1
|
Heise M, Jarzemski P, Nowak D, Bąk A, Junkiert-Czarnecka A, Pilarska-Deltow M, Borysiak M, Pilarska B, Haus O. Clinical Significance of Gene Mutations and Polymorphic Variants and their Association with Prostate Cancer Risk in Polish Men. Cancer Control 2022; 29:10732748211062342. [PMID: 35638715 PMCID: PMC9160909 DOI: 10.1177/10732748211062342] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Objectives: We tested the association of germline variants in BRCA1, BRCA2, CHEK2, CDKN2A, CYP1B1, HOXB13, MLH1, NBS1, NOD2 andPALB2 genes, as well as in 8q24 region, with prostate cancer (PC) risk and estimated their impact on disease clinical course, including overall survival time in Polish men with localized PC qualified for radical treatment.Materials and Methods: DNA of 110 patients with localized prostate cancer treated with radical prostatectomy (RP), from each age group and with different stages of the disease. DNA samples of the control group consisted of 111 men, volunteers, without PC (age-matched to study group). Sanger sequencing, AS-PCR, RFLP-PCR, and multiplex-PCR were used for variants detection.Results: The percentage of men with ≥1 germline variant was higher in PC group (52.7%) than in healthy men (37.8%) (P = .03). The presence of ≥2 variants was associated with shorter survival than the presence of one or no variant in the PC group (P = .14, trend). The HOXB13 G84E was detected in 2.9% of PC men and in no healthy men (P = .19, trend, OR = 7.21). A CHEK2 truncating mutation (1100delC or IVS2+1G>A) was detected in 2/110 (1.8%) PC patients and in no healthy men (P = .29, OR=5.14). The NBS1 I171V was detected in 2/110 (1.8%) PC patients and in no men from the control group (OR=5.14, P = .29, NS).Conclusions: We conclude that the presence of more than 2 germline variants was probably associated with shorter survival of patients with localized prostate cancer qualified for radical treatment. The HOXB13 (G84E), CHEK2 (1100delC or IVS2+1G>A) truncating variants and NBS1 (I171V) are associated with PC and hereditary form of the disease. The HOXB13 (G84E) and NOD2 (3020insC) single variants are associated with shorter and CYP1B1 (48CC, 119GG) single genotypes with longer overall survival.
Collapse
Affiliation(s)
- Marta Heise
- Faculty of Medicine, Department of Clinical Genetics, Collegium Medicum in Bydgoszcz, 49604Nicolaus Copernicus University in Toruń, Poland
| | - Piotr Jarzemski
- Faculty of Health Sciences, Department of Urology, Collegium Medicum in Bydgoszcz, 49604Nicolaus Copernicus University in Toruń, Jan Biziel University Hospital in Bydgoszcz, Poland
| | - Dagmara Nowak
- Faculty of Medicine, Department of Clinical Genetics, Collegium Medicum in Bydgoszcz, 49604Nicolaus Copernicus University in Toruń, Poland
| | - Aneta Bąk
- Faculty of Medicine, Department of Clinical Genetics, Collegium Medicum in Bydgoszcz, 49604Nicolaus Copernicus University in Toruń, Poland
| | - Anna Junkiert-Czarnecka
- Faculty of Medicine, Department of Clinical Genetics, Collegium Medicum in Bydgoszcz, 49604Nicolaus Copernicus University in Toruń, Poland
| | - Maria Pilarska-Deltow
- Faculty of Medicine, Department of Clinical Genetics, Collegium Medicum in Bydgoszcz, 49604Nicolaus Copernicus University in Toruń, Poland
| | - Maciej Borysiak
- Faculty of Health Sciences, Department of Urology, Collegium Medicum in Bydgoszcz, 49604Nicolaus Copernicus University in Toruń, Jan Biziel University Hospital in Bydgoszcz, Poland
| | - Beata Pilarska
- Faculty of Health Sciences, Department of Urology, Collegium Medicum in Bydgoszcz, 49604Nicolaus Copernicus University in Toruń, Jan Biziel University Hospital in Bydgoszcz, Poland
| | - Olga Haus
- Faculty of Medicine, Department of Clinical Genetics, Collegium Medicum in Bydgoszcz, 49604Nicolaus Copernicus University in Toruń, Poland
| |
Collapse
|
2
|
Kim EH, Cao D, Mahajan NP, Andriole GL, Mahajan K. ACK1-AR and AR-HOXB13 signaling axes: epigenetic regulation of lethal prostate cancers. NAR Cancer 2020; 2:zcaa018. [PMID: 32885168 PMCID: PMC7454006 DOI: 10.1093/narcan/zcaa018] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2020] [Revised: 07/22/2020] [Accepted: 08/13/2020] [Indexed: 12/24/2022] Open
Abstract
The androgen receptor (AR) is a critical transcription factor in prostate cancer (PC) pathogenesis. Its activity in malignant cells is dependent on interactions with a diverse set of co-regulators. These interactions fluctuate depending on androgen availability. For example, the androgen depletion increases the dependence of castration-resistant PCs (CRPCs) on the ACK1 and HOXB13 cell survival pathways. Activated ACK1, an oncogenic tyrosine kinase, phosphorylates cytosolic and nuclear proteins, thereby avoiding the inhibitory growth consequences of androgen depletion. Notably, ACK1-mediated phosphorylation of histone H4, which leads to epigenetic upregulation of AR expression, has emerged as a critical mechanism of CRPC resistance to anti-androgens. This resistance can be targeted using the ACK1-selective small-molecule kinase inhibitor (R)- 9b. CRPCs also deploy the bromodomain and extra-terminal domain protein BRD4 to epigenetically increase HOXB13 gene expression, which in turn activates the MYC target genes AURKA/AURKB. HOXB13 also facilitates ligand-independent recruitment of the AR splice variant AR-V7 to chromatin, compensating for the loss of the chromatin remodeling protein, CHD1, and restricting expression of the mitosis control gene HSPB8. These studies highlight the crosstalk between AR-ACK1 and AR-HOXB13 pathways as key mediators of CRPC recurrence.
Collapse
Affiliation(s)
- Eric H Kim
- Division of Urologic Surgery, Washington University in St. Louis, St. Louis, MO 63110, USA
| | - Dengfeng Cao
- Siteman Cancer Center, Washington University in St. Louis, St. Louis, MO 63110, USA
| | - Nupam P Mahajan
- Division of Urologic Surgery, Washington University in St. Louis, St. Louis, MO 63110, USA
| | - Gerald L Andriole
- Division of Urologic Surgery, Washington University in St. Louis, St. Louis, MO 63110, USA
| | - Kiran Mahajan
- Division of Urologic Surgery, Washington University in St. Louis, St. Louis, MO 63110, USA
| |
Collapse
|
3
|
Beebe-Dimmer JL, Kapron AL, Fraser AM, Smith KR, Cooney KA. Risk of Prostate Cancer Associated With Familial and Hereditary Cancer Syndromes. J Clin Oncol 2020; 38:1807-1813. [PMID: 32208047 DOI: 10.1200/jco.19.02808] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
PURPOSE Recently developed clinical guidelines suggest that men in families with specific cancer syndromes, such as hereditary breast and ovarian cancer (HBOC), consider genetic testing, especially in the setting of aggressive disease. However, although a family history (FH) of the same disease among close relatives is an established risk factor for prostate cancer (PC), a direct comparison of PC risk for men with each syndrome in a single population is needed. METHODS The Utah Population Database was used to identify 619,630 men, age ≥ 40 years, who were members of a pedigree that included at least 3 consecutive generations. Each man was evaluated for FH of hereditary PC (HPC), HBOC, and Lynch syndrome (LS) and for his own PC status. PC occurrences (N = 36,360) were classified into one or more subtypes: early onset (EO), lethal, and/or clinically significant. Relative risks (RRs) associated with each subtype, adjusted for important covariables, were calculated in STATA using a modified Poisson regression with robust error variances to obtain corresponding RR CIs for each FH definition. RESULTS An FH of HPC conveyed the greatest relative risk for all PC subtypes combined (RR, 2.30; 95% CI, 2.22 to 2.40), followed by HBOC and LS (both with 1 < RR < 2 and statistically significant). The strongest risks associated with FH were observed for EO disease in all pedigree types, consistent with the contribution of genetic factors to disease occurrence. CONCLUSION In this large, population-based, family database, the risk of PC varied by cancer FH and was most strongly associated with EO disease. These results are critically valuable in understanding and targeting high-risk populations that would benefit from genetic screening and enhanced surveillance.
Collapse
Affiliation(s)
- Jennifer L Beebe-Dimmer
- Department of Oncology, Wayne State University School of Medicine, Detroit, MI.,Barbara Ann Karmanos Cancer Institute, Detroit, MI
| | | | - Alison M Fraser
- University of Utah, Salt Lake City, UT.,Huntsman Cancer Institute, University of Utah, Salt Lake City, UT
| | - Ken R Smith
- University of Utah, Salt Lake City, UT.,Huntsman Cancer Institute, University of Utah, Salt Lake City, UT
| | | |
Collapse
|
4
|
The Genomic and Molecular Pathology of Prostate Cancer: Clinical Implications for Diagnosis, Prognosis, and Therapy. Adv Anat Pathol 2020; 27:11-19. [PMID: 31503032 DOI: 10.1097/pap.0000000000000245] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Prostate cancer (PCa) is the most common noncutaneous malignancy affecting American men and the second most common cause of cancer death. The traditional risk classification schemes for PCa are limited due to the vast clinical and molecular heterogeneity of the disease. Fortunately, recent advancements in sequencing technologies have provided us with valuable insight into the genomics of PCa. To date, a wide array of recurrent genomic alterations in PCa have been identified. Incorporating these distinct molecular subtypes of PCa into prediction models provides opportunities for improved risk stratification and ultimately better patient outcomes. In this review, we summarize the key molecular subtypes of PCa and focus on those genomic alterations that have clinical implications for diagnosis, prognosis, and therapeutic response.
Collapse
|
5
|
Pilarski R. The Role of BRCA Testing in Hereditary Pancreatic and Prostate Cancer Families. Am Soc Clin Oncol Educ Book 2019; 39:79-86. [PMID: 31099688 DOI: 10.1200/edbk_238977] [Citation(s) in RCA: 75] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Beyond breast and ovarian cancers, mutations in the BRCA1 and BRCA2 genes increase risks for pancreatic and prostate cancers and contribute to the prevalence of these cancers. Mutations in a number of other genes have also been shown to increase the risk for these cancers as well. Genetic testing is playing an increasingly important role in the treatment of patients with pancreatic and prostate cancer and is now recommended for all patients with pancreatic or metastatic prostate cancer, as well as patients with high Gleason grade prostate cancer and a remarkable family history. Identification of an inherited mutation can direct evaluation of the patient for other cancer risks as well as identification and management of disease in at-risk relatives. Growing evidence suggests improved responses to PARP inhibitors and other therapies in patients with mutations in the BRCA and other DNA repair genes. Although more work must be done to clarify the prevalence and penetrance of mutations in genes other than BRCA1 and BRCA2 in patients with pancreatic and prostate cancer, in most cases, testing is now being done with a panel of multiple genes. Because of the complexities in panel testing and the increased likelihood of finding variants of uncertain significance, pre- and post-test genetic counseling are essential.
Collapse
Affiliation(s)
- Robert Pilarski
- 1 Division of Human Genetics and Comprehensive Cancer Center, The Ohio State University, Columbus, OH
| |
Collapse
|
6
|
Mijuskovic M, Saunders EJ, Leongamornlert DA, Wakerell S, Whitmore I, Dadaev T, Cieza-Borrella C, Govindasami K, Brook MN, Haiman CA, Conti DV, Eeles RA, Kote-Jarai Z. Rare germline variants in DNA repair genes and the angiogenesis pathway predispose prostate cancer patients to develop metastatic disease. Br J Cancer 2018; 119:96-104. [PMID: 29915322 PMCID: PMC6035259 DOI: 10.1038/s41416-018-0141-7] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Revised: 05/01/2018] [Accepted: 05/17/2018] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Prostate cancer (PrCa) demonstrates a heterogeneous clinical presentation ranging from largely indolent to lethal. We sought to identify a signature of rare inherited variants that distinguishes between these two extreme phenotypes. METHODS We sequenced germline whole exomes from 139 aggressive (metastatic, age of diagnosis < 60) and 141 non-aggressive (low clinical grade, age of diagnosis ≥60) PrCa cases. We conducted rare variant association analyses at gene and gene set levels using SKAT and Bayesian risk index techniques. GO term enrichment analysis was performed for genes with the highest differential burden of rare disruptive variants. RESULTS Protein truncating variants (PTVs) in specific DNA repair genes were significantly overrepresented among patients with the aggressive phenotype, with BRCA2, ATM and NBN the most frequently mutated genes. Differential burden of rare variants was identified between metastatic and non-aggressive cases for several genes implicated in angiogenesis, conferring both deleterious and protective effects. CONCLUSIONS Inherited PTVs in several DNA repair genes distinguish aggressive from non-aggressive PrCa cases. Furthermore, inherited variants in genes with roles in angiogenesis may be potential predictors for risk of metastases. If validated in a larger dataset, these findings have potential for future clinical application.
Collapse
Affiliation(s)
- Martina Mijuskovic
- Oncogenetics, Division of Genetics and Epidemiology, The Institute of Cancer Research, London, SW7 3RP, UK
| | - Edward J Saunders
- Oncogenetics, Division of Genetics and Epidemiology, The Institute of Cancer Research, London, SW7 3RP, UK
| | - Daniel A Leongamornlert
- Oncogenetics, Division of Genetics and Epidemiology, The Institute of Cancer Research, London, SW7 3RP, UK
| | - Sarah Wakerell
- Oncogenetics, Division of Genetics and Epidemiology, The Institute of Cancer Research, London, SW7 3RP, UK
| | - Ian Whitmore
- Oncogenetics, Division of Genetics and Epidemiology, The Institute of Cancer Research, London, SW7 3RP, UK
| | - Tokhir Dadaev
- Oncogenetics, Division of Genetics and Epidemiology, The Institute of Cancer Research, London, SW7 3RP, UK
| | - Clara Cieza-Borrella
- Oncogenetics, Division of Genetics and Epidemiology, The Institute of Cancer Research, London, SW7 3RP, UK
| | - Koveela Govindasami
- Oncogenetics, Division of Genetics and Epidemiology, The Institute of Cancer Research, London, SW7 3RP, UK
| | - Mark N Brook
- Oncogenetics, Division of Genetics and Epidemiology, The Institute of Cancer Research, London, SW7 3RP, UK
| | - Christopher A Haiman
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California/Norris Comprehensive Cancer Center, Los Angeles, CA, 90015, USA
| | - David V Conti
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California/Norris Comprehensive Cancer Center, Los Angeles, CA, 90015, USA
| | - Rosalind A Eeles
- Oncogenetics, Division of Genetics and Epidemiology, The Institute of Cancer Research, London, SW7 3RP, UK
- The Royal Marsden NHS Foundation Trust, London, SW3 6JJ, UK
| | - Zsofia Kote-Jarai
- Oncogenetics, Division of Genetics and Epidemiology, The Institute of Cancer Research, London, SW7 3RP, UK.
| |
Collapse
|
7
|
Prostate Cancer Genomics: Recent Advances and the Prevailing Underrepresentation from Racial and Ethnic Minorities. Int J Mol Sci 2018; 19:ijms19041255. [PMID: 29690565 PMCID: PMC5979433 DOI: 10.3390/ijms19041255] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2018] [Revised: 04/15/2018] [Accepted: 04/15/2018] [Indexed: 02/07/2023] Open
Abstract
Prostate cancer (CaP) is the most commonly diagnosed non-cutaneous cancer and the second leading cause of male cancer deaths in the United States. Among African American (AA) men, CaP is the most prevalent malignancy, with disproportionately higher incidence and mortality rates. Even after discounting the influence of socioeconomic factors, the effect of molecular and genetic factors on racial disparity of CaP is evident. Earlier studies on the molecular basis for CaP disparity have focused on the influence of heritable mutations and single-nucleotide polymorphisms (SNPs). Most CaP susceptibility alleles identified based on genome-wide association studies (GWAS) were common, low-penetrance variants. Germline CaP-associated mutations that are highly penetrant, such as those found in HOXB13 and BRCA2, are usually rare. More recently, genomic studies enabled by Next-Gen Sequencing (NGS) technologies have focused on the identification of somatic mutations that contribute to CaP tumorigenesis. These studies confirmed the high prevalence of ERG gene fusions and PTEN deletions among Caucasian Americans and identified novel somatic alterations in SPOP and FOXA1 genes in early stages of CaP. Individuals with African ancestry and other minorities are often underrepresented in these large-scale genomic studies, which are performed primarily using tumors from men of European ancestry. The insufficient number of specimens from AA men and other minority populations, together with the heterogeneity in the molecular etiology of CaP across populations, challenge the generalizability of findings from these projects. Efforts to close this gap by sequencing larger numbers of tumor specimens from more diverse populations, although still at an early stage, have discovered distinct genomic alterations. These research findings can have a direct impact on the diagnosis of CaP, the stratification of patients for treatment, and can help to address the disparity in incidence and mortality of CaP. This review examines the progress of understanding in CaP genetics and genomics and highlight the need to increase the representation from minority populations.
Collapse
|
8
|
Impact of the G84E variant on HOXB13 gene and protein expression in formalin-fixed, paraffin-embedded prostate tumours. Sci Rep 2017; 7:17778. [PMID: 29259341 PMCID: PMC5736598 DOI: 10.1038/s41598-017-18217-w] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2017] [Accepted: 12/05/2017] [Indexed: 01/01/2023] Open
Abstract
The HOXB13 G84E variant is associated with risk of prostate cancer (PCa), however the role this variant plays in PCa development is unknown. This study examined 751 cases, 450 relatives and 355 controls to determine the contribution of this variant to PCa risk in Tasmania and investigated HOXB13 gene and protein expression in tumours from nine G84E heterozygote variant and 13 wild-type carriers. Quantitative PCR and immunohistochemistry showed that HOXB13 gene and protein expression did not differ between tumour samples from variant and wild-type carriers. Allele-specific transcription revealed that two of seven G84E carriers transcribed both the variant and wild-type allele, while five carriers transcribed the wild-type allele. Methylation of surrounding CpG sites was lower in the variant compared to the wild-type allele, however overall methylation across the region was very low. Notably, tumour characteristics were less aggressive in the two variant carriers that transcribed the variant allele compared to the five that did not. This study has shown that HOXB13 expression does not differ between tumour tissue of G84E variant carriers and non-carriers. Intriguingly, the G84E variant allele was rarely transcribed in carriers, suggesting that HOXB13 expression may be driven by the wild-type allele in the majority of carriers.
Collapse
|
9
|
Lotan TL, Torres A, Zhang M, Tosoian JJ, Guedes LB, Fedor H, Hicks J, Ewing CM, Isaacs SD, Johng D, De Marzo AM, Isaacs WB. Somatic molecular subtyping of prostate tumors from HOXB13 G84E carriers. Oncotarget 2017; 8:22772-22782. [PMID: 28186998 PMCID: PMC5410261 DOI: 10.18632/oncotarget.15196] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2016] [Accepted: 01/21/2017] [Indexed: 11/25/2022] Open
Abstract
A recurrent germline mutation (G84E) in the HOXB13 gene is associated with early onset and family history-positive prostate cancer in patients of European descent, occurring in up to 5% of prostate cancer families. To date, the molecular features of prostate tumors occurring in HOXB13 G84E carriers have not been studied in a large cohort of patients. We identified 101 heterozygous carriers of G84E who underwent radical prostatectomy for prostate cancer between 1985 and 2011 and matched these men by race, age and tumor grade to 99 HOXB13 wild-type controls. Immunostaining for HOXB13, PTEN, ERG, p53 and SPINK1 as well as RNA in situ hybridization for ETV1/4/5 were performed using genetically validated assays. Tumors from G84E carriers generally expressed HOXB13 protein at a level comparable to benign and wild-type glands. ETS gene expression (either ERG or ETV1/4/5) was seen in 36% (36/101) of tumors from G84E carriers compared to 68% (65/96) of the controls (p < 0.0001). PTEN was lost in 11% (11/101) of G84E carriers compared to 25% (25/99) of the controls (p = 0.014). PTEN loss was enriched among ERG-positive compared to ERG-negative tumors in both groups of patients. Nuclear accumulation of the p53 protein, indicative of underlying TP53 missense mutations, was uncommon in both groups, occurring in 1% (1/101) of the G84E carriers versus 2% (2/92) of the controls (p = NS). Taken together, these data suggest that genes other than ERG and PTEN may drive carcinogenesis/progression in the majority of men with germline HOXB13 mutations.
Collapse
Affiliation(s)
- Tamara L Lotan
- Departments of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Departments of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Alba Torres
- Departments of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Miao Zhang
- Departments of Pathology, MD Anderson Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Jeffrey J Tosoian
- Departments of Brady Urological Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Liana B Guedes
- Departments of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Helen Fedor
- Departments of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Jessica Hicks
- Departments of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Charles M Ewing
- Departments of Brady Urological Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Sarah D Isaacs
- Departments of Brady Urological Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Dorhyun Johng
- Departments of Brady Urological Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Angelo M De Marzo
- Departments of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Departments of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Departments of Brady Urological Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - William B Isaacs
- Departments of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Departments of Brady Urological Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
10
|
Affiliation(s)
- Kathleen A Cooney
- Departments of Internal Medicine and Urology, The University of Michigan, Comprehensive Cancer Center, Ann Arbor
| | - Jennifer L Beebe-Dimmer
- Department of Oncology, Wayne State University School of Medicine, Karmanos Cancer Institute, Detroit, MI, USA
| |
Collapse
|
11
|
Abstract
Prostate cancer is the most commonly diagnosed cancer among men in the United States as well as most Western countries. A significant proportion of men report having a positive family history of prostate cancer in a first-degree relative (father, brother, son), which is important in that family history is one of the only established risk factors for the disease and plays a role in decision-making for prostate cancer screening. Familial aggregation of prostate cancer is considered a surrogate marker of genetic susceptibility to developing the disease, but shared environment cannot be excluded as an explanation for clustering of cases among family members. Prostate cancer is both a clinically and genetically heterogeneous disease with inherited factors predicted to account for 40%-50% of cases, comprised of both rare highly to moderately penetrant gene variants, as well as common genetic variants of low penetrance. Most notably, HOXB13 and BRCA2 mutations have been consistently shown to increase prostate cancer risk, and are more commonly observed among patients diagnosed with early-onset disease. A recurrent mutation in HOXB13 has been shown to predispose to hereditary prostate cancer (HPC), and BRCA2 mutations to hereditary breast and ovarian cancer (HBOC). Genome-wide association studies (GWAS) have also identified approximately 100 loci that associate with modest (odds ratios <2.0) increases in prostate cancer risk, only some of which have been replicated in subsequent studies. Despite these efforts, genetic testing in prostate cancer lags behind other common tumors like breast and colorectal cancer. To date, National Comprehensive Cancer Network (NCCN) guidelines have highly selective criteria for BRCA1/2 testing for men with prostate cancer based on personal history and/or specific family cancer history. Tumor sequencing is also leading to the identification of germline mutations in prostate cancer patients, informing the scope of inheritance. Advances in genetic testing for inherited and familial prostate cancer (FPC) are needed to inform personalized cancer risk screening and treatment approaches.
Collapse
Affiliation(s)
- Veda N Giri
- Cancer Risk Assessment and Clinical Cancer Genetics Program, Division of Population Science, Department of Medical Oncology, Center of Excellence for Cancer Risk, Prevention, and Control Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA.
| | - Jennifer L Beebe-Dimmer
- Barbara Ann Karmanos Cancer Institute, Wayne State University School of Medicine Department of Oncology, Detroit, MI
| |
Collapse
|
12
|
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
|
13
|
Padmanabhan A, Rao V, De Marzo AM, Bieberich CJ. Regulating NKX3.1 stability and function: Post-translational modifications and structural determinants. Prostate 2016; 76:523-33. [PMID: 26841725 DOI: 10.1002/pros.23144] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2015] [Accepted: 12/15/2015] [Indexed: 01/22/2023]
Abstract
BACKGROUND The androgen-regulated homeodomain transcription factor NKX3.1 plays roles in early prostate development and functions as a prostate-specific tumor suppressor. Decreased expression of NKX3.1 protein is common in primary prostate cancer. Discordance between NKX3.1 mRNA and protein levels during prostate carcinogenesis suggested a key role for post-transcriptional modifications in regulating NKX3.1 protein levels in prostate epithelial cells. Subsequent studies revealed NKX3.1 to be modified post-translationally at multiple sites. METHODS We reviewed published literature to identify and summarize post-translational modifications and structural elements critical in regulating NKX3.1 stability and levels in prostate epithelial cells. RESULTS NKX3.1 is modified post-translationally at multiple sites by different protein kinases. These modifications together with several structural determinants were identified to play an important role in NKX3.1 stability and biology. CONCLUSIONS In this review, we provide a comprehensive overview of the known post-translational modifications and structural features that impact NKX3.1. Defining factors that regulate NKX3.1 in prostate epithelial cells will extend our understanding of molecular changes that may contribute to prostate cancer initiation and progression.
Collapse
Affiliation(s)
- Achuth Padmanabhan
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, Connecticut
| | - Varsha Rao
- Department of Genetics, Stanford University, Palo Alto, California
| | - Angelo M De Marzo
- Departments of Pathology, Oncology and Urology, The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins and the Brady Urological Research Institute at Johns Hopkins, The Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Charles J Bieberich
- Department of Biological Sciences, University of Maryland Baltimore County, Baltimore, Maryland
| |
Collapse
|
14
|
Diler SB, Öden A. The T-786C, G894T, and intron 4 VNTR (4a/b) polymorphisms of the endothelial nitric oxide synthase gene in prostate cancer cases. RUSS J GENET+ 2016. [DOI: 10.1134/s1022795416020022] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
15
|
Storebjerg TM, Høyer S, Kirkegaard P, Bro F, Ørntoft TF, Borre M, Sørensen KD. Prevalence of the HOXB13 G84E mutation in Danish men undergoing radical prostatectomy and its correlations with prostate cancer risk and aggressiveness. BJU Int 2016; 118:646-53. [PMID: 26779768 DOI: 10.1111/bju.13416] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
OBJECTIVES To determine the prevalence of the HOXB13 G84E mutation (rs138213197) in Danish men with or without prostate cancer (PCa) and to investigate possible correlations between HOXB13 mutation status and clinicopathological characteristics associated with tumour aggressiveness. MATERIALS AND METHODS We conducted a case-control study including 995 men with PCa (cases) who underwent radical prostatectomy (RP) between 1997 and 2011 at the Department of Urology, Aarhus University Hospital, Denmark. As controls, we used 1622 healthy men with a normal prostate specific antigen (PSA) level. RESULTS The HOXB13 G84E mutation was identified in 0.49% of controls and in 2.51% of PCa cases. The mutation was associated with a 5.12-fold increased relative risk (RR) of PCa (95% confidence interval [CI] 2.26-13.38; P = 13 × 10(-6) ). Furthermore, carriers of the risk allele were significantly more likely to have a higher PSA level at diagnosis (mean PSA 19.9 vs 13.6 ng/mL; P = 0.032), a pathological Gleason score ≥7 (83.3 vs 60.9%; P = 0.032), and positive surgical margins (56.0 vs 28.5%; P = 0.006) than non-carriers. Risk allele carriers were also more likely to have aggressive disease (54.2 vs 28.6%; P = 0.011), as defined by a preoperative PSA ≥20 ng/mL, pathological Gleason score ≥ (4+3) and/or presence of regional/distant disease. At a mean follow-up of 7 months, we found no significant association between HOXB13 mutation status and biochemical recurrence in this cohort of men who underwent RP. CONCLUSIONS This is the first study to investigate the HOXB13 G84E mutation in Danish men. The mutation was detected in 0.49% of controls and in 2.51% of cases, and was associated with 5.12-fold increased RR of being diagnosed with PCa. In our RP cohort, HOXB13 mutation carriers were more likely to develop aggressive PCa. Further studies are needed to assess the potential of HOXB13 for future targeted screening approaches.
Collapse
Affiliation(s)
- Tine M Storebjerg
- Department of Urology, Aarhus University Hospital, Aarhus, Denmark.,Department of Pathology, Aarhus University Hospital, Aarhus, Denmark.,Department of Molecular Medicine, Aarhus University Hospital, Aarhus, Denmark
| | - Søren Høyer
- Department of Pathology, Aarhus University Hospital, Aarhus, Denmark
| | - Pia Kirkegaard
- Research Unit for General Practice and Research Centre for Cancer Diagnosis in Primary Care, Aarhus University, Aarhus, Denmark
| | - Flemming Bro
- Research Unit for General Practice and Research Centre for Cancer Diagnosis in Primary Care, Aarhus University, Aarhus, Denmark
| | | | - Torben F Ørntoft
- Department of Molecular Medicine, Aarhus University Hospital, Aarhus, Denmark
| | - Michael Borre
- Department of Urology, Aarhus University Hospital, Aarhus, Denmark
| | - Karina D Sørensen
- Department of Molecular Medicine, Aarhus University Hospital, Aarhus, Denmark.
| |
Collapse
|
16
|
Falchook AD, Martin NE, Basak R, Smith AB, Milowsky MI, Chen RC. Stage at presentation and survival outcomes of patients with Gleason 8-10 prostate cancer and low prostate-specific antigen. Urol Oncol 2015; 34:119.e19-26. [PMID: 26526383 DOI: 10.1016/j.urolonc.2015.09.014] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2015] [Revised: 09/21/2015] [Accepted: 09/23/2015] [Indexed: 11/17/2022]
Abstract
OBJECTIVE To evaluate outcomes for men with high Gleason score and low prostate-specific antigen (PSA) prostate cancer. Low PSA levels among men with Gleason 8-10 prostate cancer may be owing to cellular dedifferentiation rather than low disease burden. We hypothesized that men with Gleason 8-10 prostate cancer and low PSA levels have increased risk for advanced disease and worse survival. MATERIALS AND METHODS Men diagnosed from 2004 to 2007 with Gleason 8-10 prostate adenocarcinoma in the National Cancer Data Base were included. Patients were stratified by PSA levels at diagnosis: 0.1 to 3.9, 4.0 to 9.9, 10.0 to 19.9, and≥20.0ng/ml. Outcomes were clinical TNM category, pathologic stage (for prostatectomy patients), and overall survival (OS). Kaplan-Meier analysis and Cox proportional hazards models were used. RESULTS A total of 37,283 patients were included. Men with PSA levels of<4.0ng/ml were more likely than those with PSA levels of 4 to 9.9ng/ml to present with clinical T3-4 disease (15% vs. 10%, P<0.001), nodal (4% vs. 2%, P<0.001) and distant (5% vs. 3%, P<0.001) metastasis. However, among patients treated with prostatectomy, lower PSA levels were not associated with increased likelihood of pathologic T3-4 disease or nodal metastasis. Six-year OS was 89.1% (PSA: 0.1-3.9ng/ml) vs. 91.0% (PSA: 4.0-9.9ng/ml) for prostatectomy (log-rank P<0.001), and 75.8% vs. 81.0% for radiotherapy (P<0.001). Multivariable analyses showed OS of patients with PSA levels of 0.1 to 3.9ng/ml to be similar to those with PSA levels of 10 to 19.9ng/ml. CONCLUSIONS Patients with Gleason 8-10 cancer and PSA levels of<4.0ng/ml have more aggressive disease than those with PSA levels of 4 to 9.9ng/ml; these low PSA cancers behave more like those with PSA levels of 10 to 19.9ng/ml. Further study is needed to evaluate potential biological differences in these patients with low PSA-producing cancers.
Collapse
Affiliation(s)
- Aaron D Falchook
- Department of Radiation Oncology, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Neil E Martin
- Department of Radiation Oncology, Dana-Farber Cancer Institute and Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Ramsankar Basak
- Department of Radiation Oncology, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Angela B Smith
- Department of Urology, University of North Carolina at Chapel Hill, Chapel Hill, NC; University of North Carolina-Lineberger Comprehensive Cancer Center, Chapel Hill, NC
| | - Matthew I Milowsky
- University of North Carolina-Lineberger Comprehensive Cancer Center, Chapel Hill, NC; Department of Medicine, Division of Hematology/Oncology, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Ronald C Chen
- Department of Radiation Oncology, University of North Carolina at Chapel Hill, Chapel Hill, NC; University of North Carolina-Lineberger Comprehensive Cancer Center, Chapel Hill, NC; Sheps Center for Health Services Research, University of North Carolina at Chapel Hill, Chapel Hill, NC.
| |
Collapse
|
17
|
Randazzo M, Müller A, Carlsson S, Eberli D, Huber A, Grobholz R, Manka L, Mortezavi A, Sulser T, Recker F, Kwiatkowski M. A positive family history as a risk factor for prostate cancer in a population-based study with organised prostate-specific antigen screening: results of the Swiss European Randomised Study of Screening for Prostate Cancer (ERSPC, Aarau). BJU Int 2015; 117:576-83. [PMID: 26332304 DOI: 10.1111/bju.13310] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
OBJECTIVE To assess the value of a positive family history (FH) as a risk factor for prostate cancer incidence and grade among men undergoing organised prostate-specific antigen (PSA) screening in a population-based study. SUBJECTS AND METHODS The study cohort comprised all attendees of the Swiss arm of the European Randomised Study of Screening for Prostate Cancer (ERSPC) with systematic PSA level tests every 4 years. Men reporting first-degree relative(s) diagnosed with prostate cancer were considered to have a positive FH. Biopsy was exclusively PSA triggered at a PSA level threshold of 3 ng/mL. The primary endpoint was prostate cancer diagnosis. Kaplan-Meier and Cox regression analyses were used. RESULTS Of 4 932 attendees with a median (interquartile range, IQR) age of 60.9 (57.6-65.1) years, 334 (6.8%) reported a positive FH. The median (IQR) follow-up duration was 11.6 (10.3-13.3) years. Cumulative prostate cancer incidence was 60/334 (18%, positive FH) and 550/4 598 (12%, negative FH) [odds ratio 1.6, 95% confidence interval (CI) 1.2-2.2, P = 0.001). In both groups, most prostate cancer diagnosed was low grade. There were no significant differences in PSA level at diagnosis, biopsy Gleason score or Gleason score on pathological specimen among men who underwent radical prostatectomy between both groups. On multivariable analysis, age (hazard ratio [HR] 1.04, 95% CI 1.02-1.06), baseline PSA level (HR 1.13, 95% CI 1.12-1.14), and FH (HR 1.6, 95% CI 1.24-2.14) were independent predictors for overall prostate cancer incidence (all P < 0.001). Only baseline PSA level (HR 1.14, 95% CI 1.12-1.16, P < 0.001) was an independent predictor of Gleason score ≥7 prostate cancer on prostate biopsy. The proportion of interval prostate cancer diagnosed in-between the screening rounds was not significantly different. CONCLUSION Irrespective of the FH status, the current PSA-based screening setting detects the majority of aggressive prostate cancers and missed only a minority of interval cancers with a 4-year screening algorithm. Our results suggest that men with a positive FH are at increased risk of low-grade but not aggressive prostate cancer.
Collapse
Affiliation(s)
- Marco Randazzo
- Department of Urology, University Hospital Zürich, Zürich, Switzerland.,Department of Urology, Cantonal Hospital Aarau, Aarau, Switzerland
| | - Alexander Müller
- Department of Urology, University Hospital Zürich, Zürich, Switzerland
| | - Sigrid Carlsson
- Department of Surgery (Urology Service), Memorial Sloan Kettering Cancer Center, New York, NY, USA.,Department of Urology, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Daniel Eberli
- Department of Urology, University Hospital Zürich, Zürich, Switzerland
| | - Andreas Huber
- Department of Laboratory Medicine, Cantonal Hospital Aarau, Aarau, Switzerland
| | - Rainer Grobholz
- Department of Pathology, Cantonal Hospital Aarau, Aarau, Switzerland
| | - Lukas Manka
- Department of Urology, Academic Hospital Braunschweig, Braunschweig, Germany
| | - Ashkan Mortezavi
- Department of Urology, University Hospital Zürich, Zürich, Switzerland
| | - Tullio Sulser
- Department of Urology, University Hospital Zürich, Zürich, Switzerland
| | - Franz Recker
- Department of Urology, Cantonal Hospital Aarau, Aarau, Switzerland
| | - Maciej Kwiatkowski
- Department of Urology, Cantonal Hospital Aarau, Aarau, Switzerland.,Department of Urology, Academic Hospital Braunschweig, Braunschweig, Germany
| |
Collapse
|
18
|
Beebe-Dimmer JL, Hathcock M, Yee C, Okoth LA, Ewing CM, Isaacs WB, Cooney KA, Thibodeau SN. The HOXB13 G84E Mutation Is Associated with an Increased Risk for Prostate Cancer and Other Malignancies. Cancer Epidemiol Biomarkers Prev 2015; 24:1366-72. [PMID: 26108461 DOI: 10.1158/1055-9965.epi-15-0247] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2015] [Accepted: 06/11/2015] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND A rare nonconservative substitution (G84E) in the HOXB13 gene has been shown to be associated with risk of prostate cancer. DNA samples from male patients included in the Mayo Clinic Biobank (MCB) were genotyped to determine the frequency of the G84E mutation and its association with various cancers. METHODS Subjects were genotyped using a custom TaqMan (Applied Biosystems) assay for G84E (rs138213197). In addition to donating a blood specimen, all MCB participants completed a baseline questionnaire to collect information on medical history and family history of cancer. RESULTS Forty-nine of 9,012 male patients were carriers of G84E (0.5%). Thirty-one percent (n = 2,595) of participants had been diagnosed with cancer, including 51.1% of G84E carriers compared with just 30.6% of noncarriers (P = 0.004). G84E was most frequently observed among men with prostate cancer compared with men without cancer (P < 0.0001). However, the mutation was also more commonly observed in men with bladder cancer (P = 0.06) and leukemia (P = 0.01). G84E carriers were more likely to have a positive family history of prostate cancer in a first-degree relative compared to noncarriers (36.2% vs. 16.0%, P = 0.0003). CONCLUSIONS Our study confirms the association between the HOXB13 G84E variant and prostate cancer and suggests a novel association between G84E and leukemia and a suggestive association with bladder cancer. Future investigation is warranted to confirm these associations in order to improve our understanding of the role of germline HOXB13 mutations in human cancer. IMPACT The associations between HOXB13 and prostate, leukemia, and bladder suggest that this gene is important in carcinogenesis.
Collapse
Affiliation(s)
- Jennifer L Beebe-Dimmer
- Department of Oncology, Wayne State University School of Medicine, Detroit, Michigan. Barbara Ann Karmanos Cancer Institute, Detroit, Michigan.
| | - Matthew Hathcock
- Department of Health Science Research, Mayo Clinic, Rochester, Minnesota
| | - Cecilia Yee
- Department of Oncology, Wayne State University School of Medicine, Detroit, Michigan. Barbara Ann Karmanos Cancer Institute, Detroit, Michigan
| | - Linda A Okoth
- Departments of Internal Medicine and Urology, University of Michigan School of Medicine, Ann Arbor, Michigan. University of Michigan Comprehensive Cancer Center, Ann Arbor, Michigan
| | - Charles M Ewing
- Department of Urology, Brady Urological Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - William B Isaacs
- Department of Urology, Brady Urological Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Kathleen A Cooney
- Departments of Internal Medicine and Urology, University of Michigan School of Medicine, Ann Arbor, Michigan. University of Michigan Comprehensive Cancer Center, Ann Arbor, Michigan
| | - Stephen N Thibodeau
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota
| |
Collapse
|
19
|
Kote-Jarai Z, Mikropoulos C, Leongamornlert DA, Dadaev T, Tymrakiewicz M, Saunders EJ, Jones M, Jugurnauth-Little S, Govindasami K, Guy M, Hamdy FC, Donovan JL, Neal DE, Lane JA, Dearnaley D, Wilkinson RA, Sawyer EJ, Morgan A, Antoniou AC, Eeles RA. Prevalence of the HOXB13 G84E germline mutation in British men and correlation with prostate cancer risk, tumour characteristics and clinical outcomes. Ann Oncol 2015; 26:756-761. [PMID: 25595936 DOI: 10.1093/annonc/mdv004] [Citation(s) in RCA: 74] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
BACKGROUND A rare recurrent missense variant in HOXB13 (rs138213197/G84E) was recently reported to be associated with hereditary prostate cancer. Population-based studies have established that, since the frequency of this single-nucleotide polymorphism (SNP) varies between geographic regions, the associated proportion of prostate cancer (PrCa) risk contribution is also highly variable by country. PATIENTS AND METHODS This is the largest comprehensive case-control study assessing the prevalence of the HOXB13 G84E variant to date and is the first in the UK population. We genotyped 8652 men diagnosed with PrCa within the UK Genetic Prostate Cancer Study (UKGPCS) and 5252 healthy men from the UK ProtecT study. RESULTS HOXB13 G84E was identified in 0.5% of the healthy controls and 1.5% of the PrCa cases, and it was associated with a 2.93-fold increased risk of PrCa [95% confidence interval (CI) 1.94-4.59; P = 6.27 × 10(-8)]. The risk was even higher among men with family history of PrCa [odds ratio (OR) = 4.53, 95% CI 2.86-7.34; P = 3.1 × 10(-8)] and in young-onset PrCa (diagnosed up to the age of 55 years; OR = 3.11, 95% CI 1.98-5.00; P = 6.1 × 10(-7)). There was no significant association between Gleason Score, presenting prostate specific antigen, tumour-node-metastasis (TNM) stage or NCCN risk group and carrier status. HOXB13 G84E was not associated with overall or cancer-specific survival. We found that the polygenic PrCa risk score (PR score), calculated using the 71 known single-nucleotide polymorphisms (SNPs) associated with PrCa and the HOXB13 G84E variant act multiplicatively on PrCa risk. Based on the estimated prevalence and risk, this rare variant explains ∼1% of the familial risk of PrCa in the UK population. CONCLUSIONS The clinical importance of HOXB13 G84E in PrCa management has not been established. This variant was found to have no effect on prognostic implications but could be used for stratifying screening, by identifying men at high risk. CLINICAL TRIALS NUMBERS Prostate Testing for Cancer and Treatment (ProtecT): NCT02044172. UK GENETIC PROSTATE CANCER STUDY Epidemiology and Molecular Genetics Studies (UKGPCS): NCT01737242.
Collapse
Affiliation(s)
| | | | | | - T Dadaev
- Institute of Cancer Research, London
| | | | | | - M Jones
- Institute of Cancer Research, London
| | | | | | - M Guy
- Institute of Cancer Research, London
| | - F C Hamdy
- Nuffield Department of Surgical Sciences, University of Oxford, Oxford; Faculty of Medical Science, University of Oxford, John Radcliffe Hospital, Oxford
| | - J L Donovan
- School of Social and Community Medicine, University of Bristol, Bristol
| | - D E Neal
- Surgical Oncology (Uro-Oncology: S4), University of Cambridge, Addenbrooke's Hospital, Cambridge; Cancer Research UK, Cambridge Research Institute, Li Ka Shing Centre, Cambridge
| | - J A Lane
- Surgical Oncology (Uro-Oncology: S4), University of Cambridge, Addenbrooke's Hospital, Cambridge; Cancer Research UK, Cambridge Research Institute, Li Ka Shing Centre, Cambridge
| | | | | | | | - A Morgan
- Institute of Cancer Research, London
| | - A C Antoniou
- Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, University of Cambridge, Strangeways Laboratory, Worts Causeway, Cambridge
| | - R A Eeles
- Institute of Cancer Research, London; The Royal Marsden NHS Foundation Trust, London, UK
| |
Collapse
|
20
|
Zuhlke KA, Johnson AM, Tomlins SA, Palanisamy N, Carpten JD, Lange EM, Isaacs WB, Cooney KA. Identification of a novel germline SPOP mutation in a family with hereditary prostate cancer. Prostate 2014; 74:983-90. [PMID: 24796539 PMCID: PMC4230298 DOI: 10.1002/pros.22818] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2014] [Accepted: 04/11/2014] [Indexed: 11/12/2022]
Abstract
BACKGROUND Family history of prostate cancer is a well-recognized risk factor. Previous linkage studies have reported a putative prostate cancer susceptibility locus at chromosome 17q21-22. SPOP (Speckle-type POZ protein) maps to the 17q21-22 candidate linkage region and is one of the most frequently mutated genes in sporadic prostate cancers. METHODS We performed targeted next generation sequencing to analyze 2009 exons from 202 genes in a candidate linkage region on chromosome 17q21-22 using 94 unrelated familial prostate cancer cases from the University of Michigan Prostate Cancer Genetics Project (n=54) and Johns Hopkins University (n=40) including the exons and UTRs of SPOP. RESULTS We identified a novel SPOP missense mutation (N296I) in a man with prostate cancer diagnosed at age 43. This mutation completely segregates with prostate cancer affection status among the men in this family. The N296I mutation resides within the evolutionarily conserved Bric-a-brac, Tramtrack, Broad-complex (BTB) domain, involved in recruiting targets to Cul3 for degradation. Analysis of the prostate tumor from this individual verified the presence of heterozygous N296I as well as an ERG fusion. CONCLUSIONS We have discovered a novel mutation in SPOP that tracks with prostate cancer within a family and is predicted to be deleterious. Taken together, our results implicate SPOP as a candidate gene for hereditary prostate cancer.
Collapse
Affiliation(s)
- Kimberly A. Zuhlke
- Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Anna M. Johnson
- Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Scott A. Tomlins
- Michigan Center for Translational Pathology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Nallasivam Palanisamy
- Michigan Center for Translational Pathology, University of Michigan Medical School, Ann Arbor, MI, USA
| | | | - Ethan M. Lange
- Department of Genetics, University of North Carolina School of Medicine, Chapel Hill, NC, USA
| | - William B. Isaacs
- Department of Urology, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Kathleen A. Cooney
- Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, USA
| |
Collapse
|