1
|
Kavanaugh A, Coates LC, Mease PJ, Nowak M, Hippeli L, Lehman T, Banerjee S, Merola JF. Deucravacitinib, a selective, TYK2 inhibitor, in psoriatic arthritis: achievement of minimal disease activity components in a phase 2 trial. Rheumatology (Oxford) 2025; 64:2557-2564. [PMID: 39423145 PMCID: PMC12048048 DOI: 10.1093/rheumatology/keae580] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Revised: 08/29/2024] [Accepted: 09/27/2024] [Indexed: 10/21/2024] Open
Abstract
OBJECTIVES Deucravacitinib is a novel, oral, selective, allosteric tyrosine kinase 2 (TYK2) inhibitor belonging to a distinct class of enzyme inhibitors. In a phase 2 trial in psoriatic arthritis (NCT03881059), deucravacitinib was significantly more efficacious than placebo across multiple endpoints, including achieving minimal disease activity (MDA). This post hoc analysis further evaluated the achievement of individual components of the MDA criteria with deucravacitinib treatment and the time course of responses in the phase 2 trial. METHODS Patients (N = 203) were randomized 1:1:1 to once daily treatment with placebo, deucravacitinib 6 mg or deucravacitinib 12 mg. The proportions of patients achieving MDA and each of the seven individual MDA components through week 16 were assessed. RESULTS At baseline, although some patients met criteria for individual MDA components, none of the patients met the composite MDA criterion, and all components were balanced overall across treatment arms. Treatment with deucravacitinib was associated with a numerically greater mean reduction from baseline in all MDA components vs placebo over 16 weeks of treatment. At week 16, a greater percentage of patients treated with either dose of deucravacitinib vs placebo achieved the threshold criteria for meeting MDA in each of the components. CONCLUSIONS More patients treated with deucravacitinib met each of the MDA components vs placebo, along with a higher rate of MDA response, after 16 weeks of treatment.
Collapse
Affiliation(s)
- Arthur Kavanaugh
- Department of Medicine, University of California, San Diego, School of Medicine, La Jolla, CA, USA
| | - Laura C Coates
- Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, UK
| | - Philip J Mease
- Department of Rheumatology, Swedish Medical Center/Providence St Joseph Health and University of Washington, Seattle, WA, USA
| | | | | | | | | | - Joseph F Merola
- Department of Dermatology and Department of Medicine, Division of Rheumatology, UT Southwestern Medical Center, Dallas, TX, USA
| |
Collapse
|
2
|
Renkli NÖ, Kleinrensink NJ, Spierings J, Mastbergen S, Vonkeman HE, Mooij SC, Schipper LG, Herman A, ten Katen I, Nap FJ, Hol ME, de Jong PA, Jansen MP, Foppen W. Multimodal imaging of structural damage and inflammation in psoriatic arthritis: a comparison of DMARD-naive and DMARD-failure patients. Rheumatology (Oxford) 2025; 64:1760-1769. [PMID: 39153007 PMCID: PMC11962931 DOI: 10.1093/rheumatology/keae450] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Revised: 07/15/2024] [Accepted: 07/23/2024] [Indexed: 08/19/2024] Open
Abstract
OBJECTIVES To compare inflammatory and structural differences in active PsA between DMARD-naive and DMARD-failure patients using diverse imaging approaches for future analyses. Additionally, to explore the influence of patient characteristics (clinical and demographic variables) on imaging findings. METHODS Of the 80 patients included from the first cohort of the ongoing multicentre TOFA-PREDICT trial, 40 were DMARD-naive and 40 were DMARD-failure (csDMARD failure; one prior bDMARD excluding etanercept was allowed), all meeting classification criteria for PsA with a minimum disease duration of eight weeks. Baseline conventional radiographs of hands and feet, MRIs of both ankles, and whole-body [18F]-fluorodeoxyglucose PET/CT (18F-FDG PET/CT) were evaluated for inflammatory and structural imaging parameters, including Sharp-van der Heijde (SHS), Heel Enthesitis Magnetic Resonance Imaging Scoring System (HEMRIS) and Deauville synovitis scoring. Differences between groups and the influence of patient characteristics were examined with multiple linear regression. RESULTS At baseline, patient characteristics were similar between groups. Imaging parameters showed limited inflammation and structural damage. Inflammatory imaging parameters were not significantly different (P > 0.200). Among structural parameters, only HEMRIS Achilles tendon structural damage was significantly different (P = 0.024, R2 = 0.071) and SHS Joint Space Narrowing was not statistically significant (P = 0.050, R2 = 0.048) with higher values for both in DMARD failures. After correction of patient characteristics, these differences in imaging disappeared (both P > 0.600). CONCLUSION At baseline, PsA patient groups were comparable concerning structural and inflammatory imaging parameters, especially after correcting for patient characteristics. Thus, DMARD-naive and DMARD-failure patient groups may be combined in future PsA progression and treatment decision studies. TRIAL REGISTRATION www.clinicaltrialsregister.eu. EudraCT: 2017-003900-28.
Collapse
Affiliation(s)
- Nağme Ö Renkli
- Department of Rheumatology and Clinical Immunology, UMC Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Nienke J Kleinrensink
- Department of Rheumatology and Clinical Immunology, UMC Utrecht, Utrecht University, Utrecht, The Netherlands
- Department of Radiology and Nuclear Medicine, UMC Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Julia Spierings
- Department of Rheumatology and Clinical Immunology, UMC Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Simon Mastbergen
- Department of Rheumatology and Clinical Immunology, UMC Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Harald E Vonkeman
- Department of Rheumatology, Medisch Spectrum Twente, Enschede, The Netherlands
- Department of Psychology, Health and Technology, University of Twente, Enschede, The Netherlands
| | - Shasti C Mooij
- Department of Rheumatology, Medisch Spectrum Twente, Enschede, The Netherlands
| | - Lydia G Schipper
- Department of Rheumatology, Elisabeth- TweeSteden Hospital, Tilburg, The Netherlands
| | - Amin Herman
- Department of Rheumatology, Antonius Hospital, Utrecht, The Netherlands
| | - Iris ten Katen
- Department of Radiology and Nuclear Medicine, UMC Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Frank J Nap
- Department of Radiology and Nuclear Medicine, UMC Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Marjolein E Hol
- Department of Radiology and Nuclear Medicine, UMC Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Pim A de Jong
- Department of Radiology and Nuclear Medicine, UMC Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Mylène P Jansen
- Department of Rheumatology and Clinical Immunology, UMC Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Wouter Foppen
- Department of Radiology and Nuclear Medicine, UMC Utrecht, Utrecht University, Utrecht, The Netherlands
| |
Collapse
|
3
|
Hani U, Paramshetti S, Angolkar M, Alqathanin WK, Alghaseb RS, Al Asmari SM, Alsaab AA, Fatima F, Osmani RAM, Gundawar R. Cyclodextrin-Nanosponge-Loaded Cyclo-Oxygenase-2 Inhibitor-Based Topical Gel for Treatment of Psoriatic Arthritis: Formulation Design, Development, and In vitro Evaluations. Pharmaceuticals (Basel) 2024; 17:1598. [PMID: 39770440 PMCID: PMC11676863 DOI: 10.3390/ph17121598] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Revised: 11/22/2024] [Accepted: 11/24/2024] [Indexed: 01/11/2025] Open
Abstract
Background: Psoriatic arthritis (PsA), a chronic inflammatory disease, mainly affects the joints, with approximately 30% of psoriasis patients eventually developing PsA. Characterized by both innate and adaptive immune responses, PsA poses significant challenges for effective treatment. Recent advances in drug delivery systems have sparked interest in developing novel formulations to improve therapeutic outcomes. The current research focuses on the development and evaluation of a nanosponge-loaded, cyclo-oxygenase-2 (COX-2) inhibitor-based topical gel for the treatment of PsA. Methods: Nanosponges (NSs) were prepared by using beta-cyclodextrin as a polymer and dimethyl carbonate (DMC) as a crosslinker by melting, and gels were prepared by employing carbopol and badam gum as polymers. Results: Solubility studies confirmed that the prepared nanosponges were highly soluble. FT-IR studies confirmed the formation of hydrogen bonds between lumiracoxib and beta-cyclodextrin. SEM confirmed that the prepared formulations were roughly spherical and porous in nature. The average particle size was 190.5 ± 0.02 nm, with a zeta potential of -18.9 mv. XRD studies showed that the crystallinity of lumiracoxib decreased after encapsulation, which helped to increase its solubility. The optimized nanosponges (NS2) were incorporated in an optimized gel (FG10) to formulate a nanosponge-loaded topical gel. The optimized gel formulation exhibited a homogeneous consistency, with a pH of 6.8 and a viscosity of 1.15 PaS, indicating its suitability for topical application and stability. The in vitro diffusion studies for the topical gel showed drug release of 82.32% in 24 h. The optimized formulation demonstrated significant antipsoriatic activity, as confirmed through cytotoxicity studies conducted on HaCaT cells. Conclusions: On the basis of the findings, it can be concluded that the prepared nanosponge-loaded topical gel formulation presents a promising solution for the effective management of PsA, offering enhanced drug solubility, sustained release, and improved therapeutic potential.
Collapse
Affiliation(s)
- Umme Hani
- Department of Pharmaceutics, College of Pharmacy, King Khalid University, Abha 62529, Saudi Arabia;
| | - Sharanya Paramshetti
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education and Research (JSS AHER), Mysuru 570015, Karnataka, India; (S.P.); (M.A.)
| | - Mohit Angolkar
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education and Research (JSS AHER), Mysuru 570015, Karnataka, India; (S.P.); (M.A.)
| | - Wajan Khalid Alqathanin
- Department of Doctor of Pharmacy, College of Pharmacy, King Khalid University, Abha 62529, Saudi Arabia; (W.K.A.); (R.S.A.); (S.M.A.A.)
| | - Reema Saeed Alghaseb
- Department of Doctor of Pharmacy, College of Pharmacy, King Khalid University, Abha 62529, Saudi Arabia; (W.K.A.); (R.S.A.); (S.M.A.A.)
| | - Saja Mohammed Al Asmari
- Department of Doctor of Pharmacy, College of Pharmacy, King Khalid University, Abha 62529, Saudi Arabia; (W.K.A.); (R.S.A.); (S.M.A.A.)
| | - Alhanouf A. Alsaab
- Pharmacist at Abha International Private Hospital, Abha 62521, Saudi Arabia;
| | - Farhat Fatima
- Department of Pharmaceutics, College of Pharmacy, Prince Sattam bin Abdulaziz University, Al-Kharj 11942, Saudi Arabia;
| | - Riyaz Ali M. Osmani
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education and Research (JSS AHER), Mysuru 570015, Karnataka, India; (S.P.); (M.A.)
| | - Ravi Gundawar
- Department of Pharmaceutical Quality Assurance, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education (MAHE), Manipal 576104, Karnataka, India;
| |
Collapse
|
4
|
Yi RC, Gantz HY, Shah SC, Moran SK, Klionsky YE, Feldman SR. Pharmacotherapeutic management of psoriatic disease: addressing psoriatic arthritis and cutaneous manifestations. Expert Opin Pharmacother 2024; 25:2203-2212. [PMID: 39422251 DOI: 10.1080/14656566.2024.2419563] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Revised: 09/27/2024] [Accepted: 10/17/2024] [Indexed: 10/19/2024]
Abstract
INTRODUCTION Psoriasis and psoriatic arthritis (PsA) are interrelated autoimmune conditions sharing similar genetic and immunological pathways. PsA often develops within 10 years of psoriasis onset, though it may precede cutaneous symptoms in some patients. Effective management of these conditions requires a multidisciplinary approach to address skin, bone, joint, and vascular manifestations. AREAS COVERED The review summarizes the current pharmacotherapies and to provide treatment guidelines for managing cutaneous psoriasis and PsA in psoriatic disease. EXPERT OPINION The management for mild psoriasis and mild PsA flare-ups can be addressed with topical treatments and with NSAIDs or intra-articular glucocorticoid injections. For more persistent or severe cases, systemic treatments with oral small molecules (Methotrexate, Apremilast, Janus kinase inhibitors) or with biologics (TNF inhibitors, IL-17 inhibitors, IL-23 inhibitors, CTLA-4 Ig) are effective in managing both psoriasis and PsA. With many treatment options, providers can tailor management, which considers patient disease severity, preference, comorbidities, and other factors. Early detection and a multidisciplinary management strategy can optimize patient quality of life and improve health outcomes.
Collapse
Affiliation(s)
- Robin C Yi
- Center for Dermatology Research, Department of Dermatology, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Hannah Y Gantz
- Center for Dermatology Research, Department of Dermatology, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Shailey C Shah
- Center for Dermatology Research, Department of Dermatology, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Shannon K Moran
- Center for Dermatology Research, Department of Dermatology, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Yael E Klionsky
- Department of Internal Medicine, Section of Rheumatology, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Steven R Feldman
- Center for Dermatology Research, Department of Dermatology, Wake Forest University School of Medicine, Winston-Salem, NC, USA
- Department of Pathology, Wake Forest University School of Medicine, Winston-Salem, NC, USA
- Department of Social Sciences & Health Policy, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| |
Collapse
|
5
|
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
|
6
|
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
|
7
|
Semele R, Grewal S, Jeengar MK, Singh TG, Swami R. From Traditional Medicine to Advanced Therapeutics: The Renaissance of Phyto-nano Interventions in Psoriasis. RECENT ADVANCES IN INFLAMMATION & ALLERGY DRUG DISCOVERY 2024; 18:27-42. [PMID: 37921124 DOI: 10.2174/0127722708265612231012080047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 07/26/2023] [Accepted: 08/11/2023] [Indexed: 11/04/2023]
Abstract
Psoriasis is an autoimmune systemic chronic inflammatory disease that exhibits characteristic detrimental effects on the skin, often leading to infections or comorbid conditions. The multifaceted nature of psoriasis has made it very challenging to treat, especially with current chemotherapy options. Therefore, it is essential to consider phytoconstituents as novel alternatives. However, despite demonstrating higher anti-inflammatory, anti-psoriasis, and immunomodulatory potential, their clinical usage is hindered due to their poor physicochemical properties. To address these drawbacks, nanoparticulate drug delivery systems have been developed, helping to achieve better permeation of phytoconstituents through topical administration. This has breathed new life into traditional systems of medicine, particularly in the context of treating psoriasis. In this current review, we present a detailed, comprehensive, and up-to-date analysis of the literature, which will contribute to affirming the clinical role of phyto-nano interventions against psoriasis.
Collapse
Affiliation(s)
- Rajneesh Semele
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | - Sonam Grewal
- Maharishi Markandeshwar College of Pharmacy, MMDU, Mullana, Haryana, India
| | - Manish Kumar Jeengar
- Department of Pharmacology, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, Kochi, Kerala 682041, India
| | | | - Rajan Swami
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| |
Collapse
|
8
|
Vahidy AS, Niaz F, Tariq S, Fatima I, Afzal Y, Nashwan AJ. IL-23 Inhibitors to treat psoriatic arthritis: A systematic review & meta-analysis of randomized controlled trials. CLINICAL IMMUNOLOGY COMMUNICATIONS 2023; 4:7-22. [DOI: 10.1016/j.clicom.2023.07.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/11/2025]
|
9
|
Broadwell A, Schechtman J, Conaway D, Kivitz A, Shiff NJ, Black S, Xu S, Langholff W, Schwartzman S, Curtis JR. Effectiveness and safety of intravenous golimumab with and without concomitant methotrexate in patients with rheumatoid arthritis in the prospective, noninterventional AWARE study. BMC Rheumatol 2023; 7:5. [PMID: 36973741 PMCID: PMC10045110 DOI: 10.1186/s41927-023-00329-8] [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: 05/23/2022] [Accepted: 03/07/2023] [Indexed: 03/29/2023] Open
Abstract
BACKGROUND Biologic therapies are often prescribed for patients with rheumatoid arthritis (RA) who have inadequate responses to or are intolerant of methotrexate (MTX) and patients with poor prognostic indicators. This post hoc analysis evaluated effectiveness and safety of intravenous golimumab + MTX vs golimumab without MTX in RA patients. METHODS AWARE, a real-world, prospective and pragmatic, Phase 4 study, compared effectiveness and safety of golimumab and infliximab in biologic-naïve and biologic-experienced patients. All treatment decisions were at the discretion of the treating rheumatologist. Effectiveness was evaluated by mean change in CDAI scores at Months 6 and 12. Safety was monitored through approximately 1 year. RESULTS Among 685 golimumab-treated patients, 420 (61%) received concomitant MTX during the study and 265 (39%) did not receive MTX after enrollment; 63% and 72%, respectively, discontinued the study. Relative to golimumab without MTX, golimumab + MTX patients had shorter mean disease duration (8.7 vs 10.0 years) and a lower proportion received prior biologics (60% vs 72%); mean ± standard deviation (SD) baseline CDAI scores were similar (30.8 ± 15.1 and 32.6 ± 15.4). Mean ± SD changes from baseline in CDAI scores at Months 6 and 12, respectively, were similar with golimumab + MTX (- 10.2 ± 14.2 and - 10.8 ± 13.8) and golimumab without MTX (- 9.6 ± 12.9 and - 9.9 ± 13.1). The incidence of adverse events/100 patient-years (PY) (95% confidence interval [CI]) was 155.6 (145.6, 166.1) for golimumab + MTX and 191.2 (176.2, 207.1) for golimumab without MTX; infections were the most common type. The incidence of infusion reactions/100PY (95% CI) was 2.1 (1.1, 3.6) for golimumab + MTX versus 5.1 (2.9, 8.3) for golimumab without MTX; none were considered serious. For golimumab + MTX versus golimumab without MTX, rates/100PY (95% CI) of serious infections, opportunistic infections, and malignancies were 2.6 (1.5, 4.3) versus 7.0 (4.4, 10.6), 0.9 (0.3, 2.0) versus 2.6 (1.1, 5.0), and 3.0 (1.7, 4.7) versus 1.0 (0.2, 2.8), respectively. CONCLUSIONS Mean change in CDAI score in the golimumab without MTX group was generally similar to that of the golimumab + MTX group through 1 year, regardless of prior biologic therapy. Adverse events were consistent with the known IV golimumab safety profile. These results provide real world evidential data that may assist healthcare providers and patients with RA in making informed treatment decisions. TRIAL REGISTRATION clinicaltrials.gov NCT02728934 05/04/2016.
Collapse
Affiliation(s)
- Aaron Broadwell
- Rheumatology and Osteoporosis Specialists, 820 Jordan Street, Suite 201, Shreveport, LA, 71101, USA.
| | | | | | - Alan Kivitz
- Altoona Center for Clinical Research, Duncansville, PA, USA
| | - Natalie J Shiff
- Janssen Scientific Affairs, LLC, Horsham, PA, USA
- Adjunct, Community Health and Epidemiology, University of Saskatchewan, Saskatoon, Canada
| | - Shawn Black
- Janssen Research and Development, LLC, Spring House, PA, USA
| | - Stephen Xu
- Janssen Research and Development, LLC, Spring House, PA, USA
| | - Wayne Langholff
- Janssen Research and Development, LLC, Spring House, PA, USA
| | | | | |
Collapse
|
10
|
Gottlieb AB, McInnes IB, Rahman P, Kollmeier AP, Xu XL, Jiang Y, Sheng S, Shawi M, Chakravarty SD, Lavie F, Mease PJ. Low rates of radiographic progression associated with clinical efficacy following up to 2 years of treatment with guselkumab: results from a phase 3, randomised, double-blind, placebo-controlled study of biologic-naïve patients with active psoriatic arthritis. RMD Open 2023; 9:rmdopen-2022-002789. [PMID: 36828643 PMCID: PMC9972414 DOI: 10.1136/rmdopen-2022-002789] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Accepted: 02/02/2023] [Indexed: 02/26/2023] Open
Abstract
OBJECTIVE Evaluate relationship between radiographic progression and clinical outcomes in post hoc analyses of patients with psoriatic arthritis (PsA) receiving up to 2 years of guselkumab therapy in the phase 3, placebo-controlled, randomised trial, DISCOVER-2. METHODS Biologic-naïve adults with active PsA (≥5 swollen joints /≥5 tender joints ; C reactive protein ≥0.6 mg/dL) were randomised to guselkumab 100 mg every 4 weeks (Q4W); guselkumab 100 mg at week 0, week 4, then every 8 weeks (Q8W); or placebo→guselkumab 100 mg Q4W (week 24). Radiographs (hands/feet) at week 0, week 24, week 52 and week 100 were scored via PsA-modified van der Heijde-Sharp (vdH-S) methodology. In these post hoc analyses, mean changes in vdH-S scores were summarised according to achievement of American College of Rheumatology 20/50/70 response; low disease activity (LDA) defined by Disease Activity in Psoriatic Arthritis (DAPSA) ≤14 or Psoriatic ArthritiS Disease Activity Score (PASDAS) ≤3.2, or minimal/very low disease activity (MDA/VLDA); and normalised physical function (Health Assessment Questionnaire-Disability Index (HAQ-DI) ≤0.5). Response rates for achieving MDA/VLDA and each component were determined among patients with and without radiographic progression (change in total vdH-S score >0.5). No formal hypothesis testing was performed. RESULTS 664 of 739 treated patients in DISCOVER-2 continued study treatment at week 52 and were included in these analyses. Mean changes in vdH-S scores from weeks 0 to 100 among all patients in the Q4W and Q8W groups were 1.7 and 1.5, respectively. Among all guselkumab-randomised patients, those who achieved ACR20/50/70, DAPSA LDA, PASDAS LDA, MDA, VLDA and HAQ-DI ≤0.5 (normalised physical function) had smaller mean changes in vdH-S scores than did non-responders at week 52 (0.2-1.2 vs 1.7-4.1) and week 100 (0.3-1.2 vs 2.0-4.6). Relative to patients with radiographic progression, those without progression were more likely to achieve the MDA criteria related to swollen and tender joint counts, patient-reported pain and global assessment, and normalised physical function through week 100. CONCLUSION In these post hoc analyses, the achievement of low levels of disease activity, including MDA, was associated with diminished rates of radiographic progression observed in patients receiving up to 2 years of guselkumab. Radiographic non-progressors were more likely to achieve patient-reported MDA criteria of minimal pain and normalised physical function compared with radiographic non-responders. TRIAL REGISTRATION NUMBER NCT03158285.
Collapse
Affiliation(s)
- Alice B Gottlieb
- Dermatology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Iain B McInnes
- College of Medical Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Proton Rahman
- Rheumatology, Memorial University, St John's, Newfoundland, Canada
| | - Alexa P Kollmeier
- Immunology, Janssen Research & Development, San Diego, California, USA
| | - Xie L Xu
- Immunology, Janssen Research & Development, San Diego, California, USA
| | - Yusang Jiang
- Cytel Inc on behalf of Janssen Research & Development LLC, Spring House, Pennsylvania, USA
| | - Shihong Sheng
- Janssen Research & Development, Spring House, Pennsylvania, USA
| | - May Shawi
- Immunology Global Medical Affairs, Janssen Pharmaceutical Companies of Johnson & Johnson, Horsham, Pennsylvania, USA
| | - Soumya D Chakravarty
- Immunology, Janssen Scientific Affairs, Horsham, Pennsylvania, USA,Rheumatology, Drexel University College of Medicine, Philadelphia, Pennsylvania, USA
| | - Frederic Lavie
- Immunology Global Medical Affairs, Janssen Cilag Global Medical Affairs, Issy les Moulineaux, France
| | - Philip J Mease
- School of Medicine, Swedish Medical Center and University of Washington, Seattle, Washington, USA
| |
Collapse
|
11
|
Alnaqbi KA, Hannawi S, Namas R, Alshehhi W, Badsha H, Al‐Saleh J. Consensus statements for pharmacological management, monitoring of therapies, and comorbidity management of psoriatic arthritis in the United Arab Emirates. Int J Rheum Dis 2022; 25:1107-1122. [PMID: 35916205 PMCID: PMC9804226 DOI: 10.1111/1756-185x.14406] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Revised: 06/28/2022] [Accepted: 07/17/2022] [Indexed: 01/05/2023]
Abstract
OBJECTIVE Psoriatic arthritis (PsA), a chronic inflammatory disease characterized by heterogeneous clinical manifestations, substantially impacts the quality of life of affected individuals. This article aims at developing consensus recommendations for the management of PsA and associated comorbidities and screening and monitoring requirements of PsA therapies in the United Arab Emirates (UAE) population. METHODS An extensive review of present international and regional guidelines and publications on the pharmacological management, monitoring of therapies in the context of PsA was performed. Key findings from guidelines and literature were reviewed by a panel of experts from the UAE at several meetings to align with current clinical practices. Consensus statements were formulated based on collective agreement of the experts and members of Emirates Society for Rheumatology. RESULTS The consensus recommendations were developed to aid practitioners in clinical decision-making with respect to dosage recommendations for pharmacological therapies for PsA, including conventional drugs, non-biologic, and biologic therapies. Consensus recommendations for therapeutic options for the treatment of PsA domains, including peripheral arthritis, axial disease, enthesitis, dactylitis, psoriasis, and nail disease, were developed. The panel emphasized the importance of monitoring PsA therapies and arrived at a consensus on monitoring requirements for PsA therapies. The expert panel proposed recommendations for the management of common comorbidities associated with PsA. CONCLUSION These consensus recommendations can guide physicians and healthcare professionals in the UAE in making proper treatment decisions, as well as efficiently managing comorbidities and monitoring therapies in patients with PsA.
Collapse
Affiliation(s)
- Khalid A. Alnaqbi
- Department of RheumatologyTawam HospitalAl AinUAE
- College of Medicine and Health SciencesUAE UniversityAl AinUAE
| | - Suad Hannawi
- Emirates Health Services (EHS)DubaiUAE
- Ministry of Health and PreventionDubaiUAE
| | - Rajaie Namas
- Division of Rheumatology, Department of Internal MedicineCleveland Clinic Abu DhabiUAE
| | | | | | | |
Collapse
|
12
|
Faria RJ, Cordeiro FJR, dos Santos JBR, Alvares-Teodoro J, Guerra Júnior AA, Acurcio FDA, da Silva MRR. Conventional Synthetic Disease-Modifying Anti-rheumatic Drugs for Psoriatic Arthritis: Findings and Implications From a Patient Centered Longitudinal Study in Brazil. Front Pharmacol 2022; 13:878972. [PMID: 35559237 PMCID: PMC9086188 DOI: 10.3389/fphar.2022.878972] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Accepted: 03/24/2022] [Indexed: 12/04/2022] Open
Abstract
Background: Conventional synthetic disease-modifying antirheumatic drugs are the first-line treatment to inhibit the progression of psoriatic arthritis. Despite their widespread clinical use, few studies have been conducted to compare these drugs for psoriatic arthritis. Methods: a longitudinal study was carried out based on a centered patient national database in Brazil. Market share of drugs, medication persistence, drug costs, and cost per response were evaluated. Results: a total of 1,999 individuals with psoriatic arthritis were included. Methotrexate was the most used drug (44.4%), followed by leflunomide (40.6%), ciclosporin (8.2%), and sulfasalazine (6.8%). Methotrexate and leflunomide had a greater market share than ciclosporin and sulfasalazine over years. Medication persistence was higher for leflunomide (58.9 and 28.2%), followed by methotrexate (51.6 and 25.4%) at six and 12 months, respectively. Leflunomide was deemed the most expensive drug, with an average annual cost of $317.25, followed by sulfasalazine ($106.47), ciclosporin ($97.64), and methotrexate ($40.23). Methotrexate was the drug being the lowest cost per response. Conclusion: Methotrexate had the best cost per response ratio, owing to its lower cost and a slightly lower proportion of persistent patients when compared to leflunomide. Leflunomide had a slightly higher medication persistence than methotrexate, but it was the most expensive drug.
Collapse
Affiliation(s)
- Ronaldo José Faria
- Pharmaceutical Services Graduate Program, Federal University of Espírito Santo, Alegre, Brazil
- Health Assessment, Technology, and Economy Group, Federal University of Espírito Santo, Alegre, Brazil
| | | | - Jéssica Barreto Ribeiro dos Santos
- Pharmaceutical Services Graduate Program, Federal University of Espírito Santo, Alegre, Brazil
- Health Assessment, Technology, and Economy Group, Federal University of Espírito Santo, Alegre, Brazil
| | | | | | | | - Michael Ruberson Ribeiro da Silva
- Pharmaceutical Services Graduate Program, Federal University of Espírito Santo, Alegre, Brazil
- Health Assessment, Technology, and Economy Group, Federal University of Espírito Santo, Alegre, Brazil
| |
Collapse
|
13
|
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
|
14
|
Al Rayes H, Alazmi M, Attar S, Alderaan K, Alghamdi M, Alghanim N, Alhazmi A, Alkhadhrawi N, Almohideb M, Alzahrani Z, Bedaiwi M, Halabi H. Consensus-based recommendations on the diagnosis, referral and clinical management of patients with psoriatic arthritis. Rheumatol Int 2021; 42:391-401. [PMID: 34716785 DOI: 10.1007/s00296-021-05029-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Accepted: 10/08/2021] [Indexed: 12/15/2022]
Abstract
Psoriatic arthritis (PsA) is a highly heterogeneous disease with complex manifestations. Limited understanding of the disease and non-availability of local guidelines pose challenges in the management of PsA in Saudi Arabia. Therefore, this expert consensus is aimed to provide recommendations on the management of patients with PsA, including referral pathway, definition of remission and treat-to-target (T2T) approach. A Delphi technique of consensus development was used involving an expert panel comprised of 10 rheumatologists, one dermatologist and one family physician. Based on the review of available published evidence and the opinions of clinical experts, key recommendations were developed. A consensus was achieved in defining the following: management guideline adaptable for Saudi Arabia, most useful screening tool, laboratory investigations, imaging tests and criteria for referring suspected PsA patients to a rheumatologist. In addition, an agreement was achieved in defining the T2T strategy and remission for the clinical management of PsA. Overall, these recommendations provide an evidence-based framework for the management of PsA patients in Saudi Arabia.
Collapse
Affiliation(s)
- Hanan Al Rayes
- Department of Medicine, Prince Sultan Military Medical City, Riyadh, 21577, Saudi Arabia.
| | - Mansour Alazmi
- Department of Rheumatology, Prince Mohammed Medical City, Al Jouf, Saudi Arabia
| | - Suzan Attar
- Department of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Khaled Alderaan
- Department of Rheumatology, King Fahad Specialist Hospital, Dammam, Saudi Arabia
| | - Mushabab Alghamdi
- Department of Rheumatology, University of Bisha, Bisha, Saudi Arabia
| | - Nayef Alghanim
- Department of Rheumatology, King Saud Medical City, Riyadh, Saudi Arabia
| | - Ahmed Alhazmi
- Department of Medicine, King Fahad Armed Forces Hospital, Jeddah, Saudi Arabia
| | - Nadeer Alkhadhrawi
- Department of Family Medicine, Prince Sultan Military Medical City, Riyadh, Saudi Arabia
| | - Mohammad Almohideb
- Department of Dermatology, King Saud Bin Abdulaziz University for Health Sciences and King Abdulaziz Medical City, Riyadh, Saudi Arabia
| | - Zeyad Alzahrani
- Department of Medicine, King Abdulaziz Medical City, Jeddah, Saudi Arabia
| | - Mohamed Bedaiwi
- Department of Medicine, King Saud University Medical City, Riyadh, Saudi Arabia
| | - Hussein Halabi
- Department of Medicine, King Faisal Specialist Hospital and Research Center, Jeddah, Saudi Arabia
| |
Collapse
|
15
|
Gottlieb AB, Mease PJ, Kirkham B, Nash P, Balsa AC, Combe B, Rech J, Zhu X, James D, Martin R, Ligozio G, Abrams K, Pricop L. Secukinumab Efficacy in Psoriatic Arthritis: Machine Learning and Meta-analysis of Four Phase 3 Trials. J Clin Rheumatol 2021; 27:239-247. [PMID: 32015257 PMCID: PMC8389345 DOI: 10.1097/rhu.0000000000001302] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND Using a machine learning approach, the study investigated if specific baseline characteristics could predict which psoriatic arthritis (PsA) patients may gain additional benefit from a starting dose of secukinumab 300 mg over 150 mg. We also report results from individual patient efficacy meta-analysis (IPEM) in 2049 PsA patients from the FUTURE 2 to 5 studies to evaluate the efficacy of secukinumab 300 mg, 150 mg with and without loading regimen versus placebo at week 16 on achievement of several clinically relevant difficult-to-achieve (higher hurdle) endpoints. METHODS Machine learning employed Bayesian elastic net to analyze baseline data of 2148 PsA patients investigating 275 predictors. For IPEM, results were presented as difference in response rates versus placebo at week 16. RESULTS Machine learning showed secukinumab 300 mg has additional benefits in patients who are anti-tumor necrosis factor-naive, treated with 1 prior anti-tumor necrosis factor agent, not receiving methotrexate, with enthesitis at baseline, and with shorter PsA disease duration. For IPEM, at week 16, all secukinumab doses had greater treatment effect (%) versus placebo for higher hurdle endpoints in the overall population and in all subgroups; 300-mg dose had greater treatment effect than 150 mg for all endpoints in overall population and most subgroups. CONCLUSIONS Machine learning identified predictors for additional benefit of secukinumab 300 mg compared with 150 mg dose. Individual patient efficacy meta-analysis showed that secukinumab 300 mg provided greater improvements compared with 150 mg in higher hurdle efficacy endpoints in patients with active PsA in the overall population and most subgroups with various levels of baseline disease activity and psoriasis.
Collapse
Affiliation(s)
- Alice B. Gottlieb
- From the Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Philip J. Mease
- Swedish Medical Centre and University of Washington, Seattle, WA
| | - Bruce Kirkham
- Guy's & St Thomas' NHS Foundation Trust, London, United Kingdom
| | - Peter Nash
- Department of Medicine, Griffith University, Brisbane, Australia
| | - Alejandro C. Balsa
- Hospital Universitario La Paz, Instituto de Investigación Sanitaria (IdiPAZ), Universidad Autonoma Madrid, Madrid, Spain
| | - Bernard Combe
- CHU Montpellier, Montpellier University, Montpellier, France
| | - Jürgen Rech
- Friedrich-Alexander-University Erlangen-Nürnberg (FAU), Department of Internal Medicine 3, Rheumatology and Immunology, Universitätsklinikum Erlangen, Erlangen, Germany
| | - Xuan Zhu
- Novartis Pharmaceuticals Corporation, East Hanover, NJ
| | - David James
- Novartis Pharmaceuticals Corporation, East Hanover, NJ
| | - Ruvie Martin
- Novartis Pharmaceuticals Corporation, East Hanover, NJ
| | | | - Ken Abrams
- Novartis Pharmaceuticals Corporation, East Hanover, NJ
| | | |
Collapse
|
16
|
Ainatzoglou A, Stamoula E, Dardalas I, Siafis S, Papazisis G. The Effects of PDE Inhibitors on Multiple Sclerosis: a Review of in vitro and in vivo Models. Curr Pharm Des 2021; 27:2387-2397. [PMID: 33655851 DOI: 10.2174/1381612827666210303142356] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Accepted: 02/11/2021] [Indexed: 11/22/2022]
Abstract
BACKGROUND Multiple sclerosis (MS) is a chronic inflammatory and immune-mediated disease, whose current therapeutic means are mostly effective in the relapsing-remitting form of MS, where inflammation is still prominent, but fall short of preventing long term impairment. However, apart from inflammationmediated demyelination, autoimmune mechanisms play a major role in MS pathophysiology, constituting a promising pharmacological target. Phosphodiesterase (PDE) inhibitors have been approved for clinical use in psoriasis and have undergone trials suggesting their neuroprotective effects, rendering them eligible as an option for accessory MS therapy. OBJECTIVE In this review, we discuss the potential role of PDE inhibitors as a complementary MS therapy. METHODS We conducted a literature search through which we screened and comparatively assessed papers on the effects of PDE inhibitor use, both in vitro and in animal models of MS, taking into account a number of inclusion and exclusion criteria. RESULTS In vitro studies indicated that PDE inhibitors promote remyelination and axonal sustenance, while curbing inflammatory cell infiltration, hindering oligodendrocyte and neuronal loss and suppressing cytokine production. In vivo studies underlined that these agents alleviate symptoms and reduce disease scores in MS animal models. CONCLUSION PDE inhibitors proved to be effective in addressing various aspects of MS pathogenesis both in vitro and in vivo models. Given the latest clinical trials proving that the PDE4 inhibitor Ibudilast exerts neuroprotective effects in patients with progressive MS, research on this field should be intensified and selective PDE4 inhibitors with enhanced safety features should be seriously considered as prospective complementary MS therapy.
Collapse
Affiliation(s)
- Alexandra Ainatzoglou
- Department of Clinical Pharmacology, School of Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Eleni Stamoula
- Department of Clinical Pharmacology, School of Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Ioannis Dardalas
- Department of Clinical Pharmacology, School of Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Spyridon Siafis
- Department of Clinical Pharmacology, School of Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Georgios Papazisis
- Department of Clinical Pharmacology, School of Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece
| |
Collapse
|
17
|
Lamb YN. Guselkumab in psoriatic arthritis: a profile of its use. DRUGS & THERAPY PERSPECTIVES 2021. [DOI: 10.1007/s40267-021-00840-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
18
|
Gottlieb AB, Merola JF, Reich K, Behrens F, Nash P, Griffiths CEM, Bao W, Pellet P, Pricop L, McInnes IB. Efficacy of secukinumab and adalimumab in patients with psoriatic arthritis and concomitant moderate-to-severe plaque psoriasis: results from EXCEED, a randomized, double-blind head-to-head monotherapy study. Br J Dermatol 2021; 185:1124-1134. [PMID: 33913511 PMCID: PMC9291158 DOI: 10.1111/bjd.20413] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/22/2021] [Indexed: 11/28/2022]
Abstract
Background Secukinumab [an interleukin (IL)‐17A inhibitor] has demonstrated significantly higher efficacy vs. etanercept (a tumour necrosis factor inhibitor) and ustekinumab (an IL‐12/23 inhibitor) in patients with moderate‐to‐severe plaque psoriasis. Objectives To report 52‐week results from a prespecified analysis of patients with active psoriatic arthritis (PsA) having concomitant moderate‐to‐severe plaque psoriasis from the head‐to‐head EXCEED monotherapy study comparing secukinumab with adalimumab. Methods Patients were randomized to receive secukinumab 300 mg via subcutaneous injection at baseline, week 1–4, and then every 4 weeks until week 48 or adalimumab 40 mg via subcutaneous injection every 2 weeks from baseline until week 50. Assessments in patients with concomitant moderate‐to‐severe psoriasis, defined as having affected body surface area > 10% or Psoriasis Area and Severity Index (PASI) ≥ 10 at baseline, included musculoskeletal, skin and quality‐of‐life outcomes. Missing data were handled using multiple imputation. Results Of the 853 patients [secukinumab (N = 426), adalimumab (N = 427)], 211 (24·7%) had concomitant moderate‐to‐severe psoriasis [secukinumab (N = 110, 25·8%), adalimumab (N = 101, 23·7%)]. Up to week 50, 5·5% of patients discontinued secukinumab vs.17·8% in the adalimumab group. The proportion of patients who achieved American College of Rheumatology (ACR) 20 response was 76·4% with secukinumab vs. 68·3% with adalimumab (P = 0·175), PASI 100 response was 39·1% vs. 23·8% (P = 0·013), and simultaneous improvement in ACR 50 and PASI 100 response at week 52 was 28·2% vs. 17·7%, respectively (P = 0·06). Secukinumab demonstrated consistently higher responses vs. adalimumab across skin endpoints. Conclusions This prespecified analysis in PsA patients with concomitant moderate‐to‐severe plaque psoriasis in the EXCEED study provides further evidence that IL‐17 inhibitors offer a comprehensive biological treatment to manage the concomitant features of psoriasis and PsA.
What is already known about this topic?
Secukinumab, an interleukin‐17A inhibitor, has previously been reported to have significantly higher efficacy in head‐to‐head trials vs. etanercept and ustekinumab in patients with moderate‐to‐severe plaque psoriasis.
What does this study add?The results of the study provide valuable head‐to‐head data on the efficacy of two biologics with different mechanisms of action (secukinumab and adalimumab) as first‐line biological monotherapy for patients with psoriatic arthritis and concomitant moderate‐to‐severe plaque psoriasis. The findings of this study can further help physicians to make informed and evidence‐based decisions for the treatment of patients with active psoriatic arthritis who have concomitant moderate‐to‐severe plaque psoriasis.
Linked Comment: E. Sbidian and L. Pina‐Vegas. Br J Dermatol 2021; 185:1085.
Collapse
Affiliation(s)
- A B Gottlieb
- Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - J F Merola
- Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - K Reich
- Translational Research in Inflammatory Skin Diseases, Institute for Health Services Research in Dermatology and Nursing, University Medical Center Hamburg- Eppendorf, Hamburg, Germany
| | - F Behrens
- Rheumatology University Hospital and Fraunhofer Institute for Molecular Biology and Applied Ecology IME, Branch for Translational Medicine and Pharmacology TMP and Fraunhofer Cluster of Excellence for Immune-Mediated Diseases CIMD, Goethe University, Frankfurt, Germany
| | - P Nash
- Department of Medicine, Griffith University, Brisbane, QLD, Australia
| | - C E M Griffiths
- The Dermatology Centre, Salford Royal NHS Foundation Trust, NIHR Manchester Biomedical Research Centre, University of Manchester, Manchester, UK
| | - W Bao
- Novartis Pharmaceuticals Corporation, East Hanover, NJ, USA
| | - P Pellet
- Novartis Pharma AG, Basel, Switzerland
| | - L Pricop
- Novartis Pharmaceuticals Corporation, East Hanover, NJ, USA
| | | |
Collapse
|
19
|
Thanigaimani S, Phie J, Krishna SM, Moxon J, Golledge J. Effect of disease modifying anti-rheumatic drugs on major cardiovascular events: a meta-analysis of randomized controlled trials. Sci Rep 2021; 11:6627. [PMID: 33758292 PMCID: PMC7987985 DOI: 10.1038/s41598-021-86128-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Accepted: 03/05/2021] [Indexed: 02/08/2023] Open
Abstract
Disease modifying anti-rheumatic drugs (DMARDs) were developed to treat joint inflammation. There is growing evidence that anti-inflammatory drugs prevent major cardiovascular events (MACE). The aim of this systematic review and meta-analysis was to examine whether DMARDs reduce the risk of MACE. A systematic literature search was performed to identify randomized controlled trials (RCTs) testing the effect of DMARDs on cardiovascular events. The primary outcome was MACE defined as the first occurrence of non-fatal myocardial infarction (MI), non-fatal stroke or cardiovascular death. Secondary outcomes were myocardial infarction or stroke alone and all-cause mortality. Safety was assessed by fatal or life threatening infection. Meta-analyses were performed using random effect models and reported as risk ratios (RR) and 95% confidence intervals (CI). Study quality and publication bias were assessed using the Cochrane Collaboration’s tool for assessing risk of bias and funnel plots. Twelve RCTs involving 18,056 participants testing three different DMARDs subclasses (Tumor Necrosis Factor inhibitors—4 trials; Janus Kinase inhibitors—5 trials; Interleukin inhibitors—3 trials) were included. Meta-analysis suggested that none of the DMARD subclasses had any effect on MACE, MI alone, stroke alone, risk of fatal or life threatening infection or death. Risk of bias was high, low and unclear in five, six and one studies respectively. Funnel plots suggested a low possibility of publication bias. This meta-analysis suggests that DMARDs do not affect the incidence of MACE. More trials are needed for firm conclusions.
Collapse
Affiliation(s)
- Shivshankar Thanigaimani
- The Queensland Research Centre for Peripheral Vascular Disease (QRC-PVD), College of Medicine and Dentistry, James Cook University, Townsville, QLD, 4811, Australia.,The Australian Institute of Tropical Health and Medicine, James Cook University, Townsville, QLD, Australia
| | - James Phie
- The Queensland Research Centre for Peripheral Vascular Disease (QRC-PVD), College of Medicine and Dentistry, James Cook University, Townsville, QLD, 4811, Australia.,The Australian Institute of Tropical Health and Medicine, James Cook University, Townsville, QLD, Australia
| | - Smriti Murali Krishna
- The Queensland Research Centre for Peripheral Vascular Disease (QRC-PVD), College of Medicine and Dentistry, James Cook University, Townsville, QLD, 4811, Australia.,The Australian Institute of Tropical Health and Medicine, James Cook University, Townsville, QLD, Australia
| | - Joseph Moxon
- The Queensland Research Centre for Peripheral Vascular Disease (QRC-PVD), College of Medicine and Dentistry, James Cook University, Townsville, QLD, 4811, Australia.,The Australian Institute of Tropical Health and Medicine, James Cook University, Townsville, QLD, Australia
| | - Jonathan Golledge
- The Queensland Research Centre for Peripheral Vascular Disease (QRC-PVD), College of Medicine and Dentistry, James Cook University, Townsville, QLD, 4811, Australia. .,The Australian Institute of Tropical Health and Medicine, James Cook University, Townsville, QLD, Australia. .,The Department of Vascular and Endovascular Surgery, Townsville University Hospital, Townsville, QLD, Australia.
| |
Collapse
|
20
|
Iragorri N, Hazlewood G, Manns B, Bojke L, Spackman E. Model to Determine the Cost-Effectiveness of Screening Psoriasis Patients for Psoriatic Arthritis. Arthritis Care Res (Hoboken) 2021; 73:266-274. [PMID: 31733035 DOI: 10.1002/acr.24110] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2019] [Accepted: 11/12/2019] [Indexed: 12/20/2022]
Abstract
OBJECTIVE Screening psoriasis patients for psoriatic arthritis (PsA) is intended to identify patients at earlier stages of the disease. Early treatment is expected to slow disease progression and delay the need for biologic therapy. Our objective was to determine the cost-effectiveness of screening for PsA in patients with psoriasis in Canada. METHODS A Markov model was built to estimate the costs and quality-adjusted life years (QALYs) of screening tools for PsA in psoriasis patients. The screening tools included the Toronto Psoriatic Arthritis Screen, Psoriasis Epidemiology Screening Tool, Psoriatic Arthritis Screening and Evaluation, and Early Psoriatic Arthritis Screening Questionnaire (EARP) questionnaires. States of health were defined by disability levels as measured by the Health Assessment Questionnaire. State transitions were modeled based on annual disease progression. Incremental cost-effectiveness ratios and incremental net monetary benefits were estimated. Sensitivity analyses were undertaken to account for parameter uncertainty and to test model assumptions. RESULTS Screening was cost-effective compared to no screening. The EARP tool had the lowest total cost ($2,000 per patient per year saved compared to no screening) and the highest total QALYs (additional 0.18 per patient compared to no screening). The results were most sensitive to test accuracy and the efficacy of disease-modifying antirheumatic drugs (DMARDs). No screening was cost-effective (at $50,000 per QALY) relative to screening when DMARDs failed to slow disease progression. CONCLUSION If early therapy with DMARDs delays biologic treatment, implementing screening in patients with psoriasis in Canada is expected to represent a cost savings of $220 million per year and improve the quality of life.
Collapse
Affiliation(s)
- Nicolas Iragorri
- Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Glen Hazlewood
- Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Braden Manns
- Cumming School of Medicine, University of Calgary, Calgary, and Alberta Health Services, Edmonton, Alberta, Canada
| | | | - Eldon Spackman
- Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | | |
Collapse
|
21
|
Giles JT, Ogdie A, Gomez Reino JJ, Helliwell P, Germino R, Stockert L, Young P, Joseph W, Mundayat R, Graham D, Ritchlin C. Impact of baseline body mass index on the efficacy and safety of tofacitinib in patients with psoriatic arthritis. RMD Open 2021; 7:e001486. [PMID: 33452181 PMCID: PMC7813423 DOI: 10.1136/rmdopen-2020-001486] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Revised: 12/21/2020] [Accepted: 12/30/2020] [Indexed: 12/21/2022] Open
Abstract
OBJECTIVES This post-hoc analysis explored the impact of body mass index (BMI) on tofacitinib efficacy/safety in patients with active psoriatic arthritis (PsA). METHODS Data were pooled from two phase 3 studies (NCT01877668; NCT01882439). Analyses included patients randomised to tofacitinib 5/10 mg twio times a day or placebo, stratified by baseline BMI: <25 kg/m2, ≥25-<30 kg/m2, ≥30-<35 kg/m2 or ≥35 kg/m2. Endpoints (month 3): American College of Rheumatology (ACR20/50/70), Health Assessment Questionnaire-Disability Index (HAQ-DI) and Psoriasis Area and Severity Index (PASI) 75 response rates; dactylitis/enthesitis resolution rates; changes from baseline Short Form-36 Health Survey version 2 (SF-36v2) Physical/Mental Component Summary (PCS) scores and Functional Assessment of Chronic Illness Therapy-Fatigue (FACIT-F) total score. Safety was also reported. RESULTS Analysis included 710 patients; 43.8% were obese (BMI ≥30 kg/m2). Tofacitinib demonstrated higher efficacy response rates at month 3, compared with placebo, regardless of baseline BMI. Generally, ACR20/50/70 and HAQ-DI response rates, enthesitis resolution rates and changes from baseline in SF-36v2 PCS score and FACIT-F total score (month 3) were reduced in patients with baseline BMI ≥35 kg/m2 versus patients with lower BMIs. Elevated alanine aminotransferase/aspartate aminotransferase levels were reported in patients with baseline BMI ≥35 kg/m2 receiving tofacitinib 5 mg but not 10 mg two times a day. CONCLUSION Tofacitinib demonstrated greater efficacy than placebo in patients with PsA, regardless of baseline BMI. For all treatment arms, reduced efficacy was observed in patients with baseline BMI ≥35 kg/m2. Safety was generally comparable across BMI categories, although the effect of tofacitinib on liver enzymes in patients with baseline BMI ≥35 kg/m2 was inconclusive.
Collapse
Affiliation(s)
- Jon T Giles
- Department of Medicine/Division of Rheumatology, Columbia University Vagelos College of Physicians and Surgeons, New York, New York, USA
| | - Alexis Ogdie
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Juan J Gomez Reino
- Fundación del Instituto de Investigación Sanitaria, Hospital Clínico Universitario, Santiago de Compostela, Spain
| | - Philip Helliwell
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Leeds, UK
| | | | | | | | | | | | | | - Christopher Ritchlin
- Department of Medicine/Division of Rheumatology, University of Rochester Medical Center, Rochester, New York, USA
| |
Collapse
|
22
|
Ighani A, Georgakopoulos JR, Yeung J. Tofacitinib for the treatment of psoriasis and psoriatic arthritis. GIORN ITAL DERMAT V 2020; 155:400-410. [PMID: 32348084 DOI: 10.23736/s0392-0488.20.06643-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
INTRODUCTION Tofacitinib is a novel oral janus kinase (JAK) inhibitor approved for the treatment of psoriatic arthritis. It was also investigated for the treatment of plaque psoriasis, although it is not approved by the Food and Drug Administration for this indication. This article aimed to summarize the efficacy and safety data of tofacitinib for treatment of psoriatic disease. EVIDENCE ACQUISITION A comprehensive review of literature was conducted on the PubMed database using the search terms: "((tofacitinib) AND (psoriasis)) OR ((tofacitinib) AND (psoriatic arthritis))". Data from the pivotal clinical trials evaluating tofacitinib in the treatment of psoriatic disease were summarized. EVIDENCE SYNTHESIS The Oral-treatment Psoriasis Trial (OPT) study series demonstrated that tofacitinib is efficacious in the treatment of plaque psoriasis with an acceptable safety profile. The OPT studies also demonstrated the non-inferiority of tofacitinib 10 mg twice daily compared to etanercept. The Oral Psoriatic Arthritis Trial (OPAL) study series demonstrated that tofacitinib significantly improved psoriatic arthritis outcomes compared to placebo and had an acceptable safety profile. Its efficacy is comparable to adalimumab in psoriatic arthritis. CONCLUSIONS Patients treated with tofacitinib should be monitored for herpes zoster, malignancies, blood clots, and changes in laboratory values. Overall, tofacitinib is a useful new systemic agent in the treatment of psoriatic disease. Its oral route of administration, novel JAK pathway target, short half-life, and strong recapture rates after treatment interruption make it a unique new tool for psoriatic disease management. Further long-term studies will help determine its role in the treatment algorithm for psoriatic arthritis and its indication for plaque psoriasis.
Collapse
Affiliation(s)
- Arvin Ighani
- Division of Dermatology, Department of Medicine, University of Toronto, Toronto, ON, Canada
| | - Jorge R Georgakopoulos
- Division of Dermatology, Department of Medicine, University of Toronto, Toronto, ON, Canada
| | - Jensen Yeung
- Division of Dermatology, Department of Medicine, University of Toronto, Toronto, ON, Canada - .,Division of Dermatology, Department of Medicine, Women's College Hospital, Toronto, ON, Canada.,Sunnybrook Health Sciences Centre, Toronto, ON, Canada.,Probity Medical Research Inc., Waterloo, ON, Canada
| |
Collapse
|
23
|
Abstract
PURPOSE OF THE REVIEW To provide a general overview and current challenges regarding the genetics of psoriatic disease. With the use of integrative medicine, multiple candidate loci identified to date in psoriatic disease will be annotated, summarized, and visualized. Recent studies reporting differences in genetic architecture between psoriatic arthritis and cutaneous-only psoriasis will be highlighted. RECENT FINDINGS Focusing on functional pathways that connect previously identified genetic variants can increase our understanding of psoriatic diseases. The genetic architecture differs between psoriatic arthritis and cutaneous-only psoriasis with arthritis-specific signals in linkage disequilibrium independent of the published psoriasis signals. Integrative medicine is helpful in understanding cellular mechanisms of psoriatic diseases. Careful selection of the psoriatic disease cohort has translated into mechanistic differences among psoriatic arthritis and cutaneous psoriasis.
Collapse
Affiliation(s)
- Sara Rahmati
- Department of Medicine, Faculty of Medicine, Memorial University, St. John's, Newfoundland and Labrador, A1B 3X9, Canada
- Krembil Research Institute, University Health Network, Toronto, Ontario, M5S 1A8, Canada
| | - Lam Tsoi
- Department of Computational Medicine & Bioinformatics, Department of Biostatistics, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Darren O'Rielly
- Department of Medicine, Faculty of Medicine, Memorial University, St. John's, Newfoundland and Labrador, A1B 3X9, Canada
| | - Vinod Chandran
- Department of Medicine, Faculty of Medicine, Memorial University, St. John's, Newfoundland and Labrador, A1B 3X9, Canada
- Krembil Research Institute, University Health Network, Toronto, Ontario, M5S 1A8, Canada
- Institute of Medical Science, University of Toronto, Toronto, Ontario, M5S 1A8, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, M5S 1A8, Canada
- Department of Medicine, Division of Rheumatology, University of Toronto, Toronto, Ontario, M5S 1A8, Canada
| | - Proton Rahman
- Department of Medicine, Faculty of Medicine, Memorial University, St. John's, Newfoundland and Labrador, A1B 3X9, Canada.
| |
Collapse
|
24
|
Pouw J, Leijten E, Radstake T, Boes M. Emerging molecular biomarkers for predicting therapy response in psoriatic arthritis: A review of literature. Clin Immunol 2020; 211:108318. [DOI: 10.1016/j.clim.2019.108318] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Revised: 11/25/2019] [Accepted: 11/25/2019] [Indexed: 02/07/2023]
|
25
|
Cañete JD, Nolla JM, Queiro R, Rodríguez MJ, Ruiz M, Lizán L. Expert Consensus on a Set of Outcomes to Assess the Effectiveness of Biologic Treatment in Psoriatic Arthritis: The MERECES Study. J Rheumatol 2020; 47:1637-1643. [DOI: 10.3899/jrheum.191056] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/16/2020] [Indexed: 02/06/2023]
Abstract
Objective.To reach a consensus on the instruments to be used in clinical practice to evaluate the effectiveness of biological disease-modifying antirheumatic drug (bDMARD) treatment in patients with psoriatic arthritis (PsA) in the short to medium term (3–6 mos), and to establish the minimum health outcomes for treatment continuation.Methods.A 2-round Delphi questionnaire was developed based on both the information gathered in the literature review and 4 discussion groups. The suitability and feasibility of the proposed sets of instruments were assessed on a 7-point Likert scale. Consensus was established when at least 75% of healthcare professionals (HCP) reached agreement. To define a minimum health outcome to continue treatment, a combination of 4 disease activity states and 3 health-related quality of life states were defined for 3 hypothetical patient profiles. HCP were given a dichotomous choice (yes/no) to respond to whether they would continue treatment in each case.Results.The second round was completed by 106 HCP. Consensus was reached on the use of (1) Disease Activity in Psoriatic Arthritis + Psoriatic Arthritis Impact of Disease (PsAID12) or minimal disease activity + PsAID12 + C-reactive protein, in peripheral PsA; and (2) Ankylosing Spondylitis Disease Activity Score + PsAID12, in axial PsA. Health outcomes considered sufficient to continue treatment were stricter for bDMARD-naive patients than for patients who failed several bDMARD.Conclusion.To the best of our knowledge, this is the first multidisciplinary consensus on a set of outcomes for the evaluation of bDMARD effectiveness in PsA, in routine clinical practice.
Collapse
|
26
|
Kivitz AJ, Nash P, Tahir H, Everding A, Mann H, Kaszuba A, Pellet P, Widmer A, Pricop L, Abrams K. Efficacy and Safety of Subcutaneous Secukinumab 150 mg with or Without Loading Regimen in Psoriatic Arthritis: Results from the FUTURE 4 Study. Rheumatol Ther 2019; 6:393-407. [PMID: 31228101 PMCID: PMC6702584 DOI: 10.1007/s40744-019-0163-5] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Indexed: 12/17/2022] Open
Abstract
INTRODUCTION To assess the efficacy and safety of the subcutaneous (s.c.) secukinumab 150 mg with loading (150 mg) or without loading (150 mg no-load) regimen through 104 weeks in patients with active psoriatic arthritis (PsA) in the FUTURE 4 (NCT02294227) study. METHODS Patients with PsA (N = 341) were randomized to s.c. secukinumab 150 mg, 150 mg no-load or placebo at baseline, weeks 1, 2, 3 and every 4 weeks thereafter. All placebo patients were reassigned to secukinumab 150 mg no-load at either week 16 (non-responders) or week 24 (responders). The primary end point was ACR20 at week 16. Patients could have their dose escalated from 150 to 300 mg based on their physician's decision starting at week 36. Pre- and post-escalation ACR and PASI responses were also assessed. RESULTS A total of 95.6% (326/341), 84.5% (288/341) and 79.8% (272/341) patients completed 16, 52 and 104 weeks of treatment, respectively. The primary end point was met; ACR20 response rate at week 16 was 41.2% and 39.8% with the 150 mg and 150 mg no-load groups, respectively, versus placebo (18.4%; adjusted P value = 0.0003 for both treatment arms). Efficacy responses observed at week 16 in both treatment regimens were sustained up to week 52 and 104, with many patients continuing to show improvements up to week 104. After dose escalation to 300 mg, the proportion of patients with non-/low-level ACR/PASI response decreased with increasing proportions of patients having higher ACR/PASI responses. No new or unexpected safety signals were reported. CONCLUSION The secukinumab 150 mg or 150 mg no-load regimen demonstrated significant and sustained improvements in the signs and symptoms of psoriatic arthritis through 104 weeks; the loading regimen was associated with numerically higher and earlier responses for some high-hurdle end points. Improved efficacy was observed upon dose escalation from 150 to 300 mg. The safety profile was consistent with previous reports. TRIAL REGISTRATION ClinicalTrials.gov identifier, NCT02294227. FUNDING Novartis Pharma AG, Basel, Switzerland.
Collapse
Affiliation(s)
- Alan J Kivitz
- Altoona Centre for Clinical Research, Duncansville, PA, USA.
| | - Peter Nash
- University of Queensland, Brisbane, Australia
| | | | | | - Heřman Mann
- Institute of Rheumatology and Department of Rheumatology, Charles University, Prague, Czech Republic
| | - Andrzej Kaszuba
- Specjalistyczne Gabinety Lekarskie "DERMED" Anna Kaszuba, Lodz, Poland
| | | | | | | | - Ken Abrams
- Novartis Pharmaceutical Corporation, East Hanover, NJ, USA
| |
Collapse
|
27
|
Bedaiwi M, Al-Homood IA, El-Garf A, Uthman I, Sunna N, Nassier R, Mohamed H, Al Saleh J. Disease burden and treatment challenges of psoriatic arthritis in Africa and the Middle East. Rheumatol Int 2019; 39:1321-1329. [PMID: 31134289 DOI: 10.1007/s00296-019-04319-3] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Accepted: 05/03/2019] [Indexed: 01/17/2023]
Abstract
Psoriatic arthritis (PsA) is a chronic, inflammatory arthropathy occurring in up to 30% of patients with psoriasis, and is characterized by multiple manifestations including peripheral arthritis, enthesitis, dactylitis, spondylitis, and psoriatic skin and nail disease. This complex and heterogeneous disease is poorly understood and its diagnosis and treatment are suboptimal, particularly in Africa and the Middle East, where very few studies into the impact of PsA have been carried out. This article aims to highlight the disease burden of PsA in the region as well as to identify unmet clinical needs. A non-systematic review was carried out in the PubMed database and the most relevant publications were selected. Expert rheumatologists practicing in Africa and the Middle East provide an insight into the challenges of treating PsA in daily practice, along with recommendations for improvements.
Collapse
Affiliation(s)
- Mohamed Bedaiwi
- Division of Rheumatology, College of Medicine, King Saud University Medical City, King Saud University, Riyadh, Saudi Arabia.
| | - Ibrahim A Al-Homood
- Department of Medical Specialties, Rheumatology Section, King Fahad Medical City, Riyadh, Saudi Arabia
| | - Ayman El-Garf
- Rheumatology Department, Cairo University Hospitals, Cairo University, Cairo, Egypt
| | - Imad Uthman
- Division of Rheumatology, American University of Beirut Medical Center, Beirut, Lebanon
| | | | | | | | - Jamal Al Saleh
- Rheumatology Unit, Dubai Hospital, Dubai Health Authority, Dubai, United Arab Emirates
| |
Collapse
|
28
|
Aiello E, Bianculli PM, Bhattacharyya D, Gunda P, Citera G. Cost-Effectiveness of Secukinumab Versus Other Biologics in the Treatment of Psoriatic Arthritis: An Argentinean Perspective. Value Health Reg Issues 2019; 20:86-94. [PMID: 31103950 DOI: 10.1016/j.vhri.2019.03.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Revised: 02/20/2019] [Accepted: 03/04/2019] [Indexed: 01/20/2023]
Abstract
OBJECTIVE Psoriatic arthritis (PsA) is a chronic, systemic inflammatory disease. This study assessed the cost-effectiveness of secukinumab, an interleukin-17A inhibitor, versus other biologics in PsA from the Argentinean social security perspective. METHODS A semi-Markov model evaluated subcutaneous (sc) treatment with secukinumab 150 mg and 300 mg against other sc treatments such as adalimumab, certolizumab pegol, etanercept, golimumab, ustekinumab, and intravenous treatment infliximab in biologic-naïve (with or without moderate to severe psoriasis) and biologic-experienced PsA patients over a lifetime horizon. Response to treatments was determined using the PsA Response Criteria (PsARC) at 12 weeks. Model inputs were derived from randomized controlled trials, network meta-analyses, published literature, and other Argentinean sources. Model outcomes included quality-adjusted life years (QALYs) gained and incremental cost-effectiveness ratios. Sensitivity analyses and alternative scenarios with a higher cost option were also conducted. RESULTS Among biologic-naïve PsA patients without psoriasis, secukinumab 150 mg provided the highest QALYs (7.18) versus all sc biologics at the lowest cost ($3 755 678 Argentine peso), thus dominating them. Among biologic-naïve PsA patients with psoriasis and biologic-experienced PsA patients, secukinumab 300 mg provided highest QALYs (6.99 and 7.53, respectively), dominated infliximab, and was cost-effective versus other sc biologics. Deterministic sensitivity analyses indicated sensitivity of results to variation in PsARC rates, drug acquisition costs, Health Assessment Questionnaire change, and utilities. A probabilistic sensitivity analysis showed maximum net monetary benefits with both secukinumab doses. Results from an alternative scenario analysis were similar to base-case analysis. CONCLUSIONS For both biologic-naïve and experienced PsA patients, secukinumab is either a dominant or cost-effective treatment option compared with other biologics in Argentina.
Collapse
Affiliation(s)
| | | | | | - Praveen Gunda
- Novartis Healthcare Private Limited, Hyderabad, India
| | - Gustavo Citera
- Instituto de Rehabilitación Psicofísica, Buenos Aires, Argentina
| |
Collapse
|
29
|
Dressler C, Eisert L, Pham P, Nast A. Efficacy and safety of systemic treatments in psoriatic arthritis: a systematic review, meta‐analysis and
GRADE
evaluation. J Eur Acad Dermatol Venereol 2019; 33:1249-1260. [DOI: 10.1111/jdv.15482] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2018] [Accepted: 12/19/2018] [Indexed: 02/04/2023]
Affiliation(s)
- C. Dressler
- Berlin Institute of Health Department of Dermatology Venerology and Allergy Division of Evidence‐Based Medicine (dEBM) Charité ‐ Universitätsmedizin Berlin Corporate Member of Freie Universität Berlin Humboldt‐Universität zu Berlin Berlin Germany
| | - L. Eisert
- Berlin Institute of Health Department of Dermatology Venerology and Allergy Division of Evidence‐Based Medicine (dEBM) Charité ‐ Universitätsmedizin Berlin Corporate Member of Freie Universität Berlin Humboldt‐Universität zu Berlin Berlin Germany
| | - P.A. Pham
- Berlin Institute of Health Department of Dermatology Venerology and Allergy Division of Evidence‐Based Medicine (dEBM) Charité ‐ Universitätsmedizin Berlin Corporate Member of Freie Universität Berlin Humboldt‐Universität zu Berlin Berlin Germany
| | - A. Nast
- Berlin Institute of Health Department of Dermatology Venerology and Allergy Division of Evidence‐Based Medicine (dEBM) Charité ‐ Universitätsmedizin Berlin Corporate Member of Freie Universität Berlin Humboldt‐Universität zu Berlin Berlin Germany
| |
Collapse
|
30
|
Furst DE, Belasco J, Louie JS. Genetic and inflammatory factors associated with psoriatic arthritis: Relevance to diagnosis and management. Clin Immunol 2019; 202:59-75. [DOI: 10.1016/j.clim.2019.02.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Revised: 01/21/2019] [Accepted: 02/04/2019] [Indexed: 12/22/2022]
|
31
|
A Review for Physician Assistants and Nurse Practitioners on the Considerations for Diagnosing and Treating Psoriatic Arthritis. Rheumatol Ther 2018; 6:5-21. [PMID: 30570707 PMCID: PMC6393269 DOI: 10.1007/s40744-018-0133-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2018] [Indexed: 02/08/2023] Open
Abstract
Psoriatic arthritis (PsA) is a clinically heterogeneous form of progressive inflammatory arthritis that affects up to 30% of patients with psoriasis. The rapid rate of progression associated with PsA makes early-disease diagnosis and treatment crucial to patients' quality of life and long-term health. With the aim of providing clinical guidance to physician assistants and nurse practitioners, this article gives an overview of the different PsA clinical domains, including peripheral arthritis, axial disease, enthesitis, dactylitis, skin disease, and nail dystrophy, which should be considered as part of diagnosis and treatment strategies. The efficacy of different therapies across these PsA domains is reviewed within the context of current PsA treatment guidelines while considering more recent data on newly approved therapies for PsA.Funding: Novartis Pharmaceuticals Corp., East Hanover, NJ, USA.
Collapse
|
32
|
Purmonen T, Puolakka K, Bhattacharyya D, Jain M, Martikainen J. Cost-effectiveness analysis of secukinumab versus other biologics and apremilast in the treatment of active Psoriatic arthritis: a Finnish perspective. COST EFFECTIVENESS AND RESOURCE ALLOCATION 2018; 16:56. [PMID: 30479574 PMCID: PMC6240184 DOI: 10.1186/s12962-018-0162-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Accepted: 11/08/2018] [Indexed: 01/21/2023] Open
Abstract
OBJECTIVE To study cost-effectiveness of an interleukin (IL)-17A inhibitor secukinumab, with other biologics and apremilast in patients with Psoriatic arthritis (PsA) from payer perspective in Finland. METHODS In this semi-Markov model, subcutaneous (SC) secukinumab was compared with SC treatments etanercept and its biosimilar, certolizumab pegol, adalimumab and its biosimilar, golimumab, ustekinumab, intravenous (IV) treatment infliximab, as well as oral non-biologic apremilast. Patients without prior exposure (naïve) to biologics and without moderate to severe psoriasis were considered for secukinumab 150 mg group. Secukinumab 300 mg group included naïve patients with moderate to severe psoriasis and all patients with prior biologic exposure. The PsA Response Criteria (PsARC) at 12-week was primary criteria for treatment response. Other clinical as well as cost related model inputs were derived from relevant clinical trials as well as Finnish publications. The key model outcomes were quality-adjusted life years and incremental cost-effectiveness ratio. An annual 3% discount rate was applied to all future costs and benefits. Model input variations were assessed through sensitivity analyses and alternative scenario analyses. RESULTS For a lifetime horizon (60 years), secukinumab 150 mg dominated all branded SC biologics and apremilast with highest QALY of 8.01 and lowest lifetime cost of €187,776, while it was cost-effective against IV infliximab among biologic-naïve patients without moderate to severe psoriasis. Secukinumab 300 mg was cost-effective against all branded SC biologics and apremilast and dominated IV infliximab among biologic-naïve patients with moderate to severe psoriasis, while it was cost-effective in biologic experienced patients. With the one-way sensitivity analysis, PsARC response, drug acquisition cost, and health assessment questionnaire score were the most important parameters affecting the outcomes. Across all treatment groups, patients on secukinumab were most likely to achieve highest net monetary benefit than other competitors in probabilistic sensitivity analysis. With alternative scenario analysis, results largely remained unchanged. CONCLUSIONS Secukinumab is a cost-effective treatment for PsA patients from a Finnish payer's perspective.
Collapse
Affiliation(s)
| | - Kari Puolakka
- South Karelia Social and Health Care District, Lappeenranta, Finland
| | - Devarshi Bhattacharyya
- Novartis Product Life Cycle Services-NBS, Novartis Healthcare Private Limited, Hyderabad, India
| | - Minal Jain
- Novartis Product Life Cycle Services-NBS, Novartis Healthcare Private Limited, Hyderabad, India
| | | |
Collapse
|
33
|
Are Adverse Events Induced by the Acute Administration of Calcineurin Inhibitor Cyclosporine A Behaviorally Conditioned in Healthy Male Volunteers? Clin Ther 2018; 40:1868-1877. [PMID: 30376962 DOI: 10.1016/j.clinthera.2018.09.008] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2018] [Revised: 09/14/2018] [Accepted: 09/15/2018] [Indexed: 10/28/2022]
Abstract
PURPOSE The learned immunosuppressive placebo response has been demonstrated in experimental animals, healthy humans, and patients, and is suggested as a therapy for improving immunopharmacologic treatment. It remains unclear, however, whether potential adverse events induced by the drug are also behaviorally conditioned. Employing an established taste-immune learning paradigm in healthy humans using the calcineurin inhibitor and immunosuppressive drug cyclosporine A (CsA) as an unconditioned stimulus, we investigated whether and to what extent perceived adverse events induced by acute CsA administration are behaviorally conditioned. METHODS A total of 68 healthy male subjects were exposed to the established taste-immune learning paradigm, receiving either placebo or CsA (10 mg/kg) as an unconditioned stimulus, and a novel-tasting drink as a conditioned stimulus. FINDINGS Subjects repeatedly receiving CsA during acquisition reported significantly more adverse events than did placebo-receiving subjects. However, during reexposure to the conditioned stimulus, the reported adverse events did not differ from those in the placebo control condition. IMPLICATIONS These data indicate that acute adverse events are not behaviorally conditioned during the learned immunosuppressive response. Our results further strengthen the great potential clinical relevance of employing the learned immunosuppressive placebo response as a therapy to support immunopharmacologic regimens, ultimately aiming to reduce the medical dosages required, thereby minimizing adverse drug events while maximizing the therapeutic benefit in patients. German Clinical Trial Register (www.drks.de) identifier: DRKS00007693.
Collapse
|
34
|
Walsh JA, Adejoro O, Chastek B, Palmer JB, Hur P. Treatment Patterns Among Patients with Psoriatic Arthritis Treated with a Biologic in the United States: Descriptive Analyses from an Administrative Claims Database. J Manag Care Spec Pharm 2018; 24:623-631. [PMID: 29952704 PMCID: PMC10397599 DOI: 10.18553/jmcp.2018.24.7.623] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
BACKGROUND In patients with psoriatic arthritis (PsA), limited data exist regarding patterns of biologic therapy use. OBJECTIVE To examine treatment patterns and therapy modifications in U.S. patients with PsA receiving a tumor necrosis factor inhibitor (TNFi) or an anti-interleukin (IL)-12/23 inhibitor. METHODS Adults with PsA who newly initiated a biologic therapy (index biologic) between January 1, 2013, and January 31, 2015, were included from the Optum Research Database. Biologic therapies comprised those that were approved by the FDA for the treatment of PsA at the time of the study initiation (adalimumab, certolizumab pegol, etanercept, golimumab, infliximab, or ustekinumab). Outcomes included adherence, persistence, and discontinuation of the index biologic; initiation of adjunctive medications (nonbiologics, including those commonly used for pain and/or inflammation); and dose escalation of the index biologic during the 12-month follow-up period. RESULTS Of the 1,235 patients included, 52.5% were female, and mean (SD) age was 50.3 (12.1) years. The mean (SD) duration of persistence with a newly initiated index biologic (etanercept [48.1%], adalimumab [24.0%], infliximab [10.4%], golimumab [8.3%], ustekinumab [7.2%], or certolizumab pegol [2.0%]) was 246 (128) days; 44.5% of patients persisted with the index biologic for ≥ 12 months. During the 12-month follow-up period, 22.9% of patients switched to a different biologic, 26.8% discontinued without switching or restarting, and 5.8% discontinued and restarted the index biologic. Of the 1,010 patients who persisted with the index biologic for > 90 days, 45.6% received ≥ 1 adjunctive medication during the period from 90 days after the index date to the end of persistence or 12 months. The most commonly initiated adjunctive medications were corticosteroids (22.0%), opioids (17.1%), and nonsteroidal anti-inflammatory drugs (12.9%). Overall, 9.6% of patients had a dose escalation of the index biologic in the immediate 12-month post-index period. CONCLUSIONS This real-world study of treatment patterns for PsA, which used a large U.S. claims database, demonstrated that the majority of patients with PsA discontinued their index biologic (TNFi or anti-IL-12/23 inhibitor) before 12 months. Nearly half of patients initiated an adjunctive medication, many of which were pain and conventional anti-inflammatory medications. DISCLOSURES This study was sponsored by Novartis Pharmaceuticals. Optum was commissioned by Novartis to conduct this study, but employment was not contingent on results of the study. Walsh is a paid consultant for Novartis. Adejoro was an employee of Optum at the time of the study and writing of the manuscript. Chastek is an employee of Optum. Palmer and Hur are employees of Novartis. Results of this study were presented as an abstract and poster at the Academy of Managed Care Pharmacy Nexus 2017; October 16-19, 2017; Dallas, TX; and the EULAR 2017 Annual European Congress of Rheumatology; June 14-17, 2017; Madrid, Spain.
Collapse
Affiliation(s)
- Jessica A Walsh
- 1 University of Utah School of Medicine and Salt Lake City Veterans Affairs Medical Center, Salt Lake City, Utah
| | | | | | | | - Peter Hur
- 3 Novartis Pharmaceuticals, East Hanover, New Jersey
| |
Collapse
|
35
|
Merola JF, Espinoza LR, Fleischmann R. Distinguishing rheumatoid arthritis from psoriatic arthritis. RMD Open 2018; 4:e000656. [PMID: 30167326 PMCID: PMC6109814 DOI: 10.1136/rmdopen-2018-000656] [Citation(s) in RCA: 85] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Revised: 05/25/2018] [Accepted: 05/25/2018] [Indexed: 12/19/2022] Open
Abstract
Rheumatoid arthritis (RA) and psoriatic arthritis (PsA) have key differences in clinical presentation, radiographic findings, comorbidities and pathogenesis to distinguish between these common forms of chronic inflammatory arthritis. Joint involvement is typically, but not always, asymmetric in PsA, while it is predominantly symmetric in RA. Bone erosions, without new bone growth, and cervical spine involvement are distinctive of RA, while axial spine involvement, psoriasis and nail dystrophy are distinctive of PsA. Patients with PsA typically have seronegative test findings for rheumatoid factor (RF) and cyclic citrullinated peptide (CCP) antibodies, while approximately 80% of patients with RA have positive findings for RF and CCP antibodies. Although there is overlap in the pathogenesis of PsA and RA, differences are also present that affect the efficacy of treatment. In PsA, levels of interleukin (IL)-1β, IL-6, IL-17, IL-22, IL-23, interferon-γ and tumour necrosis factor-α (TNF-α) are elevated, and in RA, levels of IL-1, IL-6, IL-22, IL-33, TNF-α, chemokine ligand 11 and chemokine C-X-C motif ligand 13 are elevated. Differences in the pathogenesis of RA and PsA translate into some variances in the specificity and efficacy of therapies.
Collapse
Affiliation(s)
- Joseph F Merola
- Department of Dermatology, Medicine and Rheumatology, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Luis R Espinoza
- Section of Rheumatology, LSU Health Sciences Center at New Orleans, New Orleans, Louisiana, USA
| | - Roy Fleischmann
- Department of Medicine, University of Texas Southwestern Medical Center, Metroplex Clinical Research Center, Dallas, Texas, USA
| |
Collapse
|
36
|
Bergman M, Lundholm A. Managing morbidity and treatment-related toxicity in patients with ankylosing spondylitis. Rheumatology (Oxford) 2018; 57:419-428. [PMID: 28977661 PMCID: PMC5850804 DOI: 10.1093/rheumatology/kex292] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2016] [Indexed: 12/17/2022] Open
Abstract
AS is the prototypical member of the family of spondyloarthropathies, and is characterized by seronegativity, axial predominance and new bone formation, which underlie symptoms of inflammatory back pain, enthesopathy and extra-articular manifestations, including anterior uveitis, psoriasis and colitis. Patients with AS typically experience a wide variety of morbidities. These include both morbidities related to the disease itself—most prominently progressive, irreversible, structural damage to the axial or peripheral skeleton—and morbidities stemming from treatments for the disease, including toxicities from NSAID use, and increased risk of infections and immunogenicity concerns with biologics. AS is also associated with a number of comorbidities. We review the risks associated with AS, its comorbidities and its treatments, as well as strategies that can be used to mitigate these risks in patients with AS.
Collapse
Affiliation(s)
- Martin Bergman
- Department of Medicine, Drexel University College of Medicine, PA, USA
| | - Amy Lundholm
- Rheumatology, Lankenau Medical Center, Wynnewood, PA, USA
| |
Collapse
|
37
|
Mease PJ, Karki C, Liu M, Kavanaugh A, Ritchlin CT, Huynh DH, Palmer JB, Greenberg JD. Baseline patient characteristics associated with response to biologic therapy in patients with psoriatic arthritis enrolled in the Corrona Psoriatic Arthritis/Spondyloarthritis Registry. RMD Open 2018; 4:e000638. [PMID: 29707232 PMCID: PMC5916237 DOI: 10.1136/rmdopen-2017-000638] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Revised: 03/05/2018] [Accepted: 03/06/2018] [Indexed: 01/11/2023] Open
Abstract
Objectives To compare baseline characteristics between patients with psoriatic arthritis (PsA) who achieved and did not achieve minimal disease activity (MDA) with biologic therapy in the US-based Corrona Psoriatic Arthritis/Spondyloarthritis Registry. Methods Patients with PsA aged ≥18 years enrolled between March 2013 and March 2016 who were receiving biologics at enrolment (baseline), not in MDA and had ≥2 follow-up visits were included. Patients were classified as those who remained on their index biologic and achieved MDA at the second follow-up visit (MDA achievers (MDA-A)) and those who did not (MDA non-achievers (MDA-NA)). Demographics, clinical characteristics, patient-reported outcomes and medication history were compared between groups. Results Of 148 patients with PsA who met the inclusion criteria, 34 (23.0%) and 114 (77.0%) were classified as MDA-A and MDA-NA, respectively. At baseline, most patients (96.6%) were receiving tumour necrosis factor inhibitors, and both groups were similar in age, sex, race, medication history, enthesitis and dactylitis counts, disease duration and comorbidities. Compared with MDA-A, MDA-NA had significantly worse mean tender joint count (7.2 vs 3.4), patient-reported pain (51.2 vs 35.7), patient-reported fatigue (54.1 vs 42.4), physical function (Health Assessment Questionnaire, 1.0 vs 0.6), Bath Ankylosing Disease Activity Index (5.0 vs 3.4) and Bath Ankylosing Spondylitis Functional Index (4.0 vs 2.0) scores (all p<0.05). Conclusions Approximately one in four patients achieved MDA with their index biologic at the time of the second follow-up visit. Both groups were similar in several baseline demographic and clinical features; however, patients who did not achieve MDA generally had worse tender joint counts and patient-reported outcomes.
Collapse
Affiliation(s)
- Philip J Mease
- Rheumatology Clinical Research Division, Swedish Medical Center and University of Washington, Seattle, Washington, USA
| | | | - Mei Liu
- Corrona, Waltham, Massachusetts, USA
| | | | - Christopher T Ritchlin
- Allergy, Immunology and Rheumatology Division, University of Rochester Medical Center, Rochester, New York, USA
| | | | | | - Jeffrey D Greenberg
- Corrona, Waltham, Massachusetts, USA.,Department of Medicine, Division of Rheumatology, New York University School of Medicine, New York City, New York, USA
| |
Collapse
|
38
|
Secukinumab Versus Adalimumab for Psoriatic Arthritis: Comparative Effectiveness up to 48 Weeks Using a Matching-Adjusted Indirect Comparison. Rheumatol Ther 2018; 5:99-122. [PMID: 29605841 DOI: 10.1007/s40744-018-0106-6] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2018] [Indexed: 12/20/2022] Open
Abstract
INTRODUCTION Secukinumab and adalimumab are approved for adults with active psoriatic arthritis (PsA). In the absence of direct randomized controlled trial (RCT) data, matching-adjusted indirect comparison can estimate the comparative effectiveness in anti-tumor necrosis factor (TNF)-naïve populations. METHODS Individual patient data from the FUTURE 2 RCT (secukinumab vs. placebo; N = 299) were adjusted to match baseline characteristics of the ADEPT RCT (adalimumab vs. placebo; N = 313). Logistic regression determined adjustment weights for age, body weight, sex, race, methotrexate use, psoriasis affecting ≥ 3% of body surface area, Psoriasis Area and Severity Index score, Health Assessment Questionnaire Disability Index score, presence of dactylitis and enthesitis, and previous anti-TNF therapy. Recalculated secukinumab outcomes were compared with adalimumab outcomes at weeks 12 (placebo-adjusted), 16, 24, and 48 (nonplacebo-adjusted). RESULTS After matching, the effective sample size for FUTURE 2 was 101. Week 12 American College of Rheumatology (ACR) response rates were not significantly different between secukinumab and adalimumab. Week 16 ACR 20 and 50 response rates were higher for secukinumab 150 mg than for adalimumab (P = 0.017, P = 0.033), as was ACR 50 for secukinumab 300 mg (P = 0.030). Week 24 ACR 20 and 50 were higher for secukinumab 150 mg than for adalimumab (P = 0.001, P = 0.019), as was ACR 20 for secukinumab 300 mg (P = 0.048). Week 48 ACR 20 was higher for secukinumab 150 and 300 mg than for adalimumab (P = 0.002, P = 0.027), as was ACR 50 for secukinumab 300 mg (P = 0.032). CONCLUSIONS In our analysis, patients with PsA receiving secukinumab were more likely to achieve higher ACR responses through 1 year (weeks 16-48) than those treated with adalimumab. Although informative, these observations rely on a subgroup of patients from FUTURE 2 and thus should be considered interim until the ongoing head-to-head RCT EXCEED can validate these findings. FUNDING Novartis Pharma AG.
Collapse
|
39
|
Walsh JA, Adejoro O, Chastek B, Palmer JB, Hur P. Treatment Patterns Among Patients with Psoriatic Arthritis Treated with a Biologic in the United States: Descriptive Analyses from an Administrative Claims Database. J Manag Care Spec Pharm 2018:1-11. [PMID: 29557701 DOI: 10.18553/jmcp.2018.17388] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
BACKGROUND In patients with psoriatic arthritis (PsA), limited data exist regarding patterns of biologic therapy use. OBJECTIVE To examine treatment patterns and therapy modifications in U.S. patients with PsA receiving a tumor necrosis factor inhibitor (TNFi) or an anti-interleukin (IL)-12/23 inhibitor. METHODS Adults with PsA who newly initiated a biologic therapy (index biologic) between January 1, 2013, and January 31, 2015, were included from the Optum Research Database. Biologic therapies comprised those that were approved by the FDA for the treatment of PsA at the time of the study initiation (adalimumab, certolizumab pegol, etanercept, golimumab, infliximab, or ustekinumab). Outcomes included adherence, persistence, and discontinuation of the index biologic; initiation of adjunctive medications (nonbiologics, including those commonly used for pain and/or inflammation); and dose escalation of the index biologic during the 12-month follow-up period. RESULTS Of the 1,235 patients included, 52.5% were female, and mean (SD) age was 50.3 (12.1) years. The mean (SD) duration of persistence with a newly initiated index biologic (etanercept [48.1%], adalimumab [24.0%], infliximab [10.4%], golimumab [8.3%], ustekinumab [7.2%], or certolizumab pegol [2.0%]) was 246 (128) days; 44.5% of patients persisted with the index biologic for ≥ 12 months. During the 12-month follow-up period, 22.9% of patients switched to a different biologic, 26.8% discontinued without switching or restarting, and 5.8% discontinued and restarted the index biologic. Of the 1,010 patients who persisted with the index biologic for > 90 days, 45.6% received ≥ 1 adjunctive medication during the period from 90 days after the index date to the end of persistence or 12 months. The most commonly initiated adjunctive medications were corticosteroids (22.0%), opioids (17.1%), and nonsteroidal anti-inflammatory drugs (12.9%). Overall, 9.6% of patients had a dose escalation of the index biologic in the immediate 12-month post-index period. CONCLUSIONS This real-world study of treatment patterns for PsA, which used a large U.S. claims database, demonstrated that the majority of patients with PsA discontinued their index biologic (TNFi or anti-IL-12/23 inhibitor) before 12 months. Nearly half of patients initiated an adjunctive medication, many of which were pain and conventional anti-inflammatory medications. DISCLOSURES This study was sponsored by Novartis Pharmaceuticals. Optum was commissioned by Novartis to conduct this study, but employment was not contingent on results of the study. Walsh is a paid consultant for Novartis. Adejoro was an employee of Optum at the time of the study and writing of the manuscript. Chastek is an employee of Optum. Palmer and Hur are employees of Novartis. Study concept and design were contributed by Walsh, Chastek, Adejoro, Palmer, and Hur. Adejoro, Chastek, Walsh, Palmer, and Hur collected the data. Data interpretation was performed by Walsh, Palmer, Adejoro, Chastek, and Hur. The manuscript was written and revised by Walsh and Hur, along with the other authors. Results of this study were presented as an abstract and poster at the Academy of Managed Care Pharmacy Nexus 2017; October 16-19, 2017; Dallas, Texas; and the EULAR 2017 Annual European Congress of Rheumatology; June 14-17, 2017; Madrid, Spain.
Collapse
Affiliation(s)
- Jessica A Walsh
- 1 University of Utah School of Medicine and Salt Lake City Veterans Affairs Medical Center, Salt Lake City, Utah
| | | | | | | | - Peter Hur
- 3 Novartis Pharmaceuticals, East Hanover, New Jersey
| |
Collapse
|
40
|
Abstract
Golimumab (Simponi®), a fully human monoclonal antibody against tumour necrosis factor-alpha (TNFα), is given once monthly by subcutaneous injection. In the EU, golimumab is approved as monotherapy and/or in combination with methotrexate for the treatment of inflammatory arthritis, including rheumatoid arthritis (RA), psoriatic arthritis (PsA) and axial spondyloarthritis [comprising ankylosing spondylitis (AS) and non-radiographic axial spondyloarthritis (nr-axSpA)] in adults, and polyarticular juvenile idiopathic arthritis (pJIA) in children. These approvals were based on the observations that golimumab was generally well tolerated and conferred some or all of the following benefits in pivotal studies in these settings: reduced signs and symptoms of arthritis; improved physical functioning and health-related quality of life; and slowed radiographic progression. Of note, data from long-term extensions (LTEs) of pivotal studies in RA, PsA and AS have confirmed that the safety and efficacy of golimumab are sustained through 5 years of treatment; the long-term tolerability profile of the drug is similar to that of other TNFα inhibitors (TNFis). Like other subcutaneous TNFis, golimumab offers patients the convenience and flexibility of home-based self-injection, although it has the added potential advantage of requiring less frequent administration, in particular compared with older, first generation agents, such as etanercept and adalimumab. Thus, golimumab is an effective, generally well tolerated and potentially convenient option for the treatment of RA, PsA, AS and nr-axSpA in adults, and pJIA in children.
Collapse
Affiliation(s)
- James E Frampton
- Springer, Private Bag 65901, Mairangi Bay, Auckland, 0754, New Zealand.
| |
Collapse
|
41
|
Goeree R, Chiva-Razavi S, Gunda P, Graham CN, Miles L, Nikoglou E, Jugl SM, Gladman DD. Cost-effectiveness analysis of secukinumab for the treatment of active psoriatic arthritis: a Canadian perspective. J Med Econ 2018; 21:163-173. [PMID: 28945143 DOI: 10.1080/13696998.2017.1384737] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
OBJECTIVE The study evaluates the cost-effectiveness of secukinumab, a fully human monoclonal antibody that selectively neutralizes interleukin (IL)-17A, vs currently licensed biologic treatments in patients with active psoriatic arthritis (PsA) from a Canadian healthcare system perspective. METHODS A decision analytic semi-Markov model evaluated the cost-effectiveness of secukinumab 150 mg and 300 mg compared to subcutaneous biologics adalimumab, certolizumab pegol, etanercept, golimumab, and ustekinumab, and intravenous biologics infliximab and infliximab biosimilar in biologic-naive and biologic-experienced patients over a lifetime horizon. The response to treatments was evaluated after 12 weeks by PsA Response Criteria (PsARC) response rates. Non-responders or patients discontinuing initial-line of biologic treatment were allowed to switch to subsequent-line biologics. Model input parameters (Psoriasis Area Severity Index [PASI], Health Assessment Questionnaire [HAQ], withdrawal rates, costs, and resource use) were collected from clinical trials, published literature, and other Canadian sources. Benefits were expressed as quality-adjusted life years (QALYs). An annual discount rate of 5% was applied to costs and benefits. The robustness of the study findings were evaluated via sensitivity analyses. RESULTS Biologic-naive patients treated with secukinumab achieved the highest number of QALYs (8.54) at the lowest cost (CAD 925,387) over a lifetime horizon vs all comparators. Secukinumab dominated all treatments, except for infliximab and its biosimilar, which achieved minimally more QALYs (8.58). However, infliximab and its biosimilar incurred more costs than secukinumab (infliximab: CAD 1,015,437; infliximab biosimilar: CAD 941,004), resulting in higher cost-effectiveness estimates relative to secukinumab. In the biologic-experienced population, secukinumab dominated all treatments as it generated more QALYs (8.89) at lower costs (CAD 954,692). Deterministic sensitivity analyses indicated the results were most sensitive to variation in PsARC response rates, change in HAQ, and utility values in both populations. CONCLUSIONS Secukinumab is either dominant or cost-effective vs all licensed biologics for the treatment of active PsA in biologic-naive and biologic-experienced populations in Canada.
Collapse
MESH Headings
- Adult
- Antibodies, Monoclonal/administration & dosage
- Antibodies, Monoclonal/economics
- Antibodies, Monoclonal, Humanized/administration & dosage
- Antibodies, Monoclonal, Humanized/economics
- Arthritis, Psoriatic/diagnostic imaging
- Arthritis, Psoriatic/drug therapy
- Arthritis, Psoriatic/economics
- Biological Products/administration & dosage
- Biological Products/economics
- Biological Products/pharmacology
- Biosimilar Pharmaceuticals/administration & dosage
- Biosimilar Pharmaceuticals/economics
- Canada
- Cost-Benefit Analysis
- Decision Support Techniques
- Drug Administration Schedule
- Drug Costs
- Female
- Follow-Up Studies
- Humans
- Injections, Subcutaneous
- Male
- Markov Chains
- Quality-Adjusted Life Years
- Severity of Illness Index
- Time Factors
- Treatment Outcome
Collapse
Affiliation(s)
- Ron Goeree
- a Goeree Consulting Ltd , Hamilton , Ontario , Canada
- b McMaster University , Hamilton , Ontario , Canada
| | | | - Praveen Gunda
- d Novartis Product Life Cycle Services-NBS, Novartis Healthcare Private Limited , Hyderabad , India
| | | | - LaStella Miles
- e RTI Health Solutions , Research Triangle Park , NC , USA
| | - Efthalia Nikoglou
- f Novartis Product Lifecycle Services - NBS, Novartis Global Service Center , Dublin , Ireland
| | | | - Dafna D Gladman
- h Toronto Western Hospital, University of Toronto , Toronto , Ontario , Canada
| |
Collapse
|
42
|
Wang EA, Suzuki E, Maverakis E, Adamopoulos IE. Targeting IL-17 in psoriatic arthritis. Eur J Rheumatol 2017; 4:272-277. [PMID: 29308283 DOI: 10.5152/eurjrheum.2017.17037] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Accepted: 08/23/2017] [Indexed: 12/14/2022] Open
Abstract
Psoriatic arthritis (PsA) is a chronic and progressive inflammatory arthritis intimately associated with psoriasis, and can be an impairing disease that leads to reduced quality of life and significant morbidity. Treatment often requires TNF antagonists, yet many patients with PsA are not responsive to the standard anti-TNF therapies. The interleukin-17 (IL-17)/IL-17 receptor (IL-17R) family has recently been implicated in the pathogenesis of PsA and psoriasis. Much enthusiasm has been generated for the development of biologics that target the IL-17 signaling pathway directly or indirectly, many of which have produced striking results in the setting of psoriasis and PsA. Herein, we review the role of IL-17 and the IL-17 receptor (IL-17R) in the pathogenesis of PsA, as well as the clinical evidence for IL-17 and IL-17R targeted therapeutics.
Collapse
Affiliation(s)
| | - Erika Suzuki
- Department of Internal Medicine, Division of Rheumatology, Allergy and Clinical Immunology, University of California at Davis, CA, USA
| | | | - Iannis E Adamopoulos
- Department of Internal Medicine, Division of Rheumatology, Allergy and Clinical Immunology, University of California at Davis, CA, USA.,Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children Northern California, CA, USA
| |
Collapse
|
43
|
Harrold LR, Stolshek BS, Rebello S, Collier DH, Mutebi A, Wade SW, Malley W, Greenberg JD, Etzel CJ. Rebound in Measures of Disease Activity and Symptoms in Corrona Registry Patients with Psoriatic Arthritis Who Discontinue Tumor Necrosis Factor Inhibitor Therapy after Achieving Low Disease Activity. J Rheumatol 2017; 45:78-82. [PMID: 28966209 DOI: 10.3899/jrheum.161567] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/19/2017] [Indexed: 02/08/2023]
Abstract
OBJECTIVE Rebound may occur in patients with psoriatic arthritis (PsA) who discontinue TNF inhibitor (TNFi) therapy in low disease activity (LDA). METHODS Using physician and patient reports, we quantified rebound following TNFi discontinuation [defined as Clinical Disease Activity Index (CDAI) score > 10 or TNFi restart] and time to rebound in adults with PsA in LDA (CDAI score ≤ 10) at TNFi discontinuation. RESULTS Rebound occurred in 73% (69/94) of patients soon after discontinuation (median time to rebound 8.0 mos, 95% CI 6.0-12.0). CONCLUSION Rebound occurred frequently in patients with PsA after TNFi discontinuation. TNFi discontinuation after achieving LDA should be carefully considered.
Collapse
Affiliation(s)
- Leslie R Harrold
- From Corrona LLC, Southborough; Department of Medicine, University of Massachusetts Medical School, Worcester, Massachusetts; Amgen Inc., Thousand Oaks, California; Wade Outcomes Research and Consulting, Salt Lake City, Utah; New York University School of Medicine, New York, New York; Department of Epidemiology, University of Texas, MD Anderson Cancer Center, Houston, Texas, USA. .,L.R. Harrold, MD, MPH, Corrona LLC, and Department of Medicine, University of Massachusetts Medical School; B.S. Stolshek, PharmD, Amgen Inc.; S. Rebello, MPH, Corrona LLC; D.H. Collier, MD, Amgen Inc.; A. Mutebi, PhD, Amgen Inc.; S.W. Wade, MPH, Wade Outcomes Research and Consulting; W. Malley, MS, Corrona LLC, and New York University School of Medicine; J.D. Greenberg, MD, MPH, Corrona LLC; C.J. Etzel, PhD, Corrona LLC, and Department of Epidemiology, University of Texas, MD Anderson Cancer Center.
| | - Bradley S Stolshek
- From Corrona LLC, Southborough; Department of Medicine, University of Massachusetts Medical School, Worcester, Massachusetts; Amgen Inc., Thousand Oaks, California; Wade Outcomes Research and Consulting, Salt Lake City, Utah; New York University School of Medicine, New York, New York; Department of Epidemiology, University of Texas, MD Anderson Cancer Center, Houston, Texas, USA.,L.R. Harrold, MD, MPH, Corrona LLC, and Department of Medicine, University of Massachusetts Medical School; B.S. Stolshek, PharmD, Amgen Inc.; S. Rebello, MPH, Corrona LLC; D.H. Collier, MD, Amgen Inc.; A. Mutebi, PhD, Amgen Inc.; S.W. Wade, MPH, Wade Outcomes Research and Consulting; W. Malley, MS, Corrona LLC, and New York University School of Medicine; J.D. Greenberg, MD, MPH, Corrona LLC; C.J. Etzel, PhD, Corrona LLC, and Department of Epidemiology, University of Texas, MD Anderson Cancer Center
| | - Sabrina Rebello
- From Corrona LLC, Southborough; Department of Medicine, University of Massachusetts Medical School, Worcester, Massachusetts; Amgen Inc., Thousand Oaks, California; Wade Outcomes Research and Consulting, Salt Lake City, Utah; New York University School of Medicine, New York, New York; Department of Epidemiology, University of Texas, MD Anderson Cancer Center, Houston, Texas, USA.,L.R. Harrold, MD, MPH, Corrona LLC, and Department of Medicine, University of Massachusetts Medical School; B.S. Stolshek, PharmD, Amgen Inc.; S. Rebello, MPH, Corrona LLC; D.H. Collier, MD, Amgen Inc.; A. Mutebi, PhD, Amgen Inc.; S.W. Wade, MPH, Wade Outcomes Research and Consulting; W. Malley, MS, Corrona LLC, and New York University School of Medicine; J.D. Greenberg, MD, MPH, Corrona LLC; C.J. Etzel, PhD, Corrona LLC, and Department of Epidemiology, University of Texas, MD Anderson Cancer Center
| | - David H Collier
- From Corrona LLC, Southborough; Department of Medicine, University of Massachusetts Medical School, Worcester, Massachusetts; Amgen Inc., Thousand Oaks, California; Wade Outcomes Research and Consulting, Salt Lake City, Utah; New York University School of Medicine, New York, New York; Department of Epidemiology, University of Texas, MD Anderson Cancer Center, Houston, Texas, USA.,L.R. Harrold, MD, MPH, Corrona LLC, and Department of Medicine, University of Massachusetts Medical School; B.S. Stolshek, PharmD, Amgen Inc.; S. Rebello, MPH, Corrona LLC; D.H. Collier, MD, Amgen Inc.; A. Mutebi, PhD, Amgen Inc.; S.W. Wade, MPH, Wade Outcomes Research and Consulting; W. Malley, MS, Corrona LLC, and New York University School of Medicine; J.D. Greenberg, MD, MPH, Corrona LLC; C.J. Etzel, PhD, Corrona LLC, and Department of Epidemiology, University of Texas, MD Anderson Cancer Center
| | - Alex Mutebi
- From Corrona LLC, Southborough; Department of Medicine, University of Massachusetts Medical School, Worcester, Massachusetts; Amgen Inc., Thousand Oaks, California; Wade Outcomes Research and Consulting, Salt Lake City, Utah; New York University School of Medicine, New York, New York; Department of Epidemiology, University of Texas, MD Anderson Cancer Center, Houston, Texas, USA.,L.R. Harrold, MD, MPH, Corrona LLC, and Department of Medicine, University of Massachusetts Medical School; B.S. Stolshek, PharmD, Amgen Inc.; S. Rebello, MPH, Corrona LLC; D.H. Collier, MD, Amgen Inc.; A. Mutebi, PhD, Amgen Inc.; S.W. Wade, MPH, Wade Outcomes Research and Consulting; W. Malley, MS, Corrona LLC, and New York University School of Medicine; J.D. Greenberg, MD, MPH, Corrona LLC; C.J. Etzel, PhD, Corrona LLC, and Department of Epidemiology, University of Texas, MD Anderson Cancer Center
| | - Sally W Wade
- From Corrona LLC, Southborough; Department of Medicine, University of Massachusetts Medical School, Worcester, Massachusetts; Amgen Inc., Thousand Oaks, California; Wade Outcomes Research and Consulting, Salt Lake City, Utah; New York University School of Medicine, New York, New York; Department of Epidemiology, University of Texas, MD Anderson Cancer Center, Houston, Texas, USA.,L.R. Harrold, MD, MPH, Corrona LLC, and Department of Medicine, University of Massachusetts Medical School; B.S. Stolshek, PharmD, Amgen Inc.; S. Rebello, MPH, Corrona LLC; D.H. Collier, MD, Amgen Inc.; A. Mutebi, PhD, Amgen Inc.; S.W. Wade, MPH, Wade Outcomes Research and Consulting; W. Malley, MS, Corrona LLC, and New York University School of Medicine; J.D. Greenberg, MD, MPH, Corrona LLC; C.J. Etzel, PhD, Corrona LLC, and Department of Epidemiology, University of Texas, MD Anderson Cancer Center
| | - Wendi Malley
- From Corrona LLC, Southborough; Department of Medicine, University of Massachusetts Medical School, Worcester, Massachusetts; Amgen Inc., Thousand Oaks, California; Wade Outcomes Research and Consulting, Salt Lake City, Utah; New York University School of Medicine, New York, New York; Department of Epidemiology, University of Texas, MD Anderson Cancer Center, Houston, Texas, USA.,L.R. Harrold, MD, MPH, Corrona LLC, and Department of Medicine, University of Massachusetts Medical School; B.S. Stolshek, PharmD, Amgen Inc.; S. Rebello, MPH, Corrona LLC; D.H. Collier, MD, Amgen Inc.; A. Mutebi, PhD, Amgen Inc.; S.W. Wade, MPH, Wade Outcomes Research and Consulting; W. Malley, MS, Corrona LLC, and New York University School of Medicine; J.D. Greenberg, MD, MPH, Corrona LLC; C.J. Etzel, PhD, Corrona LLC, and Department of Epidemiology, University of Texas, MD Anderson Cancer Center
| | - Jeffrey D Greenberg
- From Corrona LLC, Southborough; Department of Medicine, University of Massachusetts Medical School, Worcester, Massachusetts; Amgen Inc., Thousand Oaks, California; Wade Outcomes Research and Consulting, Salt Lake City, Utah; New York University School of Medicine, New York, New York; Department of Epidemiology, University of Texas, MD Anderson Cancer Center, Houston, Texas, USA.,L.R. Harrold, MD, MPH, Corrona LLC, and Department of Medicine, University of Massachusetts Medical School; B.S. Stolshek, PharmD, Amgen Inc.; S. Rebello, MPH, Corrona LLC; D.H. Collier, MD, Amgen Inc.; A. Mutebi, PhD, Amgen Inc.; S.W. Wade, MPH, Wade Outcomes Research and Consulting; W. Malley, MS, Corrona LLC, and New York University School of Medicine; J.D. Greenberg, MD, MPH, Corrona LLC; C.J. Etzel, PhD, Corrona LLC, and Department of Epidemiology, University of Texas, MD Anderson Cancer Center
| | - Carol J Etzel
- From Corrona LLC, Southborough; Department of Medicine, University of Massachusetts Medical School, Worcester, Massachusetts; Amgen Inc., Thousand Oaks, California; Wade Outcomes Research and Consulting, Salt Lake City, Utah; New York University School of Medicine, New York, New York; Department of Epidemiology, University of Texas, MD Anderson Cancer Center, Houston, Texas, USA.,L.R. Harrold, MD, MPH, Corrona LLC, and Department of Medicine, University of Massachusetts Medical School; B.S. Stolshek, PharmD, Amgen Inc.; S. Rebello, MPH, Corrona LLC; D.H. Collier, MD, Amgen Inc.; A. Mutebi, PhD, Amgen Inc.; S.W. Wade, MPH, Wade Outcomes Research and Consulting; W. Malley, MS, Corrona LLC, and New York University School of Medicine; J.D. Greenberg, MD, MPH, Corrona LLC; C.J. Etzel, PhD, Corrona LLC, and Department of Epidemiology, University of Texas, MD Anderson Cancer Center
| |
Collapse
|
44
|
Bergman M, Lundholm A. Mitigation of disease- and treatment-related risks in patients with psoriatic arthritis. Arthritis Res Ther 2017; 19:63. [PMID: 28320454 PMCID: PMC5359960 DOI: 10.1186/s13075-017-1265-5] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Psoriatic arthritis is a part of the family of diseases referred to as spondyloarthropathies, a diverse group of chronic inflammatory disorders with common clinical, radiographic, and genetic features. Peripheral arthritis is the most common symptom of psoriatic arthritis and patients also frequently experience involvement of the entheses, spine, skin, and nails. Due to the diverse clinical spectrum of disease severity, tissues affected, and associated comorbidities, the treatment of psoriatic arthritis can be challenging and it is necessary to mitigate risks associated with both the disease and its treatment. These risks include disease-specific, treatment-related, and psychological risks. Disease-specific risks include those associated with disease progression that can limit functional status and be mitigated through early diagnosis and initiation of treatment. Risks also arise from comorbidities that are associated with psoriatic arthritis such as cardiovascular disease, obesity, diabetes mellitus, and gastrointestinal inflammation. Patient outcomes can be affected by the treatment strategy employed and the pharmacologic agents administered. Additionally, it is important for physicians to be aware of risks specific to each therapeutic option. The impact of psoriatic arthritis is not limited to the skin and joints and it is common for patients to experience quality-of-life impairment. Patients are also more likely to have depression, anxiety, and alcoholism. This article reviews the many risks associated with psoriatic arthritis and provides guidance on mitigating these risks.
Collapse
Affiliation(s)
- Martin Bergman
- Taylor Hospital, 8 Morton Avenue, Suite 304, Ridley Park, PA, 19078, USA.
| | | |
Collapse
|
45
|
Mease PJ, Lesperance T, Liu M, Collier DH, Mason M, Deveikis S, Accortt NA. Changes in Treatment Patterns in Patients with Psoriatic Arthritis Initiating Biologic and Nonbiologic Therapy in a Clinical Registry. J Rheumatol 2017; 44:184-192. [PMID: 28089969 DOI: 10.3899/jrheum.160343] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/21/2016] [Indexed: 12/18/2022]
Abstract
OBJECTIVE Treatment options for psoriatic arthritis (PsA) have increased and improved in the past decade; treatment patterns in PsA remain poorly understood. Understanding current practices would aid in treatment management of patients with PsA. METHODS This observational study was based on data from the Corrona registry of adult patients with PsA in North America collected between January 1, 2004, and December 31, 2012. Patients were divided among 3 therapy cohorts: tumor necrosis factor inhibitor (TNFi) monotherapy, methotrexate (MTX) monotherapy, and TNFi and MTX combination therapy. Patients were further divided among 3 study periods to understand changes over time: 2004-2006, 2007-2009, and 2010-2012. Data were collected on persistence, discontinuation, restarting, switching, adding/dropping therapy, and dose stretching. RESULTS This study included 520 patients: 190 TNFi monotherapy, 217 MTX monotherapy, and 113 combination therapy; 110 from 2004 to 2006, 192 from 2007 to 2009, and 218 from 2010 to 2012. Over time, the proportion of patients initiating TNFi monotherapy decreased, while the proportion initiating combination therapy remained constant. The percentage of patients who were persistent decreased over time across all therapy cohorts, but remained higher in TNFi monotherapy than in other cohorts. Duration of persistence decreased over time. Patients initiating MTX monotherapy were more likely than their TNFi counterparts to add therapy. CONCLUSION Treatment patterns in patients with PsA have changed from 2004 to 2012. Physicians are not more likely to initiate TNFi monotherapy, although clinical evidence supporting its effectiveness has increased over this study period, and patients remain more persistent with it.
Collapse
Affiliation(s)
- Philip J Mease
- From the Swedish Medical Center and University of Washington, Seattle, Washington; Amgen Inc., Thousand Oaks, California; Corrona LLC, Southborough, Massachusetts, USA. .,Sponsored by Corrona LLC and funded by Amgen Inc. The Corrona Rheumatoid Arthritis registry has been supported through contracted subscriptions in the last 2 years by AbbVie, Amgen Inc., AstraZeneca, Bristol-Myers Squibb, Genentech, Horizon Pharma USA, Janssen, Eli Lilly, Novartis, Pfizer, and UCB. P.J. Mease has received research grants from AbbVie, Amgen Inc., Bristol-Myers Squibb, Eli Lilly, Novartis, Pfizer, and UCB; consulting fees from AbbVie, Amgen Inc., Bristol-Myers Squibb, Corrona, Crescendo, Eli Lilly, Merck, Novartis, Pfizer, and UCB; and speaker fees from AbbVie, Amgen Inc., Biogen Idec, Bristol-Myers Squibb, Crescendo, Eli Lilly, Pfizer, and UCB. T. Lesperance is a contractor for and has received salary from Amgen Inc. M. Liu is an employee of Corrona LLC. D.H. Collier is an employee and stockholder of Amgen Inc. M. Mason is an employee of Corrona LLC. S. Deveikis is an employee of Corrona LLC. N.A. Accortt is an employee and stockholder of Amgen Inc. P.J. Mease, MD, Swedish Medical Center and University of Washington; T. Lesperance, MPH, Amgen Inc.; M. Liu, PhD, Corrona LLC; D.H. Collier, MD, Amgen Inc.; M. Mason, PhD, Corrona LLC; S. Deveikis, MPH, Corrona LLC; N.A. Accortt, PhD, Amgen Inc.
| | - Tamara Lesperance
- From the Swedish Medical Center and University of Washington, Seattle, Washington; Amgen Inc., Thousand Oaks, California; Corrona LLC, Southborough, Massachusetts, USA.,Sponsored by Corrona LLC and funded by Amgen Inc. The Corrona Rheumatoid Arthritis registry has been supported through contracted subscriptions in the last 2 years by AbbVie, Amgen Inc., AstraZeneca, Bristol-Myers Squibb, Genentech, Horizon Pharma USA, Janssen, Eli Lilly, Novartis, Pfizer, and UCB. P.J. Mease has received research grants from AbbVie, Amgen Inc., Bristol-Myers Squibb, Eli Lilly, Novartis, Pfizer, and UCB; consulting fees from AbbVie, Amgen Inc., Bristol-Myers Squibb, Corrona, Crescendo, Eli Lilly, Merck, Novartis, Pfizer, and UCB; and speaker fees from AbbVie, Amgen Inc., Biogen Idec, Bristol-Myers Squibb, Crescendo, Eli Lilly, Pfizer, and UCB. T. Lesperance is a contractor for and has received salary from Amgen Inc. M. Liu is an employee of Corrona LLC. D.H. Collier is an employee and stockholder of Amgen Inc. M. Mason is an employee of Corrona LLC. S. Deveikis is an employee of Corrona LLC. N.A. Accortt is an employee and stockholder of Amgen Inc. P.J. Mease, MD, Swedish Medical Center and University of Washington; T. Lesperance, MPH, Amgen Inc.; M. Liu, PhD, Corrona LLC; D.H. Collier, MD, Amgen Inc.; M. Mason, PhD, Corrona LLC; S. Deveikis, MPH, Corrona LLC; N.A. Accortt, PhD, Amgen Inc
| | - Mei Liu
- From the Swedish Medical Center and University of Washington, Seattle, Washington; Amgen Inc., Thousand Oaks, California; Corrona LLC, Southborough, Massachusetts, USA.,Sponsored by Corrona LLC and funded by Amgen Inc. The Corrona Rheumatoid Arthritis registry has been supported through contracted subscriptions in the last 2 years by AbbVie, Amgen Inc., AstraZeneca, Bristol-Myers Squibb, Genentech, Horizon Pharma USA, Janssen, Eli Lilly, Novartis, Pfizer, and UCB. P.J. Mease has received research grants from AbbVie, Amgen Inc., Bristol-Myers Squibb, Eli Lilly, Novartis, Pfizer, and UCB; consulting fees from AbbVie, Amgen Inc., Bristol-Myers Squibb, Corrona, Crescendo, Eli Lilly, Merck, Novartis, Pfizer, and UCB; and speaker fees from AbbVie, Amgen Inc., Biogen Idec, Bristol-Myers Squibb, Crescendo, Eli Lilly, Pfizer, and UCB. T. Lesperance is a contractor for and has received salary from Amgen Inc. M. Liu is an employee of Corrona LLC. D.H. Collier is an employee and stockholder of Amgen Inc. M. Mason is an employee of Corrona LLC. S. Deveikis is an employee of Corrona LLC. N.A. Accortt is an employee and stockholder of Amgen Inc. P.J. Mease, MD, Swedish Medical Center and University of Washington; T. Lesperance, MPH, Amgen Inc.; M. Liu, PhD, Corrona LLC; D.H. Collier, MD, Amgen Inc.; M. Mason, PhD, Corrona LLC; S. Deveikis, MPH, Corrona LLC; N.A. Accortt, PhD, Amgen Inc
| | - David H Collier
- From the Swedish Medical Center and University of Washington, Seattle, Washington; Amgen Inc., Thousand Oaks, California; Corrona LLC, Southborough, Massachusetts, USA.,Sponsored by Corrona LLC and funded by Amgen Inc. The Corrona Rheumatoid Arthritis registry has been supported through contracted subscriptions in the last 2 years by AbbVie, Amgen Inc., AstraZeneca, Bristol-Myers Squibb, Genentech, Horizon Pharma USA, Janssen, Eli Lilly, Novartis, Pfizer, and UCB. P.J. Mease has received research grants from AbbVie, Amgen Inc., Bristol-Myers Squibb, Eli Lilly, Novartis, Pfizer, and UCB; consulting fees from AbbVie, Amgen Inc., Bristol-Myers Squibb, Corrona, Crescendo, Eli Lilly, Merck, Novartis, Pfizer, and UCB; and speaker fees from AbbVie, Amgen Inc., Biogen Idec, Bristol-Myers Squibb, Crescendo, Eli Lilly, Pfizer, and UCB. T. Lesperance is a contractor for and has received salary from Amgen Inc. M. Liu is an employee of Corrona LLC. D.H. Collier is an employee and stockholder of Amgen Inc. M. Mason is an employee of Corrona LLC. S. Deveikis is an employee of Corrona LLC. N.A. Accortt is an employee and stockholder of Amgen Inc. P.J. Mease, MD, Swedish Medical Center and University of Washington; T. Lesperance, MPH, Amgen Inc.; M. Liu, PhD, Corrona LLC; D.H. Collier, MD, Amgen Inc.; M. Mason, PhD, Corrona LLC; S. Deveikis, MPH, Corrona LLC; N.A. Accortt, PhD, Amgen Inc
| | - Marc Mason
- From the Swedish Medical Center and University of Washington, Seattle, Washington; Amgen Inc., Thousand Oaks, California; Corrona LLC, Southborough, Massachusetts, USA.,Sponsored by Corrona LLC and funded by Amgen Inc. The Corrona Rheumatoid Arthritis registry has been supported through contracted subscriptions in the last 2 years by AbbVie, Amgen Inc., AstraZeneca, Bristol-Myers Squibb, Genentech, Horizon Pharma USA, Janssen, Eli Lilly, Novartis, Pfizer, and UCB. P.J. Mease has received research grants from AbbVie, Amgen Inc., Bristol-Myers Squibb, Eli Lilly, Novartis, Pfizer, and UCB; consulting fees from AbbVie, Amgen Inc., Bristol-Myers Squibb, Corrona, Crescendo, Eli Lilly, Merck, Novartis, Pfizer, and UCB; and speaker fees from AbbVie, Amgen Inc., Biogen Idec, Bristol-Myers Squibb, Crescendo, Eli Lilly, Pfizer, and UCB. T. Lesperance is a contractor for and has received salary from Amgen Inc. M. Liu is an employee of Corrona LLC. D.H. Collier is an employee and stockholder of Amgen Inc. M. Mason is an employee of Corrona LLC. S. Deveikis is an employee of Corrona LLC. N.A. Accortt is an employee and stockholder of Amgen Inc. P.J. Mease, MD, Swedish Medical Center and University of Washington; T. Lesperance, MPH, Amgen Inc.; M. Liu, PhD, Corrona LLC; D.H. Collier, MD, Amgen Inc.; M. Mason, PhD, Corrona LLC; S. Deveikis, MPH, Corrona LLC; N.A. Accortt, PhD, Amgen Inc
| | - Sabrina Deveikis
- From the Swedish Medical Center and University of Washington, Seattle, Washington; Amgen Inc., Thousand Oaks, California; Corrona LLC, Southborough, Massachusetts, USA.,Sponsored by Corrona LLC and funded by Amgen Inc. The Corrona Rheumatoid Arthritis registry has been supported through contracted subscriptions in the last 2 years by AbbVie, Amgen Inc., AstraZeneca, Bristol-Myers Squibb, Genentech, Horizon Pharma USA, Janssen, Eli Lilly, Novartis, Pfizer, and UCB. P.J. Mease has received research grants from AbbVie, Amgen Inc., Bristol-Myers Squibb, Eli Lilly, Novartis, Pfizer, and UCB; consulting fees from AbbVie, Amgen Inc., Bristol-Myers Squibb, Corrona, Crescendo, Eli Lilly, Merck, Novartis, Pfizer, and UCB; and speaker fees from AbbVie, Amgen Inc., Biogen Idec, Bristol-Myers Squibb, Crescendo, Eli Lilly, Pfizer, and UCB. T. Lesperance is a contractor for and has received salary from Amgen Inc. M. Liu is an employee of Corrona LLC. D.H. Collier is an employee and stockholder of Amgen Inc. M. Mason is an employee of Corrona LLC. S. Deveikis is an employee of Corrona LLC. N.A. Accortt is an employee and stockholder of Amgen Inc. P.J. Mease, MD, Swedish Medical Center and University of Washington; T. Lesperance, MPH, Amgen Inc.; M. Liu, PhD, Corrona LLC; D.H. Collier, MD, Amgen Inc.; M. Mason, PhD, Corrona LLC; S. Deveikis, MPH, Corrona LLC; N.A. Accortt, PhD, Amgen Inc
| | - Neil A Accortt
- From the Swedish Medical Center and University of Washington, Seattle, Washington; Amgen Inc., Thousand Oaks, California; Corrona LLC, Southborough, Massachusetts, USA.,Sponsored by Corrona LLC and funded by Amgen Inc. The Corrona Rheumatoid Arthritis registry has been supported through contracted subscriptions in the last 2 years by AbbVie, Amgen Inc., AstraZeneca, Bristol-Myers Squibb, Genentech, Horizon Pharma USA, Janssen, Eli Lilly, Novartis, Pfizer, and UCB. P.J. Mease has received research grants from AbbVie, Amgen Inc., Bristol-Myers Squibb, Eli Lilly, Novartis, Pfizer, and UCB; consulting fees from AbbVie, Amgen Inc., Bristol-Myers Squibb, Corrona, Crescendo, Eli Lilly, Merck, Novartis, Pfizer, and UCB; and speaker fees from AbbVie, Amgen Inc., Biogen Idec, Bristol-Myers Squibb, Crescendo, Eli Lilly, Pfizer, and UCB. T. Lesperance is a contractor for and has received salary from Amgen Inc. M. Liu is an employee of Corrona LLC. D.H. Collier is an employee and stockholder of Amgen Inc. M. Mason is an employee of Corrona LLC. S. Deveikis is an employee of Corrona LLC. N.A. Accortt is an employee and stockholder of Amgen Inc. P.J. Mease, MD, Swedish Medical Center and University of Washington; T. Lesperance, MPH, Amgen Inc.; M. Liu, PhD, Corrona LLC; D.H. Collier, MD, Amgen Inc.; M. Mason, PhD, Corrona LLC; S. Deveikis, MPH, Corrona LLC; N.A. Accortt, PhD, Amgen Inc
| |
Collapse
|
46
|
Saeki H, Nakagawa H, Nakajo K, Ishii T, Morisaki Y, Aoki T, Cameron GS, Osuntokun OO. Efficacy and safety of ixekizumab treatment for Japanese patients with moderate to severe plaque psoriasis, erythrodermic psoriasis and generalized pustular psoriasis: Results from a 52-week, open-label, phase 3 study (UNCOVER-J). J Dermatol 2016; 44:355-362. [PMID: 27726163 PMCID: PMC5412888 DOI: 10.1111/1346-8138.13622] [Citation(s) in RCA: 148] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2016] [Accepted: 08/18/2016] [Indexed: 11/30/2022]
Abstract
Psoriasis, a chronic, immune‐mediated skin disease characterized by red, scaly plaques, affects approximately 0.3% of the population in Japan. The aim of this open‐label study was to evaluate the long‐term efficacy and safety of ixekizumab, a humanized, anti‐interleukin‐17A monoclonal antibody, in Japanese patients with plaque psoriasis (n = 78, including 11 psoriatic arthritis), erythrodermic psoriasis (n = 8) and generalized pustular psoriasis (n = 5). Ixekizumab was administrated s.c. at baseline (week 0, 160 mg), from weeks 2 to 12 (80 mg every 2 weeks), and from weeks 16 to 52 (80 mg every 4 weeks). At week 52, 92.3% of patients with plaque psoriasis achieved Psoriasis Area and Severity Index (PASI) 75, 80.8% achieved PASI 90, 48.7% achieved PASI 100, and 52.6% had remission of plaques (by static Physician Global Assessment, sPGA [0]). Difficult to treat areas of psoriasis (nail or scalp) also responded to ixekizumab. All patients with psoriatic arthritis who were assessed (5/5) achieved an American College of Rheumatology 20 response. Most patients with erythrodermic psoriasis or generalized pustular psoriasis responded to ixekizumab and the clinical outcome was maintained over 52 weeks (75% and 60% of patients achieved sPGA [0, 1] at week 52, respectively). Mostly mild or moderate treatment‐emergent adverse events were reported by 79 of 91 patients; the most common were nasopharyngitis, eczema, seborrheic dermatitis, urticaria and injection site reactions. In conclusion, 52‐week ixekizumab treatment was efficacious and well tolerated in Japanese patients with plaque psoriasis. Efficacy was also observed in patients with erythrodermic psoriasis, generalized pustular psoriasis and psoriatic arthritis.
Collapse
Affiliation(s)
- Hidehisa Saeki
- Department of Dermatology, Nippon Medical School, Tokyo, Japan
| | - Hidemi Nakagawa
- Department of Dermatology, The Jikei University School of Medicine, Tokyo, Japan
| | - Ko Nakajo
- Lilly Research Laboratories, Eli Lilly Japan K.K., Kobe, Japan
| | - Taeko Ishii
- Lilly Research Laboratories, Eli Lilly Japan K.K., Kobe, Japan
| | - Yoji Morisaki
- Lilly Research Laboratories, Eli Lilly Japan K.K., Kobe, Japan
| | - Takehiro Aoki
- Lilly Research Laboratories, Eli Lilly Japan K.K., Kobe, Japan
| | - Gregory S Cameron
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana, USA
| | - Olawale O Osuntokun
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana, USA
| | | |
Collapse
|
47
|
Hernández-Flórez D, Valor L. Selective Phosphodiesterase Inhibitors: A New Therapeutic Option in Inflammation and Autoimmunity. ACTA ACUST UNITED AC 2016; 12:303-306. [PMID: 27567299 DOI: 10.1016/j.reuma.2016.07.011] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2016] [Revised: 07/20/2016] [Accepted: 07/22/2016] [Indexed: 12/24/2022]
Affiliation(s)
- Diana Hernández-Flórez
- Servicio de Reumatología, Hospital General Universitario Gregorio Marañón, Madrid, España; Instituto de Investigación Biomédica, Hospital Gregorio Marañón, Madrid, España
| | - Lara Valor
- Servicio de Reumatología, Hospital General Universitario Gregorio Marañón, Madrid, España; Instituto de Investigación Biomédica, Hospital Gregorio Marañón, Madrid, España.
| |
Collapse
|
48
|
Torres T, Faria R. Ustekinumab: The "New Kid on the Block" in the Treatment of Psoriatic Arthritis. Drug Dev Res 2015; 76:428-31. [PMID: 26372543 DOI: 10.1002/ddr.21279] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2015] [Accepted: 08/23/2015] [Indexed: 11/07/2022]
Abstract
Psoriatic arthritis (PsA) is a chronic inflammatory seronegative spondyloarthritis associated with psoriasis. While TNF-α inhibitors have revolutionized the treatment of rheumatic diseases, including PsA, not all patients respond to these agents while others are unsuitable or intolerant to them. Thus, there is a need for additional treatment modalities with a novel mechanism of action. In the past years, the IL-23/Th17 axis has emerged as an important mechanism in the pathogenesis of PsA. Ustekinumab, a fully human IgG1κ monoclonal antibody that targets the common subunit p40 of IL-12 and IL-23, has been shown in clinical trials, to be well-tolerated and effective in patients with active PsA. It improved signs and symptoms of PsA, inhibited radiographic progression and was effective in dactylitis, enthesitis, and skin disease, with a safety profile consistent with the one observed in patients with psoriasis. Moreover, it was to be effective in anti-TNF-α experienced patients, definitely fulfilling an unmet need in the management of PsA.
Collapse
Affiliation(s)
- Tiago Torres
- Department of Dermatology, Centro Hospitalar do Porto, Porto, Portugal.,Instituto de Ciências Biomédicas Abel Salazar, University of Porto, Porto, Portugal
| | - Raquel Faria
- Instituto de Ciências Biomédicas Abel Salazar, University of Porto, Porto, Portugal.,Clinical Immunology Unit, Centro Hospitalar do Porto, Porto, Portugal
| |
Collapse
|