Efficacy and safety of namilumab, a human monoclonal antibody against granulocyte-macrophage colony-stimulating factor (GM-CSF) ligand in patients with rheumatoid arthritis (RA) with either an inadequate response to background methotrexate therapy or an inadequate response or intolerance to an anti-TNF (tumour necrosis factor) biologic therapy: a randomized, controlled trial

Peter C Taylor, Didier Saurigny, Jiri Vencovsky, Tsutomu Takeuchi, Tadashi Nakamura, Galina Matsievskaia, Barbara Hunt, Thomas Wagner, Bernard Souberbielle, NEXUS Study Group, Peter C Taylor, Didier Saurigny, Jiri Vencovsky, Tsutomu Takeuchi, Tadashi Nakamura, Galina Matsievskaia, Barbara Hunt, Thomas Wagner, Bernard Souberbielle, NEXUS Study Group

Abstract

Background: Namilumab (AMG203), an immunoglobulin G1 monoclonal antibody that binds with high affinity to granulocyte-macrophage colony-stimulating factor (GM-CSF), was evaluated in a phase II randomized, double-blind, placebo-controlled study to investigate the efficacy and safety in patients with rheumatoid arthritis (RA) with an inadequate response to methotrexate (MTX-IR) or anti-tumour necrosis factor therapy (TNF-IR).

Methods: Subcutaneous namilumab (20, 80, or 150 mg) or placebo was administered at baseline and weeks 2, 6, and 10 in patients on stable background methotrexate therapy who were with MTX-IR or TNF-IR. Primary endpoint was mean change from baseline in the 28-joint Disease Activity Score, C-reactive protein version (DAS28-CRP) at week 12 comparing each of the three doses of namilumab to placebo. Safety and tolerability were assessed by adverse events (AEs) and pulmonary parameters. Results were analysed using the per-protocol population.

Results: One hundred eight patients from Europe and Japan (48.4 ± 12.02 years old; 77.8% female; mean DAS28-CRP 5.60-5.79; rheumatoid factor/anti-citrullinated protein antibodies + 75%) were randomized to placebo or namilumab 20, 80, or 150 mg (n = 27, 28, 25, and 28, respectively). Ninety-two were MTX-IR; 16 were TNF-IR. At week 12, a statistically significant difference in DAS28-CRP (p = 0.005) was seen for namilumab 150 mg versus placebo and separation was seen as early as week 2 for namilumab 150 mg (p < 0.05), with higher ACR50 and response rates versus placebo at week 12. A dose-response effect was observed across the DAS28-CRP endpoint with separation versus placebo evident from week 2. The most common treatment-emergent AEs were nasopharyngitis (18.5%, 17.9%, 4.0%, 14.3%), dyspnoea (0.0%, 3.6%, 8.0%, 10.7%), bronchitis (7.4%, 3.6%, 4.0%, 3.6%), and headache (3.7%, 3.6%, 12.0%, 0.0%) for placebo and 20, 80, or 150 mg of namilumab, respectively. No serious infections were observed. One serious AE (myocardial infarction) was observed with 150 mg of namilumab. There was no apparent dose relationship for AEs. A biomarker-based disease activity score showed a dose-dependent decrease at week 12.

Conclusions: This phase II study demonstrates the benefit of inhibiting macrophage activity targeting the GM-CSF for RA. The study met its primary endpoint with a clear dose-response effect. An acceptable tolerability profile was demonstrated over the 12-week study.

Trial registration: ClinicalTrials.gov, NEXUS; NCT02379091 , submitted November 28, 2014.

Keywords: GM-CSF; Namilumab; Rheumatoid arthritis.

Conflict of interest statement

Ethics approval and consent to participate

Institutional review boards or ethics committees at the participating investigational centres approved the study, which was conducted according to the principles set out in the Declaration of Helsinki, International Conference on Harmonisation Guidelines for Good Clinical Practice, and additional local regulations.

Consent for publication

Not applicable.

Competing interests

Dr. Wagner was an employee of Takeda Pharmaceuticals International GmbH. Both Bernard Souberbielle and Didier Saurigny were employees of Takeda at the time of the study. Barbara Hunt is an employee of Takeda International, Deerfield, IL, USA. The other authors declare that they have no competing interests.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Figures

Fig. 1
Fig. 1
Subject disposition. The primary analysis was based on 106 subjects (full analysis set population) and 88 subjects (per-protocol set population). SF screening failure
Fig. 2
Fig. 2
Analysis of change from baseline in DAS28-CRP score. Change from baseline in DAS28-CRP score by visit for the a per-protocol set [p < 0.05 are shown by asterisk; week 12, p = 0.022] and b full analysis set [p < 0.05 are shown by asterisk; week 6, p = 0.026; week 10, p = 0.031; week 12, p = 0.015]. DAS28-CRP 28-joint Disease Activity Score, C-reactive protein version
Fig. 3
Fig. 3
DAS28-CRP-adjusted mean change from baseline (per-protocol set). Significant p values compared to placebo are shown (p < 0.05 as one asterisk and p < 0.01 as two asterisks) [150 mg at week 2, p = 0.021; at week 6, p = 0.037; at week 10, p = 0.018; and at week 12, p = 0.005]. DAS28-CRP, 28-joint Disease Activity Score, C-reactive protein version; PBO placebo
Fig. 4
Fig. 4
ACR20, ACR50, and ACR70 at week 12 (per-protocol set). Significant p values compared to placebo are shown by an asterisk (ACR20 for 20 mg, p = 0.031, and ACR50 for 150 mg, p = 0.049). ACR American College of Rheumatology, ACR20 ≥ 20% improvement, ACR50 ≥ 50% improvement, ACR70 ≥ 70% improvement, PBO placebo
Fig. 5
Fig. 5
Percentage of ACR20/50/70 and DAS28-CRP remission (≤ 2.6) by study visit (PPS). ACR, American College of Rheumatology; ACR20, ≥ 20% improvement; ACR50, ≥ 50% improvement; ACR70, ≥ 70% improvement; DAS28-CRP, 28-joint Disease Activity Score, C-reactive protein version; PBO, placebo; PPS, per-protocol set. p value of less than 0.05 is shown by an asterisk
Fig. 6
Fig. 6
Mean change in MBDA from baseline. MBDA multibiomarker disease activity
Fig. 7
Fig. 7
Change in C1M from baseline. Values are mean ± SE. ns not significant. *Significant p = 0.0227

References

    1. Scott DL, Wolfe F, Huizinga TW. Rheumatoid arthritis. Lancet. 2010;376:1094–1108. doi: 10.1016/S0140-6736(10)60826-4.
    1. Kievit W, Fransen J, Oerlemans AJ, Kuper HH, van der Laar MA, de Rooij DJ, et al. The efficacy of anti-TNF in rheumatoid arthritis, a comparison between randomised controlled trials and clinical practice. Ann Rheum Dis. 2007;66:1473–1478. doi: 10.1136/ard.2007.072447.
    1. Hetland ML, Christensen IJ, Tarp U, Dreyer L, Hansen A, Hansen IT, et al. Direct comparison of treatment responses, remission rates, and drug adherence in patients with rheumatoid arthritis treated with adalimumab, etanercept, or infliximab: results from eight years of surveillance of clinical practice in the nationwide Danish DANBIO registry. Arthritis Rheum. 2010;62:22–32. doi: 10.1002/art.27227.
    1. Fleetwood AJ, Cook AD, Hamilton JA. Functions of granulocyte-macrophage colony-stimulating factor. Crit Rev Immunol. 2005;25:405–428. doi: 10.1615/CritRevImmunol.v25.i5.50.
    1. Hamilton JA. Colony-stimulating factors in inflammation and autoimmunity. Nat Rev Immunol. 2008;8:533–544. doi: 10.1038/nri2356.
    1. Griffin JD, Cannistra SA, Sullivan R, Demetri GD, Ernst TJ, Kanakura Y. The biology of GM-CSF: regulation of production and interaction with its receptor. Int J Cell Cloning. 1990;8(Suppl 1):35–44. doi: 10.1002/stem.5530080705.
    1. Gasson JC. Molecular physiology of granulocyte-macrophage colony-stimulating factor. Blood. 1991;77:1131–1145.
    1. Hamilton JA. GM-CSF as a target in inflammatory/autoimmune disease: current evidence and future therapeutic potential. Expert Rev Clin Immunol. 2015;11:457–465. doi: 10.1586/1744666X.2015.1024110.
    1. Hamilton JA, Anderson GP. GM-CSF biology. Growth Factors. 2004;22:225–231. doi: 10.1080/08977190412331279881.
    1. Zvaifler NJ, Firestein GS. Cytokines in chronic inflammatory synovitis. Scand J Rheumatol. 1988;17:203–210. doi: 10.3109/03009748809102970.
    1. Xu WD, Firestein GS, Taetle R, Kaushansky K, Zvaifler NJ. Cytokines in chronic inflammatory arthritis. II. Granulocyte-macrophage colony-stimulating factor in rheumatoid synovial effusions. J Clin Invest. 1989;83:876–882. doi: 10.1172/JCI113971.
    1. Alvaro-Gracia JM, Zvaifler NJ, Brown CB, Kaushansky K, Firestein GS. Cytokines in chronic inflammatory arthritis. VI. Analysis of the synovial cells involved in granulocyte-macrophage colony-stimulating factor production and gene expression in rheumatoid arthritis and its regulation by IL-1 and tumor necrosis factor-alpha. J Immunol. 1991;146:3365–3371.
    1. Fiehn C, Wermann M, Pezzutto A, Hufner M, Heilig B. Plasma GM-CSF concentrations in rheumatoid arthritis, systemic lupus erythematosus and spondyloarthropathy. Z Rheumatol. 1992;51:121–126.
    1. Ulfgren AK, Lindblad S, Klareskog L, Andersson J, Andersson U. Detection of cytokine producing cells in the synovial membrane from patients with rheumatoid arthritis. Ann Rheum Dis. 1995;54:654–661. doi: 10.1136/ard.54.8.654.
    1. Chu CQ, Field M, Allard S, Abney E, Feldmann M, Maini RN. Detection of cytokines at the cartilage/pannus junction in patients with rheumatoid arthritis: implications for the role of cytokines in cartilage destruction and repair. Br J Rheumatol. 1992;31:653–661. doi: 10.1093/rheumatology/31.10.653.
    1. Campbell IK, Rich MJ, Bischof RJ, Dunn AR, Grail D, Hamilton JA. Protection from collagen-induced arthritis in granulocyte-macrophage colony-stimulating factor-deficient mice. J Immunol. 1998;161:3639–3644.
    1. Cook AD, Braine EL, Campbell IK, Rich MJ, Hamilton JA. Blockade of collagen-induced arthritis post-onset by antibody to granulocyte-macrophage colony-stimulating factor (GM-CSF): requirement for GM-CSF in the effector phase of disease. Arthritis Res. 2001;3:293–298. doi: 10.1186/ar318.
    1. Hashimoto M, Hirota K, Yoshitomi H, Maeda S, Teradaira S, Akizuki S, et al. Complement drives Th17 cell differentiation and triggers autoimmune arthritis. J Exp Med. 2010;207:1135–1143. doi: 10.1084/jem.20092301.
    1. Cook AD, Turner AL, Braine EL, Pobjoy J, Lenzo JC, Hamilton JA. Regulation of systemic and local myeloid cell subpopulations by bone marrow cell-derived granulocyte-macrophage colony-stimulating factor in experimental inflammatory arthritis. Arthritis Rheum. 2011;63:2340–2351. doi: 10.1002/art.30354.
    1. Cook AD, Pobjoy J, Sarros S, Steidl S, Durr M, Lacey DC, et al. Granulocyte-macrophage colony-stimulating factor is a key mediator in inflammatory and arthritic pain. Ann Rheum Dis. 2013;72:265–270. doi: 10.1136/annrheumdis-2012-201703.
    1. De Vries EGE, Willemse PHB, Biesma B, Stern AC, Limburg PC, Vellenga E. Flare-up of rheumatoid arthritis during GM-CSF treatment after chemotherapy. Lancet. 1991;338:517–518. doi: 10.1016/0140-6736(91)90594-F.
    1. Hazenberg BP, Van Leeuwen MA, Van Rijswijk MH, Stern AC, Vellenga E. Correction of granulocytopenia in Felty’s syndrome by granulocyte-macrophage colony-stimulating factor. Simultaneous induction of interleukin-6 release and flare-up of the arthritis. Blood. 1989;74:2769–2770.
    1. Burmester GR, Feist E, Sleeman MA, Wang B, White B, Magrini F. Mavrilimumab, a human monoclonal antibody targeting GM-CSF receptor-alpha, in subjects with rheumatoid arthritis: a randomised, double-blind, placebo-controlled, phase I, first-in-human study. Ann Rheum Dis. 2011;70:1542–1549. doi: 10.1136/ard.2010.146225.
    1. Burmester GR, Weinblatt ME, McInnes IB, Porter D, Barbarash O, Vatutin M, et al. Efficacy and safety of mavrilimumab in subjects with rheumatoid arthritis. Ann Rheum Dis. 2013;72:1445–1452. doi: 10.1136/annrheumdis-2012-202450.
    1. Takeuchi T, Tanaka Y, Close D, Godwood A, Wu CY, Saurigny D. Efficacy and safety of mavrilimumab in Japanese subjects with rheumatoid arthritis: findings from a phase IIa study. Mod Rheumatol. 2015;25:21–30. doi: 10.3109/14397595.2014.896448.
    1. Burmester GR, McInnes IB, Kremer JM, Miranda P, Mariusz Korkosz JV, Rubbert-Roth A, et al. Efficacy and safety/tolerability of mavrilimumab, a human GM-CSF monoclonal antibody in patients with rheumatoid arthritis. Arthritis Rheum. 2014;66(abstract):2821.
    1. Behrens F, Tak PP, Ostergaard M, Stoilov R, Wiland P, Huizinga TW, et al. MOR103, a human monoclonal antibody to granulocyte-macrophage colony-stimulating factor, in the treatment of patients with moderate rheumatoid arthritis: results of a phase Ib/IIa randomised, double-blind, placebo-controlled, dose-escalation trial. Ann Rheum Dis. 2015;74:1058–1064. doi: 10.1136/annrheumdis-2013-204816.
    1. Krinner EM, Raum T, Petsch S, Bruckmaier S, Schuster I, Petersen L, et al. A human monoclonal IgG1 potently neutralizing the pro-inflammatory cytokine GM-CSF. Mol Immunol. 2007;44:916–925. doi: 10.1016/j.molimm.2006.03.020.
    1. Plater-Zyberk C, Joosten LA, Helsen MM, Hepp J, Baeuerle PA, van den Berg WB. GM-CSF neutralisation suppresses inflammation and protects cartilage in acute streptococcal cell wall arthritis of mice. Ann Rheum Dis. 2007;66:452–457. doi: 10.1136/ard.2006.057182.
    1. Huizinga TW, Batalov A, Stoilov R, Lloyd E, Wagner T, Saurigny D, Souberbielle B, Esfandiari E. Phase 1b randomized, double-blind study of namilumab, an anti-granulocyte macrophage colony-stimulating factor monoclonal antibody, in mild-to-moderate rheumatoid arthritis. Arthritis Res Ther. 2017;19:53. doi: 10.1186/s13075-017-1267-3.
    1. Ware JE, Kosinski M. Interpreting SF-36 summary health measures: a response. Qual Life Res. 2001;10:405–13.
    1. Inoue Y, Trapnell BC, Tazawa R, et al. Characteristics of a large cohort of patients with autoimmune pulmonary alveolar proteinosis in Japan. Am J Respir Crit Care Med. 2008;177:752–762. doi: 10.1164/rccm.200708-1271OC.
    1. Curtis JR, van der Helm-van Mil AH, Knevel R, Huizinga TW, Haney DJ, Shen Y, et al. Validation of a novel multibiomarker test to assess rheumatoid arthritis disease activity. Arthritis Care Res (Hoboken) 2012;64:1794–1803. doi: 10.1002/acr.21767.
    1. Siebuhr AS, Bay-Jensen AC, Leeming DJ, Plat A, Byrialsen I, Christiansen C, et al. Serological identification of fast progressors of structural damage with rheumatoid arthritis. Arthritis Res Ther. 2013;15:R86. doi: 10.1186/ar4266.
    1. Mandema JW, Gibbs M, Boyd RA, Wada DR, Pfister M. Model-based meta-analysis for comparative efficacy and safety: application in drug development and beyond. Clin Pharmacol Ther. 2011;90:766–769. doi: 10.1038/clpt.2011.242.
    1. Landewé RB, et al. Efficacy assessed in follow-ups of clinical studies: methodological conundrum. Arthritis Res Ther. 2010;12:132. doi: 10.1186/ar3080.
    1. Burmester Gerd R, McInnes Iain B, Kremer Joel, Miranda Pedro, Korkosz Mariusz, Vencovsky Jiri, Rubbert-Roth Andrea, Mysler Eduardo, Sleeman Matthew A, Godwood Alex, Sinibaldi Dominic, Guo Xiang, White Wendy I, Wang Bing, Wu Chi-Yuan, Ryan Patricia C, Close David, Weinblatt Michael E. A randomised phase IIb study of mavrilimumab, a novel GM–CSF receptor alpha monoclonal antibody, in the treatment of rheumatoid arthritis. Annals of the Rheumatic Diseases. 2017;76(6):1020–1030. doi: 10.1136/annrheumdis-2016-210624.
    1. Bathon JM, Martin RW, Fleishmann RM, Tesser JR, Schiff MH, Keystone EC, et al. A comparison of etanercept and methotrexate in patients with early rheumatoid arthritis. N Engl J Med. 2000;343:1586–1593. doi: 10.1056/NEJM200011303432201.

Source: PubMed

Подписаться