Dr Zsofia Kote-Jarai

Senior Staff Scientist:

Email: [email protected]

Location: Sutton

Email: [email protected]

Location: Sutton

Biography and research overview

Dr Zsofia Kote-Jarai is a senior staff scientist in the Division of Genetics and Epidemiology. Dr Kote-Jarai has led and supported numerous research projects with the aim of identifying key elements of genetic predisposition to prostate cancer. Alongside Professor Ros Eeles and many other UK and international colleagues in the PRACTICAL Consortium, she has coordinated a large multistage genome-wide association study (GWAS) which has identified many common genetic variants affecting prostate cancer risk and led to several high-impact publications.

Dr Kote-Jarai joined The Institute of Cancer Research, London, in 1998 as a postdoctoral fellow, before becoming a staff scientist and, since 2009, has led projects as a senior staff scientist.

Dr Kote-Jarai’s work has focused on identifying common genetic variants affecting the risk of prostate cancer. She coordinated a large multistage GWAS which identified many common genetic variants and led to a major publication in Nature Genetics. She is currently leading projects using fine-mapping and pathway analysis to better understand the molecular basis through which these newly identified genetic variants modulate prostate cancer risk.

To identify rarer germline susceptibility variants, the group is using targeted next-generation sequencing (NGS) approaches, either by sequencing whole exomes or a selected set of candidate genes in cohorts enriched for family history of prostate cancer or advanced disease. The group has previously found that the breast cancer gene BRCA2 also predisposes to prostate cancer and is now extending this observation to investigate whether germline mutations in additional genes in related pathways also predispose to the disease. This project also seeks to establish whether specific mutations give rise to particular disease phenotypes; especially whether certain germline mutational signatures can be correlated with predisposition to more aggressive disease.

Currently Dr Kote-Jarai is one of the leaders of a large international GWAS follow-up study (OncoArray), analysing 600,000 variants in approximately 100,000 samples, which will identify further common predisposition variants. These, alongside the rare variants being discovered by the NGS studies, will have the potential to explain a significant proportion of prostate cancer heritability, and ultimately could enable targeted screening and intervention strategies to identify men who are at higher risk of developing clinically significant disease.

Types of Publications

Journal articles

Eeles, R.A. Olama, A.A.A. Benlloch, S. Saunders, E.J. Leongamornlert, D.A. Tymrakiewicz, M. Ghoussaini, M. Luccarini, C. Dennis, J. Jugurnauth-Little, S. Dadaev, T. Neal, D.E. Hamdy, F.C. Donovan, J.L. Muir, K. Giles, G.G. Severi, G. Wiklund, F. Gronberg, H. Haiman, C.A. Schumacher, F. Henderson, B.E. Le Marchand, L. Lindstrom, S. Kraft, P. Hunter, D.J. Gapstur, S. Chanock, S.J. Berndt, S.I. Albanes, D. Andriole, G. Schleutker, J. Weischer, M. Canzian, F. Riboli, E. Key, T.J. Travis, R.C. Campa, D. Ingles, S.A. John, E.M. Hayes, R.B. Pharoah, P.D.P. Pashayan, N. Khaw, K.-.T. Stanford, J.L. Ostrander, E.A. Signorello, L.B. Thibodeau, S.N. Schaid, D. Maier, C. Vogel, W. Kibel, A.S. Cybulski, C. Lubinski, J. Cannon-Albright, L. Brenner, H. Park, J.Y. Kaneva, R. Batra, J. Spurdle, A.B. Clements, J.A. Teixeira, M.R. Dicks, E. Lee, A. Dunning, A.M. Baynes, C. Conroy, D. Maranian, M.J. Ahmed, S. Govindasami, K. Guy, M. Wilkinson, R.A. Sawyer, E.J. Morgan, A. Dearnaley, D.P. Horwich, A. Huddart, R.A. Khoo, V.S. Parker, C.C. Van As, N.J. Woodhouse, C.J. Thompson, A. Dudderidge, T. Ogden, C. Cooper, C.S. Lophatananon, A. Cox, A. Southey, M.C. Hopper, J.L. English, D.R. Aly, M. Adolfsson, J. Xu, J. Zheng, S.L. Yeager, M. Kaaks, R. Diver, W.R. Gaudet, M.M. Stern, M.C. Corral, R. Joshi, A.D. Shahabi, A. Wahlfors, T. Tammela, T.L.J. Auvinen, A. Virtamo, J. Klarskov, P. Nordestgaard, B.G. Røder, M.A. Nielsen, S.F. Bojesen, S.E. Siddiq, A. Fitzgerald, L.M. Kolb, S. Kwon, E.M. Karyadi, D.M. Blot, W.J. Zheng, W. Cai, Q. McDonnell, S.K. Rinckleb, A.E. Drake, B. Colditz, G. Wokolorczyk, D. Stephenson, R.A. Teerlink, C. Muller, H. Rothenbacher, D. Sellers, T.A. Lin, H.-.Y. Slavov, C. Mitev, V. Lose, F. Srinivasan, S. Maia, S. Paulo, P. Lange, E. Cooney, K.A. Antoniou, A.C. Vincent, D. Bacot, F. Tessier, D.C. COGS–Cancer Research UK GWAS–ELLIPSE (part of GAME-ON) Initiative, . Australian Prostate Cancer Bioresource, . UK Genetic Prostate Cancer Study Collaborators/British Association of Urological Surgeons' Section of Oncology, . UK ProtecT (Prostate testing for cancer and Treatment) Study Collaborators, . PRACTICAL (Prostate Cancer Association Group to Investigate Cancer-Associated Alterations in the Genome) Consortium, . Kote-Jarai, Z. Easton, D.F (2013) Identification of 23 new prostate cancer susceptibility loci using the iCOGS custom genotyping array.. Show Abstract full text

Prostate cancer is the most frequently diagnosed cancer in males in developed countries. To identify common prostate cancer susceptibility alleles, we genotyped 211,155 SNPs on a custom Illumina array (iCOGS) in blood DNA from 25,074 prostate cancer cases and 24,272 controls from the international PRACTICAL Consortium. Twenty-three new prostate cancer susceptibility loci were identified at genome-wide significance (P < 5 × 10(-8)). More than 70 prostate cancer susceptibility loci, explaining ∼30% of the familial risk for this disease, have now been identified. On the basis of combined risks conferred by the new and previously known risk loci, the top 1% of the risk distribution has a 4.7-fold higher risk than the average of the population being profiled. These results will facilitate population risk stratification for clinical studies.

Kote-Jarai, Z. Saunders, E.J. Leongamornlert, D.A. Tymrakiewicz, M. Dadaev, T. Jugurnauth-Little, S. Ross-Adams, H. Al Olama, A.A. Benlloch, S. Halim, S. Russell, R. Dunning, A.M. Luccarini, C. Dennis, J. Neal, D.E. Hamdy, F.C. Donovan, J.L. Muir, K. Giles, G.G. Severi, G. Wiklund, F. Gronberg, H. Haiman, C.A. Schumacher, F. Henderson, B.E. Le Marchand, L. Lindstrom, S. Kraft, P. Hunter, D.J. Gapstur, S. Chanock, S. Berndt, S.I. Albanes, D. Andriole, G. Schleutker, J. Weischer, M. Canzian, F. Riboli, E. Key, T.J. Travis, R.C. Campa, D. Ingles, S.A. John, E.M. Hayes, R.B. Pharoah, P. Khaw, K.-.T. Stanford, J.L. Ostrander, E.A. Signorello, L.B. Thibodeau, S.N. Schaid, D. Maier, C. Vogel, W. Kibel, A.S. Cybulski, C. Lubinski, J. Cannon-Albright, L. Brenner, H. Park, J.Y. Kaneva, R. Batra, J. Spurdle, A. Clements, J.A. Teixeira, M.R. Govindasami, K. Guy, M. Wilkinson, R.A. Sawyer, E.J. Morgan, A. Dicks, E. Baynes, C. Conroy, D. Bojesen, S.E. Kaaks, R. Vincent, D. Bacot, F. Tessier, D.C. COGS-CRUK GWAS-ELLIPSE (Part of GAME-ON) Initiative, . UK Genetic Prostate Cancer Study Collaborators/British Association of Urological Surgeons' Section of Oncology, . UK ProtecT Study Collaborators, . PRACTICAL Consortium, . Easton, D.F. Eeles, R.A (2013) Fine-mapping identifies multiple prostate cancer risk loci at 5p15, one of which associates with TERT expression.. Show Abstract full text

Associations between single nucleotide polymorphisms (SNPs) at 5p15 and multiple cancer types have been reported. We have previously shown evidence for a strong association between prostate cancer (PrCa) risk and rs2242652 at 5p15, intronic in the telomerase reverse transcriptase (TERT) gene that encodes TERT. To comprehensively evaluate the association between genetic variation across this region and PrCa, we performed a fine-mapping analysis by genotyping 134 SNPs using a custom Illumina iSelect array or Sequenom MassArray iPlex, followed by imputation of 1094 SNPs in 22 301 PrCa cases and 22 320 controls in The PRACTICAL consortium. Multiple stepwise logistic regression analysis identified four signals in the promoter or intronic regions of TERT that independently associated with PrCa risk. Gene expression analysis of normal prostate tissue showed evidence that SNPs within one of these regions also associated with TERT expression, providing a potential mechanism for predisposition to disease.

Types of Publications

Journal articles

Eeles, R.A. Kote-Jarai, Z. Giles, G.G. Olama, A.A.A. Guy, M. Jugurnauth, S.K. Mulholland, S. Leongamornlert, D.A. Edwards, S.M. Morrison, J. Field, H.I. Southey, M.C. Severi, G. Donovan, J.L. Hamdy, F.C. Dearnaley, D.P. Muir, K.R. Smith, C. Bagnato, M. Ardern-Jones, A.T. Hall, A.L. O'Brien, L.T. Gehr-Swain, B.N. Wilkinson, R.A. Cox, A. Lewis, S. Brown, P.M. Jhavar, S.G. Tymrakiewicz, M. Lophatananon, A. Bryant, S.L. UK Genetic Prostate Cancer Study Collaborators, . British Association of Urological Surgeons' Section of Oncology, . UK ProtecT Study Collaborators, . Horwich, A. Huddart, R.A. Khoo, V.S. Parker, C.C. Woodhouse, C.J. Thompson, A. Christmas, T. Ogden, C. Fisher, C. Jamieson, C. Cooper, C.S. English, D.R. Hopper, J.L. Neal, D.E. Easton, D.F (2008) Multiple newly identified loci associated with prostate cancer susceptibility.. Show Abstract full text

Prostate cancer is the most common cancer affecting males in developed countries. It shows consistent evidence of familial aggregation, but the causes of this aggregation are mostly unknown. To identify common alleles associated with prostate cancer risk, we conducted a genome-wide association study (GWAS) using blood DNA samples from 1,854 individuals with clinically detected prostate cancer diagnosed at </=60 years or with a family history of disease, and 1,894 population-screened controls with a low prostate-specific antigen (PSA) concentration (<0.5 ng/ml). We analyzed these samples for 541,129 SNPs using the Illumina Infinium platform. Initial putative associations were confirmed using a further 3,268 cases and 3,366 controls. We identified seven loci associated with prostate cancer on chromosomes 3, 6, 7, 10, 11, 19 and X (P = 2.7 x 10(-8) to P = 8.7 x 10(-29)). We confirmed previous reports of common loci associated with prostate cancer at 8q24 and 17q. Moreover, we found that three of the newly identified loci contain candidate susceptibility genes: MSMB, LMTK2 and KLK3.

Kote-Jarai, Z. Easton, D.F. Stanford, J.L. Ostrander, E.A. Schleutker, J. Ingles, S.A. Schaid, D. Thibodeau, S. Dörk, T. Neal, D. Donovan, J. Hamdy, F. Cox, A. Maier, C. Vogel, W. Guy, M. Muir, K. Lophatananon, A. Kedda, M.-.A. Spurdle, A. Steginga, S. John, E.M. Giles, G. Hopper, J. Chappuis, P.O. Hutter, P. Foulkes, W.D. Hamel, N. Salinas, C.A. Koopmeiners, J.S. Karyadi, D.M. Johanneson, B. Wahlfors, T. Tammela, T.L. Stern, M.C. Corral, R. McDonnell, S.K. Schürmann, P. Meyer, A. Kuefer, R. Leongamornlert, D.A. Tymrakiewicz, M. Liu, J.-.F. O'Mara, T. Gardiner, R.A.F. Aitken, J. Joshi, A.D. Severi, G. English, D.R. Southey, M. Edwards, S.M. Al Olama, A.A. PRACTICAL Consortium, . Eeles, R.A (2008) Multiple novel prostate cancer predisposition loci confirmed by an international study: the PRACTICAL Consortium.. Show Abstract full text

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.

Al Olama, A.A. Kote-Jarai, Z. Giles, G.G. Guy, M. Morrison, J. Severi, G. Leongamornlert, D.A. Tymrakiewicz, M. Jhavar, S. Saunders, E. Hopper, J.L. Southey, M.C. Muir, K.R. English, D.R. Dearnaley, D.P. Ardern-Jones, A.T. Hall, A.L. O'Brien, L.T. Wilkinson, R.A. Sawyer, E. Lophatananon, A. UK Genetic Prostate Cancer Study Collaborators/British Association of Urological Surgeons' Section of Oncology, . UK Prostate testing for cancer and Treatment study (ProtecT Study) Collaborators, . Horwich, A. Huddart, R.A. Khoo, V.S. Parker, C.C. Woodhouse, C.J. Thompson, A. Christmas, T. Ogden, C. Cooper, C. Donovan, J.L. Hamdy, F.C. Neal, D.E. Eeles, R.A. Easton, D.F (2009) Multiple loci on 8q24 associated with prostate cancer susceptibility.. Show Abstract full text

Previous studies have identified multiple loci on 8q24 associated with prostate cancer risk. We performed a comprehensive analysis of SNP associations across 8q24 by genotyping tag SNPs in 5,504 prostate cancer cases and 5,834 controls. We confirmed associations at three previously reported loci and identified additional loci in two other linkage disequilibrium blocks (rs1006908: per-allele OR = 0.87, P = 7.9 x 10(-8); rs620861: OR = 0.90, P = 4.8 x 10(-8)). Eight SNPs in five linkage disequilibrium blocks were independently associated with prostate cancer susceptibility.

Eeles, R.A. Kote-Jarai, Z. Al Olama, A.A. Giles, G.G. Guy, M. Severi, G. Muir, K. Hopper, J.L. Henderson, B.E. Haiman, C.A. Schleutker, J. Hamdy, F.C. Neal, D.E. Donovan, J.L. Stanford, J.L. Ostrander, E.A. Ingles, S.A. John, E.M. Thibodeau, S.N. Schaid, D. Park, J.Y. Spurdle, A. Clements, J. Dickinson, J.L. Maier, C. Vogel, W. Dörk, T. Rebbeck, T.R. Cooney, K.A. Cannon-Albright, L. Chappuis, P.O. Hutter, P. Zeegers, M. Kaneva, R. Zhang, H.-.W. Lu, Y.-.J. Foulkes, W.D. English, D.R. Leongamornlert, D.A. Tymrakiewicz, M. Morrison, J. Ardern-Jones, A.T. Hall, A.L. O'Brien, L.T. Wilkinson, R.A. Saunders, E.J. Page, E.C. Sawyer, E.J. Edwards, S.M. Dearnaley, D.P. Horwich, A. Huddart, R.A. Khoo, V.S. Parker, C.C. Van As, N. Woodhouse, C.J. Thompson, A. Christmas, T. Ogden, C. Cooper, C.S. Southey, M.C. Lophatananon, A. Liu, J.-.F. Kolonel, L.N. Le Marchand, L. Wahlfors, T. Tammela, T.L. Auvinen, A. Lewis, S.J. Cox, A. FitzGerald, L.M. Koopmeiners, J.S. Karyadi, D.M. Kwon, E.M. Stern, M.C. Corral, R. Joshi, A.D. Shahabi, A. McDonnell, S.K. Sellers, T.A. Pow-Sang, J. Chambers, S. Aitken, J. Gardiner, R.A.F. Batra, J. Kedda, M.A. Lose, F. Polanowski, A. Patterson, B. Serth, J. Meyer, A. Luedeke, M. Stefflova, K. Ray, A.M. Lange, E.M. Farnham, J. Khan, H. Slavov, C. Mitkova, A. Cao, G. UK Genetic Prostate Cancer Study Collaborators/British Association of Urological Surgeons' Section of Oncology, . UK ProtecT Study Collaborators, . PRACTICAL Consortium, . Easton, D.F (2009) Identification of seven new prostate cancer susceptibility loci through a genome-wide association study.. Show Abstract full text

Prostate cancer (PrCa) is the most frequently diagnosed cancer in males in developed countries. To identify common PrCa susceptibility alleles, we previously conducted a genome-wide association study in which 541,129 SNPs were genotyped in 1,854 PrCa cases with clinically detected disease and in 1,894 controls. We have now extended the study to evaluate promising associations in a second stage in which we genotyped 43,671 SNPs in 3,650 PrCa cases and 3,940 controls and in a third stage involving an additional 16,229 cases and 14,821 controls from 21 studies. In addition to replicating previous associations, we identified seven new prostate cancer susceptibility loci on chromosomes 2, 4, 8, 11 and 22 (with P = 1.6 x 10(-8) to P = 2.7 x 10(-33)).

Kirby, R.S. Eeles, R.A. Kote-Jarai, Z. Guy, M. Easton, D. Fitzpatrick, J.M (2010) Screening for prostate cancer: the way ahead.. full text
Mitra, A.V. Bancroft, E.K. Barbachano, Y. Page, E.C. Foster, C.S. Jameson, C. Mitchell, G. Lindeman, G.J. Stapleton, A. Suthers, G. Evans, D.G. Cruger, D. Blanco, I. Mercer, C. Kirk, J. Maehle, L. Hodgson, S. Walker, L. Izatt, L. Douglas, F. Tucker, K. Dorkins, H. Clowes, V. Male, A. Donaldson, A. Brewer, C. Doherty, R. Bulman, B. Osther, P.J. Salinas, M. Eccles, D. Axcrona, K. Jobson, I. Newcombe, B. Cybulski, C. Rubinstein, W.S. Buys, S. Townshend, S. Friedman, E. Domchek, S. Ramon Y Cajal, T. Spigelman, A. Teo, S.H. Nicolai, N. Aaronson, N. Ardern-Jones, A. Bangma, C. Dearnaley, D. Eyfjord, J. Falconer, A. Grönberg, H. Hamdy, F. Johannsson, O. Khoo, V. Kote-Jarai, Z. Lilja, H. Lubinski, J. Melia, J. Moynihan, C. Peock, S. Rennert, G. Schröder, F. Sibley, P. Suri, M. Wilson, P. Bignon, Y.J. Strom, S. Tischkowitz, M. Liljegren, A. Ilencikova, D. Abele, A. Kyriacou, K. van Asperen, C. Kiemeney, L. IMPACT Study Collaborators, . Easton, D.F. Eeles, R.A (2011) Targeted prostate cancer screening in men with mutations in BRCA1 and BRCA2 detects aggressive prostate cancer: preliminary analysis of the results of the IMPACT study.. Show Abstract full text

OBJECTIVE: To evaluate the role of targeted prostate cancer screening in men with BRCA1 or BRCA2 mutations, an international study, IMPACT (Identification of Men with a genetic predisposition to ProstAte Cancer: Targeted screening in BRCA1/2 mutation carriers and controls), was established. This is the first multicentre screening study targeted at men with a known genetic predisposition to prostate cancer. A preliminary analysis of the data is reported. PATIENTS AND METHODS: Men aged 40-69 years from families with BRCA1 or BRCA2 mutations were offered annual prostate specific antigen (PSA) testing, and those with PSA > 3 ng/mL, were offered a prostate biopsy. Controls were men age-matched (± 5 years) who were negative for the familial mutation. RESULTS: In total, 300 men were recruited (205 mutation carriers; 89 BRCA1, 116 BRCA2 and 95 controls) over 33 months. At the baseline screen (year 1), 7.0% (21/300) underwent a prostate biopsy. Prostate cancer was diagnosed in ten individuals, a prevalence of 3.3%. The positive predictive value of PSA screening in this cohort was 47·6% (10/21). One prostate cancer was diagnosed at year 2. Of the 11 prostate cancers diagnosed, nine were in mutation carriers, two in controls, and eight were clinically significant. CONCLUSIONS: The present study shows that the positive predictive value of PSA screening in BRCA mutation carriers is high and that screening detects clinically significant prostate cancer. These results support the rationale for continued screening in such men.

Bancroft, E.K. Locke, I. Ardern-Jones, A. D'Mello, L. McReynolds, K. Lennard, F. Barbachano, Y. Barwell, J. Walker, L. Mitchell, G. Dorkins, H. Cummings, C. Paterson, J. Kote-Jarai, Z. Mitra, A. Jhavar, S. Thomas, S. Houlston, R. Shanley, S. Eeles, R.A (2010) The carrier clinic: an evaluation of a novel clinic dedicated to the follow-up of BRCA1 and BRCA2 carriers--implications for oncogenetics practice.. Show Abstract full text

BACKGROUND: A novel oncogenetic clinic was established in 2002 at the Royal Marsden NHS Foundation Trust offering advice and specialist follow-up for families with a germline mutation in BRCA1 or BRCA2. The remit of this multidisciplinary clinic, staffed by individuals in both oncology and genetics, is to provide individualised screening recommendations, support in decision making, risk reducing strategies, cascade testing, and an extensive research portfolio. METHODS: A retrospective analysis was performed to evaluate uptake of genetic testing, risk reducing surgery and cancer prevalence in 346 BRCA1/BRCA2 families seen between January 1996 and December 2006. RESULTS: 661 individuals attended the clinic and 406 mutation carriers were identified; 85.8% mutation carriers have chosen to attend for annual follow-up. 70% of mutation carriers elected for risk reducing bilateral salpingo-oophorectomy (RRBSO). 32% of unaffected women chose risk reducing bilateral mastectomy. 32% of women with breast cancer chose contralateral risk reducing mastectomy at time of diagnosis. Some women took over 8 years to decide to have surgery. 91% of individuals approached agreed to participate in research programmes. INTERPRETATION: A novel specialist clinic for BRCA1/2 mutation carriers has been successfully established. The number of mutation positive families is increasing. This, and the high demand for RRBSO in women over 40, is inevitably going to place an increasing demand on existing health resources. Our clinic model has subsequently been adopted in other centres and this will greatly facilitate translational studies and provide a healthcare structure for management and follow-up of such people who are at a high cancer risk.

Mitra, A.V. Jameson, C. Barbachano, Y. Sodha, N. Kote-Jarai, Z. Javed, A. Bancroft, E. Fletcher, A. Cooper, C. Peock, S. IMPACT and EMBRACE Collaborators, . Easton, D. Eeles, R. Foster, C.S (2010) Elevated expression of Ki-67 identifies aggressive prostate cancers but does not distinguish BRCA1 or BRCA2 mutation carriers.. Show Abstract full text

Prostate cancers in men with germline BRCA1 and BRCA2 mutations are more aggressive than morphologically similar cancers in men without these mutations. This study was performed to test the hypothesis that enhanced expression of Ki-67, as a surrogate of cell proliferation, is a characteristic feature of prostate cancers occurring in BRCA1 or BRCA2 mutation carriers. The study cohort comprised 20 cases of prostate cancer in mutation carriers and 126 control sporadic prostate cancers. Of the combined sample cohort, 65.7% stained only within malignant tissues while 0.7% stained in both malignant and benign tissues (p<0.001). Significantly increased expression of Ki-67 occurred in prostate cancers with higher Gleason score (p<0.001). Elevated Ki-67 expression was identified in 71% of prostate cancers in BRCA1 or BRCA2 mutation carriers and in 67% of the sporadic controls (p>0.5). Similar results were obtained when the data were analysed using a threshold set at 3.5 and 7.1%. This study shows that elevated expression of Ki-67 is associated both with aggressive prostate cancers and with high Gleason score irrespective of whether their occurrence is against a background of BRCA1 or BRCA2 mutations or as sporadic disease. The data suggest that, since elevated Ki-67 does not distinguish prostate cancers occurring in BRCA1 or BRCA2 mutation carriers from sporadic prostatic malignancies, the effects of these genetic mutations are probably independent. While all prostate cancers occurring in the presence of BRCA germline mutations are clinically aggressive, their potentially different phenotypes consistently involve maximal rates of cell proliferation.

Mitra, A. Jameson, C. Barbachano, Y. Sanchez, L. Kote-Jarai, Z. Peock, S. Sodha, N. Bancroft, E. Fletcher, A. Cooper, C. Easton, D. IMPACT Steering Committee and IMPACT and EMBRACE Collaborators, . Eeles, R. Foster, C.S (2009) Overexpression of RAD51 occurs in aggressive prostatic cancer.. Show Abstract full text

AIMS: To test the hypothesis that, in a matched series of prostatic cancers, either with or without BRCA1 or BRCA2 mutations, RAD51 protein expression is enhanced in association with BRCA mutation genotypes. METHODS AND RESULTS: RAD51 expression identified immunohistochemically was compared between prostatic cancers occurring in BRCA1 or BRCA2 mutation carriers and controls. RAD51 protein expression in the cytoplasm and nuclei of the benign tissues was significantly less than in the malignant tissues (P < 0.001). In all cancers, cytoplasmic expression of RAD51 was more prevalent and associated with higher Gleason score (P < 0.05) irrespective of BRCA mutational status, than its expression in benign tissues (P < 0.001). Although nuclear immunoreactivity was not observed in BRCA-associated cancers with Gleason score < or =7, it was significantly increased in all other groups of prostatic cancers when compared with benign tissues (P < 0.001). CONCLUSIONS: RAD51 protein is strongly expressed in high-grade prostatic cancers, whether sporadic or associated with BRCA germ-line mutations. Distinct localization of RAD51 between cytoplasm and nucleus, particularly in cancers of Gleason score < or =7, reflects distinct levels of RAD51 regulatory activity, from transcription to DNA repair. This biomarker may be of value in identifying patients requiring urgent treatment at diagnosis as well as in analysing biological mechanisms underlying aggressive phenotype of human prostatic cancer.

Guy, M. Kote-Jarai, Z. Giles, G.G. Al Olama, A.A. Jugurnauth, S.K. Mulholland, S. Leongamornlert, D.A. Edwards, S.M. Morrison, J. Field, H.I. Southey, M.C. Severi, G. Donovan, J.L. Hamdy, F.C. Dearnaley, D.P. Muir, K.R. Smith, C. Bagnato, M. Ardern-Jones, A.T. Hall, A.L. O'Brien, L.T. Gehr-Swain, B.N. Wilkinson, R.A. Cox, A. Lewis, S. Brown, P.M. Jhavar, S.G. Tymrakiewicz, M. Lophatananon, A. Bryant, S.L. UK Genetic Prostate Cancer Study Collaborators, . British Association of Urological Surgeons' Section of Oncology, . UK ProtecT Study Collaborators, . Horwich, A. Huddart, R.A. Khoo, V.S. Parker, C.C. Woodhouse, C.J. Thompson, A. Christmas, T. Ogden, C. Fisher, C. Jameson, C. Cooper, C.S. English, D.R. Hopper, J.L. Neal, D.E. Easton, D.F. Eeles, R.A (2009) Identification of new genetic risk factors for prostate cancer.. Show Abstract full text

There is evidence that a substantial part of genetic predisposition to prostate cancer (PCa) may be due to lower penetrance genes which are found by genome-wide association studies. We have recently conducted such a study and seven new regions of the genome linked to PCa risk have been identified. Three of these loci contain candidate susceptibility genes: MSMB, LMTK2 and KLK2/3. The MSMB and KLK2/3 genes may be useful for PCa screening, and the LMTK2 gene might provide a potential therapeutic target. Together with results from other groups, there are now 23 germline genetic variants which have been reported. These results have the potential to be developed into a genetic test. However, we consider that marketing of tests to the public is premature, as PCa risk can not be evaluated fully at this stage and the appropriate screening protocols need to be developed. Follow-up validation studies, as well as studies to explore the psychological implications of genetic profile testing, will be vital prior to roll out into healthcare.

Jhavar, S. Brewer, D. Edwards, S. Kote-Jarai, Z. Attard, G. Clark, J. Flohr, P. Christmas, T. Thompson, A. Parker, M. Shepherd, C. Stenman, U.-.H. Marchbank, T. Playford, R.J. Woodhouse, C. Ogden, C. Fisher, C. Kovacs, G. Corbishley, C. Jameson, C. Norman, A. De-Bono, J. Bjartell, A. Eeles, R. Cooper, C.S (2009) Integration of ERG gene mapping and gene-expression profiling identifies distinct categories of human prostate cancer.. Show Abstract full text

OBJECTIVE: To integrate the mapping of ERG alterations with the collection of expression microarray (EMA) data, as previous EMA analyses have failed to consider the genetic heterogeneity and complex patterns of ERG alteration frequently found in cancerous prostates. MATERIALS AND METHODS: We determined genome-wide expression levels with GeneChip Human Exon 1.0 ST arrays (Affymetrix, Santa Clara, CA, USA) using RNA prepared from 35 specimens of prostate cancer from 28 prostates. RESULTS: The expression profiles showed clustering, in unsupervised hierarchical analyses, into two distinct prostate cancer categories, with one group strongly associated with indicators of poor clinical outcome. The two categories are not tightly linked to ERG status. By analysis of the data we identified a subgroup of cancers lacking ERG rearrangements that showed an outlier pattern of SPINK1 mRNA expression. There was a major distinction between ERG rearranged and non-rearranged cancers that involves the levels of expression of genes linked to exposure to beta-oestradiol, and to retinoic acid. CONCLUSIONS: Expression profiling of prostate cancer samples containing single patterns of ERG alterations can provide novel insights into the mechanism of prostate cancer development, and support the view that factors other than ERG status are the major determinants of poor clinical outcome.

Mitra, A. Fisher, C. Foster, C.S. Jameson, C. Barbachanno, Y. Bartlett, J. Bancroft, E. Doherty, R. Kote-Jarai, Z. Peock, S. Easton, D. IMPACT and EMBRACE Collaborators, . Eeles, R (2008) Prostate cancer in male BRCA1 and BRCA2 mutation carriers has a more aggressive phenotype.. Show Abstract full text

There is a high and rising prevalence of prostate cancer (PRCA) within the male population of the United Kingdom. Although the relative risk of PRCA is higher in male BRCA2 and BRCA1 mutation carriers, the histological characteristics of this malignancy in these groups have not been clearly defined. We present the histopathological findings in the first UK series of BRCA1 and BRCA2 mutation carriers with PRCA. The archived histopathological tissue sections of 20 BRCA1/2 mutation carriers with PRCA were collected from histopathology laboratories in England, Ireland and Scotland. The cases were matched to a control group by age, stage and serum PSA level of PRCA cases diagnosed in the general population. Following histopathological evaluation and re-grading according to current conventional criteria, Gleason scores of PRCA developed by BRCA1/2 mutation carriers were identified to be significantly higher (Gleason scores 8, 9 or 10, P=0.012) than those in the control group. Since BRCA1/2 mutation carrier status is associated with more aggressive disease, it is a prognostic factor for PRCA outcome. Targeting screening to this population may detect disease at an earlier clinical stage which may therefore be beneficial.

Jhavar, S. Reid, A. Clark, J. Kote-Jarai, Z. Christmas, T. Thompson, A. Woodhouse, C. Ogden, C. Fisher, C. Corbishley, C. De-Bono, J. Eeles, R. Brewer, D. Cooper, C (2008) Detection of TMPRSS2-ERG translocations in human prostate cancer by expression profiling using GeneChip Human Exon 1.0 ST arrays.. Show Abstract full text

Translocation of TMPRSS2 to the ERG gene, found in a high proportion of human prostate cancer, results in overexpression of the 3'-ERG sequences joined to the 5'-TMPRSS2 promoter. The studies presented here were designed to test the ability of expression analysis on GeneChip Human Exon 1.0 ST arrays to detect 5'-TMPRSS2-ERG-3' hybrid transcripts encoded by this translocation. Monitoring the relative expression of each ERG exon revealed altered transcription of the ERG gene in 15 of a series of 27 prostate cancer samples. In all cases, exons 4 to 11 exhibited enhanced expression compared with exons 2 and 3. This pattern of expression indicated that the most abundant hybrid transcripts involve fusions to ERG exon 4, and RT-PCR analyses confirmed the joining of TMPRSS2 exon 1 to ERG exon 4 in all 15 cases. The exon expression patterns also indicated that TMPRSS2-ERG fusion transcripts commonly contain deletion of ERG exon 8. Analysis of gene-level data from the arrays allowed the identification of genes whose expression levels significantly correlated with the presence of the translocation. These studies demonstrate that expression analyses using exon arrays represent a valuable approach for detecting ETS gene translocation in prostate cancer, in parallel with analyses of gene expression profiles.

Locke, I. Kote-Jarai, Z. Fackler, M.J. Bancroft, E. Osin, P. Nerurkar, A. Izatt, L. Pichert, G. Gui, G.P.H. Eeles, R.A (2007) Gene promoter hypermethylation in ductal lavage fluid from healthy BRCA gene mutation carriers and mutation-negative controls.. Show Abstract full text

INTRODUCTION: Female germline BRCA gene mutation carriers are at increased risk for developing breast cancer. The purpose of our study was to establish whether healthy BRCA mutation carriers demonstrate an increased frequency of aberrant gene promoter hypermethylation in ductal lavage (DL) fluid, compared with predictive genetic test negative controls, that might serve as a surrogate marker of BRCA1/2 mutation status and/or breast cancer risk. METHODS: The pattern of CpG island hypermethylation within the promoter region of a panel of four genes (RAR-beta, HIN-1, Twist and Cyclin D2) was assessed by methylation-specific polymerase chain reaction using free DNA extracted from DL fluid. RESULTS: Fifty-one DL samples from 24 healthy women of known BRCA mutation status (7 BRCA1 mutation carriers, 12 BRCA2 mutation carriers and 5 controls) were available for methylation analysis. Eight of 19 (42.1%) BRCA mutation carriers were found to have at least one hypermethylated gene in the four-gene panel. Two BRCA mutation carriers, in whom aberrant methylation was found, also had duct epithelial cell atypia identified. No hypermethylation was found in DL samples from 5 negative controls (p = 0.13). CONCLUSION: We found substantial levels of aberrant methylation, with the use of a four-gene panel, in the fluid from the breasts of healthy BRCA mutation carriers compared with controls. Methylation analysis of free DNA in DL fluid may offer a useful surrogate marker for BRCA1/2 mutation status and/or breast cancer risk. Further studies are required for the evaluation of the specificity and predictive value of aberrant methylation in DL fluid for future breast cancer development in BRCA1/2 mutation carriers.

Kote-Jarai, Z. Powles, T.J. Mitchell, G. Tidy, A. Ashley, S. Easton, D. Assersohn, L. Sodha, N. Salter, J. Gusterson, B. Dowsett, M. Eeles, R (2007) BRCA1/BRCA2 mutation status and analysis of cancer family history in participants of the Royal Marsden Hospital tamoxifen chemoprevention trial.. Show Abstract full text

We have analysed the pedigrees of all 70 women who developed cancer in the Royal Marsden Hospital (RMH) tamoxifen chemoprevention trial, using the Claus model, to assess breast cancer susceptibility heterozygote risk (HR) and screened the entire coding regions of BRCA1 and 2 genes in 62 of these cases. We found a reduced incidence of breast cancers developing on tamoxifen in women who have a lower HR, but not in women with higher HR. There were too few BRCA1/2 mutations (4 cases) to be able to determine the efficacy of tamoxifen by BRCA status. Immunohistochemical analysis showed a significantly lower frequency of median ER (p=0.03) in the cancers developing in tamoxifen-treated patients. These results suggest that tamoxifen is less likely to be effective at reducing breast cancers which are ER negative and also in some individuals at higher HR.

Kote-Jarai, Z. Salmon, A. Mengitsu, T. Copeland, M. Ardern-Jones, A. Locke, I. Shanley, S. Summersgill, B. Lu, Y.-.J. Shipley, J. Eeles, R (2006) Increased level of chromosomal damage after irradiation of lymphocytes from BRCA1 mutation carriers.. Show Abstract full text

Deleterious mutations in the BRCA1 gene predispose women to an increased risk of breast and ovarian cancer. Many functional studies have suggested that BRCA1 has a role in DNA damage repair and failure in the DNA damage response pathway often leads to the accumulation of chromosomal aberrations. Here, we have compared normal lymphocytes with those heterozygous for a BRCA1 mutation. Short-term cultures were irradiated (8Gy) using a high dose rate and subsequently metaphases were analysed by 24-colour chromosome painting (M-FISH). We scored the chromosomal rearrangements in the metaphases from five BRCA1 mutation carriers and from five noncarrier control samples 6 days after irradiation. A significantly higher level of chromosomal damage was detected in the lymphocytes heterozygous for BRCA1 mutations compared with normal controls; the average number of aberrations per mitosis was 3.48 compared with 1.62 in controls (P=0.0001). This provides new evidence that heterozygous mutation carriers have a different response to DNA damage compared with noncarriers and that BRCA1 has a role in DNA damage surveillance. Our finding has implications for treatment and screening of BRCA1 mutation carriers using modalities that involve irradiation.

Kote-Jarai, Z. Williams, R.D. Cattini, N. Copeland, M. Giddings, I. Wooster, R. tePoele, R.H. Workman, P. Gusterson, B. Peacock, J. Gui, G. Campbell, C. Eeles, R (2004) Gene expression profiling after radiation-induced DNA damage is strongly predictive of BRCA1 mutation carrier status.. Show Abstract full text

PURPOSE: The impact of the presence of a germ-line BRCA1 mutation on gene expression in normal breast fibroblasts after radiation-induced DNA damage has been investigated. EXPERIMENTAL DESIGN: High-density cDNA microarray technology was used to identify differential responses to DNA damage in fibroblasts from nine heterozygous BRCA1 mutation carriers compared with five control samples without personal or family history of any cancer. Fibroblast cultures were irradiated, and their expression profile was compared using intensity ratios of the cDNA microarrays representing 5603 IMAGE clones. RESULTS: Class comparison and class prediction analysis has shown that BRCA1 mutation carriers can be distinguished from controls with high probability (approximately 85%). Significance analysis of microarrays and the support vector machine classifier identified gene sets that discriminate the samples according to their mutation status. These include genes already known to interact with BRCA1 such as CDKN1B, ATR, and RAD51. CONCLUSIONS: The results of this initial study suggest that normal cells from heterozygous BRCA1 mutation carriers display a different gene expression profile from controls in response to DNA damage. Adaptations of this pilot result to other cell types could result in the development of a functional assay for BRCA1 mutation status.

Kote-Jarai, Z. Singh, R. Durocher, F. Easton, D. Edwards, S.M. Ardern-Jones, A. Dearnaley, D.P. Houlston, R. Kirby, R. Eeles, R (2003) Association between leptin receptor gene polymorphisms and early-onset prostate cancer.. Show Abstract full text

OBJECTIVE: To report a case-control study examining the relationship between polymorphisms in the leptin receptor (OBR) gene and the development of young-onset prostate cancer, because epidemiological studies report that prostate cancer risk is associated with animal fat intake, and thus we investigated if this association occurs via this genetic mechanism. PATIENTS, SUBJECTS AND METHODS: The Lys109Arg (OBR1) and Gln223Arg (OBR2) polymorphisms in the coding region of OBR were studied in blood DNA from 271 patients with prostate cancer aged < 56 years at diagnosis and 277 geographically matched control subjects. Cases were collected through the Cancer Research UK/British Prostate Group Familial Prostate Cancer Study. Blood DNA was genotyped using the polymerase chain reaction and a restriction enzyme digest. RESULTS: There was no statistically significant association between the OBR genotype and prostate cancer risk; men homozygous for 109Arg genotype had a slightly increased risk for prostate cancer, with a relative risk (95% confidence interval) of 1.36 (0.65-2.85), and those homozygous for the 223Arg allele had some reduction in prostate cancer risk, at 0.82 (0.58-1.26), but neither was statistically significant. CONCLUSION: This case-control study showed no significant association between leptin receptor gene polymorphisms and the risk of young-onset prostate cancer, suggesting that genetic variations in OBR are unlikely to have a major role in the development of early-onset prostate cancer in the UK.

Singh, R. Eeles, R.A. Durocher, F. Simard, J. Edwards, S. Badzioch, M. Kote-Jarai, Z. Teare, D. Ford, D. Dearnaley, D. Ardern-Jones, A. Murkin, A. Dowe, A. Shearer, R. Kelly, J. Labrie, F. Easton, D. Narod, S.A. Tonin, P.N. Foulkes, W.D (2000) High risk genes predisposing to prostate cancer development-do they exist?. Show Abstract full text

There is evidence for genetic predisposition to prostate cancer. However, prostate cancer genes have been more difficult to find than genes for some of the other common cancers, such as breast and colon cancer. The reasons for this are discussed in this article and it is now becoming clear that prostate cancer is probably due to multiple genes, many of which are moderate or low penetrance. The advances in the Human Genome Project and technology, especially that of robotics, will help to overcome these problems. Prostate Cancer and Prostatic Diseases (2000) 3, 241-247

Kote-Jarai, Z. Durocher, F. Edwards, S.M. Hamoudi, R. Jackson, R.A. Ardern-Jones, A. Murkin, A. Dearnaley, D.P. Kirby, R. Houlston, R. Easton, D.F. Eeles, R. CRC/BPG UK Familial Prostrate Cancer Collaborators, (2002) Association between the GCG polymorphism of the selenium dependent GPX1 gene and the risk of young onset prostate cancer.. Show Abstract full text

Epidemiological studies have suggested an association between low selenium levels and the development of prostate cancer. Human cellular glutathione peroxidase I (hGPX1) is a selenium-dependent enzyme that protects against oxidative damage and its peroxidase activity is a plausible mechanism for cancer prevention by selenium. The GPX1 gene has a GCG repeat polymorphism in exon 1, coding for a polyalanine tract of five to seven alanine residues. To test if the GPX1 GCG repeat polymorphism associates with the risk of young-onset prostate cancer we conducted a case-control study. The GPX1Ala genotypes were determined for 267 prostate cancer cases and 260 control individuals using polymerase chain reaction (PCR) amplification with fluorescently labelled primers and an ABI 377 automated genotyper. Associations between specific genotypes and the risk of prostate cancer were examined by logistic regression. We found no significant association between the GPX1 genotypes and prostate cancer. There was however an increased frequency of the GPX1Ala6/Ala6 genotype in the prostate cancer cases compared to controls (OR: 1.67; 95% CI: 0.97-2.87). The result of this study suggests that the GPX1 genotype is unlikely to be associated with the risk of developing prostate cancer.

Edwards, S.M. Kote-Jarai, Z. Meitz, J. Hamoudi, R. Hope, Q. Osin, P. Jackson, R. Southgate, C. Singh, R. Falconer, A. Dearnaley, D.P. Ardern-Jones, A. Murkin, A. Dowe, A. Kelly, J. Williams, S. Oram, R. Stevens, M. Teare, D.M. Ponder, B.A.J. Gayther, S.A. Easton, D.F. Eeles, R.A. Cancer Research UK/Bristish Prostate Group UK Familial Prostate Cancer Study Collaborators, . British Association of Urological Surgeons Section of Oncology, (2003) Two percent of men with early-onset prostate cancer harbor germline mutations in the BRCA2 gene.. Show Abstract full text

Studies of families with breast cancer have indicated that male carriers of BRCA2 mutations are at increased risk of prostate cancer, particularly at an early age. To evaluate the contribution of BRCA2 mutations to early-onset prostate cancer, we screened the complete coding sequence of BRCA2 for germline mutations, in 263 men with diagnoses of prostate cancer who were </=55 years of age. Protein-truncating mutations were found in six men (2.3%; 95% confidence interval 0.8%-5.0%), and all of these mutations were clustered outside the ovarian-cancer cluster region. The relative risk of developing prostate cancer by age 56 years from a deleterious germline BRCA2 mutation was 23-fold. Four of the patients with mutations did not have a family history of breast or ovarian cancer. Twenty-two variants of uncertain significance were also identified. These results confirm that BRCA2 is a high-risk prostate-cancer-susceptibility gene and have potential implications for the management of early-onset prostate cancer, in both patients and their relatives.

Kote-Jarai, Z. Easton, D. Edwards, S.M. Jefferies, S. Durocher, F. Jackson, R.A. Singh, R. Ardern-Jones, A. Murkin, A. Dearnaley, D.P. Shearer, R. Kirby, R. Houlston, R. Eeles, R. CRC/BPG UK Familial Prostate Cancer Study Collaborators, (2001) Relationship between glutathione S-transferase M1, P1 and T1 polymorphisms and early onset prostate cancer.. Show Abstract full text

There is evidence suggesting that polymorphic variations in the glutathione S-transferases (GSTs) are associated with cancer susceptibility. Inter-individual differences in cancer susceptibility may be mediated in part through polymorphic variability in the bioactivation and detoxification of carcinogens. The GSTs have been consistently implicated as cancer susceptibility genes in this context. The GST supergene family includes several loci with well characterized polymorphisms. Approximately 50% of the Caucasian population are homozygous for deletions in GSTM1 and approximately 20% are homozygous for deletions in GSTT1, resulting in conjugation deficiency of mutagenic electrophiles to glutathione. The GSTP1 gene has a polymorphism at codon 105 resulting in an Ile to Val substitution which consequently alters the enzymatic activity of the protein and this has been suggested as a putative high-risk genotype in various cancers. We investigated the relationship between GST polymorphisms and young onset prostate cancer in a case-control study. GSTM1, GSTT1 and GSTP1 genotypes were determined for 275 prostate cancer patients and for 280 geographically matched control subjects. We found no significant difference in the frequency of GSTM1 or GSTT1 null genotypes between cases and controls. GSTP1 genotype was, however, significantly associated with prostate cancer risk: the Ile/Ile homozygotes had the lowest risk and there was a trend in increasing the risk with the number of 105 Val alleles: Ile/Val odds ratio (OR)= 1.30 (95% FCI 0.99-1.69), Val/Val OR = 1.80 (95% FCI 1.11-2.91); Ptrend = 0.026. These results suggest that the GSTP1 polymorphism may be a risk factor for developing young onset prostate cancer. We also found that carrying more than one putative high-risk allele in the carcinogen metabolizing GST family was associated with an elevated risk for early onset prostate cancer (OR 2.48, 95% FCI 1.22-5.04, Ptrend = 0.017).

Edwards, S.M. Kote-Jarai, Z. Hamoudi, R. Eeles, R.A (2001) An improved high throughput heteroduplex mutation detection system for screening BRCA2 mutations-fluorescent mutation detection (F-MD).. Show Abstract full text

We describe an improved, fast, automated method for screening large genes such as BRCA2 for germline genomic mutations. The method is based on heteroduplex analysis, and has been adapted for a high throughput application by combining the fluorescent technology of automated sequencers and robotic sample handling. This novel approach allows the entire BRCA2 gene to be screened with appropriate overlaps in four lanes of an ABI 377 gel. The method will detect all types of mutations, especially point mutations, more reliably and robustly than other commonly used conformational sensitive methods (e.g. CSGE). In addition we show that this approach, which relies on band shift detection, is able to detect single base substitutions that have hitherto only been detectable by direct sequencing methods.

Kote-Jarai, Z. Eeles, R.A (1999) BRCA1, BRCA2 and their possible function in DNA damage response.. full text
Edwards, S.M. Evans, D.G.R. Hope, Q. Norman, A.R. Barbachano, Y. Bullock, S. Kote-Jarai, Z. Meitz, J. Falconer, A. Osin, P. Fisher, C. Guy, M. Jhavar, S.G. Hall, A.L. O'Brien, L.T. Gehr-Swain, B.N. Wilkinson, R.A. Forrest, M.S. Dearnaley, D.P. Ardern-Jones, A.T. Page, E.C. Easton, D.F. Eeles, R.A. UK Genetic Prostate Cancer Study Collaborators and BAUS Section of Oncology, (2010) Prostate cancer in BRCA2 germline mutation carriers is associated with poorer prognosis.. Show Abstract full text

BACKGROUND: The germline BRCA2 mutation is associated with increased prostate cancer (PrCa) risk. We have assessed survival in young PrCa cases with a germline mutation in BRCA2 and investigated loss of heterozygosity at BRCA2 in their tumours. METHODS: Two cohorts were compared: one was a group with young-onset PrCa, tested for germline BRCA2 mutations (6 of 263 cases had a germline BRAC2 mutation), and the second was a validation set consisting of a clinical set from Manchester of known BRCA2 mutuation carriers (15 cases) with PrCa. Survival data were compared with a control series of patients in a single clinic as determined by Kaplan-Meier estimates. Loss of heterozygosity was tested for in the DNA of tumour tissue of the young-onset group by typing four microsatellite markers that flanked the BRCA2 gene, followed by sequencing. RESULTS: Median survival of all PrCa cases with a germline BRCA2 mutation was shorter at 4.8 years than was survival in controls at 8.5 years (P=0.002). Loss of heterozygosity was found in the majority of tumours of BRCA2 mutation carriers. Multivariate analysis confirmed that the poorer survival of PrCa in BRCA2 mutation carriers is associated with the germline BRCA2 mutation per se. CONCLUSION: BRCA2 germline mutation is an independent prognostic factor for survival in PrCa. Such patients should not be managed with active surveillance as they have more aggressive disease.

Lophatananon, A. Archer, J. Easton, D. Pocock, R. Dearnaley, D. Guy, M. Kote-Jarai, Z. O'Brien, L. Wilkinson, R.A. Hall, A.L. Sawyer, E. Page, E. Liu, J.-.F. Barratt, S. Rahman, A.A. UK Genetic Prostate Cancer Study Collaborators, . British Association of Urological Surgeons' Section of Oncology, . Eeles, R. Muir, K (2010) Dietary fat and early-onset prostate cancer risk.. Show Abstract full text

The UK incidence of prostate cancer has been increasing in men aged < 60 years. Migrant studies and global and secular variation in incidence suggest that modifiable factors, including a high-fat diet, may contribute to prostate cancer risk. The aim of the present study was to investigate the role of dietary fat intake and its derivatives on early-onset prostate cancer risk. During 1999-2004, a population-based case-control study with 512 cases and 838 controls was conducted. Cases were diagnosed with prostate cancer when < or = 60 years. Controls were sourced from UK GP practice registers. A self-administered FFQ collected data on typical past diet. A nutritional database was used to calculate daily fat intake. A positive, statistically significant risk estimate for the highest v. lowest quintile of intake of total fat, SFA, MUFA and PUFA was observed when adjusted for confounding variables: OR 2.53 (95 % CI 1.72, 3.74), OR 2.49 (95 % CI 1.69, 3.66), OR 2.69 (95 % CI 1.82, 3.96) and OR 2.34 (95 % CI 1.59, 3.46), respectively, with all P for trend < 0.001. In conclusion, there was a positive statistically significant association between prostate cancer risk and energy-adjusted intake of total fat and fat subtypes. These results potentially identify a modifiable risk factor for early-onset prostate cancer.

Collin, S.M. Metcalfe, C. Zuccolo, L. Lewis, S.J. Chen, L. Cox, A. Davis, M. Lane, J.A. Donovan, J. Smith, G.D. Neal, D.E. Hamdy, F.C. Gudmundsson, J. Sulem, P. Rafnar, T. Benediktsdottir, K.R. Eeles, R.A. Guy, M. Kote-Jarai, Z. UK Genetic Prostate Cancer Study Group, . Morrison, J. Al Olama, A.A. Stefansson, K. Easton, D.F. Martin, R.M (2009) Association of folate-pathway gene polymorphisms with the risk of prostate cancer: a population-based nested case-control study, systematic review, and meta-analysis.. Show Abstract full text

Folate-pathway gene polymorphisms have been implicated in several cancers and investigated inconclusively in relation to prostate cancer. We conducted a systematic review, which identified nine case-control studies (eight included, one excluded). We also included data from four genome-wide association studies and from a case-control study nested within the UK population-based Prostate Testing for Cancer and Treatment study. We investigated by meta-analysis the effects of eight polymorphisms: MTHFR C677T (rs1801133; 12 studies; 10,745 cases; 40,158 controls), MTHFR A1298C (rs1801131; 5 studies; 3,176 cases; 4,829 controls), MTR A2756G (rs1805087; 8 studies; 7,810 cases; 37,543 controls), MTRR A66G (rs1801394; 4 studies; 3,032 cases; 4,515 controls), MTHFD1 G1958A (rs2236225; 6 studies; 7,493 cases; 36,941 controls), SLC19A1/RFC1 G80A (rs1051266; 4 studies; 6,222 cases; 35,821 controls), SHMT1 C1420T (rs1979277; 2 studies; 2,689 cases; 4,110 controls), and FOLH1 T1561C (rs202676; 5 studies; 6,314 cases; 35,190 controls). The majority (10 of 13) of eligible studies had 100% Caucasian subjects; only one study had <90% Caucasian subjects. We found weak evidence of dominant effects of two alleles: MTR 2756A>G [random effects pooled odds ratio, 1.06 (1.00-1.12); P = 0.06 (P = 0.59 for heterogeneity across studies)] and SHMT1 1420C>T [random effects pooled odds ratio, 1.11 (1.00-1.22); P = 0.05 (P = 0.38 for heterogeneity across studies)]. We found no effect of MTHFR 677C>T or any of the other alleles in dominant, recessive or additive models, or in comparing a/a versus A/A homozygous. Neither did we find any difference in effects on advanced or localized cancers. Our meta-analysis suggests that known common folate-pathway single nucleotide polymorphisms do not have significant effects on susceptibility to prostate cancer.

Ghoussaini, M. Song, H. Koessler, T. Al Olama, A.A. Kote-Jarai, Z. Driver, K.E. Pooley, K.A. Ramus, S.J. Kjaer, S.K. Hogdall, E. DiCioccio, R.A. Whittemore, A.S. Gayther, S.A. Giles, G.G. Guy, M. Edwards, S.M. Morrison, J. Donovan, J.L. Hamdy, F.C. Dearnaley, D.P. Ardern-Jones, A.T. Hall, A.L. O'Brien, L.T. Gehr-Swain, B.N. Wilkinson, R.A. Brown, P.M. Hopper, J.L. Neal, D.E. Pharoah, P.D.P. Ponder, B.A.J. Eeles, R.A. Easton, D.F. Dunning, A.M. UK Genetic Prostate Cancer Study Collaborators/British Association of Urological Surgeons' Section of Oncology, . UK ProtecT Study Collaborators, (2008) Multiple loci with different cancer specificities within the 8q24 gene desert.. Show Abstract full text

Recent studies based on genome-wide association, linkage, and admixture scan analysis have reported associations of various genetic variants in 8q24 with susceptibility to breast, prostate, and colorectal cancer. This locus lies within a 1.18-Mb region that contains no known genes but is bounded at its centromeric end by FAM84B and at its telomeric end by c-MYC, two candidate cancer susceptibility genes. To investigate the associations of specific loci within 8q24 with specific cancers, we genotyped the nine previously reported cancer-associated single-nucleotide polymorphisms across the region in four case-control sets of prostate (1854 case subjects and 1894 control subjects), breast (2270 case subjects and 2280 control subjects), colorectal (2299 case subjects and 2284 control subjects), and ovarian (1975 case subjects and 3411 control subjects) cancer. Five different haplotype blocks within this gene desert were specifically associated with risks of different cancers. One block was solely associated with risk of breast cancer, three others were associated solely with the risk of prostate cancer, and a fifth was associated with the risk of prostate, colorectal, and ovarian cancer, but not breast cancer. We conclude that there are at least five separate functional variants in this region.

Kadouri, L. Kote-Jarai, Z. Hubert, A. Baras, M. Abeliovich, D. Hamburger, T. Peretz, T. Eeles, R.A (2008) Glutathione-S-transferase M1, T1 and P1 polymorphisms, and breast cancer risk, in BRCA1/2 mutation carriers.. Show Abstract full text

Variation in penetrance estimates for BRCA1/2 carriers suggests that other environmental and genetic factors may modify cancer risk in carriers. The GSTM1, T1 and P1 isoenzymes are involved in metabolism of environmental carcinogens. The GSTM1 and GSTT1 gene is absent in a substantial proportion of the population. In GSTP1, a single-nucleotide polymorphism that translates to Ile112Val was associated with lower activity. We studied the effect of these polymorphisms on breast cancer (BC) risk in BRCA1/2 carriers. A population of 320 BRCA1/2 carriers were genotyped; of them 262 were carriers of one of the three Ashkenazi founder mutations. Two hundred and eleven were affected with BC (20 also with ovarian cancer (OC)) and 109 were unaffected with BC (39 of them had OC). Risk analyses were conducted using Cox proportional hazard models adjusted for origin (Ashkenazi vs non-Ashkenazi). We found an estimated BC HR of 0.89 (95% CI 0.65-1.12, P=0.25) and 1.11 (95% CI 0.81-1.52, P=0.53) for the null alleles of GSTM1 and GSTT1, respectively. For GSTP1, HR for BC was 1.36 (95% CI 1.02-1.81, P=0.04) for individuals with Ile/Val, and 2.00 (95% CI 1.18-3.38) for carriers of the Val/Val genotype (P=0.01). An HR of 3.20 (95% CI 1.26-8.09, P=0.01), and younger age at BC onset (P=0.2), were found among Val/Val, BRCA2 carriers, but not among BRCA1 carriers. In conclusion, our results indicate significantly elevated risk for BC in carriers of BRCA2 mutations with GSTP1-Val allele with dosage effect, as implicated by higher risk in homozygous Val carriers. The GSTM1- and GSTT1-null allele did not seem to have a major effect.

Locke, I. Kote-Jarai, Z. Bancroft, E. Bullock, S. Jugurnauth, S. Osin, P. Nerurkar, A. Izatt, L. Pichert, G. Gui, G.P.H. Eeles, R.A (2006) Loss of heterozygosity at the BRCA1 and BRCA2 loci detected in ductal lavage fluid from BRCA gene mutation carriers and controls.. Show Abstract full text

Female BRCA gene mutation carriers are at increased risk for developing breast cancer. Ductal lavage is a novel method for sampling breast ductal fluid, providing epithelial cells for cytologic assessment and a source of free DNA for molecular analyses. Loss of heterozygosity (LOH) at the BRCA loci in ductal lavage fluid is a potential biomarker of breast cancer risk. The LOH rate was measured at the BRCA1/2 loci and compared with that at a control locus (APC) using free DNA from the ductal lavage fluid of BRCA carriers and predictive test negative controls. We evaluated the reproducibility of these analyses. Free DNA sufficient for PCR amplification was obtained from 33 ductal lavage samples of 17 healthy women of known BRCA status (14 BRCA carriers and 3 controls). LOH rates of 36.4% to 56.3% at the BRCA1 locus and 45% to 61.5% at the BRCA2 locus were found among BRCA carriers. The LOH rate at the APC locus was lower (18.5%). The interaliquot reproducibility for the D17S855 marker of the BRCA1 locus was 66.7%. Intraaliquot reproducibility was 90%. Although we successfully isolated sufficient free DNA from ductal lavage fluid for PCR amplification, the degree of reproducibility of these LOH studies raises questions about the robustness of this technique as a risk assessment tool in the evaluation of high-risk women. Further studies are required to evaluate the specificity and predictive value of LOH in ductal lavage fluid for breast cancer development.

Jefferies, S. Kote-Jarai, Z. Goldgar, D. Houlston, R. Frazer-Williams, M.-.J. A'Hern, R. Eeles, R. Henk, J. Gore, M. Rhys-Evans, P. Archer, D. Bishop, K. Solomon, E. Hodgson, S. McGurk, M. Hibbert, J. O'Connell, M. Partridge, M. Chevretton, E. Calman, F. Saunders, M. Shotton, K. Brown, A. Whittaker, S. Foulkes, W. MPT Collaborators, . MPT Collaborators:, (2005) Association between polymorphisms of the GPX1 gene and second primary tumours after index squamous cell cancer of the head and neck.. Show Abstract full text

We investigated the association between genetic polymorphisms in GPX1 gene amongst patients who had index squamous cell carcinoma (SCCHN) and a second primary tumour (SPT) after a primary SCCHN in a case-control study. GPX1 genotypes were determined for 61 patients with SPT and for 259 control subjects by a PCR technique using a fluorescent-labelled primer. Analysis was by an ABI automated fluorescent sequencer. The associations between specific genotypes and the development of SPT were examined by logistic regression. A significant difference was found between the control group and the SPT cases in allele frequencies of GPX1 ALA( *)6 and ALA( *)7 (p(trend)=0.04). These results suggest that polymorphisms in the GPX1 gene may be a marker for SPT development and further studies are indicated.

Forrest, M.S. Edwards, S.M. Houlston, R. Kote-Jarai, Z. Key, T. Allen, N. Knowles, M.A. Turner, F. Ardern-Jones, A. Murkin, A. Williams, S. Oram, R. Bishop, D.T. Eeles, R.A. CR-UK/BPG UK prostate cancer study collaborators, (2005) Association between hormonal genetic polymorphisms and early-onset prostate cancer.. Show Abstract full text

We investigated the association between seven polymorphisms in four candidate genes involved in vitamin D and androgen metabolism with early-onset prostate cancer (CaP) risk. The polymorphisms were genotyped in 288 UK males who were diagnosed with CaP at the age of 55 y or younger and up to 700 population-based controls. An additional 50 cases (not selected for age) and 76 controls were also genotyped. Short (< or =22 repeats) AR (CAG)(n) repeats were associated with a significantly reduced risk of early onset CaP (OR 0.68, 95% CI 0.50-0.91) compared with men with long (> 22) repeats. Men homozygous for the leucine variant of SRD5A2 p.89V > L were also found to be at a significantly increased risk of CaP compared with men who were homozygous for the valine allele (OR 1.84, 95% CI 1.15-2.98). No associations were found with the AR (GGC)(n), CYP17 Msp A1 I, VDR Taq I, SRD5A2 (TA)(n) and p.49A >T polymorphisms and CaP risk. These findings suggest that common polymorphisms in the AR and SRD5A2 genes may be associated with early-onset CaP in British men.

Kadouri, L. Kote-Jarai, Z. Hubert, A. Durocher, F. Abeliovich, D. Glaser, B. Hamburger, T. Eeles, R.A. Peretz, T (2004) A single-nucleotide polymorphism in the RAD51 gene modifies breast cancer risk in BRCA2 carriers, but not in BRCA1 carriers or noncarriers.. Show Abstract full text

Variation in the penetrance estimates for BRCA1 and BRCA2 mutation carriers suggests that other genetic polymorphisms may modify the cancer risk in carriers. The RAD51 gene, which participates in homologous recombination double-strand breaks (DSB) repair in the same pathway as the BRCA1 and BRCA2 gene products, is a candidate for such an effect. A single-nucleotide polymorphism (SNP), RAD51-135g-->c, in the 5' untranslated region of the gene has been found to elevate breast cancer (BC) risk among BRCA2 carriers. We genotyped 309 BRCA1/2 mutation carriers, of which 280 were of Ashkenazi origin, 166 noncarrier BC patients and 152 women unaffected with BC (a control group), for the RAD51-135g-->c SNP. Risk analyses were conducted using COX proportional hazard models for the BRCA1/2 carriers and simple logistic regression analysis for the noncarrier case-control population. BRCA2 carriers were also studied using logistic regression and Kaplan-Meier survival analyses. The estimated BC hazard ratio (HR) for RAD51-135c carriers adjusted for origin (Ashkenazi vs non-Ashkenazi) was 1.28 (95% CI 0.85-1.90, P=0.23) for BRCA1/2 carriers, and 2.09 (95% CI 1.04-4.18, P=0.04) when the analysis was restricted to BRCA2 carriers. The median BC age was younger in BCRA2-RAD51-135c carriers (45 (95% CI 36-54) vs 52 years (95% CI 48-56), P=0.05). In a logistic regression analysis, the odds ratio (OR) was 5.49 (95% CI 0.5-58.8, P=0.163). In noncarrier BC cases, carrying RAD51-135c was not associated with BC risk (0.97; 95% CI 0.47-2.00). These results indicate significantly elevated risk for BC in carriers of BRCA2 mutations who also carry a RAD51-135c allele. In BRCA1 carriers and noncarriers, no effect for this SNP was found.

Kadouri, L. Kote-Jarai, Z. Easton, D.F. Hubert, A. Hamoudi, R. Glaser, B. Abeliovich, D. Peretz, T. Eeles, R.A (2004) Polyglutamine repeat length in the AIB1 gene modifies breast cancer susceptibility in BRCA1 carriers.. Show Abstract full text

Variation in the penetrance estimates for BRCA1 and BRCA2 mutation carriers suggests that other factors may modify cancer risk from specific mutations. One possible mechanism is an epigenetic effect of polymorphisms in other genes. Genes involved in hormonal signal transduction are possible candidates. The AIB1 gene, an estrogen receptor (ER) coactivator, is frequently amplified in breast and ovarian tumors. Variation of a CAG repeat length has been reported within this gene that encodes a polyglutamine repeat in the C-terminus of the protein. Three hundred eleven BRCA1/2 mutation carriers (257 were of Ashkenazi origin) were genotyped for the AIB1 polyglutamine repeat. Relative risks (RR) were estimated using a maximum likelihood approach. The estimated breast cancer (BC) RR per average repeat length adjusted for population type (Ashkenazi vs. non-Ashkenazi) was 1.15 (95% CI = 1.02-1.30; p = 0.01) for BRCA1/2 carriers, and 1.25 (95% CI = 1.09-1.42; p = 0.001) when analysis was restricted to BRCA1 carriers. RR of BC was 1.17 (95% CI = 0.91-1.74), for individuals with 2 alleles >/=29 polyglutamine repeats and 0.78 (95% CI = 0.50-1.16) for those with at least 1 allele of </=26 repeats, compared to individuals with the common genotypes 28;28, 28;29 or 28;30. The corresponding BC RR in BRCA1 mutation carriers was 0.55 (95% CI = 0.34-0.90) and 1.29 (95% CI = 0.85-1.96) in those with </=26 and >/=29 repeats respectively (p = 0.025). These results indicate significant association of the risk for BC in carriers of BRCA1 mutations with the polyglutamine chain of the AIB1 gene. Longer repeat length correlates with elevated risk, whereas in carriers of a shorter AIB1 allele BC risk was reduced. The AIB1 polyglutamine length did not affect BC risk among BRCA2 mutation carriers.

Kadouri, L. Easton, D.F. Edwards, S. Hubert, A. Kote-Jarai, Z. Glaser, B. Durocher, F. Abeliovich, D. Peretz, T. Eeles, R.A (2001) CAG and GGC repeat polymorphisms in the androgen receptor gene and breast cancer susceptibility in BRCA1/2 carriers and non-carriers.. Show Abstract full text

Variation in the penetrance estimates for BRCA1 and BRCA2 mutations carriers suggests that other genetic polymorphisms may modify the cancer risk in carriers. A previous study has suggested that BRCA1 carriers with longer lengths of the CAG repeat in the androgen receptor (AR) gene are at increased risk of breast cancer (BC). We genotyped 188 BRCA1/2 carriers (122 affected and 66 unaffected with breast cancer), 158 of them of Ashkenazi origin, 166 BC cases without BRCA1/2 mutations and 156 Ashkenazi control individuals aged over 56 for the AR CAG and GGC repeats. In carriers, risk analyses were conducted using a variant of the log-rank test, assuming two sets of risk estimates in carriers: penetrance estimates based on the Breast Cancer Linkage Consortium (BCLC) studies of multiple case families, and lower estimates as suggested by population-based studies. We found no association of the CAG and GGC repeats with BC risk in either BRCA1/2 carriers or in the general population. Assuming BRCA1/2 penetrance estimates appropriate to the Ashkenazi population, the estimated RR per repeat adjusted for ethnic group (Ashkenazi and non-Ashkenazi) was 1.05 (95%CI 0.97-1.17) for BC and 1.00 (95%CI 0.83-1.20) for ovarian cancer (OC) for CAG repeats and 0.96 (95%CI 0.80-1.15) and 0.90 (95%CI 0.60-1.22) respectively for GGC repeats. The corresponding RR estimates for the unselected case-control series were 1.00 (95%CI 0.91-1.10) for the CAG and 1.05 (95%CI 0.90-1.22) for the GGC repeats. The estimated relative risk of BC in carriers associated with > or =28 CAG repeats was 1.08 (95%CI 0.45-2.61). Furthermore, no significant association was found if attention was restricted to the Ashkenazi carriers, or only to BRCA1 or BRCA2 carriers. We conclude that, in contrast to previous observations, if there is any effect of the AR repeat length on BRCA1 penetrance, it is likely to be weak.

Jefferies, S. Kote-Jarai, Z. Houlston, R. Fraser-Williams, M.J. A'Hern, R. Foulkes, W.D. Goldgar, D. Eeles, R.A (2000) Association between polymorphisms of the GPX1 gene and multiple primary tumours (MPT).. full text
Eeles, R.A. Powles, T.P. Ashley, S. Easton, D.F. Assersohn, L. Sodha, N. Dowsett, M. Gusterson, B. Tidy, A. Mitchell, G. Kote-Jarai, Z (2000) BRCA1, BRCA2 mutation and pedigree genetic analysis to determine genetic risk in the UK Royal Marsden Hospital tamoxifen prevention trial. full text
Eeles, R.A. Powles, T.P. Ashley, S. Easton, D.F. Assersohn, L. Sodha, N. Dowsett, M. Gusterson, B. Tidy, A. Mitchell, G. Kote-Jarai, Z (1999) BRCA1, BRCA2 and pedigree genetic analysis to determine genetic risk in the UK Royal Marsden Hospital tamoxifen prevention trial.. full text
Kote-Jarai, Z. Edwards, S. Easton, D. Jackson, R. Ardern-Jones, A. Murkin, A. Dearnaley, D (1999) Association between the GSTM1, GSTP1 and GSTT1 gene polymorphisms and young onset prostate cancer in the UK.. full text
Whitaker, H.C. Kote-Jarai, Z. Ross-Adams, H. Warren, A.Y. Burge, J. George, A. Bancroft, E. Jhavar, S. Leongamornlert, D. Tymrakiewicz, M. Saunders, E. Page, E. Mitra, A. Mitchell, G. Lindeman, G.J. Evans, D.G. Blanco, I. Mercer, C. Rubinstein, W.S. Clowes, V. Douglas, F. Hodgson, S. Walker, L. Donaldson, A. Izatt, L. Dorkins, H. Male, A. Tucker, K. Stapleton, A. Lam, J. Kirk, J. Lilja, H. Easton, D. Cooper, C. Eeles, R. Neal, D.E (2010) The rs10993994 Risk Allele for Prostate Cancer Results in Clinically Relevant Changes in Microseminoprotein-Beta Expression in Tissue and Urine.
Kote-Jarai, Z. Amin Al Olama, A. Leongamornlert, D. Tymrakiewicz, M. Saunders, E. Guy, M. Giles, G.G. Severi, G. Southey, M. Hopper, J.L. Sit, K.C. Harris, J.M. Batra, J. Spurdle, A.B. Clements, J.A. Hamdy, F. Neal, D. Donovan, J. Muir, K. Pharoah, P.D.P. Chanock, S.J. Brown, N. Benlloch, S. Castro, E. Mahmud, N. O'Brien, L. Hall, A. Sawyer, E. Wilkinson, R. Easton, D.F. Eeles, R.A (2011) Identification of a novel prostate cancer susceptibility variant in the KLK3 gene transcript.. Show Abstract full text

Genome-wide association studies (GWAS) have identified more than 30 prostate cancer (PrCa) susceptibility loci. One of these (rs2735839) is located close to a plausible candidate susceptibility gene, KLK3, which encodes prostate-specific antigen (PSA). PSA is widely used as a biomarker for PrCa detection and disease monitoring. To refine the association between PrCa and variants in this region, we used genotyping data from a two-stage GWAS using samples from the UK and Australia, and the Cancer Genetic Markers of Susceptibility (CGEMS) study. Genotypes were imputed for 197 and 312 single nucleotide polymorphisms (SNPs) from HapMap2 and the 1000 Genome Project, respectively. The most significant association with PrCa was with a previously unidentified SNP, rs17632542 (combined P = 3.9 × 10(-22)). This association was confirmed by direct genotyping in three stages of the UK/Australian GWAS, involving 10,405 cases and 10,681 controls (combined P = 1.9 × 10(-34)). rs17632542 is also shown to be associated with PSA levels and it is a non-synonymous coding SNP (Ile179Thr) in KLK3. Using molecular dynamic simulation, we showed evidence that this variant has the potential to introduce alterations in the protein or affect RNA splicing. We propose that rs17632542 may directly influence PrCa risk.

Schumacher, F.R. Berndt, S.I. Siddiq, A. Jacobs, K.B. Wang, Z. Lindstrom, S. Stevens, V.L. Chen, C. Mondul, A.M. Travis, R.C. Stram, D.O. Eeles, R.A. Easton, D.F. Giles, G. Hopper, J.L. Neal, D.E. Hamdy, F.C. Donovan, J.L. Muir, K. Al Olama, A.A. Kote-Jarai, Z. Guy, M. Severi, G. Grönberg, H. Isaacs, W.B. Karlsson, R. Wiklund, F. Xu, J. Allen, N.E. Andriole, G.L. Barricarte, A. Boeing, H. Bueno-de-Mesquita, H.B. Crawford, E.D. Diver, W.R. Gonzalez, C.A. Gaziano, J.M. Giovannucci, E.L. Johansson, M. Le Marchand, L. Ma, J. Sieri, S. Stattin, P. Stampfer, M.J. Tjonneland, A. Vineis, P. Virtamo, J. Vogel, U. Weinstein, S.J. Yeager, M. Thun, M.J. Kolonel, L.N. Henderson, B.E. Albanes, D. Hayes, R.B. Feigelson, H.S. Riboli, E. Hunter, D.J. Chanock, S.J. Haiman, C.A. Kraft, P (2011) Genome-wide association study identifies new prostate cancer susceptibility loci.. Show Abstract full text

Prostate cancer (PrCa) is the most common non-skin cancer diagnosed among males in developed countries and the second leading cause of cancer mortality, yet little is known regarding its etiology and factors that influence clinical outcome. Genome-wide association studies (GWAS) of PrCa have identified at least 30 distinct loci associated with small differences in risk. We conducted a GWAS in 2782 advanced PrCa cases (Gleason grade ≥ 8 or tumor stage C/D) and 4458 controls with 571 243 single nucleotide polymorphisms (SNPs). Based on in silico replication of 4679 SNPs (Stage 1, P < 0.02) in two published GWAS with 7358 PrCa cases and 6732 controls, we identified a new susceptibility locus associated with overall PrCa risk at 2q37.3 (rs2292884, P= 4.3 × 10(-8)). We also confirmed a locus suggested by an earlier GWAS at 12q13 (rs902774, P= 8.6 × 10(-9)). The estimated per-allele odds ratios for these loci (1.14 for rs2292884 and 1.17 for rs902774) did not differ between advanced and non-advanced PrCa (case-only test for heterogeneity P= 0.72 and P= 0.61, respectively). Further studies will be needed to assess whether these or other loci are differentially associated with PrCa subtypes.

Kote-Jarai, Z. Olama, A.A.A. Giles, G.G. Severi, G. Schleutker, J. Weischer, M. Campa, D. Riboli, E. Key, T. Gronberg, H. Hunter, D.J. Kraft, P. Thun, M.J. Ingles, S. Chanock, S. Albanes, D. Hayes, R.B. Neal, D.E. Hamdy, F.C. Donovan, J.L. Pharoah, P. Schumacher, F. Henderson, B.E. Stanford, J.L. Ostrander, E.A. Sorensen, K.D. Dörk, T. Andriole, G. Dickinson, J.L. Cybulski, C. Lubinski, J. Spurdle, A. Clements, J.A. Chambers, S. Aitken, J. Gardiner, R.A.F. Thibodeau, S.N. Schaid, D. John, E.M. Maier, C. Vogel, W. Cooney, K.A. Park, J.Y. Cannon-Albright, L. Brenner, H. Habuchi, T. Zhang, H.-.W. Lu, Y.-.J. Kaneva, R. Muir, K. Benlloch, S. Leongamornlert, D.A. Saunders, E.J. Tymrakiewicz, M. Mahmud, N. Guy, M. O'Brien, L.T. Wilkinson, R.A. Hall, A.L. Sawyer, E.J. Dadaev, T. Morrison, J. Dearnaley, D.P. Horwich, A. Huddart, R.A. Khoo, V.S. Parker, C.C. Van As, N. Woodhouse, C.J. Thompson, A. Christmas, T. Ogden, C. Cooper, C.S. Lophatonanon, A. Southey, M.C. Hopper, J.L. English, D.R. Wahlfors, T. Tammela, T.L.J. Klarskov, P. Nordestgaard, B.G. Røder, M.A. Tybjærg-Hansen, A. Bojesen, S.E. Travis, R. Canzian, F. Kaaks, R. Wiklund, F. Aly, M. Lindstrom, S. Diver, W.R. Gapstur, S. Stern, M.C. Corral, R. Virtamo, J. Cox, A. Haiman, C.A. Le Marchand, L. Fitzgerald, L. Kolb, S. Kwon, E.M. Karyadi, D.M. Orntoft, T.F. Borre, M. Meyer, A. Serth, J. Yeager, M. Berndt, S.I. Marthick, J.R. Patterson, B. Wokolorczyk, D. Batra, J. Lose, F. McDonnell, S.K. Joshi, A.D. Shahabi, A. Rinckleb, A.E. Ray, A. Sellers, T.A. Lin, H.-.Y. Stephenson, R.A. Farnham, J. Muller, H. Rothenbacher, D. Tsuchiya, N. Narita, S. Cao, G.-.W. Slavov, C. Mitev, V. Easton, D.F. Eeles, R.A. UK Genetic Prostate Cancer Study Collaborators/British Association of Urological Surgeons' Section of Oncology, . UK ProtecT Study Collaborators, The Australian Prostate Cancer BioResource, . PRACTICAL Consortium, (2011) Seven prostate cancer susceptibility loci identified by a multi-stage genome-wide association study.. Show Abstract full text

Prostate cancer (PrCa) is the most frequently diagnosed male cancer in developed countries. We conducted a multi-stage genome-wide association study for PrCa and previously reported the results of the first two stages, which identified 16 PrCa susceptibility loci. We report here the results of stage 3, in which we evaluated 1,536 SNPs in 4,574 individuals with prostate cancer (cases) and 4,164 controls. We followed up ten new association signals through genotyping in 51,311 samples in 30 studies from the Prostate Cancer Association Group to Investigate Cancer Associated Alterations in the Genome (PRACTICAL) consortium. In addition to replicating previously reported loci, we identified seven new prostate cancer susceptibility loci on chromosomes 2p11, 3q23, 3q26, 5p12, 6p21, 12q13 and Xq12 (P = 4.0 × 10(-8) to P = 2.7 × 10(-24)). We also identified a SNP in TERT more strongly associated with PrCa than that previously reported. More than 40 PrCa susceptibility loci, explaining ∼25% of the familial risk in this disease, have now been identified.

Rahman, A.A. Lophatananon, A. Stewart-Brown, S. Harriss, D. Anderson, J. Parker, T. Easton, D. Kote-Jarai, Z. Pocock, R. Dearnaley, D. Guy, M. O'Brien, L. Wilkinson, R.A. Hall, A.L. Sawyer, E. Page, E. Liu, J.F. Eeles, R.A. Muir, K.R (2011) Reply: 'Hand pattern indicates risk of prostate cancer'..
Rahman, A.A. Lophatananon, A. Brown, S.S. Harriss, D. Anderson, J. Parker, T. Easton, D. Kote-Jarai, Z. Pocock, R. Dearnaley, D. Guy, M. O'Brien, L. Wilkinson, R.A. Hall, A.L. Sawyer, E. Page, E. Liu, J.-.F. Eeles, R.A. Muir, K (2010) HAND PATTERN AND EARLY-ONSET PROSTATE CANCER RISK. full text
Rahman, A.A. Lophatananon, A. Stewart-Brown, S. Harriss, D. Anderson, J. Parker, T. Easton, D. Kote-Jarai, Z. Pocock, R. Dearnaley, D. Guy, M. O'Brien, L. Wilkinson, R.A. Hall, A.L. Sawyer, E. Page, E. Liu, J.-.F. UK Genetic Prostate Cancer Study Collaborators, . British Association of Urological Surgeons' Section of Oncology, . Eeles, R.A. Muir, K (2011) Hand pattern indicates prostate cancer risk.. Show Abstract full text

BACKGROUND: The ratio of digit lengths is fixed in utero, and may be a proxy indicator for prenatal testosterone levels. METHODS: We analysed the right-hand pattern and prostate cancer risk in 1524 prostate cancer cases and 3044 population-based controls. RESULTS: Compared with index finger shorter than ring finger (low 2D : 4D), men with index finger longer than ring finger (high 2D : 4D) showed a negative association, suggesting a protective effect with a 33% risk reduction (odds ratio (OR) 0.67, 95% confidence interval (CI) 0.57-0.80). Risk reduction was even greater (87%) in age group <60 (OR 0.13, 95% CI 0.09-0.21). CONCLUSION: Pattern of finger lengths may be a simple marker of prostate cancer risk, with length of 2D greater than 4D suggestive of lower risk.

Yuille, M. Condie, A. Hudson, C. Kote-Jarai, Z. Stone, E. Eeles, R. Matutes, E. Catovsky, D. Houlston, R (2002) Relationship between glutathione S-transferase M1, T1, and P1 polymorphisms and chronic lymphocytic leukemia.. Show Abstract full text

Interindividual differences in susceptibility to hematologic malignancies may be mediated in part through polymorphic variability in the bioactivation and detoxification of carcinogens. The glutathione S-transferases (GSTs) have been implicated as susceptibility genes in this context for a number of cancers. The aim of this study was to examine whether polymorphic variation in GSTs confers susceptibility to chronic lymphocytic leukemia (CLL). GSTM1, GSTT1, and GSTP1 genotypes were determined in 138 patients and 280 healthy individuals. The frequency of both GSTM1 and GSTT1 null genotypes and the GSTP1-Ile allele was higher in cases than in controls. There was evidence of a trend in increasing risk with the number of putative "high-risk" alleles of the GST family carried (P =.04). The risk of CLL associated with possession of all 3 "high-risk" genotypes was increased 2.8-fold (OR = 2.8, 95% confidence interval: 1.1-6.9). Our findings suggest that heritable GST status may influence the risk of developing CLL.

Kote-Jarai, Z. Matthews, L. Osorio, A. Shanley, S. Giddings, I. Moreews, F. Locke, I. Evans, D.G. Eccles, D. Carrier Clinic Collaborators, . Williams, R.D. Girolami, M. Campbell, C. Eeles, R (2006) Accurate prediction of BRCA1 and BRCA2 heterozygous genotype using expression profiling after induced DNA damage.. Show Abstract full text

PURPOSE: In this study, the differential gene expression changes following radiation-induced DNA damage in healthy cells from BRCA1/BRCA1 mutation carriers have been compared with controls using high-density microarray technology. We aimed to establish if BRCA1/BRCA2 mutation carriers could be distinguished from noncarriers based on expression profiling of normal cells. EXPERIMENTAL DESIGN: Short-term primary fibroblast cultures were established from skin biopsies from 10 BRCA1 and 10 BRCA2 mutation carriers and 10 controls, all of whom had previously had breast cancer. The cells were subjected to 15 Gy ionizing irradiation to induce DNA damage. RNA was extracted from all cell cultures, preirradiation and at 1 hour postirradiation. For expression profiling, 15 K spotted cDNA microarrays manufactured by the Cancer Research UK DNA Microarray Facility were used. Statistical feature selection was used with a support vector machine (SVM) classifier to determine the best feature set for predicting BRCA1 or BRCA2 heterozygous genotype. To investigate prediction accuracy, a nonprobabilistic classifier (SVM) and a probabilistic Gaussian process classifier were used. RESULTS: In the task of distinguishing BRCA1 and BRCA2 mutation carriers from noncarriers and from each other following radiation-induced DNA damage, the SVM achieved 90%, and the Gaussian process classifier achieved 100% accuracy. This effect could not be achieved without irradiation. In addition, the SVM identified a set of BRCA genotype predictor genes. CONCLUSIONS: We conclude that after irradiation-induced DNA damage, BRCA1 and BRCA2 mutation carrier cells have a distinctive expression phenotype, and this may have a future role in predicting genotypes, with application to clinical detection and classification of mutations.

Kote-Jarai, Z. Jugurnauth, S. Mulholland, S. Leongamornlert, D.A. Guy, M. Edwards, S. Tymrakiewitcz, M. O'Brien, L. Hall, A. Wilkinson, R. Al Olama, A.A. Morrison, J. Muir, K. Neal, D. Donovan, J. Hamdy, F. Easton, D.F. Eeles, R. UKGPCS Collaborators, . British Association of Urological Surgeons' Section of Oncology, (2009) A recurrent truncating germline mutation in the BRIP1/FANCJ gene and susceptibility to prostate cancer.. Show Abstract full text

Although prostate cancer (PrCa) is one of the most common cancers in men in Western countries, little is known about the inherited factors that influence PrCa risk. On the basis of the fact that BRIP1/FANCJ interacts with BRCA1 and functions as a regulator of DNA double-strand break repair pathways, and that germline mutations within the BRIP1/FANCJ gene predispose to breast cancer, we chose this gene as a candidate for mutation screening in familial and young-onset PrCa cases. We identified a truncating mutation, R798X, in the BRIP1/FANCJ gene in 4 out of 2714 UK PrCa cases enriched for familial (2 out of 641; 0.3%) and young-onset cases (2 out of 2073; 0.1%). On screening 2045 controls from the UK population, we found one R798X sequence alteration (0.05%; odds ratio 2.4 (95% CI 0.25-23.4)). In addition, using our data from a genome-wide association study, we analysed 25 SNPs in the genomic region of the BRIP1/FANCJ gene. Two SNPs showed evidence of association with familial and young-onset PrCa (rs6504074; P(trend)=0.04 and rs8076727; P(trend)=0.01). These results suggest that truncating mutations in BRIP1/FANCJ might confer an increased risk of PrCa and common SNPs might also contribute to the alteration of risk, but larger case-control series will be required to confirm or refute this association.

Kote-Jarai, Z. Leongamornlert, D. Tymrakiewicz, M. Field, H. Guy, M. Al Olama, A.A. Morrison, J. O'Brien, L. Wilkinson, R. Hall, A. Sawyer, E. Muir, K. Hamdy, F. Donovan, J. Neal, D. Easton, D. Eeles, R (2010) Mutation analysis of the MSMB gene in familial prostate cancer.. Show Abstract full text

BACKGROUND: MSMB, a gene coding for beta-microseminoprotein, has been identified as a candidate susceptibility gene for prostate cancer (PrCa) in two genome-wide association studies (GWAS). SNP rs10993994 is 2 bp upstream of the transcription initiation site of MSMB and was identified as an associated PrCa risk variant. The MSMB protein is underexpressed in PrCa and it was previously proposed to be an independent marker for the recurrence of cancer after radical prostatectomy. METHODS: In this study, the coding region of this gene and 1500 bp upstream of the 5'UTR has been sequenced in germline DNA in 192 PrCa patients with family history. To evaluate the possible effects of these variants we used in silico analysis. RESULTS: No deleterious mutations were identified, however, nine new sequence variants were found, most of these in the promoter and 5'UTR region. In silico analysis suggests that four of these SNPs are likely to have some effect on gene expression either by affecting ubiquitous or prostate-specific transcription factor (TF)-binding sites or modifying splicing efficiency. INTERPRETATION: We conclude that MSMB is unlikely to be a familial PrCa gene and propose that the high-risk alleles of the SNPs in the 5'UTR effect PrCa risk by modifying MSMB gene expression in response to hormones in a tissue-specific manner.

Whitaker, H.C. Warren, A.Y. Eeles, R. Kote-Jarai, Z. Neal, D.E (2010) The Potential Value of Microseminoprotein-β as a Prostate Cancer Biomarker and Therapeutic Target.
Barwell, J. Pangon, L. Georgiou, A. Kesterton, I. Langman, C. Arden-Jones, A. Bancroft, E. Salmon, A. Locke, I. Kote-Jarai, Z. MorriS, J.R. Solomon, E. Berg, J. DochertY, Z. Camplejohn, R. Eeles, R. Hodgson, S.V (2007) Lymphocyte radiosensitivity in <i>BRCA1</i> and <i>BRCA2</i> mutation carriers and implications for breast cancer susceptibility.
Eeles, R. Edwards, S. Kote-Jarai, Z. Hamoudi, R. Deamaley, D. Ardem-Jones, A. Murkin, A (2001) 2% of men with early onset prostate cancer harbour deleterious germline mutations in the BRCA2 gene.. full text
Kote-Jarai, Z. Kadouri, L. Durocher, F. Easton, D.F. Huber, A. Abeliovich, D. Glaser, B. Peretz, T. Eeles, R (2001) CAG repeat length in the gene AIB1 gene modifies breast cancer risk in BRCA1/2 mutation carriers.. full text
Macinnis, R.J. Antoniou, A.C. Eeles, R.A. Severi, G. Al Olama, A.A. McGuffog, L. Kote-Jarai, Z. Guy, M. O'Brien, L.T. Hall, A.L. Wilkinson, R.A. Sawyer, E. Ardern-Jones, A.T. Dearnaley, D.P. Horwich, A. Khoo, V.S. Parker, C.C. Huddart, R.A. Van As, N. McCredie, M.R. English, D.R. Giles, G.G. Hopper, J.L. Easton, D.F (2011) A risk prediction algorithm based on family history and common genetic variants: application to prostate cancer with potential clinical impact.. Show Abstract full text

Genome wide association studies have identified several single nucleotide polymorphisms (SNPs) that are independently associated with small increments in risk of prostate cancer, opening up the possibility for using such variants in risk prediction. Using segregation analysis of population-based samples of 4,390 families of prostate cancer patients from the UK and Australia, and assuming all familial aggregation has genetic causes, we previously found that the best model for the genetic susceptibility to prostate cancer was a mixed model of inheritance that included both a recessive major gene component and a polygenic component (P) that represents the effect of a large number of genetic variants each of small effect, where . Based on published studies of 26 SNPs that are currently known to be associated with prostate cancer, we have extended our model to incorporate these SNPs by decomposing the polygenic component into two parts: a polygenic component due to the known susceptibility SNPs, , and the residual polygenic component due to the postulated but as yet unknown genetic variants, . The resulting algorithm can be used for predicting the probability of developing prostate cancer in the future based on both SNP profiles and explicit family history information. This approach can be applied to other diseases for which population-based family data and established risk variants exist.

Rahman, A.A.U. Lophatananon, A. Lobaz, J. Robinson, F. Brown, S.S. Parker, T. Easton, D. Kote-Jarai, Z. Pocock, R. Dearnaley, D. Guy, M. Wilkinson, R.A. Hall, A.L. Sawyer, E. Page, E. Liu, J.-.F. Eeles, R.A. Muir, K.R (2011) OMEGA 3 POLYUNSATURATED FATTY ACIDS (PUFAS) AND RISK OF EARLY ONSET PROSTATE CANCER. full text
Kote-Jarai, Z. Leongamornlert, D. Saunders, E. Tymrakiewicz, M. Castro, E. Mahmud, N. Guy, M. Edwards, S. O'Brien, L. Sawyer, E. Hall, A. Wilkinson, R. Dadaev, T. Goh, C. Easton, D. Goldgar, D. Eeles, R (2011) <i>BRCA2</i> is a moderate penetrance gene contributing to young-onset prostate cancer: implications for genetic testing in prostate cancer patients.
Lose, F. Batra, J. O'Mara, T. Fahey, P. Marquart, L. Eeles, R.A. Easton, D.F. Al Olama, A.A. Kote-Jarai, Z. Guy, M. Muir, K. Lophatananon, A. Rahman, A.A. Neal, D.E. Hamdy, F.C. Donovan, J.L. Chambers, S. Gardiner, R.A. Aitken, J.F. Yaxley, J. Alexander, K. Clements, J.A. Spurdle, A.B. Kedda, M.-.A. Australian Prostate Cancer BioResource, (2013) Common variation in Kallikrein genes KLK5, KLK6, KLK12, and KLK13 and risk of prostate cancer and tumor aggressiveness.. Show Abstract full text

The human tissue Kallikrein family consists of 15 genes with the majority shown to be differentially expressed in cancers and/or indicators of cancer prognosis. We sought to elucidate the role of common genetic variation in four of the Kallikrein genes, KLK5, KLK6, KLK12, and KLK13, in prostate cancer risk and tumor aggressiveness. Genotyping of all 22 tagging single nucleotide polymorphisms (tagSNPs) in the KLK5, KLK6, KLK12, and KLK13 genes was performed in approximately 1,000 prostate cancer cases and 1,300 male controls from Australia. Data from any positive results were also accessed for 1,844 cases and 1,886 controls from a previously published prostate cancer genome-wide association study set from the United Kingdom. For one SNP in KLK12, rs3865443, there was evidence for association with prostate cancer risk of similar direction and magnitude in the replication set to that seen in the Australian cohort. We conducted genotyping of a further 309 prostate cancer cases, and combined analyses revealed an increased risk of prostate cancer for carriers of the rare homozygous genotype for rs3865443 (OR 1.28, 95% CI 1.04-1.57; P = 0.018). No other tagSNPs in the KLK5, KLK6, and KLK13 genes were consistently associated with prostate cancer risk or tumor aggressiveness. Analysis of a combined sample of 3,153 cases and 3,199 controls revealed the KLK12 tagSNP rs3865443 to be marginally statistically significantly associated with risk of prostate cancer. Considering the total number of SNPs investigated in this study, this finding should be interpreted cautiously and requires additional validation from very large datasets such as those of the Prostate Cancer Association group to investigate cancer associated alterations (PRACTICAL) Consortium.

Goh, C.L. Schumacher, F.R. Easton, D. Muir, K. Henderson, B. Kote-Jarai, Z. Eeles, R.A (2012) Genetic variants associated with predisposition to prostate cancer and potential clinical implications.. Show Abstract full text

Prostate cancer is the commonest cancer in the developed world. There is an inherited component to this disease as shown in familial and twin studies. However, the discovery of these variants has been difficult. The emergence of genome-wide association studies has led to the identification of over 46 susceptibility loci. Their clinical utility to predict risk, response to treatment, or treatment toxicity, remains undefined. Large consortia are needed to achieve adequate statistical power to answer these genetic-clinical and genetic-epidemiological questions. International collaborations are currently underway to link genetic with clinical/epidemiological data to develop risk prediction models, which could direct screening and treatment programs.

Leongamornlert, D. Mahmud, N. Tymrakiewicz, M. Saunders, E. Dadaev, T. Castro, E. Goh, C. Govindasami, K. Guy, M. O'Brien, L. Sawyer, E. Hall, A. Wilkinson, R. Easton, D. UKGPCS Collaborators, . Goldgar, D. Eeles, R. Kote-Jarai, Z (2012) Germline BRCA1 mutations increase prostate cancer risk.. Show Abstract full text

BACKGROUND: Prostate cancer (PrCa) is one of the most common cancers affecting men but its aetiology is poorly understood. Family history of PrCa, particularly at a young age, is a strong risk factor. There have been previous reports of increased PrCa risk in male BRCA1 mutation carriers in female breast cancer families, but there is a controversy as to whether this risk is substantiated. We sought to evaluate the role of germline BRCA1 mutations in PrCa predisposition by performing a candidate gene study in a large UK population sample set. METHODS: We screened 913 cases aged 36–86 years for germline BRCA1 mutation, with the study enriched for cases with an early age of onset. We analysed the entire coding region of the BRCA1 gene using Sanger sequencing. Multiplex ligation-dependent probe amplification was also used to assess the frequency of large rearrangements in 460 cases. RESULTS: We identified 4 deleterious mutations and 45 unclassified variants (UV). The frequency of deleterious BRCA1 mutation in this study is 0.45%; three of the mutation carriers were affected at age 65 years and one developed PrCa at 69 years. Using previously estimated population carrier frequencies, deleterious BRCA1 mutations confer a relative risk of PrCa of ~3.75-fold, (95% confidence interval 1.02–9.6) translating to a 8.6% cumulative risk by age 65. CONCLUSION: This study shows evidence for an increased risk of PrCa in men who harbour germline mutations in BRCA1. This could have a significant impact on possible screening strategies and targeted treatments.

Killick, E. Bancroft, E. Kote-Jarai, Z. Eeles, R (2012) Beyond prostate-specific antigen - future biomarkers for the early detection and management of prostate cancer.. Show Abstract full text

Prostate-specific antigen is currently commonly used as a screening biomarker for prostate cancer, but it has limitations in both sensitivity and specificity. The development of novel biomarkers for early cancer detection has the potential to improve survival, reduce unnecessary investigations and benefit the health economy. Here we review the use and limitations of prostate-specific antigen and its subtypes, urinary biomarkers including PCA3, alpha-methylacyl-CoA racemase, the TMPRSS2-ERG fusion gene and microseminoprotein-beta, and other novel markers in both serum and urine. Many of these biomarkers are at early stages of development and require evaluation in prospective trials to determine their potential usefulness in clinical practice. Genetic profiling may allow for the targeting of high-risk populations for screening and may offer the opportunity to combine biomarker results with genotype to aid risk assessment.

Orozco, G. Goh, C.L. Al Olama, A.A. Benlloch-Garcia, S. Govindasami, K. Guy, M. Muir, K.R. Giles, G.G. Severi, G. Neal, D.E. Hamdy, F.C. Donovan, J.L. Kote-Jarai, Z. Easton, D.F. Eyre, S. Eeles, R.A (2013) Common genetic variants associated with disease from genome-wide association studies are mutually exclusive in prostate cancer and rheumatoid arthritis.. Show Abstract full text

UNLABELLED: WHAT'S KNOWN ON THE SUBJECT? AND WHAT DOES THE STUDY ADD?: The link between inflammation and cancer has long been reported and inflammation is thought to play a role in the pathogenesis of many cancers, including prostate cancer (PrCa). Over the last 5 years, genome-wide association studies (GWAS) have reported numerous susceptibility loci that predispose individuals to many different traits. The present study aims to ascertain if there are common genetic risk profiles that might predispose individuals to both PrCa and the autoimmune inflammatory condition, rheumatoid arthritis. These results could have potential public heath impact in terms of screening and chemoprevention. OBJECTIVES: To investigate if potential common pathways exist for the pathogenesis of autoimmune disease and prostate cancer (PrCa). To ascertain if the single nucleotide polymorphisms (SNPs) reported by genome-wide association studies (GWAS) as being associated with susceptibility to PrCa are also associated with susceptibility to the autoimmune disease rheumatoid arthritis (RA). MATERIALS AND METHODS: The original Wellcome Trust Case Control Consortium (WTCCC) UK RA GWAS study was expanded to include a total of 3221 cases and 5272 controls. In all, 37 germline autosomal SNPs at genome-wide significance associated with PrCa risk were identified from a UK/Australian PrCa GWAS. Allele frequencies were compared for these 37 SNPs between RA cases and controls using a chi-squared trend test and corrected for multiple testing (Bonferroni). RESULTS: In all, 33 SNPs were able to be analysed in the RA dataset. Proxies could not be located for the SNPs in 3q26, 5p15 and for two SNPs in 17q12. After applying a Bonferroni correction for the number of SNPs tested, the SNP mapping to CCHCR1 (rs130067) retained statistically significant evidence for association (P = 6 × 10(-4) ; odds ratio [OR] = 1.15, 95% CI: 1.06-1.24); this has also been associated with psoriasis. However, further analyses showed that the association of this allele was due to confounding by RA-associated HLA-DRB1 alleles. CONCLUSIONS: There is currently no evidence that SNPs associated with PrCa at genome-wide significance are associated with the development of RA. Studies like this are important in determining if common genetic risk profiles might predispose individuals to many diseases, which could have implications for public health in terms of screening and chemoprevention.

Cheng, I. Chen, G.K. Nakagawa, H. He, J. Wan, P. Laurie, C.C. Shen, J. Sheng, X. Pooler, L.C. Crenshaw, A.T. Mirel, D.B. Takahashi, A. Kubo, M. Nakamura, Y. Al Olama, A.A. Benlloch, S. Donovan, J.L. Guy, M. Hamdy, F.C. Kote-Jarai, Z. Neal, D.E. Wilkens, L.R. Monroe, K.R. Stram, D.O. Muir, K. Eeles, R.A. Easton, D.F. Kolonel, L.N. Henderson, B.E. Le Marchand, L. Haiman, C.A (2012) Evaluating genetic risk for prostate cancer among Japanese and Latinos.. Show Abstract full text

BACKGROUND: There have been few genome-wide association studies (GWAS) of prostate cancer among diverse populations. To search for novel prostate cancer risk variants, we conducted GWAS of prostate cancer in Japanese and Latinos. In addition, we tested prostate cancer risk variants and developed genetic risk models of prostate cancer for Japanese and Latinos. METHODS: Our first-stage GWAS of prostate cancer included Japanese (cases/controls = 1,033/1,042) and Latino (cases/controls = 1,043/1,057) from the Multiethnic Cohort (MEC). Significant associations from stage I (P < 1.0 × 10(-4)) were examined in silico in GWAS of prostate cancer (stage II) in Japanese (cases/controls = 1,583/3,386) and Europeans (cases/controls = 1,854/1,894). RESULTS: No novel stage I single-nucleotide polymorphism (SNP) outside of known risk regions reached genome-wide significance. For Japanese, in stage I, the most notable putative novel association was seen with 10 SNPs (P ≤ 8.0 × 10(-6)) at chromosome 2q33; however, this was not replicated in stage II. For Latinos, the most significant association was observed with rs17023900 at the known 3p12 risk locus (stage I: OR = 1.45; P = 7.01 × 10(-5) and stage II: OR = 1.58; P = 3.05 × 10(-7)). The majority of the established risk variants for prostate cancer, 79% and 88%, were positively associated with prostate cancer in Japanese and Latinos (stage I), respectively. The cumulative effects of these variants significantly influence prostate cancer risk (OR per allele = 1.10; P = 2.71 × 10(-25) and OR = 1.07; P = 1.02 × 10(-16) for Japanese and Latinos, respectively). CONCLUSION AND IMPACT: Our GWAS of prostate cancer did not identify novel genome-wide significant variants. However, our findings show that established risk variants for prostate cancer significantly contribute to risk among Japanese and Latinos.

Amin Al Olama, A. Kote-Jarai, Z. Schumacher, F.R. Wiklund, F. Berndt, S.I. Benlloch, S. Giles, G.G. Severi, G. Neal, D.E. Hamdy, F.C. Donovan, J.L. Hunter, D.J. Henderson, B.E. Thun, M.J. Gaziano, M. Giovannucci, E.L. Siddiq, A. Travis, R.C. Cox, D.G. Canzian, F. Riboli, E. Key, T.J. Andriole, G. Albanes, D. Hayes, R.B. Schleutker, J. Auvinen, A. Tammela, T.L.J. Weischer, M. Stanford, J.L. Ostrander, E.A. Cybulski, C. Lubinski, J. Thibodeau, S.N. Schaid, D.J. Sorensen, K.D. Batra, J. Clements, J.A. Chambers, S. Aitken, J. Gardiner, R.A. Maier, C. Vogel, W. Dörk, T. Brenner, H. Habuchi, T. Ingles, S. John, E.M. Dickinson, J.L. Cannon-Albright, L. Teixeira, M.R. Kaneva, R. Zhang, H.-.W. Lu, Y.-.J. Park, J.Y. Cooney, K.A. Muir, K.R. Leongamornlert, D.A. Saunders, E. Tymrakiewicz, M. Mahmud, N. Guy, M. Govindasami, K. O'Brien, L.T. Wilkinson, R.A. Hall, A.L. Sawyer, E.J. Dadaev, T. Morrison, J. Dearnaley, D.P. Horwich, A. Huddart, R.A. Khoo, V.S. Parker, C.C. Van As, N. Woodhouse, C.J. Thompson, A. Dudderidge, T. Ogden, C. Cooper, C.S. Lophatonanon, A. Southey, M.C. Hopper, J.L. English, D. Virtamo, J. Le Marchand, L. Campa, D. Kaaks, R. Lindstrom, S. Diver, W.R. Gapstur, S. Yeager, M. Cox, A. Stern, M.C. Corral, R. Aly, M. Isaacs, W. Adolfsson, J. Xu, J. Zheng, S.L. Wahlfors, T. Taari, K. Kujala, P. Klarskov, P. Nordestgaard, B.G. Røder, M.A. Frikke-Schmidt, R. Bojesen, S.E. FitzGerald, L.M. Kolb, S. Kwon, E.M. Karyadi, D.M. Orntoft, T.F. Borre, M. Rinckleb, A. Luedeke, M. Herkommer, K. Meyer, A. Serth, J. Marthick, J.R. Patterson, B. Wokolorczyk, D. Spurdle, A. Lose, F. McDonnell, S.K. Joshi, A.D. Shahabi, A. Pinto, P. Santos, J. Ray, A. Sellers, T.A. Lin, H.-.Y. Stephenson, R.A. Teerlink, C. Muller, H. Rothenbacher, D. Tsuchiya, N. Narita, S. Cao, G.-.W. Slavov, C. Mitev, V. UK Genetic Prostate Cancer Study Collaborators/British Association of Urological Surgeons' Section of Oncology, . UK ProtecT Study Collaborators, . Australian Prostate Cancer Bioresource, . PRACTICAL Consortium, . Chanock, S. Gronberg, H. Haiman, C.A. Kraft, P. Easton, D.F. Eeles, R.A (2013) A meta-analysis of genome-wide association studies to identify prostate cancer susceptibility loci associated with aggressive and non-aggressive disease.. Show Abstract full text

Genome-wide association studies (GWAS) have identified multiple common genetic variants associated with an increased risk of prostate cancer (PrCa), but these explain less than one-third of the heritability. To identify further susceptibility alleles, we conducted a meta-analysis of four GWAS including 5953 cases of aggressive PrCa and 11 463 controls (men without PrCa). We computed association tests for approximately 2.6 million SNPs and followed up the most significant SNPs by genotyping 49 121 samples in 29 studies through the international PRACTICAL and BPC3 consortia. We not only confirmed the association of a PrCa susceptibility locus, rs11672691 on chromosome 19, but also showed an association with aggressive PrCa [odds ratio = 1.12 (95% confidence interval 1.03-1.21), P = 1.4 × 10(-8)]. This report describes a genetic variant which is associated with aggressive PrCa, which is a type of PrCa associated with a poorer prognosis.

Turnbull, C.A. Ruark, E. Seal, S. McDonald, H. Zhang, F. Elliot, A. Lau, K. Perdeaux, E. Rapley, E. Eeles, R. Peto, J. Kote-Jarai, Z. Muir, K. Nsengimana, J. Shipley, J. UKTCC, . Bishop, D.T. Stratton, M. Easton, D.F. Huddart, R. Rahman, N () Identification of nine new loci for testicular cancer, including variants located near DAZL and PRDM14, genes involved in germ cell development. Show Abstract full text

Testicular germ cell tumor (TGCT) is the most common cancer in young men and is notable for its high familial risks. To date, six loci associated with TGCT have been reported. From GWAS analysis of 307,291 SNPs in 986 cases and 4,946 controls, we selected for follow-up 694 SNPs, which we genotyped in a further 1,064 TGCT cases and 10,082 controls from the UK. We identified SNPs at nine new loci showing association with TGCT (P<5x10-8), at 1q22, 1q24.1, 3p24.3, 4q24, 5q31.1, 8q13.3, 16q12.1, 17q22 and 21q22.3, which together account for an additional 4-6% of the familial risk of TGCT. The loci include genes plausibly related to TGCT development. PRDM14, at 8q13.3, is essential for early germ cell specification whilst DAZL, at 3p24.3, is required for regulation of germ cell developmen. Furthermore, PITX1, at 5q31.1 regulates TERT expression, and is the third TGCT locus implicated in telomerase regulation.

Eeles, R.A. Olama, A.A.A. Benlloch, S. Saunders, E.J. Leongamornlert, D.A. Tymrakiewicz, M. Ghoussaini, M. Luccarini, C. Dennis, J. Jugurnauth-Little, S. Dadaev, T. Neal, D.E. Hamdy, F.C. Donovan, J.L. Muir, K. Giles, G.G. Severi, G. Wiklund, F. Gronberg, H. Haiman, C.A. Schumacher, F. Henderson, B.E. Le Marchand, L. Lindstrom, S. Kraft, P. Hunter, D.J. Gapstur, S. Chanock, S.J. Berndt, S.I. Albanes, D. Andriole, G. Schleutker, J. Weischer, M. Canzian, F. Riboli, E. Key, T.J. Travis, R.C. Campa, D. Ingles, S.A. John, E.M. Hayes, R.B. Pharoah, P.D.P. Pashayan, N. Khaw, K.-.T. Stanford, J.L. Ostrander, E.A. Signorello, L.B. Thibodeau, S.N. Schaid, D. Maier, C. Vogel, W. Kibel, A.S. Cybulski, C. Lubinski, J. Cannon-Albright, L. Brenner, H. Park, J.Y. Kaneva, R. Batra, J. Spurdle, A.B. Clements, J.A. Teixeira, M.R. Dicks, E. Lee, A. Dunning, A.M. Baynes, C. Conroy, D. Maranian, M.J. Ahmed, S. Govindasami, K. Guy, M. Wilkinson, R.A. Sawyer, E.J. Morgan, A. Dearnaley, D.P. Horwich, A. Huddart, R.A. Khoo, V.S. Parker, C.C. Van As, N.J. Woodhouse, C.J. Thompson, A. Dudderidge, T. Ogden, C. Cooper, C.S. Lophatananon, A. Cox, A. Southey, M.C. Hopper, J.L. English, D.R. Aly, M. Adolfsson, J. Xu, J. Zheng, S.L. Yeager, M. Kaaks, R. Diver, W.R. Gaudet, M.M. Stern, M.C. Corral, R. Joshi, A.D. Shahabi, A. Wahlfors, T. Tammela, T.L.J. Auvinen, A. Virtamo, J. Klarskov, P. Nordestgaard, B.G. Røder, M.A. Nielsen, S.F. Bojesen, S.E. Siddiq, A. Fitzgerald, L.M. Kolb, S. Kwon, E.M. Karyadi, D.M. Blot, W.J. Zheng, W. Cai, Q. McDonnell, S.K. Rinckleb, A.E. Drake, B. Colditz, G. Wokolorczyk, D. Stephenson, R.A. Teerlink, C. Muller, H. Rothenbacher, D. Sellers, T.A. Lin, H.-.Y. Slavov, C. Mitev, V. Lose, F. Srinivasan, S. Maia, S. Paulo, P. Lange, E. Cooney, K.A. Antoniou, A.C. Vincent, D. Bacot, F. Tessier, D.C. COGS–Cancer Research UK GWAS–ELLIPSE (part of GAME-ON) Initiative, . Australian Prostate Cancer Bioresource, . UK Genetic Prostate Cancer Study Collaborators/British Association of Urological Surgeons' Section of Oncology, . UK ProtecT (Prostate testing for cancer and Treatment) Study Collaborators, . PRACTICAL (Prostate Cancer Association Group to Investigate Cancer-Associated Alterations in the Genome) Consortium, . Kote-Jarai, Z. Easton, D.F (2013) Identification of 23 new prostate cancer susceptibility loci using the iCOGS custom genotyping array.. Show Abstract full text

Prostate cancer is the most frequently diagnosed cancer in males in developed countries. To identify common prostate cancer susceptibility alleles, we genotyped 211,155 SNPs on a custom Illumina array (iCOGS) in blood DNA from 25,074 prostate cancer cases and 24,272 controls from the international PRACTICAL Consortium. Twenty-three new prostate cancer susceptibility loci were identified at genome-wide significance (P < 5 × 10(-8)). More than 70 prostate cancer susceptibility loci, explaining ∼30% of the familial risk for this disease, have now been identified. On the basis of combined risks conferred by the new and previously known risk loci, the top 1% of the risk distribution has a 4.7-fold higher risk than the average of the population being profiled. These results will facilitate population risk stratification for clinical studies.

Kote-Jarai, Z. Saunders, E.J. Leongamornlert, D.A. Tymrakiewicz, M. Dadaev, T. Jugurnauth-Little, S. Ross-Adams, H. Al Olama, A.A. Benlloch, S. Halim, S. Russell, R. Dunning, A.M. Luccarini, C. Dennis, J. Neal, D.E. Hamdy, F.C. Donovan, J.L. Muir, K. Giles, G.G. Severi, G. Wiklund, F. Gronberg, H. Haiman, C.A. Schumacher, F. Henderson, B.E. Le Marchand, L. Lindstrom, S. Kraft, P. Hunter, D.J. Gapstur, S. Chanock, S. Berndt, S.I. Albanes, D. Andriole, G. Schleutker, J. Weischer, M. Canzian, F. Riboli, E. Key, T.J. Travis, R.C. Campa, D. Ingles, S.A. John, E.M. Hayes, R.B. Pharoah, P. Khaw, K.-.T. Stanford, J.L. Ostrander, E.A. Signorello, L.B. Thibodeau, S.N. Schaid, D. Maier, C. Vogel, W. Kibel, A.S. Cybulski, C. Lubinski, J. Cannon-Albright, L. Brenner, H. Park, J.Y. Kaneva, R. Batra, J. Spurdle, A. Clements, J.A. Teixeira, M.R. Govindasami, K. Guy, M. Wilkinson, R.A. Sawyer, E.J. Morgan, A. Dicks, E. Baynes, C. Conroy, D. Bojesen, S.E. Kaaks, R. Vincent, D. Bacot, F. Tessier, D.C. COGS-CRUK GWAS-ELLIPSE (Part of GAME-ON) Initiative, . UK Genetic Prostate Cancer Study Collaborators/British Association of Urological Surgeons' Section of Oncology, . UK ProtecT Study Collaborators, . PRACTICAL Consortium, . Easton, D.F. Eeles, R.A (2013) Fine-mapping identifies multiple prostate cancer risk loci at 5p15, one of which associates with TERT expression.. Show Abstract full text

Associations between single nucleotide polymorphisms (SNPs) at 5p15 and multiple cancer types have been reported. We have previously shown evidence for a strong association between prostate cancer (PrCa) risk and rs2242652 at 5p15, intronic in the telomerase reverse transcriptase (TERT) gene that encodes TERT. To comprehensively evaluate the association between genetic variation across this region and PrCa, we performed a fine-mapping analysis by genotyping 134 SNPs using a custom Illumina iSelect array or Sequenom MassArray iPlex, followed by imputation of 1094 SNPs in 22 301 PrCa cases and 22 320 controls in The PRACTICAL consortium. Multiple stepwise logistic regression analysis identified four signals in the promoter or intronic regions of TERT that independently associated with PrCa risk. Gene expression analysis of normal prostate tissue showed evidence that SNPs within one of these regions also associated with TERT expression, providing a potential mechanism for predisposition to disease.

Castro, E. Goh, C. Olmos, D. Saunders, E. Leongamornlert, D. Tymrakiewicz, M. Mahmud, N. Dadaev, T. Govindasami, K. Guy, M. Sawyer, E. Wilkinson, R. Ardern-Jones, A. Ellis, S. Frost, D. Peock, S. Evans, D.G. Tischkowitz, M. Cole, T. Davidson, R. Eccles, D. Brewer, C. Douglas, F. Porteous, M.E. Donaldson, A. Dorkins, H. Izatt, L. Cook, J. Hodgson, S. Kennedy, M.J. Side, L.E. Eason, J. Murray, A. Antoniou, A.C. Easton, D.F. Kote-Jarai, Z. Eeles, R (2013) Germline BRCA mutations are associated with higher risk of nodal involvement, distant metastasis, and poor survival outcomes in prostate cancer.. Show Abstract full text

PURPOSE: To analyze the baseline clinicopathologic characteristics of prostate tumors with germline BRCA1 and BRCA2 (BRCA1/2) mutations and the prognostic value of those mutations on prostate cancer (PCa) outcomes. PATIENTS AND METHODS: This study analyzed the tumor features and outcomes of 2,019 patients with PCa (18 BRCA1 carriers, 61 BRCA2 carriers, and 1,940 noncarriers). The Kaplan-Meier method and Cox regression analysis were used to evaluate the associations between BRCA1/2 status and other PCa prognostic factors with overall survival (OS), cause-specific OS (CSS), CSS in localized PCa (CSS_M0), metastasis-free survival (MFS), and CSS from metastasis (CSS_M1). RESULTS: PCa with germline BRCA1/2 mutations were more frequently associated with Gleason ≥ 8 (P = .00003), T3/T4 stage (P = .003), nodal involvement (P = .00005), and metastases at diagnosis (P = .005) than PCa in noncarriers. CSS was significantly longer in noncarriers than in carriers (15.7 v 8.6 years, multivariable analyses [MVA] P = .015; hazard ratio [HR] = 1.8). For localized PCa, 5-year CSS and MFS were significantly higher in noncarriers (96% v 82%; MVA P = .01; HR = 2.6%; and 93% v 77%; MVA P = .009; HR = 2.7, respectively). Subgroup analyses confirmed the poor outcomes in BRCA2 patients, whereas the role of BRCA1 was not well defined due to the limited size and follow-up in this subgroup. CONCLUSION: Our results confirm that BRCA1/2 mutations confer a more aggressive PCa phenotype with a higher probability of nodal involvement and distant metastasis. BRCA mutations are associated with poor survival outcomes and this should be considered for tailoring clinical management of these patients.

Killick, E. Morgan, R. Launchbury, F. Bancroft, E. Page, E. Castro, E. Kote-Jarai, Z. Aprikian, A. Blanco, I. Clowes, V. Domchek, S. Douglas, F. Eccles, D. Evans, D.G. Harris, M. Kirk, J. Lam, J. Lindeman, G. Mitchell, G. Pachter, N. Selkirk, C. Tucker, K. Zgajnar, J. Eeles, R. Pandha, H (2013) Role of Engrailed-2 (EN2) as a prostate cancer detection biomarker in genetically high risk men.. Show Abstract full text

Controversy surrounds the use of PSA as a biomarker for prostate cancer detection, leaving an unmet need for a novel biomarker in this setting; urinary EN2 may identify individuals with clinically relevant prostate cancer. Male BRCA1 and BRCA2 mutation carriers are at increased risk of clinically significant prostate cancer and may benefit from screening. Urine samples from 413 BRCA1 and BRCA2 mutation carriers and controls were evaluated. Subjects underwent annual PSA screening with diagnostic biopsy triggered by PSA > 3.0 ng/ml; 21 men were diagnosed with prostate cancer. Urinary EN2 levels were measured by ELISA and had a sensitivity of 66.7% and specificity of 89.3% for cancer detection. There was no statistically significant difference in EN2 levels according to genetic status or Gleason score. Urinary EN2 may be useful as a non-invasive early biomarker for prostate cancer detection in genetically high-risk individuals.

Eeles, R. Goh, C. Castro, E. Bancroft, E. Guy, M. Al Olama, A.A. Easton, D. Kote-Jarai, Z (2014) The genetic epidemiology of prostate cancer and its clinical implications.. Show Abstract full text

Worldwide, familial and epidemiological studies have generated considerable evidence of an inherited component to prostate cancer. Indeed, rare highly penetrant genetic mutations have been implicated. Genome-wide association studies (GWAS) have also identified 76 susceptibility loci associated with prostate cancer risk, which occur commonly but are of low penetrance. However, these mutations interact multiplicatively, which can result in substantially increased risk. Currently, approximately 30% of the familial risk is due to such variants. Evaluating the functional aspects of these variants would contribute to our understanding of prostate cancer aetiology and would enable population risk stratification for screening. Furthermore, understanding the genetic risks of prostate cancer might inform predictions of treatment responses and toxicities, with the goal of personalized therapy. However, risk modelling and clinical translational research are needed before we can translate risk profiles generated from these variants into use in the clinical setting for targeted screening and treatment.

Hazelett, D.J. Rhie, S.K. Gaddis, M. Yan, C. Lakeland, D.L. Coetzee, S.G. Ellipse/GAME-ON consortium, . Practical consortium, . Henderson, B.E. Noushmehr, H. Cozen, W. Kote-Jarai, Z. Eeles, R.A. Easton, D.F. Haiman, C.A. Lu, W. Farnham, P.J. Coetzee, G.A (2014) Comprehensive functional annotation of 77 prostate cancer risk loci.. Show Abstract full text

Genome-wide association studies (GWAS) have revolutionized the field of cancer genetics, but the causal links between increased genetic risk and onset/progression of disease processes remain to be identified. Here we report the first step in such an endeavor for prostate cancer. We provide a comprehensive annotation of the 77 known risk loci, based upon highly correlated variants in biologically relevant chromatin annotations--we identified 727 such potentially functional SNPs. We also provide a detailed account of possible protein disruption, microRNA target sequence disruption and regulatory response element disruption of all correlated SNPs at r(2) ≥ 0.88%. 88% of the 727 SNPs fall within putative enhancers, and many alter critical residues in the response elements of transcription factors known to be involved in prostate biology. We define as risk enhancers those regions with enhancer chromatin biofeatures in prostate-derived cell lines with prostate-cancer correlated SNPs. To aid the identification of these enhancers, we performed genomewide ChIP-seq for H3K27-acetylation, a mark of actively engaged enhancers, as well as the transcription factor TCF7L2. We analyzed in depth three variants in risk enhancers, two of which show significantly altered androgen sensitivity in LNCaP cells. This includes rs4907792, that is in linkage disequilibrium (r(2) = 0.91) with an eQTL for NUDT11 (on the X chromosome) in prostate tissue, and rs10486567, the index SNP in intron 3 of the JAZF1 gene on chromosome 7. Rs4907792 is within a critical residue of a strong consensus androgen response element that is interrupted in the protective allele, resulting in a 56% decrease in its androgen sensitivity, whereas rs10486567 affects both NKX3-1 and FOXA-AR motifs where the risk allele results in a 39% increase in basal activity and a 28% fold-increase in androgen stimulated enhancer activity. Identification of such enhancer variants and their potential target genes represents a preliminary step in connecting risk to disease process.

Killick, E. Tymrakiewicz, M. Cieza-Borrella, C. Smith, P. Thompson, D.J. Pooley, K.A. Easton, D.F. Bancroft, E. Page, E. Leongamornlert, D. IMPACT collaborators, . Kote-Jarai, Z. Eeles, R.A (2014) Telomere length shows no association with BRCA1 and BRCA2 mutation status.. Show Abstract full text

This study aimed to determine whether telomere length (TL) is a marker of cancer risk or genetic status amongst two cohorts of BRCA1 and BRCA2 mutation carriers and controls. The first group was a prospective set of 665 male BRCA1/2 mutation carriers and controls (mean age 53 years), all healthy at time of enrollment and blood donation, 21 of whom have developed prostate cancer whilst on study. The second group consisted of 283 female BRCA1/2 mutation carriers and controls (mean age 48 years), half of whom had been diagnosed with breast cancer prior to enrollment. TL was quantified by qPCR from DNA extracted from peripheral blood lymphocytes. Weighted and unweighted Cox regressions and linear regression analyses were used to assess whether TL was associated with BRCA1/2 mutation status or cancer risk. We found no evidence for association between developing cancer or being a BRCA1 or BRCA2 mutation carrier and telomere length. It is the first study investigating TL in a cohort of genetically predisposed males and although TL and BRCA status was previously studied in females our results don't support the previous finding of association between hereditary breast cancer and shorter TL.

Bancroft, E.K. Page, E.C. Castro, E. Lilja, H. Vickers, A. Sjoberg, D. Assel, M. Foster, C.S. Mitchell, G. Drew, K. Mæhle, L. Axcrona, K. Evans, D.G. Bulman, B. Eccles, D. McBride, D. van Asperen, C. Vasen, H. Kiemeney, L.A. Ringelberg, J. Cybulski, C. Wokolorczyk, D. Selkirk, C. Hulick, P.J. Bojesen, A. Skytte, A.-.B. Lam, J. Taylor, L. Oldenburg, R. Cremers, R. Verhaegh, G. van Zelst-Stams, W.A. Oosterwijk, J.C. Blanco, I. Salinas, M. Cook, J. Rosario, D.J. Buys, S. Conner, T. Ausems, M.G. Ong, K.-.R. Hoffman, J. Domchek, S. Powers, J. Teixeira, M.R. Maia, S. Foulkes, W.D. Taherian, N. Ruijs, M. Helderman-van den Enden, A.T. Izatt, L. Davidson, R. Adank, M.A. Walker, L. Schmutzler, R. Tucker, K. Kirk, J. Hodgson, S. Harris, M. Douglas, F. Lindeman, G.J. Zgajnar, J. Tischkowitz, M. Clowes, V.E. Susman, R. Ramón y Cajal, T. Patcher, N. Gadea, N. Spigelman, A. van Os, T. Liljegren, A. Side, L. Brewer, C. Brady, A.F. Donaldson, A. Stefansdottir, V. Friedman, E. Chen-Shtoyerman, R. Amor, D.J. Copakova, L. Barwell, J. Giri, V.N. Murthy, V. Nicolai, N. Teo, S.-.H. Greenhalgh, L. Strom, S. Henderson, A. McGrath, J. Gallagher, D. Aaronson, N. Ardern-Jones, A. Bangma, C. Dearnaley, D. Costello, P. Eyfjord, J. Rothwell, J. Falconer, A. Gronberg, H. Hamdy, F.C. Johannsson, O. Khoo, V. Kote-Jarai, Z. Lubinski, J. Axcrona, U. Melia, J. McKinley, J. Mitra, A.V. Moynihan, C. Rennert, G. Suri, M. Wilson, P. Killick, E. IMPACT Collaborators, . Moss, S. Eeles, R.A (2014) Targeted prostate cancer screening in BRCA1 and BRCA2 mutation carriers: results from the initial screening round of the IMPACT study.. Show Abstract full text

BACKGROUND: Men with germline breast cancer 1, early onset (BRCA1) or breast cancer 2, early onset (BRCA2) gene mutations have a higher risk of developing prostate cancer (PCa) than noncarriers. IMPACT (Identification of Men with a genetic predisposition to ProstAte Cancer: Targeted screening in BRCA1/2 mutation carriers and controls) is an international consortium of 62 centres in 20 countries evaluating the use of targeted PCa screening in men with BRCA1/2 mutations. OBJECTIVE: To report the first year's screening results for all men at enrollment in the study. DESIGN, SETTING AND PARTICIPANTS: We recruited men aged 40-69 yr with germline BRCA1/2 mutations and a control group of men who have tested negative for a pathogenic BRCA1 or BRCA2 mutation known to be present in their families. All men underwent prostate-specific antigen (PSA) testing at enrollment, and those men with PSA >3 ng/ml were offered prostate biopsy. OUTCOME MEASUREMENTS AND STATISTICAL ANALYSIS: PSA levels, PCa incidence, and tumour characteristics were evaluated. The Fisher exact test was used to compare the number of PCa cases among groups and the differences among disease types. RESULTS AND LIMITATIONS: We recruited 2481 men (791 BRCA1 carriers, 531 BRCA1 controls; 731 BRCA2 carriers, 428 BRCA2 controls). A total of 199 men (8%) presented with PSA >3.0 ng/ml, 162 biopsies were performed, and 59 PCas were diagnosed (18 BRCA1 carriers, 10 BRCA1 controls; 24 BRCA2 carriers, 7 BRCA2 controls); 66% of the tumours were classified as intermediate- or high-risk disease. The positive predictive value (PPV) for biopsy using a PSA threshold of 3.0 ng/ml in BRCA2 mutation carriers was 48%-double the PPV reported in population screening studies. A significant difference in detecting intermediate- or high-risk disease was observed in BRCA2 carriers. Ninety-five percent of the men were white, thus the results cannot be generalised to all ethnic groups. CONCLUSIONS: The IMPACT screening network will be useful for targeted PCa screening studies in men with germline genetic risk variants as they are discovered. These preliminary results support the use of targeted PSA screening based on BRCA genotype and show that this screening yields a high proportion of aggressive disease. PATIENT SUMMARY: In this report, we demonstrate that germline genetic markers can be used to identify men at higher risk of prostate cancer. Targeting screening at these men resulted in the identification of tumours that were more likely to require treatment.

Saunders, E.J. Dadaev, T. Leongamornlert, D.A. Jugurnauth-Little, S. Tymrakiewicz, M. Wiklund, F. Al Olama, A.A. Benlloch, S. Neal, D.E. Hamdy, F.C. Donovan, J.L. Giles, G.G. Severi, G. Gronberg, H. Aly, M. Haiman, C.A. Schumacher, F. Henderson, B.E. Lindstrom, S. Kraft, P. Hunter, D.J. Gapstur, S. Chanock, S. Berndt, S.I. Albanes, D. Andriole, G. Schleutker, J. Weischer, M. Nordestgaard, B.G. Canzian, F. Campa, D. Riboli, E. Key, T.J. Travis, R.C. Ingles, S.A. John, E.M. Hayes, R.B. Pharoah, P. Khaw, K.-.T. Stanford, J.L. Ostrander, E.A. Signorello, L.B. Thibodeau, S.N. Schaid, D. Maier, C. Kibel, A.S. Cybulski, C. Cannon-Albright, L. Brenner, H. Park, J.Y. Kaneva, R. Batra, J. Clements, J.A. Teixeira, M.R. Xu, J. Mikropoulos, C. Goh, C. Govindasami, K. Guy, M. Wilkinson, R.A. Sawyer, E.J. Morgan, A. COGS-CRUK GWAS-ELLIPSE (Part of GAME-ON) Initiative, . UK Genetic Prostate Cancer Study Collaborators, . UK ProtecT Study Collaborators, . PRACTICAL Consortium, . Easton, D.F. Muir, K. Eeles, R.A. Kote-Jarai, Z (2014) Fine-mapping the HOXB region detects common variants tagging a rare coding allele: evidence for synthetic association in prostate cancer.. Show Abstract full text

The HOXB13 gene has been implicated in prostate cancer (PrCa) susceptibility. We performed a high resolution fine-mapping analysis to comprehensively evaluate the association between common genetic variation across the HOXB genetic locus at 17q21 and PrCa risk. This involved genotyping 700 SNPs using a custom Illumina iSelect array (iCOGS) followed by imputation of 3195 SNPs in 20,440 PrCa cases and 21,469 controls in The PRACTICAL consortium. We identified a cluster of highly correlated common variants situated within or closely upstream of HOXB13 that were significantly associated with PrCa risk, described by rs117576373 (OR 1.30, P = 2.62×10(-14)). Additional genotyping, conditional regression and haplotype analyses indicated that the newly identified common variants tag a rare, partially correlated coding variant in the HOXB13 gene (G84E, rs138213197), which has been identified recently as a moderate penetrance PrCa susceptibility allele. The potential for GWAS associations detected through common SNPs to be driven by rare causal variants with higher relative risks has long been proposed; however, to our knowledge this is the first experimental evidence for this phenomenon of synthetic association contributing to cancer susceptibility.

Leongamornlert, D. Saunders, E. Dadaev, T. Tymrakiewicz, M. Goh, C. Jugurnauth-Little, S. Kozarewa, I. Fenwick, K. Assiotis, I. Barrowdale, D. Govindasami, K. Guy, M. Sawyer, E. Wilkinson, R. UKGPCS Collaborators, . Antoniou, A.C. Eeles, R. Kote-Jarai, Z (2014) Frequent germline deleterious mutations in DNA repair genes in familial prostate cancer cases are associated with advanced disease.. Show Abstract full text

BACKGROUND: Prostate cancer (PrCa) is one of the most common diseases to affect men worldwide and among the leading causes of cancer-related death. The purpose of this study was to use second-generation sequencing technology to assess the frequency of deleterious mutations in 22 tumour suppressor genes in familial PrCa and estimate the relative risk of PrCa if these genes are mutated. METHODS: Germline DNA samples from 191 men with 3 or more cases of PrCa in their family were sequenced for 22 tumour suppressor genes using Agilent target enrichment and Illumina technology. Analysis for genetic variation was carried out by using a pipeline consisting of BWA, Genome Analysis Toolkit (GATK) and ANNOVAR. Clinical features were correlated with mutation status using standard statistical tests. Modified segregation analysis was used to determine the relative risk of PrCa conferred by the putative loss-of-function (LoF) mutations identified. RESULTS: We discovered 14 putative LoF mutations in 191 samples (7.3%) and these mutations were more frequently associated with nodal involvement, metastasis or T4 tumour stage (P=0.00164). Segregation analysis of probands with European ancestry estimated that LoF mutations in any of the studied genes confer a relative risk of PrCa of 1.94 (95% CI: 1.56-2.42). CONCLUSIONS: These findings show that LoF mutations in DNA repair pathway genes predispose to familial PrCa and advanced disease and therefore warrants further investigation. The clinical utility of these findings will become increasingly important as targeted screening and therapies become more widespread.

Knipe, D.W. Evans, D.M. Kemp, J.P. Eeles, R. Easton, D.F. Kote-Jarai, Z. Al Olama, A.A. Benlloch, S. Donovan, J.L. Hamdy, F.C. Neal, D.E. Smith, G.D. Lathrop, M. Martin, R.M (2014) Genetic variation in prostate-specific antigen-detected prostate cancer and the effect of control selection on genetic association studies.. Show Abstract full text

BACKGROUND: Only a minority of the genetic components of prostate cancer risk have been explained. Some observed associations of SNPs with prostate cancer might arise from associations of these SNPs with circulating prostate-specific antigen (PSA) because PSA values are used to select controls. METHODS: We undertook a genome-wide association study (GWAS) of screen-detected prostate cancer (ProtecT: 1,146 cases and 1,804 controls); meta-analyzed the results with those from the previously published UK Genetic Prostate Cancer Study (1,854 cases and 1,437 controls); investigated associations of SNPs with prostate cancer using either "low" (PSA < 0.5 ng/mL) or "high" (PSA ≥ 3 ng/mL, biopsy negative) PSA controls; and investigated associations of SNPs with PSA. RESULTS: The ProtecT GWAS confirmed previously reported associations of prostate cancer at three loci: 10q11.23, 17q24.3, and 19q13.33. The meta-analysis confirmed associations of prostate cancer with SNPs near four previously identified loci (8q24.21,10q11.23, 17q24.3, and 19q13.33). When comparing prostate cancer cases with low PSA controls, alleles at genetic markers rs1512268, rs445114, rs10788160, rs11199874, rs17632542, rs266849, and rs2735839 were associated with an increased risk of prostate cancer, but the effect-estimates were attenuated to the null when using high PSA controls (Pheterogeneity in effect-estimates < 0.04). We found a novel inverse association of rs9311171-T with circulating PSA. CONCLUSIONS: Differences in effect-estimates for prostate cancer observed when comparing low versus high PSA controls may be explained by associations of these SNPs with PSA. IMPACT: These findings highlight the need for inferences from genetic studies of prostate cancer risk to carefully consider the influence of control selection criteria.

Mikropoulos, C. Goh, C. Leongamornlert, D. Kote-Jarai, Z. Eeles, R (2014) Translating genetic risk factors for prostate cancer to the clinic: 2013 and beyond.. Show Abstract full text

Prostate cancer (PrCa) is the most commonly diagnosed cancer in the male UK population, with over 40,000 new cases per year. PrCa has a complex, polygenic predisposition, due to rare variants such as BRCA and common variants such as single nucleotide polymorphisms (SNPs). With the introduction of genome-wide association studies, 78 susceptibility loci (SNPs) associated with PrCa risk have been identified. Genetic profiling could risk-stratify a population, leading to the discovery of a higher proportion of clinically significant disease and a reduction in the morbidity related to age-based prostate-specific antigen screening. Based on the combined risk of the 78 SNPs identified so far, the top 1% of the risk distribution has a 4.7-times higher risk of developing PrCa compared with the average of the general population.

Al Olama, A.A. Kote-Jarai, Z. Berndt, S.I. Conti, D.V. Schumacher, F. Han, Y. Benlloch, S. Hazelett, D.J. Wang, Z. Saunders, E. Leongamornlert, D. Lindstrom, S. Jugurnauth-Little, S. Dadaev, T. Tymrakiewicz, M. Stram, D.O. Rand, K. Wan, P. Stram, A. Sheng, X. Pooler, L.C. Park, K. Xia, L. Tyrer, J. Kolonel, L.N. Le Marchand, L. Hoover, R.N. Machiela, M.J. Yeager, M. Burdette, L. Chung, C.C. Hutchinson, A. Yu, K. Goh, C. Ahmed, M. Govindasami, K. Guy, M. Tammela, T.L.J. Auvinen, A. Wahlfors, T. Schleutker, J. Visakorpi, T. Leinonen, K.A. Xu, J. Aly, M. Donovan, J. Travis, R.C. Key, T.J. Siddiq, A. Canzian, F. Khaw, K.-.T. Takahashi, A. Kubo, M. Pharoah, P. Pashayan, N. Weischer, M. Nordestgaard, B.G. Nielsen, S.F. Klarskov, P. Røder, M.A. Iversen, P. Thibodeau, S.N. McDonnell, S.K. Schaid, D.J. Stanford, J.L. Kolb, S. Holt, S. Knudsen, B. Coll, A.H. Gapstur, S.M. Diver, W.R. Stevens, V.L. Maier, C. Luedeke, M. Herkommer, K. Rinckleb, A.E. Strom, S.S. Pettaway, C. Yeboah, E.D. Tettey, Y. Biritwum, R.B. Adjei, A.A. Tay, E. Truelove, A. Niwa, S. Chokkalingam, A.P. Cannon-Albright, L. Cybulski, C. Wokołorczyk, D. Kluźniak, W. Park, J. Sellers, T. Lin, H.-.Y. Isaacs, W.B. Partin, A.W. Brenner, H. Dieffenbach, A.K. Stegmaier, C. Chen, C. Giovannucci, E.L. Ma, J. Stampfer, M. Penney, K.L. Mucci, L. John, E.M. Ingles, S.A. Kittles, R.A. Murphy, A.B. Pandha, H. Michael, A. Kierzek, A.M. Blot, W. Signorello, L.B. Zheng, W. Albanes, D. Virtamo, J. Weinstein, S. Nemesure, B. Carpten, J. Leske, C. Wu, S.-.Y. Hennis, A. Kibel, A.S. Rybicki, B.A. Neslund-Dudas, C. Hsing, A.W. Chu, L. Goodman, P.J. Klein, E.A. Zheng, S.L. Batra, J. Clements, J. Spurdle, A. Teixeira, M.R. Paulo, P. Maia, S. Slavov, C. Kaneva, R. Mitev, V. Witte, J.S. Casey, G. Gillanders, E.M. Seminara, D. Riboli, E. Hamdy, F.C. Coetzee, G.A. Li, Q. Freedman, M.L. Hunter, D.J. Muir, K. Gronberg, H. Neal, D.E. Southey, M. Giles, G.G. Severi, G. Breast and Prostate Cancer Cohort Consortium (BPC3), . PRACTICAL (Prostate Cancer Association Group to Investigate Cancer-Associated Alterations in the Genome) Consortium, . COGS (Collaborative Oncological Gene-environment Study) Consortium, . GAME-ON/ELLIPSE Consortium, . Cook, M.B. Nakagawa, H. Wiklund, F. Kraft, P. Chanock, S.J. Henderson, B.E. Easton, D.F. Eeles, R.A. Haiman, C.A (2014) A meta-analysis of 87,040 individuals identifies 23 new susceptibility loci for prostate cancer.. Show Abstract full text

Genome-wide association studies (GWAS) have identified 76 variants associated with prostate cancer risk predominantly in populations of European ancestry. To identify additional susceptibility loci for this common cancer, we conducted a meta-analysis of > 10 million SNPs in 43,303 prostate cancer cases and 43,737 controls from studies in populations of European, African, Japanese and Latino ancestry. Twenty-three new susceptibility loci were identified at association P < 5 × 10(-8); 15 variants were identified among men of European ancestry, 7 were identified in multi-ancestry analyses and 1 was associated with early-onset prostate cancer. These 23 variants, in combination with known prostate cancer risk variants, explain 33% of the familial risk for this disease in European-ancestry populations. These findings provide new regions for investigation into the pathogenesis of prostate cancer and demonstrate the usefulness of combining ancestrally diverse populations to discover risk loci for disease.

Litchfield, K. Sultana, R. Renwick, A. Dudakia, D. Seal, S. Ramsay, E. Powell, S. Elliott, A. Warren-Perry, M. Eeles, R. Peto, J. Kote-Jarai, Z. Muir, K. Nsengimana, J. UKTCC, . Stratton, M.R. Easton, D.F. Bishop, D.T. Huddart, R.A. Rahman, N. Turnbull, C. UKTCC, (2015) Multi-stage genome-wide association study identifies new susceptibility locus for testicular germ cell tumour on chromosome 3q25.. Show Abstract full text

Recent genome-wide association studies (GWAS) and subsequent meta-analyses have identified over 25 SNPs at 18 loci, together accounting for >15% of the genetic susceptibility to testicular germ cell tumour (TGCT). To identify further common SNPs associated with TGCT, here we report a three-stage experiment, involving 4098 cases and 18 972 controls. Stage 1 comprised previously published GWAS analysis of 307 291 SNPs in 986 cases and 4946 controls. In Stage 2, we used previously published customised Illumina iSelect genotyping array (iCOGs) data across 694 SNPs in 1064 cases and 10 082 controls. Here, we report new genotyping of eight SNPs showing some evidence of association in combined analysis of Stage 1 and Stage 2 in an additional 2048 cases of TGCT and 3944 controls (Stage 3). Through fixed-effects meta-analysis across three stages, we identified a novel locus at 3q25.31 (rs1510272) demonstrating association with TGCT [per-allele odds ratio (OR) = 1.16, 95% confidence interval (CI) = 1.06-1.27; P = 1.2 × 10(-9)].

Castro, E. Goh, C. Leongamornlert, D. Saunders, E. Tymrakiewicz, M. Dadaev, T. Govindasami, K. Guy, M. Ellis, S. Frost, D. Bancroft, E. Cole, T. Tischkowitz, M. Kennedy, M.J. Eason, J. Brewer, C. Evans, D.G. Davidson, R. Eccles, D. Porteous, M.E. Douglas, F. Adlard, J. Donaldson, A. Antoniou, A.C. Kote-Jarai, Z. Easton, D.F. Olmos, D. Eeles, R (2015) Effect of BRCA Mutations on Metastatic Relapse and Cause-specific Survival After Radical Treatment for Localised Prostate Cancer.. Show Abstract full text

BACKGROUND: Germline BRCA mutations are associated with worse prostate cancer (PCa) outcomes; however, the most appropriate management for mutation carriers has not yet been investigated. OBJECTIVE: To evaluate the response of BRCA carriers to conventional treatments for localised PCa by analysing metastasis-free survival (MFS) and cause-specific survival (CSS) following radical prostatectomy (RP) or external-beam radiation therapy (RT). DESIGN, SETTING, AND PARTICIPANTS: Tumour features and outcomes of 1302 patients with local/locally advanced PCa (including 67 BRCA mutation carriers) were analysed. RP was undergone by 535 patients (35 BRCA); 767 received RT (32 BRCA). Median follow-up was 64 mo. OUTCOME MEASUREMENTS AND STATISTICAL ANALYSIS: Median survival and 3-, 5-, and 10-yr survival rates were estimated using the Kaplan-Meier method. Generated survival curves were compared using the log-rank test. Cox regression analyses were used to assess the prognostic value of BRCA mutations. RESULTS AND LIMITATIONS: A total of 67 BRCA carriers and 1235 noncarriers were included. At 3, 5, and 10 yr after treatment, 97%, 94%, and 84% of noncarriers and 90%, 72%, and 50% of carriers were free from metastasis (p<0.001). The 3-, 5- and 10-yr CSS rates were significantly better in the noncarrier cohort (99%, 97%, and 85%, respectively) than in carriers (96%, 76%, and 61%, respectively; p<0.001). Multivariate analysis confirmed BRCA mutations as an independent prognostic factor for MFS (hazard ratio [HR]: 2.36; 95% confidence interval [CI], 1.38-4.03; p=0.002) and CSS (HR: 2.17; 95% CI, 1.16-4.07; p=0.016). CONCLUSIONS: BRCA carriers had worse outcomes than noncarriers when conventionally treated for local/locally advanced PCa. PATIENT SUMMARY: Prostate cancer patients with germline BRCA mutations had worse outcomes than noncarriers when conventionally treated with surgery or radiation therapy.

Kote-Jarai, Z. Mikropoulos, C. Leongamornlert, D.A. Dadaev, T. Tymrakiewicz, M. Saunders, E.J. Jones, M. Jugurnauth-Little, S. Govindasami, K. Guy, M. Hamdy, F.C. Donovan, J.L. Neal, D.E. Lane, J.A. Dearnaley, D. Wilkinson, R.A. Sawyer, E.J. Morgan, A. Antoniou, A.C. Eeles, R.A. UK Genetic Prostate Cancer Study Collaborators, and the ProtecT Study Group, (2015) Prevalence of the HOXB13 G84E germline mutation in British men and correlation with prostate cancer risk, tumour characteristics and clinical outcomes.. Show Abstract full text

BACKGROUND: A rare recurrent missense variant in HOXB13 (rs138213197/G84E) was recently reported to be associated with hereditary prostate cancer. Population-based studies have established that, since the frequency of this single-nucleotide polymorphism (SNP) varies between geographic regions, the associated proportion of prostate cancer (PrCa) risk contribution is also highly variable by country. PATIENTS AND METHODS: This is the largest comprehensive case-control study assessing the prevalence of the HOXB13 G84E variant to date and is the first in the UK population. We genotyped 8652 men diagnosed with PrCa within the UK Genetic Prostate Cancer Study (UKGPCS) and 5252 healthy men from the UK ProtecT study. RESULTS: HOXB13 G84E was identified in 0.5% of the healthy controls and 1.5% of the PrCa cases, and it was associated with a 2.93-fold increased risk of PrCa [95% confidence interval (CI) 1.94-4.59; P = 6.27 × 10(-8)]. The risk was even higher among men with family history of PrCa [odds ratio (OR) = 4.53, 95% CI 2.86-7.34; P = 3.1 × 10(-8)] and in young-onset PrCa (diagnosed up to the age of 55 years; OR = 3.11, 95% CI 1.98-5.00; P = 6.1 × 10(-7)). There was no significant association between Gleason Score, presenting prostate specific antigen, tumour-node-metastasis (TNM) stage or NCCN risk group and carrier status. HOXB13 G84E was not associated with overall or cancer-specific survival. We found that the polygenic PrCa risk score (PR score), calculated using the 71 known single-nucleotide polymorphisms (SNPs) associated with PrCa and the HOXB13 G84E variant act multiplicatively on PrCa risk. Based on the estimated prevalence and risk, this rare variant explains ∼1% of the familial risk of PrCa in the UK population. CONCLUSIONS: The clinical importance of HOXB13 G84E in PrCa management has not been established. This variant was found to have no effect on prognostic implications but could be used for stratifying screening, by identifying men at high risk. CLINICAL TRIALS NUMBERS: Prostate Testing for Cancer and Treatment (ProtecT): NCT02044172. UK GENETIC PROSTATE CANCER STUDY: Epidemiology and Molecular Genetics Studies (UKGPCS): NCT01737242.

Pashayan, N. Duffy, S.W. Neal, D.E. Hamdy, F.C. Donovan, J.L. Martin, R.M. Harrington, P. Benlloch, S. Amin Al Olama, A. Shah, M. Kote-Jarai, Z. Easton, D.F. Eeles, R. Pharoah, P.D (2015) Implications of polygenic risk-stratified screening for prostate cancer on overdiagnosis.. Show Abstract full text

PURPOSE: This study aimed to quantify the probability of overdiagnosis of prostate cancer by polygenic risk. METHODS: We calculated the polygenic risk score based on 66 known prostate cancer susceptibility variants for 17,012 men aged 50-69 years (9,404 men identified with prostate cancer and 7,608 with no cancer) derived from three UK-based ongoing studies. We derived the probabilities of overdiagnosis by quartiles of polygenic risk considering that the observed prevalence of screen-detected prostate cancer is a combination of underlying incidence, mean sojourn time (MST), test sensitivity, and overdiagnosis. RESULTS: Polygenic risk quartiles 1 to 4 comprised 9, 18, 25, and 48% of the cases, respectively. For a prostate-specific antigen test sensitivity of 80% and MST of 9 years, 43, 30, 25, and 19% of the prevalent screen-detected cancers in quartiles 1 to 4, respectively, were likely to be overdiagnosed cancers. Overdiagnosis decreased with increasing polygenic risk, with 56% decrease between the lowest and the highest polygenic risk quartiles. CONCLUSION: Targeting screening to men at higher polygenic risk could reduce the problem of overdiagnosis and lead to a better benefit-to-harm balance in screening for prostate cancer.

Cooper, C.S. Eeles, R. Wedge, D.C. Van Loo, P. Gundem, G. Alexandrov, L.B. Kremeyer, B. Butler, A. Lynch, A.G. Camacho, N. Massie, C.E. Kay, J. Luxton, H.J. Edwards, S. Kote-Jarai, Z. Dennis, N. Merson, S. Leongamornlert, D. Zamora, J. Corbishley, C. Thomas, S. Nik-Zainal, S. O'Meara, S. Matthews, L. Clark, J. Hurst, R. Mithen, R. Bristow, R.G. Boutros, P.C. Fraser, M. Cooke, S. Raine, K. Jones, D. Menzies, A. Stebbings, L. Hinton, J. Teague, J. McLaren, S. Mudie, L. Hardy, C. Anderson, E. Joseph, O. Goody, V. Robinson, B. Maddison, M. Gamble, S. Greenman, C. Berney, D. Hazell, S. Livni, N. ICGC Prostate Group, . Fisher, C. Ogden, C. Kumar, P. Thompson, A. Woodhouse, C. Nicol, D. Mayer, E. Dudderidge, T. Shah, N.C. Gnanapragasam, V. Voet, T. Campbell, P. Futreal, A. Easton, D. Warren, A.Y. Foster, C.S. Stratton, M.R. Whitaker, H.C. McDermott, U. Brewer, D.S. Neal, D.E (2015) Analysis of the genetic phylogeny of multifocal prostate cancer identifies multiple independent clonal expansions in neoplastic and morphologically normal prostate tissue.. Show Abstract full text

Genome-wide DNA sequencing was used to decrypt the phylogeny of multiple samples from distinct areas of cancer and morphologically normal tissue taken from the prostates of three men. Mutations were present at high levels in morphologically normal tissue distant from the cancer, reflecting clonal expansions, and the underlying mutational processes at work in morphologically normal tissue were also at work in cancer. Our observations demonstrate the existence of ongoing abnormal mutational processes, consistent with field effects, underlying carcinogenesis. This mechanism gives rise to extensive branching evolution and cancer clone mixing, as exemplified by the coexistence of multiple cancer lineages harboring distinct ERG fusions within a single cancer nodule. Subsets of mutations were shared either by morphologically normal and malignant tissues or between different ERG lineages, indicating earlier or separate clonal cell expansions. Our observations inform on the origin of multifocal disease and have implications for prostate cancer therapy in individual cases.

Gundem, G. Van Loo, P. Kremeyer, B. Alexandrov, L.B. Tubio, J.M.C. Papaemmanuil, E. Brewer, D.S. Kallio, H.M.L. Högnäs, G. Annala, M. Kivinummi, K. Goody, V. Latimer, C. O'Meara, S. Dawson, K.J. Isaacs, W. Emmert-Buck, M.R. Nykter, M. Foster, C. Kote-Jarai, Z. Easton, D. Whitaker, H.C. ICGC Prostate Group, . Neal, D.E. Cooper, C.S. Eeles, R.A. Visakorpi, T. Campbell, P.J. McDermott, U. Wedge, D.C. Bova, G.S (2015) The evolutionary history of lethal metastatic prostate cancer.. Show Abstract full text

Cancers emerge from an ongoing Darwinian evolutionary process, often leading to multiple competing subclones within a single primary tumour. This evolutionary process culminates in the formation of metastases, which is the cause of 90% of cancer-related deaths. However, despite its clinical importance, little is known about the principles governing the dissemination of cancer cells to distant organs. Although the hypothesis that each metastasis originates from a single tumour cell is generally supported, recent studies using mouse models of cancer demonstrated the existence of polyclonal seeding from and interclonal cooperation between multiple subclones. Here we sought definitive evidence for the existence of polyclonal seeding in human malignancy and to establish the clonal relationship among different metastases in the context of androgen-deprived metastatic prostate cancer. Using whole-genome sequencing, we characterized multiple metastases arising from prostate tumours in ten patients. Integrated analyses of subclonal architecture revealed the patterns of metastatic spread in unprecedented detail. Metastasis-to-metastasis spread was found to be common, either through de novo monoclonal seeding of daughter metastases or, in five cases, through the transfer of multiple tumour clones between metastatic sites. Lesions affecting tumour suppressor genes usually occur as single events, whereas mutations in genes involved in androgen receptor signalling commonly involve multiple, convergent events in different metastases. Our results elucidate in detail the complex patterns of metastatic spread and further our understanding of the development of resistance to androgen-deprivation therapy in prostate cancer.

Stegeman, S. Amankwah, E. Klein, K. O'Mara, T.A. Kim, D. Lin, H.-.Y. Permuth-Wey, J. Sellers, T.A. Srinivasan, S. Eeles, R. Easton, D. Kote-Jarai, Z. Amin Al Olama, A. Benlloch, S. Muir, K. Giles, G.G. Wiklund, F. Gronberg, H. Haiman, C.A. Schleutker, J. Nordestgaard, B.G. Travis, R.C. Neal, D. Pharoah, P. Khaw, K.-.T. Stanford, J.L. Blot, W.J. Thibodeau, S. Maier, C. Kibel, A.S. Cybulski, C. Cannon-Albright, L. Brenner, H. Kaneva, R. Teixeira, M.R. PRACTICAL Consortium, . Australian Prostate Cancer BioResource, . Spurdle, A.B. Clements, J.A. Park, J.Y. Batra, J (2015) A Large-Scale Analysis of Genetic Variants within Putative miRNA Binding Sites in Prostate Cancer.. Show Abstract full text

<h4>Unlabelled</h4>Prostate cancer is the second most common malignancy among men worldwide. Genome-wide association studies have identified 100 risk variants for prostate cancer, which can explain approximately 33% of the familial risk of the disease. We hypothesized that a comprehensive analysis of genetic variations found within the 3' untranslated region of genes predicted to affect miRNA binding (miRSNP) can identify additional prostate cancer risk variants. We investigated the association between 2,169 miRSNPs and prostate cancer risk in a large-scale analysis of 22,301 cases and 22,320 controls of European ancestry from 23 participating studies. Twenty-two miRSNPs were associated (P<2.3×10(-5)) with risk of prostate cancer, 10 of which were within 7 genes previously not mapped by GWAS studies. Further, using miRNA mimics and reporter gene assays, we showed that miR-3162-5p has specific affinity for the KLK3 rs1058205 miRSNP T-allele, whereas miR-370 has greater affinity for the VAMP8 rs1010 miRSNP A-allele, validating their functional role.<h4>Significance</h4>Findings from this large association study suggest that a focus on miRSNPs, including functional evaluation, can identify candidate risk loci below currently accepted statistical levels of genome-wide significance. Studies of miRNAs and their interactions with SNPs could provide further insights into the mechanisms of prostate cancer risk.

Amin Al Olama, A. Dadaev, T. Hazelett, D.J. Li, Q. Leongamornlert, D. Saunders, E.J. Stephens, S. Cieza-Borrella, C. Whitmore, I. Benlloch Garcia, S. Giles, G.G. Southey, M.C. Fitzgerald, L. Gronberg, H. Wiklund, F. Aly, M. Henderson, B.E. Schumacher, F. Haiman, C.A. Schleutker, J. Wahlfors, T. Tammela, T.L. Nordestgaard, B.G. Key, T.J. Travis, R.C. Neal, D.E. Donovan, J.L. Hamdy, F.C. Pharoah, P. Pashayan, N. Khaw, K.-.T. Stanford, J.L. Thibodeau, S.N. Mcdonnell, S.K. Schaid, D.J. Maier, C. Vogel, W. Luedeke, M. Herkommer, K. Kibel, A.S. Cybulski, C. Wokołorczyk, D. Kluzniak, W. Cannon-Albright, L. Brenner, H. Butterbach, K. Arndt, V. Park, J.Y. Sellers, T. Lin, H.-.Y. Slavov, C. Kaneva, R. Mitev, V. Batra, J. Clements, J.A. Spurdle, A. Teixeira, M.R. Paulo, P. Maia, S. Pandha, H. Michael, A. Kierzek, A. Govindasami, K. Guy, M. Lophatonanon, A. Muir, K. Viñuela, A. Brown, A.A. PRACTICAL Consortium, . COGS-CRUK GWAS-ELLIPSE (Part of GAME-ON) Initiative, . Australian Prostate Cancer BioResource, . UK Genetic Prostate Cancer Study Collaborators, . UK ProtecT Study Collaborators, . Freedman, M. Conti, D.V. Easton, D. Coetzee, G.A. Eeles, R.A. Kote-Jarai, Z (2015) Multiple novel prostate cancer susceptibility signals identified by fine-mapping of known risk loci among Europeans.. Show Abstract full text

Genome-wide association studies (GWAS) have identified numerous common prostate cancer (PrCa) susceptibility loci. We have fine-mapped 64 GWAS regions known at the conclusion of the iCOGS study using large-scale genotyping and imputation in 25 723 PrCa cases and 26 274 controls of European ancestry. We detected evidence for multiple independent signals at 16 regions, 12 of which contained additional newly identified significant associations. A single signal comprising a spectrum of correlated variation was observed at 39 regions; 35 of which are now described by a novel more significantly associated lead SNP, while the originally reported variant remained as the lead SNP only in 4 regions. We also confirmed two association signals in Europeans that had been previously reported only in East-Asian GWAS. Based on statistical evidence and linkage disequilibrium (LD) structure, we have curated and narrowed down the list of the most likely candidate causal variants for each region. Functional annotation using data from ENCODE filtered for PrCa cell lines and eQTL analysis demonstrated significant enrichment for overlap with bio-features within this set. By incorporating the novel risk variants identified here alongside the refined data for existing association signals, we estimate that these loci now explain ∼38.9% of the familial relative risk of PrCa, an 8.9% improvement over the previously reported GWAS tag SNPs. This suggests that a significant fraction of the heritability of PrCa may have been hidden during the discovery phase of GWAS, in particular due to the presence of multiple independent signals within the same region.

Amin Al Olama, A. Benlloch, S. Antoniou, A.C. Giles, G.G. Severi, G. Neal, D.E. Hamdy, F.C. Donovan, J.L. Muir, K. Schleutker, J. Henderson, B.E. Haiman, C.A. Schumacher, F.R. Pashayan, N. Pharoah, P.D.P. Ostrander, E.A. Stanford, J.L. Batra, J. Clements, J.A. Chambers, S.K. Weischer, M. Nordestgaard, B.G. Ingles, S.A. Sorensen, K.D. Orntoft, T.F. Park, J.Y. Cybulski, C. Maier, C. Doerk, T. Dickinson, J.L. Cannon-Albright, L. Brenner, H. Rebbeck, T.R. Zeigler-Johnson, C. Habuchi, T. Thibodeau, S.N. Cooney, K.A. Chappuis, P.O. Hutter, P. Kaneva, R.P. Foulkes, W.D. Zeegers, M.P. Lu, Y.-.J. Zhang, H.-.W. Stephenson, R. Cox, A. Southey, M.C. Spurdle, A.B. FitzGerald, L. Leongamornlert, D. Saunders, E. Tymrakiewicz, M. Guy, M. Dadaev, T. Little, S.J. Govindasami, K. Sawyer, E. Wilkinson, R. Herkommer, K. Hopper, J.L. Lophatonanon, A. Rinckleb, A.E. Kote-Jarai, Z. Eeles, R.A. Easton, D.F. UK Genetic Prostate Cancer Study Collaborators/British Association of Urological Surgeons' Section of Oncology, . UK ProtecT Study Collaborators, . PRACTICAL Consortium, (2015) Risk Analysis of Prostate Cancer in PRACTICAL, a Multinational Consortium, Using 25 Known Prostate Cancer Susceptibility Loci.. Show Abstract full text

<h4>Background</h4>Genome-wide association studies have identified multiple genetic variants associated with prostate cancer risk which explain a substantial proportion of familial relative risk. These variants can be used to stratify individuals by their risk of prostate cancer.<h4>Methods</h4>We genotyped 25 prostate cancer susceptibility loci in 40,414 individuals and derived a polygenic risk score (PRS). We estimated empirical odds ratios (OR) for prostate cancer associated with different risk strata defined by PRS and derived age-specific absolute risks of developing prostate cancer by PRS stratum and family history.<h4>Results</h4>The prostate cancer risk for men in the top 1% of the PRS distribution was 30.6 (95% CI, 16.4-57.3) fold compared with men in the bottom 1%, and 4.2 (95% CI, 3.2-5.5) fold compared with the median risk. The absolute risk of prostate cancer by age of 85 years was 65.8% for a man with family history in the top 1% of the PRS distribution, compared with 3.7% for a man in the bottom 1%. The PRS was only weakly correlated with serum PSA level (correlation = 0.09).<h4>Conclusions</h4>Risk profiling can identify men at substantially increased or reduced risk of prostate cancer. The effect size, measured by OR per unit PRS, was higher in men at younger ages and in men with family history of prostate cancer. Incorporating additional newly identified loci into a PRS should improve the predictive value of risk profiles.<h4>Impact</h4>We demonstrate that the risk profiling based on SNPs can identify men at substantially increased or reduced risk that could have useful implications for targeted prevention and screening programs.

Szulkin, R. Whitington, T. Eklund, M. Aly, M. Eeles, R.A. Easton, D. Kote-Jarai, Z.S. Amin Al Olama, A. Benlloch, S. Muir, K. Giles, G.G. Southey, M.C. Fitzgerald, L.M. Henderson, B.E. Schumacher, F. Haiman, C.A. Schleutker, J. Wahlfors, T. Tammela, T.L.J. Nordestgaard, B.G. Key, T.J. Travis, R.C. Neal, D.E. Donovan, J.L. Hamdy, F.C. Pharoah, P. Pashayan, N. Khaw, K.-.T. Stanford, J.L. Thibodeau, S.N. McDonnell, S.K. Schaid, D.J. Maier, C. Vogel, W. Luedeke, M. Herkommer, K. Kibel, A.S. Cybulski, C. Lubiński, J. Kluźniak, W. Cannon-Albright, L. Brenner, H. Butterbach, K. Stegmaier, C. Park, J.Y. Sellers, T. Lin, H.-.Y. Slavov, C. Kaneva, R. Mitev, V. Batra, J. Clements, J.A. Australian Prostate Cancer BioResource, . Spurdle, A. Teixeira, M.R. Paulo, P. Maia, S. Pandha, H. Michael, A. Michael, A. Kierzek, A. Practical Consortium, . Gronberg, H. Wiklund, F (2015) Prediction of individual genetic risk to prostate cancer using a polygenic score.. Show Abstract full text

<h4>Background</h4>Polygenic risk scores comprising established susceptibility variants have shown to be informative classifiers for several complex diseases including prostate cancer. For prostate cancer it is unknown if inclusion of genetic markers that have so far not been associated with prostate cancer risk at a genome-wide significant level will improve disease prediction.<h4>Methods</h4>We built polygenic risk scores in a large training set comprising over 25,000 individuals. Initially 65 established prostate cancer susceptibility variants were selected. After LD pruning additional variants were prioritized based on their association with prostate cancer. Six-fold cross validation was performed to assess genetic risk scores and optimize the number of additional variants to be included. The final model was evaluated in an independent study population including 1,370 cases and 1,239 controls.<h4>Results</h4>The polygenic risk score with 65 established susceptibility variants provided an area under the curve (AUC) of 0.67. Adding an additional 68 novel variants significantly increased the AUC to 0.68 (P = 0.0012) and the net reclassification index with 0.21 (P = 8.5E-08). All novel variants were located in genomic regions established as associated with prostate cancer risk.<h4>Conclusions</h4>Inclusion of additional genetic variants from established prostate cancer susceptibility regions improves disease prediction.

Han, Y. Hazelett, D.J. Wiklund, F. Schumacher, F.R. Stram, D.O. Berndt, S.I. Wang, Z. Rand, K.A. Hoover, R.N. Machiela, M.J. Yeager, M. Burdette, L. Chung, C.C. Hutchinson, A. Yu, K. Xu, J. Travis, R.C. Key, T.J. Siddiq, A. Canzian, F. Takahashi, A. Kubo, M. Stanford, J.L. Kolb, S. Gapstur, S.M. Diver, W.R. Stevens, V.L. Strom, S.S. Pettaway, C.A. Al Olama, A.A. Kote-Jarai, Z. Eeles, R.A. Yeboah, E.D. Tettey, Y. Biritwum, R.B. Adjei, A.A. Tay, E. Truelove, A. Niwa, S. Chokkalingam, A.P. Isaacs, W.B. Chen, C. Lindstrom, S. Le Marchand, L. Giovannucci, E.L. Pomerantz, M. Long, H. Li, F. Ma, J. Stampfer, M. John, E.M. Ingles, S.A. Kittles, R.A. Murphy, A.B. Blot, W.J. Signorello, L.B. Zheng, W. Albanes, D. Virtamo, J. Weinstein, S. Nemesure, B. Carpten, J. Leske, M.C. Wu, S.-.Y. Hennis, A.J.M. Rybicki, B.A. Neslund-Dudas, C. Hsing, A.W. Chu, L. Goodman, P.J. Klein, E.A. Zheng, S.L. Witte, J.S. Casey, G. Riboli, E. Li, Q. Freedman, M.L. Hunter, D.J. Gronberg, H. Cook, M.B. Nakagawa, H. Kraft, P. Chanock, S.J. Easton, D.F. Henderson, B.E. Coetzee, G.A. Conti, D.V. Haiman, C.A (2015) Integration of multiethnic fine-mapping and genomic annotation to prioritize candidate functional SNPs at prostate cancer susceptibility regions.. Show Abstract full text

Interpretation of biological mechanisms underlying genetic risk associations for prostate cancer is complicated by the relatively large number of risk variants (n = 100) and the thousands of surrogate SNPs in linkage disequilibrium. Here, we combined three distinct approaches: multiethnic fine-mapping, putative functional annotation (based upon epigenetic data and genome-encoded features), and expression quantitative trait loci (eQTL) analyses, in an attempt to reduce this complexity. We examined 67 risk regions using genotyping and imputation-based fine-mapping in populations of European (cases/controls: 8600/6946), African (cases/controls: 5327/5136), Japanese (cases/controls: 2563/4391) and Latino (cases/controls: 1034/1046) ancestry. Markers at 55 regions passed a region-specific significance threshold (P-value cutoff range: 3.9 × 10(-4)-5.6 × 10(-3)) and in 30 regions we identified markers that were more significantly associated with risk than the previously reported variants in the multiethnic sample. Novel secondary signals (P < 5.0 × 10(-6)) were also detected in two regions (rs13062436/3q21 and rs17181170/3p12). Among 666 variants in the 55 regions with P-values within one order of magnitude of the most-associated marker, 193 variants (29%) in 48 regions overlapped with epigenetic or other putative functional marks. In 11 of the 55 regions, cis-eQTLs were detected with nearby genes. For 12 of the 55 regions (22%), the most significant region-specific, prostate-cancer associated variant represented the strongest candidate functional variant based on our annotations; the number of regions increased to 20 (36%) and 27 (49%) when examining the 2 and 3 most significantly associated variants in each region, respectively. These results have prioritized subsets of candidate variants for downstream functional evaluation.

Batra, J. Lose, F. O'Mara, T. Marquart, L. Stephens, C. Alexander, K. Srinivasan, S. Eeles, R.A. Easton, D.F. Al Olama, A.A. Kote-Jarai, Z. Guy, M. Muir, K. Lophatananon, A. Rahman, A.A. Neal, D.E. Hamdy, F.C. Donovan, J.L. Chambers, S. Gardiner, R.A. Aitken, J. Yaxley, J. Kedda, M.-.A. Clements, J.A. Spurdle, A.B (2011) Association between Prostinogen (KLK15) Genetic Variants and Prostate Cancer Risk and Aggressiveness in Australia and a Meta-Analysis of GWAS Data. full text
Jin, G. Lu, L. Cooney, K.A. Ray, A.M. Zuhlke, K.A. Lange, E.M. Cannon-Albright, L.A. Camp, N.J. Teerlink, C.C. FitzGerald, L.M. Stanford, J.L. Wiley, K.E. Isaacs, S.D. Walsh, P.C. Foulkes, W.D. Giles, G.G. Hopper, J.L. Severi, G. Eeles, R. Easton, D. Kote-Jarai, Z. Guy, M. Rinckleb, A. Maier, C. Vogel, W. Cancel-Tassin, G. Egrot, C. Cussenot, O. Thibodeau, S.N. McDonnell, S.K. Schaid, D.J. Wiklund, F. Gronberg, H. Emanuelsson, M. Whittemore, A.S. Oakley-Girvan, I. Hsieh, C.-.L. Wahlfors, T. Tammela, T. Schleutker, J. Catalona, W.J. Zheng, S.L. Ostrander, E.A. Isaacs, W.B. Xu, J (2012) Validation of prostate cancer risk-related loci identified from genome-wide association studies using family-based association analysis: evidence from the International Consortium for Prostate Cancer Genetics (ICPCG). full text
Xu, J. Lange, E.M. Lu, L. Zheng, S.L. Wang, Z. Thibodeau, S.N. Cannon-Albright, L.A. Teerlink, C.C. Camp, N.J. Johnson, A.M. Zuhlke, K.A. Stanford, J.L. Ostrander, E.A. Wiley, K.E. Isaacs, S.D. Walsh, P.C. Maier, C. Luedeke, M. Vogel, W. Schleutker, J. Wahlfors, T. Tammela, T. Schaid, D. McDonnell, S.K. DeRycke, M.S. Cancel-Tassin, G. Cussenot, O. Wiklund, F. Gronberg, H. Eeles, R. Easton, D. Kote-Jarai, Z. Whittemore, A.S. Hsieh, C.-.L. Giles, G.G. Hopper, J.L. Severi, G. Catalona, W.J. Mandal, D. Ledet, E. Foulkes, W.D. Hamel, N. Mahle, L. Moller, P. Powell, I. Bailey-Wilson, J.E. Carpten, J.D. Seminara, D. Cooney, K.A. Isaacs, W.B. Genet, I.C.P.C (2013) HOXB13 is a susceptibility gene for prostate cancer: results from the International Consortium for Prostate Cancer Genetics (ICPCG). full text
Lai, J. An, J. Nelson, C. Kote-Jarai, Z. Easton, D. Amin, A. Eeles, R. Clements, J. Batra, J (2013) Genome-wide analyses reveal specific functional repetitive DNA that are proximal to prostate cancer risk alleles. full text
Goh, C.L. Saunders, E.J. Leongamornlert, D.A. Tymrakiewicz, M. Thomas, K. Selvadurai, E.D. Woode-Amissah, R. Dadaev, T. Mahmud, N. Castro, E. Olmos, D. Guy, M. Govindasami, K. O'Brien, L.T. Hall, A.L. Wilkinson, R.A. Sawyer, E.J. Al Olama, A.A. Easton, D.F. Kote-Jarai, Z. Parker, C.C. Eeles, R.A (2013) Clinical implications of family history of prostate cancer and genetic risk single nucleotide polymorphism (SNP) profiles in an active surveillance cohort. full text
Pooley, K.A. Bojesen, S.E. Weischer, M. Nielsen, S.F. Thompson, D. Al Olama, A.A. Michailidou, K. Tyrer, J.P. Benlloch, S. Brown, J. Audley, T. Luben, R. Khaw, K.-.T. Neal, D.E. Hamdy, F.C. Donovan, J.L. Kote-Jarai, Z. Baynes, C. Shah, M. Bolla, M.K. Wang, Q. Dennis, J. Dicks, E. Yang, R. Rudolph, A. Schildkraut, J. Chang-Claude, J. Burwinkel, B. Chenevix-Trench, G. Pharoah, P.D.P. Berchuck, A. Eeles, R.A. Easton, D.F. Dunning, A.M. Nordestgaard, B.G (2013) A genome-wide association scan (GWAS) for mean telomere length within the COGS project: identified loci show little association with hormone-related cancer risk. full text
Teerlink, C.C. Thibodeau, S.N. McDonnell, S.K. Schaid, D.J. Rinckleb, A. Maier, C. Vogel, W. Cancel-Tassin, G. Egrot, C. Cussenot, O. Foulkes, W.D. Giles, G.G. Hopper, J.L. Severi, G. Eeles, R. Easton, D. Kote-Jarai, Z. Guy, M. Cooney, K.A. Ray, A.M. Zuhlke, K.A. Lange, E.M. FitzGerald, L.M. Stanford, J.L. Ostrander, E.A. Wiley, K.E. Isaacs, S.D. Walsh, P.C. Isaacs, W.B. Wahlfors, T. Tammela, T. Schleutker, J. Wiklund, F. Gronberg, H. Emanuelsson, M. Carpten, J. Bailey-Wilson, J. Whittemore, A.S. Oakley-Girvan, I. Hsieh, C.-.L. Catalona, W.J. Zheng, S.L. Jin, G. Lu, L. Xu, J. Camp, N.J. Cannon-Albright, L.A. Genet, I.C.P.C (2014) Association analysis of 9,560 prostate cancer cases from the International Consortium of Prostate Cancer Genetics confirms the role of reported prostate cancer associated SNPs for familial disease. full text
Castro, E. Goh, C.L. Olmos, D. Leongamornlert, D. Saunders, E. Tymrakiewicz, M. Mahmud, N. Dadaev, T. Govindasami, K. Guy, M. OBrien, L. Sawyer, E. Hall, A. Wilkinson, R. Kote-Jarai, Z. Eeles, R.A. Collaborators, U.K.G.P.C.S (2011) Correlation of germ-line BRCA2 mutations with aggressive prostate cancer and outcome.. full text
Mao, X. Marzec, J. Li, M. Zhang, Z. Feng, N. Gou, X. Sun, Z. Xu, J. Xue, Y. Al Olama, A.A. Xu, X. Li, J. He, W. Benlloch, S. Kote-Jarai, Z. Wang, M. Chelala, C. Easton, D.F. Eeles, R.A. Lu, Y.-.J. Zhang, H. Consortium, P.R.A.C.T.I.C.A.L. Grp, C.H.I.P.G.E.C.S (2014) Population differences in the genetic risk of prostate cancer and predisposition loci in the Chinese population. full text