Effects of the anti-RANKL antibody denosumab on joint structural damage in patients with rheumatoid arthritis treated with conventional synthetic disease-modifying antirheumatic drugs (DESIRABLE study): a randomised, double-blind, placebo-controlled phase 3 trial

Tsutomu Takeuchi, Yoshiya Tanaka, Satoshi Soen, Hisashi Yamanaka, Toshiyuki Yoneda, Sakae Tanaka, Takaya Nitta, Naoki Okubo, Harry K Genant, Désirée van der Heijde, Tsutomu Takeuchi, Yoshiya Tanaka, Satoshi Soen, Hisashi Yamanaka, Toshiyuki Yoneda, Sakae Tanaka, Takaya Nitta, Naoki Okubo, Harry K Genant, Désirée van der Heijde

Abstract

Objective: To evaluate the efficacy of denosumab in suppressing joint destruction when added to conventional synthetic disease-modifying antirheumatic drug (csDMARD) therapy in patients with rheumatoid arthritis (RA).

Methods: This was a multi-centre, randomised, double-blind, parallel-group, placebo-controlled phase 3 study in Japan. Patients with RA aged ≥20 years receiving csDMARDs were randomly assigned (1:1:1) to denosumab 60 mg every 3 months (Q3M), denosumab 60 mg every 6 months (Q6M) or placebo. The change in the modified total Sharp score (mTSS) and effect on bone mineral density (BMD) at 12 months was evaluated.

Results: In total, 654 patients received the trial drugs. Denosumab groups showed significantly less progression of joint destruction. The mean changes in the mTSS at 12 months were 1.49 (95% CI 0.99 to 1.99) in the placebo group, 0.99 (95% CI 0.49 to 1.49) in the Q6M group (p=0.0235) and 0.72 (95% CI 0.41 to 1.03) in the Q3M group (p=0.0055). The mean changes in bone erosion score were 0.98 (95% CI 0.65 to 1.31) in the placebo group, 0.51 (95% CI 0.22 to 0.80) in the Q6M group (p=0.0104) and 0.22 (95% CI 0.09 to 0.34) in the Q3M group (p=0.0001). No significant between-group difference was observed in the joint space narrowing score. The per cent change in lumbar spine (L1-L4) BMD in the placebo, Q6M and Q3M groups were -1.03%, 3.99% (p<0.0001) and 4.88% (p<0.0001). No major differences were observed among safety profiles.

Conclusions: Denosumab inhibits the progression of joint destruction, increases BMD and is well tolerated in patients with RA taking csDMARD.

Keywords: denosumab; erosion; joint destruction; rheumatoid arthritis.

Conflict of interest statement

Competing interests: TT has received research grants from AbbVie, Asahi Kasei, Astellas, AYUMI, Chugai, Daiichi Sankyo, Eisai, Mitsubishi Tanabe, Nippon Kayaku, Novartis, Pfizer and Takeda and has received personal fees from AbbVie, Astellas, Astra Zeneca, Bristol-Myers Squibb, Chugai, Daiichi Sankyo, Eisai, Eli Lilly, GlaxoSmithKline, Janssen, Mitsubishi Tanabe, Nippon Kayaku, Novartis, Pfizer, Sanofi, Taiho, Taisho Toyama, Takeda, Teijin and UCB. YT has received research grants from AbbVie, Astellas, Chugai, Bristol-Myers Squibb, Daiichi Sankyo, Eisai, Kyowa Hakko Kirin, Mitsubishi Tanabe, MSD, Ono, Pfizer and Takeda and has received personal fees from Astellas, Bristol-Myers Squibb, Chugai, Daiichi Sankyo, Eli Lilly, Janssen, Mitsubishi Tanabe, Pfizer, Sanofi, UCB and YL Biologics. SS has received grant/research support from Chugai and Daiichi Sankyo and has received personal fees from Asahi-Kasei Pharma, Astellas, MSD, Chugai, Daiichi Sankyo, Eli Lilly, Mitsubishi-Tanabe, Pfizer, Takeda and Teijin. HY has received research grants from AbbVie, Astellas, AYUMI, BMS, Chugai, Daiichi Sankyo, Eisai, Kaken, Mitsubishi Tanabe, MSD, Nippon Shinyaku, Ono, Pfizer, Takeda, Teijin, Torii and UCB and has received consulting fees from Astellas, BMS, Chugai, Daiichi Sankyo, Mitsubishi Tanabe, Nippon Kayaku, Pfizer, Takeda, Teijin and YL Biologics. TY has received Grants-in-Aid for Scientific Research from the Ministry of Education, Culture, Sports, Science and Technology of Japan (MEXT #17H04377) and has received consulting fees from Daiichi Sankyo. ST has acted as a consultant for AbbVie, Asahi Kasei Pharma, Amgen, Astellas, Daiichi Sankyo, Eli Lilly, MSD, Ono and Teijin Pharma. TN is an employee of Daiichi Sankyo. NO is a shareholder and employee of Daiichi Sankyo. HKG has received consulting fees from Amgen, Agnovos, Bioclinica, Biomarin, Clementia, Daiichi Sanyo, Eli Lilly, Janssen, Medimmune, Merck, Novartis, Pfizer, Regeneron, Servier and Takeda. DvdH has received consulting fees from AbbVie, Amgen, Astellas, AstraZeneca, BMS, Boehringer Ingelheim, Celgene, Daiichi Sankyo, Eli Lilly, Galapagos, Gilead, GlaxoSmithKline, Janssen, Merck, Novartis, Pfizer, Regeneron, Roche, Sanofi, Takeda and UCB and is the director of Imaging Rheumatology BV.

© Author(s) (or their employer(s)) 2019. Re-use permitted under CC BY-NC. No commercial re-use. See rights and permissions. Published by BMJ.

Figures

Figure 1
Figure 1
Trial profile. *As one patient assigned to denosumab Q6M was administered placebo by mistake, the patient was included in the placebo group for safety analysis. †No mTSS measurements available at baseline or after the first administration of treatment. §207 (per-protocol set). ¶202 (per-protocol set). ǂ200 (per-protocol set). mTSS, modified total Sharp score; PRT, protocol; Q3M, every 3 months; Q6M, every 6 months.
Figure 2
Figure 2
Mean changes from the baseline in the radiographic scores by the van der Heijde-modified Sharp method. (A) Modified total Sharp score, (B) modified Sharp erosion score and (C) modified Sharp joint space narrowing score. Missing values were imputed using linear extrapolation/interpolation. Mean and 95% CIs are presented. P values were calculated by two-sided van Elteren stratified rank test adjusting for baseline use of glucocorticoid. BL, baseline; n, number of patients who received ≥1 dose of investigational product and had a baseline and at least one postbaseline measurement of the radiograph score; Q3M, every 3 months; Q6M, every 6 months.
Figure 3
Figure 3
Cumulative probability plots of changes from the baseline in the radiographic score at 12 months. (A) Modified total Sharp score, (B) modified Sharp erosion score and (C) modified Sharp joint space narrowing score. n, number of patients who received ≥1 dose of investigational product and had a baseline and at least one postbaseline measurement of the radiograph score; Q3M, every 3 months; Q6M, every 6 months.
Figure 4
Figure 4
Per cent change in lumbar spinal bone mineral density (BMD) at 12 months from baseline in all patients (A), patients stratified by baseline use of glucocorticoid (B) and patients stratified by osteoporosis status (C). Data are for full analysis set (observed data). Coloured bars show least square mean values. P values are calculated using the analysis of covariance model after adjusting for treatment, baseline value, machine type, baseline value-by-machine type interaction and baseline use of glucocorticoid. Q3M, every 3 months; Q6M, every 6 months.

References

    1. Smolen JS, Aletaha D, McInnes IB. Rheumatoid arthritis. Lancet 2016;388:2023–38. 10.1016/S0140-6736(16)30173-8
    1. McInnes IB, Schett G. Pathogenetic insights from the treatment of rheumatoid arthritis. Lancet 2017;389:2328–37. 10.1016/S0140-6736(17)31472-1
    1. Heinlen L, Humphrey MB. Skeletal complications of rheumatoid arthritis. Osteoporos Int 2017;28:2801–12. 10.1007/s00198-017-4170-5
    1. Gough AK, Lilley J, Eyre S, et al. . Generalised bone loss in patients with early rheumatoid arthritis. Lancet 1994;344:23–7. 10.1016/S0140-6736(94)91049-9
    1. Yeo L, Lom H, Juarez M, et al. . Expression of FcRL4 defines a pro-inflammatory, RANKL-producing B cell subset in rheumatoid arthritis. Ann Rheum Dis 2015;74:928–35. 10.1136/annrheumdis-2013-204116
    1. Burmester GR, Pope JE. Novel treatment strategies in rheumatoid arthritis. Lancet 2017;389:2338–48. 10.1016/S0140-6736(17)31491-5
    1. Molenaar ETH, Voskuyl AE, Dinant HJ, et al. . Progression of radiologic damage in patients with rheumatoid arthritis in clinical remission. Arthritis Rheum 2004;50:36–42. 10.1002/art.11481
    1. Canalis E, Mazziotti G, Giustina A, et al. . Glucocorticoid-induced osteoporosis: pathophysiology and therapy. Osteoporos Int 2007;18:1319–28. 10.1007/s00198-007-0394-0
    1. Yasuda H, Shima N, Nakagawa N, et al. . Osteoclast differentiation factor is a ligand for osteoprotegerin/osteoclastogenesis-inhibitory factor and is identical to TRANCE/RANKL. Proc Natl Acad Sci U S A 1998;95:3597–602. 10.1073/pnas.95.7.3597
    1. Kostenuik PJ, Nguyen HQ, McCabe J, et al. . Denosumab, a fully human monoclonal antibody to RANKL, inhibits bone resorption and increases BMD in knock-in mice that express chimeric (murine/human) RANKL. J Bone Miner Res 2009;24:182–95. 10.1359/jbmr.081112
    1. Cohen SB, Dore RK, Lane NE, et al. . Denosumab treatment effects on structural damage, bone mineral density, and bone turnover in rheumatoid arthritis: a twelve-month, multicenter, randomized, double-blind, placebo-controlled, phase II clinical trial. Arthritis Rheum 2008;58:1299–309. 10.1002/art.23417
    1. Takeuchi T, Tanaka Y, Ishiguro N, et al. . Effect of denosumab on Japanese patients with rheumatoid arthritis: a dose-response study of AMG 162 (Denosumab) in patients with RheumatoId arthritis on methotrexate to Validate inhibitory effect on bone Erosion (DRIVE)-a 12-month, multicentre, randomised, double-blind, placebo-controlled, phase II clinical trial. Ann Rheum Dis 2016;75:983–90. 10.1136/annrheumdis-2015-208052
    1. Arnett FC, Edworthy SM, Bloch DA, et al. . The American rheumatism association 1987 revised criteria for the classification of rheumatoid arthritis. Arthritis Rheum 1988;31:315–24. 10.1002/art.1780310302
    1. Aletaha D, Neogi T, Silman AJ, et al. . Rheumatoid arthritis classification criteria: an American College of Rheumatology/European League against rheumatism collaborative initiative. Arthritis Rheum 2010;2010:2569–81.
    1. Hochberg MC, Chang RW, Dwosh I, et al. . The American College of rheumatology 1991 revised criteria for the classification of global functional status in rheumatoid arthritis. Arthritis Rheum 1992;35:498–502. 10.1002/art.1780350502
    1. van der Heijde D. How to read radiographs according to the Sharp/van Der Heijde method. J Rheumatol 2000;27:261–3.
    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. 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. Prevoo ML, van Gestel AM, van T Hof MA, et al. . Remission in a prospective study of patients with rheumatoid arthritis. American rheumatism association preliminary remission criteria in relation to the disease activity score. Br J Rheumatol 1996;35:1101–5. 10.1093/rheumatology/35.11.1101
    1. van der Heijde D, Simon L, Smolen J, et al. . How to report radiographic data in randomized clinical trials in rheumatoid arthritis: guidelines from a roundtable discussion. Arthritis Rheum 2002;47:215–8. 10.1002/art.10181
    1. Wells GA, Tugwell P, Kraag GR, et al. . Minimum important difference between patients with rheumatoid arthritis: the patient's perspective. J Rheumatol 1993;20:557–60.
    1. Atsumi T, Yamamoto K, Takeuchi T, et al. . The first double-blind, randomised, parallel-group certolizumab pegol study in methotrexate-naive early rheumatoid arthritis patients with poor prognostic factors, C-OPERA, shows inhibition of radiographic progression. Ann Rheum Dis 2016;75:75–83. 10.1136/annrheumdis-2015-207511
    1. van der Kooi E, Klarenbeek NB, Güler-Yüksel M, et al. . A decrease in disease activity score (DAS) level is associated with a decrease in health assessment Questionnaire (HAQ) score, independent of follow-up duration, during 5 years of tightly controlled treatment: results from the best study. Ann Rheum Dis 2011;70:168–71. 10.1136/ard.2010.133132
    1. Drossaers-Bakker KW, de Buck M, van Zeben D, et al. . Long-term course and outcome of functional capacity in rheumatoid arthritis: the effect of disease activity and radiologic damage over time. Arthritis Rheum 1999;42:1854–60. 10.1002/1529-0131(199909)42:9&lt;1854::AID-ANR9&gt;;2-F
    1. Dore RK, Cohen SB, Lane NE, et al. . Effects of denosumab on bone mineral density and bone turnover in patients with rheumatoid arthritis receiving concurrent glucocorticoids or bisphosphonates. Ann Rheum Dis 2010;69:872–5. 10.1136/ard.2009.112920
    1. Breuil V, Euller-Ziegler L. Bisphosphonate therapy in rheumatoid arthritis. Joint Bone Spine 2006;73:349–54. 10.1016/j.jbspin.2005.10.019
    1. Hoes JN, Bultink IEM, Lems WF. Management of osteoporosis in rheumatoid arthritis patients. Expert Opin Pharmacother 2015;16:559–71. 10.1517/14656566.2015.997709
    1. Jarrett SJ, Conaghan PG, Sloan VS, et al. . Preliminary evidence for a structural benefit of the new bisphosphonate zoledronic acid in early rheumatoid arthritis. Arthritis Rheum 2006;54:1410–4. 10.1002/art.21824
    1. Yue J, Griffith JF, Xiao F, et al. . Repair of bone erosion in rheumatoid arthritis by denosumab: a high-resolution peripheral quantitative computed tomography study. Arthritis Care Res 2017;69:1156–63. 10.1002/acr.23133
    1. Miller PD, Pannacciulli N, Brown JP, et al. . Denosumab or zoledronic acid in postmenopausal women with osteoporosis previously treated with oral bisphosphonates. J Clin Endocrinol Metab 2016;101:3163–70. 10.1210/jc.2016-1801
    1. Baron R, Ferrari S, Russell RGG, et al. . Denosumab and bisphosphonates: different mechanisms of action and effects. Bone 2011;48:677–92. 10.1016/j.bone.2010.11.020
    1. Zerbini CAF, Clark P, Mendez-Sanchez L, et al. . Biologic therapies and bone loss in rheumatoid arthritis. Osteoporos Int 2017;28:429–46. 10.1007/s00198-016-3769-2
    1. Nakamura T, Matsumoto T, Sugimoto T, et al. . Dose-response study of denosumab on bone mineral density and bone turnover markers in Japanese postmenopausal women with osteoporosis. Osteoporos Int 2012;23:1131–40. 10.1007/s00198-011-1786-8
    1. Mahlich J, Sruamsiri R. Treatment patterns of rheumatoid arthritis in Japanese hospitals and predictors of the initiation of biologic agents. Curr Med Res Opin 2017;33:101–7. 10.1080/03007995.2016.1239191

Source: PubMed

3
購読する