Genome-wide association study and gene expression analysis identifies CD84 as a predictor of response to etanercept therapy in rheumatoid arthritis

Jing Cui, Eli A Stahl, Saedis Saevarsdottir, Corinne Miceli, Dorothee Diogo, Gosia Trynka, Towfique Raj, Maša Umiċeviċ Mirkov, Helena Canhao, Katsunori Ikari, Chikashi Terao, Yukinori Okada, Sara Wedrén, Johan Askling, Hisashi Yamanaka, Shigeki Momohara, Atsuo Taniguchi, Koichiro Ohmura, Fumihiko Matsuda, Tsuneyo Mimori, Namrata Gupta, Manik Kuchroo, Ann W Morgan, John D Isaacs, Anthony G Wilson, Kimme L Hyrich, Marieke Herenius, Marieke E Doorenspleet, Paul-Peter Tak, J Bart A Crusius, Irene E van der Horst-Bruinsma, Gert Jan Wolbink, Piet L C M van Riel, Mart van de Laar, Henk-Jan Guchelaar, Nancy A Shadick, Cornelia F Allaart, Tom W J Huizinga, Rene E M Toes, Robert P Kimberly, S Louis Bridges Jr, Lindsey A Criswell, Larry W Moreland, João Eurico Fonseca, Niek de Vries, Barbara E Stranger, Philip L De Jager, Soumya Raychaudhuri, Michael E Weinblatt, Peter K Gregersen, Xavier Mariette, Anne Barton, Leonid Padyukov, Marieke J H Coenen, Elizabeth W Karlson, Robert M Plenge, Jing Cui, Eli A Stahl, Saedis Saevarsdottir, Corinne Miceli, Dorothee Diogo, Gosia Trynka, Towfique Raj, Maša Umiċeviċ Mirkov, Helena Canhao, Katsunori Ikari, Chikashi Terao, Yukinori Okada, Sara Wedrén, Johan Askling, Hisashi Yamanaka, Shigeki Momohara, Atsuo Taniguchi, Koichiro Ohmura, Fumihiko Matsuda, Tsuneyo Mimori, Namrata Gupta, Manik Kuchroo, Ann W Morgan, John D Isaacs, Anthony G Wilson, Kimme L Hyrich, Marieke Herenius, Marieke E Doorenspleet, Paul-Peter Tak, J Bart A Crusius, Irene E van der Horst-Bruinsma, Gert Jan Wolbink, Piet L C M van Riel, Mart van de Laar, Henk-Jan Guchelaar, Nancy A Shadick, Cornelia F Allaart, Tom W J Huizinga, Rene E M Toes, Robert P Kimberly, S Louis Bridges Jr, Lindsey A Criswell, Larry W Moreland, João Eurico Fonseca, Niek de Vries, Barbara E Stranger, Philip L De Jager, Soumya Raychaudhuri, Michael E Weinblatt, Peter K Gregersen, Xavier Mariette, Anne Barton, Leonid Padyukov, Marieke J H Coenen, Elizabeth W Karlson, Robert M Plenge

Abstract

Anti-tumor necrosis factor alpha (anti-TNF) biologic therapy is a widely used treatment for rheumatoid arthritis (RA). It is unknown why some RA patients fail to respond adequately to anti-TNF therapy, which limits the development of clinical biomarkers to predict response or new drugs to target refractory cases. To understand the biological basis of response to anti-TNF therapy, we conducted a genome-wide association study (GWAS) meta-analysis of more than 2 million common variants in 2,706 RA patients from 13 different collections. Patients were treated with one of three anti-TNF medications: etanercept (n = 733), infliximab (n = 894), or adalimumab (n = 1,071). We identified a SNP (rs6427528) at the 1q23 locus that was associated with change in disease activity score (ΔDAS) in the etanercept subset of patients (P = 8 × 10(-8)), but not in the infliximab or adalimumab subsets (P>0.05). The SNP is predicted to disrupt transcription factor binding site motifs in the 3' UTR of an immune-related gene, CD84, and the allele associated with better response to etanercept was associated with higher CD84 gene expression in peripheral blood mononuclear cells (P = 1 × 10(-11) in 228 non-RA patients and P = 0.004 in 132 RA patients). Consistent with the genetic findings, higher CD84 gene expression correlated with lower cross-sectional DAS (P = 0.02, n = 210) and showed a non-significant trend for better ΔDAS in a subset of RA patients with gene expression data (n = 31, etanercept-treated). A small, multi-ethnic replication showed a non-significant trend towards an association among etanercept-treated RA patients of Portuguese ancestry (n = 139, P = 0.4), but no association among patients of Japanese ancestry (n = 151, P = 0.8). Our study demonstrates that an allele associated with response to etanercept therapy is also associated with CD84 gene expression, and further that CD84 expression correlates with disease activity. These findings support a model in which CD84 genotypes and/or expression may serve as a useful biomarker for response to etanercept treatment in RA patients of European ancestry.

Conflict of interest statement

The authors have declared that no competing interests exist.

Figures

Figure 1. GWAS results for the ΔDAS…
Figure 1. GWAS results for the ΔDAS phenotype.
Shown are strengths of association (−Log10 P-value) for each SNP versus position along chromosomes 1 to 22. A) All samples (n = 2,706). B) Etanercept-treated patients (n = 733). C) Infliximab-treated patients (n = 894). D) Adalimumab-treated patients (n = 1,071).
Figure 2. Association results and SNP annotations…
Figure 2. Association results and SNP annotations in the 1q23 CD84 locus.
A) Regional association plots with ΔDAS (top panel) and with CD84 expression (bottom panel), showing strengths of association (−Log10 P-value) versus position (Kb) along chromosome 1. B) Schematic of CD84 gene structure (RefSeq gene model, box exons connected by diagonal lines, arrow indicates direction of transcription) with strong enhancer chromatin states (orange rectangles) and SNPs in high LD (r2>0.8) with rs6427528 (vertical ticks). SNPs in enhancers are labeled below. C) Annotations of strong-enhancer rs6427528 proxy SNPs; listed are SNP rs-ID (major and minor alleles), conservation score, cell line with DNAse footprint if present, and transcription factor binding sites altered. 1- Genomic evolutionary rate profiling (GERP) conservation score, where a score >2 indicates conservation across mammals. 2- DNase footprint data are compiled from publicly available experiments by HaploReg. 3- Position weight matrix logos show transcription factor consensus binding sites with nucleotide bases proportional to binding importance. SNP position is boxed. Note that the rs10797077 AIRE_2 and the rs6427528 SREBP_4 motifs are on the minus strand (base complements correspond to SNP alleles), with the SREBP motif shown upside down to align with the rs6427528 KROX motif on the positive strand. Data are from HaploReg.
Figure 3. 1q23/CD84 genotype association plots for…
Figure 3. 1q23/CD84 genotype association plots for ΔDAS and CD84 gene expression.
Shown are ΔDAS in our GWAS in etanercept-treated patients (top panel, n = 733; n = 634 with the GG genotype and n = 99 with the GA or AA genotype) and CD84 expression in our eQTL results (bottom panel, n = 228 non-RA patients; n = 178 with the GG genotype and n = 50 with the GA or AA genotype). The rare-allele homozygous genotype AA was observed four times in our ΔDAS GWAS and was pooled with the heterozygous GA genotype for this figure; AA homozygotes were not observed in the CD84 eQTL data. Association analyses reported in the text regressed phenotype (ΔDAS, P = 8×10−8; CD84 expression, P = 1×10−11) on minor-allele dosage (range 0–2).
Figure 4. CD84 expression level and clinical…
Figure 4. CD84 expression level and clinical features.
Analyses are shown in RA patients from the BRASS and ABCoN registries, for baseline DAS (top panel, n = 210; R2 = 0.02, p = 0.02) and ΔDAS (bottom panel, n = 31; R2 = 0.001, p = 0.46). Best-fit linear regression lines are shown in black, with shaded regions showing linear regression model (slope and intercept) 95% confidence intervals. CD84 expression levels were quantile normalized, and ΔDAS values were adjusted for age, gender and baseline DAS.
Figure 5. Replication and overall results for…
Figure 5. Replication and overall results for the CD84 SNP rs6427528.
Forest plot shows each cohort, sample size and linear regression beta coefficient estimates with symbol size proportional to cohort sample size and thin horizontal lines showing beta 95% CIs. Inverse variance weighted meta-analysis results are shown in bold for GWAS, GWAS+European (Portuguese) replication samples, and for GWAS+European+Asian (Japanese) replication samples, with diamond widths indicating beta 95% CIs.

References

    1. Klareskog L, Catrina AI, Paget S (2009) Rheumatoid arthritis. Lancet 373: 659–672.
    1. Scott DL, Wolfe F, Huizinga TW (2010) Rheumatoid arthritis. Lancet 376: 1094–1108.
    1. McInnes IB, Schett G (2011) The pathogenesis of rheumatoid arthritis. N Engl J Med 365: 2205–2219.
    1. Aeberli D, Seitz M, Juni P, Villiger PM (2005) Increase of peripheral CXCR3 positive T lymphocytes upon treatment of RA patients with TNF-alpha inhibitors. Rheumatology (Oxford) 44: 172–175.
    1. Agnholt J, Dahlerup JF, Kaltoft K (2003) The effect of etanercept and infliximab on the production of tumour necrosis factor alpha, interferon-gamma and GM-CSF in in vivo activated intestinal T lymphocyte cultures. Cytokine 23: 76–85.
    1. Catrina AI, Trollmo C, af Klint E, Engstrom M, Lampa J, et al. (2005) Evidence that anti-tumor necrosis factor therapy with both etanercept and infliximab induces apoptosis in macrophages, but not lymphocytes, in rheumatoid arthritis joints: extended report. Arthritis Rheum 52: 61–72.
    1. Scallon BJ, Moore MA, Trinh H, Knight DM, Ghrayeb J (1995) Chimeric anti-TNF-alpha monoclonal antibody cA2 binds recombinant transmembrane TNF-alpha and activates immune effector functions. Cytokine 7: 251–259.
    1. Gudbrandsdottir S, Larsen R, Sorensen LK, Nielsen S, Hansen MB, et al. (2004) TNF and LT binding capacities in the plasma of arthritis patients: effect of etanercept treatment in juvenile idiopathic arthritis. Clin Exp Rheumatol 22: 118–124.
    1. Plant D, Prajapati R, Hyrich KL, Morgan AW, Wilson AG, et al. (2012) Replication of association of the PTPRC gene with response to anti-tumor necrosis factor therapy in a large UK cohort. Arthritis Rheum 64: 665–670.
    1. Prajapati R, Plant D, Barton A (2011) Genetic and genomic predictors of anti-TNF response. Pharmacogenomics 12: 1571–1585.
    1. Cui J, Saevarsdottir S, Thomson B, Padyukov L, van der Helm-Van Mil AH, et al. (2010) Rheumatoid arthritis risk allele PTPRC is also associated with response to anti-tumor necrosis factor alpha therapy. Arthritis Rheum 62: 1849–1861.
    1. Plant D, Bowes J, Potter C, Hyrich KL, Morgan AW, et al. (2011) Genome-wide association study of genetic predictors of anti-tumor necrosis factor treatment efficacy in rheumatoid arthritis identifies associations with polymorphisms at seven loci. Arthritis Rheum 63: 645–653.
    1. Liu C, Batliwalla F, Li W, Lee A, Roubenoff R, et al. (2008) Genome-wide association scan identifies candidate polymorphisms associated with differential response to anti-TNF treatment in rheumatoid arthritis. Mol Med 14: 575–581.
    1. Prevoo ML, van 't Hof MA, Kuper HH, van Leeuwen MA, van de Putte LB, et al. (1995) Modified disease activity scores that include twenty-eight-joint counts. Development and validation in a prospective longitudinal study of patients with rheumatoid arthritis. Arthritis Rheum 38: 44–48.
    1. van Gestel AM, Prevoo ML, van 't Hof MA, van Rijswijk MH, van de Putte LB, et al. (1996) Development and validation of the European League Against Rheumatism response criteria for rheumatoid arthritis. Comparison with the preliminary American College of Rheumatology and the World Health Organization/International League Against Rheumatism Criteria. Arthritis Rheum 39: 34–40.
    1. Ward LD, Kellis M (2012) HaploReg: a resource for exploring chromatin states, conservation, and regulatory motif alterations within sets of genetically linked variants. Nucleic Acids Res 40: D930–934.
    1. Ernst J, Kheradpour P, Mikkelsen TS, Shoresh N, Ward LD, et al. (2011) Mapping and analysis of chromatin state dynamics in nine human cell types. Nature 473: 43–49.
    1. Cooper GM, Stone EA, Asimenos G, Green ED, Batzoglou S, et al. (2005) Distribution and intensity of constraint in mammalian genomic sequence. Genome Res 15: 901–913.
    1. Soliman MM, Hyrich KL, Lunt M, Watson KD, Symmons DP, et al. (2012) Rituximab or a second anti-TNF therapy for rheumatoid arthritis patients who have failed their first anti-TNF? Comparative analysis from the British Society for Rheumatology Biologics Register. Arthritis Care Res (Hoboken)
    1. Tangye SG, Nichols KE, Hare NJ, van de Weerdt BC (2003) Functional requirements for interactions between CD84 and Src homology 2 domain-containing proteins and their contribution to human T cell activation. J Immunol 171: 2485–2495.
    1. Martin M, Romero X, de la Fuente MA, Tovar V, Zapater N, et al. (2001) CD84 functions as a homophilic adhesion molecule and enhances IFN-gamma secretion: adhesion is mediated by Ig-like domain 1. J Immunol 167: 3668–3676.
    1. Padyukov L, Lampa J, Heimburger M, Ernestam S, Cederholm T, et al. (2003) Genetic markers for the efficacy of tumour necrosis factor blocking therapy in rheumatoid arthritis. Ann Rheum Dis 62: 526–529.
    1. Coenen MJ, Enevold C, Barrera P, Schijvenaars MM, Toonen EJ, et al. (2010) Genetic variants in toll-like receptors are not associated with rheumatoid arthritis susceptibility or anti-tumour necrosis factor treatment outcome. PLoS ONE 5: e14326 doi:.
    1. Toonen EJ, Coenen MJ, Kievit W, Fransen J, Eijsbouts AM, et al. (2008) The tumour necrosis factor receptor superfamily member 1b 676T>G polymorphism in relation to response to infliximab and adalimumab treatment and disease severity in rheumatoid arthritis. Ann Rheum Dis 67: 1174–1177.
    1. Miceli-Richard C, Comets E, Verstuyft C, Tamouza R, Loiseau P, et al. (2008) A single tumour necrosis factor haplotype influences the response to adalimumab in rheumatoid arthritis. Ann Rheum Dis 67: 478–484.
    1. Canhao H, Faustino A, Martins F, Fonseca JE (2011) - the rheumatic diseases portuguese register. Acta Reumatol Port 36: 45–56.
    1. Okada Y, Terao C, Ikari K, Kochi Y, Ohmura K, et al. (2012) Meta-analysis identifies nine new loci associated with rheumatoid arthritis in the Japanese population. Nat Genet
    1. Stahl EA, Raychaudhuri S, Remmers EF, Xie G, Eyre S, et al. (2010) Genome-wide association study meta-analysis identifies seven new rheumatoid arthritis risk loci. Nat Genet 42: 508–514.
    1. Plenge RM, Seielstad M, Padyukov L, Lee AT, Remmers EF, et al. (2007) TRAF1-C5 as a Risk Locus for Rheumatoid Arthritis – A Genomewide Study. N Engl J Med 357: 1199–1209.
    1. Purcell S, Neale B, Todd-Brown K, Thomas L, Ferreira MA, et al. (2007) PLINK: a tool set for whole-genome association and population-based linkage analyses. Am J Hum Genet 81: 559–575.
    1. Price AL, Patterson NJ, Plenge RM, Weinblatt ME, Shadick NA, et al. (2006) Principal components analysis corrects for stratification in genome-wide association studies. Nat Genet 38: 904–909.
    1. Marchini J, Howie B, Myers S, McVean G, Donnelly P (2007) A new multipoint method for genome-wide association studies by imputation of genotypes. Nat Genet 39: 906–913.
    1. Stahl EA, Wegmann D, Trynka G, Gutierrez-Achury J, Do R, et al. (2012) Bayesian inference analyses of the polygenic architecture of rheumatoid arthritis. Nat Genet
    1. Gauthier SA, Glanz BI, Mandel M, Weiner HL (2006) A model for the comprehensive investigation of a chronic autoimmune disease: the multiple sclerosis CLIMB study. Autoimmun Rev 5: 532–536.
    1. Stegle O, Parts L, Durbin R, Winn J (2010) A Bayesian framework to account for complex non-genetic factors in gene expression levels greatly increases power in eQTL studies. PLoS Comput Biol 6: e1000770 doi:.
    1. De Jager PL, Jia X, Wang J, de Bakker PI, Ottoboni L, et al. (2009) Meta-analysis of genome scans and replication identify CD6, IRF8 and TNFRSF1A as new multiple sclerosis susceptibility loci. Nat Genet 41: 776–782.
    1. Stranger BE, Nica AC, Forrest MS, Dimas A, Bird CP, et al. (2007) Population genomics of human gene expression. Nat Genet 39: 1217–1224.
    1. Parker A, Izmailova ES, Narang J, Badola S, Le T, et al. (2007) Peripheral Blood Expression of Nuclear Factor-kappaB-Regulated Genes Is Associated with Rheumatoid Arthritis Disease Activity and Responds Differentially to Anti-Tumor Necrosis Factor-alpha versus Methotrexate. J Rheumatol 34: 1817–1822.
    1. Batliwalla FM, Baechler EC, Xiao X, Li W, Balasubramanian S, et al. (2005) Peripheral blood gene expression profiling in rheumatoid arthritis. Genes Immun 6: 388–397.
    1. Devlin B, Roeder K, Wasserman L (2001) Genomic control, a new approach to genetic-based association studies. Theor Popul Biol 60: 155–166.
    1. de Bakker PI, Ferreira MA, Jia X, Neale BM, Raychaudhuri S, et al. (2008) Practical aspects of imputation-driven meta-analysis of genome-wide association studies. Hum Mol Genet 17: R122–128.

Source: PubMed

3
Předplatit