National Cancer Institute


Familial prostate cancer is associated with certain inherited gene variants. Learn about the hereditary prostate cancer genes, genetic testing, and clinical management in this expert-reviewed summary.

Familial prostate cancer is associated with certain inherited gene variants. Learn about the hereditary prostate cancer genes, genetic testing, and clinical management in this expert-reviewed summary.

Genetics of Prostate Cancer

Introduction

Many of the medical and scientific terms used in this summary are found in the NCI Dictionary of Genetics Terms. When a linked term is clicked, the definition will appear in a separate window.

Many of the genes and conditions described in this summary are found in the Online Mendelian Inheritance in Man (OMIM) catalog. For more information, see OMIM.

A concerted effort is being made within the genetics community to shift terminology used to describe genetic variation. The shift is to use the term “variant” rather than the term “mutation” to describe a difference that exists between the person or group being studied and the reference sequence, particularly for differences that exist in the germline. Variants can then be further classified as benign (harmless), likely benign, of uncertain significance, likely pathogenic, or pathogenic (disease causing). Throughout this summary, we will use the term pathogenic variant to describe a disease-causing mutation. For more information on variant classification, see Cancer Genetics Overview.

Prostate cancer is highly heritable. Up to 60% of prostate cancer risk is caused by inherited factors. The inherited risk is comprised of risk from common genetic variants and risk from pathogenic variants in moderate-risk and high-risk genes. As with breast and colon cancers, familial clustering of prostate cancer has been reported frequently.

Prostate cancer clusters with particular intensity in some families. Highly to moderately penetrantgenetic variants are thought to be associated with prostate cancer risk in these families. Members of these families may benefit from genetic counseling. Additionally, polygenic risk scores derived from combinations of single nucleotide polymorphisms, in addition to other risk factors like family history, race, and age/stage of prostate cancer diagnosis, have also been developed. Recommendations and guidelines for genetic counseling referrals are based on an individual's age at prostate cancer diagnosis, prostate cancer stage at diagnosis, and specific patterns of cancer in the family history. However, uptake of genetic testing based on an individual's family history of prostate cancer and/or a diagnosis of prostate cancer is variably implemented across practice settings and geographical regions. For more information about genetic testing criteria for prostate cancer, see Table 2.

References

  1. Houlahan KE, Livingstone J, Fox NS, et al.: A polygenic two-hit hypothesis for prostate cancer. J Natl Cancer Inst 115 (4): 468-472, 2023.
  2. Mucci LA, Hjelmborg JB, Harris JR, et al.: Familial Risk and Heritability of Cancer Among Twins in Nordic Countries. JAMA 315 (1): 68-76, 2016.
  3. Seibert TM, Garraway IP, Plym A, et al.: Genetic Risk Prediction for Prostate Cancer: Implications for Early Detection and Prevention. Eur Urol 83 (3): 241-248, 2023.
  4. Pagadala MS, Lynch J, Karunamuni R, et al.: Polygenic risk of any, metastatic, and fatal prostate cancer in the Million Veteran Program. J Natl Cancer Inst 115 (2): 190-199, 2023.
  5. Huynh-Le MP, Karunamuni R, Fan CC, et al.: Prostate cancer risk stratification improvement across multiple ancestries with new polygenic hazard score. Prostate Cancer Prostatic Dis 25 (4): 755-761, 2022.
  6. National Comprehensive Cancer Network: NCCN Clinical Practice Guidelines in Oncology: Genetic/Familial High-Risk Assessment: Breast, Ovarian, and Pancreatic. Version 2.2024. Plymouth Meeting, Pa: National Comprehensive Cancer Network, 2023. Available online with free registration. Last accessed October 31, 2023.
  7. National Comprehensive Cancer Network: NCCN Clinical Practice Guidelines in Oncology: Prostate Cancer Early Detection. Version 2.2023. Plymouth Meeting, Pa: National Comprehensive Cancer Network, 2023. Available online with free registration. Last accessed November 30, 2023.
  8. Clark NM, Flanagan MR: ASO Author Reflections: Low Genetic Testing Utilization Among Patients with Breast, Ovarian, Pancreatic, and Prostate Cancers. Ann Surg Oncol 30 (3): 1327-1328, 2023.
  9. Giri VN, Morgan TM, Morris DS, et al.: Genetic testing in prostate cancer management: Considerations informing primary care. CA Cancer J Clin 72 (4): 360-371, 2022.
  10. Russo J, Giri VN: Germline testing and genetic counselling in prostate cancer. Nat Rev Urol 19 (6): 331-343, 2022.

Risk Factors for Prostate Cancer

Age

Prostate cancer risk correlates with age. Prostate cancer is rarely seen in men younger than 40 years. The incidence rises rapidly with each decade thereafter. For example, the probability of being diagnosed with prostate cancer is 1 in 457 for men aged 49 years or younger, 1 in 55 for men aged 50 through 59 years, 1 in 19 for men aged 60 through 69 years, and 1 in 11 for men aged 70 years and older. Lifetime risk of developing prostate cancer is 1 in 8. Approximately 10% of prostate cancer cases are diagnosed in men younger than 56 years and represent early-onset prostate cancer. Data from the Surveillance, Epidemiology, and End Results (SEER) Program show that early-onset prostate cancer diagnosis rates are increasing, and there is evidence that cases may be more aggressive in this subpopulation.

Ancestry

The risk of developing prostate cancer is dramatically higher among Black individuals (176.2 cases/100,000 men) when compared with other racial and ethnic groups in the United States:

  • White: 103.5 cases/100,000 men.
  • Asian or Pacific Islander: 57.2 cases/100,000 men.
  • American Indian or Alaska Native: 82.6 cases/100,000 men.
  • Hispanic or Latino: 87.2 cases/100,000 men.

Prostate cancer mortality rates in Black individuals (37.5/100,000 men) are higher than those in other racial and ethnic groups in the United States:

  • White: 17.8/100,000 men.
  • Asian or Pacific Islander: 8.6/100,000 men.
  • American Indian or Alaska Native: 21.9/100,000 men.
  • Hispanic or Latino: 15.3/100,000 men.

Globally, prostate cancer incidence and mortality rates also vary widely from country to country. The etiology of this variation in prostate cancer risk is likely multifactorial and may be due to biological factors, access to health care, and other social determinants of health.

Family History of Prostate Cancer and Other Cancers

Results from several large case-control studies and cohort studies representing various populations suggest that family history is a major risk factor in prostate cancer. A family history of a brother or father with prostate cancer increases the risk of prostate cancer, and the risk is inversely related to the age of the affected relative. Risk is increased when a first-degree relative (FDR) was diagnosed with prostate cancer before age 65 years.

A meta-analysis of 33 epidemiological case-control and cohort-based studies has provided detailed information regarding risk ratios related to family history of prostate cancer (for more information, see Table 1).

Table 1. Relative Risk (RR) Related to Family History of Prostate Cancera

Risk GroupRR for Prostate Cancer (95% CI)
CI = confidence interval; FDR = first-degree relative.
aAdapted from Kiciński et al.
Brother(s) with prostate cancer diagnosed at any age3.14 (2.37–4.15)
Father with prostate cancer diagnosed at any age2.35 (2.02–2.72)
One affected FDR diagnosed at any age2.48 (2.25–2.74)
Affected FDRs diagnosed <65 y2.87 (2.21–3.74)
Affected FDRs diagnosed ≥65 y1.92 (1.49–2.47)
Second-degree relatives diagnosed at any age2.52 (0.99–6.46)
Two or more affected FDRs diagnosed at any age4.39 (2.61–7.39)

A family history of breast cancer is also associated with increased prostate cancer risk. In the Health Professionals Follow-up Study (HPFS), comprising over 40,000 men, those with a family history of breast cancer had a 21% higher risk of developing prostate cancer overall and a 34% increased risk of developing a lethal form of prostate cancer. This is consistent with findings from previous cohorts, though, notably, not all series have detected this association. The HPFS and other studies have also shown that men with a family history of both prostate and breast/ovarian cancers were at an even higher risk of prostate cancer compared with men with a family history of either prostate or breast/ovarian cancer alone. A proportion of the increased prostate cancer risk associated with family history of breast cancer is likely due to pathogenic variants in the DNA damage repair pathway, most commonly BRCA2. For more information, see the BRCA1 and BRCA2 section. The association between prostate and breast cancers in families appears bidirectional. Among women, a family history of prostate cancer is likewise associated with increased risk of breast cancer.

An association also exists between prostate cancer risk and colon cancer. Men with germline variants in DNA mismatch repair genes are at increased risk of developing prostate cancer. One study reported an approximately twofold increased risk of prostate cancer among first- and second-degree relatives of probands with colorectal cancer meeting Amsterdam I or Amsterdam II criteria for Lynch syndrome. For more information on Amsterdam criteria, see the Defining Lynch syndrome families section in Genetics of Colorectal Cancer.

Family history has been shown to be a risk factor for men of different races and ethnicities. In a population-based case-control study of prostate cancer among African American, White, and Asian American individuals in the United States (Los Angeles, San Francisco, and Hawaii) and Canada (Vancouver and Toronto), 5% of controls and 13% of all cases reported a father, brother, or son with prostate cancer. These prevalence estimates were somewhat lower among Asian American individuals than among African American or White individuals. A positive family history was associated with a twofold to threefold increase in relative risk (RR) in each of the three ethnic groups. The overall odds ratio (OR) associated with a family history of prostate cancer was 2.5 (95% confidence interval [CI], 1.9–3.3) with adjustment for age and ethnicity.

Evidence shows that a family history of prostate cancer can be associated with inferior clinical outcomes. When patients were referred for prostate biopsy (typically due to elevated prostate-specific antigen [PSA]), men with a family history of the disease were at increased risk for high-grade prostate cancer when compared with patients without a family history. A large population-based study from Utah reported that men with either of the following were at an increased risk for early-onset prostate cancer: 1) three or more FDRs diagnosed with prostate cancer, or 2) two or more FDRs or second-degree relatives with prostate cancer.

Genetics

There are multiple germline pathogenic variants and single nucleotide variants that are associated with prostate cancer risk. For more information about these genetic variants, see the National Human Genome Research Institute'sGWAS catalog. Germline genetic testing may be indicated to assess prostate cancer risk and/or inform therapeutic decision-making in men diagnosed with prostate cancer. Prostate cancer risks vary depending on the specific gene and pathogenic variant involved. Prostate cancer heritability (when considering low, moderate, and high-penetrant genetic factors) can be as high 57% (95% CI, 51%–63%). Genetic variants that contribute to this risk are continuously being identified. Prostate cancer heritability rates may also vary in different racial and ethnic populations. For more information, see the Germline Genetics for Prostate Cancer section.

References

  1. American Cancer Society: Cancer Facts and Figures 2023. American Cancer Society, 2023. Available online. Last accessed Dec. 15, 2023.
  2. Salinas CA, Tsodikov A, Ishak-Howard M, et al.: Prostate cancer in young men: an important clinical entity. Nat Rev Urol 11 (6): 317-23, 2014.
  3. Sung H, Ferlay J, Siegel RL, et al.: Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA Cancer J Clin 71 (3): 209-249, 2021.
  4. Krimphove MJ, Cole AP, Fletcher SA, et al.: Evaluation of the contribution of demographics, access to health care, treatment, and tumor characteristics to racial differences in survival of advanced prostate cancer. Prostate Cancer Prostatic Dis 22 (1): 125-136, 2019.
  5. Fletcher SA, Marchese M, Cole AP, et al.: Geographic Distribution of Racial Differences in Prostate Cancer Mortality. JAMA Netw Open 3 (3): e201839, 2020.
  6. Carter BS, Beaty TH, Steinberg GD, et al.: Mendelian inheritance of familial prostate cancer. Proc Natl Acad Sci U S A 89 (8): 3367-71, 1992.
  7. Grönberg H, Damber L, Damber JE: Familial prostate cancer in Sweden. A nationwide register cohort study. Cancer 77 (1): 138-43, 1996.
  8. Cannon L, Bishop DT, Skolnick M, et al.: Genetic epidemiology of prostate cancer in the Utah Mormon genealogy. Cancer Surv 1 (1): 47-69, 1982.
  9. Saarimäki L, Tammela TL, Määttänen L, et al.: Family history in the Finnish Prostate Cancer Screening Trial. Int J Cancer 136 (9): 2172-7, 2015.
  10. Barber L, Gerke T, Markt SC, et al.: Family History of Breast or Prostate Cancer and Prostate Cancer Risk. Clin Cancer Res 24 (23): 5910-5917, 2018.
  11. Ghadirian P, Howe GR, Hislop TG, et al.: Family history of prostate cancer: a multi-center case-control study in Canada. Int J Cancer 70 (6): 679-81, 1997.
  12. Stanford JL, Ostrander EA: Familial prostate cancer. Epidemiol Rev 23 (1): 19-23, 2001.
  13. Matikaine MP, Pukkala E, Schleutker J, et al.: Relatives of prostate cancer patients have an increased risk of prostate and stomach cancers: a population-based, cancer registry study in Finland. Cancer Causes Control 12 (3): 223-30, 2001.
  14. Kiciński M, Vangronsveld J, Nawrot TS: An epidemiological reappraisal of the familial aggregation of prostate cancer: a meta-analysis. PLoS One 6 (10): e27130, 2011.
  15. Cerhan JR, Parker AS, Putnam SD, et al.: Family history and prostate cancer risk in a population-based cohort of Iowa men. Cancer Epidemiol Biomarkers Prev 8 (1): 53-60, 1999.
  16. Kalish LA, McDougal WS, McKinlay JB: Family history and the risk of prostate cancer. Urology 56 (5): 803-6, 2000.
  17. Damber L, Grönberg H, Damber JE: Familial prostate cancer and possible associated malignancies: nation-wide register cohort study in Sweden. Int J Cancer 78 (3): 293-7, 1998.
  18. Agalliu I, Karlins E, Kwon EM, et al.: Rare germline mutations in the BRCA2 gene are associated with early-onset prostate cancer. Br J Cancer 97 (6): 826-31, 2007.
  19. Edwards SM, Kote-Jarai Z, Meitz J, et al.: Two percent of men with early-onset prostate cancer harbor germline mutations in the BRCA2 gene. Am J Hum Genet 72 (1): 1-12, 2003.
  20. Ford D, Easton DF, Bishop DT, et al.: Risks of cancer in BRCA1-mutation carriers. Breast Cancer Linkage Consortium. Lancet 343 (8899): 692-5, 1994.
  21. Gayther SA, de Foy KA, Harrington P, et al.: The frequency of germ-line mutations in the breast cancer predisposition genes BRCA1 and BRCA2 in familial prostate cancer. The Cancer Research Campaign/British Prostate Group United Kingdom Familial Prostate Cancer Study Collaborators. Cancer Res 60 (16): 4513-8, 2000.
  22. Beebe-Dimmer JL, Yee C, Cote ML, et al.: Familial clustering of breast and prostate cancer and risk of postmenopausal breast cancer in the Women's Health Initiative Study. Cancer 121 (8): 1265-72, 2015.
  23. Sellers TA, Potter JD, Rich SS, et al.: Familial clustering of breast and prostate cancers and risk of postmenopausal breast cancer. J Natl Cancer Inst 86 (24): 1860-5, 1994.
  24. Dominguez-Valentin M, Sampson JR, Seppälä TT, et al.: Cancer risks by gene, age, and gender in 6350 carriers of pathogenic mismatch repair variants: findings from the Prospective Lynch Syndrome Database. Genet Med 22 (1): 15-25, 2020.
  25. Samadder NJ, Smith KR, Wong J, et al.: Cancer Risk in Families Fulfilling the Amsterdam Criteria for Lynch Syndrome. JAMA Oncol 3 (12): 1697-1701, 2017.
  26. Whittemore AS, Wu AH, Kolonel LN, et al.: Family history and prostate cancer risk in black, white, and Asian men in the United States and Canada. Am J Epidemiol 141 (8): 732-40, 1995.
  27. Clements MB, Vertosick EA, Guerrios-Rivera L, et al.: Defining the Impact of Family History on Detection of High-grade Prostate Cancer in a Large Multi-institutional Cohort. Eur Urol 82 (2): 163-169, 2022.
  28. Beebe-Dimmer JL, Kapron AL, Fraser AM, et al.: Risk of Prostate Cancer Associated With Familial and Hereditary Cancer Syndromes. J Clin Oncol 38 (16): 1807-1813, 2020.
  29. Seibert TM, Garraway IP, Plym A, et al.: Genetic Risk Prediction for Prostate Cancer: Implications for Early Detection and Prevention. Eur Urol 83 (3): 241-248, 2023.
  30. Mucci LA, Hjelmborg JB, Harris JR, et al.: Familial Risk and Heritability of Cancer Among Twins in Nordic Countries. JAMA 315 (1): 68-76, 2016.
  31. Bree KK, Hensley PJ, Pettaway CA: Germline Predisposition to Prostate Cancer in Diverse Populations. Urol Clin North Am 48 (3): 411-423, 2021.

Risk Assessment for Prostate Cancer

Risk assessment for prostate cancer primarily involves the intake of a patient's family cancer history. Family history intake includes the following:

  • Information about cancers* in male and female blood relatives on maternal and paternal sides of the family.
  • Ages at cancer diagnoses.
  • Ages of death from cancer.
  • The number of relatives with metastatic prostate cancer.
  • The number of relatives who died of prostate cancer.
  • Information on relatives who are undergoing cancer screening, if known.

*Cancers include, but are not limited to, the following: prostate, breast, pancreas, colorectal, uterine, ovarian, upper gastrointestinal (GI), and skin cancers.

Ancestry is also an important component of the family history. Ashkenazi Jewish ancestry on either side of the family may prompt greater suspicion for founder pathogenic variants in BRCA1 and BRCA2, which could lead to increased cancer risk in a family. Men of African descent (Black men) also have a higher risk for prostate cancer. Within the United States, Black men (176.2 prostate cancer cases/100,000 men) have approximately a 70% higher incidence rate of prostate cancer than White men (103.5 prostate cancer cases/100,000 men). Black men also have more than twice the rate of prostate cancer–specific death (37.5 deaths/100,000 men) than White men (17.8 deaths/100,000 men). This increased prostate cancer risk may be due to challenges, including the following: 1) access to care, 2) limited awareness of prostate cancer screening programs, 3) limited engagement in prostate cancer screening/genetic testing, and 4) the presence of specific genetic markers that can increase prostate cancer risk.

These familial risk factors are then incorporated into recommendations for prostate cancer screening. National guidelines recommend discussing prostate cancer screening with prostate-specific antigen (PSA) and digital rectal exam between the ages of 45 and 75 years for individuals at average risk for prostate cancer.

In contrast, prostate cancer screening is recommended to start at age 40 years for individuals in these high-risk groups:

Men of Black/African descent.

Men with germline pathogenic variants that increase prostate cancer risk.

Men who have family histories with features suggestive of hereditary cancer syndromes like the following:

  • Hereditary breast and ovarian cancer syndrome: Family members with ovarian cancer, pancreatic cancer, metastatic/high-risk prostate cancer, male breast cancer, and/or breast cancer diagnosed at or before age 50 years.
  • Lynch syndrome: Family members with colorectal or endometrial cancer diagnosed at or before age 50 years, ovarian cancer, pancreatic cancer, urothelial cancer, and/or upper GI cancer.
  • Hereditary prostate cancer: Multiple generations with prostate cancer, deaths from prostate cancer, and/or family members with metastatic prostate cancer.

The role of additional markers, such as polygenic risk scores, in prostate cancer risk assessment is evolving. Additional screening strategies, like multiparametric magnetic resonance imaging (mpMRI), are also being studied.

References

  1. American Cancer Society: Cancer Facts and Figures 2023. American Cancer Society, 2023. Available online. Last accessed Dec. 15, 2023.
  2. Liadi Y, Campbell T, Dike P, et al.: Prostate cancer metastasis and health disparities: a systematic review. Prostate Cancer Prostatic Dis : , 2023.
  3. Nair SS, Chakravarty D, Dovey ZS, et al.: Why do African-American men face higher risks for lethal prostate cancer? Curr Opin Urol 32 (1): 96-101, 2022.
  4. National Comprehensive Cancer Network: NCCN Clinical Practice Guidelines in Oncology: Genetic/Familial High-Risk Assessment: Breast, Ovarian, and Pancreatic. Version 2.2024. Plymouth Meeting, Pa: National Comprehensive Cancer Network, 2023. Available online with free registration. Last accessed October 31, 2023.
  5. National Comprehensive Cancer Network: NCCN Clinical Practice Guidelines in Oncology: Prostate Cancer Early Detection. Version 2.2023. Plymouth Meeting, Pa: National Comprehensive Cancer Network, 2023. Available online with free registration. Last accessed November 30, 2023.
  6. National Comprehensive Cancer Network: NCCN Clinical Practice Guidelines in Oncology: Prostate Cancer. Version 4.2023. Plymouth Meeting, Pa: National Comprehensive Cancer Network, 2023. Available online with free registration. Last accessed November 30, 2023.

Indications for Prostate Cancer Germline Genetic Testing

The criteria for consideration of genetic testing for prostate cancer varies depending on the current guidelines and expert opinion consensus, as summarized in Table 2. Hereditary prostate cancer genetic testing criteria are based on an individual's family history, personal/disease characteristics, and tumor sequencing results. The genes recommended for genetic testing vary based on national guidelines and consensus conference recommendations. Precision therapy has emerged as a major driver for germline genetic testing and may be a separate reason to pursue testing beyond the criteria stated in Table 2. The National Comprehensive Cancer Network (NCCN) Prostate Cancer guidelines recommend testing for at least BRCA1, BRCA2, ATM, CHEK2, PALB2, HOXB13,MLH1, MSH2, MSH6, and PMS2 for men meeting specific testing indications. A consensus conference in 2019 addressed the role of genetic testing for inherited prostate cancer. Family history–based indications for genetic testing included testing for BRCA1/BRCA2, HOXB13, DNA mismatch repair (MMR) genes, and ATM. Tumor sequencing that identifies variants that may be germline in origin, like variants in BRCA1/BRCA2, DNA MMR genes, or ATM and other genes, warrants confirmatory germline testing. Somatic findings for which germline testing is considered include the following:

  • Somatic mutations that are associated with germline susceptibility.
  • Hypermutated tumors, which are indicative of DNA MMR.
  • Chromosome rearrangements in specific tumors.
  • High-variant allele frequency (percent of sequence reads that have the identified variant). Variant allele frequency can be altered for reasons not associated with germline variants such as loss of heterozygosity, ploidy (copy number variants), tumor heterogeneity, and tumor sample purity.

Table 2. Indications for Prostate Cancer Genetic Testing

Philadelphia Prostate Cancer Consensus Conference (Giri et al. 2020)aGenetic/Familial High-Risk Assessment: Breast, Ovarian, and Pancreatic (Version 2.2024)bNCCN Prostate Cancer (Version 4.2023)cEuropean Advanced Prostate Cancer Consensus Conference (Gillessen et al. 2017 and Gillessen 2020 )d
dMMR = mismatch repair deficient; FDR = first-degree relative; HBOC = hereditary breast and ovarian cancer; MMR = mismatch repair; MSI = microsatellite instability; NCCN = National Comprehensive Cancer Network; SDR= second-degree relative; TDR= third-degree relative.
aGiri et al.: Specific genes to test include BRCA1/BRCA2, DNA MMR genes, ATM, and HOXB13 depending on various testing indications.
bNCCN Genetic/Familial High-Risk Assessment: Breast, Ovarian, and Pancreatic guidelines state that prostate cancer risk management is indicated for BRCA1 and BRCA2carriers, but evidence for risk management is insufficient for other genes.
cNCCN Prostate Cancer guidelines specify that germline multigene testing includes at least the following genes: BRCA1, BRCA2, ATM, PALB2, CHEK2, MLH1, MSH2, MSH6, and PMS2. Including additional genes may be appropriate based on clinical context.
dGillessen et al. endorsed the use of large panel testing including homologous recombination and DNA MMR genes.
Family History Criteria All men with prostate cancer from families meeting established testing or syndromic criteria for HBOC, hereditary prostate cancer, or Lynch syndrome Men affected with prostate cancer who have a family history of the following: ≥1 FDR, SDR, or TDR (on the same side of the family) with breast cancer at age ≤50 y or with any of the following: triple-negative breast cancer, ovarian cancer, pancreatic cancer, high- or very-high-risk prostate cancer, male breast cancer, or metastatic prostate cancer at any ageMen affected with prostate cancer who have the following: ≥1 FDR, SDR, or TDR (on the same side of the family) with breast cancer at age ≤50 y, colorectal or endometrial cancer at age ≤50 y, male breast cancer at any age, ovarian cancer at any age, exocrine pancreatic cancer at any age, or metastatic, regional, very-high-risk, high-risk prostate cancer at any age Men with a positive family history of prostate cancer
Men affected with prostate cancer who have >2 close biological relatives with a cancer associated with HBOC, hereditary prostate cancer, or Lynch syndrome Men affected with prostate cancer who have ≥3 FDRs, SDRs, or TDRs (on the same side of the family) with breast cancer or prostate cancer (any grade) at any ageMen affected with prostate cancer who have ≥1 FDR with prostate cancer at age ≤60 y (exclude relatives with clinically localized Grade Group 1 disease)Men with a positive family history of other cancer syndromes (HBOC and/or pancreatic cancer and/or Lynch syndrome)
Men with an FDR who was diagnosed with prostate cancer at <60 y Men with or without prostate cancer with an FDR who meets any of the criteria listed above (except when a man without prostate cancer has relatives who meet the above criteria solely for systemic therapy decision-making; these criteria may also be extended to an affected TDR if he/she is related to the patient through two male relatives) Men affected with prostate cancer who have ≥2 FDRs, SDRs, or TDRs (on the same side of the family) with breast cancer or prostate cancer at any age (exclude relatives with clinically localized Grade Group 1 disease)
Men with relatives who died of prostate cancerMen affected with prostate cancer who have ≥3 FDRs or SDRs (on the same side of the family) with the following Lynch syndrome-related cancers, especially if diagnosed at age <50 y: colorectal, endometrial, gastric, ovarian, exocrine pancreas, upper tract urothelial, glioblastoma, biliary tract, and small intestine
Men with a metastatic prostate cancer in an FDR
Consider genetic testing in men with prostate cancer and Ashkenazi Jewish ancestry Men with prostate cancer and Ashkenazi Jewish ancestryMen with prostate cancer and Ashkenazi Jewish ancestry
Men with prostate cancer and a known family history of a pathogenic or likely pathogenic variant in one of the following genes: BRCA1, BRCA2, ATM, PALB2, CHEK2, MLH1, MSH2, MSH6, PMS2, or EPCAM
Clinical/Pathological FeaturesMen with metastatic prostate cancerMen with metastatic prostate cancer Men with metastatic prostate cancerMen with newly diagnosed metastatic prostate cancer (62% of panel voted in favor of genetic counseling/testing in a minority of selected patients)
Men with stage T3a or higher prostate cancerMen with high- or very-high-risk prostate cancer Men with high-risk prostate cancer, very-high-risk prostate cancer, high-risk localized prostate cancer, or regional (node-positive) prostate cancer
Men with prostate cancer that has intraductal/ductal histologyTesting may be considered in men who have intermediate-risk prostate cancer with intraductal/cribriform histology at any age Germline testing may be considered in men who have intermediate-risk prostate cancer with intraductal/cribriform histology at any age
Germline testing may be considered in men with prostate cancer AND a prior personal history of any of the following cancers: exocrine pancreatic, colorectal, gastric, melanoma, upper tract urothelial, glioblastoma, biliary tract, and small intestinalMen with prostate cancer diagnosed at age <60 y
Tumor Sequencing Characteristics Men with prostate cancer whose somatic testing reveals the possibility of a germline variant in a cancer risk gene, especially BRCA2, BRCA1, ATM, and DNA MMR genesMen with a pathogenic variant found on tumor genomic testing that may have clinical implications if it is also identified in the germline Recommend tumor testing for pathogenic variants in homologous recombination genes in men with metastatic disease; consider tumor testing in men with regional prostate cancer
Recommend MSI-high or dMMR tumor testing in men with metastatic castration-resistant prostate cancer; consider testing in men with regional or castration-sensitive metastatic prostate cancer

References

  1. Giri VN, Knudsen KE, Kelly WK, et al.: Role of Genetic Testing for Inherited Prostate Cancer Risk: Philadelphia Prostate Cancer Consensus Conference 2017. J Clin Oncol 36 (4): 414-424, 2018.
  2. Gillessen S, Attard G, Beer TM, et al.: Management of Patients with Advanced Prostate Cancer: The Report of the Advanced Prostate Cancer Consensus Conference APCCC 2017. Eur Urol 73 (2): 178-211, 2018.
  3. National Comprehensive Cancer Network: NCCN Clinical Practice Guidelines in Oncology: Genetic/Familial High-Risk Assessment: Breast, Ovarian, and Pancreatic. Version 2.2024. Plymouth Meeting, Pa: National Comprehensive Cancer Network, 2023. Available online with free registration. Last accessed October 31, 2023.
  4. National Comprehensive Cancer Network: NCCN Clinical Practice Guidelines in Oncology: Prostate Cancer. Version 4.2023. Plymouth Meeting, Pa: National Comprehensive Cancer Network, 2023. Available online with free registration. Last accessed November 30, 2023.
  5. National Comprehensive Cancer Network: NCCN Clinical Practice Guidelines in Oncology: Prostate Cancer Early Detection. Version 2.2023. Plymouth Meeting, Pa: National Comprehensive Cancer Network, 2023. Available online with free registration. Last accessed November 30, 2023.
  6. Giri VN, Knudsen KE, Kelly WK, et al.: Implementation of Germline Testing for Prostate Cancer: Philadelphia Prostate Cancer Consensus Conference 2019. J Clin Oncol 38 (24): 2798-2811, 2020.
  7. Raymond VM, Gray SW, Roychowdhury S, et al.: Germline Findings in Tumor-Only Sequencing: Points to Consider for Clinicians and Laboratories. J Natl Cancer Inst 108 (4): , 2016.
  8. Gillessen S, Attard G, Beer TM, et al.: Management of Patients with Advanced Prostate Cancer: Report of the Advanced Prostate Cancer Consensus Conference 2019. Eur Urol 77 (4): 508-547, 2020.

Genetic Testing Approach for Prostate Cancer

Since next-generation sequencing (NGS) has become readily available and patent restrictions have been eliminated, several clinical laboratories offer multigene panel testing at a cost that is comparable to that of single-gene testing. Three types of genetic test results can be reported: 1) pathogenic/likely pathogenic variants, 2) variants of uncertain significance (VUS), or 3) negative results. Patients need pretest genetic counseling or informed consent to understand germline genetic testing results. For example, patients should understand that VUS can be reported, that VUS do not immediately impact care/inform cancer risk, and that VUS may be reclassified as either pathogenic/likely pathogenic or benign/likely benign when more data are acquired. For more information on genetic counseling considerations and research associated with multigene testing, see the Multigene (panel) testing section in Cancer Genetics Risk Assessment and Counseling.

Germline Genetics for Prostate Cancer

Clinically Relevant Genes for Prostate Cancer

BRCA1 and BRCA2

Studies of male carriers of BRCA1 and BRCA2 pathogenic variants demonstrate that these individuals have a higher risk of prostate cancer and other cancers. Prostate cancer, in particular, has been observed at higher rates in male carriers of BRCA2 pathogenic variants than in the general population. For more information about BRCA1 and BRCA2 pathogenic variants, see BRCA1 and BRCA2: Cancer Risks and Management.

BRCA–associated prostate cancer risk

The risk of prostate cancer in carriers of BRCA pathogenic variants has been studied in various settings.

In an effort to clarify the relationship between BRCA pathogenic variants and prostate cancer risk, findings from several case series are summarized in Table 3.

Table 3. Case Series of BRCA Pathogenic Variants in Prostate Cancer

StudyPopulationProstate Cancer Risk (BRCA1)Prostate Cancer Risk (BRCA2)
BCLC = Breast Cancer Linkage Consortium; CDC = Centers for Disease Control and Prevention; CI = confidence interval; CIMBA = Consortium of Investigators of Modifiers of BRCA1/2; OCCR = Ovarian Cancer Cluster Region; RR = relative risk; SIR = standardized incidence ratio.
aIncludes all cancers except breast, ovarian, and nonmelanoma skin cancers.
BCLC (1999) BCLC family set that included 173 BRCA2linkage– or pathogenic variant–positive families, among which there were 3,728 individuals and 333 cancersaNot assessedOverall: RR, 4.65 (95% CI, 3.48–6.22)
Men <65 y: RR, 7.33 (95% CI, 4.66–11.52)
Thompson et al. (2001) BCLC family set that included 164 BRCA2 pathogenic variant–positive families, among which there were 3,728 individuals and 333 cancersaNot assessedOCCR: RR, 0.52 (95% CI, 0.24–1.00)
Thompson et al. (2002) BCLC family set that included 7,106 women and 4,741 men, among which 2,245 were carriers of BRCA1 pathogenic variants; 1,106 were tested noncarriers, and 8,496 were not testedOverall: RR, 1.07 (95% CI, 0.75–1.54) Not assessed
Men younger than 65 y: RR, 1.82 (95% CI, 1.01–3.29)
Mersch et al. (2015) Clinical genetics population at a single institution from 1997–2013. Compared cancer incidence with U.S. Statistics Report by CDC for general population cancer incidence SIR, 3.809 (95% CI, 0.766–11.13) (Not significant) SIR, 4.89 (95% CI, 1.959–10.075)
Silvestri et al. (2020) Cohort of 6,902 men who carried pathogenic variants in BRCA1 or BRCA2 in 53 cancer genetics groups across 33 countriesOccurred in 22.3% of carriersOccurred in 25.6% of carriers
Li et al. (2022) Cohort of 3,184 BRCA1 and 2,157 BRCA2 families from CIMBA; 34 of 1,508 men with BRCA1 pathogenic variants and 71 of 1,063 men with BRCA2 pathogenic variants had prostate cancerRR, 0.82 (95% CI, 0.54–1.27)RR, 2.22 (95% CI, 1.63–3.03)

Estimates derived from the Breast Cancer Linkage Consortium may be overestimates because the data were generated from highly selected families that had significant risks of breast and ovarian cancers and were suitable for linkage analysis. A review of the relationship between BRCA2 germline pathogenic variants and prostate cancer risk suggests that BRCA2 confers a significant increase in risk among male members of HBOC families but likely plays only a small role in site-specific, multiple-case prostate cancer families.

A meta-analysis assessed the relationship between BRCA1 and BRCA2 germline pathogenic variants and prostate cancer risk. The risk of prostate cancer was higher in BRCA2 carriers (odds ratio [OR], 2.64; 95% confidence interval [CI], 2.03–3.47) than in BRCA1 carriers (OR, 1.35; 95% CI, 1.03–1.76). Several studies cited in Table 3 were included in this meta-analysis.

Prevalence of BRCA founder pathogenic variants in men with prostate cancer

Ashkenazi Jewish population

Several studies in Israel and in North America have analyzed the frequency of BRCAfounder pathogenic variants among Ashkenazi Jewish (AJ) men with prostate cancer. Two specific BRCA1 pathogenic variants (185delAG and 5382insC) and one BRCA2 pathogenic variant (6174delT) are common in individuals of AJ ancestry. Carrier frequencies for these pathogenic variants in the general Jewish population are 0.9% (95% CI, 0.7%–1.1%) for the 185delAG pathogenic variant, 0.3% (95% CI, 0.2%–0.4%) for the 5382insC pathogenic variant, and 1.3% (95% CI, 1.0%–1.5%) for the BRCA2 6174delT pathogenic variant. In these studies, the relative risks (RRs) were commonly greater than 1, but only a few were statistically significant. Many of these studies were not sufficiently powered to rule out a lower, but clinically significant, risk of prostate cancer in carriers of Ashkenazi BRCA founder pathogenic variants.

In the Washington Ashkenazi Study (WAS), a kin-cohort analytic approach was used to estimate the cumulative risk of prostate cancer among more than 5,000 American AJ male volunteers from the Washington, District of Columbia area who carried one of the BRCA Ashkenazi founder pathogenic variants. The cumulative risk to age 70 years was estimated to be 16% (95% CI, 4%–30%) among carriers of the founder pathogenic variants and 3.8% (95% CI, 3.3%–4.4%) among noncarriers. This fourfold increase in prostate cancer risk was equal (in absolute terms) to the cumulative risk of ovarian cancer among female carriers at the same age (16% by age 70 y; 95% CI, 6%–28%). The risk of prostate cancer in male carriers in the WAS cohort was elevated by age 50 years, was statistically significantly elevated by age 67 years, and increased thereafter with age, suggesting both an overall excess in prostate cancer risk and an earlier age at diagnosis among carriers of Ashkenazi founder pathogenic variants. Prostate cancer risk differed depending on the gene, with BRCA1 pathogenic variants associated with increasing risk after age 55 to 60 years, reaching 25% by age 70 years and 41% by age 80 years. In contrast, prostate cancer risk associated with the BRCA2 pathogenic variant began to rise at later ages, reaching 5% by age 70 years and 36% by age 80 years (numeric values were provided by the author [written communication, April 2005]).

The studies summarized in Table 4 used similar case-control methods to examine the prevalence of Ashkenazi founder pathogenic variants among Jewish men with prostate cancer and found an overall positive association between carrier status of founder pathogenic variants and prostate cancer risk.

Table 4. Case-Control Studies in Ashkenazi Jewish Populations of BRCA1 and BRCA2 and Prostate Cancer Risk

StudyCases/ControlsPathogenic Variant Frequency (BRCA1)Pathogenic Variant Frequency (BRCA2)Prostate Cancer Risk (BRCA1)Prostate Cancer Risk (BRCA2)Comments
AJ = Ashkenazi Jewish; CI = confidence interval; MECC = Molecular Epidemiology of Colorectal Cancer; OR = odds ratio; WAS = Washington Ashkenazi Study.
Giusti et al. (2003) Cases: 979 consecutive AJ men from Israel diagnosed with prostate cancer between 1994 and 1995 Cases: 16 (1.7%) Cases: 14 (1.5%) 185delAG: OR, 2.52 (95% CI, 1.05–6.04)OR, 2.02 (95% CI, 0.16–5.72)There was no evidence of unique or specific histopathology findings within the pathogenic variant–associated prostate cancers
Controls: Prevalence of founder pathogenic variants compared with age-matched controls >50 y with no history of prostate cancer from the WAS study and the MECC study from IsraelControls: 11 (0.81%)Controls: 10 (0.74%5282insC: OR, 0.22 (95% CI, 0.16–5.72)
Kirchoff et al. (2004) Cases: 251 unselected AJ men treated for prostate cancer between 2000 and 2002 Cases: 5 (2.0%)Cases: 8 (3.2%)OR, 2.20 (95% CI, 0.72–6.70)OR, 4.78 (95% CI, 1.87–12.25)
Controls: 1,472 AJ men with no history of cancerControls: 12 (0.8%)Controls: 16 (1.1%)
Agalliu et al. (2009) Cases: 979 AJ men diagnosed with prostate cancer between 1978 and 2005 (mean and median year of diagnosis: 1996) Cases: 12 (1.2%)Cases: 18 (1.9%)OR, 1.39 (95% CI, 0.60–3.22)OR, 1.92 (95% CI, 0.91–4.07)Gleason score 7–10 prostate cancer was more common in carriers of BRCA1 pathogenic variants (OR, 2.23; 95% CI, 0.84–5.86) and carriers of BRCA2 pathogenic variants (OR, 3.18; 95% CI, 1.62–6.24) than in controls
Controls: 1,251 AJ men with no history of cancerControls: 11 (0.9%)Controls: 12 (1.0%)
Gallagher et al. (2010) Cases: 832 AJ men diagnosed with localized prostate cancer between 1988 and 2007Noncarriers: 806 (96.9%)Noncarriers: 447 (98.5%)OR, 0.38 (95% CI, 0.05–2.75)OR, 3.18 (95% CI, 1.52–6.66)The BRCA1 5382insC founder pathogenic variant was not tested in this series, so it is likely that some carriers of this pathogenic variant were not identified. Consequently, BRCA1-related risk may be underestimated. Gleason score 7–10 prostate cancer was more common in carriers of BRCA2 pathogenic variants (85%) than in noncarriers (57%); P = .0002. Carriers of BRCA1/BRCA2 pathogenic variants had significantly greater risk of recurrence and prostate cancer–specific death than did noncarriers
Cases: 6 (0.7%)Cases: 20 (2.4%)
Controls: 454 AJ men with no history of cancerControls: 4 (0.9%)Controls: 3 (0.7%)

These studies support the hypothesis that prostate cancer occurs excessively among carriers of AJ founder pathogenic variants and suggest that the risk may be greater among men with the BRCA2 founder pathogenic variant (6174delT) than among those with one of the BRCA1 founder pathogenic variants (185delAG; 5382insC). The magnitude of the BRCA2-associated risks differs somewhat, undoubtedly because of interstudy differences related to participant ascertainment, calendar time differences in diagnosis, and analytic methods. Some data suggest that BRCA-related prostate cancer has a significantly worse prognosis than prostate cancer that occurs among noncarriers.

Other populations

The association between prostate cancer and pathogenic variants in BRCA1 and BRCA2 has also been studied in other populations. Table 5 summarizes studies that used case-control methods to examine the prevalence of BRCA pathogenic variants among men with prostate cancer from other varied populations.

Table 5. Case-Control Studies in Varied Populations of BRCA1 and BRCA2 and Prostate Cancer Risk

StudyCases/ControlsPathogenic Variant Frequency (BRCA1)Pathogenic Variant Frequency (BRCA2)Prostate Cancer Risk (BRCA1)Prostate Cancer Risk (BRCA2)Comments
CI = confidence interval; OR = odds ratio; RR = relative risk; SIR = standardized incidence ratio.
Johannesdottir et al. (1996) Cases: 75 Icelandic men diagnosed with prostate cancer <65 y, between 1983 and 1992, with available archival tissue blocks Not assessedCases: 999del5 (2.7%) Not assessed999del5: RR, 2.5 (95% CI, 0.49–18.4)
Controls: 499 randomly selected DNA samples from the Icelandic National Diet SurveyControls: (0.4%)
Eerola et al. (2001) Cases: 107 Finnish hereditary breast cancer families defined as having three first- or second-degree relatives with breast or ovarian cancer at any ageNot assessedNot assessedSIR, 1.0 (95% CI, 0.0–3.9)SIR, 4.9 (95% CI, 1.8–11.0)
Controls: Finnish population based on gender, age, and calendar period–specific incidence rates
Cybulski et al. (2013) Cases: 3,750 Polish men with prostate cancer unselected for age or family history and diagnosed between 1999 and 2012 Cases: 14 (0.4%)Not assessedAny BRCA1 pathogenic variant: OR, 0.9 (95% CI, 0.4–1.8)Not assessedProstate cancer risk was greater in familial cases and cases diagnosed <60 y
4153delA: OR, 5.3 (95% CI, 0.6–45.2)
Controls: 3,956 Polish men with no history of cancer aged 23–90 yControls: 17 (0.4%) 5382insC: OR, 0.5 (95% CI, 0.2–1.3)
C61G: OR, 1.1 (95% CI, 1.6–2.2)

These data suggest that prostate cancer risk in carriers of BRCA1/BRCA2 pathogenic variants varies with the location of the pathogenic variant (i.e., there is a correlation between genotype and phenotype). These observations might explain some of the inconsistencies encountered in prior studies of these associations, because varied populations may have differences in the proportion of individuals with specific BRCA1/BRCA2 pathogenic variants.

Several case series have also explored the role of BRCA1 and BRCA2 pathogenic variants and prostate cancer risk.

Table 6. Case Series of BRCA1 and BRCA2 and Prostate Cancer Risk

StudyPopulationPathogenic Variant Frequency (BRCA1)Pathogenic Variant Frequency (BRCA2)Prostate Cancer Risk (BRCA1)Prostate Cancer Risk (BRCA2)Comments
CI = confidence interval; MLPA = multiplex ligation-dependent probe amplification; RR = relative risk; SIR = standardized incidence ratio; UK = United Kingdom.
aEstimate calculated using RR data in UK general population.
bRisks calculated on men with pathogenic variants diagnosed with prostate cancer.
Agalliu et al. (2007) 290 men (White, n = 257; African American, n = 33) diagnosed with prostate cancer <55 y and unselected for family historyNot assessed2 (0.69%)Not assessedRR, 7.8 (95% CI, 1.8–9.4)No pathogenic variants were found in African American men
The two men with a pathogenic variant reported no family history of breast cancer or ovarian cancer
Agalliu et al. (2007) 266 individuals from 194 hereditary prostate cancer families, including 253 men affected with prostate cancer; the median age at prostate cancer diagnosis was 58 yNot assessed0 (0%)Not assessedNot assessed31 nonsynonymous variations were identified; no truncating or pathogenic variants were detected
Tryggvadóttir et al. (2007) 527 men diagnosed with prostate cancer between 1955 and 2004Not assessed30/527 (5.7%) carried the Icelandic founder pathogenic variant 999del5Not assessedNot assessedThe BRCA2 999del5 pathogenic variant was associated with a lower mean age at prostate cancer diagnosis (69 vs. 74 y; P = .002)
Kote-Jarai et al. (2011) 1,832 men diagnosed with prostate cancer between ages 36 and 88 y who participated in the UK Genetic Prostate Cancer StudyNot assessedOverall: 19/1,832 (1.03%) Not assessedRR, 8.6a (95% CI, 5.1–12.6)MLPA was not used; therefore, the pathogenic variant frequency may be an underestimate, given the inability to detect large genomic rearrangements
Prostate cancer diagnosed ≤55 y: 8/632 (1.27%)
Leongamornlert et al. (2012) 913 men with prostate cancer who participated in the UK Genetic Prostate Cancer Study; this included 821 cases diagnosed between ages 36 and 65 y, regardless of family history, and 92 cases diagnosed >65 y with a family history of prostate cancerAll cases: 4/886 (0.45%) Not assessedRR, 3.75a (95% CI, 1.02–9.6)Not assessedQuality-control assessment after sequencing excluded 27 cases, resulting in 886 cases included in the final analysis
Cases ≤65 y: 3/802 (0.37%)
Nyberg et al. (2019) Prospective cohort of men with BRCA1 (n = 376) or BRCA2 (n = 447) pathogenic variants from the UK and Ireland; the median follow-up was 5.9 y and 5.3 y, respectively, for prostate cancer diagnosesConfirmed pathogenic variant: 16/376Confirmed pathogenic variant: 26/447SIR, 2.35 (95% CI, 1.43–3.88)SIR, 4.45 (95% CI, 2.99–6.61)Absolute prostate cancer risksb: 21% (95% CI, 13%–34%) by age 75 y and 29% (95% CI, 17%–45%) by age 85 y for BRCA1; 27% (95% CI, 17%–41%) by age 75 y and 60% (95% CI, 43%–78%) by age 85 y for BRCA2

These case series confirm that pathogenic variants in BRCA1 and BRCA2 do not play a significant role in hereditary prostate cancer. However, germline pathogenic variants in BRCA2 account for some cases of early-onset prostate cancer, although this is estimated to be less than 1% of early-onset prostate cancers in the United States.

Prostate cancer aggressiveness in carriers of BRCA pathogenic variants

The studies summarized in Table 7 used similar case-control methods to examine features of prostate cancer aggressiveness among men with prostate cancer found to harbor a BRCA1/BRCA2 pathogenic variant.

Table 7. Case-Control Studies of BRCA1 and BRCA2 and Prostate Cancer Aggressiveness

StudyCases / ControlsGleason ScoreaPSAaTumor Stage or GradeaComments
AJ = Ashkenazi Jewish; CI = confidence interval; HR = hazard ratio; OR = odds ratio; PSA = prostate-specific antigen; UK = United Kingdom.
aMeasures of prostate cancer aggressiveness.
Tryggvadóttir et al. (2007) Cases: 30 men diagnosed with prostate cancer who were carriers of BRCA2 999del5 founder pathogenic variantsGleason score 7–10:Not assessedStage IV at diagnosis:
— Cases: 84%— Cases: 55.2%
Controls: 59 men with prostate cancer matched by birth and diagnosis year and confirmed not to carry the BRCA2 999del5 pathogenic variant — Controls: 52.7% — Controls: 24.6%
Agalliu et al. (2009) Cases: 979 AJ men diagnosed with prostate cancer between 1978 and 2005 (mean and median year of diagnosis, 1996)Gleason score 7–10: Not assessedNot assessed
BRCA1 185delAG pathogenic variant: OR, 3.54 (95% CI, 1.22–10.31)
Controls: 1,251 AJ men with no history of cancerBRCA2 6174delT pathogenic variant: OR, 3.18 (95% CI, 1.37–7.34)
Edwards et al. (2010) Cases: 21 men diagnosed with prostate cancer who harbored a BRCA2 pathogenic variant; 6 with early-onset disease (≤55 y) from a UK prostate cancer study and 15 unselected for age at diagnosis from a UK clinical seriesNot assessedPSA ≥25 ng/mL: HR, 1.39 (95% CI, 1.04–1.86)Stage T3: HR, 1.19 (95% CI, 0.68–2.05)
Stage T4: HR, 1.87 (95% CI, 1.00–3.48)
Grade 2: HR, 2.24 (95% CI, 1.03–4.88)
Controls: 1,587 age- and stage-matched men with prostate cancerGrade 3: HR, 3.94 (95% CI, 1.78–8.73)
Gallagher et al. (2010) Cases: 832 AJ men diagnosed with localized prostate cancer between 1988 and 2007, of which there were 6 carriers of BRCA1 pathogenic variants and 20 carriers of BRCA2 pathogenic variantsGleason score 7–10: Not assessedNot assessedThe BRCA1 5382insC founder pathogenic variant was not tested in this series
Controls: 454 AJ men with no history of cancerBRCA2 6174delT pathogenic variant: HR, 2.63 (95% CI, 1.23–5.6; P = .001)
Thorne et al. (2011) Cases: 40 men diagnosed with prostate cancer who were carriers of BRCA2 pathogenic variants from 30 familial breast cancer families from Australia and New Zealand Gleason score ≥8:PSA 10–100 ng/mL:Stage ≥pT3 at presentation: Carriers of BRCA2 pathogenic variants were more likely to have high-risk disease by D’Amico criteria than were noncarriers (77.5% vs. 58.7%, P = .05)
BRCA2 pathogenic variants: 35% (14/40)BRCA2 pathogenic variants: 44.7% (17/38)
BRCA2 pathogenic variants: 65.8% (25/38) — Controls: 27.9% (27/97)
PSA >101 ng/mL:
Controls: 97 men from 89 familial breast cancer families from Australia and New Zealand with prostate cancer and no BRCA pathogenic variant found in the family — Controls: 33.0% (25/97) BRCA2 pathogenic variants: 10% (4/40)— Controls: 22.6% (21/97)
— Controls: 2.1% (2/97)
Castro et al. (2013) Cases: 2,019 men diagnosed with prostate cancer from the UK, of whom 18 were carriers of BRCA1 pathogenic variants and 61 were carriers of BRCA2 pathogenic variants Gleason score >8:BRCA1 median PSA: 8.9 (range, 0.7–3,000)Stage ≥pT3 at presentation:Nodal metastasis and distant metastasis were higher in men with a BRCA pathogenic variant than in controls
BRCA1 pathogenic variants: 27.8% (5/18)BRCA1: 38.9% (7/18)
BRCA2 pathogenic variants: 37.7% (23/61)BRCA2 median PSA: 15.1 (range, 0.5–761)BRCA2 : 49.2% (30/61)
Controls: 1,940 men who were BRCA1/BRCA2 noncarriers— Controls 15.4% (299/1,940)Controls median PSA: 11.3 (range, 0.2–7,800)— Controls: 31.7% (616/1,940)
Akbari et al. (2014) Cases: 4,187 men who underwent prostate biopsy for elevated PSA or abnormal exam, including 26 men with at least one BRCA coding pathogenic variant (all 26 coding exons of BRCA were sequenced for polymorphisms)Gleason score 7–10:Cases median PSA: 56.3Not fully assessed in cases and controlsThe 12-year survival for men with a BRCA2 pathogenic variant was inferior to that of men without a BRCA2 pathogenic variant (61.8% vs. 94.3%; P< 10−4). Among the men with high-grade disease (Gleason 7–9), the presence of a BRCA2 pathogenic variant was associated with an HR of 4.38 (95% CI, 1.99–9.62; P< .0001) after adjusting for age and PSA level
— Cases 96%
Controls: 1,878 men with no BRCA coding pathogenic variants (all 26 coding exons of BRCA were sequenced for polymorphisms) — Controls 54% Controls median PSA: 13.3

Men harboring pathogenic variants in the United Kingdom and Ireland were prospectively followed for prostate cancer diagnoses (BRCA1 [n = 16/376] and BRCA2 [n = 26/447]; median follow-up, 5.9 y and 5.3 y, respectively). The prostate cancers identified covered the spectrum of Gleason scores from less than 6 to greater than 8; however, they differed by gene:

  • BRCA1 Gleason score less than 6; standardized incidence ratio (SIR), 3.50 (95% CI, 1.67–7.35) and Gleason score greater than 7; SIR, 1.80 (95% CI, 0.89–3.65).
  • BRCA2 Gleason score less than 6; SIR, 3.03 (95% CI, 1.24–7.44) and Gleason score greater than 7; SIR, 5.07 (95% CI, 3.20–8.02).

These studies suggest that prostate cancer in BRCA pathogenic variant carriers may be associated with aggressive disease features including a high Gleason score, a high prostate-specific antigen (PSA) level at diagnosis, and a high tumor stage and/or grade at diagnosis. This is a finding that warrants consideration when patients undergo cancer risk assessment and genetic counseling. Research is under way to gain insight into the biological basis of aggressive prostate cancer in carriers of BRCA pathogenic variants. One study of 14 BRCA2 germline pathogenic variant carriers reported that BRCA2-associated prostate cancers harbor increased genomic instability and a mutational profile that more closely resembles metastatic prostate cancer than localized disease, with genomic and epigenomic dysregulation of the MED12L/MED12 axis similar to metastatic castration-resistant prostate cancer.

BRCA1/BRCA2 and survival outcomes

Analyses of prostate cancer cases in families with known BRCA1 or BRCA2 pathogenic variants have been examined for survival. In an unadjusted analysis performed on a case series, median survival was 4 years in 183 men with prostate cancer with a BRCA2 pathogenic variant and 8 years in 119 men with a BRCA1 pathogenic variant. The study suggests that carriers of BRCA2 pathogenic variants have a poorer survival than carriers of BRCA1 pathogenic variants. The case-control studies summarized in Table 8 further assess this observation.

Table 8. Case-Control Studies of BRCA1 and BRCA2 and Survival Outcomes

StudyCasesControlsProstate Cancer–Specific SurvivalOverall SurvivalComments
AJ = Ashkenazi Jewish; CI = confidence interval; HR = hazard ratio; PSA = prostate-specific antigen; UK = United Kingdom.
Tryggvadóttir et al. (2007) 30 men diagnosed with prostate cancer who were carriers of BRCA2 999del5 founder pathogenic variants59 men with prostate cancer matched by birth and diagnosis year and confirmed not to carry the BRCA2 999del5 pathogenic variant BRCA2 999del5 pathogenic variant was associated with a higher risk of death from prostate cancer (HR, 3.42; 95% CI, 2.12–5.51), which remained after adjustment for tumor stage and grade (HR, 2.35; 95% CI, 1.08–5.11)Not assessed
Edwards et al. (2010) 21 men diagnosed with prostate cancer who harbored a BRCA2 pathogenic variant: 6 with early-onset disease (≤55 y) from a UK prostate cancer study and 15 unselected for age at diagnosis from a UK clinical series1,587 age- and stage-matched men with prostate cancer Not assessedOverall survival was lower in carriers of BRCA2 pathogenic variants (4.8 y) than in noncarriers (8.5 y); in noncarriers, HR, 2.14 (95% CI, 1.28–3.56; P = .003)
Gallagher et al. (2010) 832 AJ men diagnosed with localized prostate cancer between 1988 and 2007, of which 6 were carriers of BRCA1 pathogenic variants and 20 carriers of BRCA2 pathogenic variants454 AJ men with no history of cancerAfter adjusting for stage, PSA, Gleason score, and therapy received: Not assessedThe BRCA1 5382insC founder pathogenic variant was not tested in this series
– Carriers of BRCA1 185delAG pathogenic variants had a greater risk of death due to prostate cancer (HR, 5.16; 95% CI, 1.09–24.53; P = .001)
— Carriers of BRCA2 6174delT pathogenic variants had a greater risk of death due to prostate cancer (HR, 5.48; 95% CI, 2.03–14.79; P = .001)
Thorne et al. (2011) 40 men diagnosed with prostate cancer who were carriers of BRCA2 pathogenic variants from 30 familial breast cancer families from Australia and New Zealand 97 men from 89 familial breast cancer families from Australia and New Zealand with prostate cancer and no BRCA pathogenic variant found in the family BRCA2 carriers were shown to have an increased risk of prostate cancer–specific mortality (HR, 4.5; 95% CI, 2.12–9.52; P = 8.9 × 10-5), compared with noncarrier controlsBRCA2 carriers were shown to have an increased risk of death (HR, 3.12; 95% CI, 1.64–6.14; P = 3.0 × 10-4), compared with noncarrier controlsThere were too few BRCA1 carriers available to include in the analysis
Castro et al. (2013) 2,019 men diagnosed with prostate cancer from the UK, of whom 18 were carriers of BRCA1 pathogenic variants and 61 were carriers of BRCA2 pathogenic variants 1,940 men who were BRCA1/ BRCA2 noncarriersProstate cancer–specific survival at 5 y: Overall survival at 5 y:For localized prostate cancer, metastasis-free survival was also higher in controls than in carriers of pathogenic variants (93% vs. 77%; HR, 2.7)
BRCA1: 80.8% (95% CI, 56.9%–100%) BRCA1: 82.5% (95% CI, 60.4%–100%)
BRCA2: 67.9% (95% CI 53.4%–82.4%)BRCA2: 57.9% (95% CI, 43.4%–72.4%)
— Controls: 90.6% (95% CI 88.8%–92.4%)— Controls: 86.4% (95% CI, 84.4%–88.4%)
Castro et al. (2015) 1,302 men from the UK with local or locally advanced prostate cancer, including 67 carriers of BRCA1/BRCA2 pathogenic variants1,235 men who were BRCA1/BRCA2 noncarriersProstate cancer–specific survival:Not assessed
BRCA1/BRCA2: 61% at 10 y
— Noncarriers: 85% at 10 y

These findings suggest overall survival (OS) and prostate cancer–specific survival may be lower in carriers of pathogenic variants than in controls.

Additional studies involving the BRCA region

A genome-wide scan for hereditary prostate cancer in 175 families from the University of Michigan Prostate Cancer Genetics Project (UM-PCGP) found evidence of linkage to chromosome 17q markers. The maximum logarithm of the odds (LOD) score in all families was 2.36, and the LOD score increased to 3.27 when only families with four or more confirmed affected men were analyzed. The linkage peak was centered over the BRCA1 gene. In follow-up, these investigators screened the entire BRCA1 gene for pathogenic variants using DNA from one individual from each of 93 pedigrees with evidence of prostate cancer linkage to 17q markers. Sixty-five of the individuals screened had wild-type BRCA1 sequence, and only one individual from a family with prostate and ovarian cancers was found to have a truncating pathogenic variant (3829delT). The remainder of the individuals harbored one or more germline BRCA1 variants, including 15 missense variants of uncertain clinical significance. The conclusion from these two reports is that there is evidence of a prostate cancer susceptibility gene on chromosome 17q near BRCA1; however, large deleterious inactivating variants in BRCA1 are not likely to be associated with prostate cancer risk in chromosome 17–linked families.

Another study from the UM-PCGP examined common genetic variation in BRCA1. Conditional logistic regression analysis and family-based association tests were performed in 323 familial prostate cancer families and early-onset prostate cancer families, which included 817 men with and without the disease, to investigate the association of single nucleotide variants (SNVs) tagging common haplotype variation in a 200-kb region surrounding and including BRCA1. Three SNVs in BRCA1 (rs1799950, rs3737559, and rs799923) were found to be associated with prostate cancer. The strongest association was observed for SNV rs1799950 (OR, 2.25; 95% CI, 1.21–4.20), which leads to a glutamine-to-arginine substitution at codon 356 (Gln356Arg) of exon 11 of BRCA1. Furthermore, SNV rs1799950 was found to contribute to the linkage signal on chromosome 17q21 originally reported by the UM-PCGP.

HOXB13

Key points

HOXB13 was the first gene found to be associated with hereditary prostate cancer. The HOXB13 G84E variant has been extensively studied because of its association with prostate cancer risk.

  • Overall risk of prostate cancer with the G84E variant ranges from 3- to 5-fold, with a higher risk of early-onset prostate cancer with the G84E variant of up to 10-fold.
  • Penetrance for carriers of the G84E variant is an approximate 60% lifetime risk of prostate cancer by age 80 years.
  • There is no clear association of the G84E variant with aggressive prostate cancer or other cancers.
  • Preliminary studies suggest additional variants in HOXB13 may be relevant for prostate cancer risk in diverse populations.

Background

Linkage to 17q21-22 was initially reported by the UM-PCGP from 175 pedigrees of families with hereditary prostate cancer. Fine-mapping of this region provided strong evidence of linkage (LOD score, 5.49) and a narrow candidate interval (15.5 Mb) for a putative susceptibility gene among 147 families with four or more affected men and average age at diagnosis of 65 years or younger. The exons of 200 genes in the 17q21-22 region were sequenced in DNA from 94 unrelated patients from hereditary prostate cancer families (from the UM-PCGP and Johns Hopkins University). Probands from four families were discovered to have a recurrent pathogenic variant (G84E) in HOXB13, and 18 men with prostate cancer from these four families carried the pathogenic variant. The pathogenic variant status was determined in 5,083 additional cases and 2,662 controls. Carrier frequencies and ORs for prostate cancer risk were as follows:

  • Men with a positive family history of prostate cancer, 2.2% versus negative, 0.8% (OR, 2.8; 95% CI, 1.6–5.1; P = 1.2 × 10-4).
  • Men younger than 55 years at diagnosis, 2.2% versus older than 55 years, 0.8% (OR, 2.7; 95% CI, 1.6–4.7; P = 1.1 × 10-4).
  • Men with a positive family history of prostate cancer and younger than 55 years at diagnosis, 3.1% versus a negative family history of prostate cancer and age at diagnosis older than 55 years, 0.6% (OR, 5.1; 95% CI, 2.4–12.2; P = 2.0 × 10-6).
  • Men with a positive family history of prostate cancer and older than 55 years at diagnosis, 1.2%.
  • Controls, 0.1% to 0.2%.

Validation and confirmatory studies

A validation study from the International Consortium of Prostate Cancer Genetics confirmed HOXB13 as a susceptibility gene for prostate cancer risk. Within carrier families, the G84E pathogenic variant was more common among men with prostate cancer than among unaffected men (OR, 4.42; 95% CI, 2.56–7.64). The G84E pathogenic variant was also significantly overtransmitted from parents to affected offspring (P = 6.5 × 10-6).

Additional studies have emerged that better define the carrier frequency and prostate cancer risk associated with the HOXB13 G84E pathogenic variant. This pathogenic variant appears to be restricted to White men, primarily of European descent. The highest carrier frequency of 6.25% was reported in Finnish early-onset cases. A pooled analysis of European Americans that included 9,016 cases and 9,678 controls found an overall G84E pathogenic variant frequency of 1.34% among cases and 0.28% among controls.

Risk of prostate cancer by HOXB13 G84E pathogenic variant status has been reported to vary by age of onset, family history, and geographical region. A validation study in an independent cohort of 9,988 cases and 61,994 controls from six studies of men of European ancestry, including 4,537 cases and 54,444 controls from Iceland whose genotypes were largely imputed, reported an OR of 7.06 (95% CI, 4.62–10.78; P = 1.5 × 10−19) for prostate cancer risk by G84E carrier status. A pooled analysis reported a prostate cancer OR of 4.86 (95% CI, 3.18–7.69; P = 3.48 × 10-17) in men with HOXB13 pathogenic variants compared with noncarriers; this increased to an OR of 8.41 (95% CI, 5.27–13.76; P = 2.72 ×10-22) among men diagnosed with prostate cancer at age 55 years or younger. The OR was 7.19 (95% CI, 4.55–11.67; P = 9.3 × 10-21) among men with a positive family history of prostate cancer and 3.09 (95% CI, 1.83–5.23; P = 6.26 × 10-6) among men with a negative family history of prostate cancer. A meta-analysis that included 24,213 cases and 73,631 controls of European descent revealed an overall OR for prostate cancer by carrier status of 4.07 (95% CI, 3.05–5.45; P< .00001). Risk of prostate cancer varied by geographical region: United States (OR, 5.10; 95% CI, 3.21–8.10; P< .00001), Canada (OR, 5.80; 95% CI, 1.27–26.51; P = .02), Northern Europe (OR, 3.61; 95% CI, 2.81–4.64; P< .00001), and Western Europe (OR, 8.47; 95% CI, 3.68–19.48; P< .00001). In addition, the association between the G84E pathogenic variant and prostate cancer risk was higher for early-onset cases (OR, 10.11; 95% CI, 5.97–17.12). There was no significant association with aggressive disease in the meta-analysis.

Another meta-analysis that included 11 case-control studies also reported higher risk estimates for prostate cancer in HOXB13 G84E carriers (OR, 4.51; 95% CI, 3.28–6.20; P< .00001) and found a stronger association between HOXB13 G84E and early-onset disease (OR, 9.73; 95% CI, 6.57–14.39; P< .00001). An additional meta-analysis of 25 studies that included 51,390 cases and 93,867 controls revealed an OR for prostate cancer of 3.248 (95% CI, 2.121–3.888). The association was most significant in White individuals (OR, 2.673; 95% CI, 1.920–3.720), especially those of European descent. No association was found for breast or colorectal cancer. One population-based, case-control study from the United States confirmed the association of the G84E pathogenic variant with prostate cancer (OR, 3.30; 95% CI, 1.21–8.96) and reported a suggestive association with aggressive disease. In addition, one study identified no men of AJ ancestry who carried the G84E pathogenic variant. A case-control study from the United Kingdom that included 8,652 cases and 5,252 controls also confirmed the association of HOXB13 G84E with prostate cancer (OR, 2.93; 95% CI, 1.94–4.59; P = 6.27 × 10-8). The risk was higher among men with a family history of the disease (OR, 4.53; 95% CI, 2.86–7.34; P = 3.1 × 10−8) and in early-onset prostate cancer (diagnosed at age 55 y or younger) (OR, 3.11; 95% CI, 1.98–5.00; P = 6.1 × 10−7). No association was found between carrier status and Gleason score, cancer stage, OS, or cancer-specific survival.

However, a 2018 publication of a study combining multiple prostate cancer cases and controls of Nordic origin along with functional analysis reported that simultaneous presence of HOXB13 (G84E) and CIP2A (R229Q) predisposes men to an increased risk of prostate cancer (OR, 21.1; P = .000024). Furthermore, dual carriers had elevated risk for high Gleason score (OR, 2.3; P = .025) and worse prostate cancer–specific survival (hazard ratio [HR], 3.9; P = .048). Clinical validation is needed.

HOXB13 pathogenic variants in diverse populations

A study of Chinese men with and without prostate cancer failed to identify the HOXB13 G84E pathogenic variant; however, there was an excess of a novel variant, G135E, in cases compared with controls. A large study of approximately 20,000 Japanese men with and without prostate cancer identified another novel HOXB13 variant, G132E, which was associated with prostate cancer with an OR of 6.08 (95% CI, 3.39–11.59).

Two studies confirmed the association between the HOXB13 X285K variant and increased prostate cancer risk in African American men after this variant was identified in Martinique. One of these was a single-institution study, which sequenced HOXB13 in a clinical patient population of 1,048 African American men undergoing prostatectomy for prostate cancer. The HOXB13 X285K variant was identified in eight patients. In a case–case analysis, X285K variant carriers were at increased risk of developing clinically significant prostate cancer (1.2% X285K carrier rate in prostate cancers with a Gleason score ≥7 vs. 0% X285K carrier rate in prostate cancers with Gleason score <7; P = .028). Similarly, X285K variant carriers also had an increased chance of developing prostate cancer at an early age (2.4% X285K carrier rate in patients <50 years vs. 0.5% X285K carrier rate in patients ≥50 years; OR, 5.25; 95% CI, 1.00–28.52; P = .03). A second study included 11,688 prostate cancer cases and 10,673 controls from multiple large consortia. The HOXB13 X285K variant was only present in men of West African ancestry and was associated with a 2.4-fold increased chance of developing prostate cancer (95% CI, 1.5–3.9; P = 2 x 10-4). Individuals with the X285K variant were also more likely to have aggressive and advanced prostate cancer (Gleason score ≥8: OR, 4.7; 95% CI, 2.3–9.5; P = 2 x 10-5; stage T3/T4: OR, 4.5; 95% CI, 2.0–10.0; P = 2 x 10-4; metastatic disease: OR, 5.1; 95% CI, 1.9–13.7; P = .001). This information is important to consider when developing genetic tests for HOXB13 pathogenic variants in broader populations.

Penetrance

Penetrance estimates for prostate cancer development in carriers of the HOXB13 G84E pathogenic variant are also being reported. One study from Sweden estimated a 33% lifetime risk of prostate cancer among G84E carriers. Another study from Australia reported an age-specific cumulative risk of prostate cancer of up to 60% by age 80 years. A study in the United Kingdom that included HOXB13 genotype data from nearly 12,000 men with prostate cancer enrolled between 1993 and 2014 reported that the average predicted risk of prostate cancer by age 85 years is 62% (95% CI, 47%–76%) for carriers of the G84E pathogenic variant. The risk of developing prostate cancer in variant carriers increased if the men had affected family members, especially those diagnosed at an early age.

Biology

HOXB13 plays a role in prostate cancer development and interacts with the androgen receptor; however, the mechanism by which it contributes to the pathogenesis of prostate cancer remains unknown. This is the first gene identified to account for a fraction of hereditary prostate cancer, particularly early-onset prostate cancer. The clinical utility and implications for genetic counseling regarding HOXB13 G84E or other pathogenic variants have yet to be defined.

DNA mismatch repair genes (Lynch syndrome)

Five genes are implicated in mismatch repair (MMR), namely MLH1, MSH2, MSH6, PMS2, and EPCAM. Germline pathogenic variants in these five genes have been associated with Lynch syndrome, which manifests by cases of nonpolyposis colorectal cancer and a constellation of other cancers in families, including endometrial, ovarian, duodenal cancers, and transitional cell cancers of the ureter and renal pelvis. For more information about other cancers that are associated with Lynch syndrome, see the Lynch syndrome section in Genetics of Colorectal Cancer. Reports have suggested that prostate cancer may be observed in men harboring an MMR gene pathogenic variant. The first quantitative study described nine cases of prostate cancer occurring in a population-based cohort of 106 Norwegian male carriers of MMR gene pathogenic variants or obligate carriers. The expected number of cases among these 106 men was 1.52 (P< .01); the men were younger at the time of diagnosis (60.4 y vs. 66.6 y; P = .006) and had more evidence of Gleason score of 8 to 10 (P< .00001) than the cases from the Norwegian Cancer Registry. Kaplan-Meier analysis revealed that the cumulative risk of prostate cancer diagnosis by age 70 years was 30% in carriers of MMR gene pathogenic variants and 8% in the general population. This finding awaits confirmation in additional populations. A population-based case-control study examined haplotype-tagging SNVs in three MMR genes (MLH1, MSH2, and PMS2). This study provided some evidence supporting the contribution of genetic variation in MLH1 and overall risk of prostate cancer. To assess the contribution of prostate cancer as a feature of Lynch syndrome, one study performed microsatellite instability (MSI) testing on prostate cancer tissue blocks from families enrolled in a prostate cancer family registry who also reported a history of colon cancer. Among 35 tissue blocks from 31 distinct families, two tumors from families with MMR gene pathogenic variants were found to be MSI-high. The authors conclude that MSI is rare in hereditary prostate cancer. Other studies are attempting to characterize rates of prostate cancer in Lynch syndrome families and correlate molecular features with prostate cancer risk.

One study that included two familial cancer registries found an increased cumulative incidence and risk of prostate cancer among 198 independent families with MMR gene pathogenic variants and Lynch syndrome. The cumulative lifetime risk of prostate cancer (to age 80 y) was 30.0% (95% CI, 16.54%–41.30%; P = .07) in carriers of MMR gene pathogenic variants, whereas it was 17.84% in the general population, according to the Surveillance, Epidemiology, and End Results (SEER) Program estimates. There was a trend of increased prostate cancer risk in carriers of pathogenic variants by age 50 years, where the risk was 0.64% (95% CI, 0.24%–1.01%; P = .06), compared with a risk of 0.26% in the general population. Overall, the HR (to age 80 y) for prostate cancer in carriers of MMR gene pathogenic variants in the combined data set was 1.99 (95% CI, 1.31–3.03; P = .0013). Among men aged 20 to 59 years, the HR was 2.48 (95% CI, 1.34–4.59; P = .0038).

A systematic review and meta-analysis that included 23 studies (6 studies with molecular characterization and 18 risk studies, of which 12 studies quantified risk for prostate cancer) reported an association of prostate cancer with Lynch syndrome. In the six molecular studies included in the analysis, 73% (95% CI, 57%–85%) of prostate cancers in carriers of MMR gene pathogenic variants were MMR deficient. The RR of prostate cancer in carriers of MMR gene pathogenic variants was estimated to be 3.67 (95% CI, 2.32–6.67). Of the twelve risk studies, the RR of prostate cancer ranged from 2.11 to 2.28, compared with that seen in the general population depending on carrier status, prior diagnosis of colorectal cancer, or unknown male carrier status from families with a known pathogenic variant.

A study from three sites participating in the Colon Cancer Family Registry examined 32 cases of prostate cancer (mean age at diagnosis, 62 y; standard deviation, 8 y) in men with a documented MMR gene pathogenic variant (23 MSH2 carriers, 5 MLH1 carriers, and 4 MSH6 carriers). Seventy-two percent (n = 23) had a previous diagnosis of colorectal cancer. Immunohistochemistry was used to assess MMR protein loss, which was observed in 22 tumors (69%); the pattern of loss of protein expression was 100% concordant with the germline pathogenic variant. The RR of prostate cancer was highest in carriers of MSH2 pathogenic variants (RR, 5.8; 95% CI, 2.6–20.9); the RRs in carriers of MLH1 and MSH6 pathogenic variants were 1.7 (95% CI, 1.1–6.7) and 1.3 (95% CI, 1.1–5.3), respectively. Gleason scores ranged from 5 to 10; two tumors had a Gleason score of 5; 22 tumors had a Gleason score of 6 or 7; and eight tumors had a Gleason score higher than 8. Sixty-seven percent (12 of 18) of the tumors were found to have perineural invasion, and 47% (9 of 19) had extracapsular invasion. A large observational cohort study, which included more than 6,000 MMR-variant carriers, reported an increased cumulative incidence of prostate cancer by age 70 years for specific MMR genes, as follows: MLH1 (7.0; 95% CI, 4.2–11.9), MSH2 (15.9; 95% CI, 11.2–22.5), and PMS2 (4.6; 95% CI, 0.8–67.5). No significant increase in prostate cancer incidence was reported for MSH6.

Although the risk of prostate cancer appears to be elevated in families with Lynch syndrome, strategies for germline testing for MMR gene pathogenic variants in index prostate cancer patients remain to be determined.

A study of 1,133 primary prostate adenocarcinomas and 43 neuroendocrine prostate cancers (NEPC) conducted screening by MSH2 immunohistochemistry with confirmation by NGS. MSI was assessed by polymerase chain reaction and NGS. Of primary adenocarcinomas and NEPC, 1.2% (14/1,176) had MSH2 loss. Overall, 8% (7/91) of adenocarcinomas with primary Gleason pattern 5 (Gleason score 9–10) had MSH2 loss compared with 0.4% (5/1,042) of tumors with any other Gleason scores (P< .05). Three patients had germline variants in MSH2, of whom two had a primary Gleason score of 5. Pending further confirmation, these findings may support universal MMR screening of prostate cancer with a Gleason score of 9 to 10 to identify men who may be eligible for immunotherapy and germline testing.

EPCAM testing has been included in some multigene panels likely due to EPCAM variants silencing MSH2. Specific large genomic rearrangement variants at the 3’ end of EPCAM (which lies near the MSH2 gene) induce methylation of the MSH2 promoter, resulting in MSH2 protein loss. Pathogenic variants in MSH2 are associated with Lynch syndrome and an increase in prostate cancer risk. For more information on EPCAM and MSH2, see the Gene-specific considerations and associated CRC risk section or the Lynch Syndrome section in Genetics of Colorectal Cancer. Thus far, studies have not found an association between increased prostate cancer risk and EPCAM pathogenic variants.

ATM

Ataxia telangiectasia (AT) is an autosomal recessive disorder characterized by neurological deterioration, telangiectasias, immunodeficiency states, and hypersensitivity to ionizing radiation. It is estimated that 1% of the general population may be heterozygous carriers of ATM pathogenic variants. In the presence of DNA damage, the ATM protein is involved in mediating cell cycle arrest, DNA repair, and apoptosis. Given evidence of other cancer risks in heterozygous ATM carriers, evidence of an association with prostate cancer susceptibility continues to emerge. A prospective case series of 10,317 Danish individuals who had a 36-year follow-up period, during which 2,056 individuals developed cancer, found that the ATM Ser49Cys variant was associated with increased prostate cancer risk (HR, 2.3; 95% CI, 1.1–5.0). A retrospective case series of 692 men with metastatic prostate cancer, who were not selected based on a family history of cancer or the patient's age at cancer diagnosis, found that 1.6% of participants (11 of 692) had an ATM pathogenic variant. Multiple independent reports have shown that the ATM P1054R variant, which is found in 2% of Europeans, is associated with increased prostate cancer risk. For example, the Prostate Cancer Association Group to Investigate Cancer Associated Alterations in the Genome (PRACTICAL) consortium found an OR of 1.16 (95% CI, 1.10–1.22) for the ATM P1054 variant's association with prostate cancer risk. A subsequent PRACTICAL consortium study had 14 groups (five from North America, six from Europe, and two from Australia) and 8,913 participants (5,560 cases and 3,353 controls). Next-generation ATM sequencing data were standardized and ClinVar classifications were used to categorize the variants as Tier 1 (likely pathogenic) or Tier 2 (potentially deleterious). Prostate cancer risk in Tier 1 variants had an OR of 4.4 (95% CI, 2.0–9.5).

CHEK2

CHEK2 has also been investigated for a potential association with prostate cancer risk. For more information on other cancers associated with CHEK2 pathogenic variants, see the CHEK2 section in Genetics of Breast and Gynecologic Cancers and the CHEK2 section in Genetics of Colorectal Cancer. A retrospective case series of 692 men with metastatic prostate cancer unselected for cancer family history or age at diagnosis found 1.9% (10 of 534 [men with data]) were found to have a CHEK2 pathogenic variant. A systematic review and meta-analysis from eight retrospective cohort studies examining the relationship between CHEK2 variants (1100delC, IVS2+1G>A, I157T) and prostate cancer confirmed the association of the 1100delC (OR, 3.29; 95% CI, 1.85–5.85; P = .00) and I157T (OR, 1.80; 95% CI, 1.51–2.14; P = .00) variants with prostate cancer susceptibility. A genome-wide association study (GWAS) focusing on African American cases and controls identified a missense variant, I448S, which is associated with prostate cancer (risk allele frequency, 1.5%; OR, 1.62; 95% CI, 1.39–1.89, P = 7.50 × 10-10). Further studies of CHEK2 in large diverse populations are warranted.

TP53

TP53 has also been investigated for a potential association with prostate cancer risk. For more information about other cancers associated with TP53 pathogenic variants, see the Li-Fraumeni Syndrome section in Genetics of Breast and Gynecologic Cancers. In a case series of 286 individuals from 107 families with a deleterious TP53 variant, 403 cancer diagnoses were reported, of which 211 were the first primary cancer including two prostate cancers diagnosed after age 45 years. Prostate cancer was also reported in 4 of 61 men with a second primary cancer. In a Dutch case series of 180 families meeting either classic Li-Fraumeni syndrome (LFS) or Li-Fraumeni–like (LFL) family history criteria, a deleterious TP53 variant was identified in 24 families with one case of prostate cancer found in each group (LFS or LFL). Prostate cancer risks varied on the basis of the family history criteria with LFS (RR, 0.50; 95% CI, 0.01–3.00) and LFL (RR, 4.90; 95% CI, 0.10–27.00). In a French case series of 415 families with a deleterious TP53 variant, four prostate cancers were reported, with a mean age at diagnosis of 63 years (range, 57–71 y).

Germline TP53 pathogenic variants have also been identified in men with prostate cancer who have undergone tumor testing. A prospective case series of 42 men with either localized, biochemically recurrent, or metastatic prostate cancer unselected for cancer family history or age at diagnosis undergoing tumor-only somatic testing found that 2 of 42 men (5%) were found to have a suspected TP53 germline pathogenic variant.

Further evidence supports an association between prostate cancer and germline TP53 pathogenic variants, although additional studies to clarify the association with this gene are warranted.

NBN/NBS1

NBN, which is also known as NBS1, has been investigated for a potential association with risk of prostate cancer. A retrospective case series of 692 men with metastatic prostate cancer unselected for cancer family history or age at diagnosis found that 0.3% (2 of 692 men) had an NBN pathogenic variant. A prospective cohort of men with prostate cancer diagnosed between 1999 and 2015 in Poland confirmed the association of the NBN 657del5 variant and prostate cancer (OR, 2.5; P< .001) and mortality (HR, 1.6; P = .001), which remained significant after adjusting for age at diagnosis, PSA, stage, and grade. The risk of prostate cancer in NBN 657del5 carriers is influenced by the genotype at the E185Q polymorphism.

Multigene testing studies in prostate cancer

Prevalence of pathogenic variants with prostate cancer risk on multigene panel testing

The following section gives information about additional genes that may be on hereditary prostate cancer panel tests.

One retrospective case series of 692 men with metastatic prostate cancer unselected for cancer family history or age at diagnosis assessed the incidence of germline pathogenic variants in 16 DNA repair genes. Pathogenic variants were identified in 11.8% (82 of 692), a rate higher than in men with localized prostate cancer (4.6%, P< .001), suggesting that genetic aberrations are more commonly observed in men with aggressive forms of disease. Two studies were published using data from a clinical testing laboratory database. The first study evaluated 1,328 men with prostate cancer and reported an overall pathogenic variant rate of 15.6%, including 10.9% in DNA repair genes. A second study involved a larger cohort of 3,607 men with prostate cancer, some of whom had been included in the prior publication. The reported pathogenic variant rate was 17.2%. Overall, pathogenic variant rates by gene were consistently reported between the two studies and were as follows: BRCA2, 4.74%; CHEK2, 2.88%; ATM, 2.03%; and BRCA1, 1.25%. The most commonly aberrant gene in this cohort was BRCA2. The first publication reported associations between family history of breast cancer and high Gleason score (≥8). The second publication focused on the percentage of men with pathogenic variants who met National Comprehensive Cancer Network national guidelines for genetic testing and found that 229 individuals (37%) with pathogenic variants in this cohort did not meet guidelines for genetic testing. A systematic evidence review examined the median prevalence of pathogenic germline variants in the DNA damage-response pathway, including ATM, ATR, BRCA1, BRCA2, CHEK2, FANCA, MLH1, MRE11A, NBN, PALB2, and RAD51C. The overall prevalence was 18.6% (range, 17.2%–19%; n = 1,712) for general prostate cancer, 11.6% (range, 11.4%–11.8%; n = 1,261) for metastatic prostate cancer, 8.3% (range, 7.5%–9.1%; n = 738) for metastatic castration-resistant prostate cancer, and 29.3% (range, 7.3%–92.67%; n = 327) for familial prostate cancer.

A case-control study in a Japanese population of 7,636 men with prostate cancer and 12,366 men without prostate cancer evaluated pathogenic variants in eight genes (BRCA1, BRCA2, CHEK2, ATM, NBN, PALB2, HOXB13, and BRIP1) for an association with prostate cancer. The study found strong associations for BRCA2 (OR, 5.65; 95% CI, 3.55–9.32), HOXB13 (OR, 4.73; 95% CI, 2.84–8.19), and ATM (OR, 2.86; 95% CI, 1.63–5.15). The study supports a population-specific assessment of the genetic contribution to prostate cancer risk.

Germline pathogenic variants associated with metastatic prostate cancer

The metastatic prostate cancer setting is also contributing insights into the germline pathogenic variant spectrum of prostate cancer. Clinical sequencing of 150 metastatic tumors from men with castrate-resistant prostate cancer identified alterations in genes involved in DNA repair in 23% of men. Interestingly, 8% of these variants were pathogenic and present in the germline. Another study focused on tumor-normal sequencing of advanced and metastatic cancers identified germline pathogenic variants in 19.6% of men (71 of 362) with prostate cancer. Germline pathogenic variants were found in BRCA1, BRCA2, MSH2, MSH6, PALB2, PMS2, ATM, BRIP1, NBN, as well as other genes. These and other studies are summarized in Table 9. The contribution of germline variants identified from large sequencing efforts to inherited prostate cancer predisposition requires molecular confirmation of genes not classically linked to prostate cancer risk.

Table 9. Summary of Tumor Sequencing Studies With Germline Findings

StudyCohortGermline Results for Prostate CancerComments
mCRPC = metastatic castration-resistant prostate cancer.
aPotential overlap of cohorts.
Robinson et al. (2015)aWhole-exome and transcriptome sequencing of bone or soft tissue tumor biopsies from a cohort of 150 men with mCRPC 8% had germline pathogenic variants:
BRCA2: 9/150 (6.0%)
ATM: 2/150 (1.3%)
BRCA1: 1/150 (0.7%)
Pritchard et al. (2016)a692 men with metastatic prostate cancer, unselected for family history; analysis focused on 20 genes involved in maintaining DNA integrity and associated with autosomal dominant cancer–predisposing syndromes 82/692 (11.8%) had germline pathogenic variants: Frequency of germline pathogenic variants in DNA repair genes among men with metastatic prostate cancer significantly exceeded the prevalence of 4.6% among 499 men with localized prostate cancer in the Cancer Genome Atlas (P < .001)
BRCA2: 37/692 (5.3%)
ATM: 11/692 (1.6%)
BRCA1: 6/692 (0.9%)
Schrader et al. (2016) 1,566 patients undergoing tumor profiling (341 genes) with matched normal DNA at a single institution; 97 cases of prostate cancer included10/97 (10.3%) had germline pathogenic variants:
BRCA2: 6/97 (6.2%)
BRCA1: 1/97 (1.0%)
MSH6: 1/97 (1.0%)
MUTYH: 1/97 (1.0%)
PMS2: 1/97 (1.0%)

Common Risk Variants and Polygenic Risk Scores for Prostate Cancer

GWAS and SNPs

  • GWAS can identify inherited genetic variants that influence a specific phenotype, such as risk of a particular disease.
  • For complex diseases, such as prostate cancer, risk of developing the disease is the product of multiple genetic and environmental factors; each individual factor contributes relatively little to overall risk.
  • To date, GWAS have discovered more than 250 common genetic variants associated with prostate cancer risk.
  • Individuals can be genotyped for all known prostate cancer risk markers relatively easily; but, to date, studies have not demonstrated that this information substantially refines risk estimates from commonly used variables, such as family history.
  • The clinical relevance of variants identified from GWAS remains unclear.

Although the statistical evidence for an association between genetic variation at these loci and prostate cancer risk is overwhelming, the clinical relevance of the variants and the mechanism(s) by which they lead to increased risk are unclear and will require further characterization. Additionally, these loci are associated with very modest risk estimates and explain only a fraction of overall inherited risk. However, when combined into a polygenic risk score (PRS), these confirmed genetic risk variants may prove to be useful for prostate cancer risk stratification and to identify men for targeted screening and early detection. Further work will include genome-wide analysis of rarer alleles catalogued via sequencing efforts. Disease-associated alleles with frequencies of less than 1% in the population may prove to be more highly penetrant and clinically useful. In addition, further work is needed to describe the landscape of genetic risk in non-European populations. Finally, until the individual and collective influences of genetic risk alleles are evaluated prospectively, their clinical utility will remain difficult to fully assess.

Beginning in 2006, multiple genome-wide studies seeking associations with prostate cancer risk converged on the same chromosomal locus, 8q24. Since that time, more than ten genetic polymorphisms, all independently associated with disease, reside within five distinct 8q24 risk regions. The population-attributable risk of prostate cancer from the 8q24 risk alleles reported to date is 9.4%.

Since prostate cancer risk loci have been discovered at 8q24, more than 250 variants have been identified at other chromosomal risk loci. These chromosomal risk loci were detected by multistage GWAS, which were comprised of thousands of cases and controls and were validated in independent cohorts. The most convincing associations reported to date for men of European ancestry are annotated in the National Human Genome Research Institute GWAS catalog.

Most prostate cancer GWAS data generated to date have been derived from populations of European descent. This shortcoming is profound, considering that linkage disequilibrium structure, SNV frequencies, and incidence of disease differ across ancestral groups. To provide meaningful genetic data to all patients, well-designed, adequately powered GWAS must be aimed at specific ethnic groups. Most work in this regard has focused on African American, Chinese, and Japanese men. The most convincing associations reported to date for men of non-European ancestry are annotated in the National Human Genome Research Institute GWAS catalog.

The African American population is of particular interest because American men with West African ancestry are at higher risk of prostate cancer than any other group. A handful of studies have sought to determine whether GWAS findings in men of European ancestry are applicable to men of African ancestry. The majority of risk alleles (approximately 83%) are shared across African American and European American populations. Three independent associations were subsequently replicated. All three variants were within or near long noncoding RNAs (lncRNAs) previously associated with prostate cancer, and two of the variants were unique to men of African ancestry.

Statistically well-powered GWAS have also been launched to examine inherited cancer risk in Japanese and Chinese populations. Investigators discovered that these populations share many risk regions observed in African American men. Additionally, risk regions that are unique to these ancestral groups were identified (for more information, see the National Human Genome Research Institute GWAS catalog). Ongoing work in larger cohorts will validate and expand upon these findings.

Polygenic risk scores for prostate cancer

Current GWAS findings account for an estimated 58% of disease risk that is heritable. About 6% of the familial RR of prostate cancer has been attributed to rare genetic variants. Ongoing research attempts to uncover the remaining portion of genetic risk. In the meantime, efforts have been made to utilize the established portions of inherited prostate cancer and create clinically useful metrics for disease risk. Risk variant burden within individuals subjects has been used to generate a PRS. Associations between PRS and disease risk clearly exist. However, it remains unclear whether screening PRSs can appreciably influence long-term outcomes.

In a 2018 study, 147 GWAS variants known to be associated with prostate cancer were used to calculate a PRS for more than 140,000 men. In 2021, this scoring system, which accounts for genetic dose (i.e., homozygosity vs. heterozygosity) and strength of prostate cancer risk association, was applied to a multi-ethnic cohort of over 100,000 prostate cancer cases and 100,000 controls. This scoring system used 269 known risk variants. In this study, the PRS was called a genetic risk score (GRS). When focusing on men in the top decile of GRSs and comparing them to men in the middle of the distribution, men with European ancestry had an OR of 5.06 (95% CI, 4.84–5.29), and men with African ancestry had an OR of 3.74 (95% CI, 3.36–4.17). When comparing across ethnic groups (with individuals of European, African, East Asian, and Hispanic ancestries) and across all deciles, men with African ancestry were at the highest risk of developing prostate cancer, with a mean GRS that was 2.18-times higher than that of men with European ancestry. Men with East Asian ancestry had a mean GRS that was 0.73-times lower than that of men with European ancestry.

The prostate cancer PRS's predictive value was maintained when it was applied to populations of men who carry deleterious variants in BRCA1 or BRCA2; this was particularly true among those in the top 95% distribution of the PRS. However, initial studies suggested that these associations were modest when compared with those in the general population. This is likely because BRCA1 and BRCA2 carriers have a substantially increased risk of developing prostate cancer than individuals in the general population.

The Stockholm-3 Model (S3M) was developed on the basis of a study of 58,000 Swedish men aged 50 to 69 years. Men were genotyped for 233 prostate cancer risk–associated variants, and these data were used with other clinical data to risk-stratify men. Compared with PSA alone (area under the curve [AUC], 0.56), the addition of SNVs to clinical factors (S3M) improved prediction (AUC, 0.75) of clinically significant (i.e., Gleason score ≥7) prostate cancer. Another community-based study (BARCODE1) of 5,000 men aged 55 to 69 years in the United Kingdom involves genotyping for 167 risk SNVs, with men in the top 10% of the PRS undergoing prostate biopsies. This study should provide additional information on the potential clinical utility of the PRS for guiding prostate cancer screening protocols. PRSs have been shown to be additive to risk attributed to rare pathogenic alleles, including BRCA1/BRCA2 and HOXB13.

In 2021, a prospective study was done on participants from the U.K. Biobank. This cohort consisted of 208,685 men (mostly of European ancestry). Results suggested that prostate cancer risk–associated single nucleotide polymorphisms (SNPs) can provide useful information when they are added to an individual's family history and rare pathogenic variant status. SNP carriers in the high-risk quartile had an increased risk to develop prostate cancer (RR, 1.97; 95% CI, 1.87–2.07). Men in the high-risk quartile also had an increased C-statistic for differentiating prostate cancer incidence when prostate cancer risk–associated SNP burden was added to family history and rare pathogenic variant status (C-statistic for family history, 0.58; C-statistic for pathogenic variant status, 0.67).

Research focused on the associated risk of prostate cancer and the predictability of PRSs is ongoing. In an independent cohort of over 13,000 men, a panel of 261 GWAS-derived risk variants significantly predicted disease risk. Disease risk was best predicted at the highest and lowest deciles, where the highest decile represented the largest risk variant burden, and the lowest decile represented the smallest risk variant burden. In the top decile, the OR for prostate cancer diagnosis was 3.81 (95% CI, 1.48–10.19) in men of African ancestry and 3.89 (95% CI, 3.24– 4.68) in men of European ancestry when compared with men who were at an average risk of developing prostate cancer. In the lowest decile, the ORs were 0.15 (95% CI, 0.01–0.92) and 0.34 (95% CI, 0.25–0.46) in men of African ancestry and men of European ancestry, respectively.

As the full picture of inherited prostate cancer risk becomes more complete, it is hoped that germline information will become clinically useful. Finally, GWAS are providing more insight into the mechanism of prostate cancer risk. It is now apparent that a large proportion of risk variants affect the activity of regulatory elements and, in turn, distal genes. As GWAS elucidate these networks, it is hoped that new therapies and chemopreventive strategies will follow.

Germline SNPs associated with prostate cancer aggressiveness

Prostate cancer is biologically and clinically heterogeneous. Many tumors are indolent and are successfully managed with observation alone. Other tumors are quite aggressive and prove deadly. Several variables are used to determine prostate cancer aggressiveness at the time of diagnosis, such as Gleason score and PSA, but these are imperfect. Additional markers are needed because sound treatment decisions depend on accurate prognostic information. Germline genetic variants are attractive markers because they are present, easily detectable, and static throughout life.

Findings regarding inherited risk of aggressive disease are considered preliminary. Further work is needed to validate findings and assess these associations prospectively.

References

  1. Thompson D, Easton DF; Breast Cancer Linkage Consortium: Cancer Incidence in BRCA1 mutation carriers. J Natl Cancer Inst 94 (18): 1358-65, 2002.
  2. Liede A, Karlan BY, Narod SA: Cancer risks for male carriers of germline mutations in BRCA1 or BRCA2: a review of the literature. J Clin Oncol 22 (4): 735-42, 2004.
  3. Mersch J, Jackson MA, Park M, et al.: Cancers associated with BRCA1 and BRCA2 mutations other than breast and ovarian. Cancer 121 (2): 269-75, 2015.
  4. Cancer risks in BRCA2 mutation carriers. The Breast Cancer Linkage Consortium. J Natl Cancer Inst 91 (15): 1310-6, 1999.
  5. Thompson D, Easton D; Breast Cancer Linkage Consortium: Variation in cancer risks, by mutation position, in BRCA2 mutation carriers. Am J Hum Genet 68 (2): 410-9, 2001.
  6. Silvestri V, Leslie G, Barnes DR, et al.: Characterization of the Cancer Spectrum in Men With Germline BRCA1 and BRCA2 Pathogenic Variants: Results From the Consortium of Investigators of Modifiers of BRCA1/2 (CIMBA). JAMA Oncol 6 (8): 1218-1230, 2020.
  7. Li S, Silvestri V, Leslie G, et al.: Cancer Risks Associated With BRCA1 and BRCA2 Pathogenic Variants. J Clin Oncol 40 (14): 1529-1541, 2022.
  8. Ostrander EA, Udler MS: The role of the BRCA2 gene in susceptibility to prostate cancer revisited. Cancer Epidemiol Biomarkers Prev 17 (8): 1843-8, 2008.
  9. Oh M, Alkhushaym N, Fallatah S, et al.: The association of BRCA1 and BRCA2 mutations with prostate cancer risk, frequency, and mortality: A meta-analysis. Prostate 79 (8): 880-895, 2019.
  10. Nastiuk KL, Mansukhani M, Terry MB, et al.: Common mutations in BRCA1 and BRCA2 do not contribute to early prostate cancer in Jewish men. Prostate 40 (3): 172-7, 1999.
  11. Vazina A, Baniel J, Yaacobi Y, et al.: The rate of the founder Jewish mutations in BRCA1 and BRCA2 in prostate cancer patients in Israel. Br J Cancer 83 (4): 463-6, 2000.
  12. Lehrer S, Fodor F, Stock RG, et al.: Absence of 185delAG mutation of the BRCA1 gene and 6174delT mutation of the BRCA2 gene in Ashkenazi Jewish men with prostate cancer. Br J Cancer 78 (6): 771-3, 1998.
  13. Struewing JP, Abeliovich D, Peretz T, et al.: The carrier frequency of the BRCA1 185delAG mutation is approximately 1 percent in Ashkenazi Jewish individuals. Nat Genet 11 (2): 198-200, 1995.
  14. Oddoux C, Struewing JP, Clayton CM, et al.: The carrier frequency of the BRCA2 6174delT mutation among Ashkenazi Jewish individuals is approximately 1%. Nat Genet 14 (2): 188-90, 1996.
  15. Roa BB, Boyd AA, Volcik K, et al.: Ashkenazi Jewish population frequencies for common mutations in BRCA1 and BRCA2. Nat Genet 14 (2): 185-7, 1996.
  16. Struewing JP, Hartge P, Wacholder S, et al.: The risk of cancer associated with specific mutations of BRCA1 and BRCA2 among Ashkenazi Jews. N Engl J Med 336 (20): 1401-8, 1997.
  17. Giusti RM, Rutter JL, Duray PH, et al.: A twofold increase in BRCA mutation related prostate cancer among Ashkenazi Israelis is not associated with distinctive histopathology. J Med Genet 40 (10): 787-92, 2003.
  18. Kirchhoff T, Kauff ND, Mitra N, et al.: BRCA mutations and risk of prostate cancer in Ashkenazi Jews. Clin Cancer Res 10 (9): 2918-21, 2004.
  19. Agalliu I, Gern R, Leanza S, et al.: Associations of high-grade prostate cancer with BRCA1 and BRCA2 founder mutations. Clin Cancer Res 15 (3): 1112-20, 2009.
  20. Gallagher DJ, Gaudet MM, Pal P, et al.: Germline BRCA mutations denote a clinicopathologic subset of prostate cancer. Clin Cancer Res 16 (7): 2115-21, 2010.
  21. Johannesdottir G, Gudmundsson J, Bergthorsson JT, et al.: High prevalence of the 999del5 mutation in icelandic breast and ovarian cancer patients. Cancer Res 56 (16): 3663-5, 1996.
  22. Eerola H, Pukkala E, Pyrhönen S, et al.: Risk of cancer in BRCA1 and BRCA2 mutation-positive and -negative breast cancer families (Finland). Cancer Causes Control 12 (8): 739-46, 2001.
  23. Cybulski C, Wokołorczyk D, Kluźniak W, et al.: An inherited NBN mutation is associated with poor prognosis prostate cancer. Br J Cancer 108 (2): 461-8, 2013.
  24. Cybulski C, Górski B, Gronwald J, et al.: BRCA1 mutations and prostate cancer in Poland. Eur J Cancer Prev 17 (1): 62-6, 2008.
  25. Agalliu I, Karlins E, Kwon EM, et al.: Rare germline mutations in the BRCA2 gene are associated with early-onset prostate cancer. Br J Cancer 97 (6): 826-31, 2007.
  26. Agalliu I, Kwon EM, Zadory D, et al.: Germline mutations in the BRCA2 gene and susceptibility to hereditary prostate cancer. Clin Cancer Res 13 (3): 839-43, 2007.
  27. Tryggvadóttir L, Vidarsdóttir L, Thorgeirsson T, et al.: Prostate cancer progression and survival in BRCA2 mutation carriers. J Natl Cancer Inst 99 (12): 929-35, 2007.
  28. Kote-Jarai Z, Leongamornlert D, Saunders E, et al.: BRCA2 is a moderate penetrance gene contributing to young-onset prostate cancer: implications for genetic testing in prostate cancer patients. Br J Cancer 105 (8): 1230-4, 2011.
  29. Leongamornlert D, Mahmud N, Tymrakiewicz M, et al.: Germline BRCA1 mutations increase prostate cancer risk. Br J Cancer 106 (10): 1697-701, 2012.
  30. Nyberg T, Frost D, Barrowdale D, et al.: Prostate Cancer Risks for Male BRCA1 and BRCA2 Mutation Carriers: A Prospective Cohort Study. Eur Urol 77 (1): 24-35, 2020.
  31. Edwards SM, Evans DG, Hope Q, et al.: Prostate cancer in BRCA2 germline mutation carriers is associated with poorer prognosis. Br J Cancer 103 (6): 918-24, 2010.
  32. Thorne H, Willems AJ, Niedermayr E, et al.: Decreased prostate cancer-specific survival of men with BRCA2 mutations from multiple breast cancer families. Cancer Prev Res (Phila) 4 (7): 1002-10, 2011.
  33. Castro E, Goh C, Olmos D, et al.: Germline BRCA mutations are associated with higher risk of nodal involvement, distant metastasis, and poor survival outcomes in prostate cancer. J Clin Oncol 31 (14): 1748-57, 2013.
  34. Akbari MR, Wallis CJ, Toi A, et al.: The impact of a BRCA2 mutation on mortality from screen-detected prostate cancer. Br J Cancer 111 (6): 1238-40, 2014.
  35. National Comprehensive Cancer Network: NCCN Clinical Practice Guidelines in Oncology: Genetic/Familial High-Risk Assessment: Breast, Ovarian, and Pancreatic. Version 2.2024. Plymouth Meeting, Pa: National Comprehensive Cancer Network, 2023. Available online with free registration. Last accessed October 31, 2023.
  36. Taylor RA, Fraser M, Livingstone J, et al.: Germline BRCA2 mutations drive prostate cancers with distinct evolutionary trajectories. Nat Commun 8: 13671, 2017.
  37. Narod SA, Neuhausen S, Vichodez G, et al.: Rapid progression of prostate cancer in men with a BRCA2 mutation. Br J Cancer 99 (2): 371-4, 2008.
  38. Castro E, Goh C, Leongamornlert D, et al.: Effect of BRCA Mutations on Metastatic Relapse and Cause-specific Survival After Radical Treatment for Localised Prostate Cancer. Eur Urol 68 (2): 186-93, 2015.
  39. Lange EM, Gillanders EM, Davis CC, et al.: Genome-wide scan for prostate cancer susceptibility genes using families from the University of Michigan prostate cancer genetics project finds evidence for linkage on chromosome 17 near BRCA1. Prostate 57 (4): 326-34, 2003.
  40. Zuhlke KA, Madeoy JJ, Beebe-Dimmer J, et al.: Truncating BRCA1 mutations are uncommon in a cohort of hereditary prostate cancer families with evidence of linkage to 17q markers. Clin Cancer Res 10 (18 Pt 1): 5975-80, 2004.
  41. Douglas JA, Levin AM, Zuhlke KA, et al.: Common variation in the BRCA1 gene and prostate cancer risk. Cancer Epidemiol Biomarkers Prev 16 (7): 1510-6, 2007.
  42. Lange EM, Robbins CM, Gillanders EM, et al.: Fine-mapping the putative chromosome 17q21-22 prostate cancer susceptibility gene to a 10 cM region based on linkage analysis. Hum Genet 121 (1): 49-55, 2007.
  43. Ewing CM, Ray AM, Lange EM, et al.: Germline mutations in HOXB13 and prostate-cancer risk. N Engl J Med 366 (2): 141-9, 2012.
  44. Xu J, Lange EM, Lu L, et al.: HOXB13 is a susceptibility gene for prostate cancer: results from the International Consortium for Prostate Cancer Genetics (ICPCG). Hum Genet 132 (1): 5-14, 2013.
  45. Chen Z, Greenwood C, Isaacs WB, et al.: The G84E mutation of HOXB13 is associated with increased risk for prostate cancer: results from the REDUCE trial. Carcinogenesis 34 (6): 1260-4, 2013.
  46. Shang Z, Zhu S, Zhang H, et al.: Germline homeobox B13 (HOXB13) G84E mutation and prostate cancer risk in European descendants: a meta-analysis of 24,213 cases and 73, 631 controls. Eur Urol 64 (1): 173-6, 2013.
  47. Handorf E, Crumpler N, Gross L, et al.: Prevalence of the HOXB13 G84E mutation among unaffected men with a family history of prostate cancer. J Genet Couns 23 (3): 371-6, 2014.
  48. Laitinen VH, Wahlfors T, Saaristo L, et al.: HOXB13 G84E mutation in Finland: population-based analysis of prostate, breast, and colorectal cancer risk. Cancer Epidemiol Biomarkers Prev 22 (3): 452-60, 2013.
  49. Witte JS, Mefford J, Plummer SJ, et al.: HOXB13 mutation and prostate cancer: studies of siblings and aggressive disease. Cancer Epidemiol Biomarkers Prev 22 (4): 675-80, 2013.
  50. Beebe-Dimmer JL, Hathcock M, Yee C, et al.: The HOXB13 G84E Mutation Is Associated with an Increased Risk for Prostate Cancer and Other Malignancies. Cancer Epidemiol Biomarkers Prev 24 (9): 1366-72, 2015.
  51. Gudmundsson J, Sulem P, Gudbjartsson DF, et al.: A study based on whole-genome sequencing yields a rare variant at 8q24 associated with prostate cancer. Nat Genet 44 (12): 1326-9, 2012.
  52. Huang H, Cai B: G84E mutation in HOXB13 is firmly associated with prostate cancer risk: a meta-analysis. Tumour Biol 35 (2): 1177-82, 2014.
  53. Cai Q, Wang X, Li X, et al.: Germline HOXB13 p.Gly84Glu mutation and cancer susceptibility: a pooled analysis of 25 epidemiological studies with 145,257 participates. Oncotarget 6 (39): 42312-21, 2015.
  54. Stott-Miller M, Karyadi DM, Smith T, et al.: HOXB13 mutations in a population-based, case-control study of prostate cancer. Prostate 73 (6): 634-41, 2013.
  55. Alanee S, Shah S, Vijai J, et al.: Prevalence of HOXB13 mutation in a population of Ashkenazi Jewish men treated for prostate cancer. Fam Cancer 12 (4): 597-600, 2013.
  56. Kote-Jarai Z, Mikropoulos C, Leongamornlert DA, et al.: Prevalence of the HOXB13 G84E germline mutation in British men and correlation with prostate cancer risk, tumour characteristics and clinical outcomes. Ann Oncol 26 (4): 756-61, 2015.
  57. Sipeky C, Gao P, Zhang Q, et al.: Synergistic Interaction of HOXB13 and CIP2A Predisposes to Aggressive Prostate Cancer. Clin Cancer Res 24 (24): 6265-6276, 2018.
  58. Lin X, Qu L, Chen Z, et al.: A novel germline mutation in HOXB13 is associated with prostate cancer risk in Chinese men. Prostate 73 (2): 169-75, 2013.
  59. Momozawa Y, Iwasaki Y, Hirata M, et al.: Germline Pathogenic Variants in 7636 Japanese Patients With Prostate Cancer and 12 366 Controls. J Natl Cancer Inst 112 (4): 369-376, 2020.
  60. Marlin R, Créoff M, Merle S, et al.: Mutation HOXB13 c.853delT in Martinican prostate cancer patients. Prostate 80 (6): 463-470, 2020.
  61. Na R, Wei J, Sample CJ, et al.: The HOXB13 variant X285K is associated with clinical significance and early age at diagnosis in African American prostate cancer patients. Br J Cancer 126 (5): 791-796, 2022.
  62. Darst BF, Hughley R, Pfennig A, et al.: A Rare Germline HOXB13 Variant Contributes to Risk of Prostate Cancer in Men of African Ancestry. Eur Urol 81 (5): 458-462, 2022.
  63. Karlsson R, Aly M, Clements M, et al.: A population-based assessment of germline HOXB13 G84E mutation and prostate cancer risk. Eur Urol 65 (1): 169-76, 2014.
  64. MacInnis RJ, Severi G, Baglietto L, et al.: Population-based estimate of prostate cancer risk for carriers of the HOXB13 missense mutation G84E. PLoS One 8 (2): e54727, 2013.
  65. Nyberg T, Govindasami K, Leslie G, et al.: Homeobox B13 G84E Mutation and Prostate Cancer Risk. Eur Urol 75 (5): 834-845, 2019.
  66. Soravia C, van der Klift H, Bründler MA, et al.: Prostate cancer is part of the hereditary non-polyposis colorectal cancer (HNPCC) tumor spectrum. Am J Med Genet 121A (2): 159-62, 2003.
  67. Haraldsdottir S, Hampel H, Wei L, et al.: Prostate cancer incidence in males with Lynch syndrome. Genet Med 16 (7): 553-7, 2014.
  68. Grindedal EM, Møller P, Eeles R, et al.: Germ-line mutations in mismatch repair genes associated with prostate cancer. Cancer Epidemiol Biomarkers Prev 18 (9): 2460-7, 2009.
  69. Langeberg WJ, Kwon EM, Koopmeiners JS, et al.: Population-based study of the association of variants in mismatch repair genes with prostate cancer risk and outcomes. Cancer Epidemiol Biomarkers Prev 19 (1): 258-64, 2010.
  70. Bauer CM, Ray AM, Halstead-Nussloch BA, et al.: Hereditary prostate cancer as a feature of Lynch syndrome. Fam Cancer 10 (1): 37-42, 2011.
  71. Dominguez-Valentin M, Joost P, Therkildsen C, et al.: Frequent mismatch-repair defects link prostate cancer to Lynch syndrome. BMC Urol 16: 15, 2016.
  72. Raymond VM, Mukherjee B, Wang F, et al.: Elevated risk of prostate cancer among men with Lynch syndrome. J Clin Oncol 31 (14): 1713-8, 2013.
  73. Ryan S, Jenkins MA, Win AK: Risk of prostate cancer in Lynch syndrome: a systematic review and meta-analysis. Cancer Epidemiol Biomarkers Prev 23 (3): 437-49, 2014.
  74. Rosty C, Walsh MD, Lindor NM, et al.: High prevalence of mismatch repair deficiency in prostate cancers diagnosed in mismatch repair gene mutation carriers from the colon cancer family registry. Fam Cancer 13 (4): 573-82, 2014.
  75. Dominguez-Valentin M, Sampson JR, Seppälä TT, et al.: Cancer risks by gene, age, and gender in 6350 carriers of pathogenic mismatch repair variants: findings from the Prospective Lynch Syndrome Database. Genet Med 22 (1): 15-25, 2020.
  76. Guedes LB, Antonarakis ES, Schweizer MT, et al.: MSH2 Loss in Primary Prostate Cancer. Clin Cancer Res 23 (22): 6863-6874, 2017.
  77. Kovacs ME, Papp J, Szentirmay Z, et al.: Deletions removing the last exon of TACSTD1 constitute a distinct class of mutations predisposing to Lynch syndrome. Hum Mutat 30 (2): 197-203, 2009.
  78. Pritchard CC, Mateo J, Walsh MF, et al.: Inherited DNA-Repair Gene Mutations in Men with Metastatic Prostate Cancer. N Engl J Med 375 (5): 443-53, 2016.
  79. Savitsky K, Bar-Shira A, Gilad S, et al.: A single ataxia telangiectasia gene with a product similar to PI-3 kinase. Science 268 (5218): 1749-53, 1995.
  80. Dombernowsky SL, Weischer M, Allin KH, et al.: Risk of cancer by ATM missense mutations in the general population. J Clin Oncol 26 (18): 3057-62, 2008.
  81. Angèle S, Falconer A, Edwards SM, et al.: ATM polymorphisms as risk factors for prostate cancer development. Br J Cancer 91 (4): 783-7, 2004.
  82. Meyer A, Wilhelm B, Dörk T, et al.: ATM missense variant P1054R predisposes to prostate cancer. Radiother Oncol 83 (3): 283-8, 2007.
  83. Schumacher FR, Al Olama AA, Berndt SI, et al.: Association analyses of more than 140,000 men identify 63 new prostate cancer susceptibility loci. Nat Genet 50 (7): 928-936, 2018.
  84. Karlsson Q, Brook MN, Dadaev T, et al.: Rare Germline Variants in ATM Predispose to Prostate Cancer: A PRACTICAL Consortium Study. Eur Urol Oncol 4 (4): 570-579, 2021.
  85. Wang Y, Dai B, Ye D: CHEK2 mutation and risk of prostate cancer: a systematic review and meta-analysis. Int J Clin Exp Med 8 (9): 15708-15, 2015.
  86. Conti DV, Wang K, Sheng X, et al.: Two Novel Susceptibility Loci for Prostate Cancer in Men of African Ancestry. J Natl Cancer Inst 109 (8): , 2017.
  87. Mai PL, Best AF, Peters JA, et al.: Risks of first and subsequent cancers among TP53 mutation carriers in the National Cancer Institute Li-Fraumeni syndrome cohort. Cancer 122 (23): 3673-3681, 2016.
  88. Ruijs MW, Verhoef S, Rookus MA, et al.: TP53 germline mutation testing in 180 families suspected of Li-Fraumeni syndrome: mutation detection rate and relative frequency of cancers in different familial phenotypes. J Med Genet 47 (6): 421-8, 2010.
  89. Bougeard G, Renaux-Petel M, Flaman JM, et al.: Revisiting Li-Fraumeni Syndrome From TP53 Mutation Carriers. J Clin Oncol 33 (21): 2345-52, 2015.
  90. Cheng HH, Klemfuss N, Montgomery B, et al.: A Pilot Study of Clinical Targeted Next Generation Sequencing for Prostate Cancer: Consequences for Treatment and Genetic Counseling. Prostate 76 (14): 1303-11, 2016.
  91. Stacey SN, Sulem P, Jonasdottir A, et al.: A germline variant in the TP53 polyadenylation signal confers cancer susceptibility. Nat Genet 43 (11): 1098-103, 2011.
  92. Mittal RD, George GP, Mishra J, et al.: Role of functional polymorphisms of P53 and P73 genes with the risk of prostate cancer in a case-control study from Northern India. Arch Med Res 42 (2): 122-7, 2011.
  93. Xu B, Xu Z, Cheng G, et al.: Association between polymorphisms of TP53 and MDM2 and prostate cancer risk in southern Chinese. Cancer Genet Cytogenet 202 (2): 76-81, 2010.
  94. Rusak B, Kluźniak W, Wokołorczykv D, et al.: Inherited NBN Mutations and Prostate Cancer Risk and Survival. Cancer Res Treat 51 (3): 1180-1187, 2019.
  95. Giri VN, Hegarty SE, Hyatt C, et al.: Germline genetic testing for inherited prostate cancer in practice: Implications for genetic testing, precision therapy, and cascade testing. Prostate 79 (4): 333-339, 2019.
  96. Nicolosi P, Ledet E, Yang S, et al.: Prevalence of Germline Variants in Prostate Cancer and Implications for Current Genetic Testing Guidelines. JAMA Oncol 5 (4): 523-528, 2019.
  97. Lang SH, Swift SL, White H, et al.: A systematic review of the prevalence of DNA damage response gene mutations in prostate cancer. Int J Oncol 55 (3): 597-616, 2019.
  98. Robinson D, Van Allen EM, Wu YM, et al.: Integrative clinical genomics of advanced prostate cancer. Cell 161 (5): 1215-28, 2015.
  99. Mandelker D, Zhang L, Kemel Y, et al.: Mutation Detection in Patients With Advanced Cancer by Universal Sequencing of Cancer-Related Genes in Tumor and Normal DNA vs Guideline-Based Germline Testing. JAMA 318 (9): 825-835, 2017.
  100. Schrader KA, Cheng DT, Joseph V, et al.: Germline Variants in Targeted Tumor Sequencing Using Matched Normal DNA. JAMA Oncol 2 (1): 104-11, 2016.
  101. Amundadottir LT, Sulem P, Gudmundsson J, et al.: A common variant associated with prostate cancer in European and African populations. Nat Genet 38 (6): 652-8, 2006.
  102. Schumacher FR, Feigelson HS, Cox DG, et al.: A common 8q24 variant in prostate and breast cancer from a large nested case-control study. Cancer Res 67 (7): 2951-6, 2007.
  103. Suuriniemi M, Agalliu I, Schaid DJ, et al.: Confirmation of a positive association between prostate cancer risk and a locus at chromosome 8q24. Cancer Epidemiol Biomarkers Prev 16 (4): 809-14, 2007.
  104. Wang L, McDonnell SK, Slusser JP, et al.: Two common chromosome 8q24 variants are associated with increased risk for prostate cancer. Cancer Res 67 (7): 2944-50, 2007.
  105. Yeager M, Orr N, Hayes RB, et al.: Genome-wide association study of prostate cancer identifies a second risk locus at 8q24. Nat Genet 39 (5): 645-9, 2007.
  106. Zheng SL, Sun J, Cheng Y, et al.: Association between two unlinked loci at 8q24 and prostate cancer risk among European Americans. J Natl Cancer Inst 99 (20): 1525-33, 2007.
  107. Savage SA, Greene MH: The evidence for prostate cancer risk loci at 8q24 grows stronger. J Natl Cancer Inst 99 (20): 1499-501, 2007.
  108. Salinas CA, Kwon E, Carlson CS, et al.: Multiple independent genetic variants in the 8q24 region are associated with prostate cancer risk. Cancer Epidemiol Biomarkers Prev 17 (5): 1203-13, 2008.
  109. Zheng SL, Hsing AW, Sun J, et al.: Association of 17 prostate cancer susceptibility loci with prostate cancer risk in Chinese men. Prostate 70 (4): 425-32, 2010.
  110. Zeegers MP, Khan HS, Schouten LJ, et al.: Genetic marker polymorphisms on chromosome 8q24 and prostate cancer in the Dutch population: DG8S737 may not be the causative variant. Eur J Hum Genet 19 (1): 118-20, 2011.
  111. Gudmundsson J, Sulem P, Manolescu A, et al.: Genome-wide association study identifies a second prostate cancer susceptibility variant at 8q24. Nat Genet 39 (5): 631-7, 2007.
  112. Haiman CA, Patterson N, Freedman ML, et al.: Multiple regions within 8q24 independently affect risk for prostate cancer. Nat Genet 39 (5): 638-44, 2007.
  113. Yeager M, Chatterjee N, Ciampa J, et al.: Identification of a new prostate cancer susceptibility locus on chromosome 8q24. Nat Genet 41 (10): 1055-7, 2009.
  114. Al Olama AA, Kote-Jarai Z, Giles GG, et al.: Multiple loci on 8q24 associated with prostate cancer susceptibility. Nat Genet 41 (10): 1058-60, 2009.
  115. Matejcic M, Saunders EJ, Dadaev T, et al.: Germline variation at 8q24 and prostate cancer risk in men of European ancestry. Nat Commun 9 (1): 4616, 2018.
  116. Conti DV, Darst BF, Moss LC, et al.: Trans-ancestry genome-wide association meta-analysis of prostate cancer identifies new susceptibility loci and informs genetic risk prediction. Nat Genet 53 (1): 65-75, 2021.
  117. Cook MB, Wang Z, Yeboah ED, et al.: A genome-wide association study of prostate cancer in West African men. Hum Genet 133 (5): 509-21, 2014.
  118. Haiman CA, Chen GK, Blot WJ, et al.: Characterizing genetic risk at known prostate cancer susceptibility loci in African Americans. PLoS Genet 7 (5): e1001387, 2011.
  119. Han Y, Signorello LB, Strom SS, et al.: Generalizability of established prostate cancer risk variants in men of African ancestry. Int J Cancer 136 (5): 1210-7, 2015.
  120. Han Y, Rand KA, Hazelett DJ, et al.: Prostate Cancer Susceptibility in Men of African Ancestry at 8q24. J Natl Cancer Inst 108 (7): , 2016.
  121. Takata R, Akamatsu S, Kubo M, et al.: Genome-wide association study identifies five new susceptibility loci for prostate cancer in the Japanese population. Nat Genet 42 (9): 751-4, 2010.
  122. Akamatsu S, Takata R, Haiman CA, et al.: Common variants at 11q12, 10q26 and 3p11.2 are associated with prostate cancer susceptibility in Japanese. Nat Genet 44 (4): 426-9, S1, 2012.
  123. Xu J, Mo Z, Ye D, et al.: Genome-wide association study in Chinese men identifies two new prostate cancer risk loci at 9q31.2 and 19q13.4. Nat Genet 44 (11): 1231-5, 2012.
  124. Takata R, Takahashi A, Fujita M, et al.: 12 new susceptibility loci for prostate cancer identified by genome-wide association study in Japanese population. Nat Commun 10 (1): 4422, 2019.
  125. Benafif S, Kote-Jarai Z, Eeles RA, et al.: A Review of Prostate Cancer Genome-Wide Association Studies (GWAS). Cancer Epidemiol Biomarkers Prev 27 (8): 845-857, 2018.
  126. Lecarpentier J, Silvestri V, Kuchenbaecker KB, et al.: Prediction of Breast and Prostate Cancer Risks in Male BRCA1 and BRCA2 Mutation Carriers Using Polygenic Risk Scores. J Clin Oncol 35 (20): 2240-2250, 2017.
  127. Barnes DR, Silvestri V, Leslie G, et al.: Breast and Prostate Cancer Risks for Male BRCA1 and BRCA2 Pathogenic Variant Carriers Using Polygenic Risk Scores. J Natl Cancer Inst 114 (1): 109-122, 2022.
  128. Ström P, Nordström T, Aly M, et al.: The Stockholm-3 Model for Prostate Cancer Detection: Algorithm Update, Biomarker Contribution, and Reflex Test Potential. Eur Urol 74 (2): 204-210, 2018.
  129. Shi Z, Platz EA, Wei J, et al.: Performance of Three Inherited Risk Measures for Predicting Prostate Cancer Incidence and Mortality: A Population-based Prospective Analysis. Eur Urol 79 (3): 419-426, 2021.
  130. Li-Sheng Chen S, Ching-Yuan Fann J, Sipeky C, et al.: Risk Prediction of Prostate Cancer with Single Nucleotide Polymorphisms and Prostate Specific Antigen. J Urol 201 (3): 486-495, 2019.
  131. Plym A, Penney KL, Kalia S, et al.: Evaluation of a Multiethnic Polygenic Risk Score Model for Prostate Cancer. J Natl Cancer Inst 114 (5): 771-774, 2022.
  132. Freedman ML, Monteiro AN, Gayther SA, et al.: Principles for the post-GWAS functional characterization of cancer risk loci. Nat Genet 43 (6): 513-8, 2011.
  133. Pomerantz MM, Beckwith CA, Regan MM, et al.: Evaluation of the 8q24 prostate cancer risk locus and MYC expression. Cancer Res 69 (13): 5568-74, 2009.
  134. Jia L, Landan G, Pomerantz M, et al.: Functional enhancers at the gene-poor 8q24 cancer-linked locus. PLoS Genet 5 (8): e1000597, 2009.
  135. Ahmadiyeh N, Pomerantz MM, Grisanzio C, et al.: 8q24 prostate, breast, and colon cancer risk loci show tissue-specific long-range interaction with MYC. Proc Natl Acad Sci U S A 107 (21): 9742-6, 2010.
  136. Sotelo J, Esposito D, Duhagon MA, et al.: Long-range enhancers on 8q24 regulate c-Myc. Proc Natl Acad Sci U S A 107 (7): 3001-5, 2010.
  137. Meyer KB, Maia AT, O'Reilly M, et al.: A functional variant at a prostate cancer predisposition locus at 8q24 is associated with PVT1 expression. PLoS Genet 7 (7): e1002165, 2011.
  138. Spisák S, Lawrenson K, Fu Y, et al.: CAUSEL: an epigenome- and genome-editing pipeline for establishing function of noncoding GWAS variants. Nat Med 21 (11): 1357-63, 2015.
  139. Hazelett DJ, Rhie SK, Gaddis M, et al.: Comprehensive functional annotation of 77 prostate cancer risk loci. PLoS Genet 10 (1): e1004102, 2014.
  140. Jiang J, Cui W, Vongsangnak W, et al.: Post genome-wide association studies functional characterization of prostate cancer risk loci. BMC Genomics 14 (Suppl 8): S9, 2013.

Prostate Cancer Genetics: Screening, Surveillance, and Treatment

This section addresses the impact of genetics on prostate cancer screening, surveillance, and treatment. For more information about prostate cancer screening, surveillance, and treatment, see Prostate Cancer Screening and Prostate Cancer Treatment.

Prostate Cancer Screening

Background

Decisions about risk-reducing interventions for patients with an inherited predisposition to prostate cancer, as with any disease, are best guided by randomized controlled clinical trials and knowledge of the underlying natural history of the process. However, existing studies of screening for prostate cancer in high-risk men (men with a positive family history of prostate cancer and African American men) are predominantly based on retrospective case series or retrospective cohort analyses. Because awareness of a positive family history can lead to more frequent work-ups for cancer and result in apparently earlier prostate cancer detection, assessments of disease progression rates and survival after diagnosis are subject to selection, lead time, and length biases. This section focuses on screening and risk reduction of prostate cancer among men predisposed to the disease; data relevant to screening in high-risk men are primarily extracted from studies performed in the general population.

Screening

Information is limited about the efficacy of commonly available screening tests such as the digital rectal exam (DRE) and serum prostate-specific antigen (PSA) in men genetically predisposed to developing prostate cancer. Furthermore, comparing the results of studies that have examined the efficacy of screening for prostate cancer is difficult because studies vary with regard to the cutoff values chosen for an elevated PSA test. For a given sensitivity and specificity of a screening test, the positive predictive value (PPV) increases as the underlying prevalence of disease rises. Therefore, it is theoretically possible that the PPV and diagnostic yield will be higher for the DRE and for PSA in men with a genetic predisposition than in average-risk populations.

Most retrospective analyses of prostate cancer screening cohorts have reported PPV for PSA, with or without DRE, among high-risk men in the range of 23% to 75%. Screening strategies (frequency of PSA measurements or inclusion of DRE) and PSA cutoff for biopsy varied among these studies, which may have influenced this range of PPV. Cancer detection rates among high-risk men have been reported to be in the range of 4.75% to 22%. Most cancers detected were of intermediate Gleason score (5–7), with Gleason scores of 8 or higher being detected in some high-risk men. Overall, there is limited information about the net benefits and harms of screening men at higher risk of prostate cancer. In addition, there is little evidence to support specific screening approaches in prostate cancer families at high risk. Risks and benefits of routine screening in the general population are discussed in Prostate Cancer Screening. On the basis of the available data, most professional societies and organizations recommend that high-risk men engage in shared decision-making with their health care providers and develop individualized plans for prostate cancer screening based on their risk factors. A summary of prostate cancer screening recommendations for high-risk men by professional organizations is shown in Table 10 and Table 11.

Table 10. Available Recommendations for Prostate Cancer Screening in BRCA1, BRCA2, and HOXB13 Carriers

Philadelphia Prostate Cancer Consensus Conference (Giri et al. 2020) Genetic/Familial High-Risk Assessment: Breast, Ovarian, and Pancreatic (Version 2.2024) NCCN Prostate Cancer Early Detection (Version 2.2023)a
NCCN = National Comprehensive Cancer Network; PSA = prostate-specific antigen.
aFor germlinepathogenic variants other than BRCA2 (including ATM and Lynch syndrome genes), it is reasonable to consider beginning shared decision-making about PSA screening at age 40 years and to consider screening at annual intervals, rather than every other year.
Screening in BRCA1CarriersConsider baseline PSA for age >40 y or 10 years before the earliest prostate cancer diagnosis in the family Consider prostate cancer screening starting at age 40 y Consider beginning shared decision-making about PSA screening at age 40 y
NCCN Genetic/Familial High-Risk Assessment guidelines suggest that individuals see the NCCN Prostate Cancer Early Detection guidelines for guidance on prostate cancer screening intervals Consider annual screening rather than screening every other year
Screening in BRCA2 CarriersRecommend baseline PSA for age >40 y or 10 years before the earliest prostate cancer diagnosis in the family Recommend prostate cancer screening starting at age 40 y Recommend PSA screening starting at age 40 y
Screening interval determined by baseline PSA level NCCN Genetic/Familial High-Risk Assessment guidelines suggest that individuals see the NCCN Prostate Cancer Early Detection guidelines for guidance on prostate cancer screening intervals Consider annual screening rather than screening every other year
Screening in HOXB13 Carriers Baseline PSA for age >40 y or 10 years before the earliest prostate cancer diagnosis in the family None provided Consider beginning shared decision-making about PSA screening at age 40 y
Screening interval determined by baseline PSA level Consider annual screening rather than screening every other year

Table 11. Summary of Prostate Cancer Screening Recommendations for Men Based on Family History, Race, and Ethnicity

Screening Recommendation SourcePopulationTestAge Screening InitiatedFrequencyComments
DRE = digital rectal exam; FDR = first-degree relative; NCCN = National Comprehensive Cancer Network; PSA = prostate-specific antigen; SDR = second-degree relative.
aDRE is recommended in addition to PSA test for men with hypogonadism.
bA suspicious family history includes, but is not limited to, an FDR or SDR with metastatic prostate cancer, ovarian cancer, male breast cancer, female breast cancer at age ≤45 y, colorectal or endometrial cancer at age ≤50 y, or pancreatic cancer; this may also include two or more FDRs or SDRs with breast, prostate (excluding clinically localized Grade Group 1 disease), colorectal, or endometrial cancer at any age.
United States Preventive Services Task Force (2018) Men aged 55–69 y PSAN/AN/AIn determining whether PSA-based screening is appropriate in individual cases, patients and clinicians should consider the benefits and harms of PSA screening based on family history, race and ethnicity, comorbid medical conditions, patient values about the benefits and harms of screening and treatment-specific outcomes, and other health needs
American Urological Association (2023) African American men, men with germline pathogenic variants in hereditary prostate cancer genes, and men with strong family histories of prostate cancerPSA40 to 45 yScreening is individualized based on the patient's personal preferences and an informed discussion regarding the uncertainty of benefit and associated harms
American Cancer Society (2023) African American menPSA with or without DREa≥45 y Screen every 2 y if PSA is <2.5 ng/mL; screen annually if PSA level is ≥2.5 ng/mL; if PSA levels are between 2.5–4.0 ng/mL, an individualized risk assessment can be performed, which incorporates other prostate cancer risk factors (particularly for high-grade cancer, which may be used for a referral recommendation)Counseling consists of a review of the benefits and limitations of testing so that a clinician-assisted, informed decision about testing can be made. It is recommended that prostate cancer screening be accompanied by an informed decision-making process
Men with an FDR who was diagnosed with prostate cancer at <65 y PSA with or without DREa≥45 y
Men with multiple FDRs who were diagnosed with prostate cancer at <65 yPSA with or without DREa≥40 y
NCCN Prostate Cancer Early Detection (Version 2.2023) African American men Baseline PSA40 y Consider screening at annual intervals rather than every other yearThe panel states that it is reasonable for African American men to consider beginning shared decision-making about PSA screening with their providers at age 40 y
Men with a suspicious family historybBaseline PSA40 yScreen every 2–4 y if PSA level <1 ng/mL, DRE normal; if the family history is concerning, NCCN recommends shared decision-making to determine the frequency of PSA screening Referral to a cancer genetics professional is recommended for those with a known or suspected pathogenic variant in a cancer susceptibility gene
Screen every 1–2 y if PSA level ≤3 ng/mL, DRE normal (if done)

Level of evidence: 5

Screening in carriers of BRCA pathogenic variants

IMPACT (Identification of Men with a genetic predisposition to ProstAte Cancer) is an international study focused on prostate cancer screening in carriers of BRCA1/BRCA2 pathogenic variants versus noncarriers. The study recruited 2,481 men (791 BRCA1 carriers, 531 BRCA1 noncarriers; 731 BRCA2 carriers, 428 BRCA2 noncarriers). A total of 199 men (8%) presented with PSA levels higher than 3.0 ng/mL, which was the study PSA cutoff for recommending a biopsy. The overall cancer detection rate was 36.4% (59 prostate cancers diagnosed among 162 biopsies). Prostate cancer by BRCA pathogenic variant status was as follows: BRCA1 carriers (n = 18), BRCA1 noncarriers (n = 10); BRCA2 carriers (n = 24), BRCA2 noncarriers (n = 7). Using published stage and grade criteria for risk classification, intermediate- or high-risk tumors were diagnosed in 11 of 18 BRCA1 carriers (61%), 8 of 10 BRCA1 noncarriers (80%), 17 of 24 BRCA2 carriers (71%), and 3 of 7 BRCA2 noncarriers (43%). The PPV of PSA with a biopsy threshold of 3.0 ng/mL was 48% in carriers of BRCA2 pathogenic variants, 33.3% in BRCA2 noncarriers, 37.5% in BRCA1 carriers, and 23.3% in BRCA1 noncarriers. Ninety-five percent of the men were White; therefore, the results cannot be generalized to all ethnic groups.

Interim results from the IMPACT study (now comprising 2,932 participants including 919 BRCA1 carriers and 902 BRCA2 carriers) demonstrated a cancer incidence rate (per 1,000 person-years) that was higher in BRCA2 carriers compared with noncarriers (19 vs. 12; P = .03). There was no statistical difference in the cancer incidence rates between BRCA1 carriers and noncarriers. Cancer in BRCA2 carriers, but not in BRCA1 carriers, was diagnosed at an earlier age and was more likely to be clinically significant.

Level of evidence (screening in carriers of BRCA pathogenic variants): 3

Impact of Germline Genetics on Management and Treatment of Metastatic Prostate Cancer

Targeted therapies on the basis of genetic results are increasingly driving options and strategies for treatment in oncology. These therapeutic approaches include candidacy for targeted therapy (such as poly [ADP-ribose] polymerase [PARP] inhibitors or immune checkpoint inhibitors), use of platinum-based chemotherapy, and sequencing of androgen-signaling therapy versus chemotherapy. Multiple genetically informed clinical trials are under way for men with prostate cancer. Table 12 summarizes some of the published precision oncology and precision management studies.

Table 12. Summary of Precision Oncology or Precision Management Studies Involving Germline Pathogenic Variant Status

StudyCohortGermline ResultsInterventionOutcomes and Comments
ADT = androgen deprivation therapy; AR = androgen receptor; CI = confidence interval; CSS = cause-specific survival; DDR = DNA damage repair; FDA = U.S. Food and Drug Administration; HR = hazard ratio; HRR = homologous recombination repair; mCRPC = metastatic castration-resistant prostate cancer; mPC = metastatic prostate cancer; ORR = objective response rate; OS = overall survival; PARP = poly (ADP-ribose) polymerase; PC = prostate cancer; PFS = progression-free survival; PSA = prostate-specific antigen; RR = relative risk.
aThis study reported both germline and somatic genetic test results.
Retrospective
Annala et al. (2017) 319 men with mCRPC; performed germline sequencing of 22 DNA repair genes; all participants previously received ADT and their PCs progressed24/319 (7.5%) had DDR germline pathogenic variants: Patients with mCRPC and a germline pathogenic variant received the following as a first-line AR-targeted therapy: docetaxel/cabazitaxel (41%), enzalutamide (23%), or abiraterone (36%)Patients with DNA repair defects had decreased responses to ADT:
BRCA2: 16/319 (5.0%)
ATM: 1/319 (0.3%)— Time from ADT initiation to mCRPC: Germline positive, 11.8 mo (n = 22) vs. germline negative, 19.0 mo (n = 113) (P = .031)
BRCA1: 1/319 (0.3%) Patients with mCRPC but without a germline pathogenic variant received the following as a first-line AR-targeted therapy: docetaxel/cabazitaxel (33%), enzalutamide (18%), abiraterone (39%), or other (10%)
PALB2: 2/319 (0.6%) — PFS on first-line AR-targeted therapy: Germline positive, 3.3 mo vs. germline negative, 6.2 mo (P = .01)
Pomerantz et al. (2017) 141 men with mCRPC treated with docetaxel8/141 (5.7%) had BRCA2 germline pathogenic variantsPatients received at least two doses of carboplatin and docetaxel6/8 men with BRCA2 germline pathogenic variants (75%) had PSA levels that declined by 50% vs. 23/133 in men without BRCA2 germline pathogenic variants (17%) (P< .001)
A small case series (n = 3) showed a response to platinum chemotherapy with biallelic inactivation of BRCA2, defined as either biallelic somatic BRCA2 pathogenic variants or a germline pathogenic variant plus a somatic BRCA2 pathogenic variant
Mateo et al. (2018) 390 men with mPC; retrospective review60/390 (15.4%) had DDR germline pathogenic variants:Patients received abiraterone, enzalutamide, and docetaxel; an exploratory subgroup analysis was done for PARP inhibitors/platinum chemotherapy Similar findings were observed for DDR pathogenic variant carriers and noncarriers for several outcome measures:
— Median OS from castration resistance (3.2 y in carriers vs 3.0 y in noncarriers; P = .73)
— Median docetaxel PFS (6.8 mo in carriers vs. 5.1 mo in noncarriers)
BRCA2: 37/390 (9.5%)— RRs for PC (61% in carriers vs. 54% in noncarriers)
— Median PFS on first-line abiraterone/enzalutamide (8.3 mo in both carriers and noncarriers)
— RR of PC on first-line abiraterone/enzalutamide (46% in carriers vs. 56% in noncarriers)
Carter et al. (2019) 1,211 men with PC on active surveillance2.1% of patients had germline pathogenic variants in BRCA1/BRCA2/ATMPatients were put on active surveillance289 patients had their PC tumor grades reclassified: 11/26 patients had pathogenic variants in BRCA1/BRCA2/ATM and 278/1,185 patients did not have a pathogenic variant in BRCA1/BRCA2/ATM (noncarriers); adjusted HR, 1.96 (95% CI, 1.004–3.84; P = .04)
Tumor reclassification occurred in 6/11 BRCA2 carriers and 283/1,200 noncarriers; adjusted HR, 2.74 (95% CI, 1.26–5.96; P = .01)
Of the men who had their PCs reclassified, 3.8% had a BRCA1, BRCA2, or ATM pathogenic variant, and 2.1% only had a BRCA2 pathogenic variant. Of the men whose PCs were not reclassified, 1.6% had a BRCA1, BRCA2, or ATM pathogenic variant, and 0.5% only had a BRCA2 pathogenic variant. The P value for BRCA1/BRCA2/ATM carriers with PCs reclassified versus those without PCs reclassified was .04. The P value for BRCA2 carriers with PCs reclassified versus those without PCs reclassified was .03
Marshall et al. (2019) 46 men with mCRPC were offered olaparib; 23 men had germline pathogenic variants (13 men were not tested)23 men had germline pathogenic variants in BRCA1/BRCA2/ATM; 2 men had BRCA1 pathogenic variants, 15 men had BRCA2 pathogenic variants, and 6 men had ATM pathogenic variants Patients received olaparibWhen patients were given olaparib, PSA levels were reduced by 50% in 13/17 (76%) men with BRCA1/BRCA2 pathogenic variants and in 0/6 (0%) men with ATM pathogenic variants (Fisher's exact test; P = .002)
Patients with BRCA1/BRCA2 pathogenic variants had a median PFS of 12.3 mo, while patients with ATM pathogenic variants had a median PFS of 2.4 mo (HR, 0.17; 95% CI, 0.05–0.57; P = .004)
Sokolova et al. (2021) 90 men with PC; 76/90 had metastatic disease when their PC was diagnosed; participants were matched for PC stage and year of germline testing; participants had similar ages, Gleason grades, and PSA levels at diagnosis45 men with ATM germline pathogenic variants; 45 men with BRCA2 germline pathogenic variantsPatients received various systemic therapiesNo changes were observed when different groups were given abiraterone, enzalutamide, or docetaxel
When patients were given PARP inhibitors, PSA levels were reduced by 50% in 0/7 men with ATM germline pathogenic variants and in 12/14 men with BRCA2 germline pathogenic variants (P< .001); this response was significant
Study limitations included the following: retrospective study, no zygosity data
Prospective
Antonarakis et al. (2018) 172 men with mCRPC began treatment with abiraterone or enzalutamide22/172 (12.8%) had DDR germline pathogenic variants:Patients received first-line hormonal therapy (abiraterone or enzalutamide)In propensity score–weighted multivariable analyses, outcomes were superior in men with germline BRCA1/BRCA2/ATM variants with respect to PSA-PFS (HR, 0.48; 95% CI, 0.25–0.92; P = .027), PFS (HR, 0.52; 95% CI, 0.28–0.98; P = .044), and OS (HR, 0.34; 95% CI, 0.12–0.99; P = .048). These results were not observed for men with non-BRCA1/BRCA2/ATM germline variants (P > .10)
BRCA1/BRCA2/ATM: 9/172 (5.2%)Study limitations included the following: only 9 patients with BRCA1/BRCA2/ATM pathogenic variants
Castro et al. (2019) 419 men with mCRPC were enrolled when they were diagnosed with mPC68/419 (16.2%) had DDR germline pathogenic variants: Patients received an androgen-signaling inhibitor (abiraterone or enzalutamide) as a first-line therapy and a taxane (docetaxel was given in 96.3% of patients) as a second-line therapy or patients received a taxane as a first-line therapy and an androgen-signaling inhibitor (abiraterone or enzalutamide) as a second-line therapyCSS between ATM/BRCA1/BRCA2/PALB2 carriers and noncarriers was not statistically significant (23.3 mo vs. 33.2 mo; P = .264)
BRCA2: 14/419 (3.3%)
ATM: 8/419 (1.9%) CSS was halved in BRCA2 carriers (17.4 mo vs. 33.2 mo; P = .027), and BRCA2 pathogenic variants were identified as an independent prognostic factor for CSS (HR, 2.11; P = .033)
BRCA1: 4/419 (1%) Significant interactions between BRCA2 status and treatment type (androgen-signaling inhibitor vs. taxane therapy) were observed (CSS-adjusted P = .014; PFS-adjusted P = .005)
PALB2: None CSS (24.0 mo vs. 17.0 mo) and PFS (18.9 mo vs. 8.6 mo) were greater in BRCA2 carriers treated with first-line abiraterone or enzalutamide when compared with first-line taxanes
de Bono et al. (2020) 387 men in the PROfound study who had mCRPC with disease progression while receiving a new hormonal agent (e.g., enzalutamide or abiraterone)Currently, the FDA has approved olaparib for use in patients with mCRPC who have a somatic or germline pathogenic variant in an HRR gene. The PROfound study cited data from Mateo et al. 2015, which discovered that about half of the HRR gene variants in patient tumors were germline in nature. Results in this study reported on olaparib response in individuals with somatic variants. Data on germline pathogenic variants will be reported in the futureRandomized, open-label, phase III trial in which patients received olaparib (300 mg twice per day) or the physician’s choice of enzalutamide (160 mg once per day) or abiraterone (1,000 mg once per day) plus prednisone (5 mg twice per day)In cohort A, imaging-based PFS was significantly longer in the olaparib group than in the control group (median, 7.4 mo vs. 3.6 mo; HR for progression or death, 0.34; 95% CI, 0.25–0.47; P< .001). The median OS in cohort A was 18.5 mo in the olaparib group and 15.1 mo in the control group; 81% of the patients in the control group who had disease progression crossed over to receive olaparib
Cohort A: 245 men with >1 somatic variant in BRCA1, BRCA2, or ATM
Cohort B: 142 men with >1 somatic variant in any of the following genes: BRIP1, BARD1, CDK12, CHEK1, CHEK2, FANCL, PALB2, PPP2R2A, RAD51B, RAD51C, RAD51D, or RAD54L
Hussain et al. (2020) 387 men with mCRPC in the PROfound study; PC progressed when taking enzalutamide, abiraterone, or bothCurrently, the FDA has approved olaparib for use in patients with mCRPC who have a somatic or germline pathogenic variant in an HRR gene. The PROfound study cited data from Mateo et al. 2015, which discovered that about half of the HRR gene variants in patient tumors were germline in nature. Results in this study reported on olaparib response in individuals with somatic variants. Data on germline pathogenic variants will be reported in the futurePatients received treatment that was randomly assigned in a 2:1 ratio for olaparib versus control therapy; control therapy consisted of the provider's choice of enzalutamide or abiraterone, plus prednisone. Crossover to olaparib was permitted when PC progressed on imagingThe median OS in cohort A was 19.1 mo with olaparib and 14.7 mo with control therapy. The HR for death (adjusted for crossover from control therapy) was 0.42 (95% CI, 0.19–0.91)
Cohort A: 245 men with >1 somatic variant in BRCA1, BRCA2, or ATMThe median OS in cohort B was 14.1 mo for olaparib and 11.5 mo for control therapy. The HR for death (adjusted for crossover from control therapy) was 0.83 (95% CI, 0.11–5.98)
Cohort B: 142 men with >1 somatic variant in any of the following genes: BRIP1, BARD1, CDK12, CHEK1, CHEK2, FANCL, PALB2, PPP2R2A, RAD51B, RAD51C, RAD51D, or RAD54L
Abida et al. (2020)a115 men with mCRPC from the TRITON2 study with a deleterious somatic or germline pathogenic variant in BRCA1/BRCA2; patients had mCRPCs that progressed after treatment with one to two lines of next-generation AR-directed therapy and one taxane-based chemotherapy44/115 (38%) had BRCA1/BRCA2 germline pathogenic variants:Patients received one or more doses of rucaparib (600 mg)The ORR was 43.5% in men with measurable disease and 50.8% in men without measurable disease. ORRs were similar for men with germline and somatic variants and for men with BRCA1/BRCA2 pathogenic variants
BRCA1: 5/115 (4%)
BRCA2: 39/115 (34%)
71/115 (62%) had BRCA1/BRCA2 somatic variants:63/115 men had a confirmed PSA response (54.8%), which differed by gene; however, the BRCA1 group was small:
BRCA1: 8/115 (7%) BRCA1: 2/13 (15.4%)
BRCA2: 63/115 (55%)BRCA2: 61/102 (59.8%)
De Bono et al. (2021)a104 men with progressive mCRPC and pathogenic variants in DDR-HRR genes; patients received at least one dose of talazoparib 25/71 (25%) patients had germline pathogenic variants: 13 in BRCA2, 4 in ATM, and 8 in other genesPatients received one or more doses of talazoparib per day (received 1 mg per day or 0.75 mg per day if the patient had moderate renal impairment)The ORR was observed in 7/28 (25%) men with germline pathogenic variants
Patients also had somatic variants in the following genes: 61 in BRCA1/2, 57 in BRCA2, 4 in PALB2, 17 in ATM, 22 in other genes (ATR, CHEK2, FANCA, MLH1, MRE11A, NBN, and RAD51C)After a median follow-up period of 16.4 mo (range, 11.1–22.1), the ORR for patients with somatic variants was 29.8% (31 of 104 patients; 95% CI, 21.2%–39.6%). Clinical benefit (defined as patients with complete response, partial response, or stable disease for ≥6 months from treatment start) varied between individuals with different pathogenic variants: BRCA1/2 (56%), BRCA2 (56%), PALB2 (25%), ATM (24%), other (0%)

References

  1. Sartor O: Early detection of prostate cancer in African-American men with an increased familial risk of disease. J La State Med Soc 148 (4): 179-85, 1996.
  2. Matikainen MP, Schleutker J, Mörsky P, et al.: Detection of subclinical cancers by prostate-specific antigen screening in asymptomatic men from high-risk prostate cancer families. Clin Cancer Res 5 (6): 1275-9, 1999.
  3. Catalona WJ, Antenor JA, Roehl KA, et al.: Screening for prostate cancer in high risk populations. J Urol 168 (5): 1980-3; discussion 1983-4, 2002.
  4. Valeri A, Cormier L, Moineau MP, et al.: Targeted screening for prostate cancer in high risk families: early onset is a significant risk factor for disease in first degree relatives. J Urol 168 (2): 483-7, 2002.
  5. Narod SA, Dupont A, Cusan L, et al.: The impact of family history on early detection of prostate cancer. Nat Med 1 (2): 99-101, 1995.
  6. Giri VN, Beebe-Dimmer J, Buyyounouski M, et al.: Prostate cancer risk assessment program: a 10-year update of cancer detection. J Urol 178 (5): 1920-4; discussion 1924, 2007.
  7. Giri VN, Knudsen KE, Kelly WK, et al.: Implementation of Germline Testing for Prostate Cancer: Philadelphia Prostate Cancer Consensus Conference 2019. J Clin Oncol 38 (24): 2798-2811, 2020.
  8. National Comprehensive Cancer Network: NCCN Clinical Practice Guidelines in Oncology: Genetic/Familial High-Risk Assessment: Breast, Ovarian, and Pancreatic. Version 2.2024. Plymouth Meeting, Pa: National Comprehensive Cancer Network, 2023. Available online with free registration. Last accessed October 31, 2023.
  9. National Comprehensive Cancer Network: NCCN Clinical Practice Guidelines in Oncology: Prostate Cancer Early Detection. Version 2.2023. Plymouth Meeting, Pa: National Comprehensive Cancer Network, 2023. Available online with free registration. Last accessed November 30, 2023.
  10. U.S. Preventative Services Task Force: Screening for prostate cancer: U.S. Preventive Services Task Force recommendation statement. Rockville, Md: U.S. Preventative Services Task Force, 2018. Available online. Last accessed November 8, 2023.
  11. Wei JT, Barocas D, Carlsson S, et al.: Early Detection of Prostate Cancer: AUA/SUO Guideline Part I: Prostate Cancer Screening. J Urol 210 (1): 46-53, 2023.
  12. American Cancer Society: American Cancer Society Recommendations for Prostate Cancer Early Detection. American Cancer Society, 2023. Available online. Last accessed January 12, 2024.
  13. Bancroft EK, Page EC, Castro E, et al.: Targeted prostate cancer screening in BRCA1 and BRCA2 mutation carriers: results from the initial screening round of the IMPACT study. Eur Urol 66 (3): 489-99, 2014.
  14. National Collaborating Centre for Cancer (UK): Prostate Cancer: Diagnosis and Treatment. Cardiff, UK: National Collaborating Centre for Cancer, 2008. Available online. Last accessed November 8, 2023.
  15. Page EC, Bancroft EK, Brook MN, et al.: Interim Results from the IMPACT Study: Evidence for Prostate-specific Antigen Screening in BRCA2 Mutation Carriers. Eur Urol 76 (6): 831-842, 2019.
  16. Carlo MI, Giri VN, Paller CJ, et al.: Evolving Intersection Between Inherited Cancer Genetics and Therapeutic Clinical Trials in Prostate Cancer: A White Paper From the Germline Genetics Working Group of the Prostate Cancer Clinical Trials Consortium. JCO Precis Oncol 2018: , 2018.
  17. Annala M, Struss WJ, Warner EW, et al.: Treatment Outcomes and Tumor Loss of Heterozygosity in Germline DNA Repair-deficient Prostate Cancer. Eur Urol 72 (1): 34-42, 2017.
  18. Pomerantz MM, Spisák S, Jia L, et al.: The association between germline BRCA2 variants and sensitivity to platinum-based chemotherapy among men with metastatic prostate cancer. Cancer 123 (18): 3532-3539, 2017.
  19. Cheng HH, Pritchard CC, Boyd T, et al.: Biallelic Inactivation of BRCA2 in Platinum-sensitive Metastatic Castration-resistant Prostate Cancer. Eur Urol 69 (6): 992-5, 2016.
  20. Mateo J, Cheng HH, Beltran H, et al.: Clinical Outcome of Prostate Cancer Patients with Germline DNA Repair Mutations: Retrospective Analysis from an International Study. Eur Urol 73 (5): 687-693, 2018.
  21. Carter HB, Helfand B, Mamawala M, et al.: Germline Mutations in ATM and BRCA1/2 Are Associated with Grade Reclassification in Men on Active Surveillance for Prostate Cancer. Eur Urol 75 (5): 743-749, 2019.
  22. Marshall CH, Sokolova AO, McNatty AL, et al.: Differential Response to Olaparib Treatment Among Men with Metastatic Castration-resistant Prostate Cancer Harboring BRCA1 or BRCA2 Versus ATM Mutations. Eur Urol 76 (4): 452-458, 2019.
  23. Sokolova AO, Marshall CH, Lozano R, et al.: Efficacy of systemic therapies in men with metastatic castration resistant prostate cancer harboring germline ATM versus BRCA2 mutations. Prostate 81 (16): 1382-1389, 2021.
  24. Antonarakis ES, Lu C, Luber B, et al.: Germline DNA-repair Gene Mutations and Outcomes in Men with Metastatic Castration-resistant Prostate Cancer Receiving First-line Abiraterone and Enzalutamide. Eur Urol 74 (2): 218-225, 2018.
  25. Castro E, Romero-Laorden N, Del Pozo A, et al.: PROREPAIR-B: A Prospective Cohort Study of the Impact of Germline DNA Repair Mutations on the Outcomes of Patients With Metastatic Castration-Resistant Prostate Cancer. J Clin Oncol 37 (6): 490-503, 2019.
  26. de Bono J, Mateo J, Fizazi K, et al.: Olaparib for Metastatic Castration-Resistant Prostate Cancer. N Engl J Med 382 (22): 2091-2102, 2020.
  27. Hussain M, Mateo J, Fizazi K, et al.: Survival with Olaparib in Metastatic Castration-Resistant Prostate Cancer. N Engl J Med 383 (24): 2345-2357, 2020.
  28. Abida W, Patnaik A, Campbell D, et al.: Rucaparib in Men With Metastatic Castration-Resistant Prostate Cancer Harboring a BRCA1 or BRCA2 Gene Alteration. J Clin Oncol 38 (32): 3763-3772, 2020.
  29. de Bono JS, Mehra N, Scagliotti GV, et al.: Talazoparib monotherapy in metastatic castration-resistant prostate cancer with DNA repair alterations (TALAPRO-1): an open-label, phase 2 trial. Lancet Oncol 22 (9): 1250-1264, 2021.

Latest Updates to This Summary (02/15/2024)

The PDQ cancer information summaries are reviewed regularly and updated as new information becomes available. This section describes the latest changes made to this summary as of the date above.

This summary was extensively revised.

This summary is written and maintained by the PDQ Cancer Genetics Editorial Board, which is editorially independent of NCI. The summary reflects an independent review of the literature and does not represent a policy statement of NCI or NIH. More information about summary policies and the role of the PDQ Editorial Boards in maintaining the PDQ summaries can be found on the About This PDQ Summary and PDQ® Cancer Information for Health Professionals pages.

About This PDQ Summary

Purpose of This Summary

This PDQ cancer information summary for health professionals provides comprehensive, peer-reviewed, evidence-based information about the genetics of prostate cancer. It is intended as a resource to inform and assist clinicians in the care of their patients. It does not provide formal guidelines or recommendations for making health care decisions.

Reviewers and Updates

This summary is reviewed regularly and updated as necessary by the PDQ Cancer Genetics Editorial Board, which is editorially independent of the National Cancer Institute (NCI). The summary reflects an independent review of the literature and does not represent a policy statement of NCI or the National Institutes of Health (NIH).

Board members review recently published articles each month to determine whether an article should:

  • be discussed at a meeting,
  • be cited with text, or
  • replace or update an existing article that is already cited.

Changes to the summaries are made through a consensus process in which Board members evaluate the strength of the evidence in the published articles and determine how the article should be included in the summary.

The lead reviewers for Genetics of Prostate Cancer are:

  • Kathleen A. Calzone, PhD, RN, AGN-BC, FAAN (National Cancer Institute)
  • Veda N. Giri, MD (Yale University)
  • Suzanne C. O'Neill, PhD (Georgetown University)
  • Susan K. Peterson, PhD, MPH (University of Texas, M.D. Anderson Cancer Center)
  • Mark Pomerantz, MD (Dana-Farber Cancer Institute)
  • John M. Quillin, PhD, MPH, MS (Virginia Commonwealth University)
  • Charite Ricker, MS, CGC (University of Southern California)
  • Catharine Wang, PhD, MSc (Boston University School of Public Health)

Any comments or questions about the summary content should be submitted to Cancer.gov through the NCI website's Email Us. Do not contact the individual Board Members with questions or comments about the summaries. Board members will not respond to individual inquiries.

Levels of Evidence

Some of the reference citations in this summary are accompanied by a level-of-evidence designation. These designations are intended to help readers assess the strength of the evidence supporting the use of specific interventions or approaches. The PDQ Cancer Genetics Editorial Board uses a formal evidence ranking system in developing its level-of-evidence designations.

Permission to Use This Summary

PDQ is a registered trademark. Although the content of PDQ documents can be used freely as text, it cannot be identified as an NCI PDQ cancer information summary unless it is presented in its entirety and is regularly updated. However, an author would be permitted to write a sentence such as “NCI’s PDQ cancer information summary about breast cancer prevention states the risks succinctly: [include excerpt from the summary].”

The preferred citation for this PDQ summary is:

PDQ® Cancer Genetics Editorial Board. PDQ Genetics of Prostate Cancer. Bethesda, MD: National Cancer Institute. Updated . Available at: https://www.cancer.gov/types/prostate/hp/prostate-genetics-pdq. Accessed . [PMID: 26389227]

Images in this summary are used with permission of the author(s), artist, and/or publisher for use within the PDQ summaries only. Permission to use images outside the context of PDQ information must be obtained from the owner(s) and cannot be granted by the National Cancer Institute. Information about using the illustrations in this summary, along with many other cancer-related images, is available in Visuals Online, a collection of over 2,000 scientific images.

Disclaimer

The information in these summaries should not be used as a basis for insurance reimbursement determinations. More information on insurance coverage is available on Cancer.gov on the Managing Cancer Care page.

Contact Us

More information about contacting us or receiving help with the Cancer.gov website can be found on our Contact Us for Help page. Questions can also be submitted to Cancer.gov through the website’s Email Us.

Blogs


May 31, 2023

A Poet’s Autobiography of Cancer

by OncoLink Team


July 19, 2022

Managing Side Effects with South Asian Foods

by OncoLink Team