Sustained and improved guselkumab response in patients with active psoriatic arthritis regardless of baseline demographic and disease characteristics: pooled results through week 52 of two phase III, randomised, placebo-controlled studies

Christopher T Ritchlin, Philip J Mease, Wolf-Henning Boehncke, John Tesser, Elena Schiopu, Soumya D Chakravarty, Alexa P Kollmeier, Xie L Xu, May Shawi, Yusang Jiang, Shihong Sheng, Yanli Wang, Stephen Xu, Joseph F Merola, Iain B McInnes, Atul Deodhar, Christopher T Ritchlin, Philip J Mease, Wolf-Henning Boehncke, John Tesser, Elena Schiopu, Soumya D Chakravarty, Alexa P Kollmeier, Xie L Xu, May Shawi, Yusang Jiang, Shihong Sheng, Yanli Wang, Stephen Xu, Joseph F Merola, Iain B McInnes, Atul Deodhar

Abstract

Objectives: To evaluate the efficacy through 52 weeks of guselkumab, an interleukin 23-p19 subunit inhibitor, in subgroups of pooled psoriatic arthritis (PsA) patients from the DISCOVER-1 and DISCOVER-2 trials defined by baseline patient characteristics.

Methods: Adults with active PsA despite standard therapies were enrolled in DISCOVER-1 (≥3 swollen and ≥3 tender joints, C reactive protein (CRP) level ≥0.3 mg/dL) and DISCOVER-2 (≥5 swollen and ≥5 tender joints, CRP ≥0.6 mg/dL, biological-naïve). Randomised patients received 100 mg guselkumab at weeks 0, 4, and then every 4 or 8 weeks (Q4W/Q8W) or placebo. Guselkumab effects on joint (ACR20/50/70), skin (IGA 0/1, IGA 0), patient-reported outcome (Health Assessment Questionnaire Disability Index/Functional Assessment of Chronic Illness Therapy-Fatigue) and disease severity (minimal disease activity/PsA Disease Activity Score low disease activity) endpoints were evaluated by patient sex, body mass index, PsA duration, swollen/tender joint counts, CRP level, percent body surface area with psoriasis, Psoriasis Area and Severity Index score, and conventional synthetic disease-modifying antirheumatic drug use at baseline.

Results: Baseline patients characteristics in DISCOVER-1 (N=381) and DISCOVER-2 (N=739) were well balanced across randomised groups. At week 24, 62% (232/373) and 60% (225/375), respectively, of guselkumab Q4W-treated and Q8W-treated patients pooled across DISCOVER-1 and DISCOVER-2 achieved the primary endpoint of ACR20 response versus 29% (109/372) of placebo-treated patients. Guselkumab treatment effect at week 24 was observed across patient subgroups. Within each patient subgroup, response rates across all disease domains were sustained or increased at week 52 with both guselkumab regimens.

Conclusions: Guselkumab Q4W and Q8W resulted in robust and sustained improvements in PsA signs and symptoms consistently in subgroups of patients defined by diverse baseline characteristics.

Trial registration numbers: NCT03162796, NCT03158285.

Keywords: arthritis, psoriatic; biological therapy; therapeutics.

Conflict of interest statement

Competing interests: CTR has received consulting fees from AbbVie, Amgen, Bristol Myers Squibb, Janssen, Lilly, MoonLake, Novartis, Pfizer and UCB Pharma; and speaker fees from Novartis, Pfizer and UCB Pharma. PJM has received research grants, consulting fees and/or speaker fees from AbbVie, Amgen, Boehringer Ingelheim, Bristol Myers Squibb, Lilly, Galapagos, Gilead, GlaxoSmithKline, Janssen, Novartis, Pfizer, SUN Pharma and UCB Pharma. W-HB has received consulting fees from AbbVie, Almirall, Bristol Myers Squibb, Celgene, Lilly, Janssen, Leo, Novartis and UCB Pharma; speaker fees from AbbVie, Almirall, Janssen, Leo and UCB Pharma; fees for expert testimony from Novartis; and fees for participation on an advisory board from AbbVie, Almirall, Janssen, Leo, Novartis and UCB Pharma. JT has received research grants from AbbVie, Amgen, Bristol Myers Squibb, Celgene, CoreVitas, Lilly, Gilead, Janssen, Pfizer and Sun Pharma; consulting fees from AbbVie, Lilly, Janssen, Novartis and Pfizer; speaker fees from AbbVie, Amgen, Bristol Myers Squibb, Lilly, Janssen and Pfizer; and advisory board fees from Bristol Myers Squibb, Lilly, Gilead, Janssen, Novartis and Pfizer. ES has received research grants and consulting fees from Janssen. SDC is an employee of Janssen Scientific Affairs, LLC and owns stock in Johnson & Johnson, of which Janssen Scientific Affairs, LLC is a wholly owned subsidiary. APK, XLX, SS and SX are employees of Janssen Research & Development, LLC and own stock in Johnson & Johnson, of which Janssen Research & Development is a wholly owned subsidiary. MS is an employee of Janssen Pharmaceutical Companies of Johnson & Johnson and owns stock in Johnson & Johnson. YJ is a consultant employed by Cytel, Inc and funded by Janssen to provide statistical support. YW is a consultant employed by IQVIA, Inc and funded by Janssen to provide statistical support. JFM has received consulting fees from AbbVie, Arena, Biogen, Bristol Myers Squibb, Dermavant, Janssen, Lilly, Novartis, Pfizer, Sun Pharma and UCB Pharma. IBM has received consulting fees from AbbVie, Amgen, Bristol Myers Squibb, Causeway Therapeutics, Gilead, Janssen, Lilly, Novartis, Pfizer, Regeneron and Sanofi; has served as a board member for NHS Greater Glasgow & Clyde; has served as Vice Principal & Head of MVLS College at University of Glasgow; and has received research grants from AbbVie, Amgen, Bristol Myers Squibb, Causeway Therapeutics, Lilly, Gilead, Janssen, Novartis and Pfizer. AD has received consulting fees from AbbVie, Amgen, Bristol Myers Squibb, GlaxoSmithKline, Janssen, Lilly, MoonLake, Novartis, Pfizer and UCB Pharma; speaker fees from Janssen, Novartis, Pfizer and UCB Pharma; and advisory board fees from AbbVie, GlaxoSmithKline, Janssen, MoonLake, Novartis and UCB Pharma.

© Author(s) (or their employer(s)) 2022. Re-use permitted under CC BY-NC. No commercial re-use. See rights and permissions. Published by BMJ.

Figures

Figure 1
Figure 1
Proportions of patients achieving ACR20 response at week 24 and week 52 by patient demographics, disease characteristics and csDMARD use at baseline. Week 24 response rates (NRI) are compared between guselkumab (GUS) and placebo (PBO) via ORs and 95% CIs (depicted via forest plot; actual values provided in online supplemental table 1). Week 52 response rates (NRI) for GUS Q4W and Q8W were determined to assess durability of ACR20 response rates within each patient subgroup. Prior to week 24, patients meeting treatment failure criteria were considered non-responders. Missing data through weeks 24 and 52 were imputed as non-response. ACR20, American College of Rheumatology 20% improvement; BMI, body mass index; CI, confidence interval; CRP, C reactive protein; csDMARD, conventional synthetic disease-modifying antirheumatic drug; MTX, methotrexate; NRI, non-responder imputation; OR, odds ratio; PsA, psoriatic arthritis; Q4W/Q8W, every 4 weeks/every 8 weeks.
Figure 2
Figure 2
Proportions of patients achieving (A) ACR50 and (B) ACR70 responses at week 24 and week 52 by patient demographics, disease characteristics and csDMARD use at baseline. Week 24 response rates (NRI) are compared between guselkumab (GUS) and placebo (PBO) via ORs and 95% CIs (depicted via forest plot; actual values provided in online supplemental table 1). Week 52 response rates (NRI) for GUS Q4W and GUS Q8W were determined to assess durability of ACR50 and ACR70 response rates within each patient subgroup. Prior to week 24, patients meeting treatment failure criteria were considered non-responders. Missing data through Weeks 24 and 52 were imputed as non-response. ACR50/70, American College of Rheumatology 50/70% improvement; BMI, body mass index; CI, confidence interval; CRP, C reactive protein; csDMARD, conventional synthetic disease-modifying antirheumatic drug; MTX, methotrexate; NRI, non-responder imputation; OR, odds ratio; PsA, psoriatic arthritis; Q4W/Q8W, every 4 weeks/every 8 weeks.
Figure 3
Figure 3
Proportions of patients with ≥3% BSA with PsO and IGA ≥2 at baseline achieving an (A) IGA 0/1 response (IGA psoriasis score of 0 (cleared) or 1 (minimal) and ≥2-grade reduction from baseline) and (B) IGA 0 response (IGA psoriasis score of 0) at week 24 and week 52 by patient demographics, disease characteristics, and csDMARD use at baseline. Week 24 response rates (NRI) are compared between guselkumab (GUS) and placebo (PBO) via ORs and 95% CIs (depicted via forest plot; actual values provided in online supplemental table 2). Week 52 response rates (NRI) for GUS Q4W and GUS Q8W were determined to assess durability of IGA 0/1 and IGA 0 response rates within each patient subgroup. Prior to week 24, patients meeting treatment failure criteria were considered non-responders. Missing data through weeks 24 and 52 were imputed as non-response. *Plot is undrawable due to NC values. BMI, body mass index; BSA, body surface area; CI, confidence interval; CRP, C reactive protein; csDMARD, conventional synthetic disease-modifying antirheumatic drug; IGA, Investigator’s Global Assessment; MTX, methotrexate; NC, no count; NRI, non-responder imputation; OR, odds ratio; PASI, Psoriasis Area Severity Index; PsA, psoriatic arthritis; Q4W/Q8W, every 4 weeks/every 8 weeks.
Figure 4
Figure 4
Proportions of patients achieving (A) FACIT-Fatigue response (≥4-point improvement in FACIT-F score from baseline) and (B) HAQ-DI response (≥0.35-point improvement in HAQ-DI score from baseline among patients with a HAQ-DI score ≥0.35 at baseline) at week 24 and week 52 by patient demographics, disease characteristics and csDMARD use at baseline. Week 24 response rates (NRI) are compared between guselkumab (GUS) and placebo (PBO) via ORs and 95% CIs (depicted via forest plot; actual values provided in online supplemental table 3). Week 52 response rates (NRI) for GUS Q4W and GUS Q8W were determined to assess durability of FACIT-F and HAQ-DI response rates within each patient subgroup. Prior to week 24, patients meeting treatment failure criteria were considered non-responders. Missing data through weeks 24 and 52 were imputed as non-response. BMI, body mass index; CI, confidence interval; CRP, C reactive protein; csDMARD, conventional synthetic disease-modifying antirheumatic drug; FACIT-F, Functional Assessment of Chronic Illness Therapy-Fatigue; HAQ-DI, Health Assessment Questionnaire Disability Index; MTX, methotrexate; NRI, non-responder imputation; OR, odds ratio; PsA, psoriatic arthritis; Q4W/Q8W, every 4 weeks/every 8 weeks.
Figure 5
Figure 5
Proportions of patients achieving (A) PASDAS Low Disease Activity (PASDAS score ≤3.2) and (B) MDA responses at week 24 and week 52 by patient demographics, disease characteristics and DMARD use at baseline. Week 24 response rates (NRI) are compared between guselkumab (GUS) and placebo (PBO) via ORs and 95% CIs (depicted via forest plot; actual values provided in online supplemental table 4). Week 52 response rates (NRI) for GUS Q4W and GUS Q8W were determined to assess durability of PASDAS LDA and MDA response rates within each patient subgroup. Prior to week 24, patients meeting treatment failure criteria were considered non-responders. Missing data through weeks 24 and 52 were imputed as non-response. *Plot is not drawable due to NC values. BMI, body mass index; BSA, body surface area; CI, confidence interval; CRP, C reactive protein; csDMARD, conventional synthetic disease-modifying antirheumatic drug; LDA, low disease activity; MDA, minimal disease activity; MTX, methotrexate; NC, no count; NRI, non-responder imputation; OR, odds ratio; PASDAS, Psoriatic Arthritis Disease Activity Score; PASI, Psoriasis Area Severity Index; PsA, psoriatic arthritis; Q4W/Q8W, every 4 weeks/every 8 weeks.

References

    1. Gossec L, Baraliakos X, Kerschbaumer A, et al. . EULAR recommendations for the management of psoriatic arthritis with pharmacological therapies: 2019 update. Ann Rheum Dis 2020;79:700–12. 10.1136/annrheumdis-2020-217159
    1. Coates LC, Kavanaugh A, Mease PJ, et al. . Group for Research and Assessment of Psoriasis and Psoriatic Arthritis 2015 treatment recommendations for psoriatic arthritis. Arthritis Rheumatol 2016;68:1060–71. 10.1002/art.39573
    1. Coates LC, Soriano E, Corp N, et al. . The Group for Research and Assessment of Psoriasis and Psoriatic Arthritis (GRAPPA) treatment recommendations 2021. Ann Rheum Dis 2021;80:139–40. 10.1136/annrheumdis-2021-eular.4091
    1. Yeung H, Wan J, Van Voorhees AS, et al. . Patient-reported reasons for the discontinuation of commonly used treatments for moderate to severe psoriasis. J Am Acad Dermatol 2013;68:64–72. 10.1016/j.jaad.2012.06.035
    1. Lorenzin M, Ortolan A, Cozzi G, et al. . Predictive factors for switching in patients with psoriatic arthritis undergoing anti-TNFα, anti-IL12/23, or anti-IL17 drugs: a 15-year monocentric real-life study. Clin Rheumatol 2021;40:4569–80. 10.1007/s10067-021-05799-0
    1. Janssen Biotech Inc . Tremfya: package insert. Horsham, PA, 2020.
    1. Boehncke W-H, Brembilla NC, Nissen MJ. Guselkumab: the first selective IL-23 inhibitor for active psoriatic arthritis in adults. Expert Rev Clin Immunol 2021;17:5–13. 10.1080/1744666X.2020.1857733
    1. Deodhar A, Helliwell PS, Boehncke W-H, et al. . Guselkumab in patients with active psoriatic arthritis who were biologic-naive or had previously received TNFα inhibitor treatment (DISCOVER-1): a double-blind, randomised, placebo-controlled phase 3 trial. Lancet 2020;395:1115–25. 10.1016/S0140-6736(20)30265-8
    1. Ritchlin CT, Helliwell PS, Boehncke W-H, et al. . Guselkumab, an inhibitor of the IL-23p19 subunit, provides sustained improvement in signs and symptoms of active psoriatic arthritis: 1 year results of a phase III randomised study of patients who were biologic-naïve or TNFα inhibitor-experienced. RMD Open 2021;7:e001457. 10.1136/rmdopen-2020-001457
    1. Mease PJ, Rahman P, Gottlieb AB, et al. . Guselkumab in biologic-naive patients with active psoriatic arthritis (DISCOVER-2): a double-blind, randomised, placebo-controlled phase 3 trial. Lancet 2020;395:1126–36. 10.1016/S0140-6736(20)30263-4
    1. McInnes IB, Rahman P, Gottlieb AB, et al. . Efficacy and safety of guselkumab, an interleukin-23p19-specific monoclonal antibody, through one year in biologic-naive patients with psoriatic arthritis. Arthritis Rheumatol 2021;73:604–16. 10.1002/art.41553
    1. McInnes IB, Rahman P, Gottlieb AB, et al. . Long-Term efficacy and safety of guselkumab, a monoclonal antibody specific to the P19 subunit of interleukin-23, through 2 years: results from a phase 3, randomized, double-blind, placebo-controlled study conducted in biologic-naïve patients with active psoriatic arthritis. Arthritis Rheumatol 2021;74:475–85. 10.1002/art.42010
    1. Reich K, Gordon KB, Strober BE, et al. . Five-year maintenance of clinical response and health-related quality of life improvements in patients with moderate-to-severe psoriasis treated with guselkumab: results from VOYAGE 1 and VOYAGE 2. Br J Dermatol 2021;185:1146–59. 10.1111/bjd.20568
    1. Blauvelt A, Tsai T-F, Langley RG, et al. . Consistent safety profile with up to 5 years of continuous treatment with guselkumab: pooled analyses from the phase 3 VOYAGE 1 and VOYAGE 2 trials of patients with moderate-to-severe psoriasis. J Am Acad Dermatol 202110.1016/j.jaad.2021.11.004. [Epub ahead of print: 17 Nov 2021].
    1. Højgaard P, Glintborg B, Kristensen LE, et al. . The influence of obesity on response to tumour necrosis factor-α inhibitors in psoriatic arthritis: results from the DANBIO and ICEBIO registries. Rheumatology (Oxford) 2016;55:2191–9. 10.1093/rheumatology/kew326
    1. Giles JT, Ogdie A, Gomez Reino JJ, et al. . Impact of baseline body mass index on the efficacy and safety of tofacitinib in patients with psoriatic arthritis. RMD Open 2021;7:e001486. 10.1136/rmdopen-2020-001486
    1. Haddad A, Gazitt T, Feldhamer I, et al. . Treatment persistence of biologics among patients with psoriatic arthritis. Arthritis Res Ther 2021;23:44. 10.1186/s13075-021-02417-x
    1. Navarini L, Costa L, Tasso M, et al. . Retention rates and identification of factors associated with anti-TNFα, anti-IL17, and anti-IL12/23R agents discontinuation in psoriatic arthritis patients: results from a real-world clinical setting. Clin Rheumatol 2020;39:2663–70. 10.1007/s10067-020-05027-1
    1. Geale K, Lindberg I, Paulsson EC, et al. . Persistence of biologic treatments in psoriatic arthritis: a population-based study in Sweden. Rheumatol Adv Pract 2020;4:rkaa070. 10.1093/rap/rkaa070
    1. Glintborg B, Loft AG, Omerovic E, et al. . To switch or not to switch: results of a nationwide guideline of mandatory switching from originator to biosimilar etanercept. One-year treatment outcomes in 2061 patients with inflammatory arthritis from the DANBIO registry. Ann Rheum Dis 2019;78:192–200. 10.1136/annrheumdis-2018-213474
    1. Harrold LR, Stolshek BS, Rebello S, et al. . Impact of prior biologic use on persistence of treatment in patients with psoriatic arthritis enrolled in the US Corrona registry. Clin Rheumatol 2017;36:895–901. 10.1007/s10067-017-3593-x
    1. Merola JF, Lockshin B, Mody EA. Switching biologics in the treatment of psoriatic arthritis. Semin Arthritis Rheum 2017;47:29–37. 10.1016/j.semarthrit.2017.02.001
    1. Gordon KB, Blauvelt A, Foley P, et al. . Efficacy of guselkumab in subpopulations of patients with moderate-to-severe plaque psoriasis: a pooled analysis of the phase III VOYAGE 1 and VOYAGE 2 studies. Br J Dermatol 2018;178:132–9. 10.1111/bjd.16008
    1. Felson DT, Anderson JJ, Boers M, et al. . American College of rheumatology. Preliminary definition of improvement in rheumatoid arthritis. Arthritis Rheum 1995;38:727–35. 10.1002/art.1780380602
    1. Felson DT, Anderson JJ, Lange ML, et al. . Should improvement in rheumatoid arthritis clinical trials be defined as fifty percent or seventy percent improvement in core set measures, rather than twenty percent? Arthritis Rheum 1998;41:1564–70. 10.1002/1529-0131(199809)41:9<1564::AID-ART6>;2-M
    1. Feldman SR, Krueger GG. Psoriasis assessment tools in clinical trials. Ann Rheum Dis 2005;64 Suppl 2:ii65–8. 10.1136/ard.2004.031237
    1. Langley RGB, Feldman SR, Nyirady J, et al. . The 5-point Investigator's Global Assessment (IGA) scale: a modified tool for evaluating plaque psoriasis severity in clinical trials. J Dermatolog Treat 2015;26:23–31. 10.3109/09546634.2013.865009
    1. Chandran V, Bhella S, Schentag C, et al. . Functional Assessment of Chronic Illness Therapy-Fatigue scale is valid in patients with psoriatic arthritis. Ann Rheum Dis 2007;66:936–9. 10.1136/ard.2006.065763
    1. Bruce B, Fries JF. The Health Assessment Questionnaire (HAQ). Clin Exp Rheumatol 2005;23:S14–8.
    1. Helliwell PS, FitzGerald O, Fransen J, et al. . The development of candidate composite disease activity and responder indices for psoriatic arthritis (GRACE project). Ann Rheum Dis 2013;72:986–91. 10.1136/annrheumdis-2012-201341
    1. Coates LC, Fransen J, Helliwell PS. Defining minimal disease activity in psoriatic arthritis: a proposed objective target for treatment. Ann Rheum Dis 2010;69:48–53. 10.1136/ard.2008.102053
    1. Nas K, Capkin E, Dagli AZ, et al. . Gender specific differences in patients with psoriatic arthritis. Mod Rheumatol 2017;27:345–9. 10.1080/14397595.2016.1193105
    1. Queiro R, Tejón P, Coto P, et al. . Clinical differences between men and women with psoriatic arthritis: relevance of the analysis of genes and polymorphisms in the major histocompatibility complex region and of the age at onset of psoriasis. Clin Dev Immunol 2013;2013:482691.10.1155/2013/482691
    1. Mease PJ. Fibromyalgia, a missed comorbidity in spondyloarthritis: prevalence and impact on assessment and treatment. Curr Opin Rheumatol 2017;29:304–10. 10.1097/BOR.0000000000000388
    1. Braaten TJ, Zhang C, Presson AP, et al. . Gender differences in psoriatic arthritis with fatigue, pain, function, and work disability. J Psoriasis Psoriatic Arthritis 2019;4:192–7. 10.1177/2475530319870776
    1. Krajewska-Włodarczyk M, Owczarczyk-Saczonek A, Placek W. Prevalence and severity of fatigue in psoriasis and psoriatic arthritis. Adv Dermatol Allergol 2020;37:46–51. 10.5114/ada.2019.83629
    1. Tobin AM, Sadlier M, Collins P, et al. . Fatigue as a symptom in psoriasis and psoriatic arthritis: an observational study. Br J Dermatol 2017;176:827–8. 10.1111/bjd.15258
    1. Passia E, Vis M, Coates LC, et al. . Sex-specific differences and how to handle them in early psoriatic arthritis. Arthritis Res Ther 2022;24:22. 10.1186/s13075-021-02680-y
    1. Cañete JD, Tasende JAP, Laserna FJR, et al. . The impact of comorbidity on patient-reported outcomes in psoriatic arthritis: a systematic literature review. Rheumatol Ther 2020;7:237–57. 10.1007/s40744-020-00202-x
    1. Gudu T, Gossec L. Quality of life in psoriatic arthritis. Expert Rev Clin Immunol 2018;14:405–17. 10.1080/1744666X.2018.1468252
    1. Singh S, Facciorusso A, Singh AG, et al. . Obesity and response to anti-tumor necrosis factor-α agents in patients with select immune-mediated inflammatory diseases: a systematic review and meta-analysis. PLoS One 2018;13:e0195123. 10.1371/journal.pone.0195123
    1. di Minno MND, Peluso R, Iervolino S, et al. . Obesity and the prediction of minimal disease activity: a prospective study in psoriatic arthritis. Arthritis Care Res (Hoboken) 2013;65:141–7. 10.1002/acr.21711
    1. Pantano I, Iacono D, Favalli EG, et al. . Secukinumab efficacy in patients with PSA is not dependent on patients' body mass index. Ann Rheum Dis 2022;81:e42. 10.1136/annrheumdis-2020-217251
    1. Pirro F, Caldarola G, Chiricozzi A, et al. . Impact of body mass index on the efficacy of biological therapies in patients with psoriasis: a real-world study. Clin Drug Investig 2021;41:917–25. 10.1007/s40261-021-01080-z
    1. McInnes IB, Chakravarty SD, Apaolaza I, et al. . Efficacy of ustekinumab in biologic-naïve patients with psoriatic arthritis by prior treatment exposure and disease duration: data from PSUMMIT 1 and PSUMMIT 2. RMD Open 2019;5:e000990. 10.1136/rmdopen-2019-000990
    1. Husted JA, Tom BD, Farewell VT, et al. . A longitudinal study of the effect of disease activity and clinical damage on physical function over the course of psoriatic arthritis: does the effect change over time? Arthritis Rheum 2007;56:840–9. 10.1002/art.22443
    1. de Vlam K, Merola JF, Birt JA, et al. . Skin involvement in psoriatic arthritis worsens overall disease activity, patient-reported outcomes, and increases healthcare resource utilization: an observational, cross-sectional study. Rheumatol Ther 2018;5:423–36. 10.1007/s40744-018-0120-8
    1. Strober B, Menter A, Leonardi C, et al. . Efficacy of risankizumab in patients with moderate-to-severe plaque psoriasis by baseline demographics, disease characteristics and prior biologic therapy: an integrated analysis of the phase III UltIMMa-1 and UltIMMa-2 studies. J Eur Acad Dermatol Venereol 2020;34:2830–8. 10.1111/jdv.16521
    1. Glintborg B, Østergaard M, Dreyer L, et al. . Treatment response, drug survival, and predictors thereof in 764 patients with psoriatic arthritis treated with anti-tumor necrosis factor α therapy: results from the nationwide Danish DANBIO registry. Arthritis Rheum 2011;63:382–90. 10.1002/art.30117
    1. Rahman P, Ritchlin CT, Helliwell PS, et al. . Pooled safety results through 1 year of 2 phase III trials of guselkumab in patients with psoriatic arthritis. J Rheumatol 2021;48:1815–23. 10.3899/jrheum.201532
    1. Coates LC, Gossec L, Theander E, et al. . Efficacy and safety of guselkumab in patients with active psoriatic arthritis who are inadequate responders to tumour necrosis factor inhibitors: results through one year of a phase IIIB, randomised, controlled study (COSMOS). Ann Rheum Dis 2022;81:359–69. 10.1136/annrheumdis-2021-220991

Source: PubMed

3
Předplatit