Gene Expression and Pharmacodynamic Changes in 1,760 Systemic Lupus Erythematosus Patients From Two Phase III Trials of BAFF Blockade With Tabalumab

Robert W Hoffman, Joan T Merrill, Marta M E Alarcón-Riquelme, Michelle Petri, Ernst R Dow, Eric Nantz, Laura K Nisenbaum, Krista M Schroeder, Wendy J Komocsar, Narayanan B Perumal, Matthew D Linnik, David C Airey, Yushi Liu, Guilherme V Rocha, Richard E Higgs, Robert W Hoffman, Joan T Merrill, Marta M E Alarcón-Riquelme, Michelle Petri, Ernst R Dow, Eric Nantz, Laura K Nisenbaum, Krista M Schroeder, Wendy J Komocsar, Narayanan B Perumal, Matthew D Linnik, David C Airey, Yushi Liu, Guilherme V Rocha, Richard E Higgs

Abstract

Objective: To characterize baseline gene expression and pharmacodynamically induced changes in whole blood gene expression in 1,760 systemic lupus erythematosus (SLE) patients from 2 phase III, 52-week, randomized, placebo-controlled, double-blind studies in which patients were treated with the BAFF-blocking IgG4 monoclonal antibody tabalumab.

Methods: Patient samples were obtained from SLE patients from the ILLUMINATE-1 and ILLUMINATE-2 studies, and control samples were obtained from healthy donors. Blood was collected in Tempus tubes at baseline, week 16, and week 52. RNA was analyzed using Affymetrix Human Transcriptome Array 2.0 and NanoString.

Results: At baseline, expression of the interferon (IFN) response gene was elevated in patients compared with controls, with 75% of patients being positive for this IFN response gene signature. There was, however, substantial heterogeneity of IFN response gene expression and complex relationships among gene networks. The IFN response gene signature was a predictor of time to disease flare, independent of anti-double-stranded DNA (anti-dsDNA) antibody and C3 and C4 levels, and overall disease activity. Pharmacodynamically induced changes in gene expression following tabalumab treatment were extensive, occurring predominantly in B cell-related and immunoglobulin genes, and were consistent with other pharmacodynamic changes including anti-dsDNA antibody, C3, and immunoglobulin levels.

Conclusion: SLE patients demonstrated increased expression of an IFN response gene signature (75% of patients had an elevated IFN response gene signature) at baseline in ILLUMINATE-1 and ILLUMINATE-2. Substantial heterogeneity of gene expression was detected among individual patients and in gene networks. The IFN response gene signature was an independent risk factor for future disease flares. Pharmacodynamic changes in gene expression were consistent with the mechanism of BAFF blockade by tabalumab.

Trial registration: ClinicalTrials.gov NCT01205438 NCT01196091.

© 2016, American College of Rheumatology.

Figures

Figure 1
Figure 1
Estimation of the interferon (IFN) response gene signature in 164 individual prespecified genes. A total of 1,760 patients (881 from ILLUMINATE‐1 and 879 from ILLUMINATE‐2) and 60 control subjects were studied. A and B, The 164 prespecified genes are represented on the x‐axes; control subjects and patients are represented on the y‐axes for ILLUMINATE‐1 (A) and ILLUMINATE‐2 (B). At least 3 broad patterns of gene expression can be visualized on both 164‐gene expression heatmaps, although when reduced to a 34‐gene signature this could be clearly discerned as high and low expression subgroups. C, Distribution of IFN response gene expression among patients and controls. Of the 1,760 patients studied, 75% had elevated expression of IFN response genes compared with healthy controls (see Patients and Methods for additional details). Data are depicted at the top as a box plot, where the box represents the 25th to 75th percentiles, the line within the box represents the median, and the lines outside the box represent the 10th and 90th percentiles. The diamond represents results from healthy controls.
Figure 2
Figure 2
Characterization of network connectivity between highly expressed interferon (IFN) response genes. As shown by the lines connecting individual genes, a network of highly correlated genes is expressed in the systemic lupus erythematosus (SLE) cohorts, the majority of which have been identified previously as IFN response genes. There is, however, substantially greater complexity of the IFN network than has been previously described (for additional information, see Patients and Methods, and Supplementary Material, available on the Arthritis & Rheumatology web site at http://onlinelibrary.wiley.com/doi/10.1002/art.39950/abstract). Briefly, the Weighted correlation network analysis module (“Yellow module”) containing this network is correlated with all 3 clinical end points: (IFN signature [ρ = 0.90, P < 2 × 10−308], Safety of Estrogens in Lupus Erythematosus National Assessment version of the SLE Disease Activity Index [ρ = 0.22, P = 5 × 10−21], anti‐DNA activity [ρ = 0.32, P = 4 × 10−43]) (see Supplementary Figure 2 and Supplementary Table 3, available on the Arthritis & Rheumatology web site at http://onlinelibrary.wiley.com/doi/10.1002/art.39950/abstract) and is enriched for the MetaCore‐defined SLE disease pathway (P = 1.14 × 10−32 by hypergeometric test) and the gene ontology (GO) “response to type I interferon” gene set (GO:0034340 [P = 6.60 × 10−36]) (see Supplementary Figure 1, available on the Arthritis & Rheumatology web site at http://onlinelibrary.wiley.com/doi/10.1002/art.39950/abstract).

References

    1. Lisnevskaia L, Murphy G, Isenberg D. Systemic lupus erythematosus. Lancet 2014;384:1878–88.
    1. D'Cruz DP, Khamashta MA, Hughes GR. Systemic lupus erythematosus. Lancet 2007;369:587–96.
    1. Schneider P, MacKay F, Steiner V, Hofmann K, Bodmer JL, Holler N, et al. BAFF, a novel ligand of the tumor necrosis factor family, stimulates B cell growth. J Exp Med 1999;189:1747–56.
    1. Cancro MP, D'Cruz DP, Khamashta MA. The role of B lymphocyte stimulator (BLyS) in systemic lupus erythematosus. J Clin Invest 2009;119:1066–73.
    1. Darce JR, Arendt BK, Wu X, Jelinek DF. Regulated expression of BAFF‐binding receptors during human B cell differentiation. J Immunol 2007;179:7276–86.
    1. Stohl W, Metyas S, Tan SM, Cheema GS, Oamar B, Roschke V, et al. B lymphocyte stimulator overexpression in patients with systemic lupus erythematosus: longitudinal observations. Arthritis Rheum 2003;48:3475–86.
    1. Petri M, Stohl W, Chatham W, McCune WJ, Chevrier M, Ryel J, et al. Association of plasma B lymphocyte stimulator levels and disease activity in systemic lupus erythematosus. Arthritis Rheum 2008;58:2453–9.
    1. Navarra SV, Guzman RM, Gallacher AE, Hall S, Levy RA, Jimenez RE, et al. Efficacy and safety of belimumab in patients with active systemic lupus erythematosus: a randomized, placebo‐controlled, phase 3 trial. Lancet 2011;377:721–31.
    1. Furie R, Petri M, Zamani O, Cervera R, Wallace DJ, Tegzová D, et al. A phase III, randomized, placebo‐controlled study of belimumab, a monoclonal antibody that inhibits B lymphocyte stimulator, in patients with systemic lupus erythematosus. Arthritis Rheum 2011;63:3918–30.
    1. Furie RA, Leon G, Thomas M, Petri MA, Chu AD, Hislop C, et al. A phase 2, randomized, placebo‐controlled clinical trial of blisibimod, an inhibitor of B cell activating factor, in patients with moderate‐to‐severe systemic lupus erythematosus, the PEARL‐SC study. Ann Rheum Dis 2015;74:1667–75.
    1. Isenberg DA, Petri M, Kalunian K, Tanaka Y, Urowitz MB, Hoffman RW, et al. Efficacy and safety of subcutaneous tabalumab in patients with systemic lupus erythematosus: results from ILLUMINATE‐1, a 52‐week, phase III, multicentre, randomised, double‐blind, placebo‐controlled study. Ann Rheum Dis 2016;75:323–31.
    1. Merrill JT, van Vollenhoven RF, Buyon JP, Furie RA, Stohl W, Morgan‐Cox M, et al. Efficacy and safety of subcutaneous tabalumab, a monoclonal antibody to B‐cell activating factor, in patients with systemic lupus erythematosus: results from ILLUMINATE‐2, a 52‐week, phase III, multicentre, randomised, double‐blind, placebo‐controlled study. Ann Rheum Dis 2016;75:332–40.
    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 systemic 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. Kirou KA, Lee C, George S, Louca K, Peterson MG, Crow MK. 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. Kennedy WP, Maciuca R, Wolslegel K, Tew W, Abbas A, Chaivorapol C, et al. Association of the interferon signature metric with serologic disease manifestations but not global activity scores in multiple cohorts of patients with SLE. Lupus Sci Med 2015;2:e000080.
    1. Carpintero MF, Martinez L, Fernandez I, Romero AC, Mejia C, Zang YJ, et al. Diagnosis and risk stratification in patients with anti‐RNP autoimmunity. Lupus 2015;24:1057–66.
    1. M Petri, MY Kim, KC Kalunian, J Grossman, BH Hahn, LR Sammaritano, 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. 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. Deng Y, Tsao BP. Genetic susceptibility to systemic lupus erythematosus in the genomic era. Nat Rev Rheumatol 2010;6:683–92.
    1. Guerra SG, Vyse TJ, Cunninghame Graham DS. The genetics of lupus: a functional perspective. Arthritis Res Ther 2012;14:211.
    1. Irizarry RA, Bolstad BM, Collin F, Cope LM, Hobbs B, Speed TP. Summaries of Affymetrix GeneChip probe level data. Nucleic Acids Res 2003;31:e15.
    1. Geiss GK, Bumgarner RE, Birditt B, Dahl T, Dowidar N, Dunaway DL, et al. Direct multiplexed measurement of gene expression with color‐coded probe pairs. Nat Biotechnol 2008;26:317–25.
    1. Langfelder P, Horvath S. WGCNA: an R package for weighted correlation network analysis. BMC Bioinformatics 2008;9:559.
    1. Langfelder P, Luo R, Oldham MC, Horvath S. Is my network module preserved and reproducible? PLoS Comput Biol 2011;7:e1001057.
    1. Benjamini Y, Hochberg Y. Controlling the false discovery rate: a practical and powerful approach to multiple testing. J Royal Statistical Soc 1995;57:289–300.
    1. Liu A, Qu Y, Tang L, Ting N, Tsong Y, editors. A framework of statistical methods for identification of subgroups with differential treatment effects in randomized trials Applied statistics in biomedicine and clinical trials design: selected papers from 2013 ICSA. New York: Springer Verlag; 2015.
    1. Leon AC, Heo M. Sample sizes required to detect interactions between two binary fixed‐effects in a mixed‐effects linear regression model. Comput Stat Data Anal 2009;53:603–8.
    1. Eisenberg E, Levanon EY. Human housekeeping genes, revisited [published erratum appears in Trends Genet 2014;30:119–20]. Trends Genet 2013;29:569–74.
    1. Kariuki SN, Ghodke‐Puranik Y, Dorschner JM, Chrabot BS, Kelly JA, Tsao BP, et al. Genetic analysis of the pathogenic molecular sub‐phenotype interferon‐α identifies multiple novel loci involved in systemic lupus erythematosus. Genes Immun 2015;16:15–23.
    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. Ramos PS, Williams AH, Ziegler JT, Comeau ME, Guy RT, Lessard CJ, et al. Genetic analyses of interferon pathway–related genes reveal multiple new loci associated with systemic lupus erythematosus. Arthritis Rheum 2011;63:2049–57.
    1. Bentham J, Morris DL, Cunninghame Graham DS, Pinder DL, Tombleson P, Behrens TW, et al. Genetic association analyses implicated aberrant regulation of innate and adaptive immunity genes in the pathogenesis of systemic lupus erythematosus. Nat Genet 2015;47:1457–64.
    1. Krueger JG, Fretzin S, Suarez‐Farinas M, Haslett PA, Phipps KM, Cameron GS, et al. IL‐17A is essential for cell activation and inflammatory gene circuits in subjects with psoriasis. J Allergy Clin Immunol 2012;130:145–54.e9.
    1. Hofmann‐Apitius M, Alarcón‐Riquelme ME, Chamberlain C, McHale D. Towards the taxonomy of human disease. Nat Rev Drug Discov 2015;14:75–6.
    1. Kalunian KC, Merrill JT, Maciuca R, McBride JM, Townsend MJ, Wei X, et al. A phase II study of the efficacy and safety of rontalizumab (rhuMAb interferon‐α) in patients with systemic lupus erythematosus (ROSE). Ann Rheum Dis 2016;75:196–202.
    1. Aranow C, Kamen DL, Dall'Era M, Massarotti EM, Mackay MC, Koumpouras F, et al. Randomized double‐blind, placebo‐controlled trial of the effect of vitamin D3 on the interferon signature in patients with systemic lupus erythematosus. Arthritis Rheumatol 2015;67:1848–57.
    1. Petri M, Singh S, Tesfasyone H, Dedrick R, Fry K, Lal P, et al. Longitudinal expression of type I interferon responsive genes in systemic lupus erythematosus. Lupus 2009;18:980–9.
    1. Landolt‐Marticorena C, Bonventi G, Lubovich A, Ferguson C, Unnithan T, Su J, et al. Lack of association between the interferon‐α signature and longitudinal changes in disease activity in systemic lupus erythematosus. Ann Rheum Dis 2009;68:1440–6.
    1. Feng X, Wu H, Grossman JM, Hanvivadhanakul P, FitzGerald JD, Park GS, et al. Association of increased interferon‐inducible gene expression with disease activity and lupus nephritis in patients with systemic lupus erythematosus. Arthritis Rheum 2006;54:2951–62.
    1. Weckerle CE, Franek BS, Kelly JA, Kumabe M, Mikolaitis RA, Green SL, et al. Network analysis of associations between serum interferon‐α activity, autoantibodies, and clinical features in systemic lupus erythematosus. Arthritis Rheum 2011;63:1044–53.
    1. Niewold TB, Hua J, Lehman TJ, Harley JB, Crow MK. High serum IFN‐α activity is a heritable risk factor for systemic lupus erythematosus. Genes Immun 2007;8:492–502.
    1. Streicher K, Morehouse CA, Groves CJ, Rajan B, Pilataxi F, Lehmann KP, et al. The plasma cell signature in autoimmune disease. Arthritis Rheumatol 2014;66:173–84.
    1. Novershtern N, Subramanian A, Lawton LN, Mak RH, Haining WN, McConkey ME, et al. Densely interconnected transcriptional circuits control cell states in human hematopoiesis. Cell 2011;144:296–309.
    1. Banchereau R, Hong S, Cantarel B, Baldwin N, Baisch J, Edens M, et al. Personalized immunomonitoring uncovers molecular networks that stratify lupus patients. Cell 2016;165:551–65.

Source: PubMed

3
Tilaa