Mavrilimumab, a human monoclonal antibody targeting GM-CSF receptor-α, in subjects with rheumatoid arthritis: a randomised, double-blind, placebo-controlled, phase I, first-in-human study

Gerd R Burmester, Eugen Feist, Matthew A Sleeman, Bing Wang, Barbara White, Fabio Magrini, Gerd R Burmester, Eugen Feist, Matthew A Sleeman, Bing Wang, Barbara White, Fabio Magrini

Abstract

Objective: To evaluate the safety, tolerability, pharmacokinetic and pharmacodynamic profiles of mavrilimumab, a human monoclonal antibody targeting the granulocyte-macrophage colony-stimulating factor receptor-α, in subjects with rheumatoid arthritis (RA).

Methods: A randomised, double-blind, placebo-controlled, dose-escalating phase I study in subjects with RA who received stable methotrexate treatment for ≥3 months before enrolment.

Subjects: received single intravenous escalating doses of mavrilimumab (0.01-10.0 mg/kg) or placebo.

Results: 32 subjects were enrolled in this study (1 unblinded subject at 0.01 mg/kg and another at 0.03 mg/kg were followed by five sequential double-blinded cohorts, n=6 each, treated with 0.1, 0.3, 1.0, 3.0 and 10.0 mg/kg, respectively). Adverse events were mild or moderate and were reported with similar frequency across all treatment cohorts. One subject (10.0 mg/kg) experienced moderate face and neck urticaria during infusion that resolved with symptomatic treatment. Systemic clearance of mavrilimumab approached that of endogenous IgG at doses >1.0 mg/kg; pharmacodynamic activity was confirmed in the 1.0 and 3.0 mg/kg cohorts by suppression of suppressor of cytokine signalling 3 mRNA transcripts. In exploratory analyses, reductions of acute phase reactants were observed in subjects with elevated C-reactive protein (>5 mg/l) and erythrocyte sedimentation rate (≥20.0 mm/h) at baseline. No significant change in Disease Activity Score 28-joint assessment (DAS28) was seen in any of the cohorts. In mavrilimumab-treated subjects (n=15) with baseline DAS28 >3.2, mean disease activity (DAS28) was significantly reduced at 4 weeks.

Conclusion: In this first-in-human study, mavrilimumab showed preliminary evidence of pharmacodynamic activity. Importantly, the safety and pharmacokinetic profiles of mavrilimumab support further clinical studies in RA.

Trial registration number: NCT00771420.

Conflict of interest statement

Competing interests GRB and EF received research funding from MedImmune to conduct this clinical study. MAS, B Wang and FM are employees of MedImmune. B White is a former employee of MedImmune.

Figures

Figure 1
Figure 1
Mean pharmacokinetic profiles of mavrilimumab (formerly known as CAM-3001) after a single intravenous dose in subjects with mild to moderate rheumatoid arthritis. Error bars represent SEM. Pharmacokinetic observations from the subject in the 10.0 mg/kg cohort who received partial infusion were excluded from mean concentration calculations. LLOQ, lower limit of quantification.
Figure 2
Figure 2
Effect of mavrilimumab on suppressor of cytokine signalling 3 (SOCS3) mRNA transcript levels induced by granulocyte-macrophage colony-stimulating factor ex vivo from peripheral blood leucocytes of subjects with mild to moderate rheumatoid arthritis. Data are presented as mean (SE). Asterisks indicate a significant change (*p

Figure 3

(A) Effect of mavrilimumab (formerly…

Figure 3

(A) Effect of mavrilimumab (formerly known as CAM-3001) on Disease Activity Score 28-joint…

Figure 3
(A) Effect of mavrilimumab (formerly known as CAM-3001) on Disease Activity Score 28-joint assessment (DAS28) levels at baseline, week 4 and week 12. Data are presented as the mean (SD) for each visit. (B) Effect of mavrilimumab on DAS28 levels within the first 4 weeks after a single intravenous dose in subjects with moderate disease (DAS28 >3.2 at baseline). (C) Effect of mavrilimumab on C-reactive protein (CRP) level at baseline (week 0), week 2 and week 4. Data are presented as the mean (SD) for each visit. (D) Effect of mavrilimumab on CRP level within the first 4 weeks after a single intravenous dose in subjects with mild rheumatoid arthritis who had elevated (>5 mg/l) CRP levels at baseline. Legend: placebo (◊) and mavrilimumab 0.1 mg/kg (▲), 0.3 mg/kg (●), 1.0 mg/kg (■), 3.0 mg/kg (○) and 10.0 mg/kg (Δ).
Figure 3
Figure 3
(A) Effect of mavrilimumab (formerly known as CAM-3001) on Disease Activity Score 28-joint assessment (DAS28) levels at baseline, week 4 and week 12. Data are presented as the mean (SD) for each visit. (B) Effect of mavrilimumab on DAS28 levels within the first 4 weeks after a single intravenous dose in subjects with moderate disease (DAS28 >3.2 at baseline). (C) Effect of mavrilimumab on C-reactive protein (CRP) level at baseline (week 0), week 2 and week 4. Data are presented as the mean (SD) for each visit. (D) Effect of mavrilimumab on CRP level within the first 4 weeks after a single intravenous dose in subjects with mild rheumatoid arthritis who had elevated (>5 mg/l) CRP levels at baseline. Legend: placebo (◊) and mavrilimumab 0.1 mg/kg (▲), 0.3 mg/kg (●), 1.0 mg/kg (■), 3.0 mg/kg (○) and 10.0 mg/kg (Δ).

References

    1. Klareskog L, van der Heijde D, de Jager JP, et al. Therapeutic effect of the combination of etanercept and methotrexate compared with each treatment alone in patients with rheumatoid arthritis: double-blind randomised controlled trial. Lancet 2004;363:675–81
    1. Maini R, St Clair EW, Breedveld F, et al. Infliximab (chimeric anti-tumour necrosis factor alpha monoclonal antibody) versus placebo in rheumatoid arthritis patients receiving concomitant methotrexate: a randomised phase III trial. ATTRACT Study Group. Lancet 1999;354:1932–9
    1. Breedveld FC, Weisman MH, Kavanaugh AF, et al. The PREMIER study: A multicenter, randomized, double-blind clinical trial of combination therapy with adalimumab plus methotrexate versus methotrexate alone or adalimumab alone in patients with early, aggressive rheumatoid arthritis who had not had previous methotrexate treatment. Arthritis Rheum 2006;54:26–37
    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
    1. Salmon-Ceron D, Tubach F, Lortholary O, et al. Drug-specific risk of non-tuberculosis opportunistic infections in patients receiving anti-TNF therapy reported to the 3-year prospective French RATIO registry. Ann Rheum Dis 2011;70:616–23
    1. Listing J, Strangfeld A, Rau R, et al. Clinical and functional remission: even though biologics are superior to conventional DMARDs overall success rates remain low–results from RABBIT, the German biologics register. Arthritis Res Ther 2006;8:R66.
    1. Bresnihan B, Gerlag DM, Rooney T, et al. Synovial macrophages as a biomarker of response to therapeutic intervention in rheumatoid arthritis: standardization and consistency across centers. J Rheumatol 2007;34:620–2
    1. Hamilton JA. Colony-stimulating factors in inflammation and autoimmunity. Nat Rev Immunol 2008;8:533–44
    1. Gamble JR, Elliott MJ, Jaipargas E, et al. Regulation of human monocyte adherence by granulocyte-macrophage colony-stimulating factor. Proc Natl Acad Sci USA 1989;86:7169–73
    1. Cheung DL, Hamilton JA. Regulation of human monocyte DNA synthesis by colony-stimulating factors, cytokines, and cyclic adenosine monophosphate. Blood 1992;79:1972–81
    1. Hart PH, Vitti GF, Burgess DR, et al. Activation of human monocytes by granulocyte-macrophage colony-stimulating factor: increased urokinase-type plasminogen activator activity. Blood 1991;77:841–8
    1. Shibata Y, Berclaz PY, Chroneos ZC, et al. GM-CSF regulates alveolar macrophage differentiation and innate immunity in the lung through PU.1. Immunity 2001;15:557–67
    1. Alsalameh S, Firestein GS, Oez S, et al. Regulation of granulocyte macrophage colony stimulating factor production by human articular chondrocytes. Induction by both tumor necrosis factor-alpha and interleukin 1, downregulation by transforming growth factor beta and upregulation by fibroblast growth factor. J Rheumatol 1994;21:993–1002
    1. Fiehn C, Wermann M, Pezzutto A, et al. [Plasma GM-CSF concentrations in rheumatoid arthritis, systemic lupus erythematosus and spondyloarthropathy]. Z Rheumatol 1992;51:121–6
    1. Nakano K, Okada Y, Saito K, et al. Rheumatoid synovial endothelial cells produce macrophage colony-stimulating factor leading to osteoclastogenesis in rheumatoid arthritis. Rheumatology (Oxford) 2007;46:597–603
    1. Berenbaum F, Rajzbaum G, Amor B, et al. Evidence for GM-CSF receptor expression in synovial tissue. An analysis by semi-quantitative polymerase chain reaction on rheumatoid arthritis and osteoarthritis synovial biopsies. Eur Cytokine Netw 1994;5:43–6
    1. Lang RA, Metcalf D, Cuthbertson RA, et al. Transgenic mice expressing a hemopoietic growth factor gene (GM-CSF) develop accumulations of macrophages, blindness, and a fatal syndrome of tissue damage. Cell 1987;51:675–86
    1. Campbell IK, Bendele A, Smith DA, et al. Granulocyte-macrophage colony stimulating factor exacerbates collagen induced arthritis in mice. Ann Rheum Dis 1997;56:364–8
    1. Bischof RJ, Zafiropoulos D, Hamilton JA, et al. Exacerbation of acute inflammatory arthritis by the colony-stimulating factors CSF-1 and granulocyte macrophage (GM)-CSF: evidence of macrophage infiltration and local proliferation. Clin Exp Immunol 2000;119:361–7
    1. Campbell IK, Rich MJ, Bischof RJ, et al. Protection from collagen-induced arthritis in granulocyte-macrophage colony-stimulating factor-deficient mice. J Immunol 1998;161:3639–44
    1. Cook AD, Braine EL, Campbell IK, et al. 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;3:293–8
    1. Yang YH, Hamilton JA. Dependence of interleukin-1-induced arthritis on granulocyte-macrophage colony-stimulating factor. Arthritis Rheum 2001;44:111–19
    1. Plater-Zyberk C, Joosten LA, Helsen MM, et al. GM-CSF neutralisation suppresses inflammation and protects cartilage in acute streptococcal cell wall arthritis of mice. Ann Rheum Dis 2007;66:452–7
    1. Hazenberg BP, Van Leeuwen MA, Van Rijswijk MH, et al. 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;74:2769–70
    1. de Vries EG, Willemse PH, Biesma B, et al. Flare-up of rheumatoid arthritis during GM-CSF treatment after chemotherapy. Lancet 1991;338:517–8
    1. Hansen G, Hercus TR, McClure BJ, et al. The structure of the GM-CSF receptor complex reveals a distinct mode of cytokine receptor activation. Cell 2008;134:496–507
    1. Fleetwood AJ, Cook AD, Hamilton JA. Functions of granulocyte-macrophage colony-stimulating factor. Crit Rev Immunol 2005;25:405–28
    1. Gearing DP, King JA, Gough NM, et al. Expression cloning of a receptor for human granulocyte-macrophage colony-stimulating factor. EMBO J 1989;8:3667–76
    1. Rajotte D, Sadowski HB, Haman A, et al. Contribution of both STAT and SRF/TCF to c-fos promoter activation by granulocyte-macrophage colony-stimulating factor. Blood 1996;88:2906–16
    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
    1. Fries JF, Spitz P, Kraines RG, et al. Measurement of patient outcome in arthritis. Arthritis Rheum 1980;23:137–45
    1. Smolen JS, Aletaha D. What should be our treatment goal in rheumatoid arthritis today? Clin Exp Rheumatol 2006;24(6 Suppl 43):S–7–13
    1. Scherer HU, Dörner T, Burmester GR. Patient-tailored therapy in rheumatoid arthritis: an editorial review. Curr Opin Rheumatol 2010;22:237–45
    1. Coenen MJ, Enevold C, Barrera P, et al. Genetic variants in toll-like receptors are not associated with rheumatoid arthritis susceptibility or anti-tumour necrosis factor treatment outcome. PLoS ONE 2010;5:e14326.
    1. Lindberg J, Wijbrandts CA, van Baarsen LG, et al. The gene expression profile in the synovium as a predictor of the clinical response to infliximab treatment in rheumatoid arthritis. PLoS ONE 2010;5:e11310.
    1. Calogiuri G, Ventura MT, Mason L, et al. Hypersensitivity reactions to last generation chimeric, humanized [correction of umanized] and human recombinant monoclonal antibodies for therapeutic use. Curr Pharm Des 2008;14:2883–91
    1. Levy G. Pharmacologic target-mediated drug disposition. Clin Pharmacol Ther 1994;56:248–52
    1. Mager DE, Jusko WJ. General pharmacokinetic model for drugs exhibiting target‑mediated drug disposition. J Pharmacokinet Pharmacodyn 2001;28:507–32
    1. Hansen RJ, Balthasar JP. Pharmacokinetic/pharmacodynamic modeling of the effects of intravenous immunoglobulin on the disposition of antiplatelet antibodies in a rat model of immune thrombocytopenia. J Pharm Sci 2003;92:1206–15
    1. Hayashi N, Tsukamoto Y, Sallas WM, et al. A mechanism-based binding model for the population pharmacokinetics and pharmacodynamics of omalizumab. Br J Clin Pharmacol 2007;63:548–61
    1. Tabrizi MA, Tseng CM, Roskos LK. Elimination mechanisms of therapeutic monoclonal antibodies. Drug Discov Today 2006;11:81–8
    1. Rowinsky EK, Schwartz GH, Gollob JA, et al. Safety, pharmacokinetics, and activity of ABX-EGF, a fully human anti-epidermal growth factor receptor monoclonal antibody in patients with metastatic renal cell cancer. J Clin Oncol 2004;22:3003–15
    1. Wang B, Arends R, Roskos L, et al. A preliminary population pharmacokinetic analysis of panitumumab, a fully human IgG2 anti-EGFR monoclonal antibody. AAPS J 2004;6(Suppl 1):abstract 2375
    1. Wang W, Wang EQ, Balthasar JP. Monoclonal antibody pharmacokinetics and pharmacodynamics. Clin Pharmacol Ther 2008;84:548–58
    1. Weisman MH, Moreland LW, Furst DE, et al. Efficacy, pharmacokinetic, and safety assessment of adalimumab, a fully human anti-tumor necrosis factor-alpha monoclonal antibody, in adults with rheumatoid arthritis receiving concomitant methotrexate: a pilot study. Clin Ther 2003;25:1700–21
    1. Nishimoto N, Yoshizaki K, Maeda K, et al. Toxicity, pharmacokinetics, and dose-finding study of repetitive treatment with the humanized anti-interleukin 6 receptor antibody MRA in rheumatoid arthritis. Phase I/II clinical study. J Rheumatol 2003;30:1426–35

Source: PubMed

3
購読する