jueves, 11 de abril de 2019

Genetics of Prostate Cancer (PDQ®) 3/5 —Health Professional Version - National Cancer Institute

Genetics of Prostate Cancer (PDQ®)—Health Professional Version - National Cancer Institute

National Cancer Institute



Genetics of Prostate Cancer (PDQ®)–Health Professional Version




Clinical Application of Genetic Testing for Inherited Prostate Cancer




Criteria for Genetic Testing in Prostate Cancer

The criteria for consideration of genetic testing for prostate cancer susceptibility varies depending on the emerging guidelines and expert opinion consensus as summarized in Table 3.[1-5] Identification of men for inherited prostate cancer genetic testing is based upon family history criteria, personal/disease characteristics, and tumor sequencingresults. Actual genes to test vary on the basis of specific guidelines or consensus conference recommendations. The National Comprehensive Cancer Network (NCCN) Genetic/Familial High-Risk Assessment: Breast and Ovarian Cancer guideline is focused on BRCA1/BRCA2 testing on the basis of various testing criteria.[3] The NCCN Prostate Cancer treatment guideline states to test BRCA1/BRCA2ATMPALB2, and FANCA for men meeting specific testing indications.[4] A 2017 consensus conference addressed the role of genetic testing for inherited prostate cancer.[1] Family history–based indications for testing included testing for BRCA1/BRCA2HOXB13, or DNA mismatch repair (MMR) genes. Tumor sequencing with potential findings of germline variants in BRCA1/BRCA2 or DNA MMR genes, as well as other genes, is recommended for confirmatory germline testing. Somaticfindings for which germline testing is considered include:
  • Somatic variants 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.[6]
HOXB13 and ATM had lower level of consensus for testing on the basis of tumor sequencing. Men with metastatic castration-resistant prostate cancer were recommended to undergo genetic testing for BRCA1/BRCA2 (higher level of consensus) and ATM (moderate level of consensus). A second consensus conference focused on advanced prostate cancer stated that among panelists that recommended genetic testing on the basis of various criteria, there was agreement to use large panel testing including homologous recombination and DNA MMR genes.[2] Available genetic testing indications from guidelines and consensus conferences are shown in Table 3.
Table 3. Indications for Genetic Testing for Prostate Cancer Risk
ENLARGE
Philadelphia Prostate Cancer Consensus Conference (Giri et al. 2018)a [1]NCCN Genetic/Familial High-Risk Assessment: Breast and Ovarian (Version 2.2019)b [3]NCCN Prostate Cancer (Version 4.2018)c [4]NCCN Prostate Cancer Early Detection (Version 2.2018)d [5]European Advanced Prostate Cancer Consensus Conference (Gillessen et al. 2017)e [2]
FDR = first-degree relative; HBOC = hereditary breast and ovarian cancer; MMR = mismatch repair; NCCN = National Comprehensive Cancer Network; PSA = prostate-specific antigen.
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 and Ovarian guideline focuses on testing for BRCA1/BRCA2pathogenic variants.
cNCCN Prostate Cancer guideline specifies that homologous recombination gene pathogenic variants and DNA MMR gene pathogenic variants include variants in the following genes: BRCA1BRCA2ATMPALB2FANCAMLH1MSH2MSH6, or PMS2.
dNCCN Prostate Cancer Early Detection guideline does not specifically state which genes to test, but describes consideration of BRCA1/BRCA2 status and the status of other cancer risk genes when discussing prostate cancer screening.
eGillessen et al. endorsed the use of large panel testing including homologous recombination and DNA MMR genes.
Family History CriteriaAll men with prostate cancer from families meeting established testing or syndromic criteria for HBOC; hereditary prostate cancer; and Lynch syndromePersonal history of Gleason score ≥7 prostate cancer with: ≥1 biologic relative with ovarian, pancreatic, or metastatic prostate cancer at any age or breast cancer <50 y; or ≥2 biologic relatives with breast or prostate cancer (any grade) at any age; or Ashkenazi Jewishancestry>1 relative with breast, ovarian, or pancreatic cancer; >1 relative with a family history suggestive of Lynch syndrome (colorectal, endometrial, gastric, ovarian, pancreatic, small bowel, urothelial, kidney, or bile duct cancer)None providedPositive family history of prostate cancer
Men affected with prostate cancer with >2 close biologic relatives with a cancer associated with HBOC; hereditary prostate cancer, and Lynch syndromePositive family history of other cancer syndromes (HBOC and/or pancreatic cancer and/or Lynch syndrome)
Prostate cancer diagnosed <55 y in an FDRBrother, father, or multiple family members with prostate cancer <60 y
Death <60 y from prostate cancer in an FDR
Disease CharacteristicsAll men with metastatic prostate cancerPersonal history of metastatic prostate cancer (radiographic evidence or biopsy proven)Men with high-/very-high-risk clinically localized, regional, or metastatic diseaseNone providedMen with newly diagnosed metastatic prostate cancer (62% of panel voted in favor of genetic counseling/testing in a minority of selected patients)
Prostate cancer diagnosed <55 yProstate cancer diagnosed <60 y
Tumor CharacteristicsMen with prostate cancer whose somatic testing reveals the possibility of a germline variant in a cancer risk geneBRCA1/BRCA2somatic variant detected in the absence of germline testingConsider tumor testing for homologous recombination gene pathogenic variants and DNA MMR gene pathogenic variants in high-risk, very-high-risk, regional, or metastatic diseaseNone provided
Screening in BRCA1 CarriersFollow NCCN Genetic/Familial High-Risk Assessment: Breast and Ovarian guidelinesConsider prostate cancer screening starting at age 45 yNone providedBRCA1/BRCA2status and status of other risk genes should be considered in screening discussions
Interval of screening determined by baseline PSA level as specified in NCCN Prostate Cancer Early Detection Version 2.2018
Screening in BRCA2 CarriersBaseline PSA >40 y or 10 years prior to the earliest age of prostate cancer in the familyRecommend prostate cancer screening starting at age 45 yNone providedBRCA1/BRCA2status and status of other risk genes should be considered in screening discussions
Interval of screening determined by baseline PSA levelInterval of screening determined by baseline PSA level as specified in NCCN Prostate Cancer Early Detection Version 2.2018
Screening in HOXB13CarriersBaseline PSA >40 y or 10 years prior to the earliest age of prostate cancer in the familyNone providedNone providedNone provided
Interval of screening determined by baseline PSA level

Multigene (Panel) Testing in Prostate Cancer

Since the availability of next-generation sequencing and the elimination of patent restrictions, several clinical laboratories now offer genetic testing through multigene panels at a cost comparable to single-gene testing. A caveat is the possible finding of a variant of uncertain significance, where the clinical significance remains unknown. (Refer to the Multigene [panel] testing section in the PDQ summary on Cancer Genetics Risk Assessment and Counseling for more information about multigene testing, including genetic education and counseling considerations, and research examining the use of multigene testing.) This section summarizes the evidence for additional genes that may be on prostate cancer susceptibility 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.[7]

Genetic Testing for Prostate Cancer Risk Assessment

Genetic testing for pathogenic variants in genes with some association with prostate cancer risk is now available and has the potential to identify men at increased risk of prostate cancer. Research from selected cohorts has reported that prostate cancer risk is elevated in men with pathogenic variants in BRCA1BRCA2, and on a smaller scale, in MMR genes. Because clinical genetic testing is available for these genes, information about risk of prostate cancer on the basis of alterations in these genes is included in this section. In addition, pathogenic variants in HOXB13 are reported to account for a small proportion of hereditary prostate cancer. This section summarizes the evidence for these genes and additional genes that may be on prostate cancer susceptibility panel tests.

BRCA1 and BRCA2

Studies of male carriers of BRCA1 [8] and BRCA2 pathogenic variants demonstrate that these individuals have a higher risk of prostate cancer and other cancers.[9] Prostate cancer in particular has been observed at higher rates in male carriers of BRCA2 pathogenic variants than in the general population.[10]
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 4.
Table 4. 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; OCCR = Ovarian Cancer Cluster Region; RR = relative risk; SIR = standardized incidence ratio.
aIncludes all cancers except breast, ovarian, and nonmelanoma skin cancers.
BCLC (1999) [11]BCLC family set that included 173 BRCA2 linkage – 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) [12]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) [8]BCLC family set that included 7,106 women and 4,741 men, among which 2,245 were carriers of BRCA1pathogenic 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) [10]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)
Estimates derived from the Breast Cancer Linkage Consortium may be overestimated because these data are generated from a highly select population of families ascertained for significant evidence of risk of breast cancer and ovarian cancer and suitability for linkage analysis. However, a review of the relationship between germline pathogenic variants in BRCA2 and prostate cancer risk supports the view that this gene confers a significant increase in risk among male members of hereditary breast and ovarian cancer families but that it likely plays only a small role, if any, in site-specific, multiple-case prostate cancer families.[13] In addition, the clinical validity and utility of BRCA testing solely on the basis of evidence for hereditary prostate cancer susceptibility has not been established.
One study has assessed the relationship between germline DNA repair gene pathogenic variants and metastatic prostate cancer. Of 692 men unselected for cancer family history or age at diagnosis, 5.3% (37 of 692) were found to have a BRCA2 pathogenic variant, and 0.9% (6 of 692) had a BRCA1 pathogenic variant.[7]
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.[14-16] Two specific BRCA1 pathogenic variants (185delAG and 5382insC) and one BRCA2pathogenic variant (6174delT) are common in individuals of AJ ancestry. Carrier frequencies for these pathogenic variants in the general Jewish population are 0.9% (95% confidence interval [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.[17-20] (Refer to the High-Penetrance Breast and/or Gynecologic Cancer Susceptibility Genes section in the PDQ summary on Genetics of Breast and Gynecologic Cancers for more information about BRCA1 and BRCA2 genes.) 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 BRCAAshkenazi 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.[20] 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 5 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 5. Case-Control Studies in Ashkenazi Jewish Populations of BRCA1 and BRCA2 and Prostate Cancer Risk
ENLARGE
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.
Guisti et al. (2003) [21]Cases: 979 consecutive AJ men from Israel diagnosed with prostate cancer between 1994 and 1995Cases: 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) [22]Cases: 251 unselected AJ men treated for prostate cancer between 2000 and 2002Cases: 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) [23]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 BRCA1pathogenic variants (OR, 2.23; 95% CI, 0.84–5.86) and carriers of BRCA2pathogenic 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) [24]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 BRCA15382insC 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 BRCA2pathogenic variants (85%) than in noncarriers (57%); P = .0002. Carriers of BRCA1/BRCA2pathogenic 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.[24]
Other populations
The association between prostate cancer and pathogenic variants in BRCA1 and BRCA2 has also been studied in other populations. Table 6 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 6. Case-Control Studies in Varied Populations of BRCA1 and BRCA2 and Prostate Cancer Risk
ENLARGE
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) [25]Cases: 75 Icelandic men diagnosed with prostate cancer <65 y, between 1983 and 1992, with available archival tissue blocksNot 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) [26]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) [27]Cases: 3,750 Polish men with prostate cancer unselected for age or family history and diagnosed between 1999 and 2012Cases: 14 (0.4%)Not assessedAny BRCA1pathogenic 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).[25,26,28] 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 7. Case Series of BRCA1 and BRCA2 and Prostate Cancer Risk
ENLARGE
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; UK = United Kingdom.
aEstimate calculated using RR data in UK general population.
Agalliu et al. (2007) [29]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) [30]266 individuals from 194 hereditary prostate cancer families, including 253 men affected with prostate cancer; median age at prostate cancer diagnosis, 58 yNot assessed0 (0%)Not assessedNot assessed31 nonsynonymous variations were identified; no truncating or pathogenic variants were detected.
Tryggvadóttir et al. (2007) [31]527 men diagnosed with prostate cancer between 1955 and 2004Not assessed30/527 (5.7%) carried the Icelandic founder pathogenic variant 999del5Not assessedNot assessedThe BRCA2999del5 pathogenic variant was associated with a lower mean age at prostate cancer diagnosis (69 vs. 74 y; P = .002)
Kote-Jarai et al. (2011) [32]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) [33]913 men with prostate cancer who participated in the UK Genetic Prostate Cancer Study; 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%)
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.[29]
Prostate cancer aggressiveness in carriers of BRCA pathogenic variants
The studies summarized in Table 8 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 8. Case-Control Studies of BRCA1 and BRCA2 and Prostate Cancer Aggressiveness
ENLARGE
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) [31]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) [23]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
– BRCA1185delAG pathogenic variant: OR, 3.54 (95% CI, 1.22–10.31)
Controls: 1,251 AJ men with no history of cancer– BRCA26174delT pathogenic variant: OR, 3.18 (95% CI, 1.37–7.34)
Edwards et al. (2010) [34]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) [24]Cases: 832 AJ men diagnosed with localized prostate cancer between 1988 and 2007, of which there were 6 carriers of BRCA1pathogenic variants and 20 carriers of BRCA2pathogenic variantsGleason score 7–10:Not assessedNot assessedThe BRCA15382insC founder pathogenic variant was not tested in this series.
Controls: 454 AJ men with no history of cancerBRCA26174delT pathogenic variant: HR, 2.63 (95% CI, 1.23–5.6; P= .001)
Thorne et al. (2011) [35]Cases: 40 men diagnosed with prostate cancer who were carriers ofBRCA2 pathogenic variants from 30 familial breast cancer families from Australia and New ZealandGleason score ≥8:PSA 10–100 ng/mL:Stage ≥pT3 at presentation:Carriers of BRCA2pathogenic variants were more likely to have high-risk disease by D’Amico criteriathan were noncarriers (77.5% vs. 58.7%, P = .05).
– BRCA2pathogenic variants: 35% (14/40)– BRCA2pathogenic variants: 44.7% (17/38)
– BRCA2pathogenic 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 BRCApathogenic variant found in the family– Controls: 33.0% (25/97)– BRCA2pathogenic variants: 10% (4/40)– Controls: 22.6% (21/97)
–Controls: 2.1% (2/97)
Castro et al. (2013) [36]Cases: 2,019 men diagnosed with prostate cancer from the UK, of whom 18 were carriers of BRCA1pathogenic variants and 61 were carriers of BRCA2pathogenic variantsGleason score >8:BRCA1median PSA: 8.9 (range, 0.7–3,000)Stage ≥pT3 at presentation:Nodal metastasis and distant metastasis were higher in men with a BRCApathogenic variant than in controls.
– BRCA1pathogenic variants: 27.8% (5/18)– BRCA1: 38.9% (7/18)
– BRCA2pathogenic 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/BRCA2noncarriers– 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) [37]Cases: 4,187 men who underwent prostate biopsy for elevated PSA or abnormal exam, including 26 men with at least one BRCAcoding 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 BRCA2pathogenic variant was inferior to that of men without a BRCA2pathogenic variant (61.8% vs. 94.3%; P < 10−4). Among the men with high-grade disease (Gleason 7–9), the presence of a BRCA2pathogenic 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
These studies suggest that prostate cancer in carriers of BRCA pathogenic variants may be associated with features of aggressive disease, including higher Gleason score, higher prostate-specific antigen (PSA) level at diagnosis, and higher tumor stage and/or grade at diagnosis , a finding that warrants consideration as patients undergo cancer risk assessment and genetic counseling.[3] Research is under way to gain insight into the biologic 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.[38]
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 BRCA2pathogenic 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.[39] The case-control studies summarized in Table 9 further assess this observation.
Table 9. Case-Control Studies of BRCA1 and BRCA2 and Survival Outcomes
ENLARGE
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) [31]30 men diagnosed with prostate cancer who were carriers of BRCA2999del5 founder pathogenic variants59 men with prostate cancer matched by birth and diagnosis year and confirmed not to carry the BRCA2999del5 pathogenic variantBRCA2999del5 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) [34]21 men diagnosed with prostate cancer who harbored a BRCA2pathogenic 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 cancerNot assessedOverall survival was lower in carriers of BRCA2pathogenic 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) [24]832 AJ men diagnosed with localized prostate cancer between 1988 and 2007, of which 6 were carriers of BRCA1pathogenic variants and 20 carriers of BRCA2pathogenic variants454 AJ men with no history of cancerAfter adjusting for stage, PSA, Gleason score, and therapy received:Not assessedThe BRCA15382insC 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 ofBRCA26174delT 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) [35]40 men diagnosed with prostate cancer who were carriers ofBRCA2 pathogenic variants from 30 familial breast cancer families from Australia and New Zealand97 men from 89 familial breast cancer families from Australia and New Zealand with prostate cancer and no BRCApathogenic variant found in the familyBRCA2carriers 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 controlsBRCA2carriers 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 BRCA1carriers available to include in the analysis.
Castro et al. (2013) [36]2,019 men diagnosed with prostate cancer from the UK, of whom 18 were carriers of BRCA1pathogenic variants and 61 were carriers of BRCA2pathogenic variants1,940 men who were BRCA1/BRCA2noncarriersProstate 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) [40]1,302 men from the UK with local or locally advanced prostate cancer, including 67 carriers ofBRCA1/BRCA2pathogenic variants1,235 men who were BRCA1/BRCA2noncarriersProstate 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
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.[41] 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.[42] 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 BRCA1variants, 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.[43] 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 polymorphisms (SNPs) tagging common haplotype variation in a 200-kb region surrounding and including BRCA1. Three SNPs in BRCA1 (rs1799950, rs3737559, and rs799923) were found to be associated with prostate cancer. The strongest association was observed for SNP rs1799950 (odds ratio [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, SNP rs1799950 was found to contribute to the linkage signal on chromosome 17q21 originally reported by the UM-PCGP.[41]

Mismatch repair (MMR) genes

Five genes are implicated in MMR, namely MLH1MSH2MSH6PMS2, 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, and duodenal cancers; and transitional cell cancers of the ureter and renal pelvis. Reports have suggested that prostate cancer may be observed in men harboring an MMR gene pathogenic variant.[44,45] 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.[46] 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 SNPs in three MMR genes (MLH1MSH2, and PMS2). This study provided some evidence supporting the contribution of genetic variation in MLH1 and overall risk of prostate cancer.[47] 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.[48] Other studies are attempting to characterize rates of prostate cancer in Lynch syndrome families and correlate molecular features with prostate cancer risk.[49]
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.[50] 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 hazard ratio (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.[51] 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).[52] 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 MSH2pathogenic variants (RR, 5.8; 95% CI, 2.6–20.9); the RRs in carriers of MLH1 and MSH6pathogenic 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.
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 indexprostate cancer patients remain to be determined.

HOXB13

Linkage to 17q21-22 was initially reported by the UM-PCGP from 175 pedigrees of families with hereditary prostate cancer.[41] 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.[53] 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).[54Probands 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; = 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; = 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%.[54]
A validation study from the International Consortium of Prostate Cancer Genetics confirmed HOXB13 as a susceptibility gene for prostate cancer risk.[55] 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, prostate cancer risk, and penetrance associated with the HOXB13 G84E pathogenic variant.[54,56-61] This pathogenic variant appears to be restricted to white men, primarily of European descent.[54,56-58] The highest carrier frequency of 6.25% was reported in Finnish early-onset cases.[59] A pooled analysis that included 9,016 cases and 9,678 controls of European Americans found an overall G84E pathogenic variant frequency of 1.34% among cases and 0.28% among controls.[60]
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.[62] 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.[60] 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).[57] 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).[63] An additional meta-analysis of 25 studies including 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 whites (OR, 2.673; 95% CI, 1.920–3.720), especially those of European descent. No association was found for breast or colorectal cancer.[64] 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.[65] In addition, one study identified no men of AJ ancestry who carried the G84E pathogenic variant.[66] 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).[67] 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.
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.[68] Another study from Australia reported an age-specific cumulative risk of prostate cancer of up to 60% by age 80 years.[69]
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 the HOXB13 G84E pathogenic variant have yet to be defined.

ATM

Ataxia telangiectasia (AT) is an autosomal recessive disorder characterized by neurologic deterioration, telangiectasias, immunodeficiency states, and hypersensitivity to ionizing radiation. It is estimated that 1% of the general population may be heterozygote carriers of ATM variants.[70] In the presence of DNA damage, the ATM protein is involved in mediating cell cycle arrest, DNA repair, and apoptosis.[71] Given evidence of other cancer risks in heterozygote carriers, evidence of an association with prostate cancer susceptibility continues to emerge. (Refer to the ATM section in the PDQ summary on Genetics of Breast and Gynecologic Cancers for more information about ATM and breast cancer.) A prospective case series of 10,317 Danish individuals with 36 years of follow-up, during which 2,056 individuals developed cancer, found that Ser49Cys was associated with prostate cancer (HR, 2.3; 95% CI, 1.1–5.0).[71] A retrospective case series of 692 men with metastatic prostate cancer unselected for cancer family history or age at diagnosis found that 1.6% (11 of 692) had an ATM pathogenic variant.[7]

CHEK2

CHEK2 has also been investigated for a potential association with prostate cancer risk. 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.[7]

TP53

TP53 has also been investigated for a potential association with prostate cancer risk. 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.[72] 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).[73] 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).[74]
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.[75]
Further evidence supports an association between prostate cancer and germline TP53pathogenic variants,[76-78] although additional studies to clarify the association with this gene are warranted.

NBN/NBS1

NBN, which is also known as NBS1 (Nijmegan breakage syndrome 1), 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 a NBN pathogenic variant.[7]

EPCAM

EPCAM (epithelial cellular adhesion molecule) 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 MSH2, induce methylation of the MSH2promoter resulting in MSH2 protein loss.[79] (Refer to the EPCAM section in the PDQ summary on Genetics of Colorectal Cancer for a more detailed discussion about EPCAM and Lynch syndrome.) Pathogenic variants in MSH2 that are associated with Lynch syndrome were found to be associated with increased risk of prostate cancer.[52] (Refer to the Mismatch Repair genes section of this summary for information about MSH2 and prostate cancer risk.) Thus far, studies ascertaining the spectrum of germline pathogenic variants in men with prostate cancer have not identified pathogenic variants in EPCAM.[7]

Germline Pathogenic Variants in Men 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.[80] 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.[81] Germline pathogenic variants were found in BRCA1BRCA2MSH2MSH6PALB2PMS2ATMBRIP1NBN, as well as other genes. These and other studies are summarized in Table 10. 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 10. Summary of Tumor Sequencing Studies With Germline Results
StudyCohortGermline Results for Prostate CancerComments
ADT = androgen deprivation therapy; AR = androgen receptor; mCRPC = metastatic castration-resistant prostate cancer; OS = overall survival; PFS = progression-free survival.
aPotential overlap of cohorts.
Robinson et al. (2015)a[80]Whole-exomeand transcriptome sequencing of bone or soft tissue tumor biopsies from a cohort of 150 men with mCRPC8% had germline pathogenic variants:
BRCA2: 9/150 (6.0%)
ATM: 2/150 (1.3%)
BRCA1: 1/150 (0.7%)
Pritchard et al. (2016)a[7]692 men with metastatic prostate cancer, unselected for family history; analysis focused on 20 genes involved in maintaining DNA integrity and associated with autosomal dominantcancer–predisposing syndromes82/692 (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 (< .001).
BRCA2: 37/692 (5.3%)
ATM: 11/692 (1.6%)
BRCA1: 6/692 (0.9%)
Schrader et al. (2016) [82]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%)
Annala et al. (2017) [83]319 men with mCRPC; performed germline sequencing of 22 DNA repair genes24/319 (7.5%) had germline pathogenic variants:Patients with DNA repair defects had decreased responses to ADT:
• BRCA2: 16/319 (5.0%)• Time from ADT initiation to mCRPC (mo): Germline positive, 11.8 (n = 22) vs. germline negative, 19.0 (n = 113) (= .031).
• ATM: 1/319 (0.3%)
• BRCA1: 1/319 (0.3%)• PFS on first-line AR-targeted therapy (mo): Germline positive, 3.3 vs. germline negative, 6.2 (= .01).
• PALB2: 2/319 (0.6%)

Genetic Testing for Prostate Cancer Precision Oncology

Targeted therapies on the basis of genetic results are increasingly driving options for treatment in oncology. With greater recognition of germline pathogenic variants in men with advanced or metastatic prostate cancer, treatment options have also expanded. Men with DNA repair pathogenic variants have been reported to have increased clinical activity to poly (ADP-ribose) polymerase (PARP) inhibitors.[84] On the basis of increasing data, olaparib was given Breakthrough Therapy Designation by the U.S. Food and Drug Administration (FDA) for BRCA1/BRCA2 or ATM mutated metastatic prostate cancer. Furthermore, pembrolizumab has received FDA approval for MSI-high and MMR-deficient cancers, which may also have an impact on treatment options for men with metastatic prostate cancer. NCCN Prostate Cancer Treatment guidelines also include tumor testing and/or germline testing of men with regional or metastatic prostate cancer for MMR and homologous recombination repair deficiency to identify men who may be candidates for pembrolizumab or olaparib.[4] The guidelines also state to consider BRCA1/BRCA2 status in active surveillance discussions for men with very low risk to unfavorable intermediate risk of disease. Thus, genetic results are increasingly informing treatment and management strategies for prostate cancer. Confirmation of somatic variants through germline testing is needed so that additional recommendations can be made regarding cancer risk for patients and families.
For a summary of available clinical practice guidelines for genetic testing in prostate cancer, refer to Table 3.


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. [PUBMED Abstract]
  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. [PUBMED Abstract]
  3. National Comprehensive Cancer Network: NCCN Clinical Practice Guidelines in Oncology: Genetic/Familial High-Risk Assessment: Breast and Ovarian. Version 2.2019. Plymouth Meeting, Pa: National Comprehensive Cancer Network, 2018. Available online with free registration. Last accessed October 22, 2018.
  4. National Comprehensive Cancer Network: NCCN Clinical Practice Guidelines in Oncology: Prostate Cancer. Version 4.2018. Plymouth Meeting, Pa: National Comprehensive Cancer Network, 2018. Available online with free registration. Last accessed October 23, 2018.
  5. National Comprehensive Cancer Network: NCCN Clinical Practice Guidelines in Oncology: Prostate Cancer Early Detection. Version 2.2018. Fort Washington, Pa: National Comprehensive Cancer Network, 2018. Available online with free registration. Last accessed December 14, 2018.
  6. 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. [PUBMED Abstract]
  7. 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. [PUBMED Abstract]
  8. Thompson D, Easton DF; Breast Cancer Linkage Consortium: Cancer Incidence in BRCA1 mutation carriers. J Natl Cancer Inst 94 (18): 1358-65, 2002. [PUBMED Abstract]
  9. 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. [PUBMED Abstract]
  10. 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. [PUBMED Abstract]
  11. Cancer risks in BRCA2 mutation carriers. The Breast Cancer Linkage Consortium. J Natl Cancer Inst 91 (15): 1310-6, 1999. [PUBMED Abstract]
  12. 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. [PUBMED Abstract]
  13. 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. [PUBMED Abstract]
  14. 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. [PUBMED Abstract]
  15. 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. [PUBMED Abstract]
  16. 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. [PUBMED Abstract]
  17. 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. [PUBMED Abstract]
  18. 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. [PUBMED Abstract]
  19. 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. [PUBMED Abstract]
  20. 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. [PUBMED Abstract]
  21. 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. [PUBMED Abstract]
  22. 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. [PUBMED Abstract]
  23. 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. [PUBMED Abstract]
  24. 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. [PUBMED Abstract]
  25. 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. [PUBMED Abstract]
  26. 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. [PUBMED Abstract]
  27. 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. [PUBMED Abstract]
  28. Cybulski C, Górski B, Gronwald J, et al.: BRCA1 mutations and prostate cancer in Poland. Eur J Cancer Prev 17 (1): 62-6, 2008. [PUBMED Abstract]
  29. 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. [PUBMED Abstract]
  30. 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. [PUBMED Abstract]
  31. 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. [PUBMED Abstract]
  32. 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. [PUBMED Abstract]
  33. Leongamornlert D, Mahmud N, Tymrakiewicz M, et al.: Germline BRCA1 mutations increase prostate cancer risk. Br J Cancer 106 (10): 1697-701, 2012. [PUBMED Abstract]
  34. 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. [PUBMED Abstract]
  35. 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. [PUBMED Abstract]
  36. 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. [PUBMED Abstract]
  37. 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. [PUBMED Abstract]
  38. Taylor RA, Fraser M, Livingstone J, et al.: Germline BRCA2 mutations drive prostate cancers with distinct evolutionary trajectories. Nat Commun 8: 13671, 2017. [PUBMED Abstract]
  39. 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. [PUBMED Abstract]
  40. 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. [PUBMED Abstract]
  41. 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. [PUBMED Abstract]
  42. 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. [PUBMED Abstract]
  43. 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. [PUBMED Abstract]
  44. 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. [PUBMED Abstract]
  45. Haraldsdottir S, Hampel H, Wei L, et al.: Prostate cancer incidence in males with Lynch syndrome. Genet Med 16 (7): 553-7, 2014. [PUBMED Abstract]
  46. 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. [PUBMED Abstract]
  47. 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. [PUBMED Abstract]
  48. 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. [PUBMED Abstract]
  49. Dominguez-Valentin M, Joost P, Therkildsen C, et al.: Frequent mismatch-repair defects link prostate cancer to Lynch syndrome. BMC Urol 16: 15, 2016. [PUBMED Abstract]
  50. 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. [PUBMED Abstract]
  51. 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. [PUBMED Abstract]
  52. 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. [PUBMED Abstract]
  53. 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. [PUBMED Abstract]
  54. 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. [PUBMED Abstract]
  55. 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. [PUBMED Abstract]
  56. 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. [PUBMED Abstract]
  57. 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. [PUBMED Abstract]
  58. 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. [PUBMED Abstract]
  59. 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. [PUBMED Abstract]
  60. 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. [PUBMED Abstract]
  61. 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. [PUBMED Abstract]
  62. 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. [PUBMED Abstract]
  63. 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. [PUBMED Abstract]
  64. 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. [PUBMED Abstract]
  65. 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. [PUBMED Abstract]
  66. 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. [PUBMED Abstract]
  67. 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. [PUBMED Abstract]
  68. 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. [PUBMED Abstract]
  69. 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. [PUBMED Abstract]
  70. 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. [PUBMED Abstract]
  71. 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. [PUBMED Abstract]
  72. 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. [PUBMED Abstract]
  73. 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. [PUBMED Abstract]
  74. 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. [PUBMED Abstract]
  75. 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. [PUBMED Abstract]
  76. 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. [PUBMED Abstract]
  77. 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. [PUBMED Abstract]
  78. 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. [PUBMED Abstract]
  79. 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. [PUBMED Abstract]
  80. Robinson D, Van Allen EM, Wu YM, et al.: Integrative clinical genomics of advanced prostate cancer. Cell 161 (5): 1215-28, 2015. [PUBMED Abstract]
  81. 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. [PUBMED Abstract]
  82. 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. [PUBMED Abstract]
  83. 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. [PUBMED Abstract]
  84. Mateo J, Carreira S, Sandhu S, et al.: DNA-Repair Defects and Olaparib in Metastatic Prostate Cancer. N Engl J Med 373 (18): 1697-708, 2015. [PUBMED Abstract]

No hay comentarios:

Publicar un comentario