1
|
Marthick JR, Raspin K, Foley GR, Blackburn NB, Banks A, Donovan S, Malley RC, Field MA, Stanford JL, Ostrander EA, FitzGerald LM, Dickinson JL. Massively parallel sequencing in hereditary prostate cancer families reveals a rare risk variant in the DNA repair gene, RAD51C. Eur J Cancer 2021; 159:52-55. [PMID: 34736042 DOI: 10.1016/j.ejca.2021.09.038] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Accepted: 09/25/2021] [Indexed: 11/28/2022]
Affiliation(s)
- James R Marthick
- Menzies Institute for Medical Research, University of Tasmania, Hobart, TAS, 7000, Australia
| | - Kelsie Raspin
- Menzies Institute for Medical Research, University of Tasmania, Hobart, TAS, 7000, Australia
| | - Georgea R Foley
- Menzies Institute for Medical Research, University of Tasmania, Hobart, TAS, 7000, Australia
| | - Nicholas B Blackburn
- Menzies Institute for Medical Research, University of Tasmania, Hobart, TAS, 7000, Australia
| | - Annette Banks
- Menzies Institute for Medical Research, University of Tasmania, Hobart, TAS, 7000, Australia
| | | | - Roslyn C Malley
- Hobart Pathology, Hobart, TAS, 7000, Australia; School of Medicine, University of Tasmania, Hobart, TAS, 7000, Australia
| | - Matthew A Field
- Centre for Tropical Bioinformatics and Molecular Biology and Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, QLD, 4878, Australia; Genome Informatics, John Curtin School of Medical Research, Australian National University, Canberra
| | - Janet L Stanford
- Fred Hutchinson Cancer Research Center, 1100 Fairview Ave. N., M4-B874, Seattle, WA 98109-1024, USA
| | - Elaine A Ostrander
- Cancer Genetics and Comparative Genomics Branch, National Human Genome Research Institute, NIH, Bethesda, MD 20892, USA
| | - Liesel M FitzGerald
- Menzies Institute for Medical Research, University of Tasmania, Hobart, TAS, 7000, Australia
| | - Joanne L Dickinson
- Menzies Institute for Medical Research, University of Tasmania, Hobart, TAS, 7000, Australia.
| |
Collapse
|
2
|
Saunders EJ, Kote-Jarai Z, Eeles RA. Identification of Germline Genetic Variants that Increase Prostate Cancer Risk and Influence Development of Aggressive Disease. Cancers (Basel) 2021; 13:760. [PMID: 33673083 PMCID: PMC7917798 DOI: 10.3390/cancers13040760] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 02/08/2021] [Accepted: 02/09/2021] [Indexed: 12/15/2022] Open
Abstract
Prostate cancer (PrCa) is a heterogeneous disease, which presents in individual patients across a diverse phenotypic spectrum ranging from indolent to fatal forms. No robust biomarkers are currently available to enable routine screening for PrCa or to distinguish clinically significant forms, therefore late stage identification of advanced disease and overdiagnosis plus overtreatment of insignificant disease both remain areas of concern in healthcare provision. PrCa has a substantial heritable component, and technological advances since the completion of the Human Genome Project have facilitated improved identification of inherited genetic factors influencing susceptibility to development of the disease within families and populations. These genetic markers hold promise to enable improved understanding of the biological mechanisms underpinning PrCa development, facilitate genetically informed PrCa screening programmes and guide appropriate treatment provision. However, insight remains largely lacking regarding many aspects of their manifestation; especially in relation to genes associated with aggressive phenotypes, risk factors in non-European populations and appropriate approaches to enable accurate stratification of higher and lower risk individuals. This review discusses the methodology used in the elucidation of genetic loci, genes and individual causal variants responsible for modulating PrCa susceptibility; the current state of understanding of the allelic spectrum contributing to PrCa risk; and prospective future translational applications of these discoveries in the developing eras of genomics and personalised medicine.
Collapse
Affiliation(s)
- Edward J. Saunders
- The Institute of Cancer Research, London SM2 5NG, UK; (Z.K.-J.); (R.A.E.)
| | - Zsofia Kote-Jarai
- The Institute of Cancer Research, London SM2 5NG, UK; (Z.K.-J.); (R.A.E.)
| | - Rosalind A. Eeles
- The Institute of Cancer Research, London SM2 5NG, UK; (Z.K.-J.); (R.A.E.)
- Royal Marsden NHS Foundation Trust, London SW3 6JJ, UK
| |
Collapse
|
3
|
Guan X, Sun D, Lu E, Urrutia JA, Reiter RE, Rettig M, Evans CP, Lara P, Gleave M, Beer TM, Thomas GV, Huang J, Aggarwal RR, Quigley DA, Foye A, Chen WS, Youngren J, Weinstein AS, Stuart JM, Feng FY, Small EJ, Xia Z, Alumkal JJ. Copy Number Loss of 17q22 Is Associated with Enzalutamide Resistance and Poor Prognosis in Metastatic Castration-Resistant Prostate Cancer. Clin Cancer Res 2020; 26:4616-4624. [PMID: 32727885 PMCID: PMC7484240 DOI: 10.1158/1078-0432.ccr-19-2303] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Revised: 05/24/2020] [Accepted: 06/29/2020] [Indexed: 12/22/2022]
Abstract
PURPOSE The purpose of this study was to measure genomic changes that emerge with enzalutamide treatment using analyses of whole-genome sequencing and RNA sequencing. EXPERIMENTAL DESIGN One hundred and one tumors from men with metastatic castration-resistant prostate cancer (mCRPC) who had not been treated with enzalutamide (n = 64) or who had enzalutamide-resistant mCRPC (n = 37) underwent whole genome sequencing. Ninety-nine of these tumors also underwent RNA sequencing. We analyzed the genomes and transcriptomes of these mCRPC tumors. RESULTS Copy number loss was more common than gain in enzalutamide-resistant tumors. Specially, we identified 124 protein-coding genes that were more commonly lost in enzalutamide-resistant samples. These 124 genes included eight putative tumor suppressors located at nine distinct genomic regions. We demonstrated that focal deletion of the 17q22 locus that includes RNF43 and SRSF1 was not present in any patient with enzalutamide-naïve mCRPC but was present in 16% (6/37) of patients with enzalutamide-resistant mCRPC. 17q22 loss was associated with lower RNF43 and SRSF1 expression and poor overall survival from time of biopsy [median overall survival of 19.3 months in 17q22 intact vs. 8.9 months in 17q22 loss, HR, 3.44 95% confidence interval (CI), 1.338-8.867, log-rank P = 0.006]. Finally, 17q22 loss was linked with activation of several targetable factors, including CDK1/2, Akt, and PLK1, demonstrating the potential therapeutic relevance of 17q22 loss in mCRPC. CONCLUSIONS Copy number loss is common in enzalutamide-resistant tumors. Focal deletion of chromosome 17q22 defines a previously unappreciated molecular subset of enzalutamide-resistant mCRPC associated with poor clinical outcome.
Collapse
Affiliation(s)
- Xiangnan Guan
- Knight Cancer Institute, Oregon Health & Science University, Portland, Oregon
| | - Duanchen Sun
- Knight Cancer Institute, Oregon Health & Science University, Portland, Oregon
| | - Eric Lu
- Knight Cancer Institute, Oregon Health & Science University, Portland, Oregon
| | - Joshua A Urrutia
- Knight Cancer Institute, Oregon Health & Science University, Portland, Oregon
| | - Robert Evan Reiter
- Institute of Urologic Oncology, University of California Los Angeles, Los Angeles, California
- Jonsson Comprehensive Cancer Center, Department of Urology, University of California Los Angeles, Los Angeles, California
| | - Matthew Rettig
- Jonsson Comprehensive Cancer Center, Department of Urology, University of California Los Angeles, Los Angeles, California
- VA Greater Los Angeles, Department of Medicine, Los Angeles, California
| | - Christopher P Evans
- University of California Davis Comprehensive Cancer Center, Sacramento, California
| | - Primo Lara
- University of California Davis Comprehensive Cancer Center, Sacramento, California
| | - Martin Gleave
- Vancouver Prostate Centre, University of British Columbia, Vancouver, British Columbia, Canada
| | - Tomasz M Beer
- Knight Cancer Institute, Oregon Health & Science University, Portland, Oregon
| | - George V Thomas
- Knight Cancer Institute, Oregon Health & Science University, Portland, Oregon
| | - Jiaoti Huang
- Duke University School of Medicine, Durham, North Carolina
| | - Rahul R Aggarwal
- University of California San Francisco, San Francisco, California
| | - David A Quigley
- University of California San Francisco, San Francisco, California
| | - Adam Foye
- University of California San Francisco, San Francisco, California
| | - William S Chen
- University of California San Francisco, San Francisco, California
| | - Jack Youngren
- University of California San Francisco, San Francisco, California
| | | | | | - Felix Y Feng
- University of California San Francisco, San Francisco, California
| | - Eric J Small
- University of California San Francisco, San Francisco, California
| | - Zheng Xia
- Knight Cancer Institute, Oregon Health & Science University, Portland, Oregon.
| | - Joshi J Alumkal
- Knight Cancer Institute, Oregon Health & Science University, Portland, Oregon.
- University of Michigan Rogel Cancer Center, Ann Arbor, Michigan
| |
Collapse
|
4
|
Paulo P, Maia S, Pinto C, Pinto P, Monteiro A, Peixoto A, Teixeira MR. Targeted next generation sequencing identifies functionally deleterious germline mutations in novel genes in early-onset/familial prostate cancer. PLoS Genet 2018; 14:e1007355. [PMID: 29659569 PMCID: PMC5919682 DOI: 10.1371/journal.pgen.1007355] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2017] [Revised: 04/26/2018] [Accepted: 04/05/2018] [Indexed: 12/23/2022] Open
Abstract
Considering that mutations in known prostate cancer (PrCa) predisposition genes, including those responsible for hereditary breast/ovarian cancer and Lynch syndromes, explain less than 5% of early-onset/familial PrCa, we have sequenced 94 genes associated with cancer predisposition using next generation sequencing (NGS) in a series of 121 PrCa patients. We found monoallelic truncating/functionally deleterious mutations in seven genes, including ATM and CHEK2, which have previously been associated with PrCa predisposition, and five new candidate PrCa associated genes involved in cancer predisposing recessive disorders, namely RAD51C, FANCD2, FANCI, CEP57 and RECQL4. Furthermore, using in silico pathogenicity prediction of missense variants among 18 genes associated with breast/ovarian cancer and/or Lynch syndrome, followed by KASP genotyping in 710 healthy controls, we identified "likely pathogenic" missense variants in ATM, BRIP1, CHEK2 and TP53. In conclusion, this study has identified putative PrCa predisposing germline mutations in 14.9% of early-onset/familial PrCa patients. Further data will be necessary to confirm the genetic heterogeneity of inherited PrCa predisposition hinted in this study.
Collapse
Affiliation(s)
- Paula Paulo
- Cancer Genetics Group, IPO Porto Research Center (CI-IPOP), Portuguese Oncology Institute of Porto (IPO Porto), Porto, Portugal
| | - Sofia Maia
- Cancer Genetics Group, IPO Porto Research Center (CI-IPOP), Portuguese Oncology Institute of Porto (IPO Porto), Porto, Portugal
| | - Carla Pinto
- Department of Genetics, Portuguese Oncology Institute of Porto (IPO Porto), Porto, Portugal
| | - Pedro Pinto
- Cancer Genetics Group, IPO Porto Research Center (CI-IPOP), Portuguese Oncology Institute of Porto (IPO Porto), Porto, Portugal
| | - Augusta Monteiro
- Cancer Genetics Group, IPO Porto Research Center (CI-IPOP), Portuguese Oncology Institute of Porto (IPO Porto), Porto, Portugal
| | - Ana Peixoto
- Department of Genetics, Portuguese Oncology Institute of Porto (IPO Porto), Porto, Portugal
| | - Manuel R. Teixeira
- Cancer Genetics Group, IPO Porto Research Center (CI-IPOP), Portuguese Oncology Institute of Porto (IPO Porto), Porto, Portugal
- Department of Genetics, Portuguese Oncology Institute of Porto (IPO Porto), Porto, Portugal
- Biomedical Sciences Institute Abel Salazar (ICBAS), University of Porto, Porto, Portugal
| |
Collapse
|
5
|
Karyadi DM, Geybels MS, Karlins E, Decker B, McIntosh L, Hutchinson A, Kolb S, McDonnell SK, Hicks B, Middha S, FitzGerald LM, DeRycke MS, Yeager M, Schaid DJ, Chanock SJ, Thibodeau SN, Berndt SI, Stanford JL, Ostrander EA. Whole exome sequencing in 75 high-risk families with validation and replication in independent case-control studies identifies TANGO2, OR5H14, and CHAD as new prostate cancer susceptibility genes. Oncotarget 2017; 8:1495-1507. [PMID: 27902461 PMCID: PMC5341753 DOI: 10.18632/oncotarget.13646] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2016] [Accepted: 11/07/2016] [Indexed: 12/30/2022] Open
Abstract
Prostate cancer (PCa) susceptibility is defined by a continuum from rare, high-penetrance to common, low-penetrance alleles. Research to date has concentrated on identification of variants at the ends of that continuum. Taking an alternate approach, we focused on the important but elusive class of low-frequency, moderately penetrant variants by performing disease model-based variant filtering of whole exome sequence data from 75 hereditary PCa families. Analysis of 341 candidate risk variants identified nine variants significantly associated with increased PCa risk in a population-based, case-control study of 2,495 men. In an independent nested case-control study of 7,121 men, there was risk association evidence for TANGO2 p.Ser17Ter and the established HOXB13 p.Gly84Glu variant. Meta-analysis combining the case-control studies identified two additional variants suggestively associated with risk, OR5H14 p.Met59Val and CHAD p.Ala342Asp. The TANGO2 and HOXB13 variants co-occurred in cases more often than expected by chance and never in controls. Finally, TANGO2 p.Ser17Ter was associated with aggressive disease in both case-control studies separately. Our analyses identified three new PCa susceptibility alleles in the TANGO2, OR5H14 and CHAD genes that not only segregate in multiple high-risk families but are also of importance in altering disease risk for men from the general population. This is the first successful study to utilize sequencing in high-risk families for the express purpose of identifying low-frequency, moderately penetrant PCa risk mutations.
Collapse
Affiliation(s)
- Danielle M. Karyadi
- National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - Milan S. Geybels
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Eric Karlins
- National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Brennan Decker
- National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - Laura McIntosh
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Amy Hutchinson
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Suzanne Kolb
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | | | - Belynda Hicks
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Sumit Middha
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN, USA
| | - Liesel M. FitzGerald
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Australia
| | - Melissa S. DeRycke
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
| | - Meredith Yeager
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Daniel J. Schaid
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN, USA
| | - Stephen J. Chanock
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Stephen N. Thibodeau
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
| | - Sonja I. Berndt
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - 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
| | - Elaine A. Ostrander
- National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
6
|
Affiliation(s)
- Patrick G Pilie
- Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX,, USA
| | - Veda N Giri
- Department of Medical Oncology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA
| | - Kathleen A Cooney
- Division of Hematology/Oncology, Department of Internal Medicine, University of Michigan Medical School and The University of Michigan Comprehensive Cancer Center, Ann Arbor, MI, USA
| |
Collapse
|
7
|
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
|
8
|
Lynch HT, Kosoko‐Lasaki O, Leslie SW, Rendell M, Shaw T, Snyder C, D'Amico AV, Buxbaum S, Isaacs WB, Loeb S, Moul JW, Powell I. Screening for familial and hereditary prostate cancer. Int J Cancer 2016; 138:2579-91. [DOI: 10.1002/ijc.29949] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2015] [Revised: 10/30/2015] [Accepted: 11/03/2015] [Indexed: 12/28/2022]
Affiliation(s)
- Henry T. Lynch
- Hereditary Cancer Center and Department of Preventive MedicineCreighton University2500 California PlazaOmaha NE
| | - Omofolasade Kosoko‐Lasaki
- Departments of Surgery, Preventive Medicine & Public HealthCreighton University2500 California PlazaOmaha NE
| | - Stephen W. Leslie
- Department of Surgery (Urology)Creighton University Medical Center601 North 30th Street, Suite 3700Omaha NE
| | - Marc Rendell
- Department of Internal MedicineCreighton University Medical Center601 North 30th Street, Suite 3700Omaha NE
| | - Trudy Shaw
- Hereditary Cancer Center and Department of Preventive MedicineCreighton University2500 California PlazaOmaha NE
| | - Carrie Snyder
- Hereditary Cancer Center and Department of Preventive MedicineCreighton University2500 California PlazaOmaha NE
| | - Anthony V. D'Amico
- Department of Radiation OncologyBrigham and Women's Hospital and Dana Farber Cancer Institute, Harvard Medical SchoolBoston MA
| | - Sarah Buxbaum
- Jackson State University School of Health Sciences350 W. Woodrow Wilson DriveJackson MS
| | - William B. Isaacs
- Departments of Urology and OncologyJohns Hopkins University School of Medicine, Marburg 115, Johns Hopkins Hospital600 N. Wolfe StBaltimore MD
| | - Stacy Loeb
- Department of Urology and Population HealthNew York University550 1st Ave VZ30 (#612)New York NY
| | - Judd W. Moul
- Duke Prostate Center, Division of Urologic Surgery, DUMC 3707‐Room 1562 Duke SouthDuke University Medical CenterDurham NC
| | - Isaac Powell
- Department of UrologyWayne State University, Karmanos Cancer Institute, University Health Center 7‐CDetroit MI
| |
Collapse
|
9
|
Abstract
Adhesion G protein-coupled receptors (aGPCRs) have a long evolutionary history dating back to very basal unicellular eukaryotes. Almost every vertebrate is equipped with a set of different aGPCRs. Genomic sequence data of several hundred extinct and extant species allows for reconstruction of aGPCR phylogeny in vertebrates and non-vertebrates in general but also provides a detailed view into the recent evolutionary history of human aGPCRs. Mining these sequence sources with bioinformatic tools can unveil many facets of formerly unappreciated aGPCR functions. In this review, we extracted such information from the literature and open public sources and provide insights into the history of aGPCR in humans. This includes comprehensive analyses of signatures of selection, variability of human aGPCR genes, and quantitative traits at human aGPCR loci. As indicated by a large number of genome-wide genotype-phenotype association studies, variations in aGPCR contribute to specific human phenotypes. Our survey demonstrates that aGPCRs are significantly involved in adaptation processes, phenotype variations, and diseases in humans.
Collapse
Affiliation(s)
- Peter Kovacs
- Integrated Research and Treatment Center (IFB) AdiposityDiseases, Medical Faculty, University of Leipzig, Liebigstr. 21, Leipzig, 04103, Germany.
| | - Torsten Schöneberg
- Institute of Biochemistry, Medical Faculty, University of Leipzig, Johannisallee 30, Leipzig, 04103, Germany.
| |
Collapse
|
10
|
|
11
|
Decker B, Ostrander EA. Dysregulation of the homeobox transcription factor gene HOXB13: role in prostate cancer. PHARMACOGENOMICS & PERSONALIZED MEDICINE 2014; 7:193-201. [PMID: 25206306 PMCID: PMC4157396 DOI: 10.2147/pgpm.s38117] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Prostate cancer (PC) is the most common noncutaneous cancer in men, and epidemiological studies suggest that about 40% of PC risk is heritable. Linkage analyses in hereditary PC families have identified multiple putative loci. However, until recently, identification of specific risk alleles has proven elusive. Cooney et al used linkage mapping and segregation analysis to identify a putative risk locus on chromosome 17q21-22. In search of causative variant(s) in genes from the candidate region, a novel, potentially deleterious G84E substitution in homeobox transcription factor gene HOXB13 was observed in multiple hereditary PC families. In follow-up testing, the G84E allele was enriched in cases, especially those with an early diagnosis or positive family history of disease. This finding was replicated by others, confirming HOXB13 as a PC risk gene. The HOXB13 protein plays diverse biological roles in embryonic development and terminally differentiated tissue. In tumor cell lines, HOXB13 participates in a number of biological functions, including coactivation and localization of the androgen receptor and FOXA1. However, no consensus role has emerged and many questions remain. All HOXB13 variants with a proposed role in PC risk are predicted to damage the protein and lie in domains that are highly conserved across species. The G84E variant has the strongest epidemiological support and lies in a highly conserved MEIS protein-binding domain, which binds cofactors required for activation. On the basis of epidemiological and biological data, the G84E variant likely modulates the interaction between the HOXB13 protein and the androgen receptor, as well as affecting FOXA1-mediated transcriptional programming. However, further studies of the mutated protein are required to clarify the mechanisms by which this translates into PC risk.
Collapse
Affiliation(s)
- Brennan Decker
- Cancer Genetics and Comparative Genomics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA ; Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, School of Clinical Medicine, University of Cambridge, Cambridge, UK
| | - Elaine A Ostrander
- Cancer Genetics and Comparative Genomics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
12
|
Helfand BT, Catalona WJ. The Epidemiology and Clinical Implications of Genetic Variation in Prostate Cancer. Urol Clin North Am 2014; 41:277-97. [DOI: 10.1016/j.ucl.2014.01.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
13
|
Saunders EJ, Dadaev T, Leongamornlert DA, Jugurnauth-Little S, Tymrakiewicz M, Wiklund F, Al Olama AA, Benlloch S, Neal DE, Hamdy FC, Donovan JL, Giles GG, Severi G, Gronberg H, Aly M, Haiman CA, Schumacher F, Henderson BE, Lindstrom S, Kraft P, Hunter DJ, Gapstur S, Chanock S, Berndt SI, Albanes D, Andriole G, Schleutker J, Weischer M, Nordestgaard BG, Canzian F, Campa D, Riboli E, Key TJ, Travis RC, Ingles SA, John EM, Hayes RB, Pharoah P, Khaw KT, Stanford JL, Ostrander EA, Signorello LB, Thibodeau SN, Schaid D, Maier C, Kibel AS, Cybulski C, Cannon-Albright L, Brenner H, Park JY, Kaneva R, Batra J, Clements JA, Teixeira MR, Xu J, Mikropoulos C, Goh C, Govindasami K, Guy M, Wilkinson RA, Sawyer EJ, Morgan A, Easton DF, Muir K, Eeles RA, Kote-Jarai Z. Fine-mapping the HOXB region detects common variants tagging a rare coding allele: evidence for synthetic association in prostate cancer. PLoS Genet 2014; 10:e1004129. [PMID: 24550738 PMCID: PMC3923678 DOI: 10.1371/journal.pgen.1004129] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2013] [Accepted: 12/06/2013] [Indexed: 02/02/2023] Open
Abstract
The HOXB13 gene has been implicated in prostate cancer (PrCa) susceptibility. We performed a high resolution fine-mapping analysis to comprehensively evaluate the association between common genetic variation across the HOXB genetic locus at 17q21 and PrCa risk. This involved genotyping 700 SNPs using a custom Illumina iSelect array (iCOGS) followed by imputation of 3195 SNPs in 20,440 PrCa cases and 21,469 controls in The PRACTICAL consortium. We identified a cluster of highly correlated common variants situated within or closely upstream of HOXB13 that were significantly associated with PrCa risk, described by rs117576373 (OR 1.30, P = 2.62×10(-14)). Additional genotyping, conditional regression and haplotype analyses indicated that the newly identified common variants tag a rare, partially correlated coding variant in the HOXB13 gene (G84E, rs138213197), which has been identified recently as a moderate penetrance PrCa susceptibility allele. The potential for GWAS associations detected through common SNPs to be driven by rare causal variants with higher relative risks has long been proposed; however, to our knowledge this is the first experimental evidence for this phenomenon of synthetic association contributing to cancer susceptibility.
Collapse
Affiliation(s)
| | - Tokhir Dadaev
- The Institute of Cancer Research, Sutton, Surrey, United Kingdom
| | | | | | | | - Fredrik Wiklund
- Department of Medical Epidemiology and Biostatistics, Karolinska Institute, Stockholm, Sweden
| | - Ali Amin Al Olama
- Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, University of Cambridge, Strangeways Laboratory, Cambridge, United Kingdom
| | - Sara Benlloch
- Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, University of Cambridge, Strangeways Laboratory, Cambridge, United Kingdom
| | - David E. Neal
- Surgical Oncology (Uro-Oncology: S4), University of Cambridge, Addenbrooke's Hospital, Cambridge and 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, and 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
| | - Graham G. Giles
- Cancer Epidemiology Centre, The Cancer Council Victoria, Carlton, Victoria, Australia and 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 and Centre for Molecular, Environmental, Genetic and Analytic Epidemiology, The University of Melbourne, Melbourne, Victoria, Australia
| | - Henrik Gronberg
- Department of Medical Epidemiology and Biostatistics, Karolinska Institute, Stockholm, Sweden
| | - Markus Aly
- Department of Medical Epidemiology and Biostatistics, Karolinska Institute, Stockholm, Sweden
| | - Christopher A. Haiman
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California/Norris Comprehensive Cancer Center, Los Angeles, California, United States of America
| | - Fredrick Schumacher
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California/Norris Comprehensive Cancer Center, Los Angeles, California, United States of America
| | - Brian E. Henderson
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California/Norris Comprehensive Cancer Center, Los Angeles, California, United States of America
| | - Sara Lindstrom
- Program in Genetic Epidemiology and Statistical Genetics, Department of Epidemiology, Harvard School of Public Health, Boston, Massachusetts, United States of America
| | - Peter Kraft
- Program in Genetic Epidemiology and Statistical Genetics, Department of Epidemiology, Harvard School of Public Health, Boston, Massachusetts, United States of America
| | - David J. Hunter
- Program in Genetic Epidemiology and Statistical Genetics, Department of Epidemiology, Harvard School of Public Health, Boston, Massachusetts, United States of America
| | - Susan Gapstur
- Epidemiology Research Program, American Cancer Society, Atlanta, Georgia, United States of America
| | - Stephen Chanock
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, NIH, Bethesda, Maryland, United States of America
| | - Sonja I. Berndt
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, NIH, Bethesda, Maryland, United States of America
| | - Demetrius Albanes
- Nutritional Epidemiology Branch, National Cancer Institute, NIH, EPS-3044, Bethesda, Maryland, United States of America
| | - Gerald Andriole
- Division of Urologic Surgery, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Johanna Schleutker
- Department of Medic Biochemistry and Genetics, University of Turku, Turku and Institute of Biomedical Technology and BioMediTech, University of Tampere and FimLab Laboratories, Tampere, Finland
| | - 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
| | - Federico Canzian
- Genomic Epidemiology Group, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Daniele Campa
- Division of Cancer Epidemiology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Elio Riboli
- Department of Epidemiology & Biostatistics, School of Public Health, Imperial College London, London, United Kingdom
| | - Tim J. Key
- Cancer Epidemiology Unit, Nuffield Department of Population Health, University of Oxford, Oxford, United Kingdom
| | - Ruth C. Travis
- Cancer Epidemiology Unit, Nuffield Department of Population Health, University of Oxford, Oxford, United Kingdom
| | - Sue A. Ingles
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California/Norris Comprehensive Cancer Center, Los Angeles, California, United States of America
| | - Esther M. John
- Cancer Prevention Institute of California, Fremont, California, United States of America, and Stanford University School of Medicine, Stanford, California, United States of America
| | - Richard B. Hayes
- Division of Epidemiology, Department of Population Health, NYU Langone Medical Center, NYU Cancer Institute, New York, New York, United States of America
| | - Paul Pharoah
- Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, University of Cambridge, Strangeways Laboratory, Cambridge, United Kingdom
| | - Kay-Tee Khaw
- Clinical Gerontology Unit, University of Cambridge, Cambridge, United Kingdom
| | - Janet L. Stanford
- Department of Epidemiology, School of Public Health, University of Washington and Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
| | - Elaine A. Ostrander
- National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Lisa B. Signorello
- International Epidemiology Institute, Rockville, Maryland, and Division of Epidemiology, Department of Medicine, Vanderbilt Epidemiology Center, Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, Tennessee, United States of America
| | | | - Daniel Schaid
- Mayo Clinic, Rochester, Minnesota, United States of America
| | - Christiane Maier
- Department of Urology, University Hospital Ulm and Institute of Human Genetics University Hospital Ulm, Ulm, Germany
| | - Adam S. Kibel
- Division of Urologic Surgery, Brigham and Women's Hospital, Dana-Farber Cancer Institute, Boston, Massachusetts, United States of America
| | - Cezary Cybulski
- 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 and George E. Wahlen Department of Veterans Affairs Medical Center, Salt Lake City, Utah, United States of America
| | - Hermann Brenner
- Division of Clinical Epidemiology and Aging Research, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Jong Y. Park
- Department of Cancer Epidemiology, H. Lee Moffitt Cancer Center, Tampa, Florida, United States of America
| | - Radka Kaneva
- Molecular Medicine Center and Department of Medical Chemistry and Biochemistry, Medical University - Sofia, 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, 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
| | - Manuel R. Teixeira
- Biomedical Sciences Institute (ICBAS), Porto University, Porto, and Department of Genetics, Portuguese Oncology Institute, Porto, Portugal
| | - Jianfeng Xu
- Center for Cancer Genomics, Wake Forest University School of Medicine, Winston-Salem, North Carolina, United States of America
| | | | - Chee Goh
- The Institute of Cancer Research, Sutton, Surrey, United Kingdom
| | | | - Michelle Guy
- The Institute of Cancer Research, Sutton, Surrey, United Kingdom
| | | | - Emma J. Sawyer
- The Institute of Cancer Research, Sutton, Surrey, United Kingdom
| | - Angela Morgan
- The Institute of Cancer Research, Sutton, Surrey, United Kingdom
| | | | | | | | | | - Douglas F. Easton
- Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, University of Cambridge, Strangeways Laboratory, Cambridge, United Kingdom
| | - Ken Muir
- Warwick Medical School, University of Warwick, Coventry, United Kingdom
| | | | | |
Collapse
|
14
|
Haddad BR, Gu L, Mirtti T, Dagvadorj A, Vogiatzi P, Hoang DT, Bajaj R, Leiby B, Ellsworth E, Blackmon S, Ruiz C, Curtis M, Fortina P, Ertel A, Liu C, Rui H, Visakorpi T, Bubendorf L, Lallas CD, Trabulsi EJ, McCue P, Gomella L, Nevalainen MT. STAT5A/B gene locus undergoes amplification during human prostate cancer progression. THE AMERICAN JOURNAL OF PATHOLOGY 2013; 182:2264-75. [PMID: 23660011 DOI: 10.1016/j.ajpath.2013.02.044] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/31/2012] [Revised: 02/22/2013] [Accepted: 02/28/2013] [Indexed: 12/17/2022]
Abstract
The molecular mechanisms underlying progression of prostate cancer (PCa) to castrate-resistant (CR) and metastatic disease are poorly understood. Our previous mechanistic work shows that inhibition of transcription factor Stat5 by multiple alternative methods induces extensive rapid apoptotic death of Stat5-positive PCa cells in vitro and inhibits PCa xenograft tumor growth in nude mice. Furthermore, STAT5A/B induces invasive behavior of PCa cells in vitro and in vivo, suggesting involvement of STAT5A/B in PCa progression. Nuclear STAT5A/B protein levels are increased in high-grade PCas, CR PCas, and distant metastases, and high nuclear STAT5A/B expression predicts early disease recurrence and PCa-specific death in clinical PCas. Based on these findings, STAT5A/B represents a therapeutic target protein for advanced PCa. The mechanisms underlying increased Stat5 protein levels in PCa are unclear. Herein, we demonstrate amplification at the STAT5A/B gene locus in a significant fraction of clinical PCa specimens. STAT5A/B gene amplification was more frequently found in PCas of high histologic grades and in CR distant metastases. Quantitative in situ analysis revealed that STAT5A/B gene amplification was associated with increased STAT5A/B protein expression in PCa. Functional studies showed that increased STAT5A/B copy numbers conferred growth advantage in PCa cells in vitro and as xenograft tumors in vivo. The work presented herein provides the first evidence of somatic STAT5A/B gene amplification in clinical PCas.
Collapse
Affiliation(s)
- Bassem R Haddad
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University, Washington, District of Columbia, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
15
|
Xu J, Sun J, Zheng SL. Prostate cancer risk-associated genetic markers and their potential clinical utility. Asian J Androl 2013; 15:314-22. [PMID: 23564047 PMCID: PMC3739659 DOI: 10.1038/aja.2013.42] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2013] [Revised: 03/16/2013] [Accepted: 03/18/2013] [Indexed: 02/02/2023] Open
Abstract
Prostate cancer (PCa) is one of the most common cancers among men in Western developed countries and its incidence has increased considerably in many other parts of the world, including China. The etiology of PCa is largely unknown but is thought to be multifactorial, where inherited genetics plays an important role. In this article, we first briefly review results from studies of familial aggregation and genetic susceptibility to PCa. We then recap key findings of rare and high-penetrance PCa susceptibility genes from linkage studies in PCa families. We devote a significant portion of this article to summarizing discoveries of common and low-penetrance PCa risk-associated single-nucleotide polymorphisms (SNPs) from genetic association studies in PCa cases and controls, especially those from genome-wide association studies (GWASs). A strong focus of this article is to review the literature on the potential clinical utility of these implicated genetic markers. Most of these published studies described PCa risk estimation using a genetic score derived from multiple risk-associated SNPs and its utility in determining the need for prostate biopsy. Finally, we comment on the newly proposed concept of genetic score; the notion is to treat it as a marker for genetic predisposition, similar to family history, rather than a diagnostic marker to discriminate PCa patients from non-cancer patients. Available evidence to date suggests that genetic score is an objective and better measurement of inherited risk of PCa than family history. Another unique feature of this article is the inclusion of genetic association studies of PCa in Chinese and Japanese populations.
Collapse
Affiliation(s)
- Jianfeng Xu
- Fudan Institute of Urology, Huashan Hospital, Fudan UniversityFudan Institute of Urology, Huashan Hospital, Fudan University, Shanghai 200040, China.
| | | | | |
Collapse
|
16
|
Stott-Miller M, Karyadi DM, King T, Kwon EM, Kolb S, Stanford JL, Ostrander EA. HOXB13 mutations in a population-based, case-control study of prostate cancer. Prostate 2013; 73:634-41. [PMID: 23129385 PMCID: PMC3612366 DOI: 10.1002/pros.22604] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2012] [Accepted: 09/24/2012] [Indexed: 12/22/2022]
Abstract
BACKGROUND Prostate cancer (PC) is the most frequently diagnosed non-skin malignancy in men in the Western world, yet few disease-associated mutations have been found. Recently, a low frequency recurring mutation in the HOXB13 gene was reported among both hereditary PC families and men from the general population. MATERIALS AND METHODS We determined the distribution and frequency of the G84E HOXB13 variant in 1,310 incipient PC cases and 1,259 age-mated controls from a population-based, case-control study of PC. RESULTS The G84E mutation was more frequent in cases than controls (1.3% vs. 0.4%, respectively), and men with the HOXB13 G84E variant had a 3.3-fold higher relative risk of PC compared with noncarriers (95% CI, 1.21-8.96). There was a stronger association between the G84E variant and PC among men with no first-degree relative with PC (OR, 4.04; 95% CI, 1.12-14.51) compared to men with a family history of PC (OR, 1.49; 95% CI, 0.30-7.50; P = 0.36 for interaction). We observed some evidence of higher risk estimates associated with the variant for men with higher versus lower Gleason score (OR, 4.13; 95% CI, 1.38-12.38 vs. OR, 2.71; 95% CI, 0.88-8.30), and advanced versus local stage (OR, 4.47; 95% CI, 1.28-15.57 vs. OR, 2.98; 95% CI, 1.04-8.49), however these differences were not statistically different. CONCLUSIONS These results confirm the association of a rare HOXB13 mutation with PC in the general population and suggest that this variant may be associated with features of more aggressive disease.
Collapse
Affiliation(s)
- Marni Stott-Miller
- Division of Public Health Sciences, 1100 Fairview Ave N., Fred Hutchinson Cancer Research Center, Seattle WA, 98109
| | - Danielle M. Karyadi
- National Human Genome Research Institute, National Institutes of Health, Bethesda MD 20892
| | - Tiffany King
- National Human Genome Research Institute, National Institutes of Health, Bethesda MD 20892
| | - Erika M. Kwon
- National Human Genome Research Institute, National Institutes of Health, Bethesda MD 20892
| | - Suzanne Kolb
- Division of Public Health Sciences, 1100 Fairview Ave N., Fred Hutchinson Cancer Research Center, Seattle WA, 98109
| | - Janet L. Stanford
- Division of Public Health Sciences, 1100 Fairview Ave N., Fred Hutchinson Cancer Research Center, Seattle WA, 98109
- Department of Epidemiology, School of Public Health, University of Washington, Seattle, WA 98195
| | - Elaine A. Ostrander
- National Human Genome Research Institute, National Institutes of Health, Bethesda MD 20892
| |
Collapse
|
17
|
Witte JS, Mefford J, Plummer SJ, Liu J, Cheng I, Klein EA, Rybicki BA, Casey G. HOXB13 mutation and prostate cancer: studies of siblings and aggressive disease. Cancer Epidemiol Biomarkers Prev 2013; 22:675-80. [PMID: 23396964 DOI: 10.1158/1055-9965.epi-12-1154] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND Recent work detected for the first time a high-risk prostate cancer mutation, in homeobox B13 (HOXB13) among European-Americans. METHODS We further evaluated this G84E missense mutation (rs138213197) in two genetic association studies of prostate cancer: a family-based study of brothers and a case-control study of more aggressive disease (N = 2,665 total). We then calculated overall impact of this mutation by pooling all published studies of European-Americans. RESULTS In our studies, the mutation was found exclusively among men with prostate cancer (carrier frequency = 1.48%) or unaffected brothers of cases carrying the mutation (frequency = 0.34%), and carrying the mutation gave an OR for disease = 4.79 (P = 0.01). The G84E mutation was more common among men with an earlier age of onset (≤55 years) or a family history of prostate cancer. We also observed for the first time an African-American case carrying the G84E mutation, although at HOXB13 both of his chromosomes were of European-American ancestry. The pooled analysis also indicated that carrying the G84E mutation results in an almost five-fold increase in risk of prostate cancer (P = 3.5 × 10(-17)), and this risk is even higher among cases with an early age of prostate cancer onset (≤55 years) or a family history of disease: a test of heterogeneity across these strata gives P < 1 × 10(-5). CONCLUSIONS The HOXB13 mutation substantially increases risk of early onset, familial prostate cancer in European-American men. IMPACT Testing for the G84E mutation in men with a positive family history may help distinguish those who merit more regular screening for prostate cancer.
Collapse
Affiliation(s)
- John S Witte
- Departments of Epidemiology & Biostatistics and Urology, University of California San Francisco, San Francisco, CA 94158, USA.
| | | | | | | | | | | | | | | |
Collapse
|
18
|
Laitinen VH, Wahlfors T, Saaristo L, Rantapero T, Pelttari LM, Kilpivaara O, Laasanen SL, Kallioniemi A, Nevanlinna H, Aaltonen L, Vessella RL, Auvinen A, Visakorpi T, Tammela TLJ, Schleutker J. HOXB13 G84E mutation in Finland: population-based analysis of prostate, breast, and colorectal cancer risk. Cancer Epidemiol Biomarkers Prev 2013; 22:452-60. [PMID: 23292082 DOI: 10.1158/1055-9965.epi-12-1000-t] [Citation(s) in RCA: 70] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND A recently identified germline mutation G84E in HOXB13 was shown to increase the risk of prostate cancer. In a family-based analysis by The International Consortium for Prostate Cancer Genetics (ICPCG), the G84E mutation was most prevalent in families from the Nordic countries of Finland (22.4%) and Sweden (8.2%). METHODS To further investigate the importance of G84E in the Finns, we determined its frequency in more than 4,000 prostate cancer cases and 5,000 controls. In addition, 986 breast cancer and 442 colorectal cancer (CRC) cases were studied. Genotyping was conducted using TaqMan, MassARRAY iPLEX, and sequencing. Statistical analyses were conducted using Fisher exact test, and overall survival was analyzed using Cox modeling. RESULTS The frequency of the G84E mutation was significantly higher among patients with prostate cancer and highest among patients with a family history of the disease, hereditary prostate cancer [8.4% vs. 1.0% in controls; OR 8.8; 95% confidence interval (CI), 4.9-15.7]. The mutation contributed significantly to younger age (≤55 years) at onset and high prostate-specific antigen (PSA; ≥20 ng/mL) at diagnosis. An association with increased prostate cancer risk in patients with prior benign prostate hyperplasia (BPH) diagnosis was also revealed. No statistically significant evidence for a contribution in CRC risk was detected, but a suggestive role for the mutation was observed in familial BRCA1/2-negative breast cancer. CONCLUSIONS These findings confirm an increased cancer risk associated with the G84E mutation in the Finnish population, particularly for early-onset prostate cancer and cases with substantially elevated PSA. IMPACT This study confirms the overall importance of the HOXB13 G84E mutation in prostate cancer susceptibility.
Collapse
Affiliation(s)
- Virpi H Laitinen
- Johanna Schleutker, Medical Biochemistry and Genetics, Institute of Biomedicine, Kiinamyllynkatu 10, FI-20014 University of Turku, Finland.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Xu J, Lange EM, Lu L, Zheng SL, Wang Z, Thibodeau SN, Cannon-Albright LA, Teerlink CC, Camp NJ, Johnson AM, Zuhlke KA, Stanford JL, Ostrander EA, Wiley KE, Isaacs SD, Walsh PC, Maier C, Luedeke M, Vogel W, Schleutker J, Wahlfors T, Tammela T, Schaid D, McDonnell SK, DeRycke MS, Cancel-Tassin G, Cussenot O, Wiklund F, Grönberg H, Eeles R, Easton D, Kote-Jarai Z, Whittemore AS, Hsieh CL, Giles GG, Hopper JL, Severi G, Catalona WJ, Mandal D, Ledet E, Foulkes WD, Hamel N, Mahle L, Moller P, Powell I, Bailey-Wilson JE, Carpten JD, Seminara D, Cooney KA, Isaacs WB. HOXB13 is a susceptibility gene for prostate cancer: results from the International Consortium for Prostate Cancer Genetics (ICPCG). Hum Genet 2013; 132:5-14. [PMID: 23064873 PMCID: PMC3535370 DOI: 10.1007/s00439-012-1229-4] [Citation(s) in RCA: 150] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2012] [Accepted: 09/15/2012] [Indexed: 11/26/2022]
Abstract
Prostate cancer has a strong familial component but uncovering the molecular basis for inherited susceptibility for this disease has been challenging. Recently, a rare, recurrent mutation (G84E) in HOXB13 was reported to be associated with prostate cancer risk. Confirmation and characterization of this finding is necessary to potentially translate this information to the clinic. To examine this finding in a large international sample of prostate cancer families, we genotyped this mutation and 14 other SNPs in or flanking HOXB13 in 2,443 prostate cancer families recruited by the International Consortium for Prostate Cancer Genetics (ICPCG). At least one mutation carrier was found in 112 prostate cancer families (4.6 %), all of European descent. Within carrier families, the G84E mutation was more common in men with a diagnosis of prostate cancer (194 of 382, 51 %) than those without (42 of 137, 30 %), P = 9.9 × 10(-8) [odds ratio 4.42 (95 % confidence interval 2.56-7.64)]. A family-based association test found G84E to be significantly over-transmitted from parents to affected offspring (P = 6.5 × 10(-6)). Analysis of markers flanking the G84E mutation indicates that it resides in the same haplotype in 95 % of carriers, consistent with a founder effect. Clinical characteristics of cancers in mutation carriers included features of high-risk disease. These findings demonstrate that the HOXB13 G84E mutation is present in ~5 % of prostate cancer families, predominantly of European descent, and confirm its association with prostate cancer risk. While future studies are needed to more fully define the clinical utility of this observation, this allele and others like it could form the basis for early, targeted screening of men at elevated risk for this common, clinically heterogeneous cancer.
Collapse
Affiliation(s)
- Jianfeng Xu
- Data Coordinating Center for the ICPCG, Wake Forest University School of Medicine, Winston-Salem, NC USA
- Center for Cancer Genomics, Wake Forest University School of Medicine, Winston-Salem, NC USA
| | - Ethan M. Lange
- University of Michigan ICPCG Group, University of Michigan Medical School, Ann Arbor, MI USA
- Departments of Genetics and Biostatistics, University of North Carolina, Chapel Hill, NC USA
| | - Lingyi Lu
- Data Coordinating Center for the ICPCG, Wake Forest University School of Medicine, Winston-Salem, NC USA
- Center for Cancer Genomics, Wake Forest University School of Medicine, Winston-Salem, NC USA
| | - Siqun L. Zheng
- Data Coordinating Center for the ICPCG, Wake Forest University School of Medicine, Winston-Salem, NC USA
- Center for Cancer Genomics, Wake Forest University School of Medicine, Winston-Salem, NC USA
| | - Zhong Wang
- Data Coordinating Center for the ICPCG, Wake Forest University School of Medicine, Winston-Salem, NC USA
- Center for Cancer Genomics, Wake Forest University School of Medicine, Winston-Salem, NC USA
| | - Stephen N. Thibodeau
- Mayo Clinic ICPGC Group, Mayo Clinic, Rochester, MN USA
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN USA
| | - Lisa A. Cannon-Albright
- University of Utah ICPCG Group, University of Utah School of Medicine, Salt Lake City, UT USA
- Department of Medicine, University of Utah School of Medicine, Salt Lake City, UT USA
| | - Craig C. Teerlink
- University of Utah ICPCG Group, University of Utah School of Medicine, Salt Lake City, UT USA
- Department of Medicine, University of Utah School of Medicine, Salt Lake City, UT USA
| | - Nicola J. Camp
- University of Utah ICPCG Group, University of Utah School of Medicine, Salt Lake City, UT USA
- Department of Medicine, University of Utah School of Medicine, Salt Lake City, UT USA
| | - Anna M. Johnson
- University of Michigan ICPCG Group, University of Michigan Medical School, Ann Arbor, MI USA
- Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI USA
| | - Kimberly A. Zuhlke
- University of Michigan ICPCG Group, University of Michigan Medical School, Ann Arbor, MI USA
- Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI USA
| | - Janet L. Stanford
- Fred Hutchinson Cancer Research Center (FHCRC) ICPCG Group, Seattle, WA USA
- Division of Public Health Sciences, FHCRC, Seattle, WA USA
| | - Elaine A. Ostrander
- Fred Hutchinson Cancer Research Center (FHCRC) ICPCG Group, Seattle, WA USA
- Cancer Genetics Branch, National Human Genome Research Institute, NIH, Bethesda, MD USA
| | - Kathleen E. Wiley
- Johns Hopkins University ICPCG Group, Baltimore, MD USA
- Department of Urology, Johns Hopkins Medical Institutions, Johns Hopkins Hospital, Marburg 115, 600 North Wolfe Street, Baltimore, MD 21287 USA
| | - Sarah D. Isaacs
- Johns Hopkins University ICPCG Group, Baltimore, MD USA
- Department of Urology, Johns Hopkins Medical Institutions, Johns Hopkins Hospital, Marburg 115, 600 North Wolfe Street, Baltimore, MD 21287 USA
| | - Patrick C. Walsh
- Johns Hopkins University ICPCG Group, Baltimore, MD USA
- Department of Urology, Johns Hopkins Medical Institutions, Johns Hopkins Hospital, Marburg 115, 600 North Wolfe Street, Baltimore, MD 21287 USA
| | - Christiane Maier
- University of Ulm ICPCG Group, University of Ulm, Ulm, Germany
- Department of Urology, University of Ulm, Ulm, Germany
| | - Manuel Luedeke
- University of Ulm ICPCG Group, University of Ulm, Ulm, Germany
- Department of Urology, University of Ulm, Ulm, Germany
| | - Walther Vogel
- University of Ulm ICPCG Group, University of Ulm, Ulm, Germany
- Institute of Human Genetics, University of Ulm, Ulm, Germany
| | - Johanna Schleutker
- University of Tampere ICPCG Group, University of Tampere and Fimlab Laboratories, Tampere, Finland
- Institute of Biomedical Technology/BioMediTech, University of Tampere and Fimlab Laboratories, Tampere, Finland
- Department of Medical Biochemistry and Genetics, University of Turku, Turku, Finland
| | - Tiina Wahlfors
- University of Tampere ICPCG Group, University of Tampere and Fimlab Laboratories, Tampere, Finland
- Institute of Biomedical Technology/BioMediTech, University of Tampere and Fimlab Laboratories, Tampere, Finland
| | - Teuvo Tammela
- University of Tampere ICPCG Group, University of Tampere and Fimlab Laboratories, Tampere, Finland
- Department of Urology, Tampere University Hospital, Tampere, Finland
| | - Daniel Schaid
- Mayo Clinic ICPGC Group, Mayo Clinic, Rochester, MN USA
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN USA
| | - Shannon K. McDonnell
- Mayo Clinic ICPGC Group, Mayo Clinic, Rochester, MN USA
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN USA
| | - Melissa S. DeRycke
- Mayo Clinic ICPGC Group, Mayo Clinic, Rochester, MN USA
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN USA
| | | | - Olivier Cussenot
- CeRePP ICPCG Group, Paris, France
- Department of Urology, APHP, Hospital Tenon, Paris, France
| | - Fredrik Wiklund
- Karolinska ICPCG Group, Karolinska Institutet, Stockholm, Sweden
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Henrik Grönberg
- Karolinska ICPCG Group, Karolinska Institutet, Stockholm, Sweden
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Ros Eeles
- ACTANE (Anglo/Canadian/Texan/Australian/Norwegian/EU Biomed) Consortium ICPCG Group, Surrey, UK
- Institute of Cancer Research and Royal Marsden NHS Foundation Trust, Surrey, UK
| | - Doug Easton
- ACTANE (Anglo/Canadian/Texan/Australian/Norwegian/EU Biomed) Consortium ICPCG Group, Surrey, UK
- Strangeways Laboratory, Department of Oncology, Centre for Cancer Genetic Epidemiology, University of Cambridge, Cambridge, UK
| | - Zsofia Kote-Jarai
- ACTANE (Anglo/Canadian/Texan/Australian/Norwegian/EU Biomed) Consortium ICPCG Group, Surrey, UK
- Institute of Cancer Research and Royal Marsden NHS Foundation Trust, Surrey, UK
| | - Alice S. Whittemore
- BC/CA/HI ICPCG Group, Stanford School of Medicine, Stanford, CA USA
- Department of Health Research and Policy, Stanford School of Medicine, Stanford, CA USA
- Stanford Comprehensive Cancer Center, Stanford School of Medicine, Stanford, CA USA
| | - Chih-Lin Hsieh
- BC/CA/HI ICPCG Group, Stanford School of Medicine, Stanford, CA USA
- Department of Urology and Department of Biochemistry and Molecular Biology, University of Southern California, Los Angeles, CA USA
| | - Graham G. Giles
- ACTANE (Anglo/Canadian/Texan/Australian/Norwegian/EU Biomed) Consortium ICPCG Group, Surrey, UK
- Cancer Epidemiology Centre, Cancer Council Victoria, Melbourne, Australia
- Centre for Molecular, Environmental, Genetic and Analytical Epidemiology, University of Melbourne, Melbourne, Australia
| | - John L. Hopper
- ACTANE (Anglo/Canadian/Texan/Australian/Norwegian/EU Biomed) Consortium ICPCG Group, Surrey, UK
- Cancer Epidemiology Centre, Cancer Council Victoria, Melbourne, Australia
- Centre for Molecular, Environmental, Genetic and Analytical Epidemiology, University of Melbourne, Melbourne, Australia
| | - Gianluca Severi
- ACTANE (Anglo/Canadian/Texan/Australian/Norwegian/EU Biomed) Consortium ICPCG Group, Surrey, UK
- Cancer Epidemiology Centre, Cancer Council Victoria, Melbourne, Australia
- Centre for Molecular, Environmental, Genetic and Analytical Epidemiology, University of Melbourne, Melbourne, Australia
| | - William J. Catalona
- Northwestern University ICPCG Group, Chicago, IL USA
- Northwestern University Feinberg School of Medicine, Chicago, IL USA
| | - Diptasri Mandal
- Louisiana State University ICPCG Group, New Orleans, LA USA
- Department of Genetics, Louisiana State University Health Sciences Center, New Orleans, LA USA
| | - Elisa Ledet
- Louisiana State University ICPCG Group, New Orleans, LA USA
- Department of Genetics, Louisiana State University Health Sciences Center, New Orleans, LA USA
| | - William D. Foulkes
- ACTANE (Anglo/Canadian/Texan/Australian/Norwegian/EU Biomed) Consortium ICPCG Group, Surrey, UK
- Program in Cancer Genetics, Departments of Oncology and Human Genetics, McGill University, Montreal, QC Canada
- Research Institute of the McGill University Health Centre, Montreal, QC Canada
| | - Nancy Hamel
- ACTANE (Anglo/Canadian/Texan/Australian/Norwegian/EU Biomed) Consortium ICPCG Group, Surrey, UK
- Program in Cancer Genetics, Departments of Oncology and Human Genetics, McGill University, Montreal, QC Canada
- Research Institute of the McGill University Health Centre, Montreal, QC Canada
| | - Lovise Mahle
- ACTANE (Anglo/Canadian/Texan/Australian/Norwegian/EU Biomed) Consortium ICPCG Group, Surrey, UK
- The Norwegian Radium Hospital, Oslo, Norway
| | - Pal Moller
- ACTANE (Anglo/Canadian/Texan/Australian/Norwegian/EU Biomed) Consortium ICPCG Group, Surrey, UK
- The Norwegian Radium Hospital, Oslo, Norway
| | - Isaac Powell
- African American Hereditary Prostate Cancer ICPCG Group, Detroit, MI USA
- Karmanos Cancer Institute, Wayne State University, Detroit, MI USA
| | - Joan E. Bailey-Wilson
- African American Hereditary Prostate Cancer ICPCG Group, Detroit, MI USA
- Inherited Disease Research Branch, National Human Genome Research Institute, NIH, Bethesda, MD USA
| | - John D. Carpten
- African American Hereditary Prostate Cancer ICPCG Group, Detroit, MI USA
- Genetic Basis of Human Disease Research Division, Translational Genomics Research Institute, Phoenix, AZ USA
| | | | - Kathleen A. Cooney
- University of Michigan ICPCG Group, University of Michigan Medical School, Ann Arbor, MI USA
- Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI USA
| | - William B. Isaacs
- Johns Hopkins University ICPCG Group, Baltimore, MD USA
- Department of Urology, Johns Hopkins Medical Institutions, Johns Hopkins Hospital, Marburg 115, 600 North Wolfe Street, Baltimore, MD 21287 USA
| | - International Consortium for Prostate Cancer Genetics
- Data Coordinating Center for the ICPCG, Wake Forest University School of Medicine, Winston-Salem, NC USA
- Center for Cancer Genomics, Wake Forest University School of Medicine, Winston-Salem, NC USA
- University of Michigan ICPCG Group, University of Michigan Medical School, Ann Arbor, MI USA
- Departments of Genetics and Biostatistics, University of North Carolina, Chapel Hill, NC USA
- Mayo Clinic ICPGC Group, Mayo Clinic, Rochester, MN USA
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN USA
- University of Utah ICPCG Group, University of Utah School of Medicine, Salt Lake City, UT USA
- Department of Medicine, University of Utah School of Medicine, Salt Lake City, UT USA
- Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI USA
- Fred Hutchinson Cancer Research Center (FHCRC) ICPCG Group, Seattle, WA USA
- Division of Public Health Sciences, FHCRC, Seattle, WA USA
- Cancer Genetics Branch, National Human Genome Research Institute, NIH, Bethesda, MD USA
- Johns Hopkins University ICPCG Group, Baltimore, MD USA
- Department of Urology, Johns Hopkins Medical Institutions, Johns Hopkins Hospital, Marburg 115, 600 North Wolfe Street, Baltimore, MD 21287 USA
- University of Ulm ICPCG Group, University of Ulm, Ulm, Germany
- Department of Urology, University of Ulm, Ulm, Germany
- Institute of Human Genetics, University of Ulm, Ulm, Germany
- University of Tampere ICPCG Group, University of Tampere and Fimlab Laboratories, Tampere, Finland
- Institute of Biomedical Technology/BioMediTech, University of Tampere and Fimlab Laboratories, Tampere, Finland
- Department of Medical Biochemistry and Genetics, University of Turku, Turku, Finland
- Department of Urology, Tampere University Hospital, Tampere, Finland
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN USA
- CeRePP ICPCG Group, Paris, France
- Department of Urology, APHP, Hospital Tenon, Paris, France
- CeRePP UPMC University, Paris, France
- Karolinska ICPCG Group, Karolinska Institutet, Stockholm, Sweden
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
- ACTANE (Anglo/Canadian/Texan/Australian/Norwegian/EU Biomed) Consortium ICPCG Group, Surrey, UK
- Institute of Cancer Research and Royal Marsden NHS Foundation Trust, Surrey, UK
- Strangeways Laboratory, Department of Oncology, Centre for Cancer Genetic Epidemiology, University of Cambridge, Cambridge, UK
- BC/CA/HI ICPCG Group, Stanford School of Medicine, Stanford, CA USA
- Department of Health Research and Policy, Stanford School of Medicine, Stanford, CA USA
- Stanford Comprehensive Cancer Center, Stanford School of Medicine, Stanford, CA USA
- Department of Urology and Department of Biochemistry and Molecular Biology, University of Southern California, Los Angeles, CA USA
- Cancer Epidemiology Centre, Cancer Council Victoria, Melbourne, Australia
- Centre for Molecular, Environmental, Genetic and Analytical Epidemiology, University of Melbourne, Melbourne, Australia
- Northwestern University ICPCG Group, Chicago, IL USA
- Northwestern University Feinberg School of Medicine, Chicago, IL USA
- Louisiana State University ICPCG Group, New Orleans, LA USA
- Department of Genetics, Louisiana State University Health Sciences Center, New Orleans, LA USA
- Program in Cancer Genetics, Departments of Oncology and Human Genetics, McGill University, Montreal, QC Canada
- Research Institute of the McGill University Health Centre, Montreal, QC Canada
- The Norwegian Radium Hospital, Oslo, Norway
- African American Hereditary Prostate Cancer ICPCG Group, Detroit, MI USA
- Karmanos Cancer Institute, Wayne State University, Detroit, MI USA
- Inherited Disease Research Branch, National Human Genome Research Institute, NIH, Bethesda, MD USA
- Genetic Basis of Human Disease Research Division, Translational Genomics Research Institute, Phoenix, AZ USA
- National Cancer Institute, NIH, Bethesda, MD USA
| |
Collapse
|
20
|
Pelttari LM, Nurminen R, Gylfe A, Aaltonen LA, Schleutker J, Nevanlinna H. Screening of Finnish RAD51C founder mutations in prostate and colorectal cancer patients. BMC Cancer 2012; 12:552. [PMID: 23176254 PMCID: PMC3522023 DOI: 10.1186/1471-2407-12-552] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2012] [Accepted: 11/13/2012] [Indexed: 01/20/2023] Open
Abstract
BACKGROUND Rare, heterozygous germline mutations in the RAD51C gene have been found in breast and ovarian cancer families. In the Finnish population, we have identified two founder mutations in RAD51C that increase the risk of ovarian cancer but not breast cancer in the absence of ovarian cancer. Risk for other cancers has not been studied. METHODS To study the role of RAD51C mutations in other common cancer types, we genotyped the Finnish RAD51C founder mutations c.837 + 1G > A and c.93delG in 1083 prostate cancer patients and 802 colorectal cancer patients using TaqMan Real-Time PCR. RESULTS No RAD51C mutations c.837 + 1G > A or c.93delG were detected among the prostate or colorectal cancer patients. CONCLUSIONS The results suggest that the RAD51C mutations do not predispose to prostate or colorectal cancer.
Collapse
Affiliation(s)
- Liisa M Pelttari
- Departments of Obstetrics and Gynecology, Helsinki University Central Hospital and University of Helsinki, Helsinki, Finland
| | | | | | | | | | | |
Collapse
|
21
|
Lynch HT, Shaw TG. Familial prostate cancer and HOXB13 founder mutations: geographic and racial/ethnic variations. Hum Genet 2012; 132:1-4. [PMID: 23001594 DOI: 10.1007/s00439-012-1226-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2012] [Accepted: 09/04/2012] [Indexed: 11/24/2022]
|
22
|
Kouprina N, Lee NCO, Pavlicek A, Samoshkin A, Kim JH, Lee HS, Varma S, Reinhold WC, Otstot J, Solomon G, Davis S, Meltzer PS, Schleutker J, Larionov V. Exclusion of the 750-kb genetically unstable region at Xq27 as a candidate locus for prostate malignancy in HPCX1-linked families. Genes Chromosomes Cancer 2012; 51:933-48. [PMID: 22733720 DOI: 10.1002/gcc.21977] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2012] [Accepted: 05/08/2012] [Indexed: 12/14/2022] Open
Abstract
Several linkage studies provided evidence for the presence of the hereditary prostate cancer locus, HPCX1, at Xq27-q28. The strongest linkage peak of prostate cancer overlies a variable region of ~750 kb at Xq27 enriched by segmental duplications (SDs), suggesting that the predisposition to prostate cancer may be a genomic disorder caused by recombinational interaction between SDs. The large size of SDs and their sequence similarity make it difficult to examine this region for possible rearrangements using standard methods. To overcome this problem, direct isolation of a set of genomic segments by in vivo recombination in yeast (a TAR cloning technique) was used to perform a mutational analysis of the 750 kb region in X-linked families. We did not detect disease-specific rearrangements within this region. In addition, transcriptome and computational analyses were performed to search for nonannotated genes within the Xq27 region, which may be associated with genetic predisposition to prostate cancer. Two candidate genes were identified, one of which is a novel gene termed SPANXL that represents a highly diverged member of the SPANX gene family, and the previously described CDR1 gene that is expressed at a high level in both normal and malignant prostate cells, and mapped 210 kb of upstream the SPANX gene cluster. No disease-specific alterations were identified in these genes. Our results exclude the 750-kb genetically unstable region at Xq27 as a candidate locus for prostate malignancy. Adjacent regions appear to be the most likely candidates to identify the elusive HPCX1 locus.
Collapse
Affiliation(s)
- Natalay Kouprina
- Laboratory of Molecular Pharmacology, NCI, NIH, Bethesda, MD, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Ledet EM, Sartor O, Rayford W, Bailey-Wilson JE, Mandal DM. Suggestive evidence of linkage identified at chromosomes 12q24 and 2p16 in African American prostate cancer families from Louisiana. Prostate 2012; 72:938-47. [PMID: 22615067 DOI: 10.1002/pros.21496] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2011] [Accepted: 09/13/2011] [Indexed: 12/24/2022]
Abstract
BACKGROUND In the United States, incidence of prostate cancer in African American men is more than twice than that of any other race. Thus far, numerous disease susceptibility loci have been identified for this cancer but definite locus-specific information is not yet established due to the tremendous amount of genetic and disease heterogeneity; additionally, despite high prevalence of prostate cancer amongst African American men, this population has been under represented in genetic studies of prostate cancer. METHODS In order to identify the susceptible locus (loci) for prostate cancer in African Americans, we have performed linkage analyses on members of 15 large high-risk families. Specifically, these families were recruited from Louisiana and represent a uniquely admixed African American population exclusive to Southern Louisiana. In addition to geographical constraints, these families were clinically homogeneous creating a well-characterized collection of large pedigrees. The families were genotyped with Illumina Infinium II SNP HumanLinkage-12 panel and extensive demographic and clinical information was documented from the hospital pathological reports and family interviews. RESULTS We identified two novel regions, 12q24 and 2p16, with suggestive evidence of linkage under the dominant model of inheritance. CONCLUSIONS This is the first time that chromosome 12q24 (HLOD = 2.21) and 2p16 (HLOD = 1.97) has been shown to be associated with prostate cancer in high-risk African American families. These results provide insight to prostate cancer in an exceptional, well-characterized African American population, and illustrate the significance of utilizing large unique, but homogenous pedigrees.
Collapse
Affiliation(s)
- Elisa M Ledet
- Department of Genetics, Louisiana State University Health Sciences Center, New Orleans, Louisiana 70112, USA
| | | | | | | | | |
Collapse
|
24
|
Replication and fine mapping for association of the C2orf43, FOXP4, GPRC6A and RFX6 genes with prostate cancer in the Chinese population. PLoS One 2012; 7:e37866. [PMID: 22662242 PMCID: PMC3360662 DOI: 10.1371/journal.pone.0037866] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2012] [Accepted: 04/26/2012] [Indexed: 11/19/2022] Open
Abstract
Background Prostate cancer represents the leading cause of male death across the world. A recent genome-wide association study (GWAS) identified five novel susceptibility loci for prostate cancer in the Japanese population. This study is to replicate and fine map the potential association of these five loci with prostate cancer in the Chinese Han population. Methods In Phase I of the study, we tested the five single nucleotide polymorphisms (SNPs) which showed the strongest association evidence in the original GWAS in Japanese. The study sample consists of 1,169 Chinese Hans, comprising 483 patients and 686 healthy controls. Then in phase II, flanking SNPs of the successfully replicated SNPs in Phase I were genotyped and tested for association with prostate cancer to fine map those significant association signals. Results We successfully replicated the association of rs13385191 (located in the C2orf43 gene, P = 8.60×10−5), rs12653946 (P = 1.33×10−6), rs1983891 (FOXP4, P = 6.22×10−5), and rs339331 (GPRC6A/RFX6, P = 1.42×10−5) with prostate cancer. The most significant odds ratio (OR) was recorded as 1.41 (95% confidence interval 1.18–1.68) for rs12653946. Rs9600079 did not show significant association (P = 8.07×10−2) with prostate cancer in this study. The Phase II study refined these association signals, and identified several SNPs showing more significant association with prostate cancer than the very SNPs tested in Phase I. Conclusions Our results provide further support for association of the C2orf43, FOXP4, GPRC6A and RFX6 genes with prostate cancer in Eastern Asian populations. This study also characterized the novel loci reported in the original GWAS with more details. Further work is still required to determine the functional variations and finally clarify the underlying biological mechanisms.
Collapse
|
25
|
Ewing CM, Ray AM, Lange EM, Zuhlke KA, Robbins CM, Tembe WD, Wiley KE, Isaacs SD, Johng D, Wang Y, Bizon C, Yan G, Gielzak M, Partin AW, Shanmugam V, Izatt T, Sinari S, Craig DW, Zheng SL, Walsh PC, Montie JE, Xu J, Carpten JD, Isaacs WB, Cooney KA. Germline mutations in HOXB13 and prostate-cancer risk. N Engl J Med 2012; 366:141-9. [PMID: 22236224 PMCID: PMC3779870 DOI: 10.1056/nejmoa1110000] [Citation(s) in RCA: 479] [Impact Index Per Article: 39.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
BACKGROUND Family history is a significant risk factor for prostate cancer, although the molecular basis for this association is poorly understood. Linkage studies have implicated chromosome 17q21-22 as a possible location of a prostate-cancer susceptibility gene. METHODS We screened more than 200 genes in the 17q21-22 region by sequencing germline DNA from 94 unrelated patients with prostate cancer from families selected for linkage to the candidate region. We tested family members, additional case subjects, and control subjects to characterize the frequency of the identified mutations. RESULTS Probands from four families were discovered to have a rare but recurrent mutation (G84E) in HOXB13 (rs138213197), a homeobox transcription factor gene that is important in prostate development. All 18 men with prostate cancer and available DNA in these four families carried the mutation. The carrier rate of the G84E mutation was increased by a factor of approximately 20 in 5083 unrelated subjects of European descent who had prostate cancer, with the mutation found in 72 subjects (1.4%), as compared with 1 in 1401 control subjects (0.1%) (P=8.5x10(-7)). The mutation was significantly more common in men with early-onset, familial prostate cancer (3.1%) than in those with late-onset, nonfamilial prostate cancer (0.6%) (P=2.0x10(-6)). CONCLUSIONS The novel HOXB13 G84E variant is associated with a significantly increased risk of hereditary prostate cancer. Although the variant accounts for a small fraction of all prostate cancers, this finding has implications for prostate-cancer risk assessment and may provide new mechanistic insights into this common cancer. (Funded by the National Institutes of Health and others.).
Collapse
Affiliation(s)
- Charles M Ewing
- Johns Hopkins University and the James Buchanan Brady Urological Institute, Baltimore, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Yuan Y, Ferguson LR. Nutrigenetics and Prostate Cancer: 2011 and Beyond. JOURNAL OF NUTRIGENETICS AND NUTRIGENOMICS 2011; 4:121-36. [DOI: 10.1159/000327902] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
27
|
|
28
|
Ray AM, Zuhlke KA, Johnson GR, Levin AM, Douglas JA, Lange EM, Cooney KA. Absence of truncating BRIP1 mutations in chromosome 17q-linked hereditary prostate cancer families. Br J Cancer 2009; 101:2043-7. [PMID: 19935797 PMCID: PMC2795448 DOI: 10.1038/sj.bjc.6605433] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Background: In a genome-wide scan (GWS) of 175 multiplex prostate cancer (PCa) families from the University of Michigan Prostate Cancer Genetics Project (PCGP), linkage was observed to markers on chromosome 17q21–24, a region that includes two breast cancer susceptibility genes, BRCA1 and BRIP1. BRIP1 is a Fanconi anaemia gene (FANCJ) that interacts with the BRCT domain of BRCA1 and has a role in DNA damage repair. Protein truncating mutations in BRIP1 have been identified in hereditary breast and ovarian cancer families, and a recent report suggested that a recurrent truncating mutation (R798X) may have a role in PCa susceptibility. Methods: We examined the role of BRIP1 mutations in hereditary PCa through sequence analysis of 94 individuals from PCGP families showing linkage to 17q. Results: A total of 24 single-nucleotide polymorphisms, including 7 missense variants but no protein truncating mutations, were observed. Conclusion: The data presented here suggest that BRIP1 truncating mutations are uncommon in PCa cases and do not account for the linkage to chromosome 17q observed in our GWS. Additional investigation is needed to determine the significance, if any, of the observed BRIP1 missense variants in hereditary PCa.
Collapse
Affiliation(s)
- A M Ray
- Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, USA
| | | | | | | | | | | | | |
Collapse
|
29
|
Liao Z, Lutz J, Nevalainen MT. Transcription factor Stat5a/b as a therapeutic target protein for prostate cancer. Int J Biochem Cell Biol 2009; 42:186-92. [PMID: 19914392 DOI: 10.1016/j.biocel.2009.11.001] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2009] [Revised: 11/02/2009] [Accepted: 11/03/2009] [Indexed: 10/20/2022]
Abstract
Prostate cancer is the most common non-cutaneous cancer in Western males. The majority of prostate cancer fatalities are caused by development of castration-resistant growth and metastatic spread of the primary tumor. The average duration of the response of primary prostate cancer to hormonal ablation is less than 3 years, and 75% of prostate cancers in the United States progress to castration-resistant disease. The existing pharmacological therapies for metastatic and/or castration-resistant prostate cancer do not provide significant survival benefit. This review summarizes the importance of transcription factor Stat5 signaling in the pathogenesis of prostate cancer and discusses the molecular basis of Stat5a/b inhibition as a therapeutic strategy for prostate cancer.
Collapse
Affiliation(s)
- Zhiyong Liao
- Department of Cancer Biology, Kimmel Cancer Center, Thomas Jefferson University, 233 S. 10th Street, Philadelphia, PA 19107, USA
| | | | | |
Collapse
|
30
|
Incidence of Cancer in Finnish Families with Clinically Aggressive and Nonaggressive Prostate Cancer. Cancer Epidemiol Biomarkers Prev 2009; 18:3049-56. [DOI: 10.1158/1055-9965.epi-09-0382] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
|
31
|
Patel AR, Klein EA. Risk factors for prostate cancer. ACTA ACUST UNITED AC 2009; 6:87-95. [DOI: 10.1038/ncpuro1290] [Citation(s) in RCA: 85] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2008] [Accepted: 12/10/2008] [Indexed: 11/09/2022]
|
32
|
Kim SH, Cho KS, Han KS, Joung JY, Seo HK, Chung J, Pak HO, Park WS, Lee KH. Familial Prostate Cancer in Three Brothers. Korean J Urol 2009. [DOI: 10.4111/kju.2009.50.2.195] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Affiliation(s)
- Sung Han Kim
- Department of Urology, Seoul National University College of Medicine, Seoul, Korea
| | - Kang Su Cho
- Urologic Oncology Clinic, National Cancer Center, Goyang, Korea
| | - Kyung Seok Han
- Urologic Oncology Clinic, National Cancer Center, Goyang, Korea
| | - Jae Young Joung
- Urologic Oncology Clinic, National Cancer Center, Goyang, Korea
| | - Ho Kyung Seo
- Urologic Oncology Clinic, National Cancer Center, Goyang, Korea
| | - Jinsoo Chung
- Urologic Oncology Clinic, National Cancer Center, Goyang, Korea
| | - Hee Ok Pak
- Gachon University of Medicine and Science, Incheon, Korea
| | - Weon Seo Park
- Department of Pathology, National Cancer Center, Goyang, Korea
| | - Kang Hyun Lee
- Urologic Oncology Clinic, National Cancer Center, Goyang, Korea
| |
Collapse
|
33
|
Carvalho R, Pinheiro M, Medeiros R. Study of 16 X-STRs in a prostate cancer population sample (preliminary results). FORENSIC SCIENCE INTERNATIONAL GENETICS SUPPLEMENT SERIES 2008. [DOI: 10.1016/j.fsigss.2007.10.168] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
|
34
|
Demogines A, Smith E, Kruglyak L, Alani E. Identification and dissection of a complex DNA repair sensitivity phenotype in Baker's yeast. PLoS Genet 2008; 4:e1000123. [PMID: 18617998 PMCID: PMC2440805 DOI: 10.1371/journal.pgen.1000123] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2008] [Accepted: 06/09/2008] [Indexed: 11/18/2022] Open
Abstract
Complex traits typically involve the contribution of multiple gene variants. In this study, we took advantage of a high-density genotyping analysis of the BY (S288c) and RM strains of Saccharomyces cerevisiae and of 123 derived spore progeny to identify the genetic loci that underlie a complex DNA repair sensitivity phenotype. This was accomplished by screening hybrid yeast progeny for sensitivity to a variety of DNA damaging agents. Both the BY and RM strains are resistant to the ultraviolet light-mimetic agent 4-nitroquinoline 1-oxide (4-NQO); however, hybrid progeny from a BYxRM cross displayed varying sensitivities to the drug. We mapped a major quantitative trait locus (QTL), RAD5, and identified the exact polymorphism within this locus responsible for 4-NQO sensitivity. By using a backcrossing strategy along with array-assisted bulk segregant analysis, we identified one other locus, MKT1, and a QTL on Chromosome VII that also link to the hybrid 4-NQO-sensitive phenotype but confer more minor effects. This work suggests an additive model for sensitivity to 4-NQO and provides a strategy for mapping both major and minor QTL that confer background-specific phenotypes. It also provides tools for understanding the effect of genetic background on sensitivity to genotoxic agents.
Collapse
Affiliation(s)
- Ann Demogines
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, New York, United States of America
| | - Erin Smith
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, New Jersey, United States of America
- Department of Ecology and Evolutionary Biology, Princeton University, Princeton, New Jersey, United States of America
| | - Leonid Kruglyak
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, New Jersey, United States of America
- Department of Ecology and Evolutionary Biology, Princeton University, Princeton, New Jersey, United States of America
| | - Eric Alani
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, New York, United States of America
- * E-mail:
| |
Collapse
|
35
|
Abril J, de Heredia ML, González L, Clèries R, Nadal M, Condom E, Aguiló F, Gómez-Zaera M, Nunes V. Altered expression of 12S/MT-RNR1, MT-CO2/COX2, and MT-ATP6 mitochondrial genes in prostate cancer. Prostate 2008; 68:1086-96. [PMID: 18409190 DOI: 10.1002/pros.20771] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
BACKGROUND Prostate cancer is one of the commonest cancers worldwide and is responsible for nearly 6% of all male cancer deaths. Despite this relevance, the mechanisms involved in the development and progression of this malignancy remain unknown. The involvement of polypeptides of the mitochondrial respiratory chain, the Krebs cycle and the glutathione antioxidant system in this type of cancer has been previously described, although no publication has focused on the expression of mitochondrial genes in the prostate of PCa patients. METHODS We have determined by reverse transcription-quantitative PCR (RT-qPCR) the relative amount of the transcripts of eight mitochondrial genes (MT-ND2, MT-ND4, MT-ND6, MT-CYB, 12S/MT-RNR1, 16S/MT-RNR2, MT-CO2/COX2, MT-ATP6), and four nuclear genes (COX11, GSR, CS, ACO2), all of them key players in the normal metabolism of mitochondria. Additionally we analyzed the expression of Cyclophilin A (PPIA). RESULTS We observed differential expression of mitochondrial 12S/MT-RNR1, MT-CO2/COX2, and MT-ATP6 transcripts in tumor samples when compared to their paired normal samples. CONCLUSIONS The amount of mitochondrial 12S/MT-RNR1, MT-CO2/COX2, and MT-ATP6 transcripts is significantly decreased in tumor samples when compared to their paired normal sample, suggesting that mitochondrial gene expression is altered in PCa.
Collapse
Affiliation(s)
- Jesús Abril
- Centre de Genètica Mèdica i Molecular-IDIBELL, Hospital Duran i Reynals, L'Hospitalet de Llobregat, Barcelona, Spain
| | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Demogines A, Wong A, Aquadro C, Alani E. Incompatibilities involving yeast mismatch repair genes: a role for genetic modifiers and implications for disease penetrance and variation in genomic mutation rates. PLoS Genet 2008; 4:e1000103. [PMID: 18566663 PMCID: PMC2413424 DOI: 10.1371/journal.pgen.1000103] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2008] [Accepted: 05/21/2008] [Indexed: 11/22/2022] Open
Abstract
Genetic background effects underlie the penetrance of most genetically determined phenotypes, including human diseases. To explore how such effects can modify a mutant phenotype in a genetically tractable system, we examined an incompatibility involving the MLH1 and PMS1 mismatch repair genes using a large population sample of geographically and ecologically diverse Saccharomyces cerevisiae strains. The mismatch repair incompatibility segregates into naturally occurring yeast strains, with no strain bearing the deleterious combination. In assays measuring the mutator phenotype conferred by different combinations of MLH1 and PMS1 from these strains, we observed a mutator phenotype only in combinations predicted to be incompatible. Surprisingly, intragenic modifiers could be mapped that specifically altered the strength of the incompatibility over a 20-fold range. Together, these observations provide a powerful model in which to understand the basis of disease penetrance and how such genetic variation, created through mating, could result in new mutations that could be the raw material of adaptive evolution in yeast populations. For many common afflictions, it is difficult to map disease-associated loci because multiple loci are involved, with some loci playing greater roles than others. To explore how complex interactions can contribute to disease, we examined an incompatibility involving the MLH1 and PMS1 DNA mismatch repair proteins in baker's yeast. In our system, an incompatibility is defined as a defect occurring when specific combinations of MLH1 and PMS1 proteins obtained from different baker's yeast strains are tested for function. We identified amino acid differences at only one site in each protein that contributed to this incompatibility. We also showed that amino acid differences that could cause such an incompatibility are found in strains collected from across the globe. No strain contained the incompatible MLH1-PMS1 combination, indicating that it was likely to be deleterious. When such a combination was created in the laboratory, we could detect a wide range of defects that were under the control of genetic modifiers. These observations provide a powerful model in which to understand the basis of disease penetrance and how segregation of defects in mismatch repair may allow for rapid yet reversible changes in genomic mutation rates that can help yeast adapt to changing or novel environments.
Collapse
Affiliation(s)
- Ann Demogines
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, New York, United States of America
| | - Alex Wong
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, New York, United States of America
| | - Charles Aquadro
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, New York, United States of America
| | - Eric Alani
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, New York, United States of America
- * E-mail:
| |
Collapse
|
37
|
Tan SH, Nevalainen MT. Signal transducer and activator of transcription 5A/B in prostate and breast cancers. Endocr Relat Cancer 2008; 15:367-90. [PMID: 18508994 PMCID: PMC6036917 DOI: 10.1677/erc-08-0013] [Citation(s) in RCA: 82] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Protein kinase signaling pathways, such as Janus kinase 2-Signal transducer and activator of transcription 5A/B (JAK2-STAT5A/B), are of significant interest in the search for new therapeutic strategies in both breast and prostate cancers. In prostate cancer, the components of the JAK2-STAT5A/B signaling pathway provide molecular targets for small-molecule inhibition of survival and growth signals of the cells. At the same time, new evidence suggests that the STAT5A/B signaling pathway is involved in the transition of organ-confined prostate cancer to hormone-refractory disease. This implies that the active JAK2-STAT5A/B signaling pathway potentially provides the means for pharmacological intervention of clinical prostate cancer progression. In addition, active STAT5A/B may serve as a prognostic marker for identification of those primary prostate cancers that are likely to progress to aggressive disease. In breast cancer, the role of STAT5A/B is more complex. STAT5A/B may have a dual role in the regulation of malignant mammary epithelium. Data accumulated from mouse models of breast cancer suggest that in early stages of breast cancer STAT5A/B may promote malignant transformation and enhance growth of the tumor. This is in contrast to established breast cancer, where STAT5A/B may mediate the critical cues for maintaining the differentiation of mammary epithelium. In addition, present data suggest that activation of STAT5A/B in breast cancer predicts favorable clinical outcome. The dual nature of STAT5A/B action in breast cancer makes the therapeutic use of STAT5 A/B more complex.
Collapse
Affiliation(s)
- Shyh-Han Tan
- Department of Cancer Biology, Kimmel Cancer Center, Thomas Jefferson University, 233 South 10th Street, BLSB 309, Philadelphia, Pennsylvania 19107, USA
| | | |
Collapse
|
38
|
Abdulghani J, Gu L, Dagvadorj A, Lutz J, Leiby B, Bonuccelli G, Lisanti MP, Zellweger T, Alanen K, Mirtti T, Visakorpi T, Bubendorf L, Nevalainen MT. Stat3 promotes metastatic progression of prostate cancer. THE AMERICAN JOURNAL OF PATHOLOGY 2008; 172:1717-28. [PMID: 18483213 DOI: 10.2353/ajpath.2008.071054] [Citation(s) in RCA: 187] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
There are currently no effective therapies for metastatic prostate cancer because the molecular mechanisms that underlie the metastatic spread of primary prostate cancer are unclear. Transcription factor Stat3 is constitutively active in malignant prostate epithelium, and its activation is associated with high histological grade and advanced cancer stage. In this work, we hypothesized that Stat3 stimulates metastatic progression of prostate cancer. We show that Stat3 is active in 77% of lymph node and 67% of bone metastases of clinical human prostate cancers. Importantly, adenoviral gene delivery of wild-type Stat3 (AdWTStat3) to DU145 human prostate cancer cells increased the number of lung metastases by 33-fold in an experimental metastasis assay compared with controls. Using various methods to inhibit Stat3, we demonstrated that Stat3 promotes human prostate cancer cell migration. Stat3 induced the formation of lamellipodia in both DU145 and PC-3 cells, further supporting the concept that Stat3 promotes a migratory phenotype of human prostate cancer cells. Moreover, Stat3 caused the rearrangement of cytoplasmic actin stress fibers and microtubules in both DU145 and PC-3 cells. Finally, inhibition of the Jak2 tyrosine kinase decreased both activation of Stat3 and prostate cancer cell motility. Collectively, these data indicate that transcription factor Stat3 is involved in metastatic behavior of human prostate cancer cells and may provide a therapeutic target to prevent metastatic spread of primary prostate cancer.
Collapse
Affiliation(s)
- Junaid Abdulghani
- Dept. of Cancer Biology, Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
A haplotype at chromosome Xq27.2 confers susceptibility to prostate cancer. Hum Genet 2008; 123:379-86. [PMID: 18350320 DOI: 10.1007/s00439-008-0486-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2008] [Accepted: 02/29/2008] [Indexed: 10/22/2022]
Abstract
We conducted an association study to identify risk variants for familial prostate cancer within the HPCX locus at Xq27 among Americans of Northern European descent. We investigated a total of 507 familial prostate cancer probands and 507 age-matched controls without a personal or family history of prostate cancer. The study population was subdivided into a set of training subjects to explore genetic variation of the locus potentially impacting risk of prostate cancer, and an independent set of test subjects to confirm the association and to assign significance, addressing multiple comparisons. We identified a 22.9 kb haplotype nominally associated with prostate cancer among training subjects (292 cases, 292 controls; chi(2) = 5.08, P = 0.020), that was confirmed among test subjects (215 cases, 215 controls; chi(2) = 3.73, P = 0.040). The haplotype predisposed to prostate cancer with an odds ratio of 3.41 (95% CI 1.04-11.17, P = 0.034) among test subjects. The haplotype extending from rs5907859 to rs1493189 is concordant with a prior study of the region within the Finnish founder population, and warrants further independent investigation.
Collapse
|
40
|
Ostrander EA, Johannesson B. Prostate cancer susceptibility loci: finding the genes. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2008; 617:179-90. [PMID: 18497042 DOI: 10.1007/978-0-387-69080-3_17] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/06/2022]
Abstract
Studies to date suggest that PC is a genetically very heterogeneous disease. High-risk families, in which multiple men are affected likely, reflect the contributions of a number of genes, some that are rare and highly penetrant, while others are more common and weakly penetrant. In this review, we have discussed only the first type of loci, and found that the identification of such genomic regions is a formidable problem. Replication between seemingly similar data sets is weak, likely reflecting the older age of onset associated with the disease, the inability to collect affected individuals from more than two generations in a family, and the variation seen in disease presentation, in addition to the underlying locus heterogeneity. Indeed, the definition of PC is ever changing, as diagnostic criteria and tools for pinpointing early lesions improve. Are we making progress? Clearly the answer is yes. The ability to divide large data sets into homogenous subset of families likely to share common genetic under-pinnings has improved power to identify loci and reproducibility between loci is now more common. Indeed, several groups report linkage to loci on chromosomes 1, 17, 19, and 22. Key to our continued success is our ever increasing ability to understand the disease. Identifying the subset of men who are likely to get clinically significant disease is the goal of genetic studies like these, and identifying the underlying loci is the key for developing diagnostics. The willingness of the community to work together has been an important factor in the successes the community has enjoyed to date, and will likely be as important as we move forward to untangle the genetics of this complex and common disorder.
Collapse
Affiliation(s)
- Elanie A Ostrander
- National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | | |
Collapse
|
41
|
Transcription Factors STAT5 and STAT3. Prostate Cancer 2008. [DOI: 10.1007/978-1-60327-079-3_12] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
|
42
|
Robbins C, Torres JB, Hooker S, Bonilla C, Hernandez W, Candreva A, Ahaghotu C, Kittles R, Carpten J. Confirmation study of prostate cancer risk variants at 8q24 in African Americans identifies a novel risk locus. Genome Res 2007; 17:1717-22. [PMID: 17978284 DOI: 10.1101/gr.6782707] [Citation(s) in RCA: 101] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Prostate cancer is a common complex disease that disproportionately affects men of African descent. Recently, several different common variants on chromosome 8q24 have been shown to be associated with prostate cancer in multiple studies and ethnic groups. The objective of this study was to confirm the association of 8q24 markers with prostate cancer in African Americans. We genotyped 24 markers along 8q24 and 80 unlinked ancestry informative markers in a hospital-based case-control sample of 1057 African American men (490 prostate cancer cases and 567 controls). Association analyses of 8q24 markers with prostate cancer risk were adjusted for both global and local 8q24 admixture stratification using estimates from ancestry informative markers. We report that rs7008482, which maps to the 8q24.13 region, is an additional independent prostate cancer risk variant (P = 5 x 10(-4)), and we also replicate the association of rs16901979 with prostate cancer (P = 0.002). Other published risk variants in the region such as rs1447295 and rs6983267 showed a similar direction and magnitude of effect, but were not significant in our population. Both rs7008482 and rs16901979 independently predicted risk and remained significant (P < 0.001) after controlling for each other. Our data combined with additional replications of 8q24 markers provide compelling support for multiple regions of risk for prostate cancer on 8q24.
Collapse
Affiliation(s)
- Christiane Robbins
- Division of Integrated Cancer Genomics, Translational Genomics Research Institute, Phoenix, Arizona 85004, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Kouprina N, Noskov VN, Solomon G, Otstot J, Isaacs W, Xu J, Schleutker J, Larionov V. Mutational analysis of SPANX genes in families with X-linked prostate cancer. Prostate 2007; 67:820-8. [PMID: 17373721 DOI: 10.1002/pros.20561] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
BACKGROUND Previous genetic linkage studies identified a locus for susceptibility to prostate cancer called HPCX at Xq27. The candidate region contains two clusters of SPANX genes. The first cluster called SPANX-A/D includes SPANX-A1, SPANX-A2, SPANX-B, SPANX-C, and SPANX-D; the second cluster called SPANX-N includes SPANX-N1, SPANX-N2, SPANX-N3, and SPANX-N4. The SPANX genes encode cancer-testis (CT) specific antigens. Previous studies identified SPANX-B and SPANX-D variants produced by gene conversion events, none of which are associated with X-linked prostate cancer. METHODS In this study we applied transformation-associated recombination cloning (TAR) in yeast to analyze sequence variations in SPANX-A1, SPANX-A2, and SPANX-C genes that are resided within large chromosomal duplications. A SPANX-N1/N4 cluster was analyzed by a routine PCR analysis. RESULTS None of the sequence variations in the coding regions of these genes is associated with susceptibility to prostate cancer. CONCLUSIONS Therefore, genetic variation in the SPANX genes is not the actual target variants explaining HPCX. However, it is possible that they play a modifying role in susceptibility to prostate cancer through complex recombinational interaction.
Collapse
Affiliation(s)
- Natalay Kouprina
- Laboratory of Molecular Pharmacology, National Cancer Institute, NIH, Bethesda, Maryland, USA.
| | | | | | | | | | | | | | | |
Collapse
|
44
|
Reynolds MA, Kastury K, Groskopf J, Schalken JA, Rittenhouse H. Molecular markers for prostate cancer. Cancer Lett 2007; 249:5-13. [PMID: 17303324 DOI: 10.1016/j.canlet.2006.12.029] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2006] [Accepted: 12/14/2006] [Indexed: 10/23/2022]
Abstract
Serum PSA testing has been used for over 20 years as an aid in the diagnosis and management of prostate cancer. Although highly sensitive, it suffers from a lack of specificity, showing elevated serum levels in a variety of other conditions including prostatitis, benign prostate hyperplasia, and non-cancerous neoplasia. During this period, numerous serum protein analytes have been investigated as alternative and/or supplemental tests for PSA, however in general these analytes have likewise suffered from a lack of specificity, often showing serum elevations in other clinical presentations. More recently, molecular assays targeting prostate disease at the DNA or RNA level have been investigated for potential diagnostic and prognostic utility. With the aid of modern genomics technologies, a variety of molecular biomarkers have been discovered that show potential for specific correlation with prostate cancer. Much of this discovery has been retrospective, using microdissected tissue from prostatectomy. The goal of current research is to apply genomic assays to noninvasive specimens such as blood and urine. Progress in this area is the subject of this review.
Collapse
Affiliation(s)
- Mark A Reynolds
- Gen-Probe Incorporated, 10210 Genetic Center Drive, San Diego, CA 92121, USA.
| | | | | | | | | |
Collapse
|
45
|
Baffoe-Bonnie AB, Kittles RA, Gillanders E, Ou L, George A, Robbins C, Ahaghotu C, Bennett J, Boykin W, Hoke G, Mason T, Pettaway C, Vijayakumar S, Weinrich S, Jones MP, Gildea D, Riedesel E, Albertus J, Moses T, Lockwood E, Klaric M, Faruque M, Royal C, Trent JM, Berg K, Collins FS, Furbert-Harris PM, Bailey-Wilson JE, Dunston GM, Powell I, Carpten JD. Genome-wide linkage of 77 families from the African American Hereditary Prostate Cancer study (AAHPC). Prostate 2007; 67:22-31. [PMID: 17031815 DOI: 10.1002/pros.20456] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
BACKGROUND The African American Hereditary Prostate Cancer (AAHPC) Study was designed to recruit families with early-onset disease fulfilling criteria of >or=4 affected. METHODS We present a approximately 10 cM genome-wide linkage (GWL) analysis on 77 families including 254 affected and 274 unaffected genotyped. RESULTS Linkage analysis revealed three chromosomal regions with GENEHUNTER multipoint HLOD scores >or=1.3 for all 77 families at 11q22, 17p11, and Xq21. One family yielded genome-wide significant evidence of linkage (LOD = 3.5) to the 17p11 region with seven other families >or=2.3 in this region. Twenty-nine families with no-male-to-male (MM) transmission gave a peak HLOD of 1.62 (alpha = 0.33) at the Xq21 locus. Two novel peaks >or=0.91 for the 16 families with '>6 affected' occurred at 2p21 and 22q12. CONCLUSIONS These chromosomal regions in the genome warrant further follow-up based on the hypothesis of multiple susceptibility genes with modest effects, or several major genes segregating in small subsets of families.
Collapse
|
46
|
Klein EA, Casey G, Silverman R. Genetic susceptibility and oxidative stress in prostate cancer: Integrated model with implications for prevention. Urology 2006; 68:1145-51. [PMID: 17169635 DOI: 10.1016/j.urology.2006.08.1074] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2005] [Revised: 04/25/2006] [Accepted: 08/15/2006] [Indexed: 11/18/2022]
Affiliation(s)
- Eric A Klein
- Section of Urologic Oncology, Glickman Urological Institute, Cleveland Clinic Foundation, Cleveland, Ohio 44122, USA.
| | | | | |
Collapse
|
47
|
Lange EM, Robbins CM, Gillanders EM, Zheng SL, Xu J, Wang Y, White KA, Chang BL, Ho LA, Trent JM, Carpten JD, Isaacs WB, Cooney KA. Fine-mapping the putative chromosome 17q21-22 prostate cancer susceptibility gene to a 10 cM region based on linkage analysis. Hum Genet 2006; 121:49-55. [PMID: 17120048 DOI: 10.1007/s00439-006-0274-2] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2006] [Accepted: 10/02/2006] [Indexed: 11/25/2022]
Abstract
Prostate cancer linkage studies have suggested the existence of a prostate cancer susceptibility gene on chromosome 17q21-22. We now report the results of an extended linkage analysis including 95 new multiplex prostate cancer families and 9 additional microsatellite markers resulting in a maximum LOD score of 2.99 at approximately 81-82 cM for all 453 pedigrees. Results from these 95 new pedigrees provide additional support for a chromosome 17q21-22 prostate cancer susceptibility gene. Inclusion of the 9 additional markers significantly reduced the size of the candidate region, as defined using a 1-LOD support interval, especially when focusing analyses on subsets of pedigrees with four or more confirmed affecteds or average age of diagnosis less than or equal to 65 years. A novel subset analysis of only those families (n = 147) that had four or more prostate cancer cases and an average age of prostate cancer diagnosis < or = 65 years results in a maximum LOD score of 5.49 at 78 cM with a 1-LOD support interval of 10 cM. This large set of pedigrees with four more prostate cancer cases characterized by early-onset disease will serve as a useful resource for identifying the putative 17q21-22 prostate cancer susceptibility gene.
Collapse
Affiliation(s)
- Ethan M Lange
- Department of Genetics, University of North Carolina, Chapel Hill, NC, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Schaid DJ, McDonnell SK, Zarfas KE, Cunningham JM, Hebbring S, Thibodeau SN, Eeles RA, Easton DF, Foulkes WD, Simard J, Giles GG, Hopper JL, Mahle L, Moller P, Badzioch M, Bishop DT, Evans C, Edwards S, Meitz J, Bullock S, Hope Q, Guy M, Hsieh CL, Halpern J, Balise RR, Oakley-Girvan I, Whittemore AS, Xu J, Dimitrov L, Chang BL, Adams TS, Turner AR, Meyers DA, Friedrichsen DM, Deutsch K, Kolb S, Janer M, Hood L, Ostrander EA, Stanford JL, Ewing CM, Gielzak M, Isaacs SD, Walsh PC, Wiley KE, Isaacs WB, Lange EM, Ho LA, Beebe-Dimmer JL, Wood DP, Cooney KA, Seminara D, Ikonen T, Baffoe-Bonnie A, Fredriksson H, Matikainen MP, Tammela TLJ, Bailey-Wilson J, Schleutker J, Maier C, Herkommer K, Hoegel JJ, Vogel W, Paiss T, Wiklund F, Emanuelsson M, Stenman E, Jonsson BA, Grönberg H, Camp NJ, Farnham J, Cannon-Albright LA, Catalona WJ, Suarez BK, Roehl KA. Pooled genome linkage scan of aggressive prostate cancer: results from the International Consortium for Prostate Cancer Genetics. Hum Genet 2006; 120:471-85. [PMID: 16932970 DOI: 10.1007/s00439-006-0219-9] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2006] [Accepted: 06/05/2006] [Indexed: 10/24/2022]
Abstract
While it is widely appreciated that prostate cancers vary substantially in their propensity to progress to a life-threatening stage, the molecular events responsible for this progression have not been identified. Understanding these molecular mechanisms could provide important prognostic information relevant to more effective clinical management of this heterogeneous cancer. Hence, through genetic linkage analyses, we examined the hypothesis that the tendency to develop aggressive prostate cancer may have an important genetic component. Starting with 1,233 familial prostate cancer families with genome scan data available from the International Consortium for Prostate Cancer Genetics, we selected those that had at least three members with the phenotype of clinically aggressive prostate cancer, as defined by either high tumor grade and/or stage, resulting in 166 pedigrees (13%). Genome-wide linkage data were then pooled to perform a combined linkage analysis for these families. Linkage signals reaching a suggestive level of significance were found on chromosomes 6p22.3 (LOD = 3.0), 11q14.1-14.3 (LOD = 2.4), and 20p11.21-q11.21 (LOD = 2.5). For chromosome 11, stronger evidence of linkage (LOD = 3.3) was observed among pedigrees with an average at diagnosis of 65 years or younger. Other chromosomes that showed evidence for heterogeneity in linkage across strata were chromosome 7, with the strongest linkage signal among pedigrees without male-to-male disease transmission (7q21.11, LOD = 4.1), and chromosome 21, with the strongest linkage signal among pedigrees that had African American ancestry (21q22.13-22.3; LOD = 3.2). Our findings suggest several regions that may contain genes which, when mutated, predispose men to develop a more aggressive prostate cancer phenotype. This provides a basis for attempts to identify these genes, with potential clinical utility for men with aggressive prostate cancer and their relatives.
Collapse
Affiliation(s)
- Daniel J Schaid
- Harwick 7, Mayo Clinic College of Medicine, Rochester, MN 55905, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
White KA, Lange EM, Ray AM, Wojno KJ, Cooney KA. Prohibitin mutations are uncommon in prostate cancer families linked to chromosome 17q. Prostate Cancer Prostatic Dis 2006; 9:298-302. [PMID: 16733518 DOI: 10.1038/sj.pcan.4500878] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
BACKGROUND Linkage studies have provided evidence for a prostate cancer susceptibility locus on chromosome 17q. The mitochondrial protein prohibitin (PHB) is a plausible candidate gene based on its chromosomal location (17q21) and known function. METHODS All coding regions and intron/exon junctions of the PHB gene were sequenced in 32 men from families participating in the University of Michigan Prostate Cancer Genetics Project that demonstrated evidence of linkage to 17q markers. RESULTS Although a number of nucleotide variants were identified, no coding region substitutions were identified in any of the 32 men with prostate cancer from 32 unrelated multiplex prostate cancer families. CONCLUSIONS PHB mutations do not appear to account for the linkage signal on 17q21-22 detected in PCGP families. Fine mapping of this region is in progress to refine the candidate region and highlight additional candidate prostate cancer susceptibility genes for sequence analysis.
Collapse
Affiliation(s)
- K A White
- Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI 48109-0946, USA
| | | | | | | | | |
Collapse
|
50
|
Kouprina N, Pavlicek A, Noskov VN, Solomon G, Otstot J, Isaacs W, Carpten JD, Trent JM, Schleutker J, Barrett JC, Jurka J, Larionov V. Dynamic structure of the SPANX gene cluster mapped to the prostate cancer susceptibility locus HPCX at Xq27. Genome Res 2006; 15:1477-86. [PMID: 16251457 PMCID: PMC1310635 DOI: 10.1101/gr.4212705] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
Genetic linkage studies indicate that germline variations in a gene or genes on chromosome Xq27-28 are implicated in prostate carcinogenesis. The linkage peak of prostate cancer overlies a region of approximately 750 kb containing five SPANX genes (SPANX-A1, -A2, -B, -C, and -D) encoding sperm proteins associated with the nucleus; their expression was also detected in a variety of cancers. SPANX genes are >95% identical and reside within large segmental duplications (SDs) with a high level of similarity, which confounds mutational analysis of this gene family by routine PCR methods. In this work, we applied transformation-associated recombination cloning (TAR) in yeast to characterize individual SPANX genes from prostate cancer patients showing linkage to Xq27-28 and unaffected controls. Analysis of genomic TAR clones revealed a dynamic nature of the replicated region of linkage. Both frequent gene deletion/duplication and homology-based sequence transfer events were identified within the region and were presumably caused by recombinational interactions between SDs harboring the SPANX genes. These interactions contribute to diversity of the SPANX coding regions in humans. We speculate that the predisposition to prostate cancer in X-linked families is an example of a genomic disease caused by a specific architecture of the SPANX gene cluster.
Collapse
Affiliation(s)
- Natalay Kouprina
- Laboratory of Biosystems and Cancer, National Cancer Institute, Bethesda, Maryland 20892, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|