Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Tracy O'Mara is active.

Publication


Featured researches published by Tracy O'Mara.


Cancer Epidemiology, Biomarkers & Prevention | 2008

Multiple Novel Prostate Cancer Predisposition Loci Confirmed by an International Study: The PRACTICAL Consortium

Zsofia Kote-Jarai; Douglas F. Easton; Janet L. Stanford; Elaine A. Ostrander; Johanna Schleutker; Sue A. Ingles; Daniel J. Schaid; Stephen N. Thibodeau; Thilo Dörk; David E. Neal; Angela Cox; Christiane Maier; Walter Vogel; Michelle Guy; Kenneth Muir; Artitaya Lophatananon; Mary-Anne Kedda; Amanda B. Spurdle; Suzanne K. Steginga; Esther M. John; Graham G. Giles; John L. Hopper; Pierre O. Chappuis; Pierre Hutter; William D. Foulkes; Nancy Hamel; Claudia A. Salinas; Joseph S. Koopmeiners; Danielle M. Karyadi; Bo Johanneson

A recent genome-wide association study found that genetic variants on chromosomes 3, 6, 7, 10, 11, 19 and X were associated with prostate cancer risk. We evaluated the most significant single-nucleotide polymorphisms (SNP) in these loci using a worldwide consortium of 13 groups (PRACTICAL). Blood DNA from 7,370 prostate cancer cases and 5,742 male controls was analyzed by genotyping assays. Odds ratios (OR) associated with each genotype were estimated using unconditional logistic regression. Six of the seven SNPs showed clear evidence of association with prostate cancer (P = 0.0007-P = 10−17). For each of these six SNPs, the estimated per-allele OR was similar to those previously reported and ranged from 1.12 to 1.29. One SNP on 3p12 (rs2660753) showed a weaker association than previously reported [per-allele OR, 1.08 (95% confidence interval, 1.00-1.16; P = 0.06) versus 1.18 (95% confidence interval, 1.06-1.31)]. The combined risks associated with each pair of SNPs were consistent with a multiplicative risk model. Under this model, and in combination with previously reported SNPs on 8q and 17q, these loci explain 16% of the familial risk of the disease, and men in the top 10% of the risk distribution have a 2.1-fold increased risk relative to general population rates. This study provides strong confirmation of these susceptibility loci in multiple populations and shows that they make an important contribution to prostate cancer risk prediction. (Cancer Epidemiol Biomarkers Prev 2008;17(8):2052–61)


Cancer Epidemiology, Biomarkers & Prevention | 2017

The OncoArray Consortium: a Network for Understanding the Genetic Architecture of Common Cancers.

Christopher I. Amos; Joe Dennis; Zhaoming Wang; Jinyoung Byun; Fredrick R. Schumacher; Simon A. Gayther; Graham Casey; David J. Hunter; Thomas A. Sellers; Stephen B. Gruber; Alison M. Dunning; Kyriaki Michailidou; Laura Fachal; Kimberly F. Doheny; Amanda B. Spurdle; Yafang Li; Xiangjun Xiao; Jane Romm; Elizabeth W. Pugh; Gerhard A. Coetzee; Dennis J. Hazelett; Stig E. Bojesen; Charlisse F. Caga-anan; Christopher A. Haiman; Ahsan Kamal; Craig Luccarini; Daniel C. Tessier; Daniel Vincent; Francois Bacot; David Van Den Berg

Background: Common cancers develop through a multistep process often including inherited susceptibility. Collaboration among multiple institutions, and funding from multiple sources, has allowed the development of an inexpensive genotyping microarray, the OncoArray. The array includes a genome-wide backbone, comprising 230,000 SNPs tagging most common genetic variants, together with dense mapping of known susceptibility regions, rare variants from sequencing experiments, pharmacogenetic markers, and cancer-related traits. Methods: The OncoArray can be genotyped using a novel technology developed by Illumina to facilitate efficient genotyping. The consortium developed standard approaches for selecting SNPs for study, for quality control of markers, and for ancestry analysis. The array was genotyped at selected sites and with prespecified replicate samples to permit evaluation of genotyping accuracy among centers and by ethnic background. Results: The OncoArray consortium genotyped 447,705 samples. A total of 494,763 SNPs passed quality control steps with a sample success rate of 97% of the samples. Participating sites performed ancestry analysis using a common set of markers and a scoring algorithm based on principal components analysis. Conclusions: Results from these analyses will enable researchers to identify new susceptibility loci, perform fine-mapping of new or known loci associated with either single or multiple cancers, assess the degree of overlap in cancer causation and pleiotropic effects of loci that have been identified for disease-specific risk, and jointly model genetic, environmental, and lifestyle-related exposures. Impact: Ongoing analyses will shed light on etiology and risk assessment for many types of cancer. Cancer Epidemiol Biomarkers Prev; 26(1); 126–35. ©2016 AACR.


PLOS ONE | 2011

The Obesity-Associated Polymorphisms FTO rs9939609 and MC4R rs17782313 and Endometrial Cancer Risk in Non-Hispanic White Women

Galina Lurie; Mia M. Gaudet; Amanda B. Spurdle; Michael E. Carney; Lynne R. Wilkens; Hannah P. Yang; Noel S. Weiss; Penelope M. Webb; Pamela J. Thompson; Keith Y. Terada; Veronica Wendy Setiawan; Timothy R. Rebbeck; Jennifer Prescott; Irene Orlow; Tracy O'Mara; Sara H. Olson; Steven A. Narod; Rayna K. Matsuno; Jolanta Lissowska; Xiaolin Liang; Douglas A. Levine; Loic Le Marchand; Laurence N. Kolonel; Brian E. Henderson; Montserrat Garcia-Closas; Jennifer A. Doherty; Immaculata De Vivo; Chu Chen; Louise A. Brinton; Mohammad Akbari

Overweight and obesity are strongly associated with endometrial cancer. Several independent genome-wide association studies recently identified two common polymorphisms, FTO rs9939609 and MC4R rs17782313, that are linked to increased body weight and obesity. We examined the association of FTO rs9939609 and MC4R rs17782313 with endometrial cancer risk in a pooled analysis of nine case-control studies within the Epidemiology of Endometrial Cancer Consortium (E2C2). This analysis included 3601 non-Hispanic white women with histologically-confirmed endometrial carcinoma and 5275 frequency-matched controls. Unconditional logistic regression models were used to assess the relation of FTO rs9939609 and MC4R rs17782313 genotypes to the risk of endometrial cancer. Among control women, both the FTO rs9939609 A and MC4R rs17782313 C alleles were associated with a 16% increased risk of being overweight (p = 0.001 and p = 0.004, respectively). In case-control analyses, carriers of the FTO rs9939609 AA genotype were at increased risk of endometrial carcinoma compared to women with the TT genotype [odds ratio (OR)  = 1.17; 95% confidence interval (CI): 1.03–1.32, p = 0.01]. However, this association was no longer apparent after adjusting for body mass index (BMI), suggesting mediation of the gene-disease effect through body weight. The MC4R rs17782313 polymorphism was not related to endometrial cancer risk (per allele OR = 0.98; 95% CI: 0.91–1.06; p = 0.68). FTO rs9939609 is a susceptibility marker for white non-Hispanic women at higher risk of endometrial cancer. Although FTO rs9939609 alone might have limited clinical or public health significance for identifying women at high risk for endometrial cancer beyond that of excess body weight, further investigation of obesity-related genetic markers might help to identify the pathways that influence endometrial carcinogenesis.


Gynecologic Oncology | 2010

Vascular endothelial growth factor gene polymorphisms and ovarian cancer survival

Felicity Lose; Christina M. Nagle; Tracy O'Mara; Jyotsna Batra; Kelly L. Bolton; Honglin Song; Susan J. Ramus; Aleksandra Gentry-Maharaj; Usha Menon; Simon A. Gayther; Paul Pharoah; Mary-Anne Kedda; Amanda B. Spurdle

OBJECTIVES We sought to evaluate the effect of polymorphisms in the VEGF (Vascular Endothelial Growth Factor) gene on overall survival in ovarian cancer patients. METHODS A sample of 319 women diagnosed with primary invasive epithelial ovarian cancer in Australia between 1985 and 1997, recruited as incident cases, were genotyped for four VEGF single nucleotide polymorphisms (three tagSNPs and one functional SNP) using the Sequenom MassARRAY platform. A SNP found to be associated with ovarian cancer survival in this sample set was then evaluated in two independent datasets in an attempt to replicate the association. RESULTS VEGF tagSNPs rs3025033 and rs2146323 were not associated with ovarian cancer survival in the Australian sample. Ovarian cancer patients homozygous for tagSNP rs833068 or the functional SNP rs2010963 displayed significantly shortened overall survival in the Australian sample (HR 2.09, 95% CI 1.16-3.78), an effect most apparent in the first 5years after diagnosis. This association was not replicated in two independent datasets. CONCLUSIONS Findings from this study provide no evidence that rs3025033 and rs2146323 VEGF polymorphisms are associated with ovarian cancer survival. Although homozygous carriers of the tagSNP rs833068 experienced significantly worse survival in our Australian dataset, we were unable to replicate this in two independent datasets.


Cancer Epidemiology, Biomarkers & Prevention | 2012

Genome-Wide Association Study Identifies a Possible Susceptibility Locus for Endometrial Cancer

Jirong Long; Wei Zheng; Yong-Bing Xiang; Felicity Lose; Deborah Thompson; Ian Tomlinson; Herbert Yu; Nicolas Wentzensen; Diether Lambrechts; Thilo Dörk; Natalia Dubrowinskaja; Marc T. Goodman; Helga B. Salvesen; Peter A. Fasching; Rodney J. Scott; Ryan J. Delahanty; Ying Zheng; Tracy O'Mara; Catherine S. Healey; Shirley Hodgson; Harvey A. Risch; Hannah P. Yang; Frédéric Amant; Nurzhan Turmanov; Anita Schwake; Galina Lurie; Jone Trovik; Matthias W. Beckmann; Katie A. Ashton; Bu-Tian Ji

Background: Genome-wide association studies (GWAS) have identified more than 100 genetic loci for various cancers. However, only one is for endometrial cancer. Methods: We conducted a three-stage GWAS including 8,492 endometrial cancer cases and 16,596 controls. After analyzing 585,963 single-nucleotide polymorphisms (SNP) in 832 cases and 2,682 controls (stage I) from the Shanghai Endometrial Cancer Genetics Study, we selected the top 106 SNPs for in silico replication among 1,265 cases and 5,190 controls from the Australian/British Endometrial Cancer GWAS (stage II). Nine SNPs showed results consistent in direction with stage I with P < 0.1. These nine SNPs were investigated among 459 cases and 558 controls (stage IIIa) and six SNPs showed a direction of association consistent with stages I and II. These six SNPs, plus two additional SNPs selected on the basis of linkage disequilibrium and P values in stage II, were investigated among 5,936 cases and 8,166 controls from an additional 11 studies (stage IIIb). Results: SNP rs1202524, near the CAPN9 gene on chromosome 1q42.2, showed a consistent association with endometrial cancer risk across all three stages, with ORs of 1.09 [95% confidence interval (CI), 1.03–1.16] for the A/G genotype and 1.17 (95% CI, 1.05–1.30) for the G/G genotype (P = 1.6 × 10−4 in combined analyses of all samples). The association was stronger when limited to the endometrioid subtype, with ORs (95% CI) of 1.11 (1.04–1.18) and 1.21 (1.08–1.35), respectively (P = 2.4 × 10−5). Conclusions: Chromosome 1q42.2 may host an endometrial cancer susceptibility locus. Impact: This study identified a potential genetic locus for endometrial cancer risk. Cancer Epidemiol Biomarkers Prev; 21(6); 980–7. ©2012 AACR.


Nature Genetics | 2016

Five endometrial cancer risk loci identified through genome-wide association analysis

Timothy Cheng; D Thompson; Tracy O'Mara; Jodie N. Painter; Dylan M. Glubb; Susanne Flach; Annabelle Lewis; Juliet D. French; Luke Freeman-Mills; David N. Church; Maggie Gorman; Lynn Martin; Shirley Hodgson; Penelope M. Webb; John Attia; Elizabeth G. Holliday; Mark McEvoy; Rodney J. Scott; Anjali K. Henders; Nicholas G. Martin; Grant W. Montgomery; Dale R. Nyholt; Shahana Ahmed; Catherine S. Healey; Mitul Shah; Joe Dennis; Peter A. Fasching; Matthias W. Beckmann; Alexander Hein; Arif B. Ekici

We conducted a meta-analysis of three endometrial cancer genome-wide association studies (GWAS) and two follow-up phases totaling 7,737 endometrial cancer cases and 37,144 controls of European ancestry. Genome-wide imputation and meta-analysis identified five new risk loci of genome-wide significance at likely regulatory regions on chromosomes 13q22.1 (rs11841589, near KLF5), 6q22.31 (rs13328298, in LOC643623 and near HEY2 and NCOA7), 8q24.21 (rs4733613, telomeric to MYC), 15q15.1 (rs937213, in EIF2AK4, near BMF) and 14q32.33 (rs2498796, in AKT1, near SIVA1). We also found a second independent 8q24.21 signal (rs17232730). Functional studies of the 13q22.1 locus showed that rs9600103 (pairwise r2 = 0.98 with rs11841589) is located in a region of active chromatin that interacts with the KLF5 promoter region. The rs9600103[T] allele that is protective in endometrial cancer suppressed gene expression in vitro, suggesting that regulation of the expression of KLF5, a gene linked to uterine development, is implicated in tumorigenesis. These findings provide enhanced insight into the genetic and biological basis of endometrial cancer.


Endocrine-related Cancer | 2016

CYP19A1 fine-mapping and Mendelian randomization: estradiol is causal for endometrial cancer

Deborah Thompson; Tracy O'Mara; Dylan M. Glubb; Jodie N. Painter; Timothy Cheng; Elizabeth Folkerd; Deborah Doody; Joe Dennis; Penelope M. Webb; Maggie Gorman; Lynn Martin; Shirley Hodgson; Kyriaki Michailidou; Jonathan Tyrer; Mel Maranian; Per Hall; Kamila Czene; Hatef Darabi; Jingmei Li; Peter A. Fasching; Alexander Hein; Matthias W. Beckmann; Arif B. Ekici; Thilo Dörk; Peter Hillemanns; Matthias Dürst; Ingo B. Runnebaum; Hui Zhao; Jeroen Depreeuw; Stefanie Schrauwen

Candidate gene studies have reported CYP19A1 variants to be associated with endometrial cancer and with estradiol (E2) concentrations. We analyzed 2937 single nucleotide polymorphisms (SNPs) in 6608 endometrial cancer cases and 37 925 controls and report the first genome wide-significant association between endometrial cancer and a CYP19A1 SNP (rs727479 in intron 2, P=4.8×10−11). SNP rs727479 was also among those most strongly associated with circulating E2 concentrations in 2767 post-menopausal controls (P=7.4×10−8). The observed endometrial cancer odds ratio per rs727479 A-allele (1.15, CI=1.11–1.21) is compatible with that predicted by the observed effect on E2 concentrations (1.09, CI=1.03–1.21), consistent with the hypothesis that endometrial cancer risk is driven by E2. From 28 candidate-causal SNPs, 12 co-located with three putative gene-regulatory elements and their risk alleles associated with higher CYP19A1 expression in bioinformatical analyses. For both phenotypes, the associations with rs727479 were stronger among women with a higher BMI (Pinteraction=0.034 and 0.066 respectively), suggesting a biologically plausible gene-environment interaction.


PLOS ONE | 2012

Genetic Association of the KLK4 Locus with Risk of Prostate Cancer

Felicity Lose; Srilakshmi Srinivasan; Tracy O'Mara; Louise Marquart; Suzanne K. Chambers; Robert A. Gardiner; Joanne F. Aitken; Amanda B. Spurdle; Jyotsna Batra; Judith A. Clements

The Kallikrein-related peptidase, KLK4, has been shown to be significantly overexpressed in prostate tumours in numerous studies and is suggested to be a potential biomarker for prostate cancer. KLK4 may also play a role in prostate cancer progression through its involvement in epithelial-mesenchymal transition, a more aggressive phenotype, and metastases to bone. It is well known that genetic variation has the potential to affect gene expression and/or various protein characteristics and hence we sought to investigate the possible role of single nucleotide polymorphisms (SNPs) in the KLK4 gene in prostate cancer. Assessment of 61 SNPs in the KLK4 locus (±10 kb) in approximately 1300 prostate cancer cases and 1300 male controls for associations with prostate cancer risk and/or prostate tumour aggressiveness (Gleason score <7 versus ≥7) revealed 7 SNPs to be associated with a decreased risk of prostate cancer at the Ptrend<0.05 significance level. Three of these SNPs, rs268923, rs56112930 and the HapMap tagSNP rs7248321, are located several kb upstream of KLK4; rs1654551 encodes a non-synonymous serine to alanine substitution at position 22 of the long isoform of the KLK4 protein, and the remaining 3 risk-associated SNPs, rs1701927, rs1090649 and rs806019, are located downstream of KLK4 and are in high linkage disequilibrium with each other (r2≥0.98). Our findings provide suggestive evidence of a role for genetic variation in the KLK4 locus in prostate cancer predisposition.


International Journal of Gynecological Cancer | 2010

Kallikrein-related peptidase 10 (KLK10) expression and single nucleotide polymorphisms in ovarian cancer survival.

Jyotsna Batra; Olivia L. Tan; Tracy O'Mara; Rebecca Zammit; Christina M. Nagle; Judith A. Clements; Mary-Anne Kedda; Amanda B. Spurdle

Introduction: Kallikrein-related peptidase 10 (KLK10) overexpression is a predictor of poor disease outcome in women with late-stage ovarian cancer. We aimed to identify whether KLK10 overexpression could be attributed to genetic variants, in particular, in hormone response elements or transcription factor binding sites. Methods: Cox regression analysis was used to assess the association between 2 tag and 1 exonic KLK10 single nucleotide polymorphisms (SNPs) and the survival of 319 patients with ovarian cancer. Four different ovarian cancer cell lines were investigated for KLK10 expression after hormone stimulation, and sequence variation in the 3.6-Kb upstream of the KLK10 start site. In silico analyses of SNPs in cell lines and from published databases were undertaken to identify further research novel and potentially functional SNPs that are not covered by tag SNPs. Results: The KLK10 SNPs investigated were not associated with ovarian cancer survival. However, steroid hormone treatment of ovarian cell lines showed KLK10 up-regulation in response to estrogen and estrogen plus progesterone treatments in the aggressive cell line PEO1 and affirmed a role for KLK10 in aggressive ovarian cancer. Potentially functional KLK10 SNPs were identified by cell line sequencing and bioinformatic analysis. Conclusion: Potentially functional candidate KLK10 SNPs require investigation in future association studies of ovarian cancer risk and survival, including rs3760738 identified in aggressive ovarian cancer cell lines and predicted to affect transcription factor binding sites.


Cancer Epidemiology, Biomarkers & Prevention | 2010

No Association between FTO or HHEX and Endometrial Cancer Risk

Mia M. Gaudet; Hannah P. Yang; Jesus Gonzalez Bosquet; Catherine S. Healey; Shahana Ahmed; Alison M. Dunning; Doug Easton; Amanda B. Spurdle; Kaltin Ferguson; Tracy O'Mara; Diether Lambrechts; Evelyn Despierre; Ignace Vergote; Frédéric Amant; James V. Lacey; J Lissowska; Beata Peplonska; Louise A. Brinton; Stephen J. Chanock; Montserrat Garcia-Closas

Introduction: Obesity and diabetes are known risk factors for endometrial cancer; thus, the genetic risk factors of these phenotypes might also be associated with endometrial cancer risk. To evaluate this hypothesis, we genotyped tag-single nucleotide polymorphisms (SNP) and candidate SNPs in FTO and HHEX in a primary set of 417 endometrial cancer cases and 406 population-based controls, and validated significant findings in a replication set of approximately 2,347 cases and 3,140 controls from three additional studies. Methods: We genotyped 189 tagSNPs in FTO (including rs8050136) and five tagSNPs in HHEX (including rs1111875) in the primary set and one SNP each in FTO (rs12927155) and HHEX (rs1111875) in the validation set. Per allele odds ratios (OR) and 95% confidence intervals (CI) were calculated to estimate the association between the genotypes of each SNPs (as an ordinal variable) and endometrial cancer risk using unconditional logistic regression models, controlling for age and site. Results: In the primary study, the most significant finding in FTO was rs12927155 (OR, 1.56; 95% CI, 1.21-2.01; P = 5.8 × 10−4), and in HHEX, it was rs1111875 (OR, 0.80; 95% CI, 0.66-0.97; P = 0.026). In the validation studies, the pooled per allele OR, adjusted for age and study for FTO, was rs12927155 (OR, 0.94; 95% CI, 0.83-1.06; P = 0.29), whereas for HHEX, it was rs1111875 (OR, 1.00; 95% CI, 0.92-1.10; P = 0.96). Conclusion: Our data indicate that common genetic variants in two genes previously related to obesity (FTO) and diabetes (HHEX) by genome-wide association scans were not associated with endometrial cancer risk. Impact: Polymorphisms in FTO and HHEX are unlikely to have large effects on endometrial cancer risk but may have weaker effects. Cancer Epidemiol Biomarkers Prev; 19(8); 2106–9. ©2010 AACR.

Collaboration


Dive into the Tracy O'Mara's collaboration.

Top Co-Authors

Avatar

Amanda B. Spurdle

QIMR Berghofer Medical Research Institute

View shared research outputs
Top Co-Authors

Avatar

Judith A. Clements

Queensland University of Technology

View shared research outputs
Top Co-Authors

Avatar

Jyotsna Batra

Queensland University of Technology

View shared research outputs
Top Co-Authors

Avatar

Penelope M. Webb

QIMR Berghofer Medical Research Institute

View shared research outputs
Top Co-Authors

Avatar

Mary-Anne Kedda

Queensland University of Technology

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Thilo Dörk

Hannover Medical School

View shared research outputs
Top Co-Authors

Avatar

Felicity Lose

QIMR Berghofer Medical Research Institute

View shared research outputs
Top Co-Authors

Avatar

Louise Marquart

QIMR Berghofer Medical Research Institute

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge