02421, USA
| |
Collapse
|
23
|
Tremblay G, Westley T, Forsythe A, Pelletier C, Briggs A. A criterion-based approach to systematic and transparent comparative effectiveness: a case study in psoriatic arthritis. J Comp Eff Res 2019; 8:1265-1298. [PMID: 31774340 DOI: 10.2217/cer-2019-0064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Aim: Indirect treatment comparisons are used when no direct comparison is available. Comparison networks should satisfy the transitivity assumption, that is, equal likelihood of treatment assignment for a given patient based on comparability of studies. Materials & methods: Seven criteria were evaluated across 18 randomized controlled trials in psoriatic arthritis: inclusion/exclusion criteria, clinical trial design and follow-up, patient-level baseline characteristics, disease severity, prior therapies, concomitant and extended-trial treatment and placebo response differences. Results: Across studies, placebo was a common comparator, and key efficacy end points were reported. Collectively, several potential sources of insufficient transitivity were identified, most often related to trial design and population differences. Conclusion: Potential challenges in satisfying transitivity occur frequently and should be evaluated thoroughly.
Collapse
Affiliation(s)
- Gabriel Tremblay
- Purple Squirrel Economics, 4 Lexington Avenue, Suite 15K, New York, NY 10010, USA
| | - Tracy Westley
- Purple Squirrel Economics, 4 Lexington Avenue, Suite 15K, New York, NY 10010, USA
| | - Anna Forsythe
- Purple Squirrel Economics, 4 Lexington Avenue, Suite 15K, New York, NY 10010, USA
| | - Corey Pelletier
- Celgene Corporation, 86 Morris Avenue, Summit, NJ 07901, USA
| | - Andrew Briggs
- Health Economics & Health Technology Assessment, University of Glasgow, 1 Lilybank Gardens, Glasgow G12 8RZ, UK
| |
Collapse
|
24
|
Kogan N, Raimondo N, Gusis SE, Izcovich A, Abarca Duran JA, Barahona-Torres L, Blanco O, Quintana GB, Briones MC, Castro C, Castro Vargas EG, Criniti J, Diez de Medina JC, Franco M, Gómez M, Levrero VP, Martínez López JE, Valenzuela F. Latin American Clinical Practice Guidelines on the Systemic Treatment of Psoriasis SOLAPSO - Sociedad Latinoamericana de Psoriasis (Latin American Psoriasis Society). Int J Dermatol 2019; 58 Suppl 1:4-28. [PMID: 31282026 DOI: 10.1111/ijd.14471] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
This Clinical Practice Guideline on the systemic treatment of Psoriasis includes the recommendations elaborated by a panel of experts from the Latin American Psoriasis Society SOLAPSO, who assessed the quality of the available evidence using the GRADE system and the PICO process to guide the literature search. To answer each question, the experts discussed the results of randomized controlled trials, observational studies and metanalysis evaluating the interventions identified (non-biologics, biologics and phototherapy) in different populations of patients with moderate to severe plaque-psoriasis, which was summarized in Tables ad-hoc. The main end-points considered to assess efficacy were PASI 50, 75, 90 and 100, PGA 0-1 and significant improvement of health-related quality of life. Specific adverse events, either severe or leading to treatment interruption, were also evaluated. The 31 recommendations included in this CPG follow the structure proposed by GRADE: direction (for or against) and strength (strong or weak). The goal of this CPG is to improve the management of patients with psoriasis by recommending interventions of proved benefit and providing a reference standard for the treating physician. Adhering to the contents of this CPG does not guarantee therapeutic success. The final decision on the specific treatment is the responsibility of the physician based on the individual circumstances and considering the values, the preferences and the opinions of the patient or caregivers.
Collapse
Affiliation(s)
- Nora Kogan
- Dermatology, Hospital Ramos Mejía, Buenos Aires, Argentina
| | | | - Simon E Gusis
- Rheumatology, Hospital Ramos Mejía, Buenos Aires, Argentina
| | - Ariel Izcovich
- Program on Evidence Based Medicine, Hospital Aleman, Buenos Aires, Argentina
| | | | | | - Orestes Blanco
- Parasitology, Institute of Tropical Medicine Pedro Kouri, La Habana, Cuba
| | | | - María C Briones
- Centro Privado de Piel "Dr. Enrique Uraga", Guayaquil, Ecuador
| | - Carla Castro
- Pediatric Dermatology, Hospital Universitario Austral, Pilar, Argentina
| | | | - Juan Criniti
- Program on Evidence Based Medicine, Hospital Aleman, Buenos Aires, Argentina
| | | | | | - Minerva Gómez
- Dermatology, University Hospital, Monterrey, Nueva León, Mexico
| | | | | | | |
Collapse
|
25
|
Silvagni E, Bortoluzzi A, Ciancio G, Govoni M. Biological and synthetic target DMARDs in psoriatic arthritis. Pharmacol Res 2019; 149:104473. [PMID: 31585178 DOI: 10.1016/j.phrs.2019.104473] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Revised: 08/28/2019] [Accepted: 09/30/2019] [Indexed: 12/29/2022]
Abstract
Psoriatic arthritis (PsA) is a chronic multi-faceted immune-mediated systemic disorder, characterized by articular, cutaneous, enthesis, nail and spine involvement. Articular manifestations of PsA are particularly common and highly disabling for patients, while the heterogeneous clinical subsets of the disease are challenging for clinicians. In recent years, research has made many advances in understanding the pathogenesis of the disease from genetic, epigenetic and molecular points of view. New drugs are now available for the treatment of this condition, and, in particular, TNF-alfa inhibitors, historically the first biologicals approved in PsA, are now juxtaposed by new biological disease modifying anti-rheumatic drugs (bDMARDs) with different modes of action. Targeting IL-12/IL-23 p40 common subunit with ustekinumab, IL-17A with secukinumab and ixekizumab, T cells co-stimulation with abatacept, is now possible, safe and effective. Moreover, targeted synthetic molecules with oral administration are available, with the possibility to interfere with phosphodiesterase-4 and JAK/STAT pathways. Indeed, new drugs are under development, with the possibility to target selectively IL-17 receptor, IL-23, and other key molecular targets in the pathogenesis of this condition. In this narrative review, we provide an up-to-date overview of the current application of biological and targeted synthetic DMARDs in the field of PsA, with particular regard to the clinical significance of this possibility to target a higher number of distinct immune-pathways.
Collapse
Affiliation(s)
- Ettore Silvagni
- Department of Medical Sciences, Section of Rheumatology, University of Ferrara and Azienda Ospedaliero-Universitaria Sant'Anna, Cona, Ferrara, Italy
| | - Alessandra Bortoluzzi
- Department of Medical Sciences, Section of Rheumatology, University of Ferrara and Azienda Ospedaliero-Universitaria Sant'Anna, Cona, Ferrara, Italy
| | - Giovanni Ciancio
- Department of Medical Sciences, Section of Rheumatology, University of Ferrara and Azienda Ospedaliero-Universitaria Sant'Anna, Cona, Ferrara, Italy.
| | - Marcello Govoni
- Department of Medical Sciences, Section of Rheumatology, University of Ferrara and Azienda Ospedaliero-Universitaria Sant'Anna, Cona, Ferrara, Italy
| |
Collapse
|
26
|
Song GG, Lee YH. Relative efficacy and safety of apremilast, secukinumab, and ustekinumab for the treatment of psoriatic arthritis. Z Rheumatol 2019; 77:613-620. [PMID: 28791450 DOI: 10.1007/s00393-017-0355-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
OBJECTIVE To assess the relative efficacy and safety of apremilast, secukinumab, and ustekinumab at different doses in patients with active psoriatic arthritis (PsA). METHOD A Bayesian network meta-analysis was conducted, which included randomized controlled trials (RCTs) that examined the efficacy and safety of apremilast 20 mg, apremilast 30 mg, secukinumab 75 mg, secukinumab 150 mg, secukinumab 300 mg, ustekinumab 45 mg, and ustekinumab 90 mg compared with placebo. RESULTS Of the RCTs 8 comprising 3289 patients met the inclusion criteria. The American College of Rheumatology (ACR) 20 response rate was significantly higher in the secukinumab 300 mg group than in the placebo group (odds ratio OR, 7.55; 95% confidence interval CI, 3.18-17.63). Secukinumab 150 mg, secukinumab 75 mg, ustekinumab 90 mg, apremilast 30 mg, apremilast 20 mg, and ustekinumab 45 mg were also more efficacious than placebo. There were no significant differences in the efficacy between the interventions. A dose-response relationship among the same drug groups was observed. The number of serious adverse events was not significantly different among the apremilast, secukinumab, ustekinumab, and placebo groups. CONCLUSION All drug treatments were more efficacious than placebo; however, there were no significant differences in the efficacy and safety between the drugs at the different doses.
Collapse
Affiliation(s)
- G G Song
- Division of Rheumatology, Department of Internal Medicine, Korea University Anam Hospital, Korea University College of Medicine, 73, Inchon-ro, Seongbuk-gu, 136-705, Seoul, Korea (Republic of)
| | - Y H Lee
- Division of Rheumatology, Department of Internal Medicine, Korea University Anam Hospital, Korea University College of Medicine, 73, Inchon-ro, Seongbuk-gu, 136-705, Seoul, Korea (Republic of).
| |
Collapse
|
27
|
Schepers M, Tiane A, Paes D, Sanchez S, Rombaut B, Piccart E, Rutten BPF, Brône B, Hellings N, Prickaerts J, Vanmierlo T. Targeting Phosphodiesterases-Towards a Tailor-Made Approach in Multiple Sclerosis Treatment. Front Immunol 2019; 10:1727. [PMID: 31396231 PMCID: PMC6667646 DOI: 10.3389/fimmu.2019.01727] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2019] [Accepted: 07/09/2019] [Indexed: 12/11/2022] Open
Abstract
Multiple sclerosis (MS) is a chronic demyelinating disease of the central nervous system (CNS) characterized by heterogeneous clinical symptoms including gradual muscle weakness, fatigue, and cognitive impairment. The disease course of MS can be classified into a relapsing-remitting (RR) phase defined by periods of neurological disabilities, and a progressive phase where neurological decline is persistent. Pathologically, MS is defined by a destructive immunological and neuro-degenerative interplay. Current treatments largely target the inflammatory processes and slow disease progression at best. Therefore, there is an urgent need to develop next-generation therapeutic strategies that target both neuroinflammatory and degenerative processes. It has been shown that elevating second messengers (cAMP and cGMP) is important for controlling inflammatory damage and inducing CNS repair. Phosphodiesterases (PDEs) have been studied extensively in a wide range of disorders as they breakdown these second messengers, rendering them crucial regulators. In this review, we provide an overview of the role of PDE inhibition in limiting pathological inflammation and stimulating regenerative processes in MS.
Collapse
Affiliation(s)
- Melissa Schepers
- Department of Neuroimmunology, European Graduate School of Neuroscience, Biomedical Research Institute, Hasselt University, Hasselt, Belgium.,Department Psychiatry and Neuropsychology, European Graduate School of Neuroscience, School for Mental Health and Neuroscience, Maastricht University, Maastricht, Netherlands
| | - Assia Tiane
- Department of Neuroimmunology, European Graduate School of Neuroscience, Biomedical Research Institute, Hasselt University, Hasselt, Belgium.,Department Psychiatry and Neuropsychology, European Graduate School of Neuroscience, School for Mental Health and Neuroscience, Maastricht University, Maastricht, Netherlands
| | - Dean Paes
- Department of Neuroimmunology, European Graduate School of Neuroscience, Biomedical Research Institute, Hasselt University, Hasselt, Belgium.,Department Psychiatry and Neuropsychology, European Graduate School of Neuroscience, School for Mental Health and Neuroscience, Maastricht University, Maastricht, Netherlands
| | - Selien Sanchez
- Department of Morphology, Biomedical Research Institute, Hasselt University, Hasselt, Belgium
| | - Ben Rombaut
- Department of Physiology, Biomedical Research Institute, Hasselt University, Hasselt, Belgium
| | - Elisabeth Piccart
- Department of Neuroimmunology, European Graduate School of Neuroscience, Biomedical Research Institute, Hasselt University, Hasselt, Belgium.,Department Psychiatry and Neuropsychology, European Graduate School of Neuroscience, School for Mental Health and Neuroscience, Maastricht University, Maastricht, Netherlands
| | - Bart P F Rutten
- Department Psychiatry and Neuropsychology, European Graduate School of Neuroscience, School for Mental Health and Neuroscience, Maastricht University, Maastricht, Netherlands
| | - Bert Brône
- Department of Physiology, Biomedical Research Institute, Hasselt University, Hasselt, Belgium
| | - Niels Hellings
- Department of Neuroimmunology, European Graduate School of Neuroscience, Biomedical Research Institute, Hasselt University, Hasselt, Belgium
| | - Jos Prickaerts
- Department Psychiatry and Neuropsychology, European Graduate School of Neuroscience, School for Mental Health and Neuroscience, Maastricht University, Maastricht, Netherlands
| | - Tim Vanmierlo
- Department of Neuroimmunology, European Graduate School of Neuroscience, Biomedical Research Institute, Hasselt University, Hasselt, Belgium.,Department Psychiatry and Neuropsychology, European Graduate School of Neuroscience, School for Mental Health and Neuroscience, Maastricht University, Maastricht, Netherlands
| |
Collapse
|
28
|
|
29
|
Comparative efficacy and safety of targeted DMARDs for active psoriatic arthritis during induction therapy: A systematic review and network meta-analysis. Semin Arthritis Rheum 2019; 49:381-388. [PMID: 31272807 DOI: 10.1016/j.semarthrit.2019.06.001] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Revised: 05/30/2019] [Accepted: 06/03/2019] [Indexed: 02/05/2023]
Abstract
OBJECTIVE To summarize and investigate the comparative efficacy and safety of targeted disease-modifying antirheumatic drugs (DMARDs) for active psoriatic arthritis (PsA). METHODS Randomized clinical trials (RCTs) evaluating efficacy and safety of targeted synthetic DMARDs (tofacitinib, apremilast) as well as biological DMARDs (guselkumab, ustekinumab, secukinumab, ixekizumab, brodalumab, clazakizumab, abatacept, adalimumab, etanercept, infliximab, certolizumab, and golimumab) were identified by systemic literature review. Traditional meta-analysis and network meta-analysis using a random effects model were performed to estimate pooled odds ratios (OR) and 95% CI to compare and rank these treatments according to ACR20 response, 75% improvement in psoriasis area and severity index (PASI75), numbers of adverse events (AE) and serious adverse events (SAE). Similar analyses were conducted among biologic-naïve population and biologic-experienced/failed population. RESULTS We deemed 29 RCTs eligible, including 10,204 participants and 17 treatments. During induction therapy (first 12-16 weeks), all treatments except clazakizumab were more efficacious than placebo in achieving ACR20 and PASI75. Although tofacitinib, apremilast, and ixekinumab 80 mg every 2 weeks had a higher rate of AE, no significant difference was revealed for SAE among all treatments. Network meta-analysis demonstrated that infliximab, golimumab, etanercept, adalimumab, guselkumab, and secukinumab 300 mg outperformed other drugs in achieving both ACR20 and PASI75. Infliximab, guselkumab, adalimumab, golimumab, secukinumab (300 mg and 150 mg), and ustekinumab (45 mg and 90 mg) are characterized by both high efficacy and safety. Similar rankings were observed in the analysis among biologic-naïve patients. Moreover, ustekinumab, secukinumab (300 mg and 150 mg), ixekizumab, abatacept, certolizumab pegol, tofacitinib, and apremilast were still associated with higher ACR20 compared to placebo while ustekinumab, secukinumab (300 mg), ixekizumab and tofacitinib with higher PASI75 among biologic-experienced/failed patients. CONCLUSION Regarding the overall risk-benefit profile, infliximab, guselkumab, adalimumab, golimumab, secukinumab, and ustekinumab may be safer and more efficacious treatments than the other targeted DMARDs for active PsA during induction therapy.
Collapse
|
30
|
Song GG, Lee YH. Comparison of the Efficacy and Safety of Tofacitinib and Apremilast in Patients with Active Psoriatic Arthritis: A Bayesian Network Meta-Analysis of Randomized Controlled Trials. Clin Drug Investig 2019; 39:421-428. [PMID: 30806969 DOI: 10.1007/s40261-019-00765-w] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
BACKGROUND Tofacitinib and apremilast have shown considerable efficacy in placebo-controlled trials of active psoriatic arthritis, but the relative efficacy and safety remain unclear because of a lack of head-to-head comparisons. OBJECTIVE The aim of this study was to assess the relative efficacy and safety of tofacitinib and apremilast at different doses in patients with active psoriatic arthritis. METHOD We performed a Bayesian network meta-analysis to combine evidence from randomized controlled trials for examination of the efficacy and safety of tofacitinib 10 mg, tofacitinib 5 mg, apremilast 30 mg, and apremilast 20 mg in psoriatic arthritis. RESULTS Eight randomized controlled trials including 3086 patients met the inclusion criteria. There were ten pairwise comparisons including six direct comparisons of five interventions. All the interventions achieved a significant American College of Rheumatology 20 response compared with placebo. Tofacitinib 10 mg and apremilast 30 mg were among the most effective treatments for active psoriatic arthritis, followed by tofacitinib 5 mg, and apremilast 20 mg. The ranking probability based on the surface under the cumulative ranking curve (SUCRA) indicated that tofacitinib 10 mg had the highest probability of being the best treatment in terms of the American College of Rheumatology 20 response rate (SUCRA = 0.785). This was followed by apremilast 30 mg (SUCRA = 0.670), tofacitinib 5 mg (SUCRA = 0.596), apremilast 20 mg (SUCRA = 0.448), and placebo (SUCRA = 0.001). We observed no significant differences in the incidence of serious adverse events after treatment with tofacitinib 10 mg, apremilast 30 mg, tofacitinib 5 mg, apremilast 20 mg, or placebo. CONCLUSIONS In patients with active psoriatic arthritis, tofacitinib 10 mg and apremilast 30 mg were the most efficacious interventions and were not associated with a significant risk of serious adverse events.
Collapse
Affiliation(s)
- Gwan Gyu Song
- Department of Rheumatology, Korea University College of Medicine, Seoul, Korea.
- Division of Rheumatology, Department of Internal Medicine, Korea University Anam Hospital, Korea University College of Medicine, 73, Inchon-ro, Seongbuk-gu, Seoul, 02841, Korea.
| | - Young Ho Lee
- Department of Rheumatology, Korea University College of Medicine, Seoul, Korea
| |
Collapse
|
31
|
Pham PA, Dressler C, Eisert L, Nast A, Werner RN. Time until onset of action when treating psoriatic arthritis: meta-analysis and novel approach of generating confidence intervals. Rheumatol Int 2019; 39:605-618. [PMID: 30684041 DOI: 10.1007/s00296-019-04244-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2018] [Accepted: 01/16/2019] [Indexed: 02/08/2023]
Abstract
Psoriatic arthritis (PsA) is associated with progressive joint destruction and reduced quality of life. The time until a drug treatment starts to show an effect (TOA) is important for preventing joint destruction. The objective was to assess the time until onset of action of drugs when treating PsA. A systematic review of PsA drug trials was performed. Outcomes were: time until 25% of patients (TOA) reached (1) ≥ 20%, (2) ≥ 50% improvement in modified American College of Rheumatology response criteria (ACR), (3) ≥ 75% reduction in Psoriasis Area and Severity Index (PASI75). 95% confidence intervals were calculated extracting data from graphs using a novel method. Meta-analysis was conducted. Two head-to-head trials show no difference between ixekizumab and adalimumab or adalimumab and tofacitinib for TOA-ACR outcomes. For PASI75, ixekizumab had a faster onset than adalimumab. Infliximab plus MTX was faster than MTX alone. Pooled results from 32 study arms for TOA-ACR20 (week [95% CI]) are: < 2 weeks: infliximab (1.18 [0.72-1.65]), ixekizumab (1.04 [0.80-1.28]), tofacitinib (10 mg 1.56 [1.14-1.98]); ≤ 4 weeks: adalimumab (1.95 [1.35-2.55]), secukinumab (75 mg 1.89 [0.16-3.62], 150 mg 2.13 [1.34-2.91], 300 mg 2.26 [1.75-2.76]), tofacitinib (5 mg 2.20 [1.41-2.99]); 4 + weeks: apremilast, ustekinumab. For TOA-ACR50, all pooled point estimates are > 4 weeks. For TOA-PASI75, the range is between 2.24 [1.65-2.84] for ixekizumab and 6.03 [3.76-8.29] for adalimumab. Indirect, mixed comparison suggest a faster onset of infliximab, ixekizumab and tofacitinib compared to apremilast, methotrexate and ustekinumab for ACR20, not ACR50. For PASI75, ixekizumab is faster than adalimumab.
Collapse
Affiliation(s)
- Phuong Anh Pham
- Division of Evidence-Based Medicine (dEBM), Department of Dermatology, Venerology und Allergy, Berlin Institute of Health, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Charitéplatz 1, 10117, Berlin, Germany
| | - Corinna Dressler
- Division of Evidence-Based Medicine (dEBM), Department of Dermatology, Venerology und Allergy, Berlin Institute of Health, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Charitéplatz 1, 10117, Berlin, Germany.
| | - Lisa Eisert
- Division of Evidence-Based Medicine (dEBM), Department of Dermatology, Venerology und Allergy, Berlin Institute of Health, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Charitéplatz 1, 10117, Berlin, Germany
| | - Alexander Nast
- Division of Evidence-Based Medicine (dEBM), Department of Dermatology, Venerology und Allergy, Berlin Institute of Health, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Charitéplatz 1, 10117, Berlin, Germany
| | - Ricardo Niklas Werner
- Division of Evidence-Based Medicine (dEBM), Department of Dermatology, Venerology und Allergy, Berlin Institute of Health, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Charitéplatz 1, 10117, Berlin, Germany
| |
Collapse
|
32
|
Vandendorpe AS, de Vlam K, Lories R. Evolution of psoriatic arthritis study patient population characteristics in the era of biological treatments. RMD Open 2019; 5:e000779. [PMID: 30740243 PMCID: PMC6347028 DOI: 10.1136/rmdopen-2018-000779] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Revised: 11/29/2018] [Accepted: 12/17/2018] [Indexed: 12/12/2022] Open
Abstract
Objectives Psoriatic arthritis is a chronic inflammatory disease that affects the musculoskeletal system. It can include arthritis, spondylitis, dactylitis and enthesitis, and is strongly associated with the presence of psoriasis. The introduction of biological therapies as a treatment option has brought a significant improvement in disease control for patients with psoriatic arthritis. Here, we aimed to detect emerging differences in demographic and clinical characteristics of the psoriatic arthritis patient study population since the introduction of biologicals. We hypothesised that evolving views on control of disease activity and increased experience in the management of psoriatic arthritis have affected the patient population considered for clinical trials and that this may serve as a proxy for changes in clinical practice. Methods We systematically searched for and selected 12 phase II and phase III trials and divided them into three treatment periods based on different time periods and working mechanisms of the particular biologicals. We made a selection of patient and disease parameters for which data were available in all three periods, calculated those data per period and looked for statistically significant differences between the treatment periods. Results Statistical analysis showed significant differences in patient characteristics, disease characteristics, disease activity, disease effects and use of prior treatments between the patient populations of the three periods. Conclusion This study shows a clear evolution of the patient population considered for clinical trials since the introduction of biologicals. Further research is needed to see if those changes can be detected in the daily clinical practice.
Collapse
Affiliation(s)
- Ann-Sophie Vandendorpe
- Department of Development and Regeneration, Skeletal Biology and Engineering Research Center, KU Leuven, Leuven, Belgium
| | - Kurt de Vlam
- Department of Development and Regeneration, Skeletal Biology and Engineering Research Center, KU Leuven, Leuven, Belgium.,Division of Rheumatology, University Hospitals Leuven, Leuven, Belgium
| | - Rik Lories
- Department of Development and Regeneration, Skeletal Biology and Engineering Research Center, KU Leuven, Leuven, Belgium.,Division of Rheumatology, University Hospitals Leuven, Leuven, Belgium
| |
Collapse
|
33
|
Singh JA, Guyatt G, Ogdie A, Gladman DD, Deal C, Deodhar A, Dubreuil M, Dunham J, Husni ME, Kenny S, Kwan-Morley J, Lin J, Marchetta P, Mease PJ, Merola JF, Miner J, Ritchlin CT, Siaton B, Smith BJ, Van Voorhees AS, Jonsson AH, Shah AA, Sullivan N, Turgunbaev M, Coates LC, Gottlieb A, Magrey M, Nowell WB, Orbai AM, Reddy SM, Scher JU, Siegel E, Siegel M, Walsh JA, Turner AS, Reston J. Special Article: 2018 American College of Rheumatology/National Psoriasis Foundation Guideline for the Treatment of Psoriatic Arthritis. Arthritis Rheumatol 2018; 71:5-32. [PMID: 30499246 DOI: 10.1002/art.40726] [Citation(s) in RCA: 309] [Impact Index Per Article: 44.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2017] [Accepted: 09/11/2018] [Indexed: 12/13/2022]
Abstract
OBJECTIVE To develop an evidence-based guideline for the pharmacologic and nonpharmacologic treatment of psoriatic arthritis (PsA), as a collaboration between the American College of Rheumatology (ACR) and the National Psoriasis Foundation (NPF). METHODS We identified critical outcomes in PsA and clinically relevant PICO (population/intervention/comparator/outcomes) questions. A Literature Review Team performed a systematic literature review to summarize evidence supporting the benefits and harms of available pharmacologic and nonpharmacologic therapies for PsA. GRADE (Grading of Recommendations Assessment, Development and Evaluation) methodology was used to rate the quality of the evidence. A voting panel, including rheumatologists, dermatologists, other health professionals, and patients, achieved consensus on the direction and the strength of the recommendations. RESULTS The guideline covers the management of active PsA in patients who are treatment-naive and those who continue to have active PsA despite treatment, and addresses the use of oral small molecules, tumor necrosis factor inhibitors, interleukin-12/23 inhibitors (IL-12/23i), IL-17 inhibitors, CTLA4-Ig (abatacept), and a JAK inhibitor (tofacitinib). We also developed recommendations for psoriatic spondylitis, predominant enthesitis, and treatment in the presence of concomitant inflammatory bowel disease, diabetes, or serious infections. We formulated recommendations for a treat-to-target strategy, vaccinations, and nonpharmacologic therapies. Six percent of the recommendations were strong and 94% conditional, indicating the importance of active discussion between the health care provider and the patient to choose the optimal treatment. CONCLUSION The 2018 ACR/NPF PsA guideline serves as a tool for health care providers and patients in the selection of appropriate therapy in common clinical scenarios. Best treatment decisions consider each individual patient situation. The guideline is not meant to be proscriptive and should not be used to limit treatment options for patients with PsA.
Collapse
Affiliation(s)
- Jasvinder A Singh
- University of Alabama at Birmingham and Birmingham Veterans Affairs Medical Center, Birmingham, Alabama
| | | | | | - Dafna D Gladman
- University of Toronto and Toronto Western Hospital, Toronto, Ontario, Canada
| | | | | | | | | | | | | | | | - Janice Lin
- Stanford University, Stanford, California
| | | | - Philip J Mease
- Swedish-Providence Health Systems and University of Washington, Seattle, Washington
| | - Joseph F Merola
- Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Julie Miner
- Comprehensive Therapy Consultants and Therapy Steps, Roswell, Georgia
| | | | | | - Benjamin J Smith
- Florida State University College of Medicine School of Physician Assistant Practice, Tallahassee
| | | | - Anna Helena Jonsson
- Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | | | | | | | | | - Alice Gottlieb
- New York Medical College at Metropolitan Hospital, New York, New York
| | | | | | | | - Soumya M Reddy
- New York University School of Medicine, New York, New York
| | - Jose U Scher
- New York University School of Medicine, New York, New York
| | - Evan Siegel
- Arthritis & Rheumatism Associates, Rockville, Maryland
| | | | - Jessica A Walsh
- University of Utah and George E. Wahlen VeteranS Affairs Medical Center, Salt Lake City, Utah
| | - Amy S Turner
- American College of Rheumatology, Atlanta, Georgia
| | | |
Collapse
|
34
|
Singh JA, Guyatt G, Ogdie A, Gladman DD, Deal C, Deodhar A, Dubreuil M, Dunham J, Husni ME, Kenny S, Kwan-Morley J, Lin J, Marchetta P, Mease PJ, Merola JF, Miner J, Ritchlin CT, Siaton B, Smith BJ, Van Voorhees AS, Jonsson AH, Shah AA, Sullivan N, Turgunbaev M, Coates LC, Gottlieb A, Magrey M, Nowell WB, Orbai AM, Reddy SM, Scher JU, Siegel E, Siegel M, Walsh JA, Turner AS, Reston J. 2018 American College of Rheumatology/National Psoriasis Foundation Guideline for the Treatment of Psoriatic Arthritis. ACTA ACUST UNITED AC 2018. [DOI: 10.1177/2475530318812244] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Objective: To develop an evidence-based guideline for the pharmacologic and nonpharmacologic treatment of psoriatic arthritis (PsA), as a collaboration between the American College of Rheumatology (ACR) and the National Psoriasis Foundation (NPF). Methods: We identified critical outcomes in PsA and clinically relevant PICO (population/intervention/comparator/outcomes) questions. A Literature Review Team performed a systematic literature review to summarize evidence supporting the benefits and harms of available pharmacologic and nonpharmacologic therapies for PsA. GRADE (Grading of Recommendations Assessment, Development and Evaluation) methodology was used to rate the quality of the evidence. A voting panel, including rheumatologists, dermatologists, other health professionals, and patients, achieved consensus on the direction and the strength of the recommendations. Results: The guideline covers the management of active PsA in patients who are treatment-naive and those who continue to have active PsA despite treatment, and addresses the use of oral small molecules, tumor necrosis factor inhibitors, interleukin-12/23 inhibitors (IL-12/23i), IL-17 inhibitors, CTLA4-Ig (abatacept), and a JAK inhibitor (tofacitinib). We also developed recommendations for psoriatic spondylitis, predominant enthesitis, and treatment in the presence of concomitant inflammatory bowel disease, diabetes, or serious infections. We formulated recommendations for a treat-to-target strategy, vaccinations, and nonpharmacologic therapies. Six percent of the recommendations were strong and 94% conditional, indicating the importance of active discussion between the health care provider and the patient to choose the optimal treatment. Conclusion: The 2018 ACR/NPF PsA guideline serves as a tool for health care providers and patients in the selection of appropriate therapy in common clinical scenarios. Best treatment decisions consider each individual patient situation. The guideline is not meant to be proscriptive and should not be used to limit treatment options for patients with PsA.
Collapse
Affiliation(s)
- Jasvinder A. Singh
- University of Alabama at Birmingham and Birmingham Veterans Affairs Medical Center, Birmingham, Alabama
| | | | - Alexis Ogdie
- University of Pennsylvania, Philadelphia, PA, USA
| | - Dafna D. Gladman
- University of Toronto and Toronto Western Hospital, Toronto, Ontario, Canada
| | - Chad Deal
- Cleveland Clinic, Cleveland, OH, USA
| | - Atul Deodhar
- Oregon Health & Science University, Portland, OR, USA
| | | | | | | | | | | | | | | | - Philip J. Mease
- Swedish-Providence Health Systems and University of Washington, Seattle, WA, USA
| | - Joseph F. Merola
- Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, USA
| | - Julie Miner
- Comprehensive Therapy Consultants and Therapy Steps, Roswell, GA, USA
| | | | | | - Benjamin J. Smith
- Florida State University College of Medicine School of Physician Assistant Practice, Tallahassee, FL, USA
| | | | | | | | | | | | | | - Alice Gottlieb
- New York Medical College at Metropolitan Hospital, New York, NY, USA
| | | | | | | | | | - Jose U. Scher
- New York University School of Medicine, New York, NY, USA
| | - Evan Siegel
- Arthritis & Rheumatism Associates, Rockville, MA, USA
| | | | - Jessica A. Walsh
- University of Utah and George E. Wahlen Veterans Affairs Medical Center, Salt Lake City, UT, USA
| | | | | |
Collapse
|
35
|
Singh JA, Guyatt G, Ogdie A, Gladman DD, Deal C, Deodhar A, Dubreuil M, Dunham J, Husni ME, Kenny S, Kwan-Morley J, Lin J, Marchetta P, Mease PJ, Merola JF, Miner J, Ritchlin CT, Siaton B, Smith BJ, Van Voorhees AS, Jonsson AH, Shah AA, Sullivan N, Turgunbaev M, Coates LC, Gottlieb A, Magrey M, Nowell WB, Orbai AM, Reddy SM, Scher JU, Siegel E, Siegel M, Walsh JA, Turner AS, Reston J. Special Article: 2018 American College of Rheumatology/National Psoriasis Foundation Guideline for the Treatment of Psoriatic Arthritis. Arthritis Care Res (Hoboken) 2018; 71:2-29. [PMID: 30499259 DOI: 10.1002/acr.23789] [Citation(s) in RCA: 180] [Impact Index Per Article: 25.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2017] [Accepted: 09/11/2018] [Indexed: 12/13/2022]
Abstract
OBJECTIVE To develop an evidence-based guideline for the pharmacologic and nonpharmacologic treatment of psoriatic arthritis (PsA), as a collaboration between the American College of Rheumatology (ACR) and the National Psoriasis Foundation (NPF). METHODS We identified critical outcomes in PsA and clinically relevant PICO (population/intervention/comparator/outcomes) questions. A Literature Review Team performed a systematic literature review to summarize evidence supporting the benefits and harms of available pharmacologic and nonpharmacologic therapies for PsA. GRADE (Grading of Recommendations Assessment, Development and Evaluation) methodology was used to rate the quality of the evidence. A voting panel, including rheumatologists, dermatologists, other health professionals, and patients, achieved consensus on the direction and the strength of the recommendations. RESULTS The guideline covers the management of active PsA in patients who are treatment-naive and those who continue to have active PsA despite treatment, and addresses the use of oral small molecules, tumor necrosis factor inhibitors, interleukin-12/23 inhibitors (IL-12/23i), IL-17 inhibitors, CTLA4-Ig (abatacept), and a JAK inhibitor (tofacitinib). We also developed recommendations for psoriatic spondylitis, predominant enthesitis, and treatment in the presence of concomitant inflammatory bowel disease, diabetes, or serious infections. We formulated recommendations for a treat-to-target strategy, vaccinations, and nonpharmacologic therapies. Six percent of the recommendations were strong and 94% conditional, indicating the importance of active discussion between the health care provider and the patient to choose the optimal treatment. CONCLUSION The 2018 ACR/NPF PsA guideline serves as a tool for health care providers and patients in the selection of appropriate therapy in common clinical scenarios. Best treatment decisions consider each individual patient situation. The guideline is not meant to be proscriptive and should not be used to limit treatment options for patients with PsA.
Collapse
Affiliation(s)
- Jasvinder A Singh
- University of Alabama at Birmingham and Birmingham Veterans Affairs Medical Center, Birmingham, Alabama
| | | | | | - Dafna D Gladman
- University of Toronto and Toronto Western Hospital, Toronto, Ontario, Canada
| | | | | | | | | | | | | | | | - Janice Lin
- Stanford University, Stanford, California
| | | | - Philip J Mease
- Swedish-Providence Health Systems and University of Washington, Seattle, Washington
| | - Joseph F Merola
- Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Julie Miner
- Comprehensive Therapy Consultants and Therapy Steps, Roswell, Georgia
| | | | | | - Benjamin J Smith
- Florida State University College of Medicine School of Physician Assistant Practice, Tallahassee
| | | | - Anna Helena Jonsson
- Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | | | | | | | | | - Alice Gottlieb
- New York Medical College at Metropolitan Hospital, New York, New York
| | | | | | | | - Soumya M Reddy
- New York University School of Medicine, New York, New York
| | - Jose U Scher
- New York University School of Medicine, New York, New York
| | - Evan Siegel
- Arthritis & Rheumatism Associates, Rockville, Maryland
| | | | - Jessica A Walsh
- University of Utah and George E. Wahlen Veterans Affairs Medical Center, Salt Lake City, Utah
| | - Amy S Turner
- American College of Rheumatology, Atlanta, Georgia
| | | |
Collapse
|
36
|
O'Rielly DD, Rahman P. A review of ixekizumab in the treatment of psoriatic arthritis. Expert Rev Clin Immunol 2018; 14:993-1002. [PMID: 30360663 DOI: 10.1080/1744666x.2018.1540931] [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] [Indexed: 01/03/2023]
Abstract
INTRODUCTION Psoriatic arthritis (PsA) is a heterogeneous inflammatory disorder with articular, peri-articular, and extra-articular features along with selected co-morbidities as a sequela to chronic inflammation. There is accumulating evidence that the Th-17 signaling pathway is of critical importance in PsA pathogenesis. Areas covered: Ixekizumab (IXE) is a humanized immunoglobulin G subclass 4 (IgG4) monoclonal antibody directed against IL-17A. Two phase III randomized clinical trials, SPIRIT-P1 and SPIRIT-P2, unequivocally demonstrated superiority of IXE (80 mg every two or 4 weeks) dosing over placebo in moderate-to-severe PsA patients that failed either NSAIDs, conventional disease-modifying anti-rheumatic drugs (csDMARDs), or tumor necrosis factor-α inhibitors (TNFi) for numerous articular and cutaneous parameters. IXE also delayed structural progression of PsA. No new safety signals were identified as compared with chronic plaque psoriasis studies which included many more patients. Expert opinion: IXE is a highly effective treatment for moderate to severe PsA patients, including those that have been previously exposed to csDMARD and TNFi. Most domains of PsA significantly improved with IXE treatment and disease modification was achieved.
Collapse
Affiliation(s)
- Darren D O'Rielly
- a Faculty of Medicine , Memorial University of Newfoundland , St. John's , Newfoundland and Labrador , Canada
| | - Proton Rahman
- a Faculty of Medicine , Memorial University of Newfoundland , St. John's , Newfoundland and Labrador , Canada
| |
Collapse
|
37
|
Elmamoun M, Leung YY, O'Sullivan D, Steinkoenig I, Chandran V, Gladman DD, FitzGerald OM, Orbai AM, Eder L. Using Acute-phase Reactants to Inform the Development of Instruments for the Updated Psoriatic Arthritis Core Outcome Measurement Set. J Rheumatol 2018; 46:266-273. [PMID: 30385708 DOI: 10.3899/jrheum.180195] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/03/2018] [Indexed: 02/08/2023]
Abstract
OBJECTIVE Systemic inflammationˆ is assessed through measurement of acute-phase reactants such as C-reactive protein (CRP) and erythrocyte sedimentation rate (ESR). With few exceptions, most randomized controlled trials (RCT) have assessed acute-phase reactants (CRP and ESR) as part of the American College of Rheumatology (ACR) 20 response criteria. As part of the Group for Research and Assessment of Psoriasis and Psoriatic Arthritis (GRAPPA)-Outcome Measures in Rheumatology (OMERACT) working group, we performed a systematic review of the literature to assess the performance of inflammatory biomarkers in psoriatic arthritis (PsA). METHODS A systematic search of PubMed and Embase was performed. The search included peer-reviewed articles and scientific meeting abstracts about RCT and longitudinal observational studies that assessed systemic inflammation using acute-phase reactants in PsA. Studies were assessed following the components of the OMERACT filter including construct validity, responsiveness, and predictive validity. RESULTS There were 2764 articles retrieved, and 71 articles were included for this systematic review. Twenty-eight articles reported CRP and/or ESR separately, and the remaining articles reported CRP and/or ESR as part of the ACR response criteria. Studies assessing OMERACT responsiveness provided conflicting reports. Inflammatory biomarkers had construct validity for more active disease. Evidence suggests that an elevation of ESR predicts cardiovascular outcomes. CONCLUSION Data regarding assessment of systemic inflammation using acute-phase reactants (CRP and ESR) are limited. There is only weak evidence to support normalization of these biomarkers in predicting good clinical outcomes/remission criteria. The predictive value for cardiovascular outcomes was generally good. Further studies to assess systemic inflammation in PsA using acute-phase reactants and other laboratory biomarkers are needed.
Collapse
Affiliation(s)
- Musaab Elmamoun
- From the Department of Rheumatology, St. Vincent's University Hospital, Dublin, Ireland; Division of Rheumatology, University of Toronto, Krembil Research Institute, Toronto Western Hospital, Toronto, Ontario, Canada; Department of Rheumatology and Immunology, Singapore General Hospital; Duke-NUS Medical School, Singapore; University Hospitals, Cleveland, Ohio, USA; Conway Institute for Biomolecular Research, University College Dublin, Dublin, Ireland; Johns Hopkins University School of Medicine, Division of Rheumatology, Baltimore, Maryland, USA; Women's College Research Institute, Women's College Hospital; Department of Medicine, University of Toronto, Toronto, Ontario, Canada.,M. Elmamoun, MBBS, MRCPI, Department of Rheumatology, St. Vincent's University Hospital, and Division of Rheumatology, University of Toronto, Krembil Research Institute, Toronto Western Hospital; Y.Y. Leung, MBChB, MD, Department of Rheumatology and Immunology, Singapore General Hospital, and Duke-NUS Medical School; D. O'Sullivan, BE, Patient Research Partner, St. Vincent's University Hospital; I. Steinkoenig, BA, Patient Research Partner, University Hospitals; V. Chandran, MBBS, MD, DM, PhD, Division of Rheumatology, University of Toronto, Krembil Research Institute, Toronto Western Hospital; D.D. Gladman, MD, FRCPC, Division of Rheumatology, University of Toronto, Krembil Research Institute, Toronto Western Hospital; O.M. FitzGerald, MD, Department of Rheumatology, St. Vincent's University Hospital, and Conway Institute for Biomolecular Research, University College Dublin; A.M. Orbai, MD, MHS, Johns Hopkins University School of Medicine, Division of Rheumatology; L. Eder, MD, PhD, Women's College Research Institute, Women's College Hospital, and Department of Medicine, University of Toronto
| | - Ying Ying Leung
- From the Department of Rheumatology, St. Vincent's University Hospital, Dublin, Ireland; Division of Rheumatology, University of Toronto, Krembil Research Institute, Toronto Western Hospital, Toronto, Ontario, Canada; Department of Rheumatology and Immunology, Singapore General Hospital; Duke-NUS Medical School, Singapore; University Hospitals, Cleveland, Ohio, USA; Conway Institute for Biomolecular Research, University College Dublin, Dublin, Ireland; Johns Hopkins University School of Medicine, Division of Rheumatology, Baltimore, Maryland, USA; Women's College Research Institute, Women's College Hospital; Department of Medicine, University of Toronto, Toronto, Ontario, Canada.,M. Elmamoun, MBBS, MRCPI, Department of Rheumatology, St. Vincent's University Hospital, and Division of Rheumatology, University of Toronto, Krembil Research Institute, Toronto Western Hospital; Y.Y. Leung, MBChB, MD, Department of Rheumatology and Immunology, Singapore General Hospital, and Duke-NUS Medical School; D. O'Sullivan, BE, Patient Research Partner, St. Vincent's University Hospital; I. Steinkoenig, BA, Patient Research Partner, University Hospitals; V. Chandran, MBBS, MD, DM, PhD, Division of Rheumatology, University of Toronto, Krembil Research Institute, Toronto Western Hospital; D.D. Gladman, MD, FRCPC, Division of Rheumatology, University of Toronto, Krembil Research Institute, Toronto Western Hospital; O.M. FitzGerald, MD, Department of Rheumatology, St. Vincent's University Hospital, and Conway Institute for Biomolecular Research, University College Dublin; A.M. Orbai, MD, MHS, Johns Hopkins University School of Medicine, Division of Rheumatology; L. Eder, MD, PhD, Women's College Research Institute, Women's College Hospital, and Department of Medicine, University of Toronto
| | - Denis O'Sullivan
- From the Department of Rheumatology, St. Vincent's University Hospital, Dublin, Ireland; Division of Rheumatology, University of Toronto, Krembil Research Institute, Toronto Western Hospital, Toronto, Ontario, Canada; Department of Rheumatology and Immunology, Singapore General Hospital; Duke-NUS Medical School, Singapore; University Hospitals, Cleveland, Ohio, USA; Conway Institute for Biomolecular Research, University College Dublin, Dublin, Ireland; Johns Hopkins University School of Medicine, Division of Rheumatology, Baltimore, Maryland, USA; Women's College Research Institute, Women's College Hospital; Department of Medicine, University of Toronto, Toronto, Ontario, Canada.,M. Elmamoun, MBBS, MRCPI, Department of Rheumatology, St. Vincent's University Hospital, and Division of Rheumatology, University of Toronto, Krembil Research Institute, Toronto Western Hospital; Y.Y. Leung, MBChB, MD, Department of Rheumatology and Immunology, Singapore General Hospital, and Duke-NUS Medical School; D. O'Sullivan, BE, Patient Research Partner, St. Vincent's University Hospital; I. Steinkoenig, BA, Patient Research Partner, University Hospitals; V. Chandran, MBBS, MD, DM, PhD, Division of Rheumatology, University of Toronto, Krembil Research Institute, Toronto Western Hospital; D.D. Gladman, MD, FRCPC, Division of Rheumatology, University of Toronto, Krembil Research Institute, Toronto Western Hospital; O.M. FitzGerald, MD, Department of Rheumatology, St. Vincent's University Hospital, and Conway Institute for Biomolecular Research, University College Dublin; A.M. Orbai, MD, MHS, Johns Hopkins University School of Medicine, Division of Rheumatology; L. Eder, MD, PhD, Women's College Research Institute, Women's College Hospital, and Department of Medicine, University of Toronto
| | - Ingrid Steinkoenig
- From the Department of Rheumatology, St. Vincent's University Hospital, Dublin, Ireland; Division of Rheumatology, University of Toronto, Krembil Research Institute, Toronto Western Hospital, Toronto, Ontario, Canada; Department of Rheumatology and Immunology, Singapore General Hospital; Duke-NUS Medical School, Singapore; University Hospitals, Cleveland, Ohio, USA; Conway Institute for Biomolecular Research, University College Dublin, Dublin, Ireland; Johns Hopkins University School of Medicine, Division of Rheumatology, Baltimore, Maryland, USA; Women's College Research Institute, Women's College Hospital; Department of Medicine, University of Toronto, Toronto, Ontario, Canada.,M. Elmamoun, MBBS, MRCPI, Department of Rheumatology, St. Vincent's University Hospital, and Division of Rheumatology, University of Toronto, Krembil Research Institute, Toronto Western Hospital; Y.Y. Leung, MBChB, MD, Department of Rheumatology and Immunology, Singapore General Hospital, and Duke-NUS Medical School; D. O'Sullivan, BE, Patient Research Partner, St. Vincent's University Hospital; I. Steinkoenig, BA, Patient Research Partner, University Hospitals; V. Chandran, MBBS, MD, DM, PhD, Division of Rheumatology, University of Toronto, Krembil Research Institute, Toronto Western Hospital; D.D. Gladman, MD, FRCPC, Division of Rheumatology, University of Toronto, Krembil Research Institute, Toronto Western Hospital; O.M. FitzGerald, MD, Department of Rheumatology, St. Vincent's University Hospital, and Conway Institute for Biomolecular Research, University College Dublin; A.M. Orbai, MD, MHS, Johns Hopkins University School of Medicine, Division of Rheumatology; L. Eder, MD, PhD, Women's College Research Institute, Women's College Hospital, and Department of Medicine, University of Toronto
| | - Vinod Chandran
- From the Department of Rheumatology, St. Vincent's University Hospital, Dublin, Ireland; Division of Rheumatology, University of Toronto, Krembil Research Institute, Toronto Western Hospital, Toronto, Ontario, Canada; Department of Rheumatology and Immunology, Singapore General Hospital; Duke-NUS Medical School, Singapore; University Hospitals, Cleveland, Ohio, USA; Conway Institute for Biomolecular Research, University College Dublin, Dublin, Ireland; Johns Hopkins University School of Medicine, Division of Rheumatology, Baltimore, Maryland, USA; Women's College Research Institute, Women's College Hospital; Department of Medicine, University of Toronto, Toronto, Ontario, Canada.,M. Elmamoun, MBBS, MRCPI, Department of Rheumatology, St. Vincent's University Hospital, and Division of Rheumatology, University of Toronto, Krembil Research Institute, Toronto Western Hospital; Y.Y. Leung, MBChB, MD, Department of Rheumatology and Immunology, Singapore General Hospital, and Duke-NUS Medical School; D. O'Sullivan, BE, Patient Research Partner, St. Vincent's University Hospital; I. Steinkoenig, BA, Patient Research Partner, University Hospitals; V. Chandran, MBBS, MD, DM, PhD, Division of Rheumatology, University of Toronto, Krembil Research Institute, Toronto Western Hospital; D.D. Gladman, MD, FRCPC, Division of Rheumatology, University of Toronto, Krembil Research Institute, Toronto Western Hospital; O.M. FitzGerald, MD, Department of Rheumatology, St. Vincent's University Hospital, and Conway Institute for Biomolecular Research, University College Dublin; A.M. Orbai, MD, MHS, Johns Hopkins University School of Medicine, Division of Rheumatology; L. Eder, MD, PhD, Women's College Research Institute, Women's College Hospital, and Department of Medicine, University of Toronto
| | - Dafna D Gladman
- From the Department of Rheumatology, St. Vincent's University Hospital, Dublin, Ireland; Division of Rheumatology, University of Toronto, Krembil Research Institute, Toronto Western Hospital, Toronto, Ontario, Canada; Department of Rheumatology and Immunology, Singapore General Hospital; Duke-NUS Medical School, Singapore; University Hospitals, Cleveland, Ohio, USA; Conway Institute for Biomolecular Research, University College Dublin, Dublin, Ireland; Johns Hopkins University School of Medicine, Division of Rheumatology, Baltimore, Maryland, USA; Women's College Research Institute, Women's College Hospital; Department of Medicine, University of Toronto, Toronto, Ontario, Canada.,M. Elmamoun, MBBS, MRCPI, Department of Rheumatology, St. Vincent's University Hospital, and Division of Rheumatology, University of Toronto, Krembil Research Institute, Toronto Western Hospital; Y.Y. Leung, MBChB, MD, Department of Rheumatology and Immunology, Singapore General Hospital, and Duke-NUS Medical School; D. O'Sullivan, BE, Patient Research Partner, St. Vincent's University Hospital; I. Steinkoenig, BA, Patient Research Partner, University Hospitals; V. Chandran, MBBS, MD, DM, PhD, Division of Rheumatology, University of Toronto, Krembil Research Institute, Toronto Western Hospital; D.D. Gladman, MD, FRCPC, Division of Rheumatology, University of Toronto, Krembil Research Institute, Toronto Western Hospital; O.M. FitzGerald, MD, Department of Rheumatology, St. Vincent's University Hospital, and Conway Institute for Biomolecular Research, University College Dublin; A.M. Orbai, MD, MHS, Johns Hopkins University School of Medicine, Division of Rheumatology; L. Eder, MD, PhD, Women's College Research Institute, Women's College Hospital, and Department of Medicine, University of Toronto
| | - Oliver M FitzGerald
- From the Department of Rheumatology, St. Vincent's University Hospital, Dublin, Ireland; Division of Rheumatology, University of Toronto, Krembil Research Institute, Toronto Western Hospital, Toronto, Ontario, Canada; Department of Rheumatology and Immunology, Singapore General Hospital; Duke-NUS Medical School, Singapore; University Hospitals, Cleveland, Ohio, USA; Conway Institute for Biomolecular Research, University College Dublin, Dublin, Ireland; Johns Hopkins University School of Medicine, Division of Rheumatology, Baltimore, Maryland, USA; Women's College Research Institute, Women's College Hospital; Department of Medicine, University of Toronto, Toronto, Ontario, Canada.,M. Elmamoun, MBBS, MRCPI, Department of Rheumatology, St. Vincent's University Hospital, and Division of Rheumatology, University of Toronto, Krembil Research Institute, Toronto Western Hospital; Y.Y. Leung, MBChB, MD, Department of Rheumatology and Immunology, Singapore General Hospital, and Duke-NUS Medical School; D. O'Sullivan, BE, Patient Research Partner, St. Vincent's University Hospital; I. Steinkoenig, BA, Patient Research Partner, University Hospitals; V. Chandran, MBBS, MD, DM, PhD, Division of Rheumatology, University of Toronto, Krembil Research Institute, Toronto Western Hospital; D.D. Gladman, MD, FRCPC, Division of Rheumatology, University of Toronto, Krembil Research Institute, Toronto Western Hospital; O.M. FitzGerald, MD, Department of Rheumatology, St. Vincent's University Hospital, and Conway Institute for Biomolecular Research, University College Dublin; A.M. Orbai, MD, MHS, Johns Hopkins University School of Medicine, Division of Rheumatology; L. Eder, MD, PhD, Women's College Research Institute, Women's College Hospital, and Department of Medicine, University of Toronto
| | - Ana-Maria Orbai
- From the Department of Rheumatology, St. Vincent's University Hospital, Dublin, Ireland; Division of Rheumatology, University of Toronto, Krembil Research Institute, Toronto Western Hospital, Toronto, Ontario, Canada; Department of Rheumatology and Immunology, Singapore General Hospital; Duke-NUS Medical School, Singapore; University Hospitals, Cleveland, Ohio, USA; Conway Institute for Biomolecular Research, University College Dublin, Dublin, Ireland; Johns Hopkins University School of Medicine, Division of Rheumatology, Baltimore, Maryland, USA; Women's College Research Institute, Women's College Hospital; Department of Medicine, University of Toronto, Toronto, Ontario, Canada.,M. Elmamoun, MBBS, MRCPI, Department of Rheumatology, St. Vincent's University Hospital, and Division of Rheumatology, University of Toronto, Krembil Research Institute, Toronto Western Hospital; Y.Y. Leung, MBChB, MD, Department of Rheumatology and Immunology, Singapore General Hospital, and Duke-NUS Medical School; D. O'Sullivan, BE, Patient Research Partner, St. Vincent's University Hospital; I. Steinkoenig, BA, Patient Research Partner, University Hospitals; V. Chandran, MBBS, MD, DM, PhD, Division of Rheumatology, University of Toronto, Krembil Research Institute, Toronto Western Hospital; D.D. Gladman, MD, FRCPC, Division of Rheumatology, University of Toronto, Krembil Research Institute, Toronto Western Hospital; O.M. FitzGerald, MD, Department of Rheumatology, St. Vincent's University Hospital, and Conway Institute for Biomolecular Research, University College Dublin; A.M. Orbai, MD, MHS, Johns Hopkins University School of Medicine, Division of Rheumatology; L. Eder, MD, PhD, Women's College Research Institute, Women's College Hospital, and Department of Medicine, University of Toronto
| | - Lihi Eder
- From the Department of Rheumatology, St. Vincent's University Hospital, Dublin, Ireland; Division of Rheumatology, University of Toronto, Krembil Research Institute, Toronto Western Hospital, Toronto, Ontario, Canada; Department of Rheumatology and Immunology, Singapore General Hospital; Duke-NUS Medical School, Singapore; University Hospitals, Cleveland, Ohio, USA; Conway Institute for Biomolecular Research, University College Dublin, Dublin, Ireland; Johns Hopkins University School of Medicine, Division of Rheumatology, Baltimore, Maryland, USA; Women's College Research Institute, Women's College Hospital; Department of Medicine, University of Toronto, Toronto, Ontario, Canada. .,M. Elmamoun, MBBS, MRCPI, Department of Rheumatology, St. Vincent's University Hospital, and Division of Rheumatology, University of Toronto, Krembil Research Institute, Toronto Western Hospital; Y.Y. Leung, MBChB, MD, Department of Rheumatology and Immunology, Singapore General Hospital, and Duke-NUS Medical School; D. O'Sullivan, BE, Patient Research Partner, St. Vincent's University Hospital; I. Steinkoenig, BA, Patient Research Partner, University Hospitals; V. Chandran, MBBS, MD, DM, PhD, Division of Rheumatology, University of Toronto, Krembil Research Institute, Toronto Western Hospital; D.D. Gladman, MD, FRCPC, Division of Rheumatology, University of Toronto, Krembil Research Institute, Toronto Western Hospital; O.M. FitzGerald, MD, Department of Rheumatology, St. Vincent's University Hospital, and Conway Institute for Biomolecular Research, University College Dublin; A.M. Orbai, MD, MHS, Johns Hopkins University School of Medicine, Division of Rheumatology; L. Eder, MD, PhD, Women's College Research Institute, Women's College Hospital, and Department of Medicine, University of Toronto.
| |
Collapse
|
38
|
McInnes IB, Nash P, Ritchlin C, Choy EH, Kanters S, Thom H, Gandhi K, Pricop L, Jugl SM. Secukinumab for psoriatic arthritis: comparative effectiveness versus licensed biologics/apremilast: a network meta-analysis. J Comp Eff Res 2018; 7:1107-1123. [PMID: 30230361 DOI: 10.2217/cer-2018-0075] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
AIM A network meta-analysis using randomized controlled trial data compared psoriatic arthritis (PsA) outcomes (American College of Rheumatology [ACR], Psoriasis Area Severity Index [PASI] and Psoriatic Arthritis Response Criteria [PsARC] response rates) at 12-16 weeks for secukinumab, adalimumab, apremilast, certolizumab, etanercept, golimumab, infliximab and ustekinumab. PATIENTS & METHODS Trials were identified by systematic review. Separate networks were developed for the full-study populations, biologic-naive patients and biologic-experienced patients. RESULTS In the full populations, secukinumab, adalimumab, golimumab and infliximab demonstrated the highest ACR response rates. Secukinumab and infliximab demonstrated the highest PASI response rates, and infliximab and etanercept demonstrated the highest PsARC response rates. CONCLUSION In the full populations, secukinumab demonstrated good efficacy across all outcomes. All treatments for active PsA included in this comprehensive network meta-analysis demonstrated superiority to placebo.
Collapse
Affiliation(s)
- Iain B McInnes
- University of Glasgow, Institute of Infection, Immunity & Inflammation, College of Medical, Veterinary & Life Sciences, University of Glasgow, Sir Graeme Davies Building, 120 University Place, Glasgow G12 8TA, UK
| | - Peter Nash
- Department of Medicine, University of Queensland, Brisbane, QLD 4072, Australia
| | - Christopher Ritchlin
- Division of Allergy, Immunology & Rheumatology, University of Rochester, 601 Elmwood Avenue, Box 695, Rochester, NY 1464, USA
| | - Ernest H Choy
- Institute of Infection & Immunity, Cardiff University School of Medicine, Tenovus Building, Heath Park Campus, Cardiff CF14 4XN, UK
| | - Steve Kanters
- Precision Xtract, 1505 West 2nd Avenue, Suite 300, Vancouver, BC V6H 3Y4, Canada
| | - Howard Thom
- Bristol Medical School: Population Health Sciences, University of Bristol, Canynge Hall, 39 Whatley Road, Bristol BS8 2PS, UK
| | - Kunal Gandhi
- Oncology Global Development Unit, Novartis Pharmaceuticals Corporation, One Health Plaza, Building 337, B04.3B, East Hanover, NJ, USA
| | - Luminita Pricop
- Immunology & Dermatology Franchise, Novartis Pharmaceuticals Corporation, One Health Plaza, Building 337, B04.3B, East Hanover, NJ, USA
| | - Steffen M Jugl
- Global Patient Access Immunology & Dermatology, Novartis Pharma AG, Asklepios 8-1.001.11, Postfach, Basel, CH-4001, Switzerland
| |
Collapse
|
39
|
[Value of combining biologics with methotrexate for treatment of psoriatic arthritis-questions remain]. Z Rheumatol 2018; 77:808-814. [PMID: 30203153 DOI: 10.1007/s00393-018-0533-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
Abstract
BACKGROUND Concomitant methotrexate (MTX) improves the therapeutic effect of biologic therapies in rheumatoid arthritis treatment. However, the influence of MTX on biologic therapy in psoriasis arthritis (PsA) has not yet been fully clarified, as data from randomized clinical studies are lacking. So far, it is only known, that PsA patients with inadequate response to MTX or non-steroidal anti-inflammatory drugs alone respond equally well to a subsequent biologic therapy. OBJECTIVES The aim of this study is to investigate whether MTX-naive patients achieve greater disease improvement with the combination of MTX and a biologic than with biologic monotherapy alone, and whether patients on MTX in whom a biologic therapy is additionally started would worsen if MTX is discontinued. METHODS The current data situation and its limitations are presented. Furthermore, an investigator-initiated multicenter randomized clinical study in patients with active PsA is introduced (MUST study), which investigates the influence of placebo-controlled MTX combination therapy with the interleukin 12/23 inhibitor ustekinumab (UST) in order to close the existing evidence gap. RESULTS The primary objective of the study is to demonstrate the non-inferiority of UST monotherapy compared to MTX/UST combination therapy as measured by mean DAS28 values at week 24. Of 196 planned patients, 77 have been included so far. Recruitment is still open. CONCLUSION The MUST study offers the ideal opportunity to investigate the influence of concomitant MTX in a controlled study design and to assess whether the addition of MTX to UST therapy or its continuation is beneficial for PsA patients.
Collapse
|
40
|
Corbett M, Chehadah F, Biswas M, Moe-Byrne T, Palmer S, Soares M, Walton M, Harden M, Ho P, Woolacott N, Bojke L. Certolizumab pegol and secukinumab for treating active psoriatic arthritis following inadequate response to disease-modifying antirheumatic drugs: a systematic review and economic evaluation. Health Technol Assess 2018; 21:1-326. [PMID: 28976302 DOI: 10.3310/hta21560] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
BACKGROUND Several biologic therapies are approved by the National Institute for Health and Care Excellence (NICE) for psoriatic arthritis (PsA) patients who have had an inadequate response to two or more synthetic disease-modifying antirheumatic drugs (DMARDs). NICE does not specifically recommend switching from one biologic to another, and only ustekinumab (UST; STELARA®, Janssen Pharmaceuticals, Inc., Horsham, PA, USA) is recommended after anti-tumour necrosis factor failure. Secukinumab (SEC; COSENTYX®, Novartis International AG, Basel, Switzerland) and certolizumab pegol (CZP; CIMZIA®, UCB Pharma, Brussels, Belgium) have not previously been appraised by NICE. OBJECTIVE To determine the clinical effectiveness and cost-effectiveness of CZP and SEC for treating active PsA in adults in whom DMARDs have been inadequately effective. DESIGN Systematic review and economic model. DATA SOURCES Fourteen databases (including MEDLINE and EMBASE) were searched for relevant studies from inception to April 2016 for CZP and SEC studies; update searches were run to identify new comparator studies. REVIEW METHODS Clinical effectiveness data from randomised controlled trials (RCTs) were synthesised using Bayesian network meta-analysis (NMA) methods to investigate the relative efficacy of SEC and CZP compared with comparator therapies. A de novo model was developed to assess the cost-effectiveness of SEC and CZP compared with the other relevant comparators. The model was specified for three subpopulations, in accordance with the NICE scope (patients who have taken one prior DMARD, patients who have taken two or more prior DMARDs and biologic-experienced patients). The models were further classified according to the level of concomitant psoriasis. RESULTS Nineteen eligible RCTs were included in the systematic review of short-term efficacy. Most studies were well conducted and were rated as being at low risk of bias. Trials of SEC and CZP demonstrated clinically important efficacy in all key clinical outcomes. At 3 months, patients taking 150 mg of SEC [relative risk (RR) 6.27, 95% confidence interval (CI) 2.55 to 15.43] or CZP (RR 3.29, 95% CI 1.94 to 5.56) were more likely to be responders than patients taking placebo. The NMA results for the biologic-naive subpopulations indicated that the effectiveness of SEC and CZP relative to other biologics and each other was uncertain. Limited data were available for the biologic-experienced subpopulation. Longer-term evidence suggested that these newer biologics reduced disease progression, with the benefits being similar to those seen for older biologics. The de novo model generated incremental cost-effectiveness ratios (ICERs) for three subpopulations and three psoriasis subgroups. In subpopulation 1 (biologic-naive patients who had taken one prior DMARD), CZP was the optimal treatment in the moderate-severe psoriasis subgroup and 150 mg of SEC was optimal in the subgroups of patients with mild-moderate psoriasis or no concomitant psoriasis. In subpopulation 2 (biologic-naive patients who had taken two or more prior DMARDs), etanercept (ETN; ENBREL®, Pfizer Inc., New York City, NY, USA) is likely to be the optimal treatment in all subgroups. The ICERs for SEC and CZP versus best supportive care are in the region of £20,000-30,000 per quality-adjusted life-year (QALY). In subpopulation 3 (biologic-experienced patients or patients in whom biologics are contraindicated), UST is likely to be the optimal treatment (ICERs are in the region of £21,000-27,000 per QALY). The optimal treatment in subpopulation 2 was sensitive to the choice of evidence synthesis model. In subpopulations 2 and 3, results were sensitive to the algorithm for Health Assessment Questionnaire-Disability Index costs. The optimal treatment is not sensitive to the use of biosimilar prices for ETN and infliximab (REMICADE®, Merck Sharp & Dohme, Kenilworth, NJ, USA). CONCLUSIONS SEC and CZP may be an effective use of NHS resources, depending on the subpopulation and subgroup of psoriasis severity. There are a number of limitations to this assessment, driven mainly by data availability. FUTURE WORK Trials are needed to inform effectiveness of biologics in biologic-experienced populations. STUDY REGISTRATION This study is registered as PROSPERO CRD42016033357. FUNDING The National Institute for Health Research Health Technology Assessment programme.
Collapse
Affiliation(s)
- Mark Corbett
- Centre for Reviews and Dissemination, University of York, York, UK
| | - Fadi Chehadah
- Centre for Health Economics, University of York, York, UK
| | - Mousumi Biswas
- Centre for Reviews and Dissemination, University of York, York, UK
| | | | - Stephen Palmer
- Centre for Health Economics, University of York, York, UK
| | - Marta Soares
- Centre for Health Economics, University of York, York, UK
| | - Matthew Walton
- Centre for Reviews and Dissemination, University of York, York, UK
| | - Melissa Harden
- Centre for Reviews and Dissemination, University of York, York, UK
| | - Pauline Ho
- The Kellgren Centre for Rheumatology, Central Manchester and Manchester Children's University Hospitals Trust, Manchester, UK
| | - Nerys Woolacott
- Centre for Reviews and Dissemination, University of York, York, UK
| | - Laura Bojke
- Centre for Health Economics, University of York, York, UK
| |
Collapse
|
41
|
Vossen ARJV, van Doorn MBA, van der Zee HH, Prens EP. Apremilast for moderate hidradenitis suppurativa: Results of a randomized controlled trial. J Am Acad Dermatol 2018; 80:80-88. [PMID: 30482392 DOI: 10.1016/j.jaad.2018.06.046] [Citation(s) in RCA: 82] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Revised: 06/20/2018] [Accepted: 06/20/2018] [Indexed: 01/19/2023]
Abstract
BACKGROUND Effective anti-inflammatory treatments for hidradenitis suppurativa (HS) are limited. OBJECTIVE To evaluate the efficacy and short-term safety of apremilast in patients with moderate HS. METHODS A total of 20 patients with moderate HS were randomized in a 3:1 ratio to receive blinded treatment with apremilast, 30 mg twice daily, or placebo for 16 weeks. The primary outcome was the Hidradenitis Suppurativa Clinical Response at week 16. Linear mixed effects modeling (analysis of covariance) was used to assess secondary clinical outcomes between treatment groups. RESULTS The HS clinical response was met in 8 of 15 patients in the apremilast group (53.3%) and none of 5 patients in the placebo group (0%) (P = .055) at week 16. Moreover, the apremilast-treated patients showed a significantly lower abscess and nodule count (mean difference, -2.6; 95% confidence interval, -6.0 to -0.9; P = .011), NRS for pain (mean difference, -2.7; 95% -4.5 to -0.9; P = .009), and itch (mean difference, -2.8; 95% confidence interval, -5.0 to -0.6; P = .015) over 16 weeks compared with the placebo-treated patients. There was no significant difference in the Dermatology Life Quality Index over time between the 2 treatment groups (mean difference, -3.4; 95% confidence interval, -9.0 to 2.3; P = .230). The most frequently reported adverse events in the apremilast-treated patients were mild-to-moderate headache and gastrointestinal symptoms, which did not result in dropouts. LIMITATIONS Small number of patients, relatively short study duration. CONCLUSION Apremilast, at a dose of 30 mg twice daily, demonstrated clinically meaningful efficacy and was generally well tolerated in patients with moderate HS.
Collapse
Affiliation(s)
- Allard R J V Vossen
- Department of Dermatology, Erasmus University Medical Center, Rotterdam, The Netherlands.
| | - Martijn B A van Doorn
- Department of Dermatology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Hessel H van der Zee
- Department of Dermatology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Errol P Prens
- Department of Dermatology, Erasmus University Medical Center, Rotterdam, The Netherlands
| |
Collapse
|
42
|
Abignano G, Fadl N, Merashli M, Wenham C, Freeston J, McGonagle D, Marzo-Ortega H. Apremilast for the treatment of active psoriatic arthritis: a single-centre real-life experience. Rheumatology (Oxford) 2018; 57:578-580. [PMID: 29272544 DOI: 10.1093/rheumatology/kex454] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2017] [Accepted: 10/25/2017] [Indexed: 12/21/2022] Open
Affiliation(s)
- Giuseppina Abignano
- NIHR Leeds Biomedical Research Centre, Leeds Teaching Hospitals NHS Trust, Leeds.,Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, UK.,Rheumatology Institute of Lucania (IReL), Rheumatology Department of Lucania, San Carlo Hospital of Potenza and Madonna delle Grazie Hospital of Matera, Potenza, Italy
| | - Nafisa Fadl
- NIHR Leeds Biomedical Research Centre, Leeds Teaching Hospitals NHS Trust, Leeds.,Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, UK
| | - Mira Merashli
- NIHR Leeds Biomedical Research Centre, Leeds Teaching Hospitals NHS Trust, Leeds.,Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, UK
| | - Claire Wenham
- NIHR Leeds Biomedical Research Centre, Leeds Teaching Hospitals NHS Trust, Leeds.,Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, UK
| | - Jane Freeston
- NIHR Leeds Biomedical Research Centre, Leeds Teaching Hospitals NHS Trust, Leeds.,Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, UK
| | - Dennis McGonagle
- NIHR Leeds Biomedical Research Centre, Leeds Teaching Hospitals NHS Trust, Leeds.,Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, UK
| | - Helena Marzo-Ortega
- NIHR Leeds Biomedical Research Centre, Leeds Teaching Hospitals NHS Trust, Leeds.,Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, UK
| |
Collapse
|
43
|
Psoriatic disease treatment nowadays: unmet needs among the “jungle of biologic drugs and small molecules”. Clin Rheumatol 2018; 37:1739-1741. [DOI: 10.1007/s10067-018-4090-6] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Revised: 03/24/2018] [Accepted: 03/28/2018] [Indexed: 12/17/2022]
|
44
|
Wells AF, Edwards CJ, Kivitz AJ, Bird P, Nguyen D, Paris M, Teng L, Aelion JA. Apremilast monotherapy in DMARD-naive psoriatic arthritis patients: results of the randomized, placebo-controlled PALACE 4 trial. Rheumatology (Oxford) 2018; 57:1253-1263. [PMID: 29635379 PMCID: PMC6014136 DOI: 10.1093/rheumatology/key032] [Citation(s) in RCA: 73] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2017] [Revised: 01/26/2018] [Indexed: 01/07/2023] Open
Abstract
Objectives The PALACE 4 trial evaluated apremilast monotherapy in patients with active PsA who were DMARD-naive. Methods Eligible patients were randomized (1:1:1) to placebo, apremilast 20 mg twice a day or apremilast 30 mg twice a day. At week 16 or 24, placebo patients were rerandomized to apremilast. Double-blind apremilast treatment continued to week 52, with extension up to 4 years. The primary endpoint was the proportion of patients achieving ⩾20% improvement in ACR response criteria (ACR20) at week 16; secondary endpoints included the mean change in the HAQ Disability Index (HAQ-DI) score at week 16. Results A total of 527 patients with mean disease duration of 3.4 years and high disease activity were randomized and received treatment. More apremilast patients achieved ACR20 response at week 16 [placebo, 15.9%; 20 mg, 28.0% (P = 0.0062); 30 mg, 30.7% (P = 0.0010)]. The mean HAQ-DI improvements were −0.17 (20 mg; P = 0.0008) and −0.21 (30 mg; P < 0.0001) vs 0.03 (placebo). Both apremilast doses showed significant ACR50 responses vs placebo at week 16 and improvements in secondary efficacy measures (swollen/tender joint counts) and psoriasis assessments, with sustained improvements through week 52. Common adverse events (AEs) over 52 weeks were diarrhoea, nausea, headache and upper respiratory tract infection; most events were mild or moderate. Serious AEs and AEs leading to discontinuation were comparable between groups. Laboratory abnormalities were infrequent and transient. Conclusions In DMARD-naive patients, apremilast monotherapy improved PsA signs/symptoms over 52 weeks and was generally well tolerated. Trial registration ClinicalTrials.gov (http://clinicaltrials.gov), NCT01307423.
Collapse
Affiliation(s)
- Alvin F Wells
- Rheumatology and Immunotherapy Center, Franklin, WI, USA
| | - Christopher J Edwards
- NIHR Wellcome Trust Clinical Research Facility, University Hospital Southampton, Southampton, UK
| | - Alan J Kivitz
- Altoona Center for Clinical Research, Duncansville, PA, USA
| | - Paul Bird
- Combined Rheumatology Practice, Kogarah, NSW, Australia
| | | | | | | | | |
Collapse
|
45
|
Abstract
Psoriasis is a chronic immune-mediated disease associated with several co-morbidities and negative impacts on a patient's quality of life. Despite the advances in biologic therapy, there are still unmet needs in the treatment of psoriasis, as current treatments are limited in terms of long-term efficacy, tolerability, safety, route of administration, and cost. Apremilast is an oral, small-molecule phosphodiesterase 4 inhibitor that works intracellularly by blocking the degradation of cyclic adenosine 3',5'-monophosphate, resulting in increased intracellular cyclic adenosine 3',5'-monophosphate levels in phosphodiesterase 4-expressing cells. This inhibition results in the reduced expression of proinflammatory mediators, and an increased expression of anti-inflammatory mediators, providing apremilast with an anti-inflammatory rather than immunosuppressive mode of action. Apremilast offers a novel therapeutic option for patients with psoriasis and psoriatic arthritis and may fulfill some of the unmet needs in patients with psoriasis. Potential advantages of apremilast include moderate activity for both psoriasis and psoriatic arthritis and efficacy in difficult-to-treat forms of psoriasis, a good safety profile, no need of laboratory prescreening or ongoing monitoring for laboratory parameters, owing to the absence of organ toxicity, a potentially advantageous weight loss effect, and a convenient oral administration and dosing. Cost effectiveness and health economics considerations will be decisive in determining the ultimate place of apremilast in the therapeutic armamentarium for psoriasis.
Collapse
Affiliation(s)
- Tiago Torres
- Department of Dermatology, Centro Hospitalar Universitário do Porto, Edifício das Consultas Externas, Ex. CICAP, Rua D. Manuel II, s/n, Porto, Portugal.
- Instituto de Ciências Biomédicas Abel Salazar, University of Porto, Porto, Portugal.
| | - Luis Puig
- Department of Dermatology, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
- Medical School, Universitat Autònoma de Barcelona, Barcelona, Spain
| |
Collapse
|
46
|
Bakirci Ureyen S, Ivory C, Kalyoncu U, Karsh J, Aydin SZ. What does evidence-based medicine tell us about treatments for different subtypes of psoriatic arthritis? A systematic literature review on randomized controlled trials. Rheumatol Adv Pract 2018; 2:rkx019. [PMID: 31431950 PMCID: PMC6649907 DOI: 10.1093/rap/rkx019] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2017] [Revised: 01/04/2018] [Indexed: 12/13/2022] Open
Abstract
OBJECTIVE PsA is a heterogeneous disease with various subtypes of joint manifestations, which can affect the homogeneity of randomized controlled trials (RCTs). The aim of this systematic literature review was to evaluate the inclusion criteria, demographics and outcomes of RCTs to see whether the whole spectrum of PsA was represented. METHODS Medline, EMBASE and Cochrane databases were screened for RCTs on the efficacy of any treatment for PsA up to 4 October 2016 to investigate the inclusion criteria, demographics, outcomes and efficacy. RESULTS Two thousand and sixty-eight abstracts were identified at screening; 76 articles and 52 conference proceedings were included in the final analysis. The main inclusion criteria always included the number of active joints and never axial symptoms, enthesitis nor dactylitis. Only 10 studies provided information about subtypes, of which symmetrical polyarthritis was the main subtype. Mean (s.d.) tender and swollen joints were between 7.8 and 35.8 (1.8-22.1) and between 5.2 and 25.2 (1.5-16.2), respectively. All studies had responses in joint counts as their primary outcome. Responses in enthesitis and dactylitis were usually secondary or tertiary outcomes. Response in BASDAI was among the outcomes in four studies. The comparison of efficacy in polyarticular vs oligoarticular disease was given in three studies, whereas no information was available for DIP joint disease or arthritis mutilans. CONCLUSION There is evidence in the literature to guide clinicians on how to treat PsA patients with polyarticular disease, but there is a gap in knowledge about the other subtypes. PROTOCOL REGISTRATION The study protocol is registered at PROSPERO (CRD42017053907).
Collapse
Affiliation(s)
| | - Catherine Ivory
- University of Ottawa, Faculty of Medicine, Rheumatology, Ottawa, ON, Canada
| | - Umut Kalyoncu
- Faculty of Medicine, Rheumatology, Hacettepe University, Ankara, Turkey
| | - Jacob Karsh
- University of Ottawa, Faculty of Medicine, Rheumatology, Ottawa, ON, Canada
| | - Sibel Zehra Aydin
- University of Ottawa, Faculty of Medicine, Rheumatology, Ottawa, ON, Canada
- Faculty of Medicine, Ottawa Hospital Research Institute, Rheumatology, University of Ottawa, Ottawa, ON, Canada
| |
Collapse
|
47
|
|
48
|
Kawalec P, Holko P, Moćko P, Pilc A. Comparative effectiveness of abatacept, apremilast, secukinumab and ustekinumab treatment of psoriatic arthritis: a systematic review and network meta-analysis. Rheumatol Int 2017; 38:189-201. [PMID: 29285605 PMCID: PMC5773655 DOI: 10.1007/s00296-017-3919-7] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2017] [Accepted: 12/18/2017] [Indexed: 01/05/2023]
Abstract
To assess the comparative effectiveness and safety of novel biologic therapies in psoriatic arthritis (PsA) and to establish the position of the non-anti-tumor necrosis factor α (TNF-α) biologic drugs in the treatment regimen of the disease. A systematic review and network meta-analysis (NMA) was conducted according to the preferred reporting items for systematic reviews and meta-analyses (PRISMA) requirements. Two investigators identified the studies, abstracted data, and assessed the risk of bias independently. The NMA was conducted for efficacy [American College of Rheumatology (ACR) criteria, ACR20 and ACR50; psoriasis area and severity index (PASI), PASI75] and safety outcomes [any adverse events (AEs) and serious adverse events (SAEs)]; treatments were ranked using the P score for each outcome. The PROSPERO registration number was 42017072200. MEDLINE/PubMed, Embase, Cochrane Library, and ClinicalTrials.gov were searched from the inception of each database to July 10, 2017. Randomized controlled trials (RCTs) for abatacept, apremilast, secukinumab or ustekinumab in adults with moderate and severe PsA were included. The overall PsA population and anti-TNF-α-naive, anti-TNF-α-failure, or anti-TNF-α-experienced subpopulations were considered. We identified eight eligible RCTs and included them in the systematic review and NMA. Significant differences in ACR20 response rate were revealed between secukinumab 150 mg and apremilast 20 mg [relative risk; RR = 2.55 (CI-confidence interval; 1.24, 5.23)] and between secukinumab 300 mg and apremilast 20 or 30 mg [RR = 3.57 CI (1.48, 8.64) and RR = 2.84 CI (1.18, 6.86), respectively]. Any AEs occurred more often in apremilast 20 and 30 mg compared with placebo [RR = 0.58 CI (0.45, 0.74) and RR = 0.58 CI (0.45, 0.75), respectively] but also compared with secukinumab 150 mg [RR = 0.54 CI (0.35, 0.81) and RR = 0.45 CI (0.35, 0.82), respectively]. No significant differences were revealed for SAEs among biologics and between biologics and placebo. In the overall population, as well as in the anti-TNF-α-naive subpopulation, secukinumab at a dose of 300 and 150 mg was ranked the highest for the ACR20 endpoint, while in the anti-TNF-α-experienced subpopulation, secukinumab 300 mg and apremilast 30 mg revealed the highest rank. Secukinumab 75 mg was the safest drug in terms of any AEs, but for SEAs the safest was ustekinumab 90 mg. Our study revealed no significant differences among non-anti-TNF-α biologics in the treatment of PsA in the comparisons performed with regards to the highest efficacy and safety. Both in the overall population and in the analyzed subpopulations, secukinumab 300 mg was ranked the highest for the ACR20 response rate. Secukinumab 300 mg was the safest drug in terms of any AEs, and ustekinumab 90 mg presented the lowest overall risk of SAEs. Head-to-head trials and evaluation of comparative efficacy and safety between non-TNF-α biologics are warranted to inform clinical decision making with a relevant treatment paradigm.
Collapse
Affiliation(s)
- P Kawalec
- Drug Management Department, Institute of Public Health, Faculty of Health Sciences, Jagiellonian University Collegium Medicum, Grzegorzecka 20, 31-531, Kraków, Poland.
| | - P Holko
- Drug Management Department, Institute of Public Health, Faculty of Health Sciences, Jagiellonian University Collegium Medicum, Grzegorzecka 20, 31-531, Kraków, Poland
| | - P Moćko
- Drug Management Department, Institute of Public Health, Faculty of Health Sciences, Jagiellonian University Collegium Medicum, Grzegorzecka 20, 31-531, Kraków, Poland
| | - A Pilc
- Drug Management Department, Institute of Public Health, Faculty of Health Sciences, Jagiellonian University Collegium Medicum, Grzegorzecka 20, 31-531, Kraków, Poland.,Institute of Pharmacology Polish Academy of Sciences, Kraków, Poland
| |
Collapse
|
49
|
Wu D, Yue J, Tam LS. Efficacy and safety of biologics targeting interleukin-6, -12/23 and -17 pathways for peripheral psoriatic arthritis: a network meta-analysis. Rheumatology (Oxford) 2017; 57:563-571. [DOI: 10.1093/rheumatology/kex452] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Indexed: 12/30/2022] Open
Affiliation(s)
- Dongze Wu
- Department of Medicine and Therapeutics, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong
| | - Jiang Yue
- Department of Medicine and Therapeutics, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong
| | - Lai-Shan Tam
- Department of Medicine and Therapeutics, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong
| |
Collapse
|
50
|
Torre Alonso JC, Díaz Del Campo Fontecha P, Almodóvar R, Cañete JD, Montilla Morales C, Moreno M, Plasencia-Rodríguez C, Ramírez García J, Queiro R. Recommendations of the Spanish Society of Rheumatology on treatment and use of systemic biological and non-biological therapies in psoriatic arthritis. ACTA ACUST UNITED AC 2017; 14:254-268. [PMID: 29111261 DOI: 10.1016/j.reuma.2017.08.007] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2017] [Accepted: 08/21/2017] [Indexed: 12/19/2022]
Abstract
OBJECTIVE The main purpose of this recommendation statement is to provide clinicians with the best available evidence and the best opinion agreed upon by the panelists for a rational use of synthetic disease modifying antirheumatic drugs (DMARDs) and biologicals in psoriatic arthritis (PsA) patients. The present document also focuses on important aspects in the management of PsA, such as early diagnosis, therapeutic objectives, comorbidities and optimization of treatment. METHODS The recommendations were agreed by consensus by a panel of 8 expert rheumatologists, previously selected by the Spanish Society of Rheumatology (SER) through an open call. The phases of the work were: identification of key areas for updating the previous consensus, analysis and synthesis of scientific evidence (modified Oxford system, Centre for Evidence-based Medicine, 2009) and formulation of recommendations based on this evidence and by consensus techniques. RESULTS Seventeen recommendations were issued for the treatment of PsA patients. Six of them were of general nature, ranging from the early diagnosis and treatment to the importance of assessing comorbidities. The other 11 were focused on the indications for DMARDs and biological therapy in the distinct clinical forms of the disease. Likewise, the situation of failure of the first biological is addressed and treatment algorithms and a table with the different biological therapies are also included. CONCLUSIONS We present the update of SER recommendations for the treatment of PsA with DMARDs and biologics.
Collapse
Affiliation(s)
| | | | - Raquel Almodóvar
- Unidad de Reumatología, Hospital Universitario Fundación Alcorcón, Alcorcón, Madrid, España
| | - Juan D Cañete
- Unidad de Artritis, Servicio de Reumatología, Hospital Clínic, Barcelona, España
| | | | - Mireia Moreno
- Servicio de Reumatología, Parc Taulí Hospital Universitari, Sabadell, Barcelona, España
| | | | - Julio Ramírez García
- Unidad de Artritis, Servicio de Reumatología, IDIBAPS y Hospital Clínic, Barcelona, España
| | - Rubén Queiro
- Sección de Reumatología, Hospital Universitario Central de Asturias, Oviedo, España.
| |
Collapse
|