151
|
IDH-mutant glioma specific association of rs55705857 located at 8q24.21 involves MYC deregulation. Sci Rep 2016; 6:27569. [PMID: 27282637 PMCID: PMC4901315 DOI: 10.1038/srep27569] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2015] [Accepted: 05/16/2016] [Indexed: 12/20/2022] Open
Abstract
The single nucleotide polymorphism rs55705857, located in a non-coding but evolutionarily conserved region at 8q24.21, is strongly associated with IDH-mutant glioma development and was suggested to be a causal variant. However, the molecular mechanism underlying this association has remained unknown. With a case control study in 285 gliomas, 316 healthy controls, 380 systemic cancers, 31 other CNS-tumors, and 120 IDH-mutant cartilaginous tumors, we identified that the association was specific to IDH-mutant gliomas. Odds-ratios were 9.25 (5.17–16.52; 95% CI) for IDH-mutated gliomas and 12.85 (5.94–27.83; 95% CI) for IDH-mutated, 1p/19q co-deleted gliomas. Decreasing strength with increasing anaplasia implied a modulatory effect. No somatic mutations were noted at this locus in 114 blood-tumor pairs, nor was there a copy number difference between risk-allele and only-ancestral allele carriers. CCDC26 RNA-expression was rare and not different between the two groups. There were only minor subtype-specific differences in common glioma driver genes. RNA sequencing and LC-MS/MS comparisons pointed to significantly altered MYC-signaling. Baseline enhancer activity of the conserved region specifically on the MYC promoter and its further positive modulation by the SNP risk-allele was shown in vitro. Our findings implicate MYC deregulation as the underlying cause of the observed association.
Collapse
|
152
|
Zhu Y, Wang HK, Qu YY, Ye DW. Prostate cancer in East Asia: evolving trend over the last decade. Asian J Androl 2016; 17:48-57. [PMID: 25080928 PMCID: PMC4291877 DOI: 10.4103/1008-682x.132780] [Citation(s) in RCA: 77] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
Prostate cancer is now becoming an emerging health priority in East Asia. Most of our current knowledge on Prostate cancer has been generated from studies conducted in Western population; however, there is considerable heterogeneity of Prostate cancer between East and West. In this article, we reviewed epidemiologic trends, risk factors, disease characteristics and management of Prostate cancer in East Asian population over the last decade. Growing evidence from East Asia suggests an important role of genetic and environmental risk factors interactions in the carcinogenesis of Prostate cancer. Exposure to westernized diet and life style and improvement in health care in combination contribute substantially to the increasing epidemic in this region. Diagnostic and treatment guidelines in East Asia are largely based on Western knowledge. Although there is a remarkable improvement in the outcome over the last decade, ample evidence suggests an inneglectable difference in diagnostic accuracy, treatment efficacy and adverse events between different populations. The knowledge from western countries should be calibrated in the Asian setting to provide a better race-based treatment approach. In this review, we intend to reveal the evolving trend of Prostate cancer in the last decade, in order to gain evidence to improve Prostate cancer prevention and control in East Asia.
Collapse
Affiliation(s)
| | | | | | - Ding-Wei Ye
- Department of Urology, Fudan University Shanghai Cancer Center; Department of Oncology, Fudan University Shanghai Medical College, Shanghai 200032, China
| |
Collapse
|
153
|
Bull CJ, Bonilla C, Holly JMP, Perks CM, Davies N, Haycock P, Yu OHY, Richards JB, Eeles R, Easton D, Kote‐Jarai Z, Amin Al Olama A, Benlloch S, Muir K, Giles GG, MacInnis RJ, Wiklund F, Gronberg H, Haiman CA, Schleutker J, Nordestgaard BG, Travis RC, Neal D, Pashayan N, Khaw K, Stanford JL, Blot WJ, Thibodeau S, Maier C, Kibel AS, Cybulski C, Cannon‐Albright L, Brenner H, Park J, Kaneva R, Batra J, Teixeira MR, Micheal A, Pandha H, Smith GD, Lewis SJ, Martin RM. Blood lipids and prostate cancer: a Mendelian randomization analysis. Cancer Med 2016; 5:1125-36. [PMID: 26992435 PMCID: PMC4924371 DOI: 10.1002/cam4.695] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2015] [Revised: 02/03/2016] [Accepted: 02/08/2016] [Indexed: 12/14/2022] Open
Abstract
Genetic risk scores were used as unconfounded instruments for specific lipid traits (Mendelian randomization) to assess whether circulating lipids causally influence prostate cancer risk. Data from 22,249 prostate cancer cases and 22,133 controls from 22 studies within the international PRACTICAL consortium were analyzed. Allele scores based on single nucleotide polymorphisms (SNPs) previously reported to be uniquely associated with each of low-density lipoprotein (LDL), high-density lipoprotein (HDL), and triglyceride (TG) levels, were first validated in an independent dataset, and then entered into logistic regression models to estimate the presence (and direction) of any causal effect of each lipid trait on prostate cancer risk. There was weak evidence for an association between the LDL genetic score and cancer grade: the odds ratio (OR) per genetically instrumented standard deviation (SD) in LDL, comparing high- (≥7 Gleason score) versus low-grade (<7 Gleason score) cancers was 1.50 (95% CI: 0.92, 2.46; P = 0.11). A genetically instrumented SD increase in TGs was weakly associated with stage: the OR for advanced versus localized cancer per unit increase in genetic risk score was 1.68 (95% CI: 0.95, 3.00; P = 0.08). The rs12916-T variant in 3-hydroxy-3-methylglutaryl-CoA reductase (HMGCR) was inversely associated with prostate cancer (OR: 0.97; 95% CI: 0.94, 1.00; P = 0.03). In conclusion, circulating lipids, instrumented by our genetic risk scores, did not appear to alter prostate cancer risk. We found weak evidence that higher LDL and TG levels increase aggressive prostate cancer risk, and that a variant in HMGCR (that mimics the LDL lowering effect of statin drugs) reduces risk. However, inferences are limited by sample size and evidence of pleiotropy.
Collapse
|
154
|
Shah K, Bradbury NA. Lemur Tyrosine Kinase 2, a novel target in prostate cancer therapy. Oncotarget 2016; 6:14233-46. [PMID: 26008968 PMCID: PMC4546463 DOI: 10.18632/oncotarget.3899] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2015] [Accepted: 04/25/2015] [Indexed: 11/25/2022] Open
Abstract
Progression from early forms of prostate cancer to castration-resistant disease is associated with an increase in signal transduction activity. The majority of castration-resistance cancers persist in the expression of the androgen receptor (AR), as well as androgen-dependent genes. The AR is regulated not only by it associated steroid hormone, but also by manifold regulatory and signaling molecules, including several kinases. We undertook evaluation of the role of Lemur Tyrosine Kinase 2 (LMTK2) in modulating AR activity, as several Genome Wide Association Studies (GWAS) have shown a marked association of LMTK2 activity with the development of prostate cancer. We confirm that not only is LMTK2 mRNA reduced in prostate cancer tissue, but also LMTK2 protein levels are markedly diminished. Knockdown of LMTK2 protein in prostate cell lines greatly increased the transcription of androgen-responsive genes. In addition, LMTK2 knockdown led to an increase in prostate cancer stem cell populations in LNCaP cells, indicative of increased tumorogenicity. Using multiple approaches, we also demonstrate that LMTK2 interacts with the AR, thus putting LMTK2 as a component of a signaling complex modulating AR activity. Our finding that LMTK2 is a negative regulator of AR activity defines a novel cellular pathway for activation of AR-responsive genes in castrate resistant-prostate cancer. Moreover, pharmacologic manipulation of LMTK2 activity will provide a novel therapeutic target for more effective treatments for patients with castrate-resistant prostate cancer.
Collapse
Affiliation(s)
- Kalpit Shah
- Department of Physiology and Biophysics, Rosalind Franklin University of Medicine & Sciences, The Chicago Medical School, North Chicago, IL 60064, USA
| | - Neil A Bradbury
- Department of Physiology and Biophysics, Rosalind Franklin University of Medicine & Sciences, The Chicago Medical School, North Chicago, IL 60064, USA
| |
Collapse
|
155
|
Marzec J, Mao X, Li M, Wang M, Feng N, Gou X, Wang G, Sun Z, Xu J, Xu H, Zhang X, Zhao SC, Ren G, Yu Y, Wu Y, Wu J, Xue Y, Zhou B, Zhang Y, Xu X, Li J, He W, Benlloch S, Ross-Adams H, Chen L, Li J, Hong Y, Kote-Jarai Z, Cui X, Hou J, Guo J, Xu L, Yin C, Zhou Y, Neal DE, Oliver T, Cao G, Zhang Z, Easton DF, Chelala C, Olama AAA, Eeles RA, Zhang H, Lu YJ. A genetic study and meta-analysis of the genetic predisposition of prostate cancer in a Chinese population. Oncotarget 2016; 7:21393-403. [PMID: 26881390 PMCID: PMC5008293 DOI: 10.18632/oncotarget.7250] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2015] [Accepted: 01/23/2016] [Indexed: 11/25/2022] Open
Abstract
Prostate cancer predisposition has been extensively investigated in European populations, but there have been few studies of other ethnic groups. To investigate prostate cancer susceptibility in the under-investigated Chinese population, we performed single-nucleotide polymorphism (SNP) array analysis on a cohort of Chinese cases and controls and then meta-analysis with data from the existing Chinese prostate cancer genome-wide association study (GWAS). Genotyping 211,155 SNPs in 495 cases and 640 controls of Chinese ancestry identified several new suggestive Chinese prostate cancer predisposition loci. However, none of them reached genome-wide significance level either by meta-analysis or replication study. The meta-analysis with the Chinese GWAS data revealed that four 8q24 loci are the main contributors to Chinese prostate cancer risk and the risk alleles from three of them exist at much higher frequencies in Chinese than European populations. We also found that several predisposition loci reported in Western populations have different effect on Chinese men. Therefore, this first extensive single-nucleotide polymorphism study of Chinese prostate cancer in comparison with European population indicates that four loci on 8q24 contribute to a great risk of prostate cancer in a considerable large proportion of Chinese men. Based on those four loci, the top 10% of the population have six- or two-fold prostate cancer risk compared with men of the bottom 10% or median risk respectively, which may facilitate the design of prostate cancer genetic risk screening and prevention in Chinese men. These findings also provide additional insights into the etiology and pathogenesis of prostate cancer.
Collapse
Affiliation(s)
- Jacek Marzec
- Centre for Molecular Oncology, Barts Cancer Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, EC1M 6BQ, UK
| | - Xueying Mao
- Centre for Molecular Oncology, Barts Cancer Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, EC1M 6BQ, UK
| | - Meiling Li
- Department of Epidemiology, Second Military Medical University, Shanghai, 200433, China
| | - Meilin Wang
- Department of Molecular and Genetic Toxicology, The Key Laboratory of Modern Toxicology, School of Public Health, Nanjing Medical University, Nanjing, 210029, China
| | - Ninghan Feng
- Department of Urology, Wuxi Second People's Hospital, Nanjing Medical University, Wuxi, 214002, China
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Xin Gou
- Department of Urology, The First Affiliated Hospital, Chongqing Medical University, Chongqing, 400016, China
| | - Guomin Wang
- Department of Urology, Zhongshan Hospital, Fudan University Medical College, Shanghai, 200032, China
| | - Zan Sun
- Liaoning People's Hospital and Center of Experiment and Technology, China Medical University, Shenyang, 110001, China
| | - Jianfeng Xu
- Program for Personalized Cancer Care, North Shore University Health System, Evanston, IL 60201, U.S.A
- Fudan Institute of Urology, Huashang Hospital, Fudan University, Shanghai, 200040, China
| | - Hua Xu
- Department of Urology, Tongji Hospital, Huazhong Science and Technology University, Wuhan, 430030, China
| | - Xiaoping Zhang
- Department of Urology, Xiehe Hospital, Huazhong Science and Technology University, Wuhan, 430022, China
| | - Shan-Chao Zhao
- Department of Urology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Guoping Ren
- Department of Pathology, The First Affiliated Hospital, Zhejiang University Medical College, Hangzhou, 310009, China
| | - Yongwei Yu
- Department of Pathology, Changhai Hospital, The Second Military Medical University, Shanghai, 200433, China
| | - Yudong Wu
- Department of Urology, First Affiliated Hospital, Medical College, Zhengzhou University, Zhengzhou, 450003, China
| | - Ji Wu
- Department of Urology, North Sichuan Medical College, Nanchong, 637000, China
| | - Yao Xue
- Department of Molecular and Genetic Toxicology, The Key Laboratory of Modern Toxicology, School of Public Health, Nanjing Medical University, Nanjing, 210029, China
| | - Bo Zhou
- Department of Nutrition Science, Shenyang Medical College, Shenyang, 110034, China
| | - Yanling Zhang
- Department of Pathology, The First Affiliated Hospital, Zhejiang University Medical College, Hangzhou, 310009, China
| | - Xingxing Xu
- Department of Epidemiology, Second Military Medical University, Shanghai, 200433, China
| | - Jie Li
- Department of Urology, The First Affiliated Hospital, Chongqing Medical University, Chongqing, 400016, China
| | - Weiyang He
- Department of Urology, The First Affiliated Hospital, Chongqing Medical University, Chongqing, 400016, China
| | - Sara Benlloch
- Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, University of Cambridge–Strangeways Research Laboratory, Cambridge, CB1 8RN, UK
| | - Helen Ross-Adams
- Cancer Research UK Cambridge Research Institute, Li Ka Shing Centre, Robinson Way, Cambridge, CB2 0RE, UK
| | - Li Chen
- Department of Urology, Xiehe Hospital, Huazhong Science and Technology University, Wuhan, 430022, China
| | - Jucong Li
- Department of Urology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Yingqia Hong
- Department of Urology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Zsofia Kote-Jarai
- Division of Genetics and Epidemiology, The Institute of Cancer Research, London, SM2 5NG, UK
| | - Xingang Cui
- Department of Urology, Changzheng Hospital, The Second Military Medical University, Shanghai, 200003, China
| | - Jianguo Hou
- Department of Urology, Changhai Hospital, The Second Military Medical University, Shanghai, 200433, China
| | - Jianming Guo
- Department of Urology, Zhongshan Hospital, Fudan University Medical College, Shanghai, 200032, China
| | - Lei Xu
- Department of Urology, Zhongshan Hospital, Fudan University Medical College, Shanghai, 200032, China
| | - Changjun Yin
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Yuanping Zhou
- Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - David E. Neal
- Cancer Research UK Cambridge Research Institute, Li Ka Shing Centre, Robinson Way, Cambridge, CB2 0RE, UK
| | - Tim Oliver
- Centre for Molecular Oncology, Barts Cancer Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, EC1M 6BQ, UK
| | - Guangwen Cao
- Department of Epidemiology, Second Military Medical University, Shanghai, 200433, China
| | - Zhengdong Zhang
- Department of Molecular and Genetic Toxicology, The Key Laboratory of Modern Toxicology, School of Public Health, Nanjing Medical University, Nanjing, 210029, China
| | - Douglas F. Easton
- Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, University of Cambridge–Strangeways Research Laboratory, Cambridge, CB1 8RN, UK
| | - Claude Chelala
- Centre for Molecular Oncology, Barts Cancer Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, EC1M 6BQ, UK
| | | | | | - Ali Amin Al Olama
- Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, University of Cambridge–Strangeways Research Laboratory, Cambridge, CB1 8RN, UK
| | - Rosalind A. Eeles
- Division of Genetics and Epidemiology, The Institute of Cancer Research, London, SM2 5NG, UK
- The Royal Marsden NHS Foundation Trust, London and Surrey, SM2 5NG, UK
| | - Hongwei Zhang
- Department of Epidemiology, Second Military Medical University, Shanghai, 200433, China
| | - Yong-Jie Lu
- Centre for Molecular Oncology, Barts Cancer Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, EC1M 6BQ, UK
| |
Collapse
|
156
|
Schmitt AM, Chang HY. Long Noncoding RNAs in Cancer Pathways. Cancer Cell 2016; 29:452-463. [PMID: 27070700 PMCID: PMC4831138 DOI: 10.1016/j.ccell.2016.03.010] [Citation(s) in RCA: 2313] [Impact Index Per Article: 289.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2016] [Revised: 03/01/2016] [Accepted: 03/14/2016] [Indexed: 12/16/2022]
Abstract
Genome-wide cancer mutation analyses are revealing an extensive landscape of functional mutations within the noncoding genome, with profound effects on the expression of long noncoding RNAs (lncRNAs). While the exquisite regulation of lncRNA transcription can provide signals of malignant transformation, we now understand that lncRNAs drive many important cancer phenotypes through their interactions with other cellular macromolecules including DNA, protein, and RNA. Recent advancements in surveying lncRNA molecular mechanisms are now providing the tools to functionally annotate these cancer-associated transcripts, making these molecules attractive targets for therapeutic intervention in the fight against cancer.
Collapse
Affiliation(s)
- Adam M Schmitt
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA.
| | - Howard Y Chang
- Center for Personal Dynamic Regulomes, Stanford University School of Medicine, Stanford, CA 94305, USA.
| |
Collapse
|
157
|
Du M, Tillmans L, Gao J, Gao P, Yuan T, Dittmar RL, Song W, Yang Y, Sahr N, Wang T, Wei GH, Thibodeau SN, Wang L. Chromatin interactions and candidate genes at ten prostate cancer risk loci. Sci Rep 2016; 6:23202. [PMID: 26979803 PMCID: PMC4793270 DOI: 10.1038/srep23202] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2015] [Accepted: 03/02/2016] [Indexed: 12/12/2022] Open
Abstract
Genome-wide association studies have identified more than 100 common single nucleotide polymorphisms (SNPs) that are associated with prostate cancer risk. However, the vast majority of these SNPs lie in noncoding regions of the genome. To test whether these risk SNPs regulate their target genes through long-range chromatin interactions, we applied capture-based 3C sequencing technology to investigate possible cis-interactions at ten prostate cancer risk loci in six cell lines. We identified significant physical interactions between risk regions and their potential target genes including CAPG at 2p11.2, C2orf43 at 2p24.1, RFX6 at 6q22.1, NFASC at 1q32.1, MYC at 8q24.1 and AGAP7P at 10q11.23. Most of the interaction peaks were co-localized to regions of active histone modification and transcription factor binding sites. Expression quantitative trait locus (eQTL) analysis showed suggestive eQTL signals at rs1446669, rs699664 and rs1078004 for CAPG (p < 0.004), rs13394027 for C2orf43 (p = 2.25E-27), rs10993994 and rs4631830 for AGAP7P (p < 8.02E-5). Further analysis revealed an enhancer activity at genomic region surrounding rs4631830 which was expected to disrupt HOXB-like DNA binding affinity. This study identifies a set of candidate genes and their potential regulatory variants, and provides additional evidence showing the role of long-range chromatin interactions in prostate cancer etiology.
Collapse
Affiliation(s)
- Meijun Du
- Department of Pathology, MCW Cancer Center, Medical College of Wisconsin, Milwaukee, 53226, WI, USA
| | - Lori Tillmans
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, 55905, MN, USA
| | - Jianzhong Gao
- Beijing 3H Medical Technology Co. Ltd., Beijing, 100176, China
| | - Ping Gao
- Faculty of Biochemistry and Molecular Medicine and Biocenter Oulu, University of Oulu, Oulu, Finland
| | - Tiezheng Yuan
- Department of Pathology, MCW Cancer Center, Medical College of Wisconsin, Milwaukee, 53226, WI, USA
| | - Rachel L Dittmar
- Department of Pathology, MCW Cancer Center, Medical College of Wisconsin, Milwaukee, 53226, WI, USA
| | - Wei Song
- Faculty of Biochemistry and Molecular Medicine and Biocenter Oulu, University of Oulu, Oulu, Finland
| | - Yuehong Yang
- Faculty of Biochemistry and Molecular Medicine and Biocenter Oulu, University of Oulu, Oulu, Finland
| | - Natasha Sahr
- Division of Biostatistics, Institute for Health &Society, Medical College of Wisconsin, Milwaukee, 53226, WI, USA
| | - Tao Wang
- Division of Biostatistics, Institute for Health &Society, Medical College of Wisconsin, Milwaukee, 53226, WI, USA
| | - Gong-Hong Wei
- Faculty of Biochemistry and Molecular Medicine and Biocenter Oulu, University of Oulu, Oulu, Finland
| | - Stephen N Thibodeau
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, 55905, MN, USA
| | - Liang Wang
- Department of Pathology, MCW Cancer Center, Medical College of Wisconsin, Milwaukee, 53226, WI, USA
| |
Collapse
|
158
|
Abstract
Although prostate cancer is the most common malignancy to affect men in the Western world, the molecular mechanisms underlying its development and progression remain poorly understood. Like all cancers, prostate cancer is a genetic disease that is characterized by multiple genomic alterations, including point mutations, microsatellite variations, and chromosomal alterations such as translocations, insertions, duplications, and deletions. In prostate cancer, but not other carcinomas, these chromosome alterations result in a high frequency of gene fusion events. The development and application of novel high-resolution technologies has significantly accelerated the detection of genomic alterations, revealing the complex nature and heterogeneity of the disease. The clinical heterogeneity of prostate cancer can be partly explained by this underlying genetic heterogeneity, which has been observed between patients from different geographical and ethnic populations, different individuals within these populations, different tumour foci within the same patient, and different cells within the same tumour focus. The highly heterogeneous nature of prostate cancer provides a real challenge for clinical disease management and a detailed understanding of the genetic alterations in all cells, including small subpopulations, would be highly advantageous.
Collapse
|
159
|
Penney KL, Pettersson A, Shui IM, Graff RE, Kraft P, Lis RT, Sesso HD, Loda M, Mucci LA. Association of Prostate Cancer Risk Variants with TMPRSS2:ERG Status: Evidence for Distinct Molecular Subtypes. Cancer Epidemiol Biomarkers Prev 2016; 25:745-9. [PMID: 26941365 DOI: 10.1158/1055-9965.epi-15-1078] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2015] [Accepted: 01/27/2016] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Numerous genetic variants have been confirmed as prostate cancer risk factors. These variants may confer susceptibility to the development of specific molecular alterations during tumor initiation and progression. The TMPRSS2:ERG gene fusion occurs in roughly 50% of prostate cancers. Genetic risk variants may influence the development of this fusion. We sought to determine whether prostate cancer risk variants are differentially associated with TMPRSS2:ERG fusion-positive and negative cancer. METHODS In the Health Professionals Follow-up Study and Physicians' Health Study Tumor Cohort, we evaluated the associations of 39 prostate cancer risk SNPs with TMPRSS2:ERG fusion status, measured by ERG protein expression. Logistic regression was performed to generate OR and 95% confidence intervals. The primary outcome was ERG(+) (n = 227) versus ERG(-) (n = 260) prostate cancer. A secondary outcome was ERG(+) or ERG(-) cancer versus controls without cancer. RESULTS Six of 39 SNPs were significantly associated (P < 0.05) with ERG(+) versus ERG(-) disease. Three SNPs were exclusively associated with the risk of ERG(+), one with risk of ERG(-), and two with associations trending in opposite directions for ERG(+) and ERG(-) Only two significant SNPs would be expected by chance. CONCLUSIONS Prostate cancer genetic risk variants are differentially associated with the development of ERG(+) and ERG(-) prostate cancer. IMPACT Our findings suggest the molecular process of prostate carcinogenesis may be distinct for men with different underlying genetic predisposition. When examining risk factors for prostate cancer, the integration of molecular subtypes may enhance understanding of the etiology of this disease. Cancer Epidemiol Biomarkers Prev; 25(5); 745-9. ©2016 AACR.
Collapse
Affiliation(s)
- Kathryn L Penney
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts. Channing Division of Network Medicine, Brigham and Women's Hospital, Boston, Massachusetts.
| | - Andreas Pettersson
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts. Clinical Epidemiology Unit, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
| | - Irene M Shui
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts. Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Rebecca E Graff
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts. Department of Epidemiology and Biostatistics, University of California, San Francisco, San Francisco, California
| | - Peter Kraft
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts. Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
| | - Rosina T Lis
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts. Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Howard D Sesso
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts. Division of Preventive Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Massimo Loda
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts. Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts. The Broad Institute, Cambridge, Massachusetts
| | - Lorelei A Mucci
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts. Channing Division of Network Medicine, Brigham and Women's Hospital, Boston, Massachusetts
| |
Collapse
|
160
|
Sjöblom L, Saramäki O, Annala M, Leinonen K, Nättinen J, Tolonen T, Wahlfors T, Nykter M, Bova GS, Schleutker J, Tammela TLJ, Lilja H, Visakorpi T. Microseminoprotein-Beta Expression in Different Stages of Prostate Cancer. PLoS One 2016; 11:e0150241. [PMID: 26939004 PMCID: PMC4777373 DOI: 10.1371/journal.pone.0150241] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2015] [Accepted: 02/02/2016] [Indexed: 11/18/2022] Open
Abstract
Microseminoprotein-beta (MSMB, MSMB) is an abundant secretory protein contributed by the prostate, and is implicated as a prostate cancer (PC) biomarker based on observations of its lower expression in cancerous cells compared with benign prostate epithelium. However, as the current literature on MSMB is inconsistent, we assessed the expression of MSMB at the protein and mRNA levels in a comprehensive set of different clinical stages of PC. Immunohistochemistry using monoclonal and polyclonal antibodies against MSMB was used to study protein expression in tissue specimens representing prostatectomies (n = 261) and in diagnostic needle biopsies from patients treated with androgen deprivation therapy (ADT) (n = 100), and in locally recurrent castration-resistant PC (CRPC) (n = 105) and CRPC metastases (n = 113). The transcript levels of MSMB, nuclear receptor co-activator 4 (NCOA4) and MSMB-NCOA4 fusion were examined by qRT-PCR in prostatectomy samples and by RNA-sequencing in benign prostatic hyperplasia, PC, and CRPC samples. We also measured serum MSMB levels and genotyped the single nucleotide polymorphism rs10993994 using DNA from the blood of 369 PC patients and 903 controls. MSMB expression in PC (29% of prostatectomies and 21% of needle biopsies) was more frequent than in CRPC (9% of locally recurrent CRPCs and 9% of CRPC metastases) (p<0.0001). Detection of MSMB protein was inversely correlated with the Gleason score in prostatectomy specimens (p = 0.024). The read-through MSMB-NCOA4 transcript was detected at very low levels in PC. MSMB levels in serum were similar in cases of PC and controls but were significantly associated with PC risk when adjusted for age at diagnosis and levels of free or total PSA (p<0.001). Serum levels of MSMB in both PC patients and controls were significantly associated with the rs10993994 genotype (p<0.0001). In conclusion, decreased expression of MSMB parallels the clinical progression of PC and adjusted serum MSMB levels are associated with PC risk.
Collapse
Affiliation(s)
- Liisa Sjöblom
- Prostate Cancer Research Center, Institute of Biosciences and Medical Technology (BioMediTech), University of Tampere, Tampere, Finland.,Fimlab Laboratories, Tampere University Hospital, Tampere, Finland
| | - Outi Saramäki
- Prostate Cancer Research Center, Institute of Biosciences and Medical Technology (BioMediTech), University of Tampere, Tampere, Finland.,Fimlab Laboratories, Tampere University Hospital, Tampere, Finland
| | - Matti Annala
- Prostate Cancer Research Center, Institute of Biosciences and Medical Technology (BioMediTech), University of Tampere, Tampere, Finland
| | - Katri Leinonen
- Prostate Cancer Research Center, Institute of Biosciences and Medical Technology (BioMediTech), University of Tampere, Tampere, Finland.,Fimlab Laboratories, Tampere University Hospital, Tampere, Finland
| | - Janika Nättinen
- Prostate Cancer Research Center, Institute of Biosciences and Medical Technology (BioMediTech), University of Tampere, Tampere, Finland
| | - Teemu Tolonen
- Department of Pathology, Fimlab Laboratories, Tampere University Hospital, Tampere, Finland
| | - Tiina Wahlfors
- Prostate Cancer Research Center, Institute of Biosciences and Medical Technology (BioMediTech), University of Tampere, Tampere, Finland
| | - Matti Nykter
- Prostate Cancer Research Center, Institute of Biosciences and Medical Technology (BioMediTech), University of Tampere, Tampere, Finland
| | - G Steven Bova
- Prostate Cancer Research Center, Institute of Biosciences and Medical Technology (BioMediTech), University of Tampere, Tampere, Finland
| | - Johanna Schleutker
- Prostate Cancer Research Center, Institute of Biosciences and Medical Technology (BioMediTech), University of Tampere, Tampere, Finland
| | - Teuvo L J Tammela
- Prostate Cancer Research Center, School of Medicine, University of Tampere, Tampere, Finland.,Department of Urology, Tampere University Hospital, Tampere, Finland
| | - Hans Lilja
- Prostate Cancer Research Center, Institute of Biosciences and Medical Technology (BioMediTech), University of Tampere, Tampere, Finland.,Departments of Laboratory Medicine, Surgery, and Medicine, Memorial Sloan Kettering Cancer Center, New York, United States of America.,Nuffield Department of Surgical Sciences, University of Oxford, Oxford, United Kingdom.,Department of Translational Medicine, Lund University, Malmö, Sweden
| | - Tapio Visakorpi
- Prostate Cancer Research Center, Institute of Biosciences and Medical Technology (BioMediTech), University of Tampere, Tampere, Finland.,Fimlab Laboratories, Tampere University Hospital, Tampere, Finland
| |
Collapse
|
161
|
Ahmed M, Eeles R. Germline genetic profiling in prostate cancer: latest developments and potential clinical applications. Future Sci OA 2016; 2:FSO87. [PMID: 28031937 PMCID: PMC5137984 DOI: 10.4155/fso.15.87] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2015] [Accepted: 11/10/2015] [Indexed: 12/16/2022] Open
Abstract
Familial and twin studies have demonstrated a significant inherited component to prostate cancer predisposition. Genome wide association studies have shown that there are 100 single nucleotide polymorphisms which have been associated with the development of prostate cancer. This review aims to discuss the scientific methods used to identify these susceptibility loci. It will also examine the current clinical utility of these loci, which include the development of risk models as well as predicting treatment efficacy and toxicity. In order to refine the clinical utility of the susceptibility loci, international consortia have been developed to combine statistical power as well as skills and knowledge to further develop models that could be used to predict risk and treatment outcomes.
Collapse
Affiliation(s)
- Mahbubl Ahmed
- The Institute of Cancer Research, London SM2 5NG, UK
| | | |
Collapse
|
162
|
Liu M, Shi X, Yang F, Wang J, Xu Y, Wei D, Yang K, Zhang Y, Wang X, Liang S, Chen X, Sun L, Zhu X, Zhao C, Zhu L, Tang L, Zheng C, Yang Z. The Cumulative Effect of Gene-Gene and Gene-Environment Interactions on the Risk of Prostate Cancer in Chinese Men. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2016; 13:162. [PMID: 26828504 PMCID: PMC4772182 DOI: 10.3390/ijerph13020162] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/24/2015] [Revised: 01/15/2016] [Accepted: 01/18/2016] [Indexed: 01/15/2023]
Abstract
Prostate cancer (PCa) is a multifactorial disease involving complex genetic and environmental factors interactions. Gene-gene and gene-environment interactions associated with PCa in Chinese men are less studied. We explored the association between 36 SNPs and PCa in 574 subjects from northern China. Body mass index (BMI), smoking, and alcohol consumption were determined through self-administered questionnaires in 134 PCa patients. Then gene-gene and gene-environment interactions among the PCa-associated SNPs were analyzed using the generalized multifactor dimensionality reduction (GMDR) and logistic regression methods. Allelic and genotypic association analyses showed that six variants were associated with PCa and the cumulative effect suggested men who carried any combination of 1, 2, or ≥3 risk genotypes had a gradually increased PCa risk (odds ratios (ORs) = 1.79–4.41). GMDR analysis identified the best gene-gene interaction model with scores of 10 for both the cross-validation consistency and sign tests. For gene-environment interactions, rs6983561 CC and rs16901966 GG in individuals with a BMI ≥ 28 had ORs of 7.66 (p = 0.032) and 5.33 (p = 0.046), respectively. rs7679673 CC + CA and rs12653946 TT in individuals that smoked had ORs of 2.77 (p = 0.007) and 3.11 (p = 0.024), respectively. rs7679673 CC in individuals that consumed alcohol had an OR of 4.37 (p = 0.041). These results suggest that polymorphisms, either individually or by interacting with other genes or environmental factors, contribute to an increased risk of PCa.
Collapse
Affiliation(s)
- Ming Liu
- Department of Cell Biology and Genetics, School of Basic Medical Science, Shanxi Medical University, Taiyuan 030001, China.
| | - Xiaohong Shi
- The Key Laboratory of Geriatrics, Beijing Hospital & Beijing Institute of Geriatrics, Ministry of Health, Beijing 100730, China.
| | - Fan Yang
- The Key Laboratory of Geriatrics, Beijing Hospital & Beijing Institute of Geriatrics, Ministry of Health, Beijing 100730, China.
- Graduate School, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100005, China.
| | - Jianye Wang
- Department of Urology and Beijing Hospital, Chinese Ministry of Health, Beijing 100730, China.
| | - Yong Xu
- Department of Urology, The Second Hospital of Tianjin Medical University, Tianjin 300211, China.
| | - Dong Wei
- Department of Urology and Beijing Hospital, Chinese Ministry of Health, Beijing 100730, China.
| | - Kuo Yang
- Department of Urology, The Second Hospital of Tianjin Medical University, Tianjin 300211, China.
| | - Yaoguang Zhang
- Department of Urology and Beijing Hospital, Chinese Ministry of Health, Beijing 100730, China.
| | - Xin Wang
- Department of Urology and Beijing Hospital, Chinese Ministry of Health, Beijing 100730, China.
| | - Siying Liang
- The Key Laboratory of Geriatrics, Beijing Hospital & Beijing Institute of Geriatrics, Ministry of Health, Beijing 100730, China.
| | - Xin Chen
- Department of Urology and Beijing Hospital, Chinese Ministry of Health, Beijing 100730, China.
| | - Liang Sun
- The Key Laboratory of Geriatrics, Beijing Hospital & Beijing Institute of Geriatrics, Ministry of Health, Beijing 100730, China.
| | - Xiaoquan Zhu
- The Key Laboratory of Geriatrics, Beijing Hospital & Beijing Institute of Geriatrics, Ministry of Health, Beijing 100730, China.
| | - Chengxiao Zhao
- Department of Cell Biology and Genetics, School of Basic Medical Science, Shanxi Medical University, Taiyuan 030001, China.
- The Key Laboratory of Geriatrics, Beijing Hospital & Beijing Institute of Geriatrics, Ministry of Health, Beijing 100730, China.
| | - Ling Zhu
- Medical Examination Centre, Beijing Hospital, Ministry of Health, Beijing 100730, China.
| | - Lei Tang
- The Key Laboratory of Geriatrics, Beijing Hospital & Beijing Institute of Geriatrics, Ministry of Health, Beijing 100730, China.
| | - Chenguang Zheng
- Guangxi Zhuang Autonomous Region Women and Children Care Hospital, Nanning, Guangxi 530003, China.
| | - Ze Yang
- The Key Laboratory of Geriatrics, Beijing Hospital & Beijing Institute of Geriatrics, Ministry of Health, Beijing 100730, China.
| |
Collapse
|
163
|
Jinga V, Csiki IE, Manolescu A, Iordache P, Mates IN, Radavoi D, Rascu S, Badescu D, Badea P, Mates D. Replication study of 34 common SNPs associated with prostate cancer in the Romanian population. J Cell Mol Med 2016; 20:594-600. [PMID: 26773531 PMCID: PMC5126261 DOI: 10.1111/jcmm.12729] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2015] [Accepted: 09/27/2015] [Indexed: 12/14/2022] Open
Abstract
Prostate cancer is the third‐most common form of cancer in men in Romania. The Romanian unscreened population represents a good sample to study common genetic risk variants. However, a comprehensive analysis has not been conducted yet. Here, we report our replication efforts in a Romanian population of 979 cases and 1027 controls, for potential association of 34 literature‐reported single nucleotide polymorphisms (SNPs) with prostate cancer. We also examined whether any SNP was differentially associated with tumour grade or stage at diagnosis, with disease aggressiveness, and with the levels of PSA (prostate specific antigen). In the allelic analysis, we replicated the previously reported risk for 19 loci on 4q24, 6q25.3, 7p15.2, 8q24.21, 10q11.23, 10q26.13, 11p15.5, 11q13.2, 11q13.3. Statistically significant associations were replicated for other six SNPs only with a particular disease phenotype: low‐grade tumour and low PSA levels (rs1512268), high PSA levels (rs401681 and rs11649743), less aggressive cancers (rs1465618, rs721048, rs17021918). The strongest association of our tested SNP's with PSA in controls was for rs2735839, with 29% increase for each copy of the major allele G, consistent with previous results. Our results suggest that rs4962416, previously associated only with prostate cancer, is also associated with PSA levels, with 12% increase for each copy of the minor allele C. The study enabled the replication of the effect for the majority of previously reported genetic variants in a set of clinically relevant prostate cancers. This is the first replication study on these loci, known to associate with prostate cancer, in a Romanian population.
Collapse
Affiliation(s)
- Viorel Jinga
- "Prof. Dr. Th. Burghele" Clinical Hospital, Urology Department, University of Medicine and Pharmacy "Carol Davila", Bucharest, Romania
| | | | - Andrei Manolescu
- School of Science and Engineering, Reykjavik University, Reykjavik, Iceland
| | - Paul Iordache
- School of Science and Engineering, Reykjavik University, Reykjavik, Iceland
| | - Ioan Nicolae Mates
- "St Mary" Clinical Hospital, General Surgery Department, University of Medicine and Pharmacy "Carol Davila", Bucharest, Romania
| | - Daniel Radavoi
- "Prof. Dr. Th. Burghele" Clinical Hospital, Urology Department, University of Medicine and Pharmacy "Carol Davila", Bucharest, Romania
| | - Stefan Rascu
- "Prof. Dr. Th. Burghele" Clinical Hospital, Urology Department, University of Medicine and Pharmacy "Carol Davila", Bucharest, Romania
| | - Daniel Badescu
- "Prof. Dr. Th. Burghele" Clinical Hospital, Urology Department, University of Medicine and Pharmacy "Carol Davila", Bucharest, Romania
| | - Paula Badea
- National Institute of Public Health, Bucharest, Romania
| | - Dana Mates
- National Institute of Public Health, Bucharest, Romania
| |
Collapse
|
164
|
Jiang Y, Lu H, Dag A, Hart-Smith G, Stenzel MH. Albumin–polymer conjugate nanoparticles and their interactions with prostate cancer cells in 2D and 3D culture: comparison between PMMA and PCL. J Mater Chem B 2016; 4:2017-2027. [DOI: 10.1039/c5tb02576a] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Using proteins as the hydrophilic moiety can dramatically improve the biodegradability and biocompatibility of self-assembled amphiphilic nanoparticles in the field of nanomedicine.
Collapse
Affiliation(s)
- Yanyan Jiang
- Centre for Advanced Macromolecular Design (CAMD)
- School of Chemical Engineering and School of Chemistry
- University of New South Wales
- Sydney
- Australia
| | - Hongxu Lu
- Centre for Advanced Macromolecular Design (CAMD)
- School of Chemical Engineering and School of Chemistry
- University of New South Wales
- Sydney
- Australia
| | - Aydan Dag
- Department of Pharmaceutical Chemistry
- Faculty of Pharmacy
- Bezmialem Vakif University
- 34093 Fatih
- Turkey
| | - Gene Hart-Smith
- Systems Biology Initiative
- School of Biotechnology and Biomolecular Sciences
- University of New South Wales
- Sydney 2052
- Australia
| | - Martina H. Stenzel
- Centre for Advanced Macromolecular Design (CAMD)
- School of Chemical Engineering and School of Chemistry
- University of New South Wales
- Sydney
- Australia
| |
Collapse
|
165
|
Mancuso N, Rohland N, Rand KA, Tandon A, Allen A, Quinque D, Mallick S, Li H, Stram A, Sheng X, Kote-Jarai Z, Easton DF, Eeles RA, Le Marchand L, Lubwama A, Stram D, Watya S, Conti DV, Henderson B, Haiman CA, Pasaniuc B, Reich D. The contribution of rare variation to prostate cancer heritability. Nat Genet 2016; 48:30-5. [PMID: 26569126 PMCID: PMC7534691 DOI: 10.1038/ng.3446] [Citation(s) in RCA: 98] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2015] [Accepted: 10/20/2015] [Indexed: 12/13/2022]
Abstract
We report targeted sequencing of 63 known prostate cancer risk regions in a multi-ancestry study of 9,237 men and use the data to explore the contribution of low-frequency variation to disease risk. We show that SNPs with minor allele frequencies (MAFs) of 0.1-1% explain a substantial fraction of prostate cancer risk in men of African ancestry. We estimate that these SNPs account for 0.12 (standard error (s.e.) = 0.05) of variance in risk (∼42% of the variance contributed by SNPs with MAF of 0.1-50%). This contribution is much larger than the fraction of neutral variation due to SNPs in this class, implying that natural selection has driven down the frequency of many prostate cancer risk alleles; we estimate the coupling between selection and allelic effects at 0.48 (95% confidence interval [0.19, 0.78]) under the Eyre-Walker model. Our results indicate that rare variants make a disproportionate contribution to genetic risk for prostate cancer and suggest the possibility that rare variants may also have an outsize effect on other common traits.
Collapse
Affiliation(s)
- Nicholas Mancuso
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California, USA
| | - Nadin Rohland
- Department of Genetics, Harvard Medical School, Boston, Massachusetts, USA
- Broad Institute, Cambridge, Massachusetts, USA
| | - Kristin A Rand
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
- Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, California, USA
| | - Arti Tandon
- Department of Genetics, Harvard Medical School, Boston, Massachusetts, USA
- Broad Institute, Cambridge, Massachusetts, USA
| | - Alexander Allen
- Department of Genetics, Harvard Medical School, Boston, Massachusetts, USA
- Broad Institute, Cambridge, Massachusetts, USA
| | - Dominique Quinque
- Department of Genetics, Harvard Medical School, Boston, Massachusetts, USA
- Broad Institute, Cambridge, Massachusetts, USA
| | - Swapan Mallick
- Department of Genetics, Harvard Medical School, Boston, Massachusetts, USA
- Broad Institute, Cambridge, Massachusetts, USA
| | - Heng Li
- Department of Genetics, Harvard Medical School, Boston, Massachusetts, USA
- Broad Institute, Cambridge, Massachusetts, USA
| | - Alex Stram
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Xin Sheng
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | | | - Douglas F Easton
- Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
| | - Rosalind A Eeles
- The Institute of Cancer Research, London, UK
- Royal Marsden National Health Service (NHS) Foundation Trust, London and Sutton, UK
| | - Loic Le Marchand
- Epidemiology Program, University of Hawaii Cancer Center, Honolulu, Hawaii, USA
| | - Alex Lubwama
- School of Public Health, Makerere University College of Health Sciences, Kampala, Uganda
| | - Daniel Stram
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
- Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, California, USA
| | - Stephen Watya
- School of Public Health, Makerere University College of Health Sciences, Kampala, Uganda
| | - David V Conti
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
- Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, California, USA
| | - Brian Henderson
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
- Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, California, USA
| | - Christopher A Haiman
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
- Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, California, USA
| | - Bogdan Pasaniuc
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California, USA
- Department of Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California, USA
| | - David Reich
- Department of Genetics, Harvard Medical School, Boston, Massachusetts, USA
- Broad Institute, Cambridge, Massachusetts, USA
| |
Collapse
|
166
|
|
167
|
Ilboudo A, Chouhan J, McNeil BK, Osborne JR, Ogunwobi OO. PVT1 Exon 9: A Potential Biomarker of Aggressive Prostate Cancer? INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2015; 13:ijerph13010012. [PMID: 26703666 PMCID: PMC4730403 DOI: 10.3390/ijerph13010012] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/12/2015] [Revised: 10/29/2015] [Accepted: 11/02/2015] [Indexed: 12/14/2022]
Abstract
Prostate cancer (PCa) is the most commonly diagnosed cancer as well as the greatest source of cancer-related mortality in males of African ancestry (MoAA). Interestingly, this has been shown to be associated with single nucleotide polymorphisms around regions 2 and 3 of the 8q24 human chromosomal region. The non-protein coding gene locus Plasmacytoma Variant Translocation 1 (PVT1) is located at 8q24 and is overexpressed in PCa and, therefore, is also a candidate biomarker to explain the well-known disparity in this group. PVT1 has at least 12 exons that make separate transcripts which may have different functions, all of which are at present unknown in PCa. Our aim was to determine if any PVT1 transcripts play a role in aggressiveness and racial disparity in PCa. We used a panel of seven PCa cell lines including three derived from MoAA. Ribonucleic acid extraction, complementary deoxyribonucleic acid synthesis, and quantitative polymerase chain reaction (qPCR) were performed to evaluate expression of all 12 PVT1 exons. Each qPCR was performed in quadruplicates. At least four separate qPCR experiments were performed. Expression of PVT1 exons was inconsistent except for exon 9. There was no significant difference in exon 9 expression between cell lines derived from Caucasian males (CM), and an indolent cell line derived from MoAA. However, exon 9 expression in the aggressive MDA PCa 2b and E006AA-hT cell lines derived from MoAA was significantly higher than in other cell lines. Consequently, we observed differential expression of exon 9 of PVT1 in a manner that suggests that PVT1 exon 9 may be associated with aggressive PCa in MoAA.
Collapse
Affiliation(s)
- Adeodat Ilboudo
- Department of Biological Sciences, Hunter College, The City University of New York, New York, NY 10065, USA.
| | - Jyoti Chouhan
- Department of Urology, State University of New York Downstate Medical Center, New York, NY 11203, USA.
| | - Brian K McNeil
- Department of Urology, State University of New York Downstate Medical Center, New York, NY 11203, USA.
| | - Joseph R Osborne
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA.
| | - Olorunseun O Ogunwobi
- Department of Biological Sciences, Hunter College, The City University of New York, New York, NY 10065, USA.
- Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medical College, Cornell University, New York, NY 10065, USA.
| |
Collapse
|
168
|
Qian DC, Byun J, Han Y, Greene CS, Field JK, Hung RJ, Brhane Y, Mclaughlin JR, Fehringer G, Landi MT, Rosenberger A, Bickeböller H, Malhotra J, Risch A, Heinrich J, Hunter DJ, Henderson BE, Haiman CA, Schumacher FR, Eeles RA, Easton DF, Seminara D, Amos CI. Identification of shared and unique susceptibility pathways among cancers of the lung, breast, and prostate from genome-wide association studies and tissue-specific protein interactions. Hum Mol Genet 2015; 24:7406-20. [PMID: 26483192 PMCID: PMC4664175 DOI: 10.1093/hmg/ddv440] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2015] [Revised: 09/11/2015] [Accepted: 10/12/2015] [Indexed: 12/18/2022] Open
Abstract
Results from genome-wide association studies (GWAS) have indicated that strong single-gene effects are the exception, not the rule, for most diseases. We assessed the joint effects of germline genetic variations through a pathway-based approach that considers the tissue-specific contexts of GWAS findings. From GWAS meta-analyses of lung cancer (12 160 cases/16 838 controls), breast cancer (15 748 cases/18 084 controls) and prostate cancer (14 160 cases/12 724 controls) in individuals of European ancestry, we determined the tissue-specific interaction networks of proteins expressed from genes that are likely to be affected by disease-associated variants. Reactome pathways exhibiting enrichment of proteins from each network were compared across the cancers. Our results show that pathways associated with all three cancers tend to be broad cellular processes required for growth and survival. Significant examples include the nerve growth factor (P = 7.86 × 10(-33)), epidermal growth factor (P = 1.18 × 10(-31)) and fibroblast growth factor (P = 2.47 × 10(-31)) signaling pathways. However, within these shared pathways, the genes that influence risk largely differ by cancer. Pathways found to be unique for a single cancer focus on more specific cellular functions, such as interleukin signaling in lung cancer (P = 1.69 × 10(-15)), apoptosis initiation by Bad in breast cancer (P = 3.14 × 10(-9)) and cellular responses to hypoxia in prostate cancer (P = 2.14 × 10(-9)). We present the largest comparative cross-cancer pathway analysis of GWAS to date. Our approach can also be applied to the study of inherited mechanisms underlying risk across multiple diseases in general.
Collapse
Affiliation(s)
- David C Qian
- Department of Biomedical Data Science, Geisel School of Medicine at Dartmouth, Hanover, NH 03755, USA
| | - Jinyoung Byun
- Department of Biomedical Data Science, Geisel School of Medicine at Dartmouth, Hanover, NH 03755, USA
| | - Younghun Han
- Department of Biomedical Data Science, Geisel School of Medicine at Dartmouth, Hanover, NH 03755, USA
| | - Casey S Greene
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - John K Field
- Department of Molecular and Clinical Cancer Medicine, University of Liverpool Cancer Research Centre, Liverpool L69 3GA, UK
| | - Rayjean J Hung
- Lunenfeld-Tanenbaum Research Institute of Mount Sinai Hospital, Toronto, ON M5G 1X5, Canada
| | - Yonathan Brhane
- Lunenfeld-Tanenbaum Research Institute of Mount Sinai Hospital, Toronto, ON M5G 1X5, Canada
| | - John R Mclaughlin
- Dalla Lana School of Public Health, University of Toronto, Toronto, ON M5T 3M7, Canada
| | - Gordon Fehringer
- Lunenfeld-Tanenbaum Research Institute of Mount Sinai Hospital, Toronto, ON M5G 1X5, Canada
| | - Maria Teresa Landi
- National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Albert Rosenberger
- Department of Genetic Epidemiology, University Medical Centre Göttingen, 37099 Göttingen, Germany
| | - Heike Bickeböller
- Department of Genetic Epidemiology, University Medical Centre Göttingen, 37099 Göttingen, Germany
| | - Jyoti Malhotra
- Division of Hematology and Oncology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Angela Risch
- Division of Epigenomics and Cancer Risk Factors, German Cancer Research Center, 69120 Heidelberg, Germany
| | - Joachim Heinrich
- Institute of Epidemiology I, German Research Center for Environmental Health, 85764 Neuherberg, Germany
| | - David J Hunter
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| | - Brian E Henderson
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Christopher A Haiman
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Fredrick R Schumacher
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Rosalind A Eeles
- Department of Cancer Genetics, Institute of Cancer Research, London SW7 3RP, UK and
| | - Douglas F Easton
- Centre for Cancer Genetic Epidemiology, Department of Oncology, University of Cambridge, Cambridge CB1 8RN, UK
| | - Daniela Seminara
- National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Christopher I Amos
- Department of Biomedical Data Science, Geisel School of Medicine at Dartmouth, Hanover, NH 03755, USA,
| |
Collapse
|
169
|
Lange EM, Ribado JV, Zuhlke KA, Johnson AM, Keele GR, Li J, Wang Y, Duan Q, Li G, Gao Z, Li Y, Xu J, Zheng SL, Cooney KA. Assessing the Cumulative Contribution of New and Established Common Genetic Risk Factors to Early-Onset Prostate Cancer. Cancer Epidemiol Biomarkers Prev 2015; 25:766-72. [PMID: 26671023 DOI: 10.1158/1055-9965.epi-14-0995] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2014] [Accepted: 12/08/2015] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND We assessed the evidence for association between 23 recently reported prostate cancer variants and early-onset prostate cancer and the aggregate value of 63 prostate cancer variants for predicting early-onset disease using 931 unrelated men diagnosed with prostate cancer prior to age 56 years and 1,126 male controls. METHODS Logistic regression models were used to test the evidence for association between the 23 new variants and early-onset prostate cancer. Weighted and unweighted sums of total risk alleles across these 23 variants and 40 established variants were constructed. Weights were based on previously reported effect size estimates. Receiver operating characteristic curves and forest plots, using defined cut-points, were constructed to assess the predictive value of the burden of risk alleles on early-onset disease. RESULTS Ten of the 23 new variants demonstrated evidence (P < 0.05) for association with early-onset prostate cancer, including four that were significant after multiple test correction. The aggregate burden of risk alleles across the 63 variants was predictive of early-onset prostate cancer (AUC = 0.71 using weighted sums), especially in men with a high burden of total risk alleles. CONCLUSIONS A high burden of risk alleles is strongly associated with early-onset prostate cancer. IMPACT Our results provide the first formal replication for several of these 23 new variants and demonstrate that a high burden of common-variant risk alleles is a major risk factor for early-onset prostate cancer. Cancer Epidemiol Biomarkers Prev; 25(5); 766-72. ©2015 AACR.
Collapse
Affiliation(s)
- Ethan M Lange
- Department of Genetics, University of North Carolina, Chapel Hill, North Carolina. Department of Biostatistics, University of North Carolina, Chapel Hill, North Carolina. Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, North Carolina.
| | - Jessica V Ribado
- Department of Genetics, University of North Carolina, Chapel Hill, North Carolina
| | - Kimberly A Zuhlke
- Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan
| | - Anna M Johnson
- Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan
| | - Gregory R Keele
- Department of Genetics, University of North Carolina, Chapel Hill, North Carolina
| | - Jin Li
- Department of Genetics, University of North Carolina, Chapel Hill, North Carolina
| | - Yunfei Wang
- Center for Translational Science, Children's National Medical Center, George Washington University, Washington, D.C
| | - Qing Duan
- Department of Genetics, University of North Carolina, Chapel Hill, North Carolina
| | - Ge Li
- Center for Genomics and Personalized Medicine Research, Wake Forest University, Winston-Salem, North Carolina
| | - Zhengrong Gao
- Center for Genomics and Personalized Medicine Research, Wake Forest University, Winston-Salem, North Carolina
| | - Yun Li
- Department of Genetics, University of North Carolina, Chapel Hill, North Carolina. Department of Biostatistics, University of North Carolina, Chapel Hill, North Carolina
| | - Jianfeng Xu
- Center for Genomics and Personalized Medicine Research, Wake Forest University, Winston-Salem, North Carolina
| | - S Lilly Zheng
- Center for Genomics and Personalized Medicine Research, Wake Forest University, Winston-Salem, North Carolina
| | - Kathleen A Cooney
- Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan. Department of Urology, University of Michigan, Ann Arbor, Michigan
| |
Collapse
|
170
|
Polymorphisms of matrix metalloproteinases and their association with metastasis and the efficacy of androgen-deprivation therapy for prostate cancer in Taiwanese men. UROLOGICAL SCIENCE 2015. [DOI: 10.1016/j.urols.2015.11.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
|
171
|
Chen X, McClelland M, Jia Z, Rahmatpanah FB, Sawyers A, Trent J, Duggan D, Mercola D. The identification of trans-associations between prostate cancer GWAS SNPs and RNA expression differences in tumor-adjacent stroma. Oncotarget 2015; 6:1865-73. [PMID: 25638161 PMCID: PMC4359337 DOI: 10.18632/oncotarget.2763] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2014] [Accepted: 11/17/2014] [Indexed: 11/25/2022] Open
Abstract
Here we tested the hypothesis that SNPs associated with prostate cancer risk, might differentially affect RNA expression in prostate cancer stroma. The most significant 35 SNP loci were selected from Genome Wide Association (GWA) studies of ~40,000 patients. We also selected 4030 transcripts previously associated with prostate cancer diagnosis and prognosis. eQTL analysis was carried out by a modified BAYES method to analyze the associations between the risk variants and expressed transcripts jointly in a single model. We observed 47 significant associations between eight risk variants and the expression patterns of 46 genes. This is the first study to identify associations between multiple SNPs and multiple in trans gene expression differences in cancer stroma. Potentially, a combination of SNPs and associated expression differences in prostate stroma may increase the power of risk assessment for individuals, and for cancer progression.
Collapse
Affiliation(s)
- Xin Chen
- Genomics Center, Loma Linda University, Loma Linda, California, 92354, United States of America
| | - Michael McClelland
- Department of Pathology and Laboratory Medicine, University of California, Irvine, California, 92697, United States of America.,Department of Microbiology and Molecular Genetics, University of California, Irvine, California, 92697, United States of America
| | - Zhenyu Jia
- Department of Pathology and Laboratory Medicine, University of California, Irvine, California, 92697, United States of America.,Department of Statistics, The University of Akron, Akron, Ohio, 44325, United States of America.,Department of Family & Community Medicine, Northeast Ohio Medical University, Rootstown, Ohio, 44272, United States of America
| | - Farah B Rahmatpanah
- Department of Pathology and Laboratory Medicine, University of California, Irvine, California, 92697, United States of America
| | - Anne Sawyers
- Department of Pathology and Laboratory Medicine, University of California, Irvine, California, 92697, United States of America
| | - Jeffrey Trent
- Genetic Basis of Human Disease Division, The Translational Genomics Research Institute, Phoenix, Arizona, 85004, United States of America
| | - David Duggan
- Integrated Cancer Genomics Division, The Translational Genomics Research Institute, Phoenix, Arizona, 85004, United States of America
| | - Dan Mercola
- Department of Pathology and Laboratory Medicine, University of California, Irvine, California, 92697, United States of America
| |
Collapse
|
172
|
Shah K, Bradbury NA. Kinase modulation of androgen receptor signaling: implications for prostate cancer. ACTA ACUST UNITED AC 2015; 2. [PMID: 28580371 DOI: 10.14800/ccm.1023] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Androgens and androgen receptors play essential roles in the development and progression of prostate cancer, a disease that claims roughly 28,000 lives annually. In addition to androgen biding, androgen receptor activity can be regulated via several post-translational modifications such as ubiquitination, acetylation, phosphorylation, methylation & SUMO-ylation. Off these modifications, phosphorylation has been the most extensively studied. Modification by phosphorylation can alter androgen receptor localization, protein stability and transcriptional activity, ultimately leading to changes in the biology of cancer cells and cancer progression. Understanding, role of phosphorylated androgen receptor species holds the key to identifying a potential therapeutic drug target for patients with prostate cancer and castrate resistant prostate cancer. Here, we present a brief review of recently discovered protein kinases phosphorylating AR, focusing on the functional role of phosphorylated androgen receptor species in prostate cancer and castrate resistant prostate cancer.
Collapse
Affiliation(s)
- Kalpit Shah
- Department of Physiology and Biophysics, The Chicago Medical School, Rosalind Franklin University of Medicine & Sciences, North Chicago, IL, 60064, USA
| | - Neil A Bradbury
- Department of Physiology and Biophysics, The Chicago Medical School, Rosalind Franklin University of Medicine & Sciences, North Chicago, IL, 60064, USA
| |
Collapse
|
173
|
Yu DD, Guo SW, Jing YY, Dong YL, Wei LX. A review on hepatocyte nuclear factor-1beta and tumor. Cell Biosci 2015; 5:58. [PMID: 26464794 PMCID: PMC4603907 DOI: 10.1186/s13578-015-0049-3] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2015] [Accepted: 10/01/2015] [Indexed: 01/06/2023] Open
Abstract
Hepatocyte nuclear factor-1beta (HNF1β) was initially identified as a liver-specific transcription factor. It is a homeobox transcription factor that functions as a homodimer or heterodimer with HNF1α. HNF1β plays an important role in organogenesis during embryonic stage, especially of the liver, kidney, and pancreas. Mutations in the HNF1β gene cause maturity-onset diabetes of the young type 5 (MODY5), renal cysts, genital malformations, and pancreas atrophy. Recently, it has been shown that the expression of HNF1β is associated with cancer risk in several tumors, including hepatocellular carcinoma, pancreatic carcinoma, renal cancer, ovarian cancer, endometrial cancer, and prostate cancer. HNF1β also regulates the expression of genes associated with stem/progenitor cells, which indicates that HNF1β may play an important role in stem cell regulation. In this review, we discuss some of the current developments about HNF1β and tumor, the relationship between HNF1β and stem/progenitor cells, and the potential pathogenesis of HNF1β in various tumors.
Collapse
Affiliation(s)
- Dan-Dan Yu
- Tumor Immunology and Gene Therapy Center, Eastern Hepatobiliary Surgery Hospital, The Second Military Medical University, 225 Changhai Road, 200438 Shanghai, China
| | - Shi-Wei Guo
- Tumor Immunology and Gene Therapy Center, Eastern Hepatobiliary Surgery Hospital, The Second Military Medical University, 225 Changhai Road, 200438 Shanghai, China
| | - Ying-Ying Jing
- Tumor Immunology and Gene Therapy Center, Eastern Hepatobiliary Surgery Hospital, The Second Military Medical University, 225 Changhai Road, 200438 Shanghai, China
| | - Yu-Long Dong
- Tumor Immunology and Gene Therapy Center, Eastern Hepatobiliary Surgery Hospital, The Second Military Medical University, 225 Changhai Road, 200438 Shanghai, China
| | - Li-Xin Wei
- Tumor Immunology and Gene Therapy Center, Eastern Hepatobiliary Surgery Hospital, The Second Military Medical University, 225 Changhai Road, 200438 Shanghai, China
| |
Collapse
|
174
|
Hurley PJ, Sundi D, Shinder B, Simons BW, Hughes RM, Miller RM, Benzon B, Faraj SF, Netto GJ, Vergara IA, Erho N, Davicioni E, Karnes RJ, Yan G, Ewing C, Isaacs SD, Berman DM, Rider JR, Jordahl KM, Mucci LA, Huang J, An SS, Park BH, Isaacs WB, Marchionni L, Ross AE, Schaeffer EM. Germline Variants in Asporin Vary by Race, Modulate the Tumor Microenvironment, and Are Differentially Associated with Metastatic Prostate Cancer. Clin Cancer Res 2015; 22:448-58. [PMID: 26446945 DOI: 10.1158/1078-0432.ccr-15-0256] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2015] [Accepted: 09/10/2015] [Indexed: 12/20/2022]
Abstract
PURPOSE Prostate cancers incite tremendous morbidity upon metastatic growth. We previously identified Asporin (ASPN) as a potential mediator of metastatic progression found within the tumor microenvironment. ASPN contains an aspartic acid (D)-repeat domain and germline polymorphisms in D-repeat-length have been associated with degenerative diseases. Associations of germline ASPN D polymorphisms with risk of prostate cancer progression to metastatic disease have not been assessed. EXPERIMENTAL DESIGN Germline ASPN D-repeat-length was retrospectively analyzed in 1,600 men who underwent radical prostatectomy for clinically localized prostate cancer and in 548 noncancer controls. Multivariable Cox proportional hazards models were used to test the associations of ASPN variations with risk of subsequent oncologic outcomes, including metastasis. Orthotopic xenografts were used to establish allele- and stroma-specific roles for ASPN D variants in metastatic prostate cancer. RESULTS Variation at the ASPN D locus was differentially associated with poorer oncologic outcomes. ASPN D14 [HR, 1.72; 95% confidence interval (CI), 1.05-2.81, P = 0.032] and heterozygosity for ASPN D13/14 (HR, 1.86; 95% CI, 1.03-3.35, P = 0.040) were significantly associated with metastatic recurrence, while homozygosity for the ASPN D13 variant was significantly associated with a reduced risk of metastatic recurrence (HR, 0.44; 95% CI, 0.21-0.94, P = 0.035) in multivariable analyses. Orthotopic xenografts established biologic roles for ASPN D14 and ASPN D13 variants in metastatic prostate cancer progression that were consistent with patient-based data. CONCLUSIONS We observed associations between ASPN D variants and oncologic outcomes, including metastasis. Our data suggest that ASPN expressed in the tumor microenvironment is a heritable modulator of metastatic progression.
Collapse
Affiliation(s)
- Paula J Hurley
- Brady Urological Institute, Department of Urology, Johns Hopkins University, Baltimore, Maryland. Department of Oncology, Johns Hopkins University, Baltimore, Maryland. Sidney Kimmel Comprehensive Cancer Institute, Johns Hopkins University, Baltimore, Maryland.
| | - Debasish Sundi
- Brady Urological Institute, Department of Urology, Johns Hopkins University, Baltimore, Maryland
| | - Brian Shinder
- Brady Urological Institute, Department of Urology, Johns Hopkins University, Baltimore, Maryland
| | - Brian W Simons
- Brady Urological Institute, Department of Urology, Johns Hopkins University, Baltimore, Maryland. Department of Molecular and Comparative Pathobiology, Johns Hopkins University, Baltimore, Maryland
| | - Robert M Hughes
- Brady Urological Institute, Department of Urology, Johns Hopkins University, Baltimore, Maryland
| | - Rebecca M Miller
- Brady Urological Institute, Department of Urology, Johns Hopkins University, Baltimore, Maryland
| | - Benjamin Benzon
- Brady Urological Institute, Department of Urology, Johns Hopkins University, Baltimore, Maryland
| | - Sheila F Faraj
- Department of Pathology, Johns Hopkins University, Baltimore, Maryland
| | - George J Netto
- Department of Pathology, Johns Hopkins University, Baltimore, Maryland
| | | | - Nicholas Erho
- Genome Dx Biosciences Inc., Vancouver, British Columbia, Canada
| | - Elai Davicioni
- Genome Dx Biosciences Inc., Vancouver, British Columbia, Canada
| | | | - Guifang Yan
- Brady Urological Institute, Department of Urology, Johns Hopkins University, Baltimore, Maryland
| | - Charles Ewing
- Brady Urological Institute, Department of Urology, Johns Hopkins University, Baltimore, Maryland
| | - Sarah D Isaacs
- Brady Urological Institute, Department of Urology, Johns Hopkins University, Baltimore, Maryland
| | - David M Berman
- Department of Pathology and Molecular Medicine and Cancer Research Institute, Queens University, Kingston, Ontario, Canada
| | - Jennifer R Rider
- Department of Epidemiology, Harvard University, T.H. Chan School of Public Health, Boston, Massachusetts
| | - Kristina M Jordahl
- Department of Epidemiology, Harvard University, T.H. Chan School of Public Health, Boston, Massachusetts
| | - Lorelei A Mucci
- Department of Epidemiology, Harvard University, T.H. Chan School of Public Health, Boston, Massachusetts
| | - Jessie Huang
- The Department of Environmental Health Sciences, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland
| | - Steven S An
- The Department of Environmental Health Sciences, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland. The Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, Baltimore, Maryland. Physical Sciences-Oncology Center, Johns Hopkins University, Baltimore, Maryland
| | - Ben H Park
- Department of Oncology, Johns Hopkins University, Baltimore, Maryland. Sidney Kimmel Comprehensive Cancer Institute, Johns Hopkins University, Baltimore, Maryland
| | - William B Isaacs
- Brady Urological Institute, Department of Urology, Johns Hopkins University, Baltimore, Maryland
| | - Luigi Marchionni
- Department of Oncology, Johns Hopkins University, Baltimore, Maryland
| | - Ashley E Ross
- Brady Urological Institute, Department of Urology, Johns Hopkins University, Baltimore, Maryland. Department of Oncology, Johns Hopkins University, Baltimore, Maryland. Sidney Kimmel Comprehensive Cancer Institute, Johns Hopkins University, Baltimore, Maryland. Department of Pathology, Johns Hopkins University, Baltimore, Maryland
| | - Edward M Schaeffer
- Brady Urological Institute, Department of Urology, Johns Hopkins University, Baltimore, Maryland. Department of Oncology, Johns Hopkins University, Baltimore, Maryland. Sidney Kimmel Comprehensive Cancer Institute, Johns Hopkins University, Baltimore, Maryland
| |
Collapse
|
175
|
Identifying New Candidate Genes and Chemicals Related to Prostate Cancer Using a Hybrid Network and Shortest Path Approach. COMPUTATIONAL AND MATHEMATICAL METHODS IN MEDICINE 2015; 2015:462363. [PMID: 26504486 PMCID: PMC4609422 DOI: 10.1155/2015/462363] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/24/2015] [Accepted: 02/24/2015] [Indexed: 12/26/2022]
Abstract
Prostate cancer is a type of cancer that occurs in the male prostate, a gland in the male reproductive system. Because prostate cancer cells may spread to other parts of the body and can influence human reproduction, understanding the mechanisms underlying this disease is critical for designing effective treatments. The identification of as many genes and chemicals related to prostate cancer as possible will enhance our understanding of this disease. In this study, we proposed a computational method to identify new candidate genes and chemicals based on currently known genes and chemicals related to prostate cancer by applying a shortest path approach in a hybrid network. The hybrid network was constructed according to information concerning chemical-chemical interactions, chemical-protein interactions, and protein-protein interactions. Many of the obtained genes and chemicals are associated with prostate cancer.
Collapse
|
176
|
Gilbert R, Martin RM, Evans DM, Tilling K, Davey Smith G, Kemp JP, Lane JA, Hamdy FC, Neal DE, Donovan JL, Metcalfe C. Incorporating Known Genetic Variants Does Not Improve the Accuracy of PSA Testing to Identify High Risk Prostate Cancer on Biopsy. PLoS One 2015; 10:e0136735. [PMID: 26431041 PMCID: PMC4592274 DOI: 10.1371/journal.pone.0136735] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2014] [Accepted: 07/24/2015] [Indexed: 11/18/2022] Open
Abstract
INTRODUCTION Prostate-specific antigen (PSA) testing is a widely accepted screening method for prostate cancer, but with low specificity at thresholds giving good sensitivity. Previous research identified four single nucleotide polymorphisms (SNPs) principally associated with circulating PSA levels rather than with prostate cancer risk (TERT rs2736098, FGFR2 rs10788160, TBX3 rs11067228, KLK3 rs17632542). Removing the genetic contribution to PSA levels may improve the ability of the remaining biologically-determined variation in PSA to discriminate between high and low risk of progression within men with identified prostate cancer. We investigate whether incorporating information on the PSA-SNPs improves the discrimination achieved by a single PSA threshold in men with raised PSA levels. MATERIALS AND METHODS Men with PSA between 3-10 ng/mL and histologically-confirmed prostate cancer were categorised as high or low risk of progression (Low risk: Gleason score≤6 and stage T1-T2a; High risk: Gleason score 7-10 or stage T2C). We used the combined genetic effect of the four PSA-SNPs to calculate a genetically corrected PSA risk score. We calculated the Area under the Curve (AUC) to determine how well genetically corrected PSA risk scores distinguished men at high risk of progression from low risk men. RESULTS The analysis includes 868 men with prostate cancer (Low risk: 684 (78.8%); High risk: 184 (21.2%)). Receiver operating characteristic (ROC) curves indicate that including the 4 PSA-SNPs does not improve the performance of measured PSA as a screening tool for high/low risk prostate cancer (measured PSA level AUC = 59.5% (95% CI: 54.7,64.2) vs additionally including information from the 4 PSA-SNPs AUC = 59.8% (95% CI: 55.2,64.5) (p-value = 0.40)). CONCLUSION We demonstrate that genetically correcting PSA for the combined genetic effect of four PSA-SNPs, did not improve discrimination between high and low risk prostate cancer in men with raised PSA levels (3-10 ng/mL). Replication and gaining more accurate estimates of the effects of the 4 PSA-SNPs and additional variants associated with PSA levels and not prostate cancer could be obtained from subsequent GWAS from larger prospective studies.
Collapse
Affiliation(s)
- Rebecca Gilbert
- School of Social and Community Medicine, University of Bristol, Bristol, United Kingdom
| | - Richard M. Martin
- School of Social and Community Medicine, University of Bristol, Bristol, United Kingdom
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, United Kingdom
| | - David M. Evans
- School of Social and Community Medicine, University of Bristol, Bristol, United Kingdom
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, United Kingdom
- University of Queensland Diamantina Institute, Translational Research Institute, Brisbane, Queensland, Australia
| | - Kate Tilling
- School of Social and Community Medicine, University of Bristol, Bristol, United Kingdom
| | - George Davey Smith
- School of Social and Community Medicine, University of Bristol, Bristol, United Kingdom
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, United Kingdom
| | - John P. Kemp
- School of Social and Community Medicine, University of Bristol, Bristol, United Kingdom
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, United Kingdom
- University of Queensland Diamantina Institute, Translational Research Institute, Brisbane, Queensland, Australia
| | - J. Athene Lane
- School of Social and Community Medicine, University of Bristol, Bristol, United Kingdom
| | - Freddie C. Hamdy
- Nuffield Department of Surgical Sciences, University of Oxford, Oxford, United Kingdom
| | - David E. Neal
- Department of Oncology, University of Cambridge, Cambridge, United Kingdom
| | - Jenny L. Donovan
- School of Social and Community Medicine, University of Bristol, Bristol, United Kingdom
| | - Chris Metcalfe
- School of Social and Community Medicine, University of Bristol, Bristol, United Kingdom
| |
Collapse
|
177
|
Han Y, Hazelett DJ, Wiklund F, Schumacher FR, Stram DO, Berndt SI, Wang Z, Rand KA, Hoover RN, Machiela MJ, Yeager M, Burdette L, Chung CC, Hutchinson A, Yu K, Xu J, Travis RC, Key TJ, Siddiq A, Canzian F, Takahashi A, Kubo M, Stanford JL, Kolb S, Gapstur SM, Diver WR, Stevens VL, Strom SS, Pettaway CA, Al Olama AA, Kote-Jarai Z, Eeles RA, Yeboah ED, Tettey Y, Biritwum RB, Adjei AA, Tay E, Truelove A, Niwa S, Chokkalingam AP, Isaacs WB, Chen C, Lindstrom S, Le Marchand L, Giovannucci EL, Pomerantz M, Long H, Li F, Ma J, Stampfer M, John EM, Ingles SA, Kittles RA, Murphy AB, Blot WJ, Signorello LB, Zheng W, Albanes D, Virtamo J, Weinstein S, Nemesure B, Carpten J, Leske MC, Wu SY, Hennis AJM, Rybicki BA, Neslund-Dudas C, Hsing AW, Chu L, Goodman PJ, Klein EA, Zheng SL, Witte JS, Casey G, Riboli E, Li Q, Freedman ML, Hunter DJ, Gronberg H, Cook MB, Nakagawa H, Kraft P, Chanock SJ, Easton DF, Henderson BE, Coetzee GA, Conti DV, Haiman CA. Integration of multiethnic fine-mapping and genomic annotation to prioritize candidate functional SNPs at prostate cancer susceptibility regions. Hum Mol Genet 2015; 24:5603-18. [PMID: 26162851 PMCID: PMC4572069 DOI: 10.1093/hmg/ddv269] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2015] [Accepted: 07/07/2015] [Indexed: 01/27/2023] Open
Abstract
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.
Collapse
Affiliation(s)
- Ying Han
- Department of Preventive Medicine, Keck School of Medicine
| | | | - Fredrik Wiklund
- Department of Medical Epidemiology and Biostatistics, Karolinska Institute, Stockholm, Sweden
| | - Fredrick R Schumacher
- Department of Preventive Medicine, Keck School of Medicine, Norris Comprehensive Cancer Center
| | - Daniel O Stram
- Department of Preventive Medicine, Keck School of Medicine, Norris Comprehensive Cancer Center
| | - Sonja I Berndt
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Zhaoming Wang
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA, Cancer Genomics Research Laboratory, NCI-DCEG, SAIC-Frederick Inc., Frederick, MD, USA
| | - Kristin A Rand
- Department of Preventive Medicine, Keck School of Medicine
| | - Robert N Hoover
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Mitchell J Machiela
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Merideth Yeager
- Cancer Genomics Research Laboratory, NCI-DCEG, SAIC-Frederick Inc., Frederick, MD, USA
| | - Laurie Burdette
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA, Cancer Genomics Research Laboratory, NCI-DCEG, SAIC-Frederick Inc., Frederick, MD, USA
| | - Charles C Chung
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Amy Hutchinson
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA, Cancer Genomics Research Laboratory, NCI-DCEG, SAIC-Frederick Inc., Frederick, MD, USA
| | - Kai Yu
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Jianfeng Xu
- Program for Personalized Cancer Care and Department of Surgery, NorthShore University HealthSystem, Evanston, IL, USA
| | - Ruth C Travis
- Cancer Epidemiology Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Timothy J Key
- Cancer Epidemiology Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Afshan Siddiq
- Department of Genomics of Common Disease, School of Public Health
| | - Federico Canzian
- Genomic Epidemiology Group, German Cancer Research Center, Heidelberg, Germany
| | | | | | - Janet L Stanford
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA, USA, Department of Epidemiology, School of Public Health, University of Washington, Seattle, WA, USA
| | - Suzanne Kolb
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Susan M Gapstur
- Epidemiology Research Program, American Cancer Society, Atlanta, GA, USA
| | - W Ryan Diver
- Epidemiology Research Program, American Cancer Society, Atlanta, GA, USA
| | - Victoria L Stevens
- Epidemiology Research Program, American Cancer Society, Atlanta, GA, USA
| | | | - Curtis A Pettaway
- Department of Urology, University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
| | - Ali Amin Al Olama
- Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
| | | | - Rosalind A Eeles
- The Institute of Cancer Research, London, UK, Royal Marsden National Health Services (NHS) Foundation Trust, London and Sutton, UK
| | - Edward D Yeboah
- Korle Bu Teaching Hospital, Accra, Ghana, University of Ghana Medical School, Accra, Ghana
| | - Yao Tettey
- Korle Bu Teaching Hospital, Accra, Ghana, University of Ghana Medical School, Accra, Ghana
| | - Richard B Biritwum
- Korle Bu Teaching Hospital, Accra, Ghana, University of Ghana Medical School, Accra, Ghana
| | - Andrew A Adjei
- Korle Bu Teaching Hospital, Accra, Ghana, University of Ghana Medical School, Accra, Ghana
| | - Evelyn Tay
- Korle Bu Teaching Hospital, Accra, Ghana, University of Ghana Medical School, Accra, Ghana
| | | | | | | | - William B Isaacs
- James Buchanan Brady Urological Institute, Johns Hopkins Hospital and Medical Institution, Baltimore, MD, USA
| | - Constance Chen
- Program in Genetic Epidemiology and Statistical Genetics, Department of Epidemiology
| | - Sara Lindstrom
- Program in Genetic Epidemiology and Statistical Genetics, Department of Epidemiology
| | - Loic Le Marchand
- Epidemiology Program, University of Hawaii Cancer Center, Honolulu, HI, USA
| | | | | | - Henry Long
- Department of Medical Oncology, Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Fugen Li
- Department of Medical Oncology, Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Jing Ma
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | | | - Esther M John
- Cancer Prevention Institute of California, Fremont, CA, USA, Division of Epidemiology, Department of Health Research and Policy, and Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Sue A Ingles
- Department of Preventive Medicine, Keck School of Medicine, Norris Comprehensive Cancer Center
| | - Rick A Kittles
- University of Arizona College of Medicine and University of Arizona Cancer Center, Tucson, AZ, USA
| | - Adam B Murphy
- Department of Urology, Northwestern University, Chicago, IL, USA
| | - William J Blot
- International Epidemiology Institute, Rockville, MD, USA, Division of Epidemiology, Department of Medicine, Vanderbilt Epidemiology Center, Vanderbilt University School of Medicine, Nashville, TN, USA
| | | | - Wei Zheng
- Division of Epidemiology, Department of Medicine, Vanderbilt Epidemiology Center, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Demetrius Albanes
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Jarmo Virtamo
- Department of Chronic Disease Prevention, National Institute for Health and Welfare, Helsinki, Finland
| | - Stephanie Weinstein
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Barbara Nemesure
- Department of Preventive Medicine, Stony Brook University, Stony Brook, NY, USA
| | - John Carpten
- The Translational Genomics Research Institute, Phoenix, AZ, USA
| | - M Cristina Leske
- Department of Preventive Medicine, Stony Brook University, Stony Brook, NY, USA
| | - Suh-Yuh Wu
- Department of Preventive Medicine, Stony Brook University, Stony Brook, NY, USA
| | - Anselm J M Hennis
- Department of Preventive Medicine, Stony Brook University, Stony Brook, NY, USA, Chronic Disease Research Centre and Faculty of Medical Sciences, University of the West Indies, Bridgetown, Barbados
| | - Benjamin A Rybicki
- Department of Public Health Sciences, Henry Ford Hospital, Detroit, MI, USA
| | | | - Ann W Hsing
- Cancer Prevention Institute of California, Fremont, CA, USA, Division of Epidemiology, Department of Health Research and Policy, and Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Lisa Chu
- Cancer Prevention Institute of California, Fremont, CA, USA, Division of Epidemiology, Department of Health Research and Policy, and Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Phyllis J Goodman
- SWOG Statistical Center, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Eric A Klein
- Department of Urology, Glickman Urological and Kidney Institute, Cleveland Clinic, Cleveland, OH, USA
| | - S Lilly Zheng
- Center for Cancer Genomics, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - John S Witte
- Department of Epidemiology and Biostatistics, Institute for Human Genetics, University of California, San Francisco, CA, USA and
| | - Graham Casey
- Department of Preventive Medicine, Keck School of Medicine, Norris Comprehensive Cancer Center
| | - Elio Riboli
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College, London, UK
| | - Qiyuan Li
- Medical College, Xiamen University, Xiamen 361102, China
| | | | - David J Hunter
- Program in Genetic Epidemiology and Statistical Genetics, Department of Epidemiology
| | - Henrik Gronberg
- Department of Medical Epidemiology and Biostatistics, Karolinska Institute, Stockholm, Sweden
| | - Michael B Cook
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Hidewaki Nakagawa
- Laboratory for Genome Sequencing Analysis, RIKEN Center for Integrative Medical Sciences, Tokyo, Japan
| | - Peter Kraft
- Program in Genetic Epidemiology and Statistical Genetics, Department of Epidemiology, Department of Biostatistics, Harvard School of Public Health, Boston, MA, USA
| | - Stephen J Chanock
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Douglas F Easton
- Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
| | - Brian E Henderson
- Department of Preventive Medicine, Keck School of Medicine, Norris Comprehensive Cancer Center
| | - Gerhard A Coetzee
- Department of Preventive Medicine, Keck School of Medicine, Norris Comprehensive Cancer Center, Department of Urology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - David V Conti
- Department of Preventive Medicine, Keck School of Medicine, Norris Comprehensive Cancer Center
| | - Christopher A Haiman
- Department of Preventive Medicine, Keck School of Medicine, Norris Comprehensive Cancer Center,
| |
Collapse
|
178
|
Amin Al Olama A, Dadaev T, Hazelett DJ, Li Q, Leongamornlert D, Saunders EJ, Stephens S, Cieza-Borrella C, Whitmore I, Benlloch Garcia S, Giles GG, Southey MC, Fitzgerald L, Gronberg H, Wiklund F, Aly M, Henderson BE, Schumacher F, Haiman CA, Schleutker J, Wahlfors T, Tammela TL, Nordestgaard BG, Key TJ, Travis RC, Neal DE, Donovan JL, Hamdy FC, Pharoah P, Pashayan N, Khaw KT, Stanford JL, Thibodeau SN, Mcdonnell SK, Schaid DJ, Maier C, Vogel W, Luedeke M, Herkommer K, Kibel AS, Cybulski C, Wokołorczyk D, Kluzniak W, Cannon-Albright L, Brenner H, Butterbach K, Arndt V, Park JY, Sellers T, Lin HY, Slavov C, Kaneva R, Mitev V, Batra J, Clements JA, Spurdle A, Teixeira MR, Paulo P, Maia S, Pandha H, Michael A, Kierzek A, Govindasami K, Guy M, Lophatonanon A, Muir K, Viñuela A, Brown AA, Freedman M, Conti DV, Easton D, Coetzee GA, Eeles RA, Kote-Jarai Z. Multiple novel prostate cancer susceptibility signals identified by fine-mapping of known risk loci among Europeans. Hum Mol Genet 2015; 24:5589-602. [PMID: 26025378 PMCID: PMC4572072 DOI: 10.1093/hmg/ddv203] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2015] [Revised: 05/26/2015] [Accepted: 05/27/2015] [Indexed: 02/02/2023] Open
Abstract
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.
Collapse
Affiliation(s)
- Ali Amin Al Olama
- Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, Strangeways Research Laboratory
| | - Tokhir Dadaev
- Division of Genetics and Epidemiology, The Institute of Cancer Research & Royal Marsden NHS Foundation Trust, London, UK
| | - Dennis J Hazelett
- Department of Urology, Norris Comprehensive Cancer Center, Keck School of Medicine, USC, Los Angeles, CA, USA, Department of Preventive Medicine, Norris Comprehensive Cancer Center, Keck School of Medicine, USC, Los Angeles, CA, USA
| | - Qiuyan Li
- Medical College, Xiamen University, Xiamen, China
| | - Daniel Leongamornlert
- Division of Genetics and Epidemiology, The Institute of Cancer Research & Royal Marsden NHS Foundation Trust, London, UK
| | - Edward J Saunders
- Division of Genetics and Epidemiology, The Institute of Cancer Research & Royal Marsden NHS Foundation Trust, London, UK
| | - Sarah Stephens
- Division of Genetics and Epidemiology, The Institute of Cancer Research & Royal Marsden NHS Foundation Trust, London, UK
| | - Clara Cieza-Borrella
- Division of Genetics and Epidemiology, The Institute of Cancer Research & Royal Marsden NHS Foundation Trust, London, UK
| | - Ian Whitmore
- Division of Genetics and Epidemiology, The Institute of Cancer Research & Royal Marsden NHS Foundation Trust, London, UK
| | - Sara Benlloch Garcia
- Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, Strangeways Research Laboratory
| | - Graham G Giles
- Cancer Epidemiology Centre, The Cancer Council Victoria, Melbourne, VIC, Australia, Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health
| | - Melissa C Southey
- Genetic Epidemiology Laboratory, Department of Pathology, The University of Melbourne, Parkville, VIC, Australia
| | | | - Henrik Gronberg
- Department of Medical Epidemiology and Biostatistics, Karolinska Institute, Stockholm, Sweden
| | - Fredrik Wiklund
- Department of Medical Epidemiology and Biostatistics, Karolinska Institute, Stockholm, Sweden
| | - Markus Aly
- Department of Medical Epidemiology and Biostatistics, Karolinska Institute, Stockholm, Sweden, Department of Clinical Sciences, Danderyds Hospital, Stockholm, Sweden
| | - Brian E Henderson
- Department of Preventive Medicine, Norris Comprehensive Cancer Center, Keck School of Medicine, USC, Los Angeles, CA, USA
| | - Fredrick Schumacher
- Department of Preventive Medicine, Norris Comprehensive Cancer Center, Keck School of Medicine, USC, Los Angeles, CA, USA
| | - Christopher A Haiman
- Department of Preventive Medicine, Norris Comprehensive Cancer Center, Keck School of Medicine, USC, Los Angeles, CA, USA
| | - Johanna Schleutker
- Department of Medical Biochemistry and Genetics Institute of Biomedicine, University of Turku, Turku, Finland, BioMediTech, University of Tampere and FimLab Laboratories, Tampere, Finland
| | - Tiina Wahlfors
- BioMediTech, University of Tampere and FimLab Laboratories, Tampere, Finland
| | - Teuvo L Tammela
- Department of Urology, Tampere University Hospital and Medical School, University of Tampere, Tampere, Finland
| | - Børge G Nordestgaard
- Department of Clinical Biochemistry, Herlev Hospital, Copenhagen University Hospital, Herlev, Denmark, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Tim J Key
- Cancer Epidemiology, Nuffield Department of Population Health
| | - Ruth C Travis
- Cancer Epidemiology, Nuffield Department of Population Health
| | - David E Neal
- Department of Oncology, Addenbrooke's Hospital, Cancer Research UK Cambridge Research Institute, Li Ka Shing Centre, Cambridge, UK
| | - Jenny L Donovan
- School of Social and Community Medicine, University of Bristol, Bristol, UK
| | - Freddie C Hamdy
- Nuffield Department of Surgical Sciences, University of Oxford, Oxford, UK, Faculty of Medical Science, John Radcliffe Hospital, University of Oxford, Oxford, UK
| | - Paul Pharoah
- Centre for Cancer Genetic Epidemiology, Department of Oncology, Strangeways Laboratory
| | - Nora Pashayan
- Centre for Cancer Genetic Epidemiology, Department of Oncology, Strangeways Laboratory, Department of Applied Health Research, University College London, London, UK
| | - Kay-Tee Khaw
- Clinical Gerontology Unit, University of Cambridge, Cambridge, UK
| | - Janet L Stanford
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA, USA, Department of Epidemiology, School of Public Health, University of Washington, Seattle, WA, USA
| | | | | | | | | | - Walther Vogel
- Institute of Human Genetics, University of Ulm, Ulm, Germany
| | - Manuel Luedeke
- Department of Urology, University Hospital Ulm, Ulm, Germany
| | - Kathleen Herkommer
- Department of Urology, Klinikum rechts der Isar der Technischen Universitaet Muenchen, Munich, Germany
| | - Adam S Kibel
- Division of Urologic Surgery, Brigham and Women's Hospital, Dana-Farber Cancer Institute, Boston, USA
| | - Cezary Cybulski
- International Hereditary Cancer Center, Department of Genetics and Pathology, Pomeranian Medical University, Szczecin, Poland
| | - Dominika Wokołorczyk
- International Hereditary Cancer Center, Department of Genetics and Pathology, Pomeranian Medical University, Szczecin, Poland
| | - Wojciech Kluzniak
- International Hereditary Cancer Center, Department of Genetics and Pathology, Pomeranian Medical University, Szczecin, Poland
| | - Lisa Cannon-Albright
- Division of Genetic Epidemiology, Department of Medicine, University of Utah School of Medicine, Salt Lake City, UT, USA, George E. Wahlen Department of Veterans Affairs Medical Center, Salt Lake City, UT, USA
| | - Hermann Brenner
- Division of Clinical Epidemiology and Aging Research, German Cancer Research Center (DKFZ), Heidelberg, Germany, German Cancer Consortium (DKTK), Heidelberg, Germany
| | - Katja Butterbach
- Division of Clinical Epidemiology and Aging Research, German Cancer Research Center (DKFZ), Heidelberg, Germany, German Cancer Consortium (DKTK), Heidelberg, Germany
| | - Volker Arndt
- Division of Clinical Epidemiology and Aging Research, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Jong Y Park
- Department of Cancer Epidemiology, Moffitt Cancer Center, Tampa, FL, USA
| | - Thomas Sellers
- Department of Cancer Epidemiology, Moffitt Cancer Center, Tampa, FL, USA
| | - Hui-Yi Lin
- Biostatistics Program, Moffitt Cancer Center, Tampa, FL, USA
| | - Chavdar Slavov
- Department of Urology and Alexandrovska University Hospital, Medical University, Sofia, Bulgaria
| | - Radka Kaneva
- Department of Medical Chemistry and Biochemistry, Molecular Medicine Center, Medical University, Sofia, Bulgaria
| | - Vanio Mitev
- Department of Medical Chemistry and Biochemistry, Molecular Medicine Center, Medical University, Sofia, Bulgaria
| | - Jyotsna Batra
- Australian Prostate Cancer Research Centre-Qld, Institute of Health and Biomedical Innovation and School of Biomedical Science, Queensland University of Technology, Brisbane, Australia
| | - Judith A Clements
- Australian Prostate Cancer Research Centre-Qld, Institute of Health and Biomedical Innovation and School of Biomedical Science, Queensland University of Technology, Brisbane, Australia
| | - Amanda Spurdle
- Molecular Cancer Epidemiology Laboratory, Queensland Institute of Medical Research, Brisbane, Australia
| | - Manuel R Teixeira
- Department of Genetics, Portuguese Oncology Institute, Porto, Portugal, Biomedical Sciences Institute (ICBAS), University of Porto, Porto, Portugal
| | - Paula Paulo
- Department of Genetics, Portuguese Oncology Institute, Porto, Portugal
| | - Sofia Maia
- Department of Genetics, Portuguese Oncology Institute, Porto, Portugal
| | | | | | | | - Koveela Govindasami
- Division of Genetics and Epidemiology, The Institute of Cancer Research & Royal Marsden NHS Foundation Trust, London, UK
| | - Michelle Guy
- Division of Genetics and Epidemiology, The Institute of Cancer Research & Royal Marsden NHS Foundation Trust, London, UK
| | - Artitaya Lophatonanon
- Institute of Population Health, University of Manchester, Manchester, UK, Warwick Medical School, University of Warwick, Coventry, UK
| | - Kenneth Muir
- Institute of Population Health, University of Manchester, Manchester, UK, Warwick Medical School, University of Warwick, Coventry, UK
| | - Ana Viñuela
- Department of Twin Research & Genetic Epidemiology, King's College London, London, UK
| | - Andrew A Brown
- NORMENT, KG Jebsen Centre for Psychosis Research, Institute of Clinical Medicine, University of Oslo, Oslo, Norway, Department of Genetic Medicine and Development, University of Geneva, Switzerland and
| | | | - David V Conti
- Department of Urology, Norris Comprehensive Cancer Center, Keck School of Medicine, USC, Los Angeles, CA, USA, Department of Preventive Medicine, Norris Comprehensive Cancer Center, Keck School of Medicine, USC, Los Angeles, CA, USA
| | - Douglas Easton
- Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, Strangeways Research Laboratory
| | - Gerhard A Coetzee
- Department of Urology, Norris Comprehensive Cancer Center, Keck School of Medicine, USC, Los Angeles, CA, USA, Department of Preventive Medicine, Norris Comprehensive Cancer Center, Keck School of Medicine, USC, Los Angeles, CA, USA
| | - Rosalind A Eeles
- Division of Genetics and Epidemiology, The Institute of Cancer Research & Royal Marsden NHS Foundation Trust, London, UK
| | - Zsofia Kote-Jarai
- Division of Genetics and Epidemiology, The Institute of Cancer Research & Royal Marsden NHS Foundation Trust, London, UK,
| |
Collapse
|
179
|
Yamoah K, Johnson MH, Choeurng V, Faisal FA, Yousefi K, Haddad Z, Ross AE, Alshalafa M, Den R, Lal P, Feldman M, Dicker AP, Klein EA, Davicioni E, Rebbeck TR, Schaeffer EM. Novel Biomarker Signature That May Predict Aggressive Disease in African American Men With Prostate Cancer. J Clin Oncol 2015; 33:2789-96. [PMID: 26195723 PMCID: PMC4550692 DOI: 10.1200/jco.2014.59.8912] [Citation(s) in RCA: 107] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
PURPOSE We studied the ethnicity-specific expression of prostate cancer (PC) -associated biomarkers to evaluate whether genetic/biologic factors affect ethnic disparities in PC pathogenesis and disease progression. PATIENTS AND METHODS A total of 154 African American (AA) and 243 European American (EA) patients from four medical centers were matched according to the Cancer of the Prostate Risk Assessment postsurgical score within each institution. The distribution of mRNA expression levels of 20 validated biomarkers reported to be associated with PC initiation and progression was compared with ethnicity using false discovery rate, adjusted Wilcoxon-Mann-Whitney, and logistic regression models. A conditional logistic regression model was used to evaluate the interaction between ethnicity and biomarkers for predicting clinicopathologic outcomes. RESULTS Of the 20 biomarkers examined, six showed statistically significant differential expression in AA compared with EA men in one or more statistical models. These include ERG (P < .001), AMACR (P < .001), SPINK1 (P = .001), NKX3-1 (P = .03), GOLM1 (P = .03), and androgen receptor (P = .04). Dysregulation of AMACR (P = .036), ERG (P = .036), FOXP1 (P = .041), and GSTP1 (P = .049) as well as loss-of-function mutations for tumor suppressors NKX3-1 (P = .025) and RB1 (P = .037) predicted risk of pathologic T3 disease in an ethnicity-dependent manner. Dysregulation of GOLM1 (P = .037), SRD5A2 (P = .023), and MKi67 (P = .023) predicted clinical outcomes, including 3-year biochemical recurrence and metastasis at 5 years. A greater proportion of AA men than EA men had triple-negative (ERG-negative/ETS-negative/SPINK1-negative) disease (51% v 35%; P = .002). CONCLUSION We have identified a subset of PC biomarkers that predict the risk of clinicopathologic outcomes in an ethnicity-dependent manner. These biomarkers may explain in part the biologic contribution to ethnic disparity in PC outcomes between EA and AA men.
Collapse
Affiliation(s)
- Kosj Yamoah
- Kosj Yamoah, Robert Den, and Adam P. Dicker, Thomas Jefferson University Hospital; Priti Lal, Michael Feldman, and Timothy R. Rebbeck, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA; Kosj Yamoah, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL; Michael H. Johnson, Farzana A. Faisal, Ashley E. Ross, and Edward M. Schaeffer, Johns Hopkins University, Baltimore, MD; Voleak Choeurng, Kasra Yousefi, Zaid Haddad, Mohammed Alshalafa, and Elai Davicioni, GenomeDx Biosciences, Vancouver, British Columbia, Canada; and Eric A. Klein, The Cleveland Clinic Foundation, Cleveland, OH.
| | - Michael H Johnson
- Kosj Yamoah, Robert Den, and Adam P. Dicker, Thomas Jefferson University Hospital; Priti Lal, Michael Feldman, and Timothy R. Rebbeck, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA; Kosj Yamoah, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL; Michael H. Johnson, Farzana A. Faisal, Ashley E. Ross, and Edward M. Schaeffer, Johns Hopkins University, Baltimore, MD; Voleak Choeurng, Kasra Yousefi, Zaid Haddad, Mohammed Alshalafa, and Elai Davicioni, GenomeDx Biosciences, Vancouver, British Columbia, Canada; and Eric A. Klein, The Cleveland Clinic Foundation, Cleveland, OH
| | - Voleak Choeurng
- Kosj Yamoah, Robert Den, and Adam P. Dicker, Thomas Jefferson University Hospital; Priti Lal, Michael Feldman, and Timothy R. Rebbeck, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA; Kosj Yamoah, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL; Michael H. Johnson, Farzana A. Faisal, Ashley E. Ross, and Edward M. Schaeffer, Johns Hopkins University, Baltimore, MD; Voleak Choeurng, Kasra Yousefi, Zaid Haddad, Mohammed Alshalafa, and Elai Davicioni, GenomeDx Biosciences, Vancouver, British Columbia, Canada; and Eric A. Klein, The Cleveland Clinic Foundation, Cleveland, OH
| | - Farzana A Faisal
- Kosj Yamoah, Robert Den, and Adam P. Dicker, Thomas Jefferson University Hospital; Priti Lal, Michael Feldman, and Timothy R. Rebbeck, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA; Kosj Yamoah, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL; Michael H. Johnson, Farzana A. Faisal, Ashley E. Ross, and Edward M. Schaeffer, Johns Hopkins University, Baltimore, MD; Voleak Choeurng, Kasra Yousefi, Zaid Haddad, Mohammed Alshalafa, and Elai Davicioni, GenomeDx Biosciences, Vancouver, British Columbia, Canada; and Eric A. Klein, The Cleveland Clinic Foundation, Cleveland, OH
| | - Kasra Yousefi
- Kosj Yamoah, Robert Den, and Adam P. Dicker, Thomas Jefferson University Hospital; Priti Lal, Michael Feldman, and Timothy R. Rebbeck, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA; Kosj Yamoah, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL; Michael H. Johnson, Farzana A. Faisal, Ashley E. Ross, and Edward M. Schaeffer, Johns Hopkins University, Baltimore, MD; Voleak Choeurng, Kasra Yousefi, Zaid Haddad, Mohammed Alshalafa, and Elai Davicioni, GenomeDx Biosciences, Vancouver, British Columbia, Canada; and Eric A. Klein, The Cleveland Clinic Foundation, Cleveland, OH
| | - Zaid Haddad
- Kosj Yamoah, Robert Den, and Adam P. Dicker, Thomas Jefferson University Hospital; Priti Lal, Michael Feldman, and Timothy R. Rebbeck, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA; Kosj Yamoah, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL; Michael H. Johnson, Farzana A. Faisal, Ashley E. Ross, and Edward M. Schaeffer, Johns Hopkins University, Baltimore, MD; Voleak Choeurng, Kasra Yousefi, Zaid Haddad, Mohammed Alshalafa, and Elai Davicioni, GenomeDx Biosciences, Vancouver, British Columbia, Canada; and Eric A. Klein, The Cleveland Clinic Foundation, Cleveland, OH
| | - Ashley E Ross
- Kosj Yamoah, Robert Den, and Adam P. Dicker, Thomas Jefferson University Hospital; Priti Lal, Michael Feldman, and Timothy R. Rebbeck, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA; Kosj Yamoah, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL; Michael H. Johnson, Farzana A. Faisal, Ashley E. Ross, and Edward M. Schaeffer, Johns Hopkins University, Baltimore, MD; Voleak Choeurng, Kasra Yousefi, Zaid Haddad, Mohammed Alshalafa, and Elai Davicioni, GenomeDx Biosciences, Vancouver, British Columbia, Canada; and Eric A. Klein, The Cleveland Clinic Foundation, Cleveland, OH
| | - Mohammed Alshalafa
- Kosj Yamoah, Robert Den, and Adam P. Dicker, Thomas Jefferson University Hospital; Priti Lal, Michael Feldman, and Timothy R. Rebbeck, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA; Kosj Yamoah, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL; Michael H. Johnson, Farzana A. Faisal, Ashley E. Ross, and Edward M. Schaeffer, Johns Hopkins University, Baltimore, MD; Voleak Choeurng, Kasra Yousefi, Zaid Haddad, Mohammed Alshalafa, and Elai Davicioni, GenomeDx Biosciences, Vancouver, British Columbia, Canada; and Eric A. Klein, The Cleveland Clinic Foundation, Cleveland, OH
| | - Robert Den
- Kosj Yamoah, Robert Den, and Adam P. Dicker, Thomas Jefferson University Hospital; Priti Lal, Michael Feldman, and Timothy R. Rebbeck, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA; Kosj Yamoah, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL; Michael H. Johnson, Farzana A. Faisal, Ashley E. Ross, and Edward M. Schaeffer, Johns Hopkins University, Baltimore, MD; Voleak Choeurng, Kasra Yousefi, Zaid Haddad, Mohammed Alshalafa, and Elai Davicioni, GenomeDx Biosciences, Vancouver, British Columbia, Canada; and Eric A. Klein, The Cleveland Clinic Foundation, Cleveland, OH
| | - Priti Lal
- Kosj Yamoah, Robert Den, and Adam P. Dicker, Thomas Jefferson University Hospital; Priti Lal, Michael Feldman, and Timothy R. Rebbeck, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA; Kosj Yamoah, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL; Michael H. Johnson, Farzana A. Faisal, Ashley E. Ross, and Edward M. Schaeffer, Johns Hopkins University, Baltimore, MD; Voleak Choeurng, Kasra Yousefi, Zaid Haddad, Mohammed Alshalafa, and Elai Davicioni, GenomeDx Biosciences, Vancouver, British Columbia, Canada; and Eric A. Klein, The Cleveland Clinic Foundation, Cleveland, OH
| | - Michael Feldman
- Kosj Yamoah, Robert Den, and Adam P. Dicker, Thomas Jefferson University Hospital; Priti Lal, Michael Feldman, and Timothy R. Rebbeck, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA; Kosj Yamoah, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL; Michael H. Johnson, Farzana A. Faisal, Ashley E. Ross, and Edward M. Schaeffer, Johns Hopkins University, Baltimore, MD; Voleak Choeurng, Kasra Yousefi, Zaid Haddad, Mohammed Alshalafa, and Elai Davicioni, GenomeDx Biosciences, Vancouver, British Columbia, Canada; and Eric A. Klein, The Cleveland Clinic Foundation, Cleveland, OH
| | - Adam P Dicker
- Kosj Yamoah, Robert Den, and Adam P. Dicker, Thomas Jefferson University Hospital; Priti Lal, Michael Feldman, and Timothy R. Rebbeck, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA; Kosj Yamoah, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL; Michael H. Johnson, Farzana A. Faisal, Ashley E. Ross, and Edward M. Schaeffer, Johns Hopkins University, Baltimore, MD; Voleak Choeurng, Kasra Yousefi, Zaid Haddad, Mohammed Alshalafa, and Elai Davicioni, GenomeDx Biosciences, Vancouver, British Columbia, Canada; and Eric A. Klein, The Cleveland Clinic Foundation, Cleveland, OH
| | - Eric A Klein
- Kosj Yamoah, Robert Den, and Adam P. Dicker, Thomas Jefferson University Hospital; Priti Lal, Michael Feldman, and Timothy R. Rebbeck, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA; Kosj Yamoah, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL; Michael H. Johnson, Farzana A. Faisal, Ashley E. Ross, and Edward M. Schaeffer, Johns Hopkins University, Baltimore, MD; Voleak Choeurng, Kasra Yousefi, Zaid Haddad, Mohammed Alshalafa, and Elai Davicioni, GenomeDx Biosciences, Vancouver, British Columbia, Canada; and Eric A. Klein, The Cleveland Clinic Foundation, Cleveland, OH
| | - Elai Davicioni
- Kosj Yamoah, Robert Den, and Adam P. Dicker, Thomas Jefferson University Hospital; Priti Lal, Michael Feldman, and Timothy R. Rebbeck, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA; Kosj Yamoah, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL; Michael H. Johnson, Farzana A. Faisal, Ashley E. Ross, and Edward M. Schaeffer, Johns Hopkins University, Baltimore, MD; Voleak Choeurng, Kasra Yousefi, Zaid Haddad, Mohammed Alshalafa, and Elai Davicioni, GenomeDx Biosciences, Vancouver, British Columbia, Canada; and Eric A. Klein, The Cleveland Clinic Foundation, Cleveland, OH
| | - Timothy R Rebbeck
- Kosj Yamoah, Robert Den, and Adam P. Dicker, Thomas Jefferson University Hospital; Priti Lal, Michael Feldman, and Timothy R. Rebbeck, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA; Kosj Yamoah, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL; Michael H. Johnson, Farzana A. Faisal, Ashley E. Ross, and Edward M. Schaeffer, Johns Hopkins University, Baltimore, MD; Voleak Choeurng, Kasra Yousefi, Zaid Haddad, Mohammed Alshalafa, and Elai Davicioni, GenomeDx Biosciences, Vancouver, British Columbia, Canada; and Eric A. Klein, The Cleveland Clinic Foundation, Cleveland, OH
| | - Edward M Schaeffer
- Kosj Yamoah, Robert Den, and Adam P. Dicker, Thomas Jefferson University Hospital; Priti Lal, Michael Feldman, and Timothy R. Rebbeck, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA; Kosj Yamoah, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL; Michael H. Johnson, Farzana A. Faisal, Ashley E. Ross, and Edward M. Schaeffer, Johns Hopkins University, Baltimore, MD; Voleak Choeurng, Kasra Yousefi, Zaid Haddad, Mohammed Alshalafa, and Elai Davicioni, GenomeDx Biosciences, Vancouver, British Columbia, Canada; and Eric A. Klein, The Cleveland Clinic Foundation, Cleveland, OH
| |
Collapse
|
180
|
Chen H, Yu H, Wang J, Zhang Z, Gao Z, Chen Z, Lu Y, Liu W, Jiang D, Zheng SL, Wei GH, Issacs WB, Feng J, Xu J. Systematic enrichment analysis of potentially functional regions for 103 prostate cancer risk-associated loci. Prostate 2015; 75:1264-76. [PMID: 26015065 DOI: 10.1002/pros.23008] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2015] [Accepted: 03/27/2015] [Indexed: 12/31/2022]
Abstract
BACKGROUND More than 100 prostate cancer (PCa) risk-associated single nucleotide polymorphisms (SNPs) have been identified by genome wide association studies (GWAS). However, the molecular mechanisms are unclear for most of these SNPs. METHODS All reported PCa risk-associated SNPs reaching the genome-wide significance level of P < 1 × 10(-7) (index SNPs), as well as SNPs in linkage disequilibrium (LD, r(2) ≥ 0.5) with them were cataloged. Genomic regions with potentially functional impact were also identified, including UCSC annotated coding regions (exon and snoRNA/miRNA) and regulatory regions, as well as binding regions for transcription factors (TFs), histone modifications (HMs), DNase I hypersensitivity (DHSs), and RNA Polymerase IIA (POLR2A) defined by ChIP-Seq in prostate cell lines and tissues. Enrichment analysis was performed to test whether PCa risk-associated SNPs are located in these functional regions more than expected. RESULTS A total of 103 PCa risk-associated index SNPs and 7,244 SNPs in LD with these index SNPs were cataloged. Genomic regions with potentially functional impact, grouped in 30 different categories of functionalities, were identified. Enrichment analysis indicated that genomic regions in the following 15 categories were enriched for the PCa risk-associated SNPs: exons, CpG regions, 6 TFs (AR, ERG, FOXA1, HOXB13, CTCF, and NR3C1), 5 HMs (H3K4me1, H3K4me2, H3K4me3, H3K27AC, and H3T11P), DHSs and POLR2A. In contrast, significantly fewer PCa risk SNPs were mapped to binding regions for H3K27me3, a repressive chromatin marker. CONCLUSIONS The PCa risk-associated SNPs discovered to date may affect PCa risk through multiple different mechanisms, especially by affecting binding regions of TFs/HMs.
Collapse
Affiliation(s)
- Haitao Chen
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai, P.R. China
- Center for Cancer Genomics, Wake Forest University School of Medicine, Winston Salem, North Carolina
- Center for Genomic Translational Medicine and Prevention, Fudan School of Public Health, Fudan University, Shanghai, P.R. China
| | - Hongjie Yu
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai, P.R. China
| | - Jianqing Wang
- Fudan Institute of Urology, Huashan Hospital, Fudan University, Shanghai, P.R. China
| | - Zheng Zhang
- Center for Cancer Genomics, Wake Forest University School of Medicine, Winston Salem, North Carolina
| | - Zhengrong Gao
- Center for Cancer Genomics, Wake Forest University School of Medicine, Winston Salem, North Carolina
| | - Zhuo Chen
- Center for Cancer Genomics, Wake Forest University School of Medicine, Winston Salem, North Carolina
| | - Yulan Lu
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai, P.R. China
| | - Wennuan Liu
- Program for Personalized Cancer Care and Department of Surgery, North Shore University Health System, Evanston, Illinois
| | - Deke Jiang
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai, P.R. China
- Program for Personalized Cancer Care and Department of Surgery, North Shore University Health System, Evanston, Illinois
| | - S Lilly Zheng
- Center for Cancer Genomics, Wake Forest University School of Medicine, Winston Salem, North Carolina
- Program for Personalized Cancer Care and Department of Surgery, North Shore University Health System, Evanston, Illinois
| | - Gong-Hong Wei
- Faculty of Biochemistry and Molecular Medicine, Biocenter Oulu, University of Oulu, Oulu, Finland
| | - William B Issacs
- Department of Urology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Junjie Feng
- Center for Cancer Genomics, Wake Forest University School of Medicine, Winston Salem, North Carolina
| | - Jianfeng Xu
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai, P.R. China
- Center for Cancer Genomics, Wake Forest University School of Medicine, Winston Salem, North Carolina
- Center for Genomic Translational Medicine and Prevention, Fudan School of Public Health, Fudan University, Shanghai, P.R. China
- Fudan Institute of Urology, Huashan Hospital, Fudan University, Shanghai, P.R. China
- Program for Personalized Cancer Care and Department of Surgery, North Shore University Health System, Evanston, Illinois
| |
Collapse
|
181
|
Analysis of Prostate Cancer Susceptibility Variants in South African Men: Replicating Associations on Chromosomes 8q24 and 10q11. Prostate Cancer 2015; 2015:465184. [PMID: 26347821 PMCID: PMC4549549 DOI: 10.1155/2015/465184] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2015] [Accepted: 08/02/2015] [Indexed: 12/11/2022] Open
Abstract
Genome-wide association studies (GWAS) have implicated single nucleotide polymorphisms (SNPs) on chromosomes 2p15, 6q25, 7p15.2, 7q21, 8q24, 10q11, 10q26, 11q13, 17q12, 17q24, 19q13, and Xp11, with prostate cancer (PCa) susceptibility and/or tumour aggressiveness, in populations of African, European, and Asian ancestry. The objective of this study was to confirm these associations in South African Mixed Ancestry and White men. We evaluated 17 prioritised GWAS SNPs in South African cases (331 Mixed Ancestry and 155 White) and controls (178 Mixed Ancestry and 145 White). The replicated SNP associations for the different South African ethnic groups were rs7008482 (8q24) (p = 2.45 × 10−5), rs6983267 (8q24) (p = 4.48 × 10−7), and rs10993994 (10q11) (p = 1.40 × 10−3) in Mixed Ancestry men and rs10993994 (p = 1.56 × 10−9) in White men. No significant associations were observed for the analyses stratified by disease aggressiveness in the individual and the combined population group analysis. The present study demonstrates that a number of known PCa susceptibility variants may contribute to disease susceptibility in South African men. Larger genetic investigations extended to other South African population groups are warranted to confirm the role of these and other SNPs in disease susceptibility.
Collapse
|
182
|
Kim K, Hong SH, Kim BJ, Kim BR, Lee SY, Kim GN, Shim TS, Kook YH, Kim BJ. Separation of Mycobacterium abscessus into subspecies or genotype level by direct application of peptide nucleic acid multi-probe- real-time PCR method into sputa samples. BMC Infect Dis 2015; 15:325. [PMID: 26259717 PMCID: PMC4531893 DOI: 10.1186/s12879-015-1076-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2015] [Accepted: 07/31/2015] [Indexed: 11/30/2022] Open
Abstract
Background Recently, we introduced a novel peptide nucleic acid (PNA) multi-probe real time PCR method targeting the hsp65 gene (hsp65 PNA RT-PCR) to distinguish Mycobacterium abscessus groups. Methods Here, we evaluated the usefulness of the hsp65 PNA RT-PCR for the direct identification of the M. abscessus group at the subspecies and genotype levels from sputa samples. The method was applied to total sputa DNA from 60 different patients who were identified as having mycobacterial infections via rpoB PCR restriction analysis of the same cultures. Results The hsp65 PNA RT-PCR method had higher sensitivity than the multi-probe real-time PCR assay targeting hsp65 (HMPRT-PCR) for the detection of M. abscessus from sputum [96.7 % (29/30 samples) vs. 70 % (21/30 samples); 100 % specificity]. Conclusions These results suggest that the PNA-based method is feasible for the detection of M. abscessus members not only from cultures but also directly from sputa. Electronic supplementary material The online version of this article (doi:10.1186/s12879-015-1076-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Kijeong Kim
- Department of Microbiology, Chung-Ang University College of Medicine, Seoul, 156-756, Republic of Korea.
| | - Seok-Hyun Hong
- Department of Biomedical Sciences, Microbiology and Immunology, Cancer Research Institute, and Liver Research Institute, Seoul National University College of Medicine, 28 Yongon-dong, Chongno-gu, Seoul, 110-799, Republic of Korea.
| | - Byoung-Jun Kim
- Department of Biomedical Sciences, Microbiology and Immunology, Cancer Research Institute, and Liver Research Institute, Seoul National University College of Medicine, 28 Yongon-dong, Chongno-gu, Seoul, 110-799, Republic of Korea.
| | - Bo-Ram Kim
- Department of Biomedical Sciences, Microbiology and Immunology, Cancer Research Institute, and Liver Research Institute, Seoul National University College of Medicine, 28 Yongon-dong, Chongno-gu, Seoul, 110-799, Republic of Korea.
| | - So-Young Lee
- Department of Biomedical Sciences, Microbiology and Immunology, Cancer Research Institute, and Liver Research Institute, Seoul National University College of Medicine, 28 Yongon-dong, Chongno-gu, Seoul, 110-799, Republic of Korea.
| | - Ga-Na Kim
- Department of Biomedical Sciences, Microbiology and Immunology, Cancer Research Institute, and Liver Research Institute, Seoul National University College of Medicine, 28 Yongon-dong, Chongno-gu, Seoul, 110-799, Republic of Korea.
| | - Tae Sun Shim
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea.
| | - Yoon-Hoh Kook
- Department of Biomedical Sciences, Microbiology and Immunology, Cancer Research Institute, and Liver Research Institute, Seoul National University College of Medicine, 28 Yongon-dong, Chongno-gu, Seoul, 110-799, Republic of Korea.
| | - Bum-Joon Kim
- Department of Biomedical Sciences, Microbiology and Immunology, Cancer Research Institute, and Liver Research Institute, Seoul National University College of Medicine, 28 Yongon-dong, Chongno-gu, Seoul, 110-799, Republic of Korea.
| |
Collapse
|
183
|
Zhang Y, Qian J, Wu M, Liu M, Zhang K, Lin Y, Guo X, Zhou Z, Hu Z, Sha J. A susceptibility locus rs7099208 is associated with non-obstructive azoospermia via reduction in the expression of FAM160B1. J Biomed Res 2015; 29:491-500. [PMID: 26668583 PMCID: PMC4662211 DOI: 10.7555/jbr.29.20150034] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2015] [Revised: 03/08/2015] [Accepted: 05/06/2015] [Indexed: 12/25/2022] Open
Abstract
Non-obstructive azoospermia (NOA) is a severe defect in male reproductive health that occurs in 1% of adult men. In a previous study, we identified that rs7099208 is located within the last intron of FAM160B1 at 10q25.3. In this study, we analysed expression Quantitative Trait Loci (eQTL) of FAM160B1, ABLIM1 and TRUB1, the three genes surrounding rs7099208. Only the expression level of FAM160B1 was reduced for the homozygous alternate genotype (GG) of rs7099208, but not for the homozygous reference or heterozygous genotypes. FAM160B1 is predominantly expressed in human testes, where it is found in spermatocytes and round spermatids. From 17 patients with NOA and five with obstructive azoospermia (OA), immunohistochemistry revealed that expression of FAM160B1 is reduced, or undetectable in NOA patients, but not in OA cases or normal men. We conclude that rs7099208 is associated with NOA via a reduction in the expression of FAM160B1.
Collapse
Affiliation(s)
- Yan Zhang
- State Key Laboratory of Reproductive Medicine ; The Center for Reproductive Medicine, The Second Affiliated Hospital of Nanjing Medical University , Nanjing, Jiangsu 210011 , China
| | - Jing Qian
- State Key Laboratory of Reproductive Medicine ; Department of Histology and Embryology
| | - Minghui Wu
- State Key Laboratory of Reproductive Medicine ; Department of Histology and Embryology
| | - Mingxi Liu
- State Key Laboratory of Reproductive Medicine ; Department of Histology and Embryology
| | - Kai Zhang
- State Key Laboratory of Reproductive Medicine ; Department of Epidemiology and Biostatistics and Key Laboratory of Modern Toxicology of Ministry of Education Nanjing Medical University , Nanjing, Jiangsu 210029 , China
| | - Yuan Lin
- State Key Laboratory of Reproductive Medicine ; Department of Epidemiology and Biostatistics and Key Laboratory of Modern Toxicology of Ministry of Education Nanjing Medical University , Nanjing, Jiangsu 210029 , China
| | - Xuejiang Guo
- State Key Laboratory of Reproductive Medicine ; Department of Histology and Embryology
| | - Zuomin Zhou
- State Key Laboratory of Reproductive Medicine ; Department of Histology and Embryology
| | - Zhibin Hu
- State Key Laboratory of Reproductive Medicine ; Department of Epidemiology and Biostatistics and Key Laboratory of Modern Toxicology of Ministry of Education Nanjing Medical University , Nanjing, Jiangsu 210029 , China
| | - Jiahao Sha
- State Key Laboratory of Reproductive Medicine ; Department of Histology and Embryology
| |
Collapse
|
184
|
Novel and known genetic variants for male breast cancer risk at 8q24.21, 9p21.3, 11q13.3 and 14q24.1: results from a multicenter study in Italy. Eur J Cancer 2015; 51:2289-95. [PMID: 26248686 DOI: 10.1016/j.ejca.2015.07.020] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2015] [Accepted: 07/16/2015] [Indexed: 11/23/2022]
Abstract
Increasing evidence indicates that common genetic variants may contribute to the heritable risk of breast cancer (BC). In this study, we investigated whether single nucleotide polymorphisms (SNPs), within the 8q24.21 multi-cancer susceptibility region and within BC-associated loci widespread in the genome, may influence the risk of BC in men, and whether they may be associated with specific clinical-pathologic characteristics of male BC (MBC). In the frame of the ongoing Italian Multicenter Study on MBC, we performed a case-control study on 386 MBC cases, including 50 BRCA1/2 mutation carriers, and 1105 healthy male controls, including 197 unaffected BRCA1/2 mutation carriers. All 1491 subjects were genotyped by Sequenom iPLEX technology for a total of 29 susceptibility SNPs. By logistic regression models, we found a significant association with MBC risk for five SNPs: rs1562430 (p=0.002) and rs445114 (p=0.026) both within the 8q24.21 region; rs1011970/9p21.3 (p=0.011), rs614367/11q13.3 (p=0.016) and rs1314913/14q24.1 (p<0.0001). Differences in the distribution of rs614367/11q13.3 genotypes according to oestrogen receptor (ER) status (p=0.006), and of rs1011970/9p21.3 genotypes according to human epidermal growth factor receptor 2 (HER2) status (p=0.002) emerged. Association of rs1011970/9p21.3 risk genotype with HER2+MBC was confirmed by a multivariate analysis. rs1314913/14q24.1 was associated with increased MBC risk in analyses restricted to male BRCA1/2 mutation carriers (p=0.041). In conclusion, we provided the first evidence that the 8q24.21 region is associated with MBC risk. Furthermore, we showed that the SNPs rs1562430/8q24.21 and rs1314913/14q24.1 strongly influence BC risk in men and suggested that the SNP rs1314913/14q24.1 may act as a risk modifier locus in male BRCA1/2 mutation carriers.
Collapse
|
185
|
Lloyd T, Hounsome L, Mehay A, Mee S, Verne J, Cooper A. Lifetime risk of being diagnosed with, or dying from, prostate cancer by major ethnic group in England 2008-2010. BMC Med 2015; 13:171. [PMID: 26224061 PMCID: PMC4520076 DOI: 10.1186/s12916-015-0405-5] [Citation(s) in RCA: 72] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND In the UK, a man's lifetime risk of being diagnosed with prostate cancer is 1 in 8. We calculated both the lifetime risk of being diagnosed with and dying from prostate cancer by major ethnic group. METHODS Public Health England provided prostate cancer incidence and mortality data for England (2008-2010) by major ethnic group. Ethnicity and mortality data were incomplete, requiring various assumptions and adjustments before lifetime risk was calculated using DevCan (percent, range). RESULTS The lifetime risk of being diagnosed with prostate cancer is approximately 1 in 8 (13.3 %, 13.2-15.0 %) for White men, 1 in 4 (29.3 %, 23.5-37.2 %) for Black men, and 1 in 13 (7.9 %, 6.3-10.5 %) for Asian men, whereas that of dying from prostate cancer is approximately 1 in 24 (4.2 %, 4.2-4.7 %) for White men, 1 in 12 (8.7 %, 7.6-10.6 %) for Black men, and 1 in 44 (2.3 %, 1.9-3.0 %) for Asian men. CONCLUSIONS In England, Black men are at twice the risk of being diagnosed with, and dying from, prostate cancer compared to White men. This is an important message to communicate to Black men. White, Black, and Asian men with a prostate cancer diagnosis are all as likely to die from the disease, independent of their ethnicity. Nonetheless, proportionally more Black men are dying from prostate cancer in England.
Collapse
Affiliation(s)
- Therese Lloyd
- Evidence Team, Prostate Cancer UK, 4th Floor Counting House, 53 Tooley Street, London, UK.
| | - Luke Hounsome
- Knowledge and Intelligence Team (South West), Public Health England, 2 Rivergate, Temple Quay, Bristol, UK.
| | - Anita Mehay
- Evidence Team, Prostate Cancer UK, 4th Floor Counting House, 53 Tooley Street, London, UK.
| | - Sarah Mee
- Evidence Team, Prostate Cancer UK, 4th Floor Counting House, 53 Tooley Street, London, UK.
| | - Julia Verne
- Knowledge and Intelligence Team (South West), Public Health England, 2 Rivergate, Temple Quay, Bristol, UK.
| | - Alison Cooper
- Evidence Team, Prostate Cancer UK, 4th Floor Counting House, 53 Tooley Street, London, UK.
| |
Collapse
|
186
|
Mhatre DR, Mahale SD, Khatkhatay MI, Achrekar SK, Desai SS, Jagtap DD, Dhabalia JV, Tongaonkar HB, Dandekar SP, Varadkar AM. The rs10993994 in the proximal MSMB promoter region is a functional polymorphism in Asian Indian subjects. SPRINGERPLUS 2015; 4:380. [PMID: 26240778 PMCID: PMC4516150 DOI: 10.1186/s40064-015-1164-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/07/2015] [Accepted: 07/17/2015] [Indexed: 12/23/2022]
Abstract
BACKGROUND The microseminoprotein gene encoding prostate secretory protein of 94 amino acids (PSP94) harbours a potential risk allele (rs10993994) for prostate cancer (PCa) in its promoter region. However, studies on rs10993994 have been sparse in Asian Indians. METHODS The present study recruited a sample population of 44 benign prostatic hyperplasia patients, 33 PCa patients and 60 healthy participants, of which, participants without other confounding risk factors for PCa were retained. The serum PSP94 (sPSP94) levels were measured by a serum-based ELISA in an earlier study. A novel RFLP technique was developed to screen for rs10993994 which was validated with direct sequencing. RESULTS Sequencing showed additional 4 SNPs (rs41274660, rs141211965, rs12770171, rs10669586) and 2 novel variants (GenBank accession nos. KM265191 and KM265192). In silico DNA topographical studies predicted that KM265192 would have higher cleavage intensity and more accessibility for binding of transcription factors. Even though, similar frequencies were observed for all the variants in all the three study groups, the risk allele 'T' (rs10993994) was seen to be associated with reduced PSP94 expression both at mRNA and protein level. Further, mRNA expression as studied by real-time PCR correlated positively with sPSP94 levels. Interestingly, CC genotype of rs10993994 showed highest sPSP94 levels in all the three study groups and was associated with Gleason score ≤7 in PCa patients. In contrast, TT genotype of rs10993994 was associated with lesser sPSP94 levels and with aggressiveness of PCa. CONCLUSION rs10993994 was found to be a functional SNP in the studied Asian Indian population.
Collapse
Affiliation(s)
- Deepa R Mhatre
- Department of Biochemistry and Clinical Nutrition, Seth G.S. Medical College and K.E.M Hospital, Parel, Mumbai, 400012 India
| | - Smita D Mahale
- Division of Structural Biology, National Institute for Research in Reproductive Health, Jehangir Merwanji Street, Parel, Mumbai, 400012 India
| | - Mohammed I Khatkhatay
- Department of Molecular Immunodiagnostics, National Institute for Research in Reproductive Health, Jehangir Merwanji Street, Parel, Mumbai, India
| | - Swati K Achrekar
- Division of Structural Biology, National Institute for Research in Reproductive Health, Jehangir Merwanji Street, Parel, Mumbai, 400012 India
| | - Swapna S Desai
- Division of Structural Biology, National Institute for Research in Reproductive Health, Jehangir Merwanji Street, Parel, Mumbai, 400012 India
| | - Dhanashree D Jagtap
- Division of Structural Biology, National Institute for Research in Reproductive Health, Jehangir Merwanji Street, Parel, Mumbai, 400012 India
| | - Jayesh V Dhabalia
- Department of Urology, Seth G.S. Medical College and K.E.M Hospital, Parel, Mumbai, India
| | - Hemant B Tongaonkar
- Department of Genitourinary Oncology, Tata Memorial Hospital, Parel, Mumbai, India
| | - Sucheta P Dandekar
- Department of Biochemistry and Clinical Nutrition, Seth G.S. Medical College and K.E.M Hospital, Parel, Mumbai, 400012 India
| | - Anand M Varadkar
- Department of Biochemistry and Clinical Nutrition, Seth G.S. Medical College and K.E.M Hospital, Parel, Mumbai, 400012 India
| |
Collapse
|
187
|
Rajendran P, Kaufmann S, Vörös J, Zenobi-Wong M, Demkó L. Femtomolar oligonucleotide detection by a one-step gold nanoparticle-based assay. Colloids Surf B Biointerfaces 2015; 135:193-200. [PMID: 26263211 DOI: 10.1016/j.colsurfb.2015.07.035] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2015] [Revised: 06/20/2015] [Accepted: 07/13/2015] [Indexed: 12/11/2022]
Abstract
A sequence-specific oligonucleotide detection method based on the tail-to-tail aggregation of functionalized gold nanoparticles in the presence of target analytes is presented together with its optimization and capabilities for detection of single nucleotide polymorphisms (SNPs). In this single-step method, capture probes are freely accessible for hybridization, resulting in an improved assay performance compared to substrate-based assays. The analytes bring the nanoparticles close to each other via hybridization, causing a red shift of the nanoparticle plasmon peak detected by a spectrophotometer or CCD camera coupled to a darkfield imaging system. Optimal conditions for the assay were found to be (i) use of capture probes complementary to the target without any gap, (ii) maximum possible probe density on the gold nanoparticles, and (iii) 1M ionic strength buffer. The optimized assay has a 1 fM limit of detection and fM to 10 pM dynamic range, with detection of perfect match sequences being three orders of magnitude more sensitive than targets with single nucleotide mismatches.
Collapse
Affiliation(s)
- Prayanka Rajendran
- Laboratory of Biosensors and Bioelectronics, Institute for Biomedical Engineering, ETH Zurich, CH-8092 Zurich, Switzerland
| | - Silvan Kaufmann
- Laboratory of Biosensors and Bioelectronics, Institute for Biomedical Engineering, ETH Zurich, CH-8092 Zurich, Switzerland
| | - János Vörös
- Laboratory of Biosensors and Bioelectronics, Institute for Biomedical Engineering, ETH Zurich, CH-8092 Zurich, Switzerland
| | - Marcy Zenobi-Wong
- Cartilage Engineering and Regeneration, Department of Health Sciences and Technology, ETH Zurich, CH-8093 Zurich, Switzerland
| | - László Demkó
- Laboratory of Biosensors and Bioelectronics, Institute for Biomedical Engineering, ETH Zurich, CH-8092 Zurich, Switzerland.
| |
Collapse
|
188
|
Ao X, Liu Y, Bai XY, Qu X, Xu Z, Hu G, Chen M, Wu H. Association between EHBP1 rs721048(A>G) polymorphism and prostate cancer susceptibility: a meta-analysis of 17 studies involving 150,678 subjects. Onco Targets Ther 2015; 8:1671-80. [PMID: 26185455 PMCID: PMC4500625 DOI: 10.2147/ott.s84034] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Background EHBP1 rs721048(A) was first identified as a prostate cancer (PCa) risk in Caucasians by genome-wide association study, but subsequent replication studies involving Caucasian and other ethnicities did not produce consistent results. The aim of this study was to obtain a more definite association between rs721048(A) and PCa risk. Methods We comprehensively searched several databases updated to September 2014, including PubMed, Web of Science, EBSCO, and Google Scholar. Two authors independently screened and reviewed the eligibility of each study. The quality of the included studies was assessed by the Newcastle–Ottawa scale. The association of rs721048(A) and PCa risk was assessed by pooling odds ratios (ORs) with 95% confidence intervals (CIs). Results A total of 17 studies, including 48,135 cases and 102,543 controls, published between 2008 and 2014 were included in the meta-analysis. Overall, the pooled analysis demonstrated that rs721048(A) was significantly associated with the risk of PCa under the allele model (OR=1.14, 95% CI=1.11–1.17, P=0.000). Subgroup analysis based on ethnicity revealed a significant association between rs721048(A) and PCa in Caucasian (OR=1.14, 95% CI=1.11–1.16, P=0.000), African descent (OR=1.11, 95% CI=1.01–1.23, P=0.025), and Asian (OR=1.35, 95% CI=1.12–1.64, P=0.002). Conclusion Our results provided strong evidence that rs721048(A) could be a risk factor for PCa.
Collapse
Affiliation(s)
- Xiang Ao
- Laboratory of Molecular Medicine & Pharmacy, School of Life Science and Biotechnology, Dalian University of Technology, Dalian, People's Republic of China
| | - Ying Liu
- Laboratory of Molecular Medicine & Pharmacy, School of Life Science and Biotechnology, Dalian University of Technology, Dalian, People's Republic of China
| | - Xiao-Yan Bai
- Laboratory of Molecular Medicine & Pharmacy, School of Life Science and Biotechnology, Dalian University of Technology, Dalian, People's Republic of China
| | - Xinjian Qu
- Laboratory of Molecular Medicine & Pharmacy, School of Life Science and Medicine, Dalian University of Technology, Panjin, Liaoning, People's Republic of China
| | - Zhaowei Xu
- Laboratory of Molecular Medicine & Pharmacy, School of Life Science and Biotechnology, Dalian University of Technology, Dalian, People's Republic of China
| | - Gaolei Hu
- Laboratory of Molecular Medicine & Pharmacy, School of Life Science and Biotechnology, Dalian University of Technology, Dalian, People's Republic of China
| | - Min Chen
- Laboratory of Molecular Medicine & Pharmacy, School of Life Science and Biotechnology, Dalian University of Technology, Dalian, People's Republic of China
| | - Huijian Wu
- Laboratory of Molecular Medicine & Pharmacy, School of Life Science and Biotechnology, Dalian University of Technology, Dalian, People's Republic of China ; Laboratory of Molecular Medicine & Pharmacy, School of Life Science and Medicine, Dalian University of Technology, Panjin, Liaoning, People's Republic of China
| |
Collapse
|
189
|
Amin Al Olama A, Benlloch S, Antoniou AC, Giles GG, Severi G, Neal DE, Hamdy FC, Donovan JL, Muir K, Schleutker J, Henderson BE, Haiman CA, Schumacher FR, Pashayan N, Pharoah PDP, Ostrander EA, Stanford JL, Batra J, Clements JA, Chambers SK, Weischer M, Nordestgaard BG, Ingles SA, Sorensen KD, Orntoft TF, Park JY, Cybulski C, Maier C, Doerk T, Dickinson JL, Cannon-Albright L, Brenner H, Rebbeck TR, Zeigler-Johnson C, Habuchi T, Thibodeau SN, Cooney KA, Chappuis PO, Hutter P, Kaneva RP, Foulkes WD, Zeegers MP, Lu YJ, Zhang HW, Stephenson R, Cox A, Southey MC, Spurdle AB, FitzGerald L, Leongamornlert D, Saunders E, Tymrakiewicz M, Guy M, Dadaev T, Little SJ, Govindasami K, Sawyer E, Wilkinson R, Herkommer K, Hopper JL, Lophatonanon A, Rinckleb AE, Kote-Jarai Z, Eeles RA, Easton DF. Risk Analysis of Prostate Cancer in PRACTICAL, a Multinational Consortium, Using 25 Known Prostate Cancer Susceptibility Loci. Cancer Epidemiol Biomarkers Prev 2015; 24:1121-9. [PMID: 25837820 PMCID: PMC4491026 DOI: 10.1158/1055-9965.epi-14-0317] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2014] [Accepted: 03/09/2015] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND 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. METHODS 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. RESULTS 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). CONCLUSIONS 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. IMPACT 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.
Collapse
Affiliation(s)
- Ali Amin Al Olama
- Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, University of Cambridge, Strangeways Research Laboratory, Cambridge, United Kingdom.
| | - Sara Benlloch
- Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, University of Cambridge, Strangeways Research Laboratory, Cambridge, United Kingdom
| | - Antonis C Antoniou
- Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, University of Cambridge, Strangeways Research Laboratory, Cambridge, United Kingdom
| | - Graham G Giles
- Cancer Epidemiology Centre, the Cancer Council Victoria, Carlton, Victoria, Australia. Centre for Molecular, Environmental, Genetic, and Analytic Epidemiology, The University of Melbourne, Melbourne, Victoria, Australia
| | - Gianluca Severi
- Cancer Epidemiology Centre, the Cancer Council Victoria, Carlton, Victoria, Australia
| | - David E Neal
- University of Cambridge, Department of Oncology, Addenbrooke's Hospital, Cambridge, United Kingdom. Cancer Research UK Cambridge Research Institute, Li Ka Shing Centre, Cambridge, United Kingdom
| | - Freddie C Hamdy
- Nuffield Department of Surgical Sciences, University of Oxford, Oxford, United Kingdom. Faculty of Medical Science, University of Oxford, John Radcliffe Hospital, Oxford, United Kingdom
| | - Jenny L Donovan
- School of Social and Community Medicine, University of Bristol, Bristol, United Kingdom
| | - Kenneth Muir
- The University of Manchester, Centre for Epidemiology, Institute of Population Health, Manchester, United Kingdom. University of Warwick, University House, Coventry, United Kingdom
| | - Johanna Schleutker
- Institute of Biomedical Technology/BioMediTech, University of Tampere and FimLab Laboratories, Tampere, Finland. Department of Medical Biochemistry and Genetics, University of Turku, Turku, Finland
| | - Brian E Henderson
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California/Norris Comprehensive Cancer Centre, Los Angeles, California
| | - Christopher A Haiman
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California/Norris Comprehensive Cancer Centre, Los Angeles, California
| | - Fredrick R Schumacher
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California/Norris Comprehensive Cancer Centre, Los Angeles, California
| | - Nora Pashayan
- Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, University of Cambridge, Strangeways Research Laboratory, Cambridge, United Kingdom. University College London, Department of Applied Health Research, London, United Kingdom
| | - Paul D P Pharoah
- Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, University of Cambridge, Strangeways Research Laboratory, Cambridge, United Kingdom
| | | | - Janet L Stanford
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Centre, Seattle, Washington. Department of Epidemiology, School of Public Health, University of Washington, Seattle, Washington
| | - Jyotsna Batra
- Australian Prostate Cancer Research Centre-Qld, Institute of Health and Biomedical Innovation and School of Biomedical Science, Queensland University of Technology, Brisbane, Queensland, Australia
| | - Judith A Clements
- Australian Prostate Cancer Research Centre-Qld, Institute of Health and Biomedical Innovation and School of Biomedical Science, Queensland University of Technology, Brisbane, Queensland, Australia
| | - Suzanne K Chambers
- Griffith Health Institute, Griffith University, Gold Coast, Queensland, Australia. Cancer Council Queensland, Brisbane, Queensland, Australia. Prostate Cancer Foundation of Australia, Sydney, Australia
| | - Maren Weischer
- Department of Clinical Biochemistry, Herlev Hospital, Copenhagen University Hospital, Herlev, Denmark
| | - Børge G Nordestgaard
- Department of Clinical Biochemistry, Herlev Hospital, Copenhagen University Hospital, Herlev, Denmark
| | - Sue A Ingles
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California/Norris Comprehensive Cancer Centre, Los Angeles, California
| | - Karina D Sorensen
- Department of Molecular Medicine (MOMA), Aarhus University Hospital, Aarhus N, Denmark
| | - Torben F Orntoft
- Department of Molecular Medicine (MOMA), Aarhus University Hospital, Aarhus N, Denmark
| | - Jong Y Park
- Division of Cancer Prevention and Control, H. Lee Moffitt Cancer Centre, Tampa, Florida
| | - Cezary Cybulski
- International Hereditary Cancer Centre, Department of Genetics and Pathology, Pomeranian Medical University, Szczecin, Poland
| | | | - Thilo Doerk
- Hannover Biomedical Research School, Hannover, Germany
| | - Joanne L Dickinson
- University of Tasmania, Menzies Research Institute Tasmania, Hobart, Tasmania, Australia
| | - Lisa Cannon-Albright
- Division of Genetic Epidemiology, Department of Medicine, University of Utah School of Medicine, Salt Lake City, Utah. George E. Wahlen Department of Veterans Affairs Medical Centre, Salt Lake City, Utah
| | - Hermann Brenner
- Division of Clinical Epidemiology and Ageing Research, German Cancer Research Centre (DKFZ), Heidelberg, Germany. German Cancer Consortium (DKTK), Heidelberg, Germany
| | | | - Charnita Zeigler-Johnson
- Division of Population Sciences, Department of Medical Oncology, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Tomonori Habuchi
- Department of Urology, Akita University School of Medicine, Akita, Japan
| | | | - Kathleen A Cooney
- Division of Hematology/Oncology, University of Michigan Medical School, Ann Arbor, Michigan
| | - Pierre O Chappuis
- Divisions of Oncology and Genetic Medicine, Geneva University Hospitals, Geneva, Switzerland
| | - Pierre Hutter
- Hopital Cantonal Universitaire de Geneve, Geneva, Switzerland
| | - Radka P Kaneva
- Department of Medical Chemistry and Biochemistry, Molecular Medicine Centre, Medical University-Sofia, Sofia, Bulgaria
| | | | - Maurice P Zeegers
- Department of Complex Genetics, Cluster of Genetics and Cell Biology, NUTRIM School for Nutrition, Toxicology, and Metabolism, Maastricht University Medical Centre+, Maastricht, the Netherlands
| | - Yong-Jie Lu
- Barts Cancer Institute, Queen Mary University of London, John Vane Science Centre, London, United Kingdom
| | - Hong-Wei Zhang
- Second Military Medical University, Shanghai, P.R. China
| | - Robert Stephenson
- Division of Urology, Department of Surgery, University of Utah School of Medicine, Salt Lake City, Utah
| | - Angela Cox
- CR-UK/YCR Sheffield Cancer Research Centre, University of Sheffield, Sheffield, United Kingdom
| | - Melissa C Southey
- Genetic Epidemiology Laboratory, Department of Pathology, The University of Melbourne, Parkville, Victoria, Australia
| | - Amanda B Spurdle
- Molecular Cancer Epidemiology Laboratory, Queensland Institute of Medical Research, Brisbane, Queensland, Australia
| | - Liesel FitzGerald
- Cancer Council Victoria, Cancer Epidemiology Centre, Melbourne, Victoria, Australia
| | | | | | | | - Michelle Guy
- The Institute of Cancer Research, London, United Kingdom
| | - Tokhir Dadaev
- The Institute of Cancer Research, London, United Kingdom
| | - Sarah J Little
- The Institute of Cancer Research, London, United Kingdom
| | | | - Emma Sawyer
- The Institute of Cancer Research, London, United Kingdom
| | | | | | - John L Hopper
- Centre for Molecular, Environmental, Genetic, and Analytic Epidemiology, The University of Melbourne, Melbourne, Victoria, Australia
| | - Aritaya Lophatonanon
- University of Warwick, University House, Coventry, United Kingdom. Institute of Biomedical Technology/BioMediTech, University of Tampere and FimLab Laboratories, Tampere, Finland
| | | | | | | | - Douglas F Easton
- Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, University of Cambridge, Strangeways Research Laboratory, Cambridge, United Kingdom.
| |
Collapse
|
190
|
Li H, Chen H, Liu F, Ren C, Wang S, Bo X, Shu W. Functional annotation of HOT regions in the human genome: implications for human disease and cancer. Sci Rep 2015; 5:11633. [PMID: 26113264 PMCID: PMC4481521 DOI: 10.1038/srep11633] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2015] [Accepted: 06/01/2015] [Indexed: 12/17/2022] Open
Abstract
Advances in genome-wide association studies (GWAS) and large-scale sequencing studies have resulted in an impressive and growing list of disease- and trait-associated genetic variants. Most studies have emphasised the discovery of genetic variation in coding sequences, however, the noncoding regulatory effects responsible for human disease and cancer biology have been substantially understudied. To better characterise the cis-regulatory effects of noncoding variation, we performed a comprehensive analysis of the genetic variants in HOT (high-occupancy target) regions, which are considered to be one of the most intriguing findings of recent large-scale sequencing studies. We observed that GWAS variants that map to HOT regions undergo a substantial net decrease and illustrate development-specific localisation during haematopoiesis. Additionally, genetic risk variants are disproportionally enriched in HOT regions compared with LOT (low-occupancy target) regions in both disease-relevant and cancer cells. Importantly, this enrichment is biased toward disease- or cancer-specific cell types. Furthermore, we observed that cancer cells generally acquire cancer-specific HOT regions at oncogenes through diverse mechanisms of cancer pathogenesis. Collectively, our findings demonstrate the key roles of HOT regions in human disease and cancer and represent a critical step toward further understanding disease biology, diagnosis, and therapy.
Collapse
Affiliation(s)
- Hao Li
- Department of Biotechnology, Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Hebing Chen
- Department of Biotechnology, Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Feng Liu
- Department of Biotechnology, Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Chao Ren
- Department of Biotechnology, Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Shengqi Wang
- Department of Biotechnology, Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Xiaochen Bo
- Department of Biotechnology, Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Wenjie Shu
- Department of Biotechnology, Beijing Institute of Radiation Medicine, Beijing 100850, China
| |
Collapse
|
191
|
Abedinzadeh M, Zare-Shehneh M, Neamatzadeh H, Abedinzadeh M, Karami H. Association between MTHFR C677T Polymorphism and Risk of Prostate Cancer: Evidence from 22 Studies with 10,832 Cases and 11,993 Controls. Asian Pac J Cancer Prev 2015; 16:4525-30. [DOI: 10.7314/apjcp.2015.16.11.4525] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
|
192
|
He Y, Gu J, Strom S, Logothetis CJ, Kim J, Wu X. The prostate cancer susceptibility variant rs2735839 near KLK3 gene is associated with aggressive prostate cancer and can stratify gleason score 7 patients. Clin Cancer Res 2015; 20:5133-5139. [PMID: 25274378 DOI: 10.1158/1078-0432.ccr-14-0661] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE Gleason score (GS) 7 prostate cancer is a heterogeneous disease with different clinical behavior. We sought to identify genetic biomarkers that may predict the aggressiveness of GS 7 diseases. EXPERIMENTAL DESIGN We genotyped 72 prostate cancer susceptibility SNPs identified in genome-wide association studies in 1,827 white men with histologically confirmed prostate adenocarcinoma. SNPs associated with disease aggressiveness were identified by comparing high-aggressive (GS ≥8) and low-aggressive (GS ≤6) cases. The significant SNPs were then tested to see whether they could further stratify GS 7 prostate cancer. RESULTS Three SNPs-rs2735839, rs10486567, and rs103294-were associated with biopsy-proven high-aggressive (GS ≥8) prostate cancer (P < 0.05). Furthermore, the frequency of the variant allele (A) at rs2735839 was significantly higher in patients with biopsy-proven GS 4+3 disease than in those with GS 3 + 4 disease (P = 0.003). In multivariate logistic regression analysis, patients carrying the A allele at rs2735839 exhibited a 1.85-fold (95% confidence interval, 1.31-2.61) increased risk of being GS 4 + 3 compared with those with GS 3 + 4. The rs2735839 is located 600 base pair downstream of the KLK3 gene (encoding PSA) on 19q13.33 and has been shown to modulate PSA level, providing strong biologic plausibility for its association with prostate cancer aggressiveness. CONCLUSIONS We confirmed the association of the rs2735839 with high-aggressive prostate cancer (GS ≥8). Moreover, we reported for the first time that rs2735839 can stratify GS 7 patients, which would be clinically important for more accurately assessing the clinical behavior of the intermediate-grade prostate cancer and for tailoring personalized treatment and posttreatment management.
Collapse
Affiliation(s)
- Yonggang He
- Department of Epidemiology, The University of Texas MD Anderson Cancer Center, Houston, Texas.,Department of Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jian Gu
- Department of Epidemiology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Sara Strom
- Department of Epidemiology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Christopher J Logothetis
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Jeri Kim
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Xifeng Wu
- Department of Epidemiology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| |
Collapse
|
193
|
Zhao CX, Liu M, Xu Y, Yang K, Wei D, Shi XH, Yang F, Zhang YG, Wang X, Liang SY, Zhao F, Zhang YR, Wang NN, Chen X, Sun L, Zhu XQ, Yuan HP, Zhu L, Yang YG, Tang L, Jiao HY, Huo ZH, Wang JY, Yang Z. 8q24 rs4242382 polymorphism is a risk factor for prostate cancer among multi-ethnic populations: evidence from clinical detection in China and a meta-analysis. Asian Pac J Cancer Prev 2015; 15:8311-7. [PMID: 25339022 DOI: 10.7314/apjcp.2014.15.19.8311] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Evidence supporting an association between the 8q24 rs4242382-A polymorphism and prostate cancer (PCa) risk has been reported in North American and Europe populations, though data from Asian populations remain limited. We therefore investigated this association by clinical detection in China, and meta-analysis in Asian, Caucasian and African-American populations. MATERIALS AND METHODS Blood samples and clinical information were collected from ethnically Chinese men from Northern China with histologically- confirmed PCa (n=335) and from age-matched normal controls (n=347). The 8q24 (rs4242382) gene polymorphism was genotyped by polymerase chain reaction-high-resolution melting analysis. We initially analyzed the associations between the risk allele and PCa and clinical covariates. A meta-analysis was then performed using genotyping data from a total of 1,793 PCa cases and 1,864 controls from our study and previously published studies in American and European populations, to determine the association between PCa and risk genotype. RESULTS The incidence of the risk allele was higher in PCa cases than controls (0.222 vs 0.140, P=7.3?10-5), suggesting that the 8q24 rs4242382-A polymorphism was associated with PCa risk in Chinese men. The genotypes in subjects were in accordance with a dominant genetic model (ORadj=2.03, 95%CI: 1.42-2.91, Padj=1.1?10-4). Presence of the risk allele rs4242382-A at 8q24 was also associated with clinical covariates including age at diagnosis ≥65 years, prostate specific antigen >10 ng/ml, Gleason score <8, tumor stage and aggressive PCa, compared with the non-risk genotype (P=4.6?10-5-3.0?10-2). Meta-analysis confirmed the association between 8q24 rs4242382-A polymorphism and PCa risk (OR=1.62, 95%CI: 1.39-1.88, P=1.0?10-5) across Asian, Caucasian and African American populations. CONCLUSIONS The replicated data suggest that the 8q24 rs4242382-A variation might be associated with increased PCa susceptibility in Asian, Caucasian and African American populations. These results imply that this polymorphism may be a useful risk biomarker for PCa in multi-ethnic populations.
Collapse
Affiliation(s)
- Cheng-Xiao Zhao
- The Key Laboratory of Geriatrics, Beijing Hospital and Beijing Institute of Geriatrics, Chinese Ministry of Health, Beijing, China E-mail : ,
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
194
|
Oh JJ, Park S, Lee SE, Hong SK, Lee S, Jo JK, Lee JK, Ho JN, Yoon S, Byun SS. Genome-wide detection of allelic genetic variation to predict advanced-stage prostate cancer after radical prostatectomy using an exome SNP chip. Urol Oncol 2015; 33:385.e7-13. [PMID: 26087972 DOI: 10.1016/j.urolonc.2015.05.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2015] [Revised: 05/11/2015] [Accepted: 05/13/2015] [Indexed: 10/23/2022]
Abstract
OBJECTIVES Genetic variations among patients with prostate cancer (PCa) who underwent radical prostatectomies were evaluated to predict advanced stage above T3 using an exome single nucleotide polymorphism (SNP) chip array. MATERIALS AND METHODS We collected data of genetic SNP variants from 820 patients with PCa who underwent radical prostatectomy (RP) using a custom HumanExome BeadChip v1.0 (Illumina Inc.). We selected the SNPs that were most significantly associated with advanced-stage PCa (≥ T3) among the 242,186 SNPs that were genotyped, and we compared the accuracies of the associations using a multivariate logistic model that incorporated clinical factors and clinicogenetic factors. RESULTS Among the total cohort, 360 patients (43.9%) had advanced pathologic stage (≥ T3) after RP, of whom 262 (32.0%) had extracapsular extensions, 79 (9.6%) had seminal vesicle invasions, and 10 (1.3%) had bladder neck invasions. The exome array analysis indicated that 5 SNPs (rs6804162, rs8055236, rs56335308, rs6104, and rs12618769) were significant for predicting T3 stage after RP in patients with PCa. These genetic markers were significant factors after adjusting for other clinical parameters, and they increased the accuracy of a multivariate model for predicting advanced stage of PCa (83.9%-87.2%, P = 0.0001). CONCLUSIONS Based on a genetic array, the selected SNPs were found to be independent predictors for advanced stage after RP, and the addition of individualized genetic information effectively enhanced the accuracy of predicting advanced-stage disease. These results should be validated in another independent cohort.
Collapse
Affiliation(s)
- Jong Jin Oh
- Department of Urology, Seoul National University Bundang Hospital, Seongnam, Republic of Korea
| | - Seunghyun Park
- Department of Electrical and Computer Engineering, Seoul National University, Seoul, Republic of Korea; School of Electrical Engineering, Korea University, Seoul, Republic of Korea
| | - Sang Eun Lee
- Department of Urology, Seoul National University Bundang Hospital, Seongnam, Republic of Korea
| | - Sung Kyu Hong
- Department of Urology, Seoul National University Bundang Hospital, Seongnam, Republic of Korea
| | - Sangchul Lee
- Department of Urology, Seoul National University Bundang Hospital, Seongnam, Republic of Korea
| | - Jung Ki Jo
- Department of Urology, Seoul National University Bundang Hospital, Seongnam, Republic of Korea
| | - Jung Keun Lee
- Department of Urology, Seoul National University Bundang Hospital, Seongnam, Republic of Korea
| | - Jin-Nyoung Ho
- Department of Urology, Seoul National University Bundang Hospital, Seongnam, Republic of Korea; Biomedical Research Institute, Seoul National University Bundang Hospital, Seongnam, Republic of Korea
| | - Sungroh Yoon
- Department of Electrical and Computer Engineering, Seoul National University, Seoul, Republic of Korea
| | - Seok-Soo Byun
- Department of Urology, Seoul National University Bundang Hospital, Seongnam, Republic of Korea.
| |
Collapse
|
195
|
Yen AMF, Auvinen A, Schleutker J, Wu YY, Fann JCY, Tammela T, Chen SLS, Chiu SYH, Chen HH. Prostate cancer screening using risk stratification based on a multi-state model of genetic variants. Prostate 2015; 75:825-35. [PMID: 25683204 DOI: 10.1002/pros.22964] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2014] [Accepted: 12/22/2014] [Indexed: 11/09/2022]
Abstract
BACKGROUND Risk-stratified screening for prostate cancer (PCa) with prostate-specific antigen (PSA) testing incorporating genetic variants has received some attention but has been scarcely investigated. We developed a model to stratify the Finnish population by different risk profiles related to genetic variants to optimize the screening policy. METHODS Data from the Finnish randomized controlled trial on screening for PCa with PSA testing were used to estimate a six-state Markov model of disease progression. Blood samples from Finnish men were used to assess the risk of PCa related to three genetic variants (rs4242382, rs138213197, and rs200331695). A risk score-based approach combined with a series of computer simulation models was applied to optimize individual screening policies. RESULTS The 10-year risk of having progressive prostate cancer detected ranged from 43% in the top 5% risk group to approximately 11% in the bottom half of the population. Using the median group, with screening every four years beginning at 55 years-old, as the reference group, the recommended age beginning screening was approximately 47 years-old for the top 5% risk group and 55 years-old for those in the lower 60% risk group. The recommended interscreening interval has been shortened for individuals in the high risk group. The increased availability of genomic information allows the proposed multistate model to be more discriminating with respect to risk stratification and the suggested screening policy, particularly for the lowest risk groups-. -- CONCLUSIONS A multi-state genetic variant-based model was developed for further application to population risk stratification to optimize the interscreening interval and the age at which to begin screening for PSA. A small sub-group of the population is likely to benefit from more intensive screening with early start and short interval, while half of the population is unlikely to benefit from such protocol (compared with four-year interval after age 55 years).
Collapse
Affiliation(s)
- Amy Ming-Fang Yen
- School of Oral Hygiene, College of Oral Medicine, Taipei Medical University, Taipei, Taiwan
| | | | | | | | | | | | | | | | | |
Collapse
|
196
|
Laitinen VH, Rantapero T, Fischer D, Vuorinen EM, Tammela TL, Wahlfors T, Schleutker J. Fine-mapping the 2q37 and 17q11.2-q22 loci for novel genes and sequence variants associated with a genetic predisposition to prostate cancer. Int J Cancer 2015; 136:2316-27. [PMID: 25335771 PMCID: PMC4355047 DOI: 10.1002/ijc.29276] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2014] [Accepted: 10/01/2014] [Indexed: 01/13/2023]
Abstract
The 2q37 and 17q12-q22 loci are linked to an increased prostate cancer (PrCa) risk. No candidate gene has been localized at 2q37 and the HOXB13 variant G84E only partially explains the linkage to 17q21-q22 observed in Finland. We screened these regions by targeted DNA sequencing to search for cancer-associated variants. Altogether, four novel susceptibility alleles were identified. Two ZNF652 (17q21.3) variants, rs116890317 and rs79670217, increased the risk of both sporadic and hereditary PrCa (rs116890317: OR = 3.3-7.8, p = 0.003-3.3 × 10(-5) ; rs79670217: OR = 1.6-1.9, p = 0.002-0.009). The HDAC4 (2q37.2) variant rs73000144 (OR = 14.6, p = 0.018) and the EFCAB13 (17q21.3) variant rs118004742 (OR = 1.8, p = 0.048) were overrepresented in patients with familial PrCa. To map the variants within 2q37 and 17q11.2-q22 that may regulate PrCa-associated genes, we combined DNA sequencing results with transcriptome data obtained by RNA sequencing. This expression quantitative trait locus (eQTL) analysis identified 272 single-nucleotide polymorphisms (SNPs) possibly regulating six genes that were differentially expressed between cases and controls. In a modified approach, prefiltered PrCa-associated SNPs were exploited and interestingly, a novel eQTL targeting ZNF652 was identified. The novel variants identified in this study could be utilized for PrCa risk assessment, and they further validate the suggested role of ZNF652 as a PrCa candidate gene. The regulatory regions discovered by eQTL mapping increase our understanding of the relationship between regulation of gene expression and susceptibility to PrCa and provide a valuable starting point for future functional research.
Collapse
Affiliation(s)
- Virpi H. Laitinen
- BioMediTech, University of Tampere and Fimlab Laboratories, FI-33520 Tampere, Finland
| | - Tommi Rantapero
- BioMediTech, University of Tampere and Fimlab Laboratories, FI-33520 Tampere, Finland
| | - Daniel Fischer
- School of Health Sciences, University of Tampere, FI-33014 Tampere, Finland
| | - Elisa M. Vuorinen
- BioMediTech, University of Tampere and Fimlab Laboratories, FI-33520 Tampere, Finland
| | - Teuvo L.J. Tammela
- Department of Urology, Tampere University Hospital and Medical School, University of Tampere, FI-33520 Tampere, Finland
| | | | - Tiina Wahlfors
- BioMediTech, University of Tampere and Fimlab Laboratories, FI-33520 Tampere, Finland
| | - Johanna Schleutker
- BioMediTech, University of Tampere and Fimlab Laboratories, FI-33520 Tampere, Finland
- Medical Biochemistry and Genetics, Institute of Biomedicine, FI-20014 University of Turku, Turku, Finland
| |
Collapse
|
197
|
Braun K, Sjoberg DD, Vickers AJ, Lilja H, Bjartell AS. A Four-kallikrein Panel Predicts High-grade Cancer on Biopsy: Independent Validation in a Community Cohort. Eur Urol 2015; 69:505-11. [PMID: 25979570 DOI: 10.1016/j.eururo.2015.04.028] [Citation(s) in RCA: 67] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2015] [Accepted: 04/18/2015] [Indexed: 11/17/2022]
Abstract
BACKGROUND A statistical model based on four kallikrein markers (total prostate-specific antigen [tPSA], free PSA [fPSA], intact PSA, and human kallikrein-related peptidase 2) in blood can predict risk of Gleason score ≥7 (high-grade) cancer at prostate biopsy. OBJECTIVE To determine the value of this model in predicting high-grade cancer at biopsy in a community-based setting in which referral criteria included percentage of fPSA to tPSA (%fPSA). DESIGN, SETTING, AND PARTICIPANTS We evaluated the model, with or without adding blood levels of microseminoprotein-β (MSMB) in a cohort of 749 men referred for prostate biopsy due to elevated PSA (≥3 ng/ml), low %fPSA (<20%), or suspicious digital rectal examination at Skåne University Hospital, Malmö, Sweden. OUTCOME MEASUREMENTS AND STATISTICAL ANALYSIS The kallikrein markers, with or without MSMB levels, measured in cryopreserved anticoagulated blood were combined with age in a published statistical model (Prostate Testing for Cancer and Treatment [ProtecT]) to predict high-grade cancer at biopsy. Predictive accuracy was compared with a base model. RESULTS AND LIMITATIONS The %fPSA was low (median: 17; interquartile range: 13-22) in this cohort because this marker was used as a referral criterion. The ProtecT model improved discrimination over age and PSA for high-grade cancer (0.777 vs 0.720; p=0.002). At one illustrative cut point, use of the panel would reduce the number of biopsies by 236 per 1000 and detect 195 of 208 (94%) but delay diagnosis of 13 of 208 high-grade cancers. MSMB levels in blood did not improve the accuracy of the panel (p=0.2). CONCLUSIONS The kallikrein model is predictive of high-grade cancer if criteria for biopsy referral also include %fPSA, and it can reduce unnecessary biopsies without missing an undue number of tumors. PATIENT SUMMARY We evaluated a published model to predict biopsy outcome in men biopsied due to low percentage of free to total prostate-specific antigen. The model helps reduce unnecessary biopsies without missing an undue number of high-grade cancers.
Collapse
Affiliation(s)
- Katharina Braun
- Department of Urology, University Hospital Ruhr-University Bochum, Marien Hospital Herne, Herne, Germany
| | - Daniel D Sjoberg
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Andrew J Vickers
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Hans Lilja
- Departments of Laboratory Medicine, Medicine (Genitourinary Oncology), and Surgery (Urology), Memorial Sloan Kettering Cancer Center, New York, NY, USA; Nuffield Department of Surgical Sciences, University of Oxford, Oxford, UK; Department of Translational Medicine, Lund University, Malmö, Sweden.
| | - Anders S Bjartell
- Department of Translational Medicine, Lund University, Malmö, Sweden
| |
Collapse
|
198
|
Berndt SI, Wang Z, Yeager M, Alavanja MC, Albanes D, Amundadottir L, Andriole G, Beane Freeman L, Campa D, Cancel-Tassin G, Canzian F, Cornu JN, Cussenot O, Diver WR, Gapstur SM, Grönberg H, Haiman CA, Henderson B, Hutchinson A, Hunter DJ, Key TJ, Kolb S, Koutros S, Kraft P, Le Marchand L, Lindström S, Machiela MJ, Ostrander EA, Riboli E, Schumacher F, Siddiq A, Stanford JL, Stevens VL, Travis RC, Tsilidis KK, Virtamo J, Weinstein S, Wilkund F, Xu J, Lilly Zheng S, Yu K, Wheeler W, Zhang H, Sampson J, Black A, Jacobs K, Hoover RN, Tucker M, Chanock SJ. Two susceptibility loci identified for prostate cancer aggressiveness. Nat Commun 2015; 6:6889. [PMID: 25939597 PMCID: PMC4422072 DOI: 10.1038/ncomms7889] [Citation(s) in RCA: 81] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2014] [Accepted: 03/10/2015] [Indexed: 01/06/2023] Open
Abstract
Most men diagnosed with prostate cancer will experience indolent disease; hence, discovering genetic variants that distinguish aggressive from nonaggressive prostate cancer is of critical clinical importance for disease prevention and treatment. In a multistage, case-only genome-wide association study of 12,518 prostate cancer cases, we identify two loci associated with Gleason score, a pathological measure of disease aggressiveness: rs35148638 at 5q14.3 (RASA1, P=6.49 × 10(-9)) and rs78943174 at 3q26.31 (NAALADL2, P=4.18 × 10(-8)). In a stratified case-control analysis, the SNP at 5q14.3 appears specific for aggressive prostate cancer (P=8.85 × 10(-5)) with no association for nonaggressive prostate cancer compared with controls (P=0.57). The proximity of these loci to genes involved in vascular disease suggests potential biological mechanisms worthy of further investigation.
Collapse
Affiliation(s)
- Sonja I Berndt
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, Maryland 20892, USA
| | - Zhaoming Wang
- 1] Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, Maryland 20892, USA [2] Cancer Genomics Research Laboratory, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Leidos Biomedical Research Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland 21701, USA
| | - Meredith Yeager
- 1] Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, Maryland 20892, USA [2] Cancer Genomics Research Laboratory, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Leidos Biomedical Research Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland 21701, USA
| | - Michael C Alavanja
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, Maryland 20892, USA
| | - Demetrius Albanes
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, Maryland 20892, USA
| | - Laufey Amundadottir
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, Maryland 20892, USA
| | - Gerald Andriole
- Division of Urologic Surgery, Washington University School of Medicine, St Louis, Missouri 63110, USA
| | - Laura Beane Freeman
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, Maryland 20892, USA
| | - Daniele Campa
- Division of Cancer Epidemiology, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | | | - Federico Canzian
- Genomic Epidemiology Group, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Jean-Nicolas Cornu
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, Maryland 20892, USA
| | - Olivier Cussenot
- CeRePP, Assistance Publique-Hôpitaux de Paris, UPMC University Paris 6, Paris, France
| | - W Ryan Diver
- Epidemiology Research Program, American Cancer Society, Atlanta, Georgia 30303, USA
| | - Susan M Gapstur
- Epidemiology Research Program, American Cancer Society, Atlanta, Georgia 30303, USA
| | - Henrik Grönberg
- Department of Medical Epidemiology and Biostatistics, Karolinska Institute, Stockholm 17177, Sweden
| | - Christopher A Haiman
- Department of Preventative Medicine, Keck School of Medicine, University of Southern California/Norris Comprehensive Cancer Center, Los Angeles, California 90033, USA
| | - Brian Henderson
- Department of Preventative Medicine, Keck School of Medicine, University of Southern California/Norris Comprehensive Cancer Center, Los Angeles, California 90033, USA
| | - Amy Hutchinson
- Cancer Genomics Research Laboratory, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Leidos Biomedical Research Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland 21701, USA
| | - David J Hunter
- Department of Epidemiology, Harvard School of Public Health, Boston, Massachusetts 02115, USA
| | - Timothy J Key
- Cancer Epidemiology Unit, Nuffield Department of Clinical Medicine, University of Oxford, Oxford OX3 7BN, UK
| | - Suzanne Kolb
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, Washington 98109, USA
| | - Stella Koutros
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, Maryland 20892, USA
| | - Peter Kraft
- Department of Epidemiology, Harvard School of Public Health, Boston, Massachusetts 02115, USA
| | - Loic Le Marchand
- Epidemiology Program, University of Hawaii Cancer Center, Honolulu, Hawaii 96813, USA
| | - Sara Lindström
- Department of Epidemiology, Harvard School of Public Health, Boston, Massachusetts 02115, USA
| | - Mitchell J Machiela
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, Maryland 20892, USA
| | - Elaine A Ostrander
- National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland 20892, USA
| | - Elio Riboli
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College, London SW7 2AZ, UK
| | - Fred Schumacher
- Department of Preventative Medicine, Keck School of Medicine, University of Southern California/Norris Comprehensive Cancer Center, Los Angeles, California 90033, USA
| | - Afshan Siddiq
- Department of Genomics of Common Disease, School of Public Health, Imperial College London, London SW7 2AZ, UK
| | - Janet L Stanford
- 1] Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, Washington 98109, USA [2] Department of Epidemiology, School of Public Health, University of Washington, Seattle, Washington 98195, USA
| | - Victoria L Stevens
- Epidemiology Research Program, American Cancer Society, Atlanta, Georgia 30303, USA
| | - Ruth C Travis
- Cancer Epidemiology Unit, Nuffield Department of Clinical Medicine, University of Oxford, Oxford OX3 7BN, UK
| | - Konstantinos K Tsilidis
- Department of Hygiene and Epidemiology, University of Ioannina School of Medicine, Ioannina 45110, Greece
| | - Jarmo Virtamo
- Department of Chronic Disease Prevention, National Institute for Health and Welfare, FI-00271 Helsinki, Finland
| | - Stephanie Weinstein
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, Maryland 20892, USA
| | - Fredrik Wilkund
- Department of Medical Epidemiology and Biostatistics, Karolinska Institute, Stockholm 17177, Sweden
| | - Jianfeng Xu
- Center for Cancer Genomics, Wake Forest University School of Medicine, Winston-Salem, North Carolina 27157, USA
| | - S Lilly Zheng
- Center for Cancer Genomics, Wake Forest University School of Medicine, Winston-Salem, North Carolina 27157, USA
| | - Kai Yu
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, Maryland 20892, USA
| | - William Wheeler
- Information Management Services Inc., Rockville, Maryland 20852, USA
| | - Han Zhang
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, Maryland 20892, USA
| | - Joshua Sampson
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, Maryland 20892, USA
| | - Amanda Black
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, Maryland 20892, USA
| | - Kevin Jacobs
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, Maryland 20892, USA
| | - Robert N Hoover
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, Maryland 20892, USA
| | - Margaret Tucker
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, Maryland 20892, USA
| | - Stephen J Chanock
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, Maryland 20892, USA
| |
Collapse
|
199
|
Panagiotou OA, Travis RC, Campa D, Berndt SI, Lindstrom S, Kraft P, Schumacher FR, Siddiq A, Papatheodorou SI, Stanford JL, Albanes D, Virtamo J, Weinstein SJ, Diver WR, Gapstur SM, Stevens VL, Boeing H, Bueno-de-Mesquita HB, Barricarte Gurrea A, Kaaks R, Khaw KT, Krogh V, Overvad K, Riboli E, Trichopoulos D, Giovannucci E, Stampfer M, Haiman C, Henderson B, Le Marchand L, Gaziano JM, Hunter DJ, Koutros S, Yeager M, Hoover RN, Chanock SJ, Wacholder S, Key TJ, Tsilidis KK. A genome-wide pleiotropy scan for prostate cancer risk. Eur Urol 2015; 67:649-57. [PMID: 25277271 PMCID: PMC4359641 DOI: 10.1016/j.eururo.2014.09.020] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2014] [Accepted: 09/13/2014] [Indexed: 01/17/2023]
Abstract
BACKGROUND No single-nucleotide polymorphisms (SNPs) specific for aggressive prostate cancer have been identified in genome-wide association studies (GWAS). OBJECTIVE To test if SNPs associated with other traits may also affect the risk of aggressive prostate cancer. DESIGN, SETTING, AND PARTICIPANTS SNPs implicated in any phenotype other than prostate cancer (p≤10(-7)) were identified through the catalog of published GWAS and tested in 2891 aggressive prostate cancer cases and 4592 controls from the Breast and Prostate Cancer Cohort Consortium (BPC3). The 40 most significant SNPs were followed up in 4872 aggressive prostate cancer cases and 24,534 controls from the Prostate Cancer Association Group to Investigate Cancer Associated Alterations in the Genome (PRACTICAL) consortium. OUTCOME MEASUREMENTS AND STATISTICAL ANALYSIS Odds ratios (ORs) and 95% confidence intervals (CIs) for aggressive prostate cancer were estimated. RESULTS AND LIMITATIONS A total of 4666 SNPs were evaluated by the BPC3. Two signals were seen in regions already reported for prostate cancer risk. rs7014346 at 8q24.21 was marginally associated with aggressive prostate cancer in the BPC3 trial (p=1.6×10(-6)), whereas after meta-analysis by PRACTICAL the summary OR was 1.21 (95% CI 1.16-1.27; p=3.22×10(-18)). rs9900242 at 17q24.3 was also marginally associated with aggressive disease in the meta-analysis (OR 0.90, 95% CI 0.86-0.94; p=2.5×10(-6)). Neither of these SNPs remained statistically significant when conditioning on correlated known prostate cancer SNPs. The meta-analysis by BPC3 and PRACTICAL identified a third promising signal, marked by rs16844874 at 2q34, independent of known prostate cancer loci (OR 1.12, 95% CI 1.06-1.19; p=4.67×10(-5)); it has been shown that SNPs correlated with this signal affect glycine concentrations. The main limitation is the heterogeneity in the definition of aggressive prostate cancer between BPC3 and PRACTICAL. CONCLUSIONS We did not identify new SNPs for aggressive prostate cancer. However, rs16844874 may provide preliminary genetic evidence on the role of the glycine pathway in prostate cancer etiology. PATIENT SUMMARY We evaluated whether genetic variants associated with several traits are linked to the risk of aggressive prostate cancer. No new such variants were identified.
Collapse
Affiliation(s)
- Orestis A Panagiotou
- Division of Cancer Epidemiology & Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Ruth C Travis
- Cancer Epidemiology Unit, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
| | - Daniele Campa
- Division of Cancer Epidemiology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Sonja I Berndt
- Division of Cancer Epidemiology & Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Sara Lindstrom
- Department of Epidemiology, Harvard School of Public Health, Harvard University, Boston, MA, USA
| | - Peter Kraft
- Department of Epidemiology, Harvard School of Public Health, Harvard University, Boston, MA, USA
| | - Fredrick R Schumacher
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Afshan Siddiq
- Department of Genomics of Common Disease, School of Public Health, Imperial College London, London, UK
| | - Stefania I Papatheodorou
- Cyprus International Institute for Environmental and Public Health, Cyprus University of Technology, Limassol, Cyprus
| | - Janet L Stanford
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Demetrius Albanes
- Division of Cancer Epidemiology & Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Jarmo Virtamo
- Department of Chronic Disease Prevention, National Institute for Health and Welfare, Helsinki, Finland
| | - Stephanie J Weinstein
- Division of Cancer Epidemiology & Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - W Ryan Diver
- Epidemiology Research Program, American Cancer Society, Atlanta, GA, USA
| | - Susan M Gapstur
- Epidemiology Research Program, American Cancer Society, Atlanta, GA, USA
| | - Victoria L Stevens
- Epidemiology Research Program, American Cancer Society, Atlanta, GA, USA
| | - Heiner Boeing
- Department of Epidemiology, German Institute of Human Nutrition Potsdam-Rehbruecke, Nuthetal, Germany
| | - H Bas Bueno-de-Mesquita
- Department of Epidemiology and Biostatistics, Imperial College School of Public Health, London, UK; Department for Determinants of Chronic Diseases (DCD), National Institute for Public Health and the Environment (RIVM), Bilthoven, Netherlands; Department of Gastroenterology and Hepatology, University Medical Centre, Utrecht, Netherlands; Department of Social and Preventive Medicine, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Aurelio Barricarte Gurrea
- Navarre Public Health Institute, Pamplona, Spain; Consortium for Biomedical Research in Epidemiology and Public Health, Madrid, Spain
| | - Rudolf Kaaks
- Division of Cancer Epidemiology, German Cancer Research Center, Heidelberg, Germany
| | - Kay-Tee Khaw
- Clinical Gerontology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
| | - Vittorio Krogh
- Epidemiology and Prevention Unit, Department of Preventive & Predictive Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Kim Overvad
- Department of Public Health, Section for Epidemiology, Aarhus University, Aarhus, Denmark
| | - Elio Riboli
- Department of Epidemiology and Biostatistics, Imperial College School of Public Health, London, UK
| | - Dimitrios Trichopoulos
- Department of Epidemiology, Harvard School of Public Health, Harvard University, Boston, MA, USA; Hellenic Health Foundation, Athens, Greece; Bureau of Epidemiologic Research, Academy of Athens, Athens, Greece; Department of Nutrition, Harvard School of Public Health, Boston, MA, USA
| | - Edward Giovannucci
- Department of Epidemiology, Harvard School of Public Health, Harvard University, Boston, MA, USA; Department of Nutrition, Harvard School of Public Health, Boston, MA, USA; Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Meir Stampfer
- Department of Epidemiology, Harvard School of Public Health, Harvard University, Boston, MA, USA
| | - Christopher Haiman
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Brian Henderson
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | | | - J Michael Gaziano
- Department of Epidemiology, Harvard School of Public Health, Harvard University, Boston, MA, USA; Department of Medicine, Harvard Medical School, Boston, MA, USA; Division of Aging, Brigham and Women's Hospital, Boston, MA, USA
| | - David J Hunter
- Department of Epidemiology, Harvard School of Public Health, Harvard University, Boston, MA, USA; Department of Nutrition, Harvard School of Public Health, Boston, MA, USA
| | - Stella Koutros
- Division of Cancer Epidemiology & Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Meredith Yeager
- Core Genotyping Facility Frederick National Laboratory for Cancer Research, Gaithersburg, MD, USA
| | - Robert N Hoover
- Division of Cancer Epidemiology & Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Stephen J Chanock
- Division of Cancer Epidemiology & Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA; Core Genotyping Facility Frederick National Laboratory for Cancer Research, Gaithersburg, MD, USA
| | - Sholom Wacholder
- Division of Cancer Epidemiology & Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Timothy J Key
- Cancer Epidemiology Unit, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
| | - Konstantinos K Tsilidis
- Cancer Epidemiology Unit, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK; Department of Hygiene and Epidemiology, University of Ioannina, School of Medicine, Ioannina, Greece.
| |
Collapse
|
200
|
Cremers RG, Galesloot TE, Aben KK, van Oort IM, Vasen HF, Vermeulen SH, Kiemeney LA. Known susceptibility SNPs for sporadic prostate cancer show a similar association with "hereditary" prostate cancer. Prostate 2015; 75:474-83. [PMID: 25560306 PMCID: PMC6680338 DOI: 10.1002/pros.22933] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/03/2014] [Accepted: 10/23/2014] [Indexed: 11/25/2022]
Abstract
BACKGROUND More than 70 single nucleotide polymorphisms (SNPs) have been reported to be associated with prostate cancer (PC) risk; these were mainly identified in the general population with predominantly sporadic PC (SPC). Previous studies have suggested similar associations between a selection of these SNPs and hereditary PC (HPC). Our aim was to evaluate the effect of all known PC risk SNPs and their discriminative value for SPC and HPC. METHODS Seventy-four PC susceptibility SNPs (reported in literature up to June 2014) were genotyped in a population-based series of 620 SPC patients, 312 HPC patients from the national Dutch registry and 1819 population-based referents. Association analyses were performed using logistic regression, focusing on directional consistency of the odds ratios (ORs) with those in the original reports, that is, whether the OR was in the same direction as in the original report. Discriminative performance was evaluated by a genetic risk score used in logistic regression and receiver operating characteristic (ROC) curve analyses. RESULTS Directional consistency was seen for 62 SNPs in SPC and 64 SNPs in HPC, 56 of which overlapped. ORs were mostly higher for HPC with 22 ORs >1.25 versus 5 for SPC. Discriminative performance was better for HPC with an area under the ROC curve of 0.73 versus 0.64 for SPC. CONCLUSIONS A large overlap was found for the associations between low-penetrance susceptibility SNPs and SPC and HPC, suggesting a similarity in genetic etiology. This warrants a reconsideration of "HPC" and a restrictive policy toward prostate-specific antigen testing in men with a positive family history. Genetic risk scores might be used for PC risk stratification on the population level.
Collapse
Affiliation(s)
- Ruben G. Cremers
- Department for Health EvidenceRadboud university medical centerNijmegenThe Netherlands
- Department of UrologyRadboud university medical centerNijmegenThe Netherlands
- The Netherlands Foundation for the Detection of Hereditary TumoursLeidenThe Netherlands
| | - Tessel E. Galesloot
- Department for Health EvidenceRadboud university medical centerNijmegenThe Netherlands
| | - Katja K. Aben
- Department for Health EvidenceRadboud university medical centerNijmegenThe Netherlands
- Comprehensive Cancer Centre the NetherlandsUtrechtThe Netherlands
| | - Inge M. van Oort
- Department of UrologyRadboud university medical centerNijmegenThe Netherlands
| | - Hans F. Vasen
- The Netherlands Foundation for the Detection of Hereditary TumoursLeidenThe Netherlands
| | - Sita H. Vermeulen
- Department for Health EvidenceRadboud university medical centerNijmegenThe Netherlands
| | - Lambertus A. Kiemeney
- Department for Health EvidenceRadboud university medical centerNijmegenThe Netherlands
- Department of UrologyRadboud university medical centerNijmegenThe Netherlands
| |
Collapse
|