Baricitinib in patients with inadequate response or intolerance to conventional synthetic DMARDs: results from the RA-BUILD study

Maxime Dougados, Désirée van der Heijde, Ying-Chou Chen, Maria Greenwald, Edit Drescher, Jiajun Liu, Scott Beattie, Sarah Witt, Inmaculada de la Torre, Carol Gaich, Terence Rooney, Douglas Schlichting, Stephanie de Bono, Paul Emery, Maxime Dougados, Désirée van der Heijde, Ying-Chou Chen, Maria Greenwald, Edit Drescher, Jiajun Liu, Scott Beattie, Sarah Witt, Inmaculada de la Torre, Carol Gaich, Terence Rooney, Douglas Schlichting, Stephanie de Bono, Paul Emery

Abstract

Background: Baricitinib is an oral, reversible, selective Janus kinase 1 and 2 inhibitor.

Methods: In this phase III, double-blind 24-week study, 684 biologic disease-modifying antirheumatic drug (DMARD)-naïve patients with rheumatoid arthritis and inadequate response or intolerance to ≥1 conventional synthetic DMARDs were randomly assigned 1:1:1 to placebo or baricitinib (2 or 4 mg) once daily, stratified by region and the presence of joint erosions. Endpoint measures included American College of Rheumatology 20% response (ACR20, primary endpoint), Disease Activity Score (DAS28) and Simplified Disease Activity Index (SDAI) score ≤3.3.

Results: More patients achieved ACR20 response at week 12 with baricitinib 4 mg than with placebo (62% vs 39%, p≤0.001). Compared with placebo, statistically significant improvements in DAS28, SDAI remission, Health Assessment Questionnaire-Disability Index, morning joint stiffness, worst joint pain and worst tiredness were observed. In a supportive analysis, radiographic progression of structural joint damage at week 24 was reduced with baricitinib versus placebo. Rates of adverse events during the treatment period and serious adverse events (SAEs), including serious infections, were similar among groups (SAEs: 5% for baricitinib 4 mg and placebo). One patient had an adverse event of tuberculosis (baricitinib 4 mg); one patient had an adverse event of non-melanoma skin cancer (baricitinib 4 mg). Two deaths and three major adverse cardiovascular events occurred (placebo). Baricitinib was associated with a decrease in neutrophils and increases in low-density and high-density lipoprotein.

Conclusions: In patients with rheumatoid arthritis and an inadequate response or intolerance to conventional synthetic DMARDs, baricitinib was associated with clinical improvement and inhibition of progression of radiographic joint damage.

Trial registration number: NCT01721057; Results.

Keywords: DMARDs (synthetic); Rheumatoid Arthritis; Treatment.

Conflict of interest statement

MD has received grant/research support or consulting support from AbbVie, Bristol Myers Squibb, Eli Lilly and Company, Novartis, Pfizer, Roche, Sanofi and UCB. PE has received grant/research support or consulting support from Abbott, AbbVie, Bristol Myers Squibb, Eli Lilly and Company, MSD, Novartis, Pfizer, Roche, Samsung, Takeda and UCB. DvdH has received grant/research support and/or consulting fees from AbbVie, Amgen, AstraZeneca, Augurex, Bristol Myers Squibb, Boehringer Ingelheim, Celgene, Centocor, Chugai, Covagen, Daiichi, Eli Lilly and Company, Galapagos, Glaxo-Smith Kline, Janssen Biologics, Merck, Novartis, Novo-Nordisk, Otsuka, Pfizer, Roche, Sanofi, UCB and Vertex and is director of Imaging Rheumatology bv. Y-CC has received speakers bureau fees and/or grant research support from AbbVie, Bristol Myers Squibb, Eli Lilly and Company and Pfizer. MG received research support from Eli Lilly and Company. JL, SB, SW, IdlT, CG, TR, DS and SdB are employees of Eli Lilly and Company and may own stock or stock options in Eli Lilly and Company.

Published by the BMJ Publishing Group Limited. For permission to use (where not already granted under a licence) please go to http://www.bmj.com/company/products-services/rights-and-licensing/.

Figures

Figure 1
Figure 1
Patient disposition through 24 weeks. Patients who were rescued or discontinued from the study or study treatment were defined as non-responders or had their last observations before rescue or discontinuation used for analyses of subsequent time points for efficacy endpoints. Nineteen patients (8%) had baseline estimated glomerular filtration rate ≥40 and 2 and therefore received baricitinib 2 mg (despite randomisation to and analysis by assigned treatment arm of baricitinib 4 mg). LTE, long-term extension; QD, once daily.
Figure 2
Figure 2
Primary and secondary efficacy analyses. The percentage of patients achieving American College of Rheumatology 20% response (ACR20) is shown in (A). The vertical line at 12 weeks indicates the primary efficacy time point. The least squares mean (LSM) change from baseline in Disease Activity Score for 28 joint counts C-reactive protein (DAS28-CRP) is shown in (B). Data reported as modified last observation carried forward (mLOCF), a form of LOCF modified to use the last observation prior to rescue or discontinuation. (C) shows the LSM change from baseline in Health Assessment Questionnaire-Disability Index (HAQ-DI) (mLOCF) with scores ranging from 0 to 3 (higher scores indicate greater disability). Analyses of change from baseline in DAS28-CRP (mBOCF) and HAQ-DI (mBOCF) at week 12 were included in the gatekeeping strategy. The percentage of patients with Simplified Disease Activity Index (SDAI) ≤3.3 at weeks 12 and 24 is shown in (D). (E–H) show the weekly diary scores for patient-reported outcomes. (E) shows the median duration of morning joint stiffness (MJS) at time points through week 12. The LSM for severity of MJS (numeric rating scale (NRS)) is shown in (F), worst tiredness (NRS) in (G) and worst joint pain (NRS) in (H). Patients recorded these measures in an electronic daily diary. MJS duration was truncated at a maximum value of 720 min. MJS severity: 0–10 NRS; 0=no joint stiffness, 10=joint stiffness as bad as you can imagine. Worst tiredness: 0–10 NRS; 0=no tiredness, 10=as bad as you can imagine. Worst joint pain: 0–10 NRS; 0=no pain, 10=pain as bad as you can imagine. *p≤0.05, **p≤0.01, ***p≤0.001 versus placebo. †For comparisons between baricitinib 4 mg versus placebo and baricitinib 2 mg versus placebo for the gated endpoints that are statistically significant based on the gatekeeping strategy with familywise error rate strongly controlled at α=0.05 for multiple comparisons.
Figure 3
Figure 3
Inhibition of radiographic progression of structural joint damage at week 24. The least squares mean (LSM) change from baseline in structural joint damage evaluated using modified Total Sharp Score (mTSS), joint space narrowing and erosion score is shown in (A). (B) shows the change from baseline in structural joint damage evaluated using the cumulative percentile change in mTSS. SDC (smallest detectable change)=1.2 units. *p≤0.05, **p≤0.01, ***p≤0.001 versus placebo.

References

    1. Smolen JS, Breedveld FC, Burmester GR, et al. . Treating rheumatoid arthritis to target: 2014 update of the recommendations of an international task force. Ann Rheum Dis 2016;75:3–15. 10.1136/annrheumdis-2015-207524
    1. Smolen JS, van der Heijde D, Machold KP, et al. . Proposal for a new nomenclature of disease-modifying antirheumatic drugs. Ann Rheum Dis 2014;73:3–5. 10.1136/annrheumdis-2013-204317
    1. O'Shea JJ, Kontzias A, Yamaoka K, et al. . Janus kinase inhibitors in autoimmune diseases. Ann Rheum Dis 2013;72:ii111–15. 10.1136/annrheumdis-2012-202576
    1. Keystone EC, Taylor PC, Drescher E, et al. . Safety and efficacy of baricitinib at 24 weeks in patients with rheumatoid arthritis who have had an inadequate response to methotrexate. Ann Rheum Dis 2015;74:333–40. 10.1136/annrheumdis-2014-206478
    1. Tanaka Y, Emoto K, Tsujimoto M, et al. . 12-week results of a phase 2b dose-ranging study of baricitinib, an oral JAK1/JAK2 inhibitor in Japanese patients with rheumatoid arthritis on background methotrexate therapy. Arthritis Rheum 2013;65:S765.
    1. Eli Lilly and Company. A Study in Participants with Moderate to Severe Rheumatoid Arthritis (RA-BEGIN). Bethesda, MD: National Library of Medicine (US), 2000. (cited 23 May 2016). NLM identifier: NCT01711359
    1. Eli Lilly and Company. A Study in Moderate to Severe Rheumatoid Arthritis (RA-BEAM). Bethesda, MD: National Library of Medicine (US), 2000. (cited 23 May 2016). NLM identifier: NCT01710358
    1. Eli Lilly and Company. A Study in Moderate to Severe Rheumatoid Arthritis Participants (RA-BUILD). Bethesda, MD: National Library of Medicine (US), 2000. (cited 23 May 2016). NLM identifier: NCT01721057
    1. Eli Lilly and Company. A Moderate to Severe Rheumatoid Arthritis Study (RA-BEACON). Bethesda, MD: National Library of Medicine (US), 2000. (cited 23 May 2016). NLM identifier: NCT01721044
    1. Genovese MC, Kremer J, Zamani O, et al. . Baricitinib in Patients with Refractory Rheumatoid Arthritis. N Engl J Med 2016;374:1243–52. 10.1056/NEJMoa1507247
    1. Eli Lilly and Company. An Extension Study in Participants with Moderate to Severe Rheumatoid Arthritis (RA-BEYOND). . Bethesda, MD: National Library of Medicine (US), 2000. (cited 23 May 2016). NLM identifier: NCT01885078
    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. Fries JF, Spitz P, Kraines RG, et al. . Measurement of patient outcome in arthritis. Arthritis Rheum 1980;23:137–45. 10.1002/art.1780230202
    1. Fries JF, Spitz PW, Young DY. The dimensions of health outcomes: the health assessment questionnaire, disability and pain scales. J Rheumatol 1982;9:789–93.
    1. Smolen JS, Breedveld FC, Schiff MH, et al. . A simplified disease activity index for rheumatoid arthritis for use in clinical practice. Rheumatology (Oxford). 2003;42:244–57.
    1. Aletaha D, Smolen J. The Simplified Disease Activity Index (SDAI) and the Clinical Disease Activity Index (CDAI): a review of their usefulness and validity in rheumatoid arthritis. Clin Exp Rheumatol 2005;23(Suppl 39): S100–8.
    1. Aletaha D, Nell VP, Stamm T, et al. . Acute phase reactants add little to composite disease activity indices for rheumatoid arthritis: validation of a clinical activity score. Arthritis Res Ther 2005;7:R796–806. 10.1186/ar1740
    1. Prevoo ML, van't Hof MA, Kuper HH, et al. . Modified disease activity scores that include twenty-eight-joint counts. Development and validation in a prospective longitudinal study of patients with rheumatoid arthritis. Arthritis Rheum. 1995;38:44–8.
    1. Disease Activity Score in rheumatoid arthritis. Nijmegen, the Netherlands: University Medical Center; (accessed 28 Apr 2016).
    1. van der Heijde D. How to read radiographs according to the Sharp/van der Heijde method. J Rheumatol 2000;27:261–3.
    1. van der Heijde D. Assessment of radiographs in longitudinal observational studies. J Rheumatol 2004;31(Suppl 69):46–7.
    1. Zhang Y, Warren MS, Zhang X, et al. . Impact on creatinine renal clearance by the interplay of multiple renal transporters: a case study with INCB039110. Drug Metab Dispos 2015;43:485–9. 10.1124/dmd.114.060673
    1. Papp KA, Menter MA, Abe M, et al. . Tofacitinib, an oral Janus kinase inhibitor, for the treatment of chronic plaque psoriasis: results from two randomized, placebo-controlled, phase III trials. Br J Dermatol 2015;173:949–61. 10.1111/bjd.14018
    1. Takeuchi T, Tanaka Y, Iwasaki M, et al. . Efficacy and safety of the oral Janus kinase inhibitor peficitinib (ASP015K) monotherapy in patients with moderate to severe rheumatoid arthritis in Japan: a 12-week, randomized, double-blind, placebo-controlled phase IIb study. Ann Rheum Dis 2016;75:1057–64. 10.1136/annrheumdis-2015-208279
    1. Genovese MC, Smolen JS, Weinblatt ME, et al. . A randomized phase 2b study of ABT-494, a selective JAK1 inhibitor in patients with rheumatoid arthritis and an inadequate response to methotrexate. Arthritis Rheumatol 2016. 10.1002/art.3980810.1002/art.39808
    1. Skoda RC. Less Jak2 makes more platelets. Blood 2014;124:2168–9. 10.1182/blood-2014-08-596361
    1. Yamane A, Nakamura T, Suzuki H, et al. . Interferon-alpha 2b-induced thrombocytopenia is caused by inhibition of platelet production but not proliferation and endomitosis in human megakaryocytes. Blood 2008;112:542–50. 10.1182/blood-2007-12-125906
    1. Felson DT, LaValley MP. The ACR20 and defining a threshold for response in rheumatic diseases: too much of a good thing. Arthritis Res Ther 2014;16:101 10.1186/ar4428
    1. Westhovens R, Alten R, Pavlova D, et al. . Filgotinib (GLPG0634), an oral JAK1 selective inhibitor is effective in combination with methotrexate in patients with active rheumatoid arthritis: results from a phase 2B dose ranging study [abstract]. Arthritis Rheumatol 2015;67(Suppl 10):1366-7. Abstract number: 1048.
    1. Kremer JM, Emery P, Camp HS, et al. . A phase 2b study of ABT-494, a selective JAK1 inhibitor, in patients with rheumatoid arthritis and an inadequate response to anti-TNF therapy. Arthritis Res Ther 2016. 10.1002/art.39801 10.1002/art.39801http://
    1. Huizinga TW, Fleischmann RM, Jasson M, et al. . Sarilumab, a fully human monoclonal antibody against IL-6Rα in patients with rheumatoid arthritis and an inadequate response to methotrexate: efficacy and safety results from the randomized SARIL-RA-MOBILITY Part A trial. Ann Rheum Dis 2014;73:1626–34. 10.1136/annrheumdis-2013-204405

Source: PubMed

3
Abonneren