A phase 1b clinical trial evaluating sifalimumab, an anti-IFN-α monoclonal antibody, shows target neutralisation of a type I IFN signature in blood of dermatomyositis and polymyositis patients

Brandon W Higgs, Wei Zhu, Chris Morehouse, Wendy I White, Philip Brohawn, Xiang Guo, Marlon Rebelatto, Chenxiong Le, Anthony Amato, David Fiorentino, Steven A Greenberg, Jorn Drappa, Laura Richman, Warren Greth, Bahija Jallal, Yihong Yao, Brandon W Higgs, Wei Zhu, Chris Morehouse, Wendy I White, Philip Brohawn, Xiang Guo, Marlon Rebelatto, Chenxiong Le, Anthony Amato, David Fiorentino, Steven A Greenberg, Jorn Drappa, Laura Richman, Warren Greth, Bahija Jallal, Yihong Yao

Abstract

Objective: To assess the pharmacodynamic effects of sifalimumab, an investigational anti-IFN-α monoclonal antibody, in the blood and muscle of adult dermatomyositis and polymyositis patients by measuring neutralisation of a type I IFN gene signature (IFNGS) following drug exposure.

Methods: A phase 1b randomised, double-blinded, placebo controlled, dose-escalation, multicentre clinical trial was conducted to evaluate sifalimumab in dermatomyositis or polymyositis patients. Blood and muscle biopsies were procured before and after sifalimumab administration. Selected proteins were measured in patient serum with a multiplex assay, in the muscle using immunohistochemistry, and transcripts were profiled with microarray and quantitative reverse transcriptase PCR assays. A 13-gene IFNGS was used to measure the pharmacological effect of sifalimumab.

Results: The IFNGS was suppressed by a median of 53-66% across three time points (days 28, 56 and 98) in blood (p=0.019) and 47% at day 98 in muscle specimens post-sifalimumab administration. Both IFN-inducible transcripts and proteins were prevalently suppressed following sifalimumab administration. Patients with 15% or greater improvement from baseline manual muscle testing scores showed greater neutralisation of the IFNGS than patients with less than 15% improvement in both blood and muscle. Pathway/functional analysis of transcripts suppressed by sifalimumab showed that leucocyte infiltration, antigen presentation and immunoglobulin categories were most suppressed by sifalimumab and highly correlated with IFNGS neutralisation in muscle.

Conclusions: Sifalimumab suppressed the IFNGS in blood and muscle tissue in myositis patients, consistent with this molecule's mechanism of action with a positive correlative trend between target neutralisation and clinical improvement. These observations will require confirmation in a larger trial powered to evaluate efficacy.

Trial registration: ClinicalTrials.gov NCT00533091.

Keywords: Cytokines; Dermatomyositis; Polymyositis.

Figures

Figure 1
Figure 1
Median target neutralisation (with median absolute deviation error bars) of the IFN gene signature (IFNGS) as calculated based on the expression of 13 genes (see supplementary material, available online only) pre-dose and post-dose up to day 98 for dose cohorts of 0.3 mg/kg (green), 1 mg/kg (blue), 3 mg/kg (orange), 10 mg/kg (purple) and placebo (red) in (A) blood and (C) muscle specimens, as well as median combined dose cohorts (blue) versus placebo treatment cohorts (red) in (B) blood and (D) muscle specimens from dermatomyositis or polymyositis patients. The y-axis represents the percentage of IFNGS remaining following treatment; each line is the median of the respective dose cohort. p Values at each time point for each dose cohort are provided in supplementary material (available online only), while those with p<0.05 are marked with an asterisk.
Figure 2
Figure 2
Target neutralisation of the IFN gene signature (IFNGS) calculated at days 28 (n=36), 56 (n=31) and 98 (n=32) post-treatment in blood specimens (first three columns) and day 98 (n=30) post-treatment in muscle specimens (last column) with either sifalimumab (blue vertical bar) or placebo (green vertical bar) on a patient level (designated on the right y-axis). Each row indicates a patient with values approaching red, representing maximum neutralisation of the IFNGS and values approaching blue, representing minimum neutralisation. Both blood and muscle comparisons are made to day 0 (pre-treatment) and all columns are sorted based on the day 28 column values. Neutralisation values in black indicate missing data points.
Figure 3
Figure 3
Stratified target neutralisation curves representing the proportion of dermatomyositis (n=14) or polymyositis (n=10) patients dosed with sifalimumab who demonstrate neutralisation of their IFN gene signature (IFNGS) at day 98 at the provided threshold value on the x-axis in (A) blood and (B) muscle specimens. Plots include all IFNGS-positive patients. Patients who exhibited a 15% or greater improvement in MMT8 score at day 98 (compared to day 0) are represented by the blue lines (blood n=8; muscle n=7), while those patients who did not are represented by the purple lines (blood n=16; muscle n=17).
Figure 4
Figure 4
Three of the pathways most suppressed by sifalimumab at day 98 in muscle specimens from dermatomyositis or polymyositis patients and unique to type I IFN signalling. Each point indicates a gene in either the (A) leucocyte extravasation pathway, (B) antigen presentation pathway, or (C) B-cell development pathway treated with either sifalimumab (blue squares) or placebo (red triangles). The y-axis represents the percentage of patients with at least 37.5% neutralisation of pathway-enricheds transcripts at day 98 in muscle tissue specimens.
Figure 5
Figure 5
Correlation between target neutralisation of the IFN gene signature (IFNGS) and neutralisation of the (A) leucocyte index, (B) MHC class I signature and (C) immunoglobulin signature in dermatomyositis (DM; red squares) or polymyositis (PM; blue circles) patient muscle specimens. Red dashed lines flanking the black dotted least squares fit line indicate 95% CI. The plots indicate that the alterations of the three gene signatures display significant negative correlation with target neutralisation of the IFNGS in muscle.

References

    1. Mastaglia FL, Garlepp MJ, Phillips BA, et al. Inflammatory myopathies: clinical, diagnostic and therapeutic aspects. Muscle Nerve 2003;27:407–25
    1. Christopher-Stine L, Plotz PH. Adult inflammatory myopathies. Best Pract Res Clin Rheumatol 2004;18:331–44
    1. Greenberg SA. Type 1 interferons and myositis. Arthritis Res Ther 2010;12(Suppl. 1):S4.
    1. Amato AA, Griggs RC. Treatment of idiopathic inflammatory myopathies. Curr Opin Neurol 2003;16:569–75
    1. Choy EH, Hoogendijk JE, Lecky B, et al. Withdrawn: immunosuppressant and immunomodulatory treatment for dermatomyositis and polymyositis. Cochrane Database Syst Rev 2009;4:CD003643.
    1. Icon Health Publications Myositis: a medical dictionary, bibliography, and annotated research guide to internet resources. San Diego, CA: Icon Health Publications, 2004
    1. Isenberg DA, Rowe D, Shearer M, et al. Localization of interferons and interleukin 2 in polymyositis and muscular dystrophy. Clin Exp Immunol 1986;63:450–8
    1. Greenberg SA, Pinkus JL, Pinkus GS, et al. Interferon-alpha/beta mediated innate immune mechanisms in dermatomyositis. Ann Neurol 2005;57:664–78
    1. Wenzel J, Schmidt R, Proelss J, et al. Type I interferon-associated skin recruitment of CXCR3+ lymphocytes in dermatomyositis. Clin Exp Dermatol 2006;31:576–82
    1. Yao Y, Richman L, Higgs BW, et al. Neutralization of interferon-alpha/beta-inducible genes and downstream effect in a phase I trial of an anti-interferon-alpha monoclonal antibody in systemic lupus erythematosus. Arthritis Rheum 2009;60:1785–96
    1. Kirou KA, Lee C, George S, et al. Activation of the interferon-α pathway identifies a subgroup of systemic lupus erythematosus patients with distinct serologic features and active disease. Arthritis Rheum 2005;52:1491–503
    1. Hua J, Kirou K, Lee C, et al. Functional assay of type I interferon in systemic lupus erythematosus plasma and association with anti-RNA binding protein autoantibodies. Arthritis Rheum 2006;54:1906–16
    1. Bengtsson AA, Sturfelt G, Truedsson L, et al. Activation of type I interferon system in systemic lupus erythematosus correlates with disease activity but not with antiretroviral antibodies. Lupus 2000;9:664–71
    1. Higgs BW, Liu Z, White B, et al. Patients with systemic lupus erythematosus, myositis, rheumatoid arthritis and scleroderma share activation of a common type I interferon pathway. Ann Rheum Dis 2011;70:2029–36
    1. Greenberg SA, Higgs BW, Morehouse C, et al. Relationship between disease activity and type 1 interferon- and other cytokine-inducible gene expression in blood in dermatomyositis and polymyositis. Genes Immun 2012;13:207–13
    1. Walsh RJ, Kong SW, Yao Y, et al. Type I interferon-inducible gene expression in blood is present and reflects disease activity in dermatomyositis and polymyositis. Arthritis Rheum 2007;56:3784–92
    1. Baechler EC, Bauer JW, Slattery CA, et al. An interferon signature in the peripheral blood of dermatomyositis patients is associated with disease activity. Mol Med 2007;13:59–68
    1. Bilgic H, Ytterberg SR, Amin S, et al. Interleukin-6 and type I interferon-regulated genes and chemokines mark disease activity in dermatomyositis. Arthritis Rheum 2009;60:3436–46
    1. Liao AP, Salajegheh M, Nazareno R, et al. Interferon β is associated with type 1 interferon-inducible gene expression in dermatomyositis. Ann Rheum Dis 2011;70:831–6
    1. Yao Y, Higgs BW, Richman L, et al. Use of type I interferon-inducible mRNAs as pharmacodynamic markers and potential diagnostic markers in trials with sifalimumab, an anti-IFNalpha antibody, in systemic lupus erythematosus. Arthritis Res Ther 2010;12(Suppl. 1):S6.
    1. Yao Y, Higgs BW, Morehouse C, et al. Development of potential pharmacodynamic and diagnostic markers for anti-IFN-α monoclonal antibody trials in systemic lupus erythematosus. Hum Genomics Proteomics 2009. Published Online First: 17 Nov 2009. pii: 374312. doi:10.4061/2009/374312
    1. McBride JM, Jiang J, Abbas AR, et al. Safety and pharmacodynamic results of rontalizumab in a phase I, placebo controlled, double blind, dose escalation study in systemic lupus erythematosus. Arthritis Rheum 2012;64:3666–76
    1. Merrill JT, Wallace DJ, Petri M, et al. Lupus Interferon Skin Activity (LISA) Study Investigators. Safety profile and clinical activity of sifalimumab, a fully human anti-interferon α monoclonal antibody, in systemic lupus erythematosus: a phase I, multicentre, double-blind randomized study. Ann Rheum Dis 2011;70:1905–13
    1. Petri M, Wallace DJ, Spindler A, et al. Sifalimumab, a human anti-interferon-a monoclonal antibody, in systemic lupus erythematosus: a phase 1 randomized controlled, dose-escalation study. Arthritis Rheum 11 Feb 2013. doi: 10.1002/art.37824
    1. Yao Y, Richman L, Morehouse C, et al. Type I interferon: potential therapeutic target for psoriasis? PLoS One 2008;3:e2737.
    1. Zhu W, Streicher K, Shen N, et al. Genomic signatures characterize leukocyte infiltration in myositis muscles. BMC Med Genomics 2012;5:53.
    1. Cappelletti C, Baggi F, Zolezzi F, et al. Type I interferon and Toll-like receptor expression characterizes inflammatory myopathies. Neurology 2011;76:2079–88
    1. Kotenko SV, Gallagher G, Baurin VV, et al. IFN-lambdas mediate antiviral protection through a distinct class II cytokine receptor complex. Nat Immunol 2003;4:69–77
    1. Yin Z, Dai J, Deng J, et al. Type III IFNs are produced by and stimulate human plasmacytoid dendritic cells. J Immunol 2012;189:2735–45
    1. Dalakas MC. Mechanisms of disease: signaling pathways and immunobiology of inflammatory myopathies. Nat Clin Pract Rheumatol 2006;2:219–27
    1. Zong M, Lundberg IE. Pathogenesis, classification and treatment of inflammatory myopathies. Nat Rev Rheumatol 2011;7:297–306
    1. Szodoray P, Alex P, Knowlton N, et al. Idiopathic inflammatory myopathies, signified by distinctive peripheral cytokines, chemokines and the TNF family members B-cell activating factor and a proliferation inducing ligand. Rheumatology (Oxford) 2010;49:1867–77
    1. Page G, Chevrel G, Miossec P. Anatomic localization of immature and mature dendritic cell subsets in dermatomyositis and polymyositis: interaction with chemokines and Th1 cytokine-producing cells. Arthritis Rheum 2004;50:199–208

Source: PubMed

3
S'abonner