Blockade of interferon-γ normalizes interferon-regulated gene expression and serum CXCL10 levels in patients with systemic lupus erythematosus

Andrew A Welcher, Michael Boedigheimer, Alan J Kivitz, Zahir Amoura, Jill Buyon, Alla Rudinskaya, Kevin Latinis, Kit Chiu, Kelly S Oliner, Michael A Damore, Gregory E Arnold, Winnie Sohn, Narendra Chirmule, Lovely Goyal, Christopher Banfield, James B Chung, Andrew A Welcher, Michael Boedigheimer, Alan J Kivitz, Zahir Amoura, Jill Buyon, Alla Rudinskaya, Kevin Latinis, Kit Chiu, Kelly S Oliner, Michael A Damore, Gregory E Arnold, Winnie Sohn, Narendra Chirmule, Lovely Goyal, Christopher Banfield, James B Chung

Abstract

Objective: To assess the safety and immunologic impact of inhibiting interferon-γ (IFNγ) with AMG 811, a human IgG1 monoclonal antibody against IFNγ, in patients with systemic lupus erythematosus (SLE).

Methods: Twenty-six patients with mild-to-moderate, stable SLE were administered placebo or a single dose of AMG 811, ranging from 2 mg to 180 mg subcutaneously or 60 mg intravenously.

Results: Similar to results previously reported following inhibition of type I IFNs, treatment of SLE patients with AMG 811 led to a dose-dependent modulation of the expression of genes associated with IFN signaling, as assessed by microarray analysis of the whole blood. The list of impacted genes overlapped with that identified by stimulating human whole blood with IFNγ and with those gene sets reported in the literature to be differentially expressed in SLE patients. Serum levels of IFNγ-induced chemokines, including IFNγ-inducible protein 10 (IP-10), were found to be elevated at baseline in SLE patients as compared to healthy volunteers. In contrast to previously reported results from studies using type I IFN-blocking agents, treatment with AMG 811 led to dose-related reductions in the serum levels of CXCL10 (IP-10).

Conclusion: The scope and nature of the biomarkers impacted by AMG 811 support targeting of IFNγ as a therapeutic strategy for SLE.

Trial registration: ClinicalTrials.gov NCT00774943 NCT02391259.

© 2015 Amgen, Inc. Arthritis & Rheumatology is published by Wiley Periodicals, Inc. on behalf of the American College of Rheumatology.

Figures

Figure 1
Figure 1
AMG 811 serum concentration–time (A) and total interferon‐γ (IFNγ) serum concentration–time (B) profiles following a single subcutaneous (SC) dose (2–180 mg) or intravenous (IV) dose (60 mg) of AMG 811 in patients with systemic lupus erythematosus. Results are the mean ± SD.
Figure 2
Figure 2
Identification of the interferon‐γ (IFNγ) signature set and comparison to published IFN signatures. A, Volcano plot shows the mean fold change in gene expression (IFNγ‐stimulated relative to unstimulated) in whole blood from healthy volunteers, plotted with the associated P values determined by analysis of variance. Circled symbols indicate the 20 genes with the largest magnitude of change in expression, exceeding a significance level of 0.001. B, Two IFNγ scores were derived from 2 published gene lists (score A from gene set A [top] and score B from gene set B [middle]), and these were compared to an IFNγ score based on a set of 432 genes (gene set C) that met the significance criteria for modulated gene expression in IFNγ‐stimulated blood samples from healthy volunteers. A random selection of 500 genes (bottom) was used as a negative control. The broken diagonal line represents a reference to indicate perfect correlation.
Figure 3
Figure 3
Interferon‐γ (IFNγ) blockade with AMG 811 leads to normalization of the IFN‐related gene expression profile in systemic lupus erythematosus patients. A, Volcano plot shows the mean fold difference in gene expression posttreatment (day 15) in blood samples from AMG 811–treated patients compared to all other samples (all samples at baseline and placebo‐treated patients on day 15). Circled symbols indicate the top 20 IFNγ signature genes. B, Relationship between the serum AMG 811 concentration and guanylate binding protein 1 (GBP‐1) transcript expression on days 1 and 15 posttreatment. The GBP‐1 gene expression levels were standardized against the levels seen in the healthy volunteers, and values were plotted against the serum AMG 811 concentration. SD = standard deviation.
Figure 4
Figure 4
Analysis of serum proteins. A, Log concentrations of interleukin‐18 (IL‐18), CXCL10, and CCL2 in healthy volunteers (HV) and systemic lupus erythematosus patients. Values are presented as box plots, where the boxes represent the 25th and 75th percentiles (the interquartile range), the lines within the boxes represent the median, and the lines outside the boxes represent the most extreme data points within 1.5 times the interquartile range. Crosses represent outliers. B, Dose‐dependent decrease in serum levels of CXCL10 in response to AMG 811 administration. Results are the mean fold change from baseline (with 95% confidence intervals) in the concentrations of CXCL10 for each dose group of AMG 811 (2–180 mg subcutaneously or 60 mg intravenously [IV]) by study day (day 15 [Dy15], day 56 [Dy56], and end of study [EOS]), using correction for small sample size 40.

References

    1. Kotzin BL. Systemic lupus erythematosus. Cell 1996;85:303–6.
    1. Rahman A, Isenberg DA. Systemic lupus erythematosus. N Engl J Med 2008;358:929–39.
    1. Baechler EC, Batliwalla FM, Karypis G, Gaffney PM, Ortmann WA, Espe KJ, et al. Interferon‐inducible gene expression signature in peripheral blood cells of patients with severe lupus. Proc Natl Acad Sci U S A 2003;100:2610–5.
    1. Bennett L, Palucka AK, Arce E, Cantrell V, Borvak J, Banchereau J, et al. Interferon and granulopoiesis signatures in systemic lupus erythematosus blood. J Exp Med 2003;197:711–23.
    1. Crow MK, George S, Paget SA, Ly N, Woodward R, Fry K, et al. Expression of an interferon‐alpha gene program in SLE [abstract]. Arthritis Rheum 2002;46 Suppl:S281.
    1. Bauer JW, Petri M, Batliwalla FM, Koeuth T, Wilson J, Slattery C, et al. Interferon‐regulated chemokines as biomarkers of systemic lupus erythematosus disease activity: a validation study. Arthritis Rheum 2009;60:3098–107.
    1. Bauer JW, Baechler EC, Petri M, Batliwalla FM, Crawford D, Ortmann WA, et al. Elevated serum levels of interferon‐regulated chemokines are biomarkers for active human systemic lupus erythematosus. PLoS Med 2006;3:e491.
    1. Lit LC, Wong CK, Tam LS, Li EK, Lam CW. Raised plasma concentration and ex vivo production of inflammatory chemokines in patients with systemic lupus erythematosus. Ann Rheum Dis 2006;65:209–15.
    1. Narumi S, Takeuchi T, Kobayashi Y, Konishi K. Serum levels of IFN‐inducible protein‐10 relating to the activity of systemic lupus erythematosus. Cytokine 2000;12:1561–5.
    1. Kong KO, Tan AW, Thong BY, Lian TY, Cheng YK, Teh CL, et al. Enhanced expression of interferon‐inducible protein‐10 correlates with disease activity and clinical manifestations in systemic lupus erythematosus. Clin Exp Immunol 2009;156:134–40.
    1. Brinkmann V, Geiger T, Alkan S, Heusser CH. Interferon α increases the frequency of interferon γ‐producing human CD4+ T cells. J Exp Med 1993;178:1655–63.
    1. Nguyen KB, Cousens LP, Doughty LA, Pien GC, Durbin JE, Biron CA. Interferon α/β‐mediated inhibition and promotion of interferon γ: STAT1 resolves a paradox. Nat Immunol 2000;1:70–6.
    1. Rayamajhi M, Humann J, Penheiter K, Andreasen K, Lenz LL. Induction of IFN‐αβ enables Listeria monocytogenes to suppress macrophage activation by IFN‐γ. J Exp Med 2010;207:327–37.
    1. Hall JC, Casciola‐Rosen L, Berger AE, Kapsogeorgou EK, Cheadle C, Tzioufas AG, et al. Precise probes of type II interferon activity define the origin of interferon signatures in target tissues in rheumatic diseases. Proc Natl Acad Sci U S A 2012;109:17609–14.
    1. Chiche L, Jourde‐Chiche N, Whalen E, Presnell S, Gersuk V, Dang K, et al. Modular transcriptional repertoire analyses of adults with systemic lupus erythematosus reveal distinct type I and type II interferon signatures. Arthritis Rheumatol 2014;66:1583–95.
    1. Balomenos D, Rumold R, Theofilopoulos AN. Interferon‐γ is required for lupus‐like disease and lymphoaccumulation in MRL‐lpr mice. J Clin Invest 1998;101:364–71.
    1. Jacob CO, van der Meide PH, McDevitt HO. In vivo treatment of (NZB × NZW)F1 lupus‐like nephritis with monoclonal antibody to γ interferon. J Exp Med 1987;166:798–803.
    1. Peng SL, Moslehi J, Craft J. Roles of interferon‐γ and interleukin‐4 in murine lupus. J Clin Invest 1997;99:1936–46.
    1. Hron JD, Peng SL. Type I IFN protects against murine lupus. J Immunol 2004;173:2134–42.
    1. Seery JP, Carroll JM, Cattell V, Watt FM. Antinuclear autoantibodies and lupus nephritis in transgenic mice expressing interferon γ in the epidermis. J Exp Med 1997;186:1451–9.
    1. Harigai M, Kawamoto M, Hara M, Kubota T, Kamatani N, Miyasaka N. Excessive production of IFN‐γ in patients with systemic lupus erythematosus and its contribution to induction of B lymphocyte stimulator/B cell‐activating factor/TNF ligand superfamily‐13B. J Immunol 2008;181:2211–9.
    1. Karonitsch T, Feierl E, Steiner CW, Dalwigk K, Korb A, Binder N, et al. Activation of the interferon‐γ signaling pathway in systemic lupus erythematosus peripheral blood mononuclear cells. Arthritis Rheum 2009;60:1463–71.
    1. Graninger WB, Hassfeld W, Pesau BB, Machold KP, Zielinski CC, Smolen JS. Induction of systemic lupus erythematosus by interferon‐γ in a patient with rheumatoid arthritis. J Rheumatol 1991;18:1621–2.
    1. McBride JM, Jiang J, Abbas AR, Morimoto A, Li J, Maciuca R, et al. Safety and pharmacodynamics of rontalizumab in patients with systemic lupus erythematosus: results of a phase I, placebo‐controlled, double‐blind, dose‐escalation study. Arthritis Rheum 2012;64:3666–76.
    1. Yao Y, Richman L, Higgs BW, Morehouse CA, de los Reyes M, Brohawn P, et al. Neutralization of interferon‐α/β–inducible genes and downstream effect in a phase I trial of an anti–interferon‐α monoclonal antibody in systemic lupus erythematosus. Arthritis Rheum 2009;60:1785–96.
    1. Petri M, Wallace DJ, Spindler A, Chindalore V, Kalunian K, Mysler E, et al. Sifalimumab, a human anti–interferon‐α monoclonal antibody, in systemic lupus erythematosus; a phase I randomized, controlled, dose‐escalation study. Arthritis Rheum 2013;65:1011–21.
    1. Tan EM, Cohen AS, Fries JF, Masi AT, McShane DJ, Rothfield NF, et al. The 1982 revised criteria for the classification of systemic lupus erythematosus. Arthritis Rheum 1982;25:1271–7.
    1. Hochberg MC, for the Diagnostic and Therapeutic Criteria Committee of the American College of Rheumatology . Updating the American College of Rheumatology revised criteria for the classification of systemic lupus erythematosus [letter]. Arthritis Rheum 1997;40:1725.
    1. Symmons DP, Coppock JS, Bacon PA, Bresnihan B, Isenberg DA, Maddison P, et al, and Members of the British Isles Lupus Assessment Group (BILAG) . Development and assessment of a computerized index of clinical disease activity in systemic lupus erythematosus. Q J Med 1988;69:927–37.
    1. Storey JD. A direct approach to false discovery rates. J R Stat Soc Ser B 2002;64:479–98.
    1. Petri M, Kim MY, Kalunian KC, Grossman J, Hahn BH, Sammaritano LR, et al, for the OC‐SELENA Trial . Combined oral contraceptives in women with systemic lupus erythematosus. N Engl J Med 2005;353:2550–8.
    1. Shankar G, Devanarayan V, Amaravadi L, Barrett YC, Bowsher R, Finco‐Kent D, et al. Recommendations for the validation of immunoassays used for detection of host antibodies against biotechnology products. J Pharm Biomed Anal 2008;48:1267–81.
    1. Gupta S, Devanarayan V, Finco D, Gunn GR, Kirshner S, Richards S, et al. Recommendations for the validation of cell‐based assays used for the detection of neutralizing antibody immune responses elicited against biological therapeutics. J Pharm Biomed Anal 2011;55:878–88.
    1. Cui X, Churchill GA. Statistical tests for differential expression in cDNA microarray experiments. Genome Biol 2003;4:210.
    1. Horwitz DA, Gray JD, Behrendsen SC, Kubin M, Rengaraju M, Ohtsuka K, et al. Decreased production of interleukin‐12 and other Th1‐type cytokines in patients with recent‐onset systemic lupus erythematosus. Arthritis Rheum 1998;41:838–44.
    1. Funauchi M, Sugishima H, Minoda M, Horiuchi A. Serum levels of interferon‐γ in autoimmune disease. Tohoku J Exp Med 1991;164:259–67.
    1. Akashi K, Hayashi S, Gondo H, Mizuno S, Harada M, Tamura K, et al. Involvement of interferon‐γ and macrophage colony‐stimulating factor in pathogenesis of haemophagocytic lymphohistiocytosis in adults. Br J Haematol 1994;87:243–50.
    1. Wendling D, Racadot E, Wijdenes J. Treatment of severe rheumatoid arthritis by anti‐interleukin 6 monoclonal antibody. J Rheumatol 1993;20:259–62.
    1. Barrera P, Joosten LA, den Broeder AA, van de Putte LB, van Riel PL, van den Berg WB. Effects of treatment with a fully human anti‐tumour necrosis factor‐α monoclonal antibody on the local and systemic homeostasis of interleukin‐1 and TNFα in patients with rheumatoid arthritis. Ann Rheum Dis 2001;60:660–9.
    1. Kackar RN, Harville DA. Approximations for standard errors of estimators of fixed and random effects in mixed linear‐models. J Am Stat Assoc 1984;79:853–62.

Source: PubMed

3
구독하다