Conoce sobre los beneficios médicos, dentales, de farmacia y voluntarios que tu empleador puede ofrecer.
This executive summary reviews the topics covered in this PDQ summary on the genetics of prostate cancer, with hyperlinks to detailed sections below that describe the evidence on each topic.
A genetic contribution to prostate cancer risk has been documented, and knowledge about the molecular genetics of the disease is increasing. Clinical management based on knowledge of inherited pathogenic variants is emerging. Factors suggestive of a genetic contribution to prostate cancer include the following: 1) multiple affected first-degree relatives (FDRs) with prostate cancer, including three successive generations with prostate cancer in the maternal or paternal lineage; 2) early-onset prostate cancer (age ≤55 y); and 3) prostate cancer with a family history of other cancers (e.g., breast, ovarian, pancreatic).
Several genes and chromosomal regions have been found to be associated with prostate cancer in various linkage analyses, case-control studies, genome-wide association studies (GWAS), next-generation sequencing (NGS), and admixture mapping studies. Pathogenic variants in genes, such as BRCA1, BRCA2, the mismatch repair genes, and HOXB13 confer modest to moderate lifetime risk of prostate cancer. Some, such as BRCA2, have emerging clinical relevance in the treatment and screening for prostate cancer. In addition, GWAS have identified more than 150 SNVs associated with the development of prostate cancer, but the clinical utility of these findings remains uncertain. Studies are ongoing to assess whether combinations of these SNVs (e.g., polygenic risk scores) may have clinical relevance in identifying individuals at increased risk of the disease. Studies analyzing the association between variants and aggressive disease are also ongoing.
Information is limited about the efficacy of commonly available screening tests such as the digital rectal exam and serum prostate-specific antigen (PSA) levels in men genetically predisposed to developing prostate cancer. Initial reports of targeted PSA screening of carriers of BRCA pathogenic variants has yielded a higher proportion of aggressive disease. 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. For example, some experts suggest initiating prostate cancer screening at age 40 years in carriers of BRCA2 pathogenic variants and consideration of screening in BRCA1 carriers. Inherited variants may influence treatment decisions, particularly for males with pathogenic variants in DNA repair genes. Studies have reported improved response rates to poly (ADP-ribose) polymerase (PARP) inhibition and platinum-based chemotherapy among males with metastatic, castrate-resistant prostate cancer carrying germline pathogenic variants in BRCA2 and other DNA repair genes.
Psychosocial research in men at increased hereditary risk of prostate cancer has focused on risk perception, interest in genetic testing, and screening behaviors. Study conclusions vary regarding whether FDRs of prostate cancer patients accurately estimate their prostate cancer risk, with some studies reporting that men with a family history of prostate cancer consider their risk to be the same as or less than that of the average man. Factors such as being married and the confusion between benign prostatic hyperplasia and prostate cancer have been found to influence perceived risk of prostate cancer. Studies conducted before the availability of genetic testing for prostate cancer susceptibility showed that factors found to positively influence men's hypothetical interest in genetic testing included the advice of their primary care physician, a combination of the emotional distress and concern about prostate cancer treatment effects, and having children. Several small studies have examined the behavioral correlates of prostate cancer screening at average and increased prostate cancer risk based on family history; in general, results appear contradictory regarding whether men with a family history are more likely to be screened than those not at risk and whether the screening is appropriate for their risk status. Research is ongoing to better understand and address psychosocial and behavioral issues in high-risk families.
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.
The public health burden of prostate cancer is substantial. A total of 268,490 new cases of prostate cancer and 34,500 deaths from the disease are anticipated in the United States in 2022, making it the most frequent nondermatological cancer among U.S. males.[
Some men with prostate cancer remain asymptomatic and die from unrelated causes rather than as a result of the cancer itself. This may be due to the advanced age of many men at the time of diagnosis, slow tumor growth, or response to therapy.[
Prostate cancer exhibits tremendous differences in incidence among populations worldwide; the annual incidence rate of prostate cancer in different regions in the world ranges from 86.4 cases per 100,000 men in Australia and New Zealand to 5.0 cases per 100,000 men in South Central Asia.[
These differences may be due in part to environmental and social influences (such as access to health care), which may affect the development and progression of the disease.[
Risk Factors for Prostate Cancer
The four most important recognized risk factors for prostate cancer in the United States are:
Age
Age is an important risk factor for prostate cancer. 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 456 for men 49 years or younger, 1 in 54 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, with an overall lifetime risk of developing prostate cancer of 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 is increasing, and there is evidence that some cases may be more aggressive.[
Ancestry
The risk of developing prostate cancer is dramatically higher among non-Hispanic Black individuals (172.6 cases/100,000 men) when compared with other racial groups in the United States (non-Hispanic White, 99.9 cases/100,000 men; Asian or Pacific Islander, 55.0 cases/100,000 men; American Indian or Alaska Native, 79.8 cases/100,000 men; and Hispanic or Latino, 85.3 cases/100,000 men).[
Family cancer history
Prostate cancer is highly heritable; the inherited risk of prostate cancer has been estimated to be as high as 60%. As with breast and colon cancer, familial clustering of prostate cancer has been reported frequently.[
Although some of the prostate cancer studies examining risks associated with family history have used hospital-based series, several studies described population-based series.[
Risk Group | RR for Prostate Cancer (95% CI) |
---|---|
CI = confidence interval; FDR = first-degree relative. | |
a Adapted from Kiciński et al.[ |
|
Brother(s) with prostate cancer diagnosed at any age | 3.14 (2.37–4.15) |
Father with prostate cancer diagnosed at any age | 2.35 (2.02–2.72) |
One affected FDR diagnosed at any age | 2.48 (2.25–2.74) |
Affected FDRs diagnosed <65 y | 2.87 (2.21–3.74) |
Affected FDRs diagnosed ≥65 y | 1.92 (1.49–2.47) |
Second-degree relativesdiagnosed at any age | 2.52 (0.99–6.46) |
Two or more affected FDRs diagnosed at any age | 4.39 (2.61–7.39) |
Among the many data sources included in this meta-analysis, those from the Swedish population-based Family-Cancer Database warrant special comment. These data were derived from a resource that contained more than 11.8 million individuals, among whom there were 26,651 men with medically verified prostate cancer, of which 5,623 were familial cases.[
A separate analysis of this Swedish database reported that the cumulative (absolute) risks of prostate cancer among men in families with two or more affected cases were 5% by age 60 years, 15% by age 70 years, and 30% by age 80 years, compared with 0.45%, 3%, and 10%, respectively, by the same ages in the general population. The risks were even higher when the affected father was diagnosed before age 70 years.[
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.[
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.[
Prostate cancer clusters with particular intensity in some families. Highly to moderately penetrant genetic variants are thought to be associated with prostate cancer risk in these families. Members of these families may benefit from genetic counseling. Emerging 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.[
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),[
There is variable evidence that family history alone is associated with inferior clinical outcomes. In a cohort of 7,690 men in Germany who were undergoing radical prostatectomy for localized prostate cancer, family history had no bearing on prostate cancer–specific survival.[
Germline genetic variants
There are multiple germline pathogenic variants and single nucleotide variants across the genome that are associated with prostate cancer risk. For more information about these genetic variants, see the National Human Genome Research Institute GWAS catalog, and for information about prostate cancer genetic testing, see the Clinical Application of Genetic Testing for Inherited Prostate Cancer section.
Multiple Primaries
The SEER Cancer Registries assessed the risk of developing a second primary cancer in 292,029 men diagnosed with prostate cancer between 1973 and 2000. Excluding subsequent prostate cancer and adjusting for the risk of death from other causes, the cumulative incidence of a second primary cancer among all patients was 15.2% at 25 years (95% CI, 15.0%–15.4%). There was a significant risk of new malignancies (all cancers combined) among men diagnosed before age 50 years, no excess or deficit in cancer risk in men aged 50 to 59 years, and a deficit in cancer risk in all older age groups. The authors suggested that this deficit may be attributable to decreased cancer surveillance in an elderly population. Excess risks of second primary cancers included cancers of the small intestine, soft tissue, bladder, thyroid, and thymus; and melanoma. Prostate cancer diagnosed in patients aged 50 years or younger was associated with an excess risk of pancreatic cancer, which may relate to the inheritance of pathogenic variants in BRCA1/BRCA2.[
A review of more than 441,000 men diagnosed with prostate cancer between 1992 and 2010 demonstrated similar findings, with an overall reduction in the risk of being diagnosed with a second primary cancer. This study also examined the risk of second primary cancers in 44,310 men (10%) by treatment modality for localized cancer. The study suggested that men who received radiation therapy had increases in bladder (standardized incidence ratio [SIR], 1.42) and rectal cancer risk (SIR, 1.70) compared with those who did not receive radiation therapy (SIRbladder, 0.76; SIRrectal, 0.74).[
One Swedish study using the nationwide Swedish Family Cancer Database assessed the role of family history in the risk of a second primary cancer after prostate cancer. Of 80,449 men with prostate cancer, 6,396 developed a second primary malignancy. Those with a family history of cancer were found to have an increased risk for a second primary cancer with the greatest risk consisting of colorectal cancer (RR, 1.78; 95% CI, 1.56–1.90), lung cancer (RR, 2.29; 95% CI, 1.65–3.18), kidney cancer (RR, 3.59; 95% CI, 1.61–7.99), bladder cancer (RR, 3.84; 95% CI, 2.63–5.60), melanoma (RR, 2.30; 95% CI, 1.86–2.93), squamous cell skin cancer (RR, 2.10; 95% CI, 1.92–2.26), and leukemia (RR, 3.88; 95% CI, 1.94–7.77). Among probands with prostate cancer with a family history of cancer, 47% of deaths were secondary to a second primary malignancy. The cumulative incidence of a second primary cancer by age 83 years was highest (35%) in those participants with a family history of cancer in contrast to those without a family history of cancer (28%).[
Data are emerging that prostate cancer patients who have at least one additional primary malignancy disproportionately harbor pathogenic variants in known cancer-predisposing genes, such as BRCA2 and MLH1.[
Risk of Other Cancers in Multiple-Case Families
Several reports have suggested an elevated risk of various other cancers among relatives within multiple-case prostate cancer families, but none of these associations have been established definitively.[
In a population-based Finnish study of 202 multiple-case prostate cancer families, no excess risk of all cancers combined (other than prostate cancer) was detected in 5,523 family members. Female family members had a marginal excess of gastric cancer (SIR, 1.9; 95% CI, 1.0–3.2). No difference in familial cancer risk was observed when families affected by clinically aggressive prostate cancers were compared with those having nonaggressive prostate cancer. These data suggest that familial prostate cancer is a cancer site–specific disorder.[
A study from the Swedish Family Cancer Database reported an increased risk of the following cancers in families where multiple members had a prostate cancer diagnosis: myeloma (RR, 2.44; 95% CI, 1.24–4.82), kidney cancer (RR, 2.32; 95% CI, 1.23–4.36), nonthyroid endocrine tumors (RR, 2.18; 95% CI, 1.06–4.49), melanoma (RR, 1.82; 95% CI, 1.18–2.80), nervous system tumors (RR, 1.77; 95% CI, 1.08–2.91), and female breast cancer (RR, 1.37; 95% CI, 1.02–1.86).[
Inheritance of Prostate Cancer Risk
Many types of epidemiologic studies (case-control, cohort, twin, family) strongly suggest that prostate cancer susceptibility genes exist in the population. Analysis of longer follow-up of the monozygotic (MZ) and dizygotic (DZ) twin pairs in Scandinavia concluded that 58% (95% CI, 52%–63%) of prostate cancer risk may be accounted for by heritable factors.[
The first segregation analysis was performed in 1992 using families from 740 consecutive probands who had radical prostatectomies between 1982 and 1989. The study results suggested that familial clustering of disease among men with early-onset prostate cancer was best explained by the presence of a rare (frequency, 0.003) autosomal dominant, highly penetrant allele(s).[
Subsequent segregation analyses generally agreed with the conclusions but differed in the details regarding frequency, penetrance, and mode of inheritance.[
References:
Various research methods have been used to understand the genetic variation that is associated with prostate cancer. Different methods can identify unique phenotypes or inheritance patterns. The sections below describe prostate cancer research, which uses various methods to uncover the genetic basis of prostate cancer. Linkage studies can help identify susceptibility genes that predispose to genetic disease. These studies are typically performed on large, high-risk families in which multiple cases of a particular disease have occurred. Typically, pathogenic variants identified through linkage analyses are rare in the population, are moderately to highly penetrant in families, and have large (e.g., relative risk [RR] >2.0) effect sizes. Pathogenic variants that are identified in linkage studies allow individuals to receive clinical treatment.
Genome-wide association studies (GWAS) are another methodology used to identify candidate loci associated with prostate cancer. Genetic variants identified from GWAS typically are common in the population and have low to modest effect sizes for prostate cancer risk. The clinical role of markers identified from GWAS is an active area of investigation. Case-control studies are useful in validating the findings of linkage studies and GWAS as well as for studying candidate gene alterations for association with prostate cancer risk, although the clinical role of findings from case-control studies needs to be further defined.
Linkage Analyses
Introduction to linkage analyses
The recognition that prostate cancer clusters within families has led many investigators to collect multiple-case families with the goal of localizing prostate cancer susceptibility genes through linkage studies.
Linkage studies are typically performed on high-risk kindreds in whom multiple cases of a particular disease have occurred in an effort to identify disease susceptibility genes. Linkage analysis statistically compares the genotypes between affected and unaffected individuals within the extended family and looks for associations between inherited genetic markers and the disease trait. If an association between a variation at a particular chromosomal region and the disease trait is found (linkage), it provides statistical evidence that the genetic locus harbors a disease susceptibility gene. Once a genomic region of interest has been identified through linkage analysis, additional studies are required to prove that there truly is a susceptibility gene at that position. Linkage analysis is influenced by the following:
Furthermore, because a standard definition of hereditary prostate cancer has not been accepted, prostate cancer linkage studies have not used consistent criteria for enrollment.[
Using these criteria, surgical series have reported that approximately 3% to 5% of men with prostate cancer will be from a family with hereditary prostate cancer.[
An additional issue in linkage studies is the high background rate of sporadic prostate cancer in the context of family studies. Because a man's lifetime risk of prostate cancer is one in eight,[
One way to address inconsistencies between linkage studies is to require inclusion criteria that define clinically significant disease (e.g., Gleason score ≥7, PSA ≥20 ng/mL) in an affected man.[
Investigators have also incorporated clinical parameters into linkage analyses with the goal of identifying genes that may influence disease severity.[
Susceptibility loci identified in linkage analyses
Linkage analyses led to the successful identification of HOXB13 at 17q21-22 as a prostate cancer susceptibility gene.[
In 2012, the first prostate cancer–specific germline genetic variant was identified in a linkage study.[
Linkage analyses in various familial phenotypes
Linkage studies have also been performed in specific populations or with specific clinical parameters to identify population-specific susceptibility genes or genes influencing disease phenotypes.
The African American Hereditary Prostate Cancer study conducted a genome-wide linkage study of 77 families with four or more affected men. Multipoint heterogeneity logarithm of the odds (HLOD) scores of 1.3 to less than 2.0 were observed using markers that map to 11q22, 17p11, and Xq21. Analysis of the 16 families with more than six men with prostate cancer provided evidence for two additional loci: 2p21 (multipoint HLOD score = 1.08) and 22q12 (multipoint HLOD score = 0.91).[
In an effort to identify loci contributing to prostate cancer aggressiveness, linkage analyses have been performed in families with clinically high-risk features such as: Gleason score 7 or higher, PSA of 20 ng/mL or higher, regional or distant cancer stage at diagnosis, or death from metastatic prostate cancer before age 65 years. One study of 123 families with two or more affected family members with aggressive prostate cancer discovered linkage at chromosome 22q11 and 22q12.3-q13.1.[
Evidence suggests that many of the prostate cancer risk loci discovered via linkage analysis account for disease in a small subset of families, which is consistent with the concept that prostate cancer exhibits locus heterogeneity. Several proposed prostate cancer susceptibility loci have been identified in families with multiple prostate cancer–affected individuals. Genes residing at risk loci discovered using linkage analysis include HPC1/RNASEL (1q25), PCAP (1q42.2-43), HPCX (Xq27-28), CAPB (1p36), and HPC20 (20q13),[
Case-Control Studies of Candidate Genes and Pathways
A case-control study evaluates factors of interest to assess for association with a condition. The design involves cases with a condition of interest, such as a specific disease or genetic variant, and a control sample without that condition. In most cases, researchers seek to match cases and controls with as many characteristics as possible (e.g., age, gender, and ethnicity) in order to isolate a particular genetic variant as the sole focus of interrogation. Limitations of case-control design with regard to identifying genetic factors include the following:[
Because of potential confounders in this line of inquiry, validation in independent datasets is required to establish a true association.[
Androgen receptor (AR) gene variants have been examined in relation to both prostate cancer risk and disease progression. The AR gene is a logical gene to interrogate because it is expressed during all stages of prostate carcinogenesis and is routinely overexpressed in advanced disease.[
Molecular epidemiology studies have also examined genetic polymorphisms of the SRD5A2 gene, which is involved in the androgen metabolism cascade. Two isozymes of 5-alpha-reductase exist. The gene that codes for 5-alpha-reductase type II (SRD5A2) is located on chromosome 2. It is expressed in the prostate, where testosterone is converted irreversibly to dihydrotestosterone by 5-alpha-reductase type II.[
Other investigators have explored the potential contribution of the variation in genes involved in the estrogen pathway. A Swedish population study of 1,415 prostate cancer cases and 801 age-matched controls examined the association of SNVs in the estrogen receptor-beta (ER-beta) gene and prostate cancer. One SNV in the promoter region of ER-beta, rs2987983, was associated with an overall prostate cancer risk of 1.23 and 1.35 for localized disease.[
Germline pathogenic variants in the tumor suppressor gene E-cadherin (CDH1) cause a hereditary form of gastric carcinoma. A SNV designated -160C/A, located in the promoter region of CDH1, has been found to alter the transcriptional activity of this gene.[
In a whole-exome germline sequencing cohort of 200 African American men and 452 European American men with aggressive prostate cancer along with ethnic- and age-matched controls, researchers found that variants in TET2 were associated with aggressive disease in the African American subpopulation. These variants were present in 24.4% of African American cases compared with 9.6% of controls.[
Several other gene groups have been the focus of case-control studies, including the steroid hormone pathway,[
Case-control studies assessed site-specific prostate cancer susceptibility in the following genes: EMSY, KLF6, AMACR, NBN, CHEK2, AR, SRD5A2, ER-beta, CDH1, and the toll-like receptor genes. The clinical validity and utility of genetic testing for any of these genes to assess risk has not been established. Validation and prospective series are needed in order to prove clinical utility.
Admixture Mapping
Admixture mapping is a method used to identify genetic variants associated with traits and/or diseases in individuals with mixed ancestry.[
Admixture mapping is a particularly attractive method for identifying genetic loci associated with increased prostate cancer risk among African American individuals. African American men are at higher risk of developing prostate cancer than are men of European ancestry, and the genomes of African American men are mosaics of regions from Africa and regions from Europe. It is therefore hypothesized that inherited variants accounting for the difference in incidence between the two groups must reside in regions enriched for African ancestry. In prostate cancer admixture studies, genetic markers for ancestry were genotyped genome-wide in African American cases and controls in a search for areas enriched for African ancestry in the men with prostate cancer. Admixture studies have identified the following chromosomal regions associated with prostate cancer:
Recent admixtures result in long stretches of linkage disequilibrium (up to hundreds of thousands of base pairs) of one particular ancestry.[
Genome-wide Association Studies
Overview
Introduction to GWAS
Genome-wide searches have successfully identified susceptibility alleles for many complex diseases,[
In a GWAS, allele frequency is compared for each SNV between cases and controls. Promising signals–in which allele frequencies deviate significantly in cases when compared with control populations–are validated in replication datasets. In order to have adequate statistical power to identify variants associated with a phenotype, large numbers of cases and controls, typically thousands of each, are studied. Because 1 million SNVs are typically evaluated in a GWAS, false-positive findings are expected to occur frequently when standard statistical thresholds are used. Therefore, stringent statistical rules are used to declare a positive finding, usually using a threshold of P < 1 × 10-7.[
To date, more than 150 variants associated with prostate cancer have been identified by well-powered GWAS. These variants have been validated in independent cohorts (for more information, see National Human Genome Research Institute GWAS catalog).[
The implications of these points are discussed in greater detail below. Additional detail can be found elsewhere.[
Susceptibility loci identified in GWAS
Beginning in 2006, multiple genome-wide studies seeking associations with prostate cancer risk converged on the same chromosomal locus, 8q24.[
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.[
GWAS in populations of non-European ancestry
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.[
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. One study interrogated 28 known prostate cancer risk loci via fine mapping in 3,425 African American cases and 3,290 African American controls.[
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.[
Clinical study of GWAS findings
Because the variants discovered by GWAS are markers of risk, there has been great interest in using genotype as a screening tool to predict the development of prostate cancer. As increasing numbers of risk SNVs have been discovered, they have been applied to clinical cohorts alongside traditional variables such as PSA and family history, although the clinical utility of this information has not been established.
An initial study of the first five known risk SNVs could not demonstrate that they added clinically meaningful data.[
In July 2012, the Agency for Healthcare Research and Quality (AHRQ) published a report that sought to address the clinical utility of germline genotyping of prostate cancer risk markers discovered by GWAS.[
Polygenic risk scores
In a 2018 study, 147 GWAS variants known to be associated with prostate cancer were used to calculate a polygenic risk score (PRS) for more than 140,000 men.[
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.[
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.[
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.[
Current GWAS findings account for only a portion of the estimated 58% of disease risk that is heritable. In addition, around 6% of the familial RR of prostate cancer has been attributed to rare genetic variants.[
In addition, other genetic polymorphisms, such as copy number variants, are becoming increasingly amenable to testing. 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. Notably, almost all reported prostate cancer risk alleles reside in nonprotein-coding regions of the genome; however, the underlying biological mechanism of disease susceptibility was initially unclear. It is now apparent that a large proportion of risk variants affect the activity of regulatory elements and, in turn, distal genes.[
Conclusions
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 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, such as the 1000 Genomes Project.[
Inherited Single Nucleotide Variants (SNVs) 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 to date regarding inherited risk of aggressive disease are considered preliminary. As described below, germline SNVs associated with prostate cancer aggressiveness are derived primarily from three methods of analysis: 1) annotation of common variants within candidate risk genes; 2) assessment of known overall prostate cancer risk SNVs for aggressiveness; and 3) GWAS for prostate cancer aggressiveness. Further work is needed to validate findings and assess these associations prospectively.
Like studies of the genetics of overall prostate cancer risk, initial studies of inherited risk of aggressive prostate cancer focused on polymorphisms in candidate genes.[
There has been great interest in launching more unbiased, genome-wide searches for inherited variants associated with indolent versus aggressive prostate cancer.
Associations between inherited variants and prostate cancer aggressiveness have been reported. A multistage, case-only GWAS led by the National Cancer Institute examined 12,518 prostate cancer cases and discovered an association between genotype and Gleason score at two polymorphisms: rs35148638 at 5q14.3 (RASA1, P = 6.49 × 10-9) and rs78943174 at 3q26.31 (NAALADL2, P = 4.18 × 10-8).[
A few GWAS designed specifically to focus on prostate cancer subjects with documented disease-related outcomes have been launched. In one study—a genome-wide analysis in which two of the largest international prostate cancer genotyped cohorts were combined for analysis (24,023 prostate cancer cases, including 3,513 disease-specific deaths)—no SNV was significantly associated with prostate cancer–specific survival.[
A GWAS of Swedish men diagnosed with prostate cancer found a genetic variant at the AOX1 locus, which was significantly associated with survival.[
References:
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 2.[
Men with metastatic castration-resistant prostate cancer were recommended to undergo genetic testing for BRCA1/BRCA2, DNA MMR genes, and ATM.[
| Philadelphia Prostate Cancer Consensus Conference (Giri et al. 2020)a[ |
Genetic/Familial High-Risk Assessment: Breast, Ovarian, and Pancreatic (Version 1.2023)b[ |
NCCN Prostate Cancer (Version 3.2022)c[ |
European Advanced Prostate Cancer Consensus Conference (Gillessen et al. 2017[ |
---|---|---|---|---|
dMMR = mismatch repair deficient; FDR = first-degree relative; HBOC = hereditary breast and ovarian cancer; MSI = microsatellite instability; NCCN = National Comprehensive Cancer Network; SDR= second-degree relative; TDR= third-degree relative. | ||||
a Giri et al.: Specific genes to test includeBRCA1/BRCA2, DNA MMR genes,ATM, andHOXB13depending on various testing indications. | ||||
b NCCN Genetic/Familial High-Risk Assessment: Breast, Ovarian, and Pancreatic guidelines state that prostate cancer risk management is indicated forBRCA1andBRCA2carriers, but evidence for risk management is insufficient for other genes. | ||||
c NCCN Prostate Cancer guidelines specify that germline multigene testing includes at least the following genes:BRCA1,BRCA2,ATM,PALB2,CHEK2,MLH1,MSH2,MSH6, andPMS2. Including additional genes may be appropriate based on clinical context. | ||||
d Gillessen 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, or metastatic prostate cancer at any age | Men 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 ≥2 FDRs, SDRs, or TDRs (on the same side of the family) with breast cancer or prostate cancer (any grade) at any age | Men 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 anFDRwho 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 from prostate cancer | Men 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 andAshkenazi Jewishancestry | Men with prostate cancer and Ashkenazi Jewish ancestry | Men 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, orEPCAM | ||||
Clinical/Pathological Features | Men with metastatic prostate cancer | Men with metastatic prostate cancer | Men with metastatic prostate cancer | Men with newly diagnosed metastatic prostate cancer (62% of panel voted in favor ofgenetic counseling /testing in a minority of selected patients)[ |
Men with stage T3a or higher prostate cancer | Men with high- or very-high-risk prostate cancer | Men with high- or very-high-risk prostate cancer | ||
Men with prostate cancer that has intraductal/ductal histology | 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 who have intermediate-risk prostate cancer with intraductal/cribriform histology | ||
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 intestinal | Men 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, especiallyBRCA2,BRCA1,ATM, and DNA mismatch repair genes | Men 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 forpathogenic variantsin homologous recombination genes in men with metastatic disease; consider tumor testing in men with regional prostate cancer | |
RecommendMSI -high or dMMR tumor testing in men with metastatic castration-resistant prostate cancer; consider testing in men with regional or castration-naïve prostate cancer |
Multigene (Panel) Testing in Prostate Cancer
Since next-generation sequencing (NGS) has become readily available and patent restrictions have been eliminated, several clinical laboratories now offer multigene panel testing at a cost that is comparable to that of single-gene testing. However, variants of uncertain significance can be found. These results should be viewed with caution, since their clinical significance is unknown. 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. 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.[
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.[
Genetic Testing for Prostate Cancer Risk Assessment
Genetic testing for pathogenic variants in prostate cancer risk genes is now available. This can identify men at increased prostate cancer risk. Research from selected cohorts has reported that prostate cancer risk is elevated in men with pathogenic variants in the BRCA1 gene, the BRCA2 gene, and on a smaller scale, the MMR genes. In addition, pathogenic variants in HOXB13 account for a small proportion of hereditary prostate cancer cases. This section summarizes the evidence for the genes mentioned above and additional genes that may be on prostate cancer susceptibility panel tests.
BRCA1andBRCA2
Studies of male carriers of BRCA1[
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.
Study | Population | Prostate 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 ofBRCA1/2; OCCR = Ovarian Cancer Cluster Region; RR = relative risk; SIR = standardized incidence ratio. | |||
a Includes all cancers except breast, ovarian, and nonmelanoma skin cancers. | |||
BCLC (1999)[ |
BCLC family set that included 173BRCA2linkage – or pathogenic variant–positive families, among which there were 3,728 individuals and 333 cancersa | Not assessed | Overall: 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 164BRCA2pathogenic variant–positive families, among which there were 3,728 individuals and 333 cancersa | Not assessed | OCCR: 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 ofBRCA1pathogenic variants; 1,106 were testednoncarriers, and 8,496 were not tested | Overall: 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 inBRCA1orBRCA2in 53 cancer genetics groups across 33 countries | Occurred in 22.3% of carriers | Occurred in 25.6% of carriers |
Li et al. (2022)[ |
Cohort of 3,184BRCA1and 2,157BRCA2families from CIMBA; 34 of 1,508 men withBRCA1pathogenic variants and 71 of 1,063 men withBRCA2pathogenic variants had prostate cancer | RR, 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 (OR, 2.64; 95% CI, 2.03–3.47) than in BRCA1 carriers (OR, 1.35; 95% CI, 1.03–1.76).[
Prevalence ofBRCAfounder 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.[
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.[
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.
Study | Cases/Controls | Pathogenic 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 Israel | Controls: 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 cancer | Controls: 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 ofBRCA1pathogenic variants (OR, 2.23; 95% CI, 0.84–5.86) and carriers ofBRCA2pathogenic variants (OR, 3.18; 95% CI, 1.62–6.24) than in controls |
Controls: 1,251 AJ men with no history of cancer | Controls: 11 (0.9%) | Controls: 12 (1.0%) | ||||
Gallagher et al. (2010)[ |
Cases: 832 AJ men diagnosed with localized prostate cancer between 1988 and 2007 | Noncarriers: 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) | TheBRCA15382insC 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 ofBRCA2pathogenic variants (85%) than in noncarriers (57%);P = .0002. Carriers ofBRCA1/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 cancer | Controls: 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.
Study | Cases/Controls | Pathogenic 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 assessed | Cases: 999del5 (2.7%) | Not assessed | 999del5: RR, 2.5 (95% CI, 0.49–18.4) | |
Controls: 499 randomly selected DNA samples from the Icelandic National Diet Survey | Controls: (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 age | Not assessed | Not assessed | SIR, 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 assessed | AnyBRCA1pathogenic variant: OR, 0.9 (95% CI, 0.4–1.8) | Not assessed | Prostate 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 y | Controls: 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).[
Several case series have also explored the role of BRCA1 and BRCA2 pathogenic variants and prostate cancer risk.
Study | Population | Pathogenic 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. | ||||||
a Estimate calculated using RR data in UK general population. | ||||||
b Risks 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 history | Not assessed | 2 (0.69%) | Not assessed | RR, 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 y | Not assessed | 0 (0%) | Not assessed | Not assessed | 31 nonsynonymous variations were identified; no truncating or pathogenic variants were detected |
Tryggvadóttir et al. (2007)[ |
527 men diagnosed with prostate cancer between 1955 and 2004 | Not assessed | 30/527 (5.7%) carried the Icelandic founder pathogenic variant 999del5 | Not assessed | Not assessed | TheBRCA2999del5 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 Study | Not assessed | Overall: 19/1,832 (1.03%) | Not assessed | RR, 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 cancer | All cases: 4/886 (0.45%) | Not assessed | RR, 3.75a(95% CI, 1.02–9.6) | Not assessed | Quality-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 withBRCA1(n = 376) orBRCA2(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 diagnoses | Confirmed pathogenic variant: 16/376 | Confirmed pathogenic variant: 26/447 | SIR, 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 forBRCA1; 27% (95% CI, 17%–41%) by age 75 y and 60% (95% CI, 43%–78%) by age 85 y forBRCA2 |
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 ofBRCApathogenic 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.
Study | Cases / Controls | Gleason Scorea | PSAa | Tumor Stage or Gradea | Comments |
---|---|---|---|---|---|
AJ = Ashkenazi Jewish; CI = confidence interval; HR = hazard ratio; OR = odds ratio; PSA = prostate-specific antigen; UK = United Kingdom. | |||||
a Measures of prostate cancer aggressiveness. | |||||
Tryggvadóttir et al. (2007)[ |
Cases: 30 men diagnosed with prostate cancer who were carriers ofBRCA2999del5 founder pathogenic variants | Gleason score 7–10: | Not assessed | Stage 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 theBRCA2999del5 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 assessed | Not 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)[ |
Cases: 21 men diagnosed with prostate cancer who harbored aBRCA2 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 series | Not assessed | PSA ≥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 cancer | Grade 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 ofBRCA1pathogenic variants and 20 carriers ofBRCA2pathogenic variants | Gleason score 7–10: | Not assessed | Not assessed | TheBRCA15382insC founder pathogenic variant was not tested in this series |
Controls: 454 AJ men with no history of cancer | —BRCA26174delT 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 ofBRCA2pathogenic variants were more likely to have high-risk disease byD'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 noBRCApathogenic 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)[ |
Cases: 2,019 men diagnosed with prostate cancer from the UK, of whom 18 were carriers ofBRCA1pathogenic variants and 61 were carriers ofBRCA2pathogenic variants | Gleason 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 aBRCApathogenic 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 wereBRCA1/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)[ |
Cases: 4,187 men who underwent prostate biopsy for elevated PSA or abnormal exam, including 26 men with at least oneBRCAcoding pathogenic variant (all 26 codingexonsofBRCAwere sequenced forpolymorphisms) | Gleason score 7–10: | Cases median PSA: 56.3 | Not fully assessed in cases and controls | The 12-year survival for men with aBRCA2pathogenic variant was inferior to that of men without aBRCA2pathogenic variant (61.8% vs. 94.3%;P< 10−4). Among the men with high-grade disease (Gleason 7–9), the presence of aBRCA2pathogenic 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 noBRCAcoding pathogenic variants (all 26 coding exons ofBRCAwere 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).[
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.[
BRCA1/BRCA2and 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.[
Study | Cases | Controls | Prostate Cancer–Specific Survival | Overall Survival | Comments |
---|---|---|---|---|---|
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 ofBRCA2999del5 founder pathogenic variants | 59 men with prostate cancer matched by birth and diagnosis year and confirmed not to carry theBRCA2999del5 pathogenic variant | BRCA2999del5 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 aBRCA2pathogenic 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 series | 1,587 age- and stage-matched men with prostate cancer | Not assessed | Overall survival was lower in carriers ofBRCA2pathogenic 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 ofBRCA1pathogenic variants and 20 carriers ofBRCA2pathogenic variants | 454 AJ men with no history of cancer | After adjusting for stage, PSA, Gleason score, and therapy received: | Not assessed | TheBRCA15382insC founder pathogenic variant was not tested in this series |
– Carriers ofBRCA1 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 BRCA26174delT 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 noBRCApathogenic variant found in the family | BRCA2carriers 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 controls | BRCA2carriers 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 controls | There were too fewBRCA1carriers 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 ofBRCA1pathogenic variants and 61 were carriers ofBRCA2pathogenic variants | 1,940 men who wereBRCA1/BRCA2noncarriers | Prostate 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 ofBRCA1/BRCA2pathogenic variants | 1,235 men who wereBRCA1/BRCA2noncarriers | Prostate 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 theBRCAregion
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.[
Another study from the UM-PCGP examined common genetic variation in BRCA1.[
HOXB13
Summary
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.
Background
Linkage to 17q21-22 was initially reported by the UM-PCGP from 175 pedigrees of families with hereditary prostate cancer.[
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.[
Additional studies have emerged that better define the carrier frequency and prostate cancer risk associated with the HOXB13 G84E pathogenic variant.[
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.[
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).[
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).[
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.[
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.[
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.[
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.
Mismatch repair (MMR) genes
Five genes are implicated in 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.[
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.[
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.[
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).[
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.[
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 heterozygous carriers of ATM pathogenic variants.[
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.[
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.[
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,[
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.[
EPCAM
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.[
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.[
Study | Cohort | Germline Results for Prostate Cancer | Comments | ||
---|---|---|---|---|---|
mCRPC = metastatic castration-resistant prostate cancer. | |||||
a Potential overlap of cohorts. | |||||
Robinson et al. (2015)a[ |
Whole-exomeand 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)a[ |
692 men with metastatic prostate cancer, unselected for family history; analysis focused on 20 genes involved in maintaining DNA integrity and associated withautosomal dominantcancer–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 included | 10/97 (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%) |
Genetic Testing for Prostate Cancer Precision Oncology
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.[
Study | Cohort | Germline Results | Intervention | Outcomes 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. | |||||
a This 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 progressed | 24/319 (7.5%) had DDR germline pathogenic variants: | Patientswith 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 butwithout 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 docetaxel | 8/141 (5.7%) hadBRCA2germline pathogenic variants | Patients received at least two doses of carboplatin and docetaxel | 6/8 men withBRCA2germline pathogenic variants (75%) had PSA levels that declined by 50% vs. 23/133 in men withoutBRCA2germline pathogenic variants (17%) (P< .001) | |
A small case series (n = 3) showed a response to platinum chemotherapy with biallelic inactivation ofBRCA2, defined as either biallelic somaticBRCA2pathogenic variants or a germline pathogenic variant plus a somaticBRCA2pathogenic variant[ |
|||||
Mateo et al. (2018)[ |
390 men with mPC; retrospective review | 60/390 (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 surveillance | 2.1% of patients had germline pathogenic variants inBRCA1/BRCA2/ATM | Patients were put on active surveillance | 289 patients had their PC tumor grades reclassified: 11/26 patients had pathogenic variants inBRCA1/BRCA2/ATMand 278/1,185 patients did not have a pathogenic variant inBRCA1/BRCA2/ATM(noncarriers); adjusted HR, 1.96 (95% CI, 1.004–3.84;P = .04) | |
Tumor reclassification occurred in 6/11BRCA2carriers 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 aBRCA1,BRCA2, orATMpathogenic variant, and 2.1% only had aBRCA2 pathogenic variant. Of the men whose PCs were not reclassified, 1.6% had aBRCA1,BRCA2, orATMpathogenic variant, and 0.5% only had aBRCA2 pathogenic variant. TheP value forBRCA1/BRCA2/ATMcarriers with PCs reclassified versus those without PCs reclassified was .04. TheP value forBRCA2carriers 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 inBRCA1/BRCA2/ATM; 2 men hadBRCA1pathogenic variants, 15 men hadBRCA2 pathogenic variants, and 6 men hadATMpathogenic variants | Patients received olaparib | When patients were given olaparib, PSA levels were reduced by 50% in 13/17 (76%) men withBRCA1/BRCA2pathogenic variants and in 0/6 (0%) men withATMpathogenic variants (Fisher's exact test;P = .002) | |
Patients withBRCA1/BRCA2 pathogenic variants had a median PFS of 12.3 mo, while patients withATMpathogenic 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 diagnosis | 45 men withATMgermline pathogenic variants; 45 men withBRCA2 germline pathogenic variants | Patients received various systemic therapies | No 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 withATMgermline pathogenic variants and in 12/14 men withBRCA2germline 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 enzalutamide | 22/172 (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 germlineBRCA1/BRCA2/ATMvariants 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/ATMgermline variants (P> .10) | |
—BRCA1/BRCA2/ATM: 9/172 (5.2%) | Study limitations included the following: only 9 patients withBRCA1/BRCA2/ATMpathogenic variants | ||||
Castro et al. (2019)[ |
419 men with mCRPC were enrolled when they were diagnosed with mPC | 68/419 (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 therapyor patients received a taxane as a first-line therapy and an androgen-signaling inhibitor (abiraterone or enzalutamide) as a second-line therapy | CSS betweenATM/BRCA1/BRCA2/PALB2carriers 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 inBRCA2carriers (17.4 mo vs. 33.2 mo;P = .027), andBRCA2pathogenic variants were identified as an independent prognostic factor for CSS (HR, 2.11;P = .033) | ||||
—BRCA1: 4/419 (1%) | Significant interactions betweenBRCA2status and treatment type (androgen-signaling inhibitor vs. taxane therapy) were observed (CSS-adjustedP = .014; PFS-adjustedP = .005) | ||||
—PALB2: None | CSS (24.0 mo vs. 17.0 mo) and PFS (18.9 mo vs. 8.6 mo) were greater inBRCA2carriers 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 fromMateo 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 future | Randomized, 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 inBRCA1,BRCA2, orATM | |||||
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, orRAD54L | |||||
Hussain et al. (2020)[ |
387 men with mCRPC in the PROfound study; PC progressed when taking enzalutamide, abiraterone, or both | 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 fromMateo 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 future | Patients 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 imaging | The 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 inBRCA1,BRCA2, orATM | The 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, orRAD54L | |||||
Abida et al. (2020)a[ |
115 men with mCRPC from the TRITON2 study with a deleterious somatic or germline pathogenic variant inBRCA1/BRCA2; patients had mCRPCs that progressed after treatment with one to two lines of next-generation AR-directed therapy and one taxane-based chemotherapy | 44/115 (38%) hadBRCA1/BRCA2germline 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 withBRCA1/BRCA2pathogenic variants | |
—BRCA1: 5/115 (4%) | |||||
—BRCA2: 39/115 (34%) | |||||
71/115 (62%) hadBRCA1/BRCA2somatic variants: | 63/115 men had a confirmed PSA response (54.8%), which differed by gene; however, theBRCA1group 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)a[ |
104 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 inBRCA2, 4 inATM, and 8 in other genes | Patients 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 inBRCA1/2, 57 inBRCA2, 4 inPALB2, 17 inATM, 22 in other genes (ATR,CHEK2,FANCA,MLH1,MRE11A,NBN, andRAD51C) | 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%) |
Genetic results are increasingly informing treatment and management strategies for prostate cancer. Confirmation of somatic mutations through germline testing is needed so that additional recommendations can be made regarding cancer risk for patients and families. For more information about available practice guidelines for prostate cancer genetic testing, see Table 2.
References:
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 workups 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%.[
| Philadelphia Prostate Cancer Consensus Conference (Giri et al. 2020)[ |
Genetic/Familial High-Risk Assessment: Breast, Ovarian, and Pancreatic (Version 1.2023)[ |
NCCN Prostate Cancer Early Detection (Version 1.2022)a[ |
---|---|---|---|
NCCN = National Comprehensive Cancer Network; PSA = prostate-specific antigen. | |||
a Forgermline pathogenic variantsother thanBRCA2(includingATMand Lynch syndromegenes), 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 BRCA1Carriers | Consider 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 Carriers | Recommend 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; there is insufficient evidence to make recommendations for prostate cancer management | 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 |
Screening Recommendation Source | Population | Test | Age Screening Initiated | Frequency | Comments |
---|---|---|---|---|---|
DRE = digital rectal exam; FDR = first-degree relative; NCCN = National Comprehensive Cancer Network; PSA = prostate-specific antigen; SDR =second-degree relative. | |||||
a DRE is recommended in addition to PSA test for men with hypogonadism. | |||||
b A 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 | PSA | N/A | N/A | In 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 College of Physicians (2013)[ |
African American men and men with anFDRdiagnosed with prostate cancer, especially at <65 y | PSA | ≥45 y | No clear evidence to establish screening frequency | Counseling includes information about the uncertainties, risks, and potential benefits associated with prostate cancer screening |
No clear evidence to perform PSA test more frequently than every 4 y | |||||
Men with multiple family members who were diagnosed with prostate cancer at <65 y | PSA | ≥40 y | |||
PSA level >2.5 µg/L may warrant annual screening | |||||
American Urological Association (2013)[ |
African American men and men with a strong family history of prostate cancer | PSA | >40 to <55 y | Screening is individualized based on the patient's personal preferences and an informed discussion regarding the uncertainty of benefit and associated harms | |
American Cancer Society (2019)[ |
African American men | PSA with or without DREa | ≥45 y | Screen every 1–2 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 family members who were diagnosed with prostate cancer at <65 y | PSA with or without DREa | ≥40 y | |||
NCCN Prostate Cancer Early Detection (Version 1.2022)[ |
African American men | Baseline PSA; strongly consider baseline DRE | 40–75 y | Consider screening at annual intervals rather than every other year | The 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 historyb | Baseline PSA; strongly consider baseline DRE | 40–75 y | Screen every 2–4 y if PSA level <1 ng/mL, DRE normal | Referral to a cancer genetics professional is recommended for those with a known or suspected pathogenic variant in a cancersusceptibility gene [ |
|
Screen every 1–2 y if PSA level 1–3 ng/mL, DRE normal (if done) |
Level of evidence: 5
Screening in carriers ofBRCApathogenic 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.[
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
Chemoprevention of Prostate Cancer With Finasteride and Dutasteride
The benefits, harms, and supporting data regarding the use of finasteride and dutasteride for the prevention of prostate cancer in the general population are discussed in Prostate Cancer Prevention.
References:
This section describes current approaches of assessing and counseling patients about prostate cancer susceptibility. Genetic counseling for men at increased risk of prostate cancer encompasses all of the elements of genetic counseling for other hereditary cancers. For more information, see Cancer Genetics Risk Assessment and Counseling. The components of genetic counseling include concepts of prostate cancer risk, reinforcing the importance of detailed family history, pedigree analysis to derive age-related risk, and offering participation in research studies to those individuals who have multiple affected family members.[
Prostate cancer will affect an estimated one in eight American men during their lifetimes.[
Families need to fulfill only one of these criteria to be considered to have hereditary prostate cancer. One study investigated attitudes regarding prostate cancer susceptibility among sons of men with prostate cancer.[
Risk Assessment and Analysis
Assessment of a man concerned about his inherited risk of prostate cancer should include taking a detailed family history; eliciting information regarding personal prostate cancer risk factors such as age, race, and dietary intake of fats and dairy products; documenting other medical problems; and evaluating genetics-related psychosocial issues.
Family history documentation is based on construction of a pedigree, and generally includes the following:
For more information, see the Documenting the family history section in Cancer Genetics Risk Assessment and Counseling.
Analysis of the family history generally consists of four components:
A number of studies have examined the accuracy of the family history of prostate cancer provided by men with prostate cancer. This has clinical importance when risk assessments are based on unverified family history information. In an Australian study of 154 unaffected men with a family history of prostate cancer, self-reported family history was verified from cancer registry data in 89.6% of cases.[
The personal health and risk-factor history includes, but is not limited to, the following:
The most definitive risk factors for prostate cancer are age, race, and family history.[
The psychosocial assessment in this context might include evaluation of the following:
One study found that psychological distress was greater among men attending prostate cancer screening who had a family history of the disease, particularly if they also reported an overestimation of prostate cancer risk. Psychological distress and elevated risk perception may influence adherence to cancer screening and risk management strategies. Consultation with a mental health professional may be valuable if serious psychosocial issues are identified.[
Genetic Testing
Multigene (panel) tests for variants in genes associated with prostate cancer susceptibility are currently available and are increasingly being used in the clinic. For more information, see the Multigene (Panel) Testing in Prostate Cancer section.
References:
Introduction
Research to date has included survey, focus group, and correlation studies on psychosocial issues related to prostate cancer risk. For more information about psychosocial issues associated with genetic counseling, see the Psychological Impact of Genetic Information/Test Results on the Individual section in Cancer Genetics Risk Assessment and Counseling. Genetic testing for pathogenic variants in genes associated with prostate cancer risk is now available and has the potential to identify those at increased risk of prostate cancer. Understanding the motivations of men who may consider genetic testing for inherited susceptibility to prostate cancer can help clinicians and researchers anticipate interest in testing. Further, these data may inform the nature and content of counseling strategies for men and their families, including consideration of the risks, benefits, decision-making issues, and informed consent for genetic testing.
Risk Perception
Knowledge about risk of prostate cancer is thought to be a factor influencing men's decisions to pursue prostate cancer screening and, possibly, genetic testing.[
Study Population | Sample Size | Proportion of Study Population That Accurately Reported Their Risk | Other Findings |
---|---|---|---|
FDR = first-degree relative. | |||
Unaffected men with a family history of prostate cancer[ |
120 men aged 40–72 y | 40% | |
FDRof men with prostate cancer[ |
105 men aged 40–70 y | 62% | |
Men with brothersaffectedwith prostate cancer[ |
111 men aged 33–78 y | Not available | 38% of men reported their risk of prostate cancer to be the same or less than the average man |
FDR of men with prostate cancer and a community sample[ |
56 men with an FDR with prostate cancer and 100 men without an FDR with prostate cancer all older than 40 y | 57% | 29% of men with an FDR thought that they were at the same risk as the average man, and 14% believed that they were at somewhat lower risk than average |
Study conclusions vary regarding whether first-degree relatives (FDRs) of prostate cancer patients accurately estimate their prostate cancer risk. Some studies found that men with a family history of prostate cancer considered their risk to be the same as or less than that of the average man.[
Anticipated Interest in Genetic Testing for Risk of Prostate Cancer
A number of studies summarized in Table 14 have examined participants' interest in genetic testing, if such a test were available for clinical use. Factors found to positively influence the interest in genetic testing include the following:
Findings from these studies were not consistent regarding the influence of race, education, marital status, employment status, family history, and age on interest in genetic testing. Study participants expressed concerns about confidentiality of test results among employers, insurers, and family and stigmatization; potential loss of insurability; and the cost of the test.[
Study Population | Sample Size | Percent Expressing Interest in Genetic Testing | Other Findings |
---|---|---|---|
FDR = first-degree relative; PSA = prostate-specific antigen. | |||
Prostate screening clinic participants[ |
342 men aged 40–97 y | 89% | 28% did not demonstrate an understanding of the concept of inherited predisposition to cancer |
General population; 9% with positive family history[ |
12 focus groups with a total of 90 men aged 18–70 y | All focus groups | |
African American men[ |
320 men aged 21–98 y | 87% | Most participants could not distinguish between genetic susceptibility testing and a prostate-specific antigen blood test |
Men with and without FDRs with prostate cancer[ |
126 men aged >40 y; mean age 52.6 y | 24% definitely; 50% probably | |
Swedish men with an FDR with prostate cancer[ |
110 men aged 40–72 y | 76% definitely; 18% probably | 89% definitely or probably wanted their sons to undergo genetic testing |
Sons of Swedish men with prostate cancer[ |
101 men aged 21–65 y | 90%; 100% of sons with two or three family members affected with prostate cancer | 60% expressed worry about having an increased risk of prostate cancer |
Healthy outpatient males with no history of prostate cancer[ |
400 men aged 40–69 y | 82% | |
Healthy African American males with no history of prostate cancer[ |
413 African American men aged 40–70 y | 87% | Belief in the efficacy of and intention to undergo prostate cancer screening was associated with testing interest |
Healthy Australian males with no history of prostate cancer[ |
473 adult men | 66% definitely; 26% probably | 73% reported that they felt diet could influence prostate cancer risk |
Males with prostate cancer and their unaffected male family members[ |
559 men with prostate cancer; 370 unaffected male relatives | 45% of men affected with cancer; 56% of unaffected men | In affected men, younger age and test familiarity were predictors of genetic testing interest. In unaffected men, older age, test familiarity, and a PSA test within the last 5 y were predictors of genetic testing interest |
Overall, these reports and a study that developed a conceptual model to look at factors associated with intention to undergo genetic testing [
In a sample comprised of undiagnosed men with and without a prostate cancer–affected FDR, older age and lower education levels were associated with lower levels of prostate cancer–specific distress (as measured by the 11-item Prostate Cancer Anxiety Subscale of the Memorial Anxiety Scale for Prostate Cancer); higher distress was associated with having more urinary symptoms.[
Screening for Prostate Cancer in Individuals at Increased Familial Risk
The proportion of prostate cancers attributed to hereditary causes is estimated to be 5% to 10%.[
Screening behaviors
In most cancers, the goal of improved knowledge of hereditary risk can be translated rather easily into a desired increase in adherence to approved and recommended (if not proven) screening behaviors. This complicates prostate cancer screening, because there is a lack of clear recommendations for both high-risk men and men in the general population. For more information, see the Screening section. In addition, controversy exists with regard to the value of early diagnosis of prostate cancer. This creates uncertainty for patients and providers. It also challenges the psychosocial factors related to screening behavior.
Several small studies have examined the behavioral correlates of prostate cancer screening at average and increased prostate cancer risk based on family history; these are summarized in Table 15. In general, results appear contradictory regarding whether men with a family history are more likely to be screened than those not at risk and whether the screening is appropriate for their risk status. Furthermore, most of the studies had relatively small numbers of subjects, and the criteria for screening were not uniform, making generalization difficult.
Study Population | Sample Size | Percent Undergoing Screening | Predictive Correlates for Screening Behavior |
---|---|---|---|
AAHPC = African American Hereditary Prostate Cancer Study Network; DRE = digital rectal exam; FDR = first-degree relative; NHIS = National Health Interview Survey; PSA = prostate-specific antigen. | |||
Unaffected men with at least one FDR with prostate cancer[ |
82 men (aged ≥40 y; mean age 50.5 y) | PSA: | Aged >50 y |
Annual income ≥ U.S. $40,000 | |||
50% reported PSA screening within the previous 14 mo | History of PSA screening before study enrollment | ||
Higher levels of self-efficacy and response efficacy for undergoing prostate cancer screening | |||
Sons of men with prostate cancer[ |
124 men (60 men with a history of prostate cancer aged 38–84 y, median age 59 y; 64 unaffected men aged 31–78 y, median age 55 y) | PSA: | 39.4% patient request |
— Unaffected men: 95.3% reported ever having a PSA test | |||
— Affected men: 71.7% reported ever having a PSA test before diagnosis | |||
DRE: | |||
— Unaffected men: 96.9% reported ever having a DRE | |||
— Affected men: 91.5% reported ever having a DRE before diagnosis | 35.6% physician request | ||
Both PSA and DRE: | |||
— Unaffected men: 93.8% had both procedures | |||
— Affected men: 70.0% reported having both procedures before diagnosis | |||
Unaffected men with and without an FDR with prostate cancer[ |
156 men aged ≥40 y (56 men with an FDR; 100 men without an FDR) | PSA: | Older age |
63% reported ever having a PSA test | |||
FDRs reported higher disease vulnerability and less belief in disease prevention, but this did not result in increased prostate cancer screening when compared with those without an FDR | |||
DRE: | |||
86% reported ever having a DRE | |||
Unaffected Swedish men from families with a 50% probability of carrying a pathogenic variant in a dominant prostate cancer susceptibility gene[ |
110 men aged 50–72 y | 68% of men aged ≥50 y were screened for prostate cancer | More relatives with prostate cancer |
Low score on the avoidance subscales of the Impact of Event Scale[ |
|||
Brothers or sons of men with prostate cancer[ |
136 men aged 40–70 y (72% were African American men) | PSA: | More relatives with prostate cancer |
72% reported ever having a PSA test | |||
— 73% within 1 y | Older age | ||
— 23% 1–2 y ago | |||
— 4% >2 y ago | |||
DRE: | Urinary symptoms | ||
90% reported ever having had a DRE | |||
— 60% within 1 y | |||
— 23% 1–2 y ago | 71% reported their physician had spoken to them about prostate cancer screening | ||
— 17% >2 y ago | |||
Unaffected men with and without an FDR with prostate cancer[ |
166 men aged 40–80 y (83 men with an FDR; 83 men with no family history) | PSA: | Family history of prostate cancer |
— FDR: 72% reported ever having had a PSA test | |||
— No family history: 53% reported ever having had a PSA test | Greater perceived vulnerability to developing prostate cancer | ||
French brothers or sons of men with prostate cancer[ |
420 men aged 40–70 y | PSA: | Younger age |
More relatives with prostate cancer | |||
Increased anxiety | |||
88% adhered to annual PSA screening | Married | ||
Higher education | |||
Previous history of prostate cancer screening | |||
Data from unaffected African American men participating in AAHPC and data from the 1998 and 2000 NHIS[ |
Unaffected men aged 40–69 y: | PSA: | Younger age |
AAHPC Cohort: | |||
— 45% reported ever having had a PSA test | |||
— AAHPC Cohort: 134 men | African American men in 2000 NHIS: | ||
— 65% reported ever having had a PSA test | |||
DRE: | |||
— NHIS 1998 Cohort: 5,583 men (683 African American, 4,900 White) | AAHPC Cohort: | Fewer relatives with prostate cancer | |
— 35% reported ever having had a DRE | |||
African American men in 1998 NHIS: | |||
— NHIS 2000 Cohort: 3,359 men (411 African American, 2,948 White) | — 45% reported ever having had a DRE | ||
Unaffected African American men who participated in the 2000 NHIS[ |
736 men aged ≥45 y | PSA: | Older age (≥50 y) |
Private or military health insurance | |||
48% reported ever having had a PSA test | Fair or poor health status | ||
Family history of prostate cancer |
Psychosocial outcomes of screening in individuals at increased familial risk
Concern about developing prostate cancer: Although up to 50% of men in some studies who were FDRs of prostate cancer patients expressed some concern about developing prostate cancer,[
Baseline distress levels: Among men who self-referred for free prostate cancer screening, general and prostate cancer–related distress did not differ significantly between men who were FDRs of prostate cancer patients and men who were not.[
Distress experienced during prostate cancer screening: A study measured the anxiety and general quality of life experienced by 220 men with a family history of prostate cancer while undergoing prostate cancer screening with PSA tests.[
A study in the United Kingdom assessed predictors of psychological morbidity and screening adherence in FDRs of men with prostate cancer participating in a PSA screening study. One hundred twenty-eight FDRs completed measures assessing psychological morbidity, barriers, benefits, knowledge of PSA screening, and perceived susceptibility to prostate cancer. Overall, 18 men (14%) scored above the threshold for psychiatric morbidity, consistent with normal population ranges. Cancer worry was positively associated with health anxiety, perceived risk, and subjective stress. However, psychological morbidity did not predict PSA screening adherence. Only past screening behavior was found to be associated with PSA screening adherence.[
References:
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.
Editorial changes were made to this summary.
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.
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:
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:
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 <MM/DD/YYYY>. Available at: https://www.cancer.gov/types/prostate/hp/prostate-genetics-pdq. Accessed <MM/DD/YYYY>. [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.
Last Revised: 2023-03-17
This information does not replace the advice of a doctor. Healthwise, Incorporated, disclaims any warranty or liability for your use of this information. Your use of this information means that you agree to the
Healthwise, Healthwise for every health decision, and the Healthwise logo are trademarks of Healthwise, Incorporated.
Los planes individuales y familiares de seguro médico y dental están asegurados por Cigna Health and Life Insurance Company (CHLIC), Cigna HealthCare of Arizona, Inc., Cigna HealthCare of Illinois, Inc., Cigna HealthCare of Georgia, Inc., Cigna HealthCare of North Carolina, Inc., Cigna HealthCare of South Carolina, Inc. y Cigna HealthCare of Texas, Inc. Los planes de beneficios de salud y de seguro de salud de grupo están asegurados o administrados por CHLIC, Connecticut General Life Insurance Company (CGLIC) o sus afiliadas (puedes ver
Todas las pólizas de seguros y los planes de beneficios de grupo contienen exclusiones y limitaciones. Para conocer la disponibilidad, los costos y detalles completos de la cobertura, comunícate con un agente autorizado o con un representante de ventas de Cigna. Este sitio web no está dirigido a los residentes de New Mexico.