ASP5094, a humanized monoclonal antibody against integrin alpha-9, did not show efficacy in patients with rheumatoid arthritis refractory to methotrexate: results from a phase 2a, randomized, double-blind, placebo-controlled trial

Tsutomu Takeuchi, Yoshiya Tanaka, Jay Erdman, Yuichiro Kaneko, Masako Saito, Chieri Higashitani, Ronald Smulders, Christopher Lademacher, Tsutomu Takeuchi, Yoshiya Tanaka, Jay Erdman, Yuichiro Kaneko, Masako Saito, Chieri Higashitani, Ronald Smulders, Christopher Lademacher

Abstract

Background: Rheumatoid arthritis (RA) is a chronic, debilitating autoimmune condition characterized by joint synovial inflammation. Current treatments include methotrexate (MTX), biologic agents, and Janus kinase (JAK) inhibitors. However, these agents are not efficacious in all patients and there are concerns regarding side effects and risk of infection as these treatments target immune-related pathways. Overexpression and activation of integrin alpha-9 (α9) on fibroblast-like synoviocytes are associated with RA disease onset and exacerbation. The humanized immunoglobulin G1 monoclonal antibody ASP5094 was designed to inhibit human α9 and is currently under investigation for the treatment of RA.

Methods: This phase 2a, multicenter, randomized, placebo-controlled, double-blind, parallel-group study (NCT03257852) evaluated the efficacy, safety, and biological activity of intravenous ASP5094 10 mg/kg in patients with moderate to severe RA that was refractory to MTX. Patients received ASP5094 or placebo every 4 weeks for a total of three administrations. Both treatment groups used concomitant MTX. The primary efficacy endpoint was the proportion of patients who responded per American College of Rheumatology 50% improvement using C-reactive protein (ACR50-CRP) after 12 weeks of treatment. Biological activity of ASP5094 was assessed via pharmacokinetics and pharmacodynamics of known downstream effectors of α9. Safety was also assessed.

Results: Sixty-six patients were enrolled and randomized to placebo (n = 33) or ASP5094 (n = 33). In the primary efficacy analysis, ACR50-CRP response rates were 6.3% and 18.2% at week 12 in the ASP5094 and placebo groups, respectively; a difference of - 11.9, which was not significant (2-sided P value = 0.258). No trends in ACR50 response rates were observed in subgroups based on demographics or baseline disease characteristics, and no significant differences between placebo and ASP5094 were identified in secondary efficacy or pharmacodynamic endpoints, despite achievement of target serum concentrations of ASP5094. Most treatment-emergent adverse events were mild to moderate in severity, and ASP5094 was considered safe and well tolerated overall.

Conclusion: Although no notable safety signals were observed in this study, ASP5094 was not efficacious in patients with moderate to severe RA with an inadequate response to MTX.

Trial registration: ClinicalTrials.gov, NCT03257852 . Registered on 22 Aug. 2017.

Keywords: Antibodies, Monoclonal, Humanized; Antirheumatic agents; Arthritis, Rheumatoid; Biological products; Extracellular matrix proteins; Integrin alpha-9, human; Integrins; Methotrexate; Synovial membrane; Synoviocytes.

Conflict of interest statement

T. Takeuchi reports grants from Astellas Pharma, Inc.; Chugai Pharmaceutical Co, Ltd.; Daiichi-Sankyo Co., Ltd.; Takeda Pharmaceutical Co., Ltd.; AbbVie GK; Asahikasei Pharma Corp.; Mitsubishi Tanabe Pharma Co.; Pfizer Japan Inc.; Eisai Co., Ltd.; AYUMI Pharmaceutical Corporation; Nipponkayaku Co. Ltd.; Novartis Pharma K.K.; and Shionogi & Co., LTD and personal fees from Astellas Pharma, Inc.; AbbVie GK; AYUMI Pharmaceutical Corporation; Eisai Co., Ltd.; Gilead Sciences, Inc.; GlaxoSmithKline K.K.; Sanofi K.K.; Taiho Pharmaceutical Co., Ltd.; Mitsubishi Tanabe Pharma Co.; Daichi Sankyo Co. Ltd.; Chugai Pharmaceutical Co., Ltd.; Taisho Pharmaceutical Co., Ltd.; Eli Lilly Japan K.K.; Novartis Pharma K.K.; Boehringer-Ingelheim Co., Ltd.; Nipponkayaku Co. Ltd.; Pfizer Japan Inc.; Bristol-Myers K.K.; Janssen Pharmaceutical K.K.; and UCB Japan Co. Ltd.

Y. Tanaka reports personal fees from Daiichi-Sankyo; Astellas Pharma, Inc.; Chugai; Eli Lilly; Pfizer; AbbVie; YL Biologics; Bristol-Myers; Takeda; Mitsubishi-Tanabe; Novartis; Eisai; Janssen; and Teijin and grants from Asahi-kasei, Mitsubishi-Tanabe, Chugai, Takeda, Sanofi, Bristol-Myers, UCB, Daiichi-Sankyo, Eisai, Ono.

C. Lademacher, J. Erdman, Y. Kaneko, M. Saito, C. Higashitani, and R. Smulders are employees of Astellas Pharma, Inc.

Figures

Fig. 1
Fig. 1
Study design
Fig. 2
Fig. 2
Patient disposition. aProvided informed consent
Fig. 3
Fig. 3
Change from baseline in a DAS28-CRP, b DAS28-ESR, c SDAI, and d CDAI scores over time. CDAI, Clinical Disease Activity Index; CRP, c-reactive protein; DAS28, Disease Activity Score in 28 joints; ESR, erythrocyte sedimentation rate; SDAI, Simplified Disease Activity Index

References

    1. Amaya-Amaya J, et al. Autoimmunity: from bench to bedside. Bogota: El Rosario University Press; 2013. Rheumatoid arthritis.
    1. Schett G, Gravallese E. Bone erosion in rheumatoid arthritis: mechanisms, diagnosis and treatment. Nat Rev Rheumatol. 2012;8(11):656–664. doi: 10.1038/nrrheum.2012.153.
    1. Scott DL, Wolfe F, Huizinga TW. Rheumatoid arthritis. Lancet. 2010;376(9746):1094–1108. doi: 10.1016/S0140-6736(10)60826-4.
    1. Haraoui B, et al. Treating rheumatoid arthritis to target: multinational recommendations assessment questionnaire. Ann Rheum Dis. 2011;70(11):1999–2002. doi: 10.1136/ard.2011.154179.
    1. Kameda H, et al. Etanercept (ETN) with methotrexate (MTX) is better than ETN monotherapy in patients with active rheumatoid arthritis despite MTX therapy: a randomized trial. Mod Rheumatol. 2010;20(6):531–538. doi: 10.3109/s10165-010-0324-4.
    1. Kaneko Y, et al. Comparison of adding tocilizumab to methotrexate with switching to tocilizumab in patients with rheumatoid arthritis with inadequate response to methotrexate: 52-week results from a prospective, randomised, controlled study (SURPRISE study) Ann Rheum Dis. 2016;75(11):1917–1923. doi: 10.1136/annrheumdis-2015-208426.
    1. Genovese MC, et al. VX-509 (decernotinib), an oral selective Janus kinase 3 inhibitor, in combination with methotrexate in patients with rheumatoid arthritis. Arthritis Rheumatol. 2015. 10.1002/art.39473 [Epub ahead of print].
    1. Singh JA, et al. 2015 American College of Rheumatology Guideline for the treatment of rheumatoid arthritis. Arthritis Rheumatol. 2016;68(1):1–26. doi: 10.1002/art.39480.
    1. Kaneko Y, Takeuchi T. Targeted antibody therapy and relevant novel biomarkers for precision medicine for rheumatoid arthritis. Int Immunol. 2017;29(11):511–517. doi: 10.1093/intimm/dxx055.
    1. Takeuchi T. Treatment of rheumatoid arthritis with biological agents — as a typical and common immune-mediated inflammatory disease. Proc Japan Acad Series B. 2017;93(8):600–608. doi: 10.2183/pjab.93.038.
    1. Saeki Y, et al. Current treatments of rheumatoid arthritis: from the ‘NinJa’ registry. Expert Rev Clin Immunol. 2012;8(5):455–465. doi: 10.1586/eci.12.35.
    1. Kaneko Y, Kondo H, Takeuchi T. American College of Rheumatology/European League Against Rheumatism remission criteria for rheumatoid arthritis maintain reliable performance when evaluated in 44 joints. J Rheumatol. 2013;40(8):1254–8. doi: 10.3899/jrheum.130166.
    1. SIMPONI ARIA (golimumab) [package insert]. Horsham: Janssen Biotech, Inc; 2017.
    1. XELJANZ (tofacitinib) [package insert]. Pfizer Inc: New York; 2018.
    1. RINVOQ (upadacitinib) [package insert]. Chicago: AbbVie Inc; 2019.
    1. ACTEMRA (tocilizumab) [package insert]. South San Francisco: Genentech, Inc; 2019.
    1. Tasaki S, et al. Multi-omics monitoring of drug response in rheumatoid arthritis in pursuit of molecular remission. Nat Commun. 2018;9(1):2755. doi: 10.1038/s41467-018-05044-4.
    1. Mor A, Abramson SB, Pillinger MH. The fibroblast-like synovial cell in rheumatoid arthritis: a key player in inflammation and joint destruction. Clin Immunol. 2005;115(2):118–128. doi: 10.1016/j.clim.2004.12.009.
    1. Neumann E, et al. Rheumatoid arthritis progression mediated by activated synovial fibroblasts. Trends Mol Med. 2010;16(10):458–468. doi: 10.1016/j.molmed.2010.07.004.
    1. Noss EH, Brenner MB. The role and therapeutic implications of fibroblast-like synoviocytes in inflammation and cartilage erosion in rheumatoid arthritis. Immunol Rev. 2008;223:252–270. doi: 10.1111/j.1600-065X.2008.00648.x.
    1. Koch AE, et al. Epithelial neutrophil activating peptide-78: a novel chemotactic cytokine for neutrophils in arthritis. J Clin Invest. 1994;94(3):1012–1018. doi: 10.1172/JCI117414.
    1. Leech MT, Morand EF. Fibroblasts and synovial immunity. Curr Opin Pharmacol. 2013;13(4):565–569. doi: 10.1016/j.coph.2013.04.001.
    1. Croft AP, et al. Distinct fibroblast subsets drive inflammation and damage in arthritis. Nature. 2019;570(7760):246–251. doi: 10.1038/s41586-019-1263-7.
    1. Wynn TA. Two types of fibroblast drive arthritis. Nature. 2019;570(7760):169–170. doi: 10.1038/d41586-019-01594-9.
    1. Zhang F, et al. Defining inflammatory cell states in rheumatoid arthritis joint synovial tissues by integrating single-cell transcriptomics and mass cytometry. Nat Immunol. 2019;20(7):928–942. doi: 10.1038/s41590-019-0378-1.
    1. Miranti CK, Brugge JS. Sensing the environment: a historical perspective on integrin signal transduction. Nat Cell Biol. 2002;4(4):E83–E90. doi: 10.1038/ncb0402-e83.
    1. Yokosaki Y, et al. The integrin alpha 9 beta 1 mediates cell attachment to a non-RGD site in the third fibronectin type III repeat of tenascin. J Biol Chem. 1994;269(43):26691–26696.
    1. Smith LL, et al. Osteopontin N-terminal domain contains a cryptic adhesive sequence recognized by alpha9beta1 integrin. J Biol Chem. 1996;271(45):28485–28491. doi: 10.1074/jbc.271.45.28485.
    1. Taooka Y, et al. The integrin alpha9beta1 mediates adhesion to activated endothelial cells and transendothelial neutrophil migration through interaction with vascular cell adhesion molecule-1. J Cell Biol. 1999;145(2):413–420. doi: 10.1083/jcb.145.2.413.
    1. Gupta SK, Vlahakis NE. Integrin alpha9beta1: unique signaling pathways reveal diverse biological roles. Cell Adhes Migr. 2010;4(2):194–198. doi: 10.4161/cam.4.2.10900.
    1. Hoye AM, et al. The newcomer in the integrin family: integrin alpha9 in biology and cancer. Adv Biol Regul. 2012;52(2):326–339. doi: 10.1016/j.jbior.2012.03.004.
    1. Asano T, et al. Alpha9beta1 integrin acts as a critical intrinsic regulator of human rheumatoid arthritis. Rheumatology (Oxford) 2014;53(3):415–424. doi: 10.1093/rheumatology/ket371.
    1. Emori T, et al. Constitutive activation of integrin alpha9 augments self-directed hyperplastic and proinflammatory properties of fibroblast-like synoviocytes of rheumatoid arthritis. J Immunol. 2017;199(10):3427–3436. doi: 10.4049/jimmunol.1700941.
    1. Kanayama M, et al. α9β1 integrin-mediated signaling serves as an intrinsic regulator of pathogenic Th17 cell generation. J Immunol. 2011;187(11):5851–5864. doi: 10.4049/jimmunol.1101524.
    1. Yamamoto N, et al. Essential role of the cryptic epitope SLAYGLR within osteopontin in a murine model of rheumatoid arthritis. J Clin Invest. 2003;112(2):181–188. doi: 10.1172/JCI17778.
    1. Uede T. Osteopontin, intrinsic tissue regulator of intractable inflammatory diseases. Pathol Int. 2011;61(5):265–280. doi: 10.1111/j.1440-1827.2011.02649.x.
    1. Sugahara S, Hanaoka K, Yamamoto N. Integrin, alpha9 subunit blockade suppresses collagen-induced arthritis with minimal systemic immunomodulation. Eur J Pharmacol. 2018;833:320–327. doi: 10.1016/j.ejphar.2018.06.021.
    1. Huang J, et al. Multiple dose safety, tolerability, pharmacokinetics, and pharmacodynamics of ASP5094, an anti-alpha9 integrin monoclonal antibody, in patients with rheumatoid arthritis on methotrexate [abstract]. Arthritis Rheumatol. 2018;70(suppl 10).
    1. Tanaka Y, et al. Efficacy and safety of Baricitinib in Japanese patients with active rheumatoid arthritis receiving background methotrexate therapy: a 12-week, double-blind, randomized placebo-controlled study. J Rheumatol. 2016;43(3):504–511. doi: 10.3899/jrheum.150613.
    1. Yamamoto K, et al. Efficacy and safety of certolizumab pegol plus methotrexate in Japanese rheumatoid arthritis patients with an inadequate response to methotrexate: the J-RAPID randomized, placebo-controlled trial. Mod Rheumatol. 2014;24(5):715–724. doi: 10.3109/14397595.2013.864224.
    1. Tanaka Y, et al. Golimumab in combination with methotrexate in Japanese patients with active rheumatoid arthritis: results of the GO-FORTH study. Ann Rheum Dis. 2012;71(6):817–824. doi: 10.1136/ard.2011.200317.
    1. Kanayama M, et al. Alpha9 integrin and its ligands constitute critical joint microenvironments for development of autoimmune arthritis. J Immunol. 2009;182(12):8015–8025. doi: 10.4049/jimmunol.0900725.
    1. Ohshima S, et al. Enhanced local production of osteopontin in rheumatoid joints. J Rheumatol. 2002;29(10):2061–2067.

Source: PubMed

3
購読する