Meta-analysis of three genome-wide association studies identifies two loci that predict survival and treatment outcome in breast cancer

The majority of breast cancers are driven by the female hormone oestrogen via oestrogen receptor (ER) alpha. ER-positive patients are commonly treated with adjuvant endocrine therapy, however, resistance is a common occurrence and aside from ER-status, no unequivocal predictive biomarkers are currently in clinical use. In this study, we aimed to identify constitutional genetic variants influencing breast cancer survival among ER-positive patients and specifically, among endocrine-treated patients. We conducted a meta-analysis of three genome-wide association studies comprising in total 3,136 patients with ER-positive breast cancer of which 2,751 had received adjuvant endocrine therapy. We identified a novel locus (rs992531 at 8p21.2) associated with reduced survival among the patients with ER-positive breast cancer (P = 3.77 × 10−8). Another locus (rs7701292 at 5q21.3) was associated with reduced survival among the endocrine-treated patients (P = 2.13 × 10−8). Interaction analysis indicated that the survival association of rs7701292 is treatment-specific and independent of conventional prognostic markers. In silico functional studies suggest plausible biological mechanisms for the observed survival associations and a functional link between the putative target genes of the rs992531 and rs7701292 (RHOBTB2 and RAB9P1, respectively). We further explored the genetic interaction between rs992531 and rs7701292 and found a significant, treatment-specific interactive effect on survival among ER-positive, endocrine-treated patients (hazard ratio = 6.97; 95% confidence interval, 1.79–27.08, Pinteraction = 0.036). This is the first study to identify a genetic interaction that specifically predicts treatment outcome. These findings may provide predictive biomarkers based on germ line genotype informing more personalized treatment selection.

[1]  Sofia Khan,et al.  Polymorphism at 19q13.41 Predicts Breast Cancer Survival Specifically after Endocrine Therapy , 2015, Clinical Cancer Research.

[2]  Peter Kraft,et al.  Identification of Novel Genetic Markers of Breast Cancer Survival , 2015, Journal of the National Cancer Institute.

[3]  Qian Zhu,et al.  Tissue-aware data integration approach for the inference of pathway interactions in metazoan organisms , 2015, Bioinform..

[4]  P. Fasching,et al.  The SNP rs6500843 in 16p13.3 is associated with survival specifically among chemotherapy-treated breast cancer patients , 2015, Oncotarget.

[5]  F. Couch,et al.  A Genome Wide Meta-Analysis Study for Identification of Common Variation Associated with Breast Cancer Prognosis , 2014, PloS one.

[6]  Yan Zhang,et al.  Comparative analysis of pseudogenes across three phyla , 2014, Proceedings of the National Academy of Sciences.

[7]  Y. Wu,et al.  Regulation of autophagy by the Rab GTPase network , 2014, Cell Death and Differentiation.

[8]  Masato Kimura,et al.  NCBI’s Database of Genotypes and Phenotypes: dbGaP , 2013, Nucleic Acids Res..

[9]  Andrea Sottoriva,et al.  The shaping and functional consequences of the microRNA landscape in breast cancer , 2013, Nature.

[10]  F. Couch,et al.  Identification of inherited genetic variations influencing prognosis in early-onset breast cancer. , 2013, Cancer research.

[11]  M. Seabra,et al.  Novel functions for Rab GTPases in multiple aspects of tumour progression , 2012, Biochemical Society transactions.

[12]  S. Hamada,et al.  The small GTPases Rab9A and Rab23 function at distinct steps in autophagy during Group A Streptococcus infection , 2012, Cellular microbiology.

[13]  Raymond K. Auerbach,et al.  An Integrated Encyclopedia of DNA Elements in the Human Genome , 2012, Nature.

[14]  F. Markowetz,et al.  The genomic and transcriptomic architecture of 2,000 breast tumours reveals novel subgroups , 2012, Nature.

[15]  Zoltan Szallasi,et al.  Jetset: selecting the optimal microarray probe set to represent a gene , 2011, BMC Bioinformatics.

[16]  Manolis Kellis,et al.  HaploReg: a resource for exploring chromatin states, conservation, and regulatory motif alterations within sets of genetically linked variants , 2011, Nucleic Acids Res..

[17]  Yong-mei Yang,et al.  RhoBTB2 (DBC2) functions as tumor suppressor via inhibiting proliferation, preventing colony formation and inducing apoptosis in breast cancer cells. , 2011, Gene.

[18]  Anbupalam Thalamuthu,et al.  A combined analysis of genome-wide association studies in breast cancer , 2011, Breast Cancer Research and Treatment.

[19]  Rachel Schiff,et al.  Mechanisms of endocrine resistance in breast cancer. , 2011, Annual review of medicine.

[20]  Avi Ma'ayan,et al.  ChEA: transcription factor regulation inferred from integrating genome-wide ChIP-X experiments , 2010, Bioinform..

[21]  Z. Szallasi,et al.  An online survival analysis tool to rapidly assess the effect of 22,277 genes on breast cancer prognosis using microarray data of 1,809 patients , 2010, Breast Cancer Research and Treatment.

[22]  P. Pandolfi,et al.  A coding-independent function of gene and pseudogene mRNAs regulates tumour biology , 2010, Nature.

[23]  Xun Qu,et al.  A novel tumor suppressor gene RhoBTB2 (DBC2): Frequent loss of expression in sporadic breast cancer , 2010, Molecular carcinogenesis.

[24]  P. Schoenlein,et al.  Autophagy facilitates the progression of ERα-positive breast cancer cells to antiestrogen resistance , 2009, Autophagy.

[25]  F. Rivero,et al.  Characterization of RhoBTB-dependent Cul3 ubiquitin ligase complexes--evidence for an autoregulatory mechanism. , 2008, Experimental cell research.

[26]  Manuel A. R. Ferreira,et al.  PLINK: a tool set for whole-genome association and population-based linkage analyses. , 2007, American journal of human genetics.

[27]  D. Altman,et al.  Prospective study of Outcomes in Sporadic versus Hereditary breast cancer (POSH): study protocol , 2007, BMC Cancer.

[28]  Takashi Yoshihara,et al.  Cyclin D1 down-regulation is essential for DBC2's tumor suppressor function. , 2007, Biochemical and biophysical research communications.

[29]  Päivi Heikkilä,et al.  Correlation of CHEK2 protein expression and c.1100delC mutation status with tumor characteristics among unselected breast cancer patients , 2005, International journal of cancer.

[30]  K Holli,et al.  Population-based study of BRCA1 and BRCA2 mutations in 1035 unselected Finnish breast cancer patients. , 2000, Journal of the National Cancer Institute.

[31]  H. Nevanlinna,et al.  Familial breast cancer in southern Finland: how prevalent are breast cancer families and can we trust the family history reported by patients? , 2000, European journal of cancer.

[32]  C K Osborne,et al.  Estrogen receptor status by immunohistochemistry is superior to the ligand-binding assay for predicting response to adjuvant endocrine therapy in breast cancer. , 1999, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[33]  R. Bernards,et al.  CDK-Independent Activation of Estrogen Receptor by Cyclin D1 , 1997, Cell.

[34]  Massimo Cristofanilli,et al.  Palbociclib in Hormone-Receptor-Positive Advanced Breast Cancer. , 2015, The New England journal of medicine.

[35]  Melissa C. Greven,et al.  An integrated encyclopedia of DNA elements in the human genome , 2014 .

[36]  Tanya M. Teslovich,et al.  LocusZoom: regional visualization of genome-wide association scan results , 2010, Bioinform..

[37]  H. Nevanlinna,et al.  Cyclin D1 expression is associated with poor prognostic features in estrogen receptor positive breast cancer , 2008, Breast Cancer Research and Treatment.

[38]  Robert Clarke,et al.  Antiestrogens, aromatase inhibitors, and apoptosis in breast cancer. , 2005, Vitamins and hormones.

[39]  Yusuke Nakamura,et al.  A genome-wide association study identifies locus at 10q22 associated with clinical outcomes of adjuvant tamoxifen therapy for breast cancer patients in Japanese , 2022 .