Abstract
The vast majority of new prostate cancer diagnoses are low-grade tumors that are monitored by active surveillance rather than undergoing immediate treatment. However, a subset of men will progress to advanced prostate cancer which may result in lethality, and these men are likely to benefit from early intervention to prevent or delay such progression. For this high-risk group, which includes aged men, men of African descent, and those with a hereditary predisposition to prostate cancer, informed risk stratification can be the cornerstone of clinical decision making and treatment intervention. In this review, we discuss the importance of a precision intervention approach that considers the cumulative risk for any given patient or population to develop prostate cancer or to progress to lethal disease, with particular focus on the interplay of major determinants of high-risk disease.
Keywords: Risk factors, genetic variants, mitochondrial DNA, active surveillance, precision prevention and treatment
1. Introduction
Prostate cancer is the most frequently diagnosed cancer in more than half of the world’s nations, accounting for more than 7% of the global cancer burden, exceeded only by lung and colorectal cancers [1–3]. In the United States, the incidence of prostate cancer has risen sharply over the last two decades, due in part to the increase in the ageing population, since ageing is a major risk factor for prostate cancer (see below). Given the increasing life-expectancy, particularly in Western countries but also worldwide, the incidence of prostate cancer is expected to further increase in the future.
Another major factor contributing to the increased incidence of prostate cancer is the wide-spread screening for prostate-specific antigen (PSA) [4], which has resulted in a substantial increase in prostate cancer detection, albeit a significant level of overdiagnoses [5–7]. Indeed, high PSA can occur in benign as well as malignant contexts, and is not necessarily indicative of lethal disease. Many prostate cancers are relatively indolent and unlikely to progress to lethal disease within the patient’s lifetime. Furthermore, overdiagnosis can lead to overtreatment of non-lethal tumors via surgery or radiation therapy, which may result in significant morbidities, including persistent urinary, sexual and/or bowel problems. It has therefore become common practice for men who are diagnosed with low-grade, localized prostate cancer to be monitored by active surveillance, which involves close observation without immediate intervention unless there are signs of disease progression [8, 9]. Notably, the implementation of targeted biopsies via imaging-guided modalities, such as multi-parametric MRI, has improved detection of prostate cancer and thereby increased confidence in accurate monitoring of disease progression [10–12].
For patients that exhibit signs of progression during active surveillance, radical prostatectomy or radiotherapy can often be curative [13]. However, a significant percentage will fail local treatment and further progress to aggressive prostate cancer, which is likely to metastasize quickly and can be lethal over time. The majority of recent investigations define aggressive prostate cancer as any tumor that would require treatment according to National Comprehensive Cancer Network (NCCN) guidelines and would therefore not be eligible for active surveillance [14]. Currently, it is difficult to accurately identify men at greatest risk of progression to more aggressive and potentially lethal disease when it is early enough to implement appropriate interventive measures that would improve their survival chances. Thus, the real challenge of active surveillance is achieving the appropriate balance between minimizing the adverse consequences of overtreatment, while maximizing the opportunity to identify potentially lethal tumors at the earliest possible point to ensure their most effective treatment.
Considering the wide range of clinical outcomes from indolence to lethality, a “one size fits all” approach is unlikely to be effective for preventive management of prostate cancer. Rather, it would be beneficial to implement a “precision intervention” approach wherein for any given patient, a disease management plan would be adopted that is tailored to their individual risk, with the goal of following a conservative monitoring plan for those at lower risk, and more active and earlier intervention for those at moderate or higher risk for aggressive prostate cancer [15, 16]. While this may seem to be an obvious approach, in practice it has been more difficult to achieve since it requires an accurate understanding of potential risk factors and how their cumulative interactions impact the chances of disease onset or progression for any given individual or population. In this review, we discuss the current status of our understanding of the major factors associated with prostate cancer risk (Figure 1), and consider how these can be used in a precision intervention paradigm to more accurately anticipate patient outcomes, and help stratify men for more effective management of prostate cancer.
Figure 1. Prostate cancer progression and risk factors.

Progression pathway for prostate cancer. The stages of prostate cancer development and progression are shown, together with the risk factors likely to be significant at each stage and their suggested contribution to increasing prostate cancer risk (from low to high risk). Specific major genetic alterations are depicted for the risk of genetic predisposition.
2.0. Factors associating with prostate cancer risk
We use the term “risk factors” to refer to anything that may increase the probability of prostate cancer development or progression to aggressive disease. Throughout this review we distinguish risk that is associated with the development or onset of prostate cancer, or its progression to aggressive disease, and we consider how a knowledge of key risk factors can be used for precision interventions. Many studies have identified factors associated with prostate cancer risk, of which race, age, and genetic determinants are among the most significant [2, 17, 18]. However, despite an abundance of basic, translational, and clinical studies that have expanded our understanding of the processes underlying prostate cancer initiation and progression and have improved its early detection and treatment, significant gaps remain in meaningful risk assessment that will allow for more accurate stratification for given individuals or populations.
2.1. Age
Age constitutes the most significant risk factor associated with prostate cancer. Indeed, the incidence of prostate cancer displays a steep association with age, much more so than other epithelial cancers. Less than 25% of prostate cancers are detected before the age of 65 while more than 60% occur in men 70 years or older, and more than 80% in men 80 years or older [2, 17, 18], suggesting that the risk of developing prostate cancer increases exponentially with age. Older men are also more likely to be diagnosed with high-grade prostate cancer and tend to have worse outcome [19, 20]. It is conceivable that such outcome differences reflect their health at the time of diagnosis; in particular, men are more likely to undergo treatment if they are otherwise healthy, whereas those with co-morbidities that are common in aged men may necessitate more conservative management. These observations suggest that although age is a crucial risk factor for prostate cancer, it should not be the sole indicator for guiding treatment decisions and highlights the importance of taking into account associated risk factors, such as those discussed below.
2.2. Oxidative Stress and Inflammation
Aberrant oxidative stress is another key risk factor for prostate cancer, and particularly associated with aging [21, 22]. For reasons that are largely unknown, the prostate is highly susceptible to oxidative stress, which is a hallmark of aggressive, lethal prostate cancer and manifested as an excess of reactive oxygen species (ROS) [23]. The effects of oxidative stress are potentiated by aging in several respects. On the one hand, accumulation of excess ROS in the form of hydroxyl radicals, superoxides or peroxides can accelerate aging of prostatic epithelium [24]. On the other hand, antioxidant defense systems are often compromised in elderly individuals making them more susceptible to oxidative stress conditions that produce excess ROS, which in turn can trigger and accelerate tumorigenesis [22, 25]. Increased ROS production and oxidative stress can be caused by various factors, including metabolic alterations, androgen receptor activation and mutation-induced mitochondrial dysfunctions [22].
Another major source of age-associated oxidative stress is inflammation. The presence of chronic inflammation known as prostatitis, is prevalent in benign prostatic intraepithelial neoplasia in elderly men [26–28]. This condition is often accompanied by immune cell infiltrate into the prostate, the presence of which has been correlated with increased cancer risk and poor prognosis [29–33]. Notably, an increase in activated inflammatory cells has been associated with increased ROS levels in the prostate, which may in turn promote somatic mutations and mitochondrial dysfunction [22, 34]. These changes can promote tissue damage resulting in impaired differentiation and compensatory epithelial cell proliferation, therefore potentiating prostate carcinogenesis [35, 36]. Overall, these observations suggest that the aging prostate acquires an inflammatory microenvironment that promotes aberrant oxidative stress, which in turn increases the risk of developing clinically-relevant prostate cancer.
Although oxidative stress and inflammation have been associated with prostate cancer progression, in practice, chemopreventive approaches using agents that target these processes have not proven to be promising [37–39]. Nonetheless, while the mass implementation of such agents may not be effective or may even have opposing effects [40], their individualized use in certain contexts may be beneficial. For example, individuals with elevated levels of oxidative stress or reduced expression of the prostate-specific homeobox gene, NKX3.1, may benefit from anti-oxidant interventions in a precision prevention paradigm (see for example [41]).
2.3. Race
Epidemiological data suggest that racial differences contribute significantly to increased risk for prostate cancer onset and aggressiveness. In the United States, for example, men of African descent experience an approximately 60% higher incidence of prostate cancer and a 144% higher death rate compared to caucasian Americans [18, 42], while men of African descent from Caribbean and sub-Saharan African countries exhibit high rates of prostate cancer mortality and have higher tumor stage at diagnosis, respectively, compared to other racial groups [43]. In the United States, African American men are often diagnosed with more advanced and lethal prostate cancer compared to other racial groups [44, 45]. Although most prostate cancer patients are diagnosed at 55 years of age or older, the average age of diagnosis for African Americans is 40 years, and they are also about two times more likely to die from prostate cancer than Caucasian men [46, 47].
Exogenous factors that have been implicated as contributing to these observed racial disparities include differences in access to health care that are associated with demographic or socioeconomic factors [48–50]. In particular, epidemiologic evidence shows that African Americans residing in racially segregated areas with persistent poverty, stagnant social mobility and poor healthcare access, have worse outcomes than their Caucasian counterparts [51]. However, it is unlikely that disparities in prostate cancer outcome are solely due to exogenous factors [52]. Recent studies have identified transcriptomic and genomic differences associated with differential susceptibility men of African ancestry to prostate cancer [47, 52–54]. In particular, genome-wide association studies (GWAS) have identified susceptibility loci that are independently linked with prostate cancer risk (see below and [55]). In our view, the potential interplay between socioeconomic and genetic factors that may contribute to racial differences in prostate cancer risk is an important area for future investigation.
3.0. Genetic risk in prostate cancer onset
Another significant risk factor is genetic predisposition; notably, men with a familial history of prostate cancer tend to have earlier onset and more aggressive disease [56–58]. To clarify, familial prostate cancer refers to disease that occurs in with a family history of the disease but without an evident genetic basis, whereas hereditary prostate cancer refers to disease that occurs in individuals having germline pathogenic variants that are associated with higher risk for disease onset or progression [59–61]. Hereditary prostate cancer accounts for 10–15% of all cases, and is generally defined as occurring in three successive generations, or at least two first-degree relatives, each diagnosed with early-onset disease [17, 62]. Although individual germline variants may influence onset or mortality, prostate cancer is more likely polygenic, shaped by the cumulative effect of multiple genetic variants each with a relatively small effect. In particular, GWAS have identified numerous single nucleotide polymorphisms (SNPs) that cumulatively associate with increased incidence or lethality of prostate cancer [55, 63–65], while linkage analyses have identified rare variants of known genes that associate with a greater lifetime risk of developing prostate cancer [66–69].
3.1. Single nuclear polymorphisms associated with prostate cancer risk
Supporting the notion that genetic predisposition for prostate cancer is driven by the cumulative interactions of multiple alleles, each associating with low to modest risk, several SNPs have been identified that co-occur in hereditary prostate cancer [70–74]. Initial GWAS studies identified 16 SNPs that strongly associate with a family history of prostate cancer [74], which paved the way for subsequent, more comprehensive GWAS that have led to the identification of more than 170 loci linked to prostate cancer susceptibility [64, 75]. Some of these SNPs occur in known genes, including BRCA1, BRCA2, ELAC2 (locus HPC2), RNase L, and NKX3.1, which have been further associated with increased incidence of aggressive prostate cancer (see below and [76–81]). However, the majority of SNPs are located in non-coding regions of the genome and do not have an easily discernable mechanism by which they might contribute to prostate cancer incidence or progression. For example, SNPs on chromosomes 15q13 and 17p12, which are not associated with any known genes, occur at significantly higher frequency in men with high grade invasive prostate cancer compared to those with lower grade or indolent disease [82, 83]. Another SNP (rs11672691) of as yet unknown function, has been shown to significantly associate with increased risk for prostate cancer mortality [84]. In-depth functional characterization of these genetic variants is warranted to better understand whether or how they contribute to increased susceptibility for aggressive prostate cancer.
Notably, certain SNPs are present at a higher frequency in African American men as compared to men of other races. For example, several germline variants of BRCA1 and BRCA2 have been observed in African American men more frequently than in Caucasian American men [85], associating with disease aggressiveness (see below). Additionally, a novel SNP (rs7918885), localized on chromosomal region 10p14 within an intron of a long non-coding region of the GATA3 gene, has been detected exclusively in African American men [86, 87], which is notable since loss of function of GATA3 has been functionally implicated in prostate cancer progression [88]. Therefore, analyses of SNPs may inform on differences in the incidence of aggressive prostate cancer observed in African American men (Figure 1).
A promising application of these SNP analyses has been the introduction of a polygenic risk score (PRS) or a polygenic hazard score (PHS), which take into account the total number of genetic variants in a given individual to assess his risk of prostate cancer development or progression [89–92]. The PRS of prostate cancer patients at the time of diagnosis has been associated with their respective survival outcomes [93], and in combination with PSA levels, may help to identify men who are at high risk for developing lethal prostate cancer, thereby further informing PSA screening [70, 94]. Of note, PRS assessment has proven to be effective for predicting the risk for developing aggressive prostate cancer in men who carry a germline variant of BRCA1 [95]. One recent study also identified an association between PRS and metastatic prostate cancer at the time of diagnosis [90], likely due to the inclusion of SNPs known to associated with metastatic prostate cancer risk [96, 97]. Despite progress in establishing PRS models to predict disease progression, the performance of PRS for predicting metastatic disease remains limited compared to overall prostate cancer incidence or mortality.
The PHS, another iteration of an SNP-based polygenic risk calculator, has proven to be a significant predictor of aggressive prostate cancer at the age of diagnosis [92]. More specifically, in the Prostate testing for Cancer and Treatment (ProtecT) clinical trial, men with a high PHS exhibited a 3-fold greater risk of developing higher grade prostate cancer compared to those with average PHS. Thus, in combination with assessment of other risk factors, models based on analyses of SNPs may facilitate earlier and more accurate detection of patients at high-risk for developing prostate cancer and for identifying those at greater risk of progression to lethal disease.
With that in mind, most polygenic risk assessment models were constructed using data from Caucasian men raising the concern that they may be under-representative of genetic variants that are more relevant for men of other races [93]. Comprehensive multi-ethnic studies revealed that modified PRS or PHS can be applied to accurately assess the risk of prostate cancer onset in African American men [91, 93, 98, 99]. In the course of these studies, three new SNPs on chromosome 8 were identified that associate with increased risk of prostate cancer onset almost exclusively in men of African ancestry [99]. Interestingly, one of them (SNP rs76229939) is located in the protein-coding region of the PCAT2 gene and the other two (rs74421890 and rs5013678) are in the non-coding region of the PRNCR1 gene, neither of which have as yet well-known functions in prostate cancer. However, no polygenic risk or hazard score has been shown to accurately discriminate men at risk of aggressive prostate cancer from those at risk of indolent prostate cancer within African American populations, warranting further investigations to improve SNP-based risk stratification for aggressive prostate cancer.
3.2. Germline variants associated with prostate cancer risk
Linkage studies have identified several germline variants that cluster in relatively predictable heritance patterns in kindreds at high-risk for prostate cancer [100–102]. Following these early studies, several other genes have subsequently been shown to be associated with prostate cancer risk. In particular, variants of BRCA1 and BRCA2, which are tumor suppressor genes that have distinct roles in DNA repair [103], have been shown to associate with high risk of prostate cancer, especially in men under the age of 65 [104]. BRCA1 and BRCA2 were originally identified on the basis of their hereditary susceptibility to breast and ovarian cancer [105, 106]. In fact, the first evidence of the association of their germline mutations with prostate cancer emerged from the Breast Cancer Linkage Consortium, which reported an increased incidence of prostate cancer in kindreds with a high incidence of breast and ovarian cancers [107].
Men with pathogenic variants of BRCA1 or BRCA2 tend to present with prostate cancer having higher grade and stage at diagnosis, including local metastases, and tend to have worse outcome after prostatectomy or radiation therapy compared to non-carriers [108, 109]; however, men with BRCA2 variants appear to be at a higher risk for lethal prostate cancer than those with BRCA1 variants [110, 111]. Recent interim data from the IMPACT study, a clinical trial in which annual PSA screening is used to determine the prostate cancer incidence in men who harbor these germline variants, revealed that men with alterations of BRCA2 but not BRCA1 are more likely be diagnosed with clinically-significant prostate cancer [112]. Interestingly, a rare protein-truncating variant in BRCA2 was recently identified in African American men diagnosed with early-onset prostate cancer, providing further support that BRCA2 variants and, consequently, DNA damage repair gene defects, may be are associated with the more aggressive prostate cancer phenotypes observed in men of African descent [113].
In addition, a substantial proportion of men undergoing treatment for locally advanced prostate cancer harbor potentially pathogenic variants of TP53 [114]. While somatic inactivation of TP53 is a late event during prostate cancer progression [115, 116], emerging evidence indicates that TP53 alterations occur at a relatively high frequency in primary and, especially, in castration-naïve metastatic prostate cancer [114, 117, 118]. Moreover, a recent study revealed that germline variants and somatic mutations of TP53 were the most prevalent alterations in approximately 25% of the primary tumors of patients who later developed aggressive metastatic disease [119]. The relative risk of carrying germline TP53 pathogenic variants is comparable to that of BRCA2 mutations, predisposing men to lethal prostate cancer: five germline missense TP53 variants, 158R>H, 181R>H, 252R>Q, 283R>C and 337R>H, have been reported to be significantly enriched in prostate cancer cohorts compared to men with benign prostate [120].
Another key gene associated with prostate cancer susceptibility is the HOXB13 homeobox gene that harbors the germline mutation 84G<E, or G84E as reported in literature, which has been associated with a 3 to 4-fold increase risk of prostate cancer onset [67, 121]. This germline variant was first identified in four prostate cancer families after sequencing over 200 genes on chromosome 17 [67], and subsequent studies have shown its strong association with hereditary prostate cancer [67, 122]. Men harboring this variant have an almost 20-fold greater probability of developing prostate cancer compared to the general population [67, 123], with subsequent meta-analyses establishing a strong association between HOXB13 G84E and early onset prostate cancer [124]. Follow-up studies have identified additional HOXB13 variants that have more modest association with increased risk of advanced prostate cancer [123]. Importantly, men carrying the G84E variant have significantly higher PSA levels at diagnosis and higher Gleason scores compared to non-carriers, suggesting a possible association between the HOXB13 G84E variant and aggressive prostate cancer [125]. On the other hand, one study showed that the G48E variant was associated with a 2.9-fold increase for developing prostate cancer, but not correlated with clinical stage, Gleason score or PSA levels [126], suggesting that HOXB13 G84E may identify men at high risk of developing prostate cancer but not those at greater risk of progression to aggressive disease. Two additional HOXB13 variants (R229G and G216C), harboring missense mutations, were exclusively detected in African American, and linked to higher stage and more aggressive prostate cancer [67]. Notably, HOXB13 is known to function in prostate development and contributes to androgen-independent prostate cancer [127, 128]; however, the mechanisms by which its germline variants, and specifically G84E, may promote prostate carcinogenesis remains unknown.
Another homeobox gene associated with prostate cancer risk is NKX3.1, a prostate-specific homeobox gene that regulates prostatic differentiation and whose reduced expression promotes prostate cancer initiation [129]. Linkage and GWAS analyses have identified several germline variants of NKX3.1 [78, 80, 130]. Of these, the rs11781886 variant, located 15 bases upstream of the start codon, has been shown to associate with increased prostate cancer susceptibility potentially by reducing NKX3.1 expression. Retrospective analyses in the Selenium and Vitamin E Cancer Prevention Trial (SELECT) also revealed a role for the rs11781886 variant in modifying prostate cancer risk [77]. Another germline NKX3.1 variant (454A>G), encodes a variant protein, NKX3.1 T164A, which has a mutation in the homeodomain that results in impaired DNA binding activity [78]. A second NKX3.1 variant, 154C>T encoding for the NKX3.1 R52C variant protein, has been associated with increased risk of higher grade prostate cancer [80]. Although none of these variants appear to be associated with racial disparities [80], downregulation of NKX3.1 expression levels has been linked to increased aggressiveness and mortality in African American men [47, 131]. Recently we reported a novel function for NKX3.1 in mitochondria in response to oxidative stress [132] and found that, unlike wild-type NKX3.1, neither the NKX3.1 T164A nor the NKX3.1 R52C variant proteins were capable of protecting against oxidative stress or suppression of prostate cancer initiation.
4.0. Genetic risk in metastatic prostate cancer
In general, prostate cancer has a low mortality rate since most patients are diagnosed at a localized or locoregional stage. Men diagnosed with localized disease that progresses to more aggressive disease undergo surgery or radiation treatment, which is often curative. However, treatment failure is common and is manifested as rising PSA levels, termed biochemical recurrence (BCR) [133]. Additionally, some men will present at diagnosis with metastatic disease, which is called metastatic hormone-sensitive (mHSPC). For the past 65 years, androgen deprivation therapy (ADT) has been on the forefront of treatment for men that fail primary interventions or those with mHSPC [14]. While most men initially respond to ADT, nearly all will eventually develop castration resistance. New treatment options for men with metastatic castration-resistant prostate cancer (mCRPC) have improved clinical outcome [134]; however, there have been no significant advancements in the treatment of mHSPC. This necessitates a clear need to improve on current risk-stratification guidelines to better define treatment paradigms for patients with mCRPC and mHSPC.
Several studies have attempted to address this issue by analyzing the genomic profiles of primary tumor biopsies from lethal prostate cancers, either presenting as mCRPC or mHSPC, to identify somatic alterations that are associated with disease progression [119, 135–138]. With regards to mCRPC, somatic alterations of MYC, CCND1, and PRKDC copy number gains as well as TP53 and ZNRF3 loss have been shown to have prognostic value for predicting metastatic relapse of localized disease [138]. Interestingly, somatic loss of NKX3.1 was the most frequent genomic alternation event in localized prostate cancer and also present in around 70% of mCRPC tumors. Another study revealed that alterations in DNA damage repair pathway genes were present from initial diagnosis to metastatic relapse, with 11 truncating mutations in BRCA2, CDK12, ATM, MSH6, and PALB2 present in both primary mCRPC biopsies [119]. Interestingly, in the same cohort, two patients harbored pathogenic germline variants of BRCA2 that manifested as biallelic BRCA2 loss, while the remaining deleterious DNA damage repair gene mutations were only detected in somatic DNA. These data support the use of genomic profiling for diagnostic purposes in prostate cancer biopsies, focusing on DNA repair gene defects [139], as the prevalence BRCA2 alterations (both somatic and germline) in primary prostate tumors was similar to that reported for mCRPC.
Given that mHSPC tumors share common genomic alterations with mCRPC [139–141], it is reasonable to assume that patients with mHSPC could also benefit from molecular-guided patient stratification. In that respect, germline or/and somatic genomic alterations in DNA damage repair genes occur in 18% of mHSPC tumors, with BRCA2 variants occupying the top spot as the most altered gene at 7% [135]. In addition, a small number of mHSPC patients were shown to harbor somatic TP53, PTEN, RB1 and CTNNB1 mutations associated with a higher risk of progression to lethal prostate cancer [118]. A recent study demonstrated that men with high-volume mHSPC and a higher frequency of NOTCH and cell-cycle pathway genes exhibited shorter times to mCRPC and reduced survival [137]. Therefore, the established association between certain molecular signatures, specific clinical phenotypes and outcomes in mCRPC may also open new avenues for similar risk stratification approaches in mHSPC patients.
5.0. Mitochondrial genetic variation associated with cancer risk
Although less well studied than variants of nuclear DNA, alterations of mitochondrial DNA (mtDNA) have been associated with prostate cancer risk [142, 143], particularly in African American men [143]. The absence of genomic recombination of mtDNA and its strict maternal inheritance leads to accumulation of heritable mtDNA mutations that can define population-specific mtDNA genomic profiles, which are known as haplogroups. Based on correlation analyses between population haplogroups and prostate cancer risk, African American men have been found to be at highest risk [144]. Furthermore, reduced mtDNA content as well as increased mtDNA mutational load have been associated with higher grade prostate cancer and poorer prognosis in men of African descent [145, 146].
An association between the total number of somatic mtDNA variants and prostate cancer stage at diagnosis is evidenced by the correlation between higher mtDNA mutational load and risk of biochemical relapse after surgery [142, 146, 147]. Meta-analyses of these studies revealed that increased mtDNA mutational load is correlated with higher Gleason score, elevated PSA levels and age at diagnosis [148], suggesting that total mtDNA mutational load, rather than individual mutations may be a stronger indicator of prostate cancer risk. However, a recent study based on sequencing mtDNA from 384 individuals with localized prostate cancer found that each individual had an average of at least one mitochondrial SNP (mtSNP) [149]. Some of these individual mtSNPs, found in recurring mutational foci, were predominantly present in men diagnosed with locally invasive or metastatic disease at older age. It is therefore possible that analyses of specific mtDNA alterations can inform prostate cancer risk predictions, particularly when combined with other genetic indicators of high-risk prostate cancer. For example, a SNP located in an mtDNA transcriptional control region was found to co-occur with MYC oncogenic gain or NKX3.1 loss, and such correlation was associated with adverse outcome [149], suggesting the interplay of variants of both the mitochondrial and nuclear genomes in promoting risk of disease progression.
With respect to mechanism, knowledge of the aging process and accompanying cellular dysfunction suggests an important role for disruption of mitochondrial function in age-related decline and predisposition to cancer [150]. In particular, persistent oxidative stress may ultimately promote mutations in the mitochondrial genome that give rise to mitochondrial dysfunction, which can induce or aggravate prostatic tumorigenesis [132, 149, 151]. As one example, an inherited mtDNA variant in the CO1 gene (6124T>C) has been shown to alter ROS production and proliferation in patient-derived prostate cancer cells, implicating a functional role for mtDNA variants in prostate tumorigenesis [152]. Additional analyses of mutational signatures in patient-derived prostate cancer cells revealed two additional inherited mtDNA mutations (8993T>G and 6142T>C) that lead to increased oxidative stress via ROS accumulation [143]. Although a recent study identified more than 80 conserved mtDNA mutations among patients with metastatic prostate cancer, only a small percentage are predicted to impact the corresponding protein function [148]. Further investigation of the functional significance of mtDNA alterations and their association with oxidative stress and prostate tumorigenesis may identify novel pathways of aggressive prostate cancer, leading to improved patient risk assessment.
6.0. Precision prostate cancer intervention in practice
The overarching goal of precision intervention is to capitalize on the knowledge regarding risk factors associated with early onset, increased incidence, or increased tendency toward more aggressive prostate cancer to enhance patient stratification with the ultimate goal of identifying, as early as possible, those men who are at risk of developing clinically relevant prostate cancer, while minimizing unnecessary treatment for those at lower risk (Figure 2).
Figure 2. Cumulative prostate cancer risk assessment model for improved precision intervention.

A comprehensive risk assessment is employed for the population at risk for prostate cancer, which incorporates genetic risk profiling, to determine the cumulative risk for prostate cancer (low to high) for each individual or population. Of note is that genetic risk profiling includes the assessment of both nuclear (e.g., PRS, HOXB13 G84E, BRCA2, NKX3.1 154C<T) as well as mitochondrial (e.g., mtDNA mutations, mtSNPs) alterations. Patient stratification to the most effective mode of intervention based on their cumulative prostate cancer risk is depicted that highlights a precision intervention paradigm for prostate cancer.
6.1. Risk stratification for prevention of prostate cancer onset and progression
Moving towards reliance on risk-based strategies particularly for men at high risk of developing prostate cancer, including those of advanced age, of African descent or with a family history of prostate cancer, could help to inform PSA testing. For example, the Rotterdam prostate cancer risk calculator incorporates available pre-biopsy information, such as ultrasound volume and digital rectal examination results that complement PSA testing, resulting in a significant reduction of unnecessary biopsies while improving detection of clinically important prostate cancer [153]. However, this approach does not consider individual risk and may benefit from the incorporation of genetic screening to assess nuclear and mitochondrial variants to identify individuals at a higher risk for developing prostate cancer.
Indeed, as emerging evidence supports the importance of the germline genomic landscape for development or progression of prostate cancer, significant advancements have been made to establish guidelines for the genetic risk assessment of prostate cancer patients [154]. The most notable update has been the implementation of genetic testing for all men with a strong family history of prostate cancer and high-risk germline alterations, such as pathogenic variants BRCA1 or BRCA2 [155]. Moreover, certain histopathological features (i.e. intraductal carcinoma) have been associated with pathogenic germline DNA repair gene alterations and therefore used as an indicator for genetic testing [156]. Recent studies indicate that non-hereditary genetic testing can be successfully applied to prostate biopsies from prostate cancer patients to predict absence of adverse pathologic features [157–159]. In that regard, histopathological changes associated with increased oxidative stress, which is also associated with higher risk, have been linked to reduced expression levels of NKX3.1 [132]. Therefore, a key opportunity is to use these approaches to better define the characteristics that make patients eligible for active surveillance [160], and in conjugation with existing criteria such as low grade tumor on biopsy and low PSA levels, guide appropriate prevention interventions.
Chemopreventive strategies have gained focus in an effort to decrease the incidence morbidity and mortality of prostate cancer, as well as treatment-related complications [161–163]. However, this approach has not been promising, as exemplified by a large-scale study of finasteride, an inhibitor of 5-α-reductase type 2 (5-ARI) that halts the conversion of testosterone to dihydrotestosterone. The Prostate Cancer Prevention Trial (PCPT) revealed that finasteride intake reduced prostate cancer prevalence by almost 30% over 7 years [162]. However, follow-up analyses did not show any clear indication that finasteride could improve the long-term (15-year) survival of prostate cancer patients, while its administration was associated with an increased occurrence of side effects, such as urinary and bowel incontinence, erectile dysfunction and gynecomastia [163]. Interestingly, our co-clinical studies identified NKX3.1 expression status as a predictor of response to finasteride [41], suggesting that genetic features might help identify men most likely to benefit from such chemoprevention interventions. This also begs the question as to whether men harboring pathogenic germline NKX3.1 alterations, indicative of a potentially more aggressive prostate cancer (see above), could also have better responses to finasteride during active surveillance.
Increasingly, a plethora of agents have shown promising anti-cancer activities but their efficacy in men on active surveillance remains unclear. One such example is metformin, an anti-diabetic drug that has been shown to have anti-cancer properties and good tolerance [164, 165]. A randomized study aimed at evaluating the effect of metformin in delaying prostate cancer progression in men undergoing active surveillance is on-going Metformin Active Surveillance Trial (MAST) (NCT01864096), expected to be finalized in 2024. Given the antioxidant activity of metformin [166], one would also assume that it would be a suitable preventative intervention for prostate cancer, where oxidative stress is a prominent risk factor. Of note is that men with specific genetic alterations, such as increased mtDNA mutation load or NKX3.1 aberrations, have been shown to foster increased levels of prostatic oxidative stress and higher risk for aggressive disease (see above). Therefore, adopting a precision prevention approach of tailoring intervention approaches, like metformin, for each individual patient or population based on a cumulative prostate cancer risk that incorporates genetic risk profiling, could significantly improve prostate cancer prevention (Figure 2).
6.2. Risk stratification for diagnosis and treatment prostate cancer
Several nomograms have been created for estimation of BCR risk, such as the CAPRA-S and the Walz nomograms [167]. The most recent update of the latter provides refined estimates for BCR risk at one and two years post-radical prostatectomy based on PSA, Gleason score, disease stage and metastasis status [168], albeit with no clear correlation to prostate cancer mortality rates. In that respect, the application of non-hereditary genetic testing based on the differential expression of genes involved in distinct biological pathways of prostate tumorigenesis have been beneficial to predict disease outcome. The most widely used genetic classifier in clinical practice is the Decipher test, which aims at assessing the expression of a 22-gene panel in patients newly diagnosed with localized prostate cancer at the time of biopsy, as well as men who have undergone radical prostatectomy [159]. Decipher has been successfully applied to identify subsets of patients with histologically low risk prostate cancer at diagnosis that qualify for active surveillance [158], suggesting that molecular profiling of newly diagnosed prostate cancer can improve risk stratification and help guide treatment decisions. Additionally, OncotypeDx, a 17-gene Genomic Prostate Score (GPS) signature based on prostate cancer biopsies, was shown to be predictive of adverse prostate cancer pathology beyond conventional clinical factors [169]. Interestingly, a recent study found that the accuracy of prostate cancer outcome prediction by the presence of rare genetic variants, like HOXB13 G84E, could significantly be enhanced by the incorporation of an individual’s PRS profile [89]. We further suggest that incorporation of mitochondrial genome screening in conjugation with established nuclear genetic tests, may further improve risk assessment. Although currently there is less known about the frequency and functional relevance of mtDNA alterations, compelling evidence suggest collaboration between mitochondrial and nuclear mutational profiles in defining prostate cancer aggressiveness and outcome (see above).
To date, testosterone suppression using novel ADT therapies is the mainstay of initial treatment for patients with mHSPC [14]. The trend has been to follow clinical rather than molecular factors to aid in decision making when it comes to incorporating additional agents targeting the androgen receptor axis, such as abiraterone or enzalutamide. However, treatment intensification may be preferentially used for individuals with the more aggressive mHSPC phenotypes harboring distinct genetic alterations, such as TP53 or RB1 mutations, sparing overtreatment and toxicity for those who are less likely to benefit and eventually develop mCRPC. Although the choice for further treatment following the development of castration resistance is unclear, recent evidence suggests the need for routine genetic testing to inform sequential treatment in mCRPC patients [170]. For example, testing for germline DNA damage repair defects can help identify patients who could benefit from targeted treatments, while determining prostate cancer risk in family members. If DNA damage repair genetic alterations, such as pathogenic BRCA2 variants, are already detectable in the primary tumor, there is a strong rationale for testing synthetic lethal strategies with PARP inhibitors or platinum chemotherapy in metastatic prostate cancer, where the magnitude of benefit for patients could be larger.
7.0. Concluding remarks
Precision intervention is intended to incorporate an individuals’ risk profile, which can be defined as the culmination of several risk factors contributing to increased prostate cancer incidence and mortality (Figure 2). In particular, improvement of precision screening and intervention at different stages of prostate cancer may establish a successful connection between specific genetic factors, such high-risk germline genetic alterations, and other non-genetic factors, such as age or race. This could allow for a more accurate assessment of individual or population risk for lethal prostate cancer and fulfil the need for better stratification strategies in chemoprevention or therapeutic interventions, where men with high risk benefit the most. We believe that more comprehensive genetic screening approaches could improve the impact of current prostate cancer risk assessment approaches and counterbalance the risk of overdiagnosis and overtreatment morbidity.
Highlights:
Age, race, oxidative stress and inflammation influence prostate cancer risk
Genetic factors play a pivotal role in predicting prostate cancer onset and outcome
Nuclear and mitochondrial genetic alterations point to prostate cancer susceptibility
Integrative risk assessment may improve precision prostate cancer intervention
Acknowledgments:
Figures 1 and 2 were created with BioRender.com using an institutional license sponsored by Columbia University’s VP&S Office for Research.
Funding:
Research in the Abate-Shen laboratory is supported by grants from the National Institute of Health (NIH) CA233176, CA183929, CA265768-01 and CA173481 (to C.A.S.) and Prostate Cancer Foundation 21YOUN32 (to A.P.).
Footnotes
Declaration of competing interest:
The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper.
References
- [1].Giona S, The Epidemiology of Prostate Cancer, in: Bott SRJ, Ng KL (Eds.) Prostate Cancer, Brisbane (AU), 2021. [PubMed] [Google Scholar]
- [2].Rawla P, Epidemiology of Prostate Cancer, World J Oncol, 10 (2019) 63–89. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [3].Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A, Bray F, Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries, CA Cancer J Clin, 71 (2021) 209–249. [DOI] [PubMed] [Google Scholar]
- [4].Catalona WJ, Smith DS, Ratliff TL, Dodds KM, Coplen DE, Yuan JJ, Petros JA, Andriole GL, Measurement of prostate-specific antigen in serum as a screening test for prostate cancer, N Engl J Med, 324 (1991) 1156–1161. [DOI] [PubMed] [Google Scholar]
- [5].Ilic D, Djulbegovic M, Jung JH, Hwang EC, Zhou Q, Cleves A, Agoritsas T, Dahm P, Prostate cancer screening with prostate-specific antigen (PSA) test: a systematic review and meta-analysis, BMJ, 362 (2018) k3519. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [6].Merriel SWD, Funston G, Hamilton W, Prostate Cancer in Primary Care, Adv Ther, 35 (2018) 1285–1294. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [7].Vickers AJ, Sjoberg DD, Ulmert D, Vertosick E, Roobol MJ, Thompson I, Heijnsdijk EA, De Koning H, Atoria-Swartz C, Scardino PT, Lilja H, Empirical estimates of prostate cancer overdiagnosis by age and prostate-specific antigen, BMC Med, 12 (2014) 26. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [8].Cooperberg MR, Carroll PR, Klotz L, Active surveillance for prostate cancer: progress and promise, J Clin Oncol, 29 (2011) 3669–3676. [DOI] [PubMed] [Google Scholar]
- [9].Romero-Otero J, Garcia-Gomez B, Duarte-Ojeda JM, Rodriguez-Antolin A, Vilaseca A, Carlsson SV, Touijer KA, Active surveillance for prostate cancer, Int J Urol, 23 (2016) 211–218. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [10].Frye TP, George AK, Kilchevsky A, Maruf M, Siddiqui MM, Kongnyuy M, Muthigi A, Han H, Parnes HL, Merino M, Choyke PL, Turkbey B, Wood B, Pinto PA, Magnetic Resonance Imaging-Transrectal Ultrasound Guided Fusion Biopsy to Detect Progression in Patients with Existing Lesions on Active Surveillance for Low and Intermediate Risk Prostate Cancer, J Urol, 197 (2017) 640–646. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [11].Eklund M, Jaderling F, Discacciati A, Bergman M, Annerstedt M, Aly M, Glaessgen A, Carlsson S, Gronberg H, Nordstrom T, consortium S., MRI-Targeted or Standard Biopsy in Prostate Cancer Screening, N Engl J Med, 385 (2021) 908–920. [DOI] [PubMed] [Google Scholar]
- [12].Herlemann A, Overland MR, Washington SL 3rd, Cowan JE, Westphalen AC, Carroll PR, Nguyen HG, Shinohara K, Cooperberg MR, How Often Does Magnetic Resonance Imaging Detect Prostate Cancer Missed by Transrectal Ultrasound?, Eur Urol Focus, (2020). [DOI] [PubMed] [Google Scholar]
- [13].Artibani W, Porcaro AB, De Marco V, Cerruto MA, Siracusano S, Management of Biochemical Recurrence after Primary Curative Treatment for Prostate Cancer: A Review, Urol Int, 100 (2018) 251–262. [DOI] [PubMed] [Google Scholar]
- [14].Sandhu S, Moore CM, Chiong E, Beltran H, Bristow RG, Williams SG, Prostate cancer, Lancet, 398 (2021) 1075–1090. [DOI] [PubMed] [Google Scholar]
- [15].Lippman SM, Abate-Shen C, Colbert Maresso KL, Colditz GA, Dannenberg AJ, Davidson NE, Disis ML, DuBois RN, Szabo E, Giuliano AR, Hait WN, Lee JJ, Kensler TW, Kramer BS, Limburg P, Maitra A, Martinez ME, Rebbeck TR, Schmitz KH, Vilar E, Hawk ET, AACR White Paper: Shaping the Future of Cancer Prevention - A Roadmap for Advancing Science and Public Health, Cancer Prev Res (Phila), 11 (2018) 735–778. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [16].Rebbeck TR, Precision prevention of cancer, Cancer Epidemiol Biomarkers Prev, 23 (2014) 2713–2715. [DOI] [PubMed] [Google Scholar]
- [17].Gandaglia G, Leni R, Bray F, Fleshner N, Freedland SJ, Kibel A, Stattin P, Van Poppel H, La Vecchia C, Epidemiology and Prevention of Prostate Cancer, Eur Urol Oncol, (2021). [DOI] [PubMed] [Google Scholar]
- [18].Layne TM, Graubard BI, Ma X, Mayne ST, Albanes D, Prostate cancer risk factors in black and white men in the NIH-AARP Diet and Health Study, Prostate Cancer Prostatic Dis, 22 (2019) 91–100. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [19].Bechis SK, Carroll PR, Cooperberg MR, Impact of age at diagnosis on prostate cancer treatment and survival, J Clin Oncol, 29 (2011) 235–241. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [20].Konety BR, Cowan JE, Carroll PR, Ca PI, Patterns of primary and secondary therapy for prostate cancer in elderly men: analysis of data from CaPSURE, J Urol, 179 (2008) 1797–1803; discussion 1803. [DOI] [PubMed] [Google Scholar]
- [21].Bostwick DG, Burke HB, Djakiew D, Euling S, Ho SM, Landolph J, Morrison H, Sonawane B, Shifflett T, Waters DJ, Timms B, Human prostate cancer risk factors, Cancer, 101 (2004) 2371–2490. [DOI] [PubMed] [Google Scholar]
- [22].Paschos A, Pandya R, Duivenvoorden WC, Pinthus JH, Oxidative stress in prostate cancer: changing research concepts towards a novel paradigm for prevention and therapeutics, Prostate Cancer Prostatic Dis, 16 (2013) 217–225. [DOI] [PubMed] [Google Scholar]
- [23].Kumar B, Koul S, Khandrika L, Meacham RB, Koul HK, Oxidative stress is inherent in prostate cancer cells and is required for aggressive phenotype, Cancer Res, 68 (2008) 1777–1785. [DOI] [PubMed] [Google Scholar]
- [24].Desai N, Sabanegh E Jr., Kim T, Agarwal A, Free radical theory of aging: implications in male infertility, Urology, 75 (2010) 14–19. [DOI] [PubMed] [Google Scholar]
- [25].Szewczyk-Golec K, Tyloch J, Czuczejko J, Antioxidant defense system in prostate adenocarcinoma and benign prostate hyperplasia of elderly patients, Neoplasma, 62 (2015) 119–123. [DOI] [PubMed] [Google Scholar]
- [26].De Nunzio C, Salonia A, Gacci M, Ficarra V, Inflammation is a target of medical treatment for lower urinary tract symptoms associated with benign prostatic hyperplasia, World J Urol, 38 (2020) 2771–2779. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [27].Ficarra V, Rossanese M, Zazzara M, Giannarini G, Abbinante M, Bartoletti R, Mirone V, Scaglione F, The role of inflammation in lower urinary tract symptoms (LUTS) due to benign prostatic hyperplasia (BPH) and its potential impact on medical therapy, Curr Urol Rep, 15 (2014) 463. [DOI] [PubMed] [Google Scholar]
- [28].Nickel JC, Inflammation and benign prostatic hyperplasia, Urol Clin North Am, 35 (2008) 109–115; vii. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [29].Bahmad HF, Jalloul M, Azar J, Moubarak MM, Samad TA, Mukherji D, Al-Sayegh M, Abou-Kheir W, Tumor Microenvironment in Prostate Cancer: Toward Identification of Novel Molecular Biomarkers for Diagnosis, Prognosis, and Therapy Development, Front Genet, 12 (2021) 652747. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [30].De Marzo AM, Platz EA, Sutcliffe S, Xu J, Gronberg H, Drake CG, Nakai Y, Isaacs WB, Nelson WG, Inflammation in prostate carcinogenesis, Nat Rev Cancer, 7 (2007) 256–269. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [31].McArdle PA, Canna K, McMillan DC, McNicol AM, Campbell R, Underwood MA, The relationship between T-lymphocyte subset infiltration and survival in patients with prostate cancer, Br J Cancer, 91 (2004) 541–543. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [32].Nonomura N, Takayama H, Nakayama M, Nakai Y, Kawashima A, Mukai M, Nagahara A, Aozasa K, Tsujimura A, Infiltration of tumour-associated macrophages in prostate biopsy specimens is predictive of disease progression after hormonal therapy for prostate cancer, BJU Int, 107 (2011) 1918–1922. [DOI] [PubMed] [Google Scholar]
- [33].Watanabe M, Kanao K, Suzuki S, Muramatsu H, Morinaga S, Kajikawa K, Kobayashi I, Nishikawa G, Kato Y, Zennami K, Nakamura K, Tsuzuki T, Yoshikawa K, Ueda R, Sumitomo M, Increased infiltration of CCR4-positive regulatory T cells in prostate cancer tissue is associated with a poor prognosis, Prostate, 79 (2019) 1658–1665. [DOI] [PubMed] [Google Scholar]
- [34].Espey MG, Miranda KM, Thomas DD, Xavier S, Citrin D, Vitek MP, Wink DA, A chemical perspective on the interplay between NO, reactive oxygen species, and reactive nitrogen oxide species, Ann N Y Acad Sci, 962 (2002) 195–206. [DOI] [PubMed] [Google Scholar]
- [35].Le Magnen C, Virk RK, Dutta A, Kim JY, Panja S, Lopez-Bujanda ZA, Califano A, Drake CG, Mitrofanova A, Abate-Shen C, Cooperation of loss of NKX3.1 and inflammation in prostate cancer initiation, Dis Model Mech, 11 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- [36].Liu X, Grogan TR, Hieronymus H, Hashimoto T, Mottahedeh J, Cheng D, Zhang L, Huang K, Stoyanova T, Park JW, Shkhyan RO, Nowroozizadeh B, Rettig MB, Sawyers CL, Elashoff D, Horvath S, Huang J, Witte ON, Goldstein AS, Low CD38 Identifies Progenitor-like Inflammation-Associated Luminal Cells that Can Initiate Human Prostate Cancer and Predict Poor Outcome, Cell Rep, 17 (2016) 2596–2606. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [37].Klein EA, Thompson IM Jr., Tangen CM, Crowley JJ, Lucia MS, Goodman PJ, Minasian LM, Ford LG, Parnes HL, Gaziano JM, Karp DD, Lieber MM, Walther PJ, Klotz L, Parsons JK, Chin JL, Darke AK, Lippman SM, Goodman GE, Meyskens FL Jr., Baker LH, Vitamin E and the risk of prostate cancer: the Selenium and Vitamin E Cancer Prevention Trial (SELECT), JAMA, 306 (2011) 1549–1556. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [38].Veitonmaki T, Murtola TJ, Maattanen L, Taari K, Stenman UH, Tammela TL, Auvinen A, Prostate cancer risk and nonsteroidal antiinflammatory drug use in the Finnish prostate cancer screening trial, Br J Cancer, 111 (2014) 1421–1431. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [39].Vidal AC, Howard LE, Moreira DM, Castro-Santamaria R, Andriole GL, Freedland SJ, Aspirin, NSAIDs, and risk of prostate cancer: results from the REDUCE study, Clin Cancer Res, 21 (2015) 756–762. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [40].Tasdogan A, Ubellacker JM, Morrison SJ, Redox Regulation in Cancer Cells during Metastasis, Cancer Discov, 11 (2021) 2682–2692. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [41].Dutta A, Panja S, Virk RK, Kim JY, Zott R, Cremers S, Golombos DM, Liu D, Mosquera JM, Mostaghel EA, Barbieri CE, Mitrofanova A, Abate-Shen C, Co-clinical Analysis of a Genetically Engineered Mouse Model and Human Prostate Cancer Reveals Significance of NKX3.1 Expression for Response to 5alpha-reductase Inhibition, Eur Urol, 72 (2017) 499–506. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [42].Fuletra JG, Kamenko A, Ramsey F, Eun DD, Reese AC, African-American men with prostate cancer have larger tumor volume than Caucasian men despite no difference in serum prostate specific antigen, Can J Urol, 25 (2018) 9193–9198. [PubMed] [Google Scholar]
- [43].McGinley KF, Tay KJ, Moul JW, Prostate cancer in men of African origin, Nat Rev Urol, 13 (2016) 99–107. [DOI] [PubMed] [Google Scholar]
- [44].Khani F, Mosquera JM, Park K, Blattner M, O’Reilly C, MacDonald TY, Chen Z, Srivastava A, Tewari AK, Barbieri CE, Rubin MA, Robinson BD, Evidence for molecular differences in prostate cancer between African American and Caucasian men, Clin Cancer Res, 20 (2014) 4925–4934. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [45].Williams VL, Awasthi S, Fink AK, Pow-Sang JM, Park JY, Gerke T, Yamoah K, African-American men and prostate cancer-specific mortality: a competing risk analysis of a large institutional cohort, 1989–2015, Cancer Med, 7 (2018) 2160–2171. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [46].Jones RA, Underwood SM, Rivers BM, Reducing prostate cancer morbidity and mortality in African American men: issues and challenges, Clin J Oncol Nurs, 11 (2007) 865–872. [DOI] [PubMed] [Google Scholar]
- [47].Yamoah K, Johnson MH, Choeurng V, Faisal FA, Yousefi K, Haddad Z, Ross AE, Alshalafa M, Den R, Lal P, Feldman M, Dicker AP, Klein EA, Davicioni E, Rebbeck TR, Schaeffer EM, Novel Biomarker Signature That May Predict Aggressive Disease in African American Men With Prostate Cancer, J Clin Oncol, 33 (2015) 2789–2796. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [48].Darcey E, Pereira G, Salter A, Fritschi L, Leavy J, Ambrosini GL, Boyle T, The Impact of Lifestyle-related Factors on Survival After a Prostate Cancer Diagnosis, Eur Urol, 75 (2019) 884–885. [DOI] [PubMed] [Google Scholar]
- [49].Kirby JB, Kaneda T, Neighborhood socioeconomic disadvantage and access to health care, J Health Soc Behav, 46 (2005) 15–31. [DOI] [PubMed] [Google Scholar]
- [50].White K, Haas JS, Williams DR, Elucidating the role of place in health care disparities: the example of racial/ethnic residential segregation, Health Serv Res, 47 (2012) 1278–1299. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [51].Poulson MR, Helrich SA, Kenzik KM, Dechert TA, Sachs TE, Katz MH, The impact of racial residential segregation on prostate cancer diagnosis and treatment, BJU Int, 127 (2021) 636–644. [DOI] [PubMed] [Google Scholar]
- [52].Nair SS, Chakravarty D, Dovey ZS, Zhang X, Tewari AK, Why do African-American men face higher risks for lethal prostate cancer?, Curr Opin Urol, 32 (2022) 96–101. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [53].Han Y, Rand KA, Hazelett DJ, Ingles SA, Kittles RA, Strom SS, Rybicki BA, Nemesure B, Isaacs WB, Stanford JL, Zheng W, Schumacher FR, Berndt SI, Wang Z, Xu J, Rohland N, Reich D, Tandon A, Pasaniuc B, Allen A, Quinque D, Mallick S, Notani D, Rosenfeld MG, Jayani RS, Kolb S, Gapstur SM, Stevens VL, Pettaway CA, Yeboah ED, Tettey Y, Biritwum RB, Adjei AA, Tay E, Truelove A, Niwa S, Chokkalingam AP, John EM, Murphy AB, Signorello LB, Carpten J, Leske MC, Wu SY, Hennis AJM, Neslund-Dudas C, Hsing AW, Chu L, Goodman PJ, Klein EA, Zheng SL, Witte JS, Casey G, Lubwama A, Pooler LC, Sheng X, Coetzee GA, Cook MB, Chanock SJ, Stram DO, Watya S, Blot WJ, Conti DV, Henderson BE, Haiman CA, Prostate Cancer Susceptibility in Men of African Ancestry at 8q24, J Natl Cancer Inst, 108 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- [54].Koga Y, Song H, Chalmers ZR, Newberg J, Kim E, Carrot-Zhang J, Piou D, Polak P, Abdulkadir SA, Ziv E, Meyerson M, Frampton GM, Campbell JD, Huang FW, Genomic Profiling of Prostate Cancers from Men with African and European Ancestry, Clin Cancer Res, 26 (2020) 4651–4660. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [55].Hoffmann TJ, Passarelli MN, Graff RE, Emami NC, Sakoda LC, Jorgenson E, Habel LA, Shan J, Ranatunga DK, Quesenberry CP, Chao CR, Ghai NR, Aaronson D, Presti J, Nordstrom T, Wang Z, Berndt SI, Chanock SJ, Mosley JD, Klein RJ, Middha M, Lilja H, Melander O, Kvale MN, Kwok PY, Schaefer C, Risch N, Van Den Eeden SK, Witte JS, Genome-wide association study of prostate-specific antigen levels identifies novel loci independent of prostate cancer, Nat Commun, 8 (2017) 14248. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [56].Bancroft EK, Raghallaigh HN, Page EC, Eeles RA, Updates in Prostate Cancer Research and Screening in Men at Genetically Higher Risk, Curr Genet Med Rep, 9 (2021) 47–58. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [57].Bratt O, Drevin L, Akre O, Garmo H, Stattin P, Family History and Probability of Prostate Cancer, Differentiated by Risk Category: A Nationwide Population-Based Study, J Natl Cancer Inst, 108 (2016). [DOI] [PubMed] [Google Scholar]
- [58].Zeegers MP, Jellema A, Ostrer H, Empiric risk of prostate carcinoma for relatives of patients with prostate carcinoma: a meta-analysis, Cancer, 97 (2003) 1894–1903. [DOI] [PubMed] [Google Scholar]
- [59].Das S, Salami SS, Spratt DE, Kaffenberger SD, Jacobs MF, Morgan TM, Bringing Prostate Cancer Germline Genetics into Clinical Practice, J Urol, 202 (2019) 223–230. [DOI] [PubMed] [Google Scholar]
- [60].Na R, Zheng SL, Han M, Yu H, Jiang D, Shah S, Ewing CM, Zhang L, Novakovic K, Petkewicz J, Gulukota K, Helseth DL Jr., Quinn M, Humphries E, Wiley KE, Isaacs SD, Wu Y, Liu X, Zhang N, Wang CH, Khandekar J, Hulick PJ, Shevrin DH, Cooney KA, Shen Z, Partin AW, Carter HB, Carducci MA, Eisenberger MA, Denmeade SR, McGuire M, Walsh PC, Helfand BT, Brendler CB, Ding Q, Xu J, Isaacs WB, Germline Mutations in ATM and BRCA1/2 Distinguish Risk for Lethal and Indolent Prostate Cancer and are Associated with Early Age at Death, Eur Urol, 71 (2017) 740–747. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [61].Nicolosi P, Ledet E, Yang S, Michalski S, Freschi B, O’Leary E, Esplin ED, Nussbaum RL, Sartor O, Prevalence of Germline Variants in Prostate Cancer and Implications for Current Genetic Testing Guidelines, JAMA Oncol, 5 (2019) 523–528. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [62].Zhen JT, Syed J, Nguyen KA, Leapman MS, Agarwal N, Brierley K, Llor X, Hofstatter E, Shuch B, Genetic testing for hereditary prostate cancer: Current status and limitations, Cancer, 124 (2018) 3105–3117. [DOI] [PubMed] [Google Scholar]
- [63].Amundadottir LT, Sulem P, Gudmundsson J, Helgason A, Baker A, Agnarsson BA, Sigurdsson A, Benediktsdottir KR, Cazier JB, Sainz J, Jakobsdottir M, Kostic J, Magnusdottir DN, Ghosh S, Agnarsson K, Birgisdottir B, Le Roux L, Olafsdottir A, Blondal T, Andresdottir M, Gretarsdottir OS, Bergthorsson JT, Gudbjartsson D, Gylfason A, Thorleifsson G, Manolescu A, Kristjansson K, Geirsson G, Isaksson H, Douglas J, Johansson JE, Balter K, Wiklund F, Montie JE, Yu X, Suarez BK, Ober C, Cooney KA, Gronberg H, Catalona WJ, Einarsson GV, Barkardottir RB, Gulcher JR, Kong A, Thorsteinsdottir U, Stefansson K, A common variant associated with prostate cancer in European and African populations, Nat Genet, 38 (2006) 652–658. [DOI] [PubMed] [Google Scholar]
- [64].Eeles RA, Olama AA, Benlloch S, Saunders EJ, Leongamornlert DA, Tymrakiewicz M, Ghoussaini M, Luccarini C, Dennis J, Jugurnauth-Little S, Dadaev T, Neal DE, Hamdy FC, Donovan JL, Muir K, Giles GG, Severi G, Wiklund F, Gronberg H, Haiman CA, Schumacher F, Henderson BE, Le Marchand L, Lindstrom S, Kraft P, Hunter DJ, Gapstur S, Chanock SJ, Berndt SI, Albanes D, Andriole G, Schleutker J, Weischer M, Canzian F, Riboli E, Key TJ, Travis RC, Campa D, Ingles SA, John EM, Hayes RB, Pharoah PD, Pashayan N, Khaw KT, Stanford JL, Ostrander EA, Signorello LB, Thibodeau SN, Schaid D, Maier C, Vogel W, Kibel AS, Cybulski C, Lubinski J, Cannon-Albright L, Brenner H, Park JY, Kaneva R, Batra J, Spurdle AB, Clements JA, Teixeira MR, Dicks E, Lee A, Dunning AM, Baynes C, Conroy D, Maranian MJ, Ahmed S, Govindasami K, Guy M, Wilkinson RA, Sawyer EJ, Morgan A, Dearnaley DP, Horwich A, Huddart RA, Khoo VS, Parker CC, Van As NJ, Woodhouse CJ, Thompson A, Dudderidge T, Ogden C, Cooper CS, Lophatananon A, Cox A, Southey MC, Hopper JL, English DR, Aly M, Adolfsson J, Xu J, Zheng SL, Yeager M, Kaaks R, Diver WR, Gaudet MM, Stern MC, Corral R, Joshi AD, Shahabi A, Wahlfors T, Tammela TL, Auvinen A, Virtamo J, Klarskov P, Nordestgaard BG, Roder MA, Nielsen SF, Bojesen SE, Siddiq A, Fitzgerald LM, Kolb S, Kwon EM, Karyadi DM, Blot WJ, Zheng W, Cai Q, McDonnell SK, Rinckleb AE, Drake B, Colditz G, Wokolorczyk D, Stephenson RA, Teerlink C, Muller H, Rothenbacher D, Sellers TA, Lin HY, Slavov C, Mitev V, Lose F, Srinivasan S, Maia S, Paulo P, Lange E, Cooney KA, Antoniou AC, Vincent D, Bacot F, Tessier DC, C.O.-C.R.U.G.-E. Initiative, B. Australian Prostate Cancer, U.K.G.P.C.S.C.B.A.o.U.S.S.o. Oncology, U.K.P.S. Collaborators, P. Consortium, Kote-Jarai Z, Easton DF, Identification of 23 new prostate cancer susceptibility loci using the iCOGS custom genotyping array, Nat Genet, 45 (2013) 385–391, 391e381–382. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [65].Schumacher FR, Al Olama AA, Berndt SI, Benlloch S, Ahmed M, Saunders EJ, Dadaev T, Leongamornlert D, Anokian E, Cieza-Borrella C, Goh C, Brook MN, Sheng X, Fachal L, Dennis J, Tyrer J, Muir K, Lophatananon A, Stevens VL, Gapstur SM, Carter BD, Tangen CM, Goodman PJ, Thompson IM Jr., Batra J, Chambers S, Moya L, Clements J, Horvath L, Tilley W, Risbridger GP, Gronberg H, Aly M, Nordstrom T, Pharoah P, Pashayan N, Schleutker J, Tammela TLJ, Sipeky C, Auvinen A, Albanes D, Weinstein S, Wolk A, Hakansson N, West CML, Dunning AM, Burnet N, Mucci LA, Giovannucci E, Andriole GL, Cussenot O, Cancel-Tassin G, Koutros S, Beane Freeman LE, Sorensen KD, Orntoft TF, Borre M, Maehle L, Grindedal EM, Neal DE, Donovan JL, Hamdy FC, Martin RM, Travis RC, Key TJ, Hamilton RJ, Fleshner NE, Finelli A, Ingles SA, Stern MC, Rosenstein BS, Kerns SL, Ostrer H, Lu YJ, Zhang HW, Feng N, Mao X, Guo X, Wang G, Sun Z, Giles GG, Southey MC, MacInnis RJ, FitzGerald LM, Kibel AS, Drake BF, Vega A, Gomez-Caamano A, Szulkin R, Eklund M, Kogevinas M, Llorca J, Castano-Vinyals G, Penney KL, Stampfer M, Park JY, Sellers TA, Lin HY, Stanford JL, Cybulski C, Wokolorczyk D, Lubinski J, Ostrander EA, Geybels MS, Nordestgaard BG, Nielsen SF, Weischer M, Bisbjerg R, Roder MA, Iversen P, Brenner H, Cuk K, Holleczek B, Maier C, Luedeke M, Schnoeller T, Kim J, Logothetis CJ, John EM, Teixeira MR, Paulo P, Cardoso M, Neuhausen SL, Steele L, Ding YC, De Ruyck K, De Meerleer G, Ost P, Razack A, Lim J, Teo SH, Lin DW, Newcomb LF, Lessel D, Gamulin M, Kulis T, Kaneva R, Usmani N, Singhal S, Slavov C, Mitev V, Parliament M, Claessens F, Joniau S, Van den Broeck T, Larkin S, Townsend PA, Aukim-Hastie C, Gago-Dominguez M, Castelao JE, Martinez ME, Roobol MJ, Jenster G, van Schaik RHN, Menegaux F, Truong T, Koudou YA, Xu J, Khaw KT, Cannon-Albright L, Pandha H, Michael A, Thibodeau SN, McDonnell SK, Schaid DJ, Lindstrom S, Turman C, Ma J, Hunter DJ, Riboli E, Siddiq A, Canzian F, Kolonel LN, Le Marchand L, Hoover RN, Machiela MJ, Cui Z, Kraft P, Amos CI, Conti DV, Easton DF, Wiklund F, Chanock SJ, Henderson BE, Kote-Jarai Z, Haiman CA, Eeles RA, Profile S, Australian Prostate Cancer B, Study I, Canary PI, Breast C Prostate Cancer Cohort, P. Consortium, S. Cancer of the Prostate in, L. Prostate Cancer Genome-wide Association Study of Uncommon Susceptibility, Genetic A, C. Mechanisms in Oncology/Elucidating Loci Involved in Prostate Cancer Susceptibility, Association analyses of more than 140,000 men identify 63 new prostate cancer susceptibility loci, Nat Genet, 50 (2018) 928–936. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [66].Dong X, Wang L, Taniguchi K, Wang X, Cunningham JM, McDonnell SK, Qian C, Marks AF, Slager SL, Peterson BJ, Smith DI, Cheville JC, Blute ML, Jacobsen SJ, Schaid DJ, Tindall DJ, Thibodeau SN, Liu W, Mutations in CHEK2 associated with prostate cancer risk, Am J Hum Genet, 72 (2003) 270–280. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [67].Ewing CM, Ray AM, Lange EM, Zuhlke KA, Robbins CM, Tembe WD, Wiley KE, Isaacs SD, Johng D, Wang Y, Bizon C, Yan G, Gielzak M, Partin AW, Shanmugam V, Izatt T, Sinari S, Craig DW, Zheng SL, Walsh PC, Montie JE, Xu J, Carpten JD, Isaacs WB, Cooney KA, Germline mutations in HOXB13 and prostate-cancer risk, N Engl J Med, 366 (2012) 141–149. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [68].Grindedal EM, Moller P, Eeles R, Stormorken AT, Bowitz-Lothe IM, Landro SM, Clark N, Kvale R, Shanley S, Maehle L, Germ-line mutations in mismatch repair genes associated with prostate cancer, Cancer Epidemiol Biomarkers Prev, 18 (2009) 2460–2467. [DOI] [PubMed] [Google Scholar]
- [69].Mitra A, Fisher C, Foster CS, Jameson C, Barbachanno Y, Bartlett J, Bancroft E, Doherty R, Kote-Jarai Z, Peock S, Easton D, Impact, E. Collaborators, Eeles R, Prostate cancer in male BRCA1 and BRCA2 mutation carriers has a more aggressive phenotype, Br J Cancer, 98 (2008) 502–507. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [70].Eeles R, Goh C, Castro E, Bancroft E, Guy M, Al Olama AA, Easton D, Kote-Jarai Z, The genetic epidemiology of prostate cancer and its clinical implications, Nat Rev Urol, 11 (2014) 18–31. [DOI] [PubMed] [Google Scholar]
- [71].Helfand BT, Loeb S, Hu Q, Cooper PR, Roehl KA, McGuire BB, Baumann NA, Catalona WJ, Personalized prostate specific antigen testing using genetic variants may reduce unnecessary prostate biopsies, J Urol, 189 (2013) 1697–1701. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [72].Jin HJ, Jung S, DebRoy AR, Davuluri RV, Identification and validation of regulatory SNPs that modulate transcription factor chromatin binding and gene expression in prostate cancer, Oncotarget, 7 (2016) 54616–54626. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [73].Sun J, Kader AK, Hsu FC, Kim ST, Zhu Y, Turner AR, Jin T, Zhang Z, Adolfsson J, Wiklund F, Zheng SL, Isaacs WB, Gronberg H, Xu J, Inherited genetic markers discovered to date are able to identify a significant number of men at considerably elevated risk for prostate cancer, Prostate, 71 (2011) 421–430. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [74].Zheng SL, Sun J, Wiklund F, Smith S, Stattin P, Li G, Adami HO, Hsu FC, Zhu Y, Balter K, Kader AK, Turner AR, Liu W, Bleecker ER, Meyers DA, Duggan D, Carpten JD, Chang BL, Isaacs WB, Xu J, Gronberg H, Cumulative association of five genetic variants with prostate cancer, N Engl J Med, 358 (2008) 910–919. [DOI] [PubMed] [Google Scholar]
- [75].Helfand BT, Roehl KA, Cooper PR, McGuire BB, Fitzgerald LM, Cancel-Tassin G, Cornu JN, Bauer S, Van Blarigan EL, Chen X, Duggan D, Ostrander EA, Gwo-Shu M, Zhang ZF, Chang SC, Jeong S, Fontham ET, Smith G, Mohler JL, Berndt SI, McDonnell SK, Kittles R, Rybicki BA, Freedman M, Kantoff PW, Pomerantz M, Breyer JP, Smith JR, Rebbeck TR, Mercola D, Isaacs WB, Wiklund F, Cussenot O, Thibodeau SN, Schaid DJ, Cannon-Albright L, Cooney KA, Chanock SJ, Stanford JL, Chan JM, Witte J, Xu J, Bensen JT, Taylor JA, Catalona WJ, Associations of prostate cancer risk variants with disease aggressiveness: results of the NCI-SPORE Genetics Working Group analysis of 18,343 cases, Hum Genet, 134 (2015) 439–450. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [76].Alvarez-Cubero MJ, Pascual-Geler M, Martinez-Gonzalez LJ, Exposito Ruiz M, Saiz M, Cozar JM, Lorente JA, Association between RNASEL, MSR1, and ELAC2 single nucleotide polymorphisms and gene expression in prostate cancer risk, Urol Oncol, 34 (2016) 431 e431–438. [DOI] [PubMed] [Google Scholar]
- [77].Martinez EE, Darke AK, Tangen CM, Goodman PJ, Fowke JH, Klein EA, Abdulkadir SA, A functional variant in NKX3.1 associated with prostate cancer risk in the Selenium and Vitamin E Cancer Prevention Trial (SELECT), Cancer Prev Res (Phila), 7 (2014) 950–957. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [78].Zheng SL, Ju JH, Chang BL, Ortner E, Sun J, Isaacs SD, Sun J, Wiley KE, Liu W, Zemedkun M, Walsh PC, Ferretti J, Gruschus J, Isaacs WB, Gelmann EP, Xu J, Germ-line mutation of NKX3.1 cosegregates with hereditary prostate cancer and alters the homeodomain structure and function, Cancer Res, 66 (2006) 69–77. [DOI] [PubMed] [Google Scholar]
- [79].Douglas JA, Levin AM, Zuhlke KA, Ray AM, Johnson GR, Lange EM, Wood DP, Cooney KA, Common variation in the BRCA1 gene and prostate cancer risk, Cancer Epidemiol Biomarkers Prev, 16 (2007) 1510–1516. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [80].Gelmann EP, Steadman DJ, Ma J, Ahronovitz N, Voeller HJ, Swope S, Abbaszadegan M, Brown KM, Strand K, Hayes RB, Stampfer MJ, Occurrence of NKX3.1 C154T polymorphism in men with and without prostate cancer and studies of its effect on protein function, Cancer Res, 62 (2002) 2654–2659. [PubMed] [Google Scholar]
- [81].Rodriguez Ortner E, Hayes RB, Weissfeld J, Gelmann EP, Effect of homeodomain protein NKX3.1 R52C polymorphism on prostate gland size, Urology, 67 (2006) 311–315. [DOI] [PubMed] [Google Scholar]
- [82].FitzGerald LM, Kwon EM, Conomos MP, Kolb S, Holt SK, Levine D, Feng Z, Ostrander EA, Stanford JL, Genome-wide association study identifies a genetic variant associated with risk for more aggressive prostate cancer, Cancer Epidemiol Biomarkers Prev, 20 (2011) 1196–1203. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [83].Xu J, Zheng SL, Isaacs SD, Wiley KE, Wiklund F, Sun J, Kader AK, Li G, Purcell LD, Kim ST, Hsu FC, Stattin P, Hugosson J, Adolfsson J, Walsh PC, Trent JM, Duggan D, Carpten J, Gronberg H, Isaacs WB, Inherited genetic variant predisposes to aggressive but not indolent prostate cancer, Proc Natl Acad Sci U S A, 107 (2010) 2136–2140. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [84].Shui IM, Lindstrom S, Kibel AS, Berndt SI, Campa D, Gerke T, Penney KL, Albanes D, Berg C, Bueno-de-Mesquita HB, Chanock S, Crawford ED, Diver WR, Gapstur SM, Gaziano JM, Giles GG, Henderson B, Hoover R, Johansson M, Le Marchand L, Ma J, Navarro C, Overvad K, Schumacher FR, Severi G, Siddiq A, Stampfer M, Stevens VL, Travis RC, Trichopoulos D, Vineis P, Mucci LA, Yeager M, Giovannucci E, Kraft P, Prostate cancer (PCa) risk variants and risk of fatal PCa in the National Cancer Institute Breast and Prostate Cancer Cohort Consortium, Eur Urol, 65 (2014) 1069–1075. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [85].Petrovics G, Price DK, Lou H, Chen Y, Garland L, Bass S, Jones K, Kohaar I, Ali A, Ravindranath L, Young D, Cullen J, Dorsey TH, Sesterhenn IA, Brassell SA, Rosner IL, Ross D, Dahut W, Ambs S, Figg WD, Srivastava S, Dean M, Increased frequency of germline BRCA2 mutations associates with prostate cancer metastasis in a racially diverse patient population, Prostate Cancer Prostatic Dis, 22 (2019) 406–410. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [86].Cook MB, Wang Z, Yeboah ED, Tettey Y, Biritwum RB, Adjei AA, Tay E, Truelove A, Niwa S, Chung CC, Chokkalingam AP, Chu LW, Yeager M, Hutchinson A, Yu K, Rand KA, Haiman CA, African GC Ancestry Prostate Cancer, Hoover RN, Hsing AW, Chanock SJ, A genome-wide association study of prostate cancer in West African men, Hum Genet, 133 (2014) 509–521. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [87].Heidegger I, Tsaur I, Borgmann H, Surcel C, Kretschmer A, Mathieu R, Visschere P, Valerio M, van den Bergh RCN, Ost P, Tilki D, Gandaglia G, Ploussard G, Party E-YPCW, Hereditary prostate cancer - Primetime for genetic testing?, Cancer Treat Rev, 81 (2019) 101927. [DOI] [PubMed] [Google Scholar]
- [88].Nguyen AH, Tremblay M, Haigh K, Koumakpayi IH, Paquet M, Pandolfi PP, Mes-Masson AM, Saad F, Haigh JJ, Bouchard M, Gata3 antagonizes cancer progression in Pten-deficient prostates, Hum Mol Genet, 22 (2013) 2400–2410. [DOI] [PubMed] [Google Scholar]
- [89].Darst BF, Sheng X, Eeles RA, Kote-Jarai Z, Conti DV, Haiman CA, Combined Effect of a Polygenic Risk Score and Rare Genetic Variants on Prostate Cancer Risk, Eur Urol, 80 (2021) 134–138. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [90].Sipeky C, Talala KM, Tammela TLJ, Taari K, Auvinen A, Schleutker J, Prostate cancer risk prediction using a polygenic risk score, Sci Rep, 10 (2020) 17075. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [91].Conti DV, Darst BF, Moss LC, Saunders EJ, Sheng X, Chou A, Schumacher FR, Olama AAA, Benlloch S, Dadaev T, Brook MN, Sahimi A, Hoffmann TJ, Takahashi A, Matsuda K, Momozawa Y, Fujita M, Muir K, Lophatananon A, Wan P, Le Marchand L, Wilkens LR, Stevens VL, Gapstur SM, Carter BD, Schleutker J, Tammela TLJ, Sipeky C, Auvinen A, Giles GG, Southey MC, MacInnis RJ, Cybulski C, Wokolorczyk D, Lubinski J, Neal DE, Donovan JL, Hamdy FC, Martin RM, Nordestgaard BG, Nielsen SF, Weischer M, Bojesen SE, Roder MA, Iversen P, Batra J, Chambers S, Moya L, Horvath L, Clements JA, Tilley W, Risbridger GP, Gronberg H, Aly M, Szulkin R, Eklund M, Nordstrom T, Pashayan N, Dunning AM, Ghoussaini M, Travis RC, Key TJ, Riboli E, Park JY, Sellers TA, Lin HY, Albanes D, Weinstein SJ, Mucci LA, Giovannucci E, Lindstrom S, Kraft P, Hunter DJ, Penney KL, Turman C, Tangen CM, Goodman PJ, Thompson IM Jr., Hamilton RJ, Fleshner NE, Finelli A, Parent ME, Stanford JL, Ostrander EA, Geybels MS, Koutros S, Freeman LEB, Stampfer M, Wolk A, Hakansson N, Andriole GL, Hoover RN, Machiela MJ, Sorensen KD, Borre M, Blot WJ, Zheng W, Yeboah ED, Mensah JE, Lu YJ, Zhang HW, Feng N, Mao X, Wu Y, Zhao SC, Sun Z, Thibodeau SN, McDonnell SK, Schaid DJ, West CML, Burnet N, Barnett G, Maier C, Schnoeller T, Luedeke M, Kibel AS, Drake BF, Cussenot O, Cancel-Tassin G, Menegaux F, Truong T, Koudou YA, John EM, Grindedal EM, Maehle L, Khaw KT, Ingles SA, Stern MC, Vega A, Gomez-Caamano A, Fachal L, Rosenstein BS, Kerns SL, Ostrer H, Teixeira MR, Paulo P, Brandao A, Watya S, Lubwama A, Bensen JT, Fontham ETH, Mohler J, Taylor JA, Kogevinas M, Llorca J, Castano-Vinyals G, Cannon-Albright L, Teerlink CC, Huff CD, Strom SS, Multigner L, Blanchet P, Brureau L, Kaneva R, Slavov C, Mitev V, Leach RJ, Weaver B, Brenner H, Cuk K, Holleczek B, Saum KU, Klein EA, Hsing AW, Kittles RA, Murphy AB, Logothetis CJ, Kim J, Neuhausen SL, Steele L, Ding YC, Isaacs WB, Nemesure B, Hennis AJM, Carpten J, Pandha H, Michael A, De Ruyck K, De Meerleer G, Ost P, Xu J, Razack A, Lim J, Teo SH, Newcomb LF, Lin DW, Fowke JH, Neslund-Dudas C, Rybicki BA, Gamulin M, Lessel D, Kulis T, Usmani N, Singhal S, Parliament M, Claessens F, Joniau S, Van den Broeck T, Gago-Dominguez M, Castelao JE, Martinez ME, Larkin S, Townsend PA, Aukim-Hastie C, Bush WS, Aldrich MC, Crawford DC, Srivastava S, Cullen JC, Petrovics G, Casey G, Roobol MJ, Jenster G, van Schaik RHN, Hu JJ, Sanderson M, Varma R, McKean-Cowdin R, Torres M, Mancuso N, Berndt SI, Van Den Eeden SK, Easton DF, Chanock SJ, Cook MB, Wiklund F, Nakagawa H, Witte JS, Eeles RA, Kote-Jarai Z, Haiman CA, Trans-ancestry genome-wide association meta-analysis of prostate cancer identifies new susceptibility loci and informs genetic risk prediction, Nat Genet, 53 (2021) 65–75. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [92].Seibert TM, Fan CC, Wang Y, Zuber V, Karunamuni R, Parsons JK, Eeles RA, Easton DF, Kote-Jarai Z, Al Olama AA, Garcia SB, Muir K, Gronberg H, Wiklund F, Aly M, Schleutker J, Sipeky C, Tammela TL, Nordestgaard BG, Nielsen SF, Weischer M, Bisbjerg R, Roder MA, Iversen P, Key TJ, Travis RC, Neal DE, Donovan JL, Hamdy FC, Pharoah P, Pashayan N, Khaw KT, Maier C, Vogel W, Luedeke M, Herkommer K, Kibel AS, Cybulski C, Wokolorczyk D, Kluzniak W, Cannon-Albright L, Brenner H, Cuk K, Saum KU, Park JY, Sellers TA, Slavov C, Kaneva R, Mitev V, Batra J, Clements JA, Spurdle A, Teixeira MR, Paulo P, Maia S, Pandha H, Michael A, Kierzek A, Karow DS, Mills IG, Andreassen OA, Dale AM, Consortium* P., Polygenic hazard score to guide screening for aggressive prostate cancer: development and validation in large scale cohorts, BMJ, 360 (2018) j5757. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [93].Huynh-Le MP, Fan CC, Karunamuni R, Thompson WK, Martinez ME, Eeles RA, Kote-Jarai Z, Muir K, Schleutker J, Pashayan N, Batra J, Gronberg H, Neal DE, Donovan JL, Hamdy FC, Martin RM, Nielsen SF, Nordestgaard BG, Wiklund F, Tangen CM, Giles GG, Wolk A, Albanes D, Travis RC, Blot WJ, Zheng W, Sanderson M, Stanford JL, Mucci LA, West CML, Kibel AS, Cussenot O, Berndt SI, Koutros S, Sorensen KD, Cybulski C, Grindedal EM, Menegaux F, Khaw KT, Park JY, Ingles SA, Maier C, Hamilton RJ, Thibodeau SN, Rosenstein BS, Lu YJ, Watya S, Vega A, Kogevinas M, Penney KL, Huff C, Teixeira MR, Multigner L, Leach RJ, Cannon-Albright L, Brenner H, John EM, Kaneva R, Logothetis CJ, Neuhausen SL, De Ruyck K, Pandha H, Razack A, Newcomb LF, Fowke JH, Gamulin M, Usmani N, Claessens F, Gago-Dominguez M, Townsend PA, Bush WS, Roobol MJ, Parent ME, Hu JJ, Mills IG, Andreassen OA, Dale AM, Seibert TM, U. collaborators, Apcb, N.-L.P. Investigators, I.S.S. Committee, Collaborators, P.I. Canary, C. Profile Study Steering, P. Consortium, Polygenic hazard score is associated with prostate cancer in multi-ethnic populations, Nat Commun, 12 (2021) 1236. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [94].Pashayan N, Duffy SW, Neal DE, Hamdy FC, Donovan JL, Martin RM, Harrington P, Benlloch S, Amin Al Olama A, Shah M, Kote-Jarai Z, Easton DF, Eeles R, Pharoah PD, Implications of polygenic risk-stratified screening for prostate cancer on overdiagnosis, Genet Med, 17 (2015) 789–795. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [95].Barnes DR, Silvestri V, Leslie G, McGuffog L, Dennis J, Yang X, Adlard J, Agnarsson BA, Ahmed M, Aittomaki K, Andrulis IL, Arason A, Arnold N, Auber B, Azzollini J, Balmana J, Barkardottir RB, Barrowdale D, Barwell J, Belotti M, Benitez J, Berthet P, Boonen SE, Borg A, Bozsik A, Brady A, Brennan P, Brewer C, Brunet J, Bucalo A, Buys SS, Caldes T, Caligo MA, Campbell I, Cassingham H, Lotte Christensen L, Cini G, Claes KBM, G.S. Collaborators, E. Collaborators, Cook J, Coppa A, Cortesi L, Damante G, Darder E, Davidson R, de la Hoya M, De Leeneer K, de Putter R, Del Valle J, Diez O, Chun Ding Y, Domchek SM, Donaldson A, Eason J, Eeles R, Engel C, Gareth Evans D, Feliubadalo L, Fostira F, Frone M, Frost D, Gallagher D, Gehrig A, Giraud S, Glendon G, Godwin AK, Goldgar DE, Greene MH, Gregory H, Gross E, Hahnen E, Hamann U, Hansen TVO, Hanson H, Hentschel J, Horvath J, K.C. Investigators, H. Investigators, Izatt L, Izquierdo A, James PA, Janavicius R, Birk Jensen U, Johannsson OT, John EM, Kramer G, Kroeldrup L, Kruse TA, Lautrup C, Lazaro C, Lesueur F, Lopez-Fernandez A, Mai PL, Manoukian S, Matrai Z, Matricardi L, Maxwell KN, Mebirouk N, Meindl A, Montagna M, Monteiro AN, Morrison PJ, Muranen TA, Murray A, Nathanson KL, Neuhausen SL, Nevanlinna H, Nguyen-Dumont T, Niederacher D, Olah E, Olopade OI, Palli D, Parsons MT, Sokilde Pedersen I, Peissel B, Perez-Segura P, Peterlongo P, Petersen AH, Pinto P, Porteous ME, Pottinger C, Angel Pujana M, Radice P, Ramser J, Rantala J, Robson M, Rogers MT, Ronlund K, Rump A, Maria A, de Abajo Sanchez, Shah PD, Sharif S, Side LE, Singer CF, Stadler Z, Steele L, Stoppa-Lyonnet D, Sutter C, Yen Tan Y, Teixeira MR, Teule A, Thull DL, Tischkowitz M, Toland AE, Tommasi S, Toss A, Trainer AH, Tripathi V, Valentini V, van Asperen CJ, Venturelli M, Viel A, Vijai J, Walker L, Wang-Gohrke S, Wappenschmidt B, Whaite A, Zanna I, Offit K, Thomassen M, Couch FJ, Schmutzler RK, Simard J, Easton DF, Chenevix-Trench G, Antoniou AC, Ottini L, B. Consortium of Investigators of Modifiers of, Brca, Breast and Prostate Cancer Risks for Male BRCA1 and BRCA2 Pathogenic Variant Carriers Using Polygenic Risk Scores, J Natl Cancer Inst, (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- [96].Ahn J, Kibel AS, Park JY, Rebbeck TR, Rennert H, Stanford JL, Ostrander EA, Chanock S, Wang MH, Mittal RD, Isaacs WB, Platz EA, Hayes RB, Prostate cancer predisposition loci and risk of metastatic disease and prostate cancer recurrence, Clin Cancer Res, 17 (2011) 1075–1081. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [97].Gallagher DJ, Vijai J, Cronin AM, Bhatia J, Vickers AJ, Gaudet MM, Fine S, Reuter V, Scher HI, Hallden C, Dutra-Clarke A, Klein RJ, Scardino PT, Eastham JA, Lilja H, Kirchhoff T, Offit K, Susceptibility loci associated with prostate cancer progression and mortality, Clin Cancer Res, 16 (2010) 2819–2832. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [98].Fritsche LG, Ma Y, Zhang D, Salvatore M, Lee S, Zhou X, Mukherjee B, On cross-ancestry cancer polygenic risk scores, PLoS Genet, 17 (2021) e1009670. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [99].Karunamuni RA, Huynh-Le MP, Fan CC, Thompson W, Eeles RA, Kote-Jarai Z, Muir K, U. Collaborators, Lophatananon A, Tangen CM, Goodman PJ, Thompson IM Jr., Blot WJ, Zheng W, Kibel AS, Drake BF, Cussenot O, Cancel-Tassin G, Menegaux F, Truong T, Park JY, Lin HY, Bensen JT, Fontham ETH, Mohler JL, Taylor JA, Multigner L, Blanchet P, Brureau L, Romana M, Leach RJ, John EM, Fowke J, Bush WS, Aldrich M, Crawford DC, Srivastava S, Cullen JC, Petrovics G, Parent ME, Hu JJ, Sanderson M, Mills IG, Andreassen OA, Dale AM, Seibert TM, Consortium P., African-specific improvement of a polygenic hazard score for age at diagnosis of prostate cancer, Int J Cancer, 148 (2021) 99–105. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [100].Xu J, Zheng SL, Carpten JD, Nupponen NN, Robbins CM, Mestre J, Moses TY, Faith DA, Kelly BD, Isaacs SD, Wiley KE, Ewing CM, Bujnovszky P, Chang B, Bailey-Wilson J, Bleecker ER, Walsh PC, Trent JM, Meyers DA, Isaacs WB, Evaluation of linkage and association of HPC2/ELAC2 in patients with familial or sporadic prostate cancer, Am J Hum Genet, 68 (2001) 901–911. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [101].Xu J, Zheng SL, Hawkins GA, Faith DA, Kelly B, Isaacs SD, Wiley KE, Chang B, Ewing CM, Bujnovszky P, Carpten JD, Bleecker ER, Walsh PC, Trent JM, Meyers DA, Isaacs WB, Linkage and association studies of prostate cancer susceptibility: evidence for linkage at 8p22–23, Am J Hum Genet, 69 (2001) 341–350. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [102].Maier C, Herkommer K, Hoegel J, Vogel W, Paiss T, A genomewide linkage analysis for prostate cancer susceptibility genes in families from Germany, Eur J Hum Genet, 13 (2005) 352–360. [DOI] [PubMed] [Google Scholar]
- [103].Roy R, Chun J, Powell SN, BRCA1 and BRCA2: different roles in a common pathway of genome protection, Nat Rev Cancer, 12 (2011) 68–78. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [104].Nyberg T, Frost D, Barrowdale D, Evans DG, Bancroft E, Adlard J, Ahmed M, Barwell J, Brady AF, Brewer C, Cook J, Davidson R, Donaldson A, Eason J, Gregory H, Henderson A, Izatt L, Kennedy MJ, Miller C, Morrison PJ, Murray A, Ong KR, Porteous M, Pottinger C, Rogers MT, Side L, Snape K, Walker L, Tischkowitz M, Eeles R, Easton DF, Antoniou AC, Prostate Cancer Risks for Male BRCA1 and BRCA2 Mutation Carriers: A Prospective Cohort Study, Eur Urol, 77 (2020) 24–35. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [105].Wooster R, Bignell G, Lancaster J, Swift S, Seal S, Mangion J, Collins N, Gregory S, Gumbs C, Micklem G, Identification of the breast cancer susceptibility gene BRCA2, Nature, 378 (1995) 789–792. [DOI] [PubMed] [Google Scholar]
- [106].Miki Y, Swensen J, Shattuck-Eidens D, Futreal PA, Harshman K, Tavtigian S, Liu Q, Cochran C, Bennett LM, Ding W, et al. , A strong candidate for the breast and ovarian cancer susceptibility gene BRCA1, Science, 266 (1994) 66–71. [DOI] [PubMed] [Google Scholar]
- [107].Breast Cancer Linkage C, Cancer risks in BRCA2 mutation carriers, J Natl Cancer Inst, 91 (1999) 1310–1316. [DOI] [PubMed] [Google Scholar]
- [108].Castro E, Goh C, Leongamornlert D, Saunders E, Tymrakiewicz M, Dadaev T, Govindasami K, Guy M, Ellis S, Frost D, Bancroft E, Cole T, Tischkowitz M, Kennedy MJ, Eason J, Brewer C, Evans DG, Davidson R, Eccles D, Porteous ME, Douglas F, Adlard J, Donaldson A, Antoniou AC, Kote-Jarai Z, Easton DF, Olmos D, Eeles R, Effect of BRCA Mutations on Metastatic Relapse and Cause-specific Survival After Radical Treatment for Localised Prostate Cancer, Eur Urol, 68 (2015) 186–193. [DOI] [PubMed] [Google Scholar]
- [109].Castro E, Goh C, Olmos D, Saunders E, Leongamornlert D, Tymrakiewicz M, Mahmud N, Dadaev T, Govindasami K, Guy M, Sawyer E, Wilkinson R, Ardern-Jones A, Ellis S, Frost D, Peock S, Evans DG, Tischkowitz M, Cole T, Davidson R, Eccles D, Brewer C, Douglas F, Porteous ME, Donaldson A, Dorkins H, Izatt L, Cook J, Hodgson S, Kennedy MJ, Side LE, Eason J, Murray A, Antoniou AC, Easton DF, Kote-Jarai Z, Eeles R, Germline BRCA mutations are associated with higher risk of nodal involvement, distant metastasis, and poor survival outcomes in prostate cancer, J Clin Oncol, 31 (2013) 1748–1757. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [110].Gallagher DJ, Gaudet MM, Pal P, Kirchhoff T, Balistreri L, Vora K, Bhatia J, Stadler Z, Fine SW, Reuter V, Zelefsky M, Morris MJ, Scher HI, Klein RJ, Norton L, Eastham JA, Scardino PT, Robson ME, Offit K, Germline BRCA mutations denote a clinicopathologic subset of prostate cancer, Clin Cancer Res, 16 (2010) 2115–2121. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [111].Narod SA, Neuhausen S, Vichodez G, Armel S, Lynch HT, Ghadirian P, Cummings S, Olopade O, Stoppa-Lyonnet D, Couch F, Wagner T, Warner E, Foulkes WD, Saal H, Weitzel J, Tulman A, Poll A, Nam R, Sun P, Hereditary G Breast Cancer Study, Danquah J, Domchek S, Tung N, Ainsworth P, Horsman D, Kim-Sing C, Maugard C, Eisen A, Daly M, McKinnon W, Wood M, Isaacs C, Gilchrist D, Karlan B, Nedelcu R, Meschino W, Garber J, Pasini B, Manoukian S, Bellati C, Rapid progression of prostate cancer in men with a BRCA2 mutation, Br J Cancer, 99 (2008) 371–374. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [112].Page EC, Bancroft EK, Brook MN, Assel M, Hassan Al Battat M, Thomas S, Taylor N, Chamberlain A, Pope J, Raghallaigh HN, Evans DG, Rothwell J, Maehle L, Grindedal EM, James P, Mascarenhas L, McKinley J, Side L, Thomas T, van Asperen C, Vasen H, Kiemeney LA, Ringelberg J, Jensen TD, Osther PJS, Helfand BT, Genova E, Oldenburg RA, Cybulski C, Wokolorczyk D, Ong KR, Huber C, Lam J, Taylor L, Salinas M, Feliubadalo L, Oosterwijk JC, van Zelst-Stams W, Cook J, Rosario DJ, Domchek S, Powers J, Buys S, O’Toole K, Ausems M, Schmutzler RK, Rhiem K, Izatt L, Tripathi V, Teixeira MR, Cardoso M, Foulkes WD, Aprikian A, van Randeraad H, Davidson R, Longmuir M, Ruijs MWG, Helderman van den Enden A, Adank M, Williams R, Andrews L, Murphy DG, Halliday D, Walker L, Liljegren A, Carlsson S, Azzabi A, Jobson I, Morton C, Shackleton K, Snape K, Hanson H, Harris M, Tischkowitz M, Taylor A, Kirk J, Susman R, Chen-Shtoyerman R, Spigelman A, Pachter N, Ahmed M, Ramon YCT, Zgajnar J, Brewer C, Gadea N, Brady AF, van Os T, Gallagher D, Johannsson O, Donaldson A, Barwell J, Nicolai N, Friedman E, Obeid E, Greenhalgh L, Murthy V, Copakova L, Saya S, McGrath J, Cooke P, Ronlund K, Richardson K, Henderson A, Teo SH, Arun B, Kast K, Dias A, Aaronson NK, Ardern-Jones A, Bangma CH, Castro E, Dearnaley D, Eccles DM, Tricker K, Eyfjord J, Falconer A, Foster C, Gronberg H, Hamdy FC, Stefansdottir V, Khoo V, Lindeman GJ, Lubinski J, Axcrona K, Mikropoulos C, Mitra A, Moynihan C, Rennert G, Suri M, Wilson P, Dudderidge T, I.S. Collaborators, Offman J, Kote-Jarai Z, Vickers A, Lilja H, Eeles RA, Interim Results from the IMPACT Study: Evidence for Prostate-specific Antigen Screening in BRCA2 Mutation Carriers, Eur Urol, 76 (2019) 831–842. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [113].Beebe-Dimmer JL, Zuhlke KA, Johnson AM, Liesman D, Cooney KA, Rare germline mutations in African American men diagnosed with early-onset prostate cancer, Prostate, 78 (2018) 321–326. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [114].Nientiedt C, Budczies J, Endris V, Kirchner M, Schwab C, Jurcic C, Behnisch R, Hoveida S, Lantwin P, Kaczorowski A, Geisler C, Dieffenbacher S, Falkenbach F, Franke D, Gortz M, Heller M, Himmelsbach R, Pecqueux C, Rath M, Reimold P, Schutz V, Simunovic I, Walter E, Hofer L, Gasch C, Schonberg G, Pursche L, Hatiboglu G, Nyarangi-Dix J, Sultmann H, Zschabitz S, Koerber SA, Jager D, Debus J, Duensing A, Schirmacher P, Hohenfellner M, Stenzinger A, Duensing S, Mutations in TP53 or DNA damage repair genes define poor prognostic subgroups in primary prostate cancer, Urol Oncol, 40 (2022) 8 e11–18 e18. [DOI] [PubMed] [Google Scholar]
- [115].Navone NM, Troncoso P, Pisters LL, Goodrow TL, Palmer JL, Nichols WW, von Eschenbach AC, Conti CJ, p53 protein accumulation and gene mutation in the progression of human prostate carcinoma, J Natl Cancer Inst, 85 (1993) 1657–1669. [DOI] [PubMed] [Google Scholar]
- [116].Heidenberg HB, Sesterhenn IA, Gaddipati JP, Weghorst CM, Buzard GS, Moul JW, Srivastava S, Alteration of the tumor suppressor gene p53 in a high fraction of hormone refractory prostate cancer, J Urol, 154 (1995) 414–421. [DOI] [PubMed] [Google Scholar]
- [117].Nientiedt C, Endris V, Jenzer M, Mansour J, Sedehi NTP, Pecqueux C, Volckmar AL, Leichsenring J, Neumann O, Kirchner M, Hoveida S, Lantwin P, Kaltenecker K, Dieffenbacher S, Gasch C, Hofer L, Franke D, Tosev G, Gortz M, Schutz V, Radtke JP, Nyarangi-Dix J, Hatiboglu G, Simpfendorfer T, Schonberg G, Isaac S, Teber D, Koerber SA, Christofi G, Czink E, Kreuter R, Apostolidis L, Kratochwil C, Giesel F, Haberkorn U, Debus J, Sultmann H, Zschabitz S, Jager D, Duensing A, Schirmacher P, Grullich C, Hohenfellner M, Stenzinger A, Duensing S, High prevalence of DNA damage repair gene defects and TP53 alterations in men with treatment-naive metastatic prostate cancer -Results from a prospective pilot study using a 37 gene panel, Urol Oncol, 38 (2020) 637 e617–637 e627. [DOI] [PubMed] [Google Scholar]
- [118].Hamid AA, Gray KP, Shaw G, MacConaill LE, Evan C, Bernard B, Loda M, Corcoran NM, Van Allen EM, Choudhury AD, Sweeney CJ, Compound Genomic Alterations of TP53, PTEN, and RB1 Tumor Suppressors in Localized and Metastatic Prostate Cancer, Eur Urol, 76 (2019) 89–97. [DOI] [PubMed] [Google Scholar]
- [119].Mateo J, Seed G, Bertan C, Rescigno P, Dolling D, Figueiredo I, Miranda S, Nava Rodrigues D, Gurel B, Clarke M, Atkin M, Chandler R, Messina C, Sumanasuriya S, Bianchini D, Barrero M, Petermolo A, Zafeiriou Z, Fontes M, Perez-Lopez R, Tunariu N, Fulton B, Jones R, McGovern U, Ralph C, Varughese M, Parikh O, Jain S, Elliott T, Sandhu S, Porta N, Hall E, Yuan W, Carreira S, de Bono JS, Genomics of lethal prostate cancer at diagnosis and castration resistance, J Clin Invest, 130 (2020) 1743–1751. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [120].Maxwell KN, Cheng HH, Powers J, Gulati R, Ledet EM, Morrison C, Le A, Hausler R, Stopfer J, Hyman S, Kohlmann W, Naumer A, Vagher J, Greenberg SE, Naylor L, Laurino M, Konnick EQ, Shirts BH, AlDubayan SH, Van Allen EM, Nguyen B, Vijai J, Abida W, Carlo MI, Dubard-Gault M, Lee DJ, Maese LD, Mandelker D, Montgomery B, Morris MJ, Nicolosi P, Nussbaum RL, Schwartz LE, Stadler Z, Garber JE, Offit K, Schiffman JD, Nelson PS, Sartor O, Walsh MF, Pritchard CC, Inherited TP53 Variants and Risk of Prostate Cancer, Eur Urol, 81 (2022) 243–250. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [121].Wei J, Shi Z, Na R, Wang CH, Resurreccion WK, Zheng SL, Hulick PJ, Cooney KA, Helfand BT, Isaacs WB, Xu J, Germline HOXB13 G84E mutation carriers and risk to twenty common types of cancer: results from the UK Biobank, Br J Cancer, 123 (2020) 1356–1359. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [122].Xu J, Lange EM, Lu L, Zheng SL, Wang Z, Thibodeau SN, Cannon-Albright LA, Teerlink CC, Camp NJ, Johnson AM, Zuhlke KA, Stanford JL, Ostrander EA, Wiley KE, Isaacs SD, Walsh PC, Maier C, Luedeke M, Vogel W, Schleutker J, Wahlfors T, Tammela T, Schaid D, McDonnell SK, DeRycke MS, Cancel-Tassin G, Cussenot O, Wiklund F, Gronberg H, Eeles R, Easton D, Kote-Jarai Z, Whittemore AS, Hsieh CL, Giles GG, Hopper JL, Severi G, Catalona WJ, Mandal D, Ledet E, Foulkes WD, Hamel N, Mahle L, Moller P, Powell I, Bailey-Wilson JE, Carpten JD, Seminara D, Cooney KA, Isaacs WB, International G Consortium for Prostate Cancer, HOXB13 is a susceptibility gene for prostate cancer: results from the International Consortium for Prostate Cancer Genetics (ICPCG), Hum Genet, 132 (2013) 5–14. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [123].Beebe-Dimmer JL, Hathcock M, Yee C, Okoth LA, Ewing CM, Isaacs WB, Cooney KA, Thibodeau SN, The HOXB13 G84E Mutation Is Associated with an Increased Risk for Prostate Cancer and Other Malignancies, Cancer Epidemiol Biomarkers Prev, 24 (2015) 1366–1372. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [124].Shang Z, Zhu S, Zhang H, Li L, Niu Y, Germline homeobox B13 (HOXB13) G84E mutation and prostate cancer risk in European descendants: a meta-analysis of 24,213 cases and 73, 631 controls, Eur Urol, 64 (2013) 173–176. [DOI] [PubMed] [Google Scholar]
- [125].Storebjerg TM, Hoyer S, Kirkegaard P, Bro F, LuCamp Study G, Orntoft TF, Borre M, Sorensen KD, Prevalence of the HOXB13 G84E mutation in Danish men undergoing radical prostatectomy and its correlations with prostate cancer risk and aggressiveness, BJU Int, 118 (2016) 646–653. [DOI] [PubMed] [Google Scholar]
- [126].Kote-Jarai Z, Mikropoulos C, Leongamornlert DA, Dadaev T, Tymrakiewicz M, Saunders EJ, Jones M, Jugurnauth-Little S, Govindasami K, Guy M, Hamdy FC, Donovan JL, Neal DE, Lane JA, Dearnaley D, Wilkinson RA, Sawyer EJ, Morgan A, Antoniou AC, Eeles RA, U.K.G.P.C.S. Collaborators, the Protec TSG, Prevalence of the HOXB13 G84E germline mutation in British men and correlation with prostate cancer risk, tumour characteristics and clinical outcomes, Ann Oncol, 26 (2015) 756–761. [DOI] [PubMed] [Google Scholar]
- [127].Economides KD, Capecchi MR, Hoxb13 is required for normal differentiation and secretory function of the ventral prostate, Development, 130 (2003) 2061–2069. [DOI] [PubMed] [Google Scholar]
- [128].Kim YR, Oh KJ, Park RY, Xuan NT, Kang TW, Kwon DD, Choi C, Kim MS, Nam KI, Ahn KY, Jung C, HOXB13 promotes androgen independent growth of LNCaP prostate cancer cells by the activation of E2F signaling, Mol Cancer, 9 (2010) 124. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [129].Bhatia-Gaur R, Donjacour AA, Sciavolino PJ, Kim M, Desai N, Young P, Norton CR, Gridley T, Cardiff RD, Cunha GR, Abate-Shen C, Shen MM, Roles for Nkx3.1 in prostate development and cancer, Genes Dev, 13 (1999) 966–977. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [130].Akamatsu S, Takata R, Ashikawa K, Hosono N, Kamatani N, Fujioka T, Ogawa O, Kubo M, Nakamura Y, Nakagawa H, A functional variant in NKX3.1 associated with prostate cancer susceptibility down-regulates NKX3.1 expression, Hum Mol Genet, 19 (2010) 4265–4272. [DOI] [PubMed] [Google Scholar]
- [131].Barnabas N, Xu L, Savera A, Hou Z, Barrack ER, Chromosome 8 markers of metastatic prostate cancer in African American men: gain of the MIR151 gene and loss of the NKX3–1 gene, Prostate, 71 (2011) 857–871. [DOI] [PubMed] [Google Scholar]
- [132].Papachristodoulou A, Rodriguez-Calero A, Panja S, Margolskee E, Virk RK, Milner TA, Martina LP, Kim JY, Di Bernardo M, Williams AB, Maliza EA, Caputo JM, Haas C, Wang V, De Castro GJ, Wenske S, Hibshoosh H, McKiernan JM, Shen MM, Rubin MA, Mitrofanova A, Dutta A, Abate-Shen C, NKX3.1 Localization to Mitochondria Suppresses Prostate Cancer Initiation, Cancer Discov, 11 (2021) 2316–2333. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [133].Mottet N, van den Bergh RCN, Briers E, Van den Broeck T, Cumberbatch MG, De Santis M, Fanti S, Fossati N, Gandaglia G, Gillessen S, Grivas N, Grummet J, Henry AM, van der Kwast TH, Lam TB, Lardas M, Liew M, Mason MD, Moris L, Oprea-Lager DE, van der Poel HG, Rouviere O, Schoots IG, Tilki D, Wiegel T, Willemse PM, Cornford P, EAU-EANM-ESTRO-ESUR-SIOG Guidelines on Prostate Cancer-2020 Update. Part 1: Screening, Diagnosis, and Local Treatment with Curative Intent, Eur Urol, 79 (2021) 243–262. [DOI] [PubMed] [Google Scholar]
- [134].Teo MY, Rathkopf DE, Kantoff P, Treatment of Advanced Prostate Cancer, Annu Rev Med, 70 (2019) 479–499. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [135].Van der Eecken K, Vanwelkenhuyzen J, Deek MP, Tran PT, Warner E, Wyatt AW, Kwan EM, Verbeke S, Van Dorpe J, Fonteyne V, Lumen N, De Laere B, Ost P, Tissue- and Blood-derived Genomic Biomarkers for Metastatic Hormone-sensitive Prostate Cancer: A Systematic Review, Eur Urol Oncol, 4 (2021) 914–923. [DOI] [PubMed] [Google Scholar]
- [136].Deek MP, Van der Eecken K, Phillips R, Parikh NR, Isaacsson Velho P, Lotan TL, Kishan AU, Maurer T, Consortium G.A.P., Boutros PC, Hovens C, Abramowtiz M, Pollack A, Desai N, Stish B, Feng FY, Eisenberger M, Carducci M, Pienta KJ, Markowski M, Paller CJ, Antonarakis ES, Berlin A, Ost P, Tran PT, The Mutational Landscape of Metastatic Castration-sensitive Prostate Cancer: The Spectrum Theory Revisited, Eur Urol, 80 (2021) 632–640. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [137].Stopsack KH, Nandakumar S, Wibmer AG, Haywood S, Weg ES, Barnett ES, Kim CJ, Carbone EA, Vasselman SE, Nguyen B, Hullings MA, Scher HI, Morris MJ, Solit DB, Schultz N, Kantoff PW, Abida W, Oncogenic Genomic Alterations, Clinical Phenotypes, and Outcomes in Metastatic Castration-Sensitive Prostate Cancer, Clin Cancer Res, 26 (2020) 3230–3238. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [138].Fraser M, Livingstone J, Wrana JL, Finelli A, He HH, van der Kwast T, Zlotta AR, Bristow RG, Boutros PC, Somatic driver mutation prevalence in 1844 prostate cancers identifies ZNRF3 loss as a predictor of metastatic relapse, Nat Commun, 12 (2021) 6248. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [139].Abida W, Armenia J, Gopalan A, Brennan R, Walsh M, Barron D, Danila D, Rathkopf D, Morris M, Slovin S, McLaughlin B, Curtis K, Hyman DM, Durack JC, Solomon SB, Arcila ME, Zehir A, Syed A, Gao J, Chakravarty D, Vargas HA, Robson ME, Joseph V, Offit K, Donoghue MTA, Abeshouse AA, Kundra R, Heins ZJ, Penson AV, Harris C, Taylor BS, Ladanyi M, Mandelker D, Zhang L, Reuter VE, Kantoff PW, Solit DB, Berger MF, Sawyers CL, Schultz N, Scher HI, Prospective Genomic Profiling of Prostate Cancer Across Disease States Reveals Germline and Somatic Alterations That May Affect Clinical Decision Making, JCO Precis Oncol, 2017 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- [140].Kohli M, Tan W, Zheng T, Wang A, Montesinos C, Wong C, Du P, Jia S, Yadav S, Horvath LG, Mahon KL, Kwan EM, Fettke H, Yu J, Azad AA, Clinical and genomic insights into circulating tumor DNA-based alterations across the spectrum of metastatic hormone-sensitive and castrate-resistant prostate cancer, EBioMedicine, 54 (2020) 102728. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [141].Abida W, Cyrta J, Heller G, Prandi D, Armenia J, Coleman I, Cieslik M, Benelli M, Robinson D, Van Allen EM, Sboner A, Fedrizzi T, Mosquera JM, Robinson BD, De Sarkar N, Kunju LP, Tomlins S, Wu YM, Nava Rodrigues D, Loda M, Gopalan A, Reuter VE, Pritchard CC, Mateo J, Bianchini D, Miranda S, Carreira S, Rescigno P, Filipenko J, Vinson J, Montgomery RB, Beltran H, Heath EI, Scher HI, Kantoff PW, Taplin ME, Schultz N, deBono JS, Demichelis F, Nelson PS, Rubin MA, Chinnaiyan AM, Sawyers CL, Genomic correlates of clinical outcome in advanced prostate cancer, Proc Natl Acad Sci U S A, 116 (2019) 11428–11436. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [142].Kalsbeek AM, Chan EF, Grogan J, Petersen DC, Jaratlerdsiri W, Gupta R, Lyons RJ, Haynes AM, Horvath LG, Kench JG, Stricker PD, Hayes VM, Mutational load of the mitochondrial genome predicts pathological features and biochemical recurrence in prostate cancer, Aging (Albany NY), 8 (2016) 2702–2712. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [143].Petros JA, Baumann AK, Ruiz-Pesini E, Amin MB, Sun CQ, Hall J, Lim S, Issa MM, Flanders WD, Hosseini SH, Marshall FF, Wallace DC, mtDNA mutations increase tumorigenicity in prostate cancer, Proc Natl Acad Sci U S A, 102 (2005) 719–724. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [144].Booker LM, Habermacher GM, Jessie BC, Sun QC, Baumann AK, Amin M, Lim SD, Fernandez-Golarz C, Lyles RH, Brown MD, Marshall FF, Petros JA, North American white mitochondrial haplogroups in prostate and renal cancer, J Urol, 175 (2006) 468–472; discussion 472–463. [DOI] [PubMed] [Google Scholar]
- [145].Koochekpour S, Marlowe T, Singh KK, Attwood K, Chandra D, Reduced mitochondrial DNA content associates with poor prognosis of prostate cancer in African American men, PLoS One, 8 (2013) e74688. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [146].McCrow JP, Petersen DC, Louw M, Chan EK, Harmeyer K, Vecchiarelli S, Lyons RJ, Bornman MS, Hayes VM, Spectrum of mitochondrial genomic variation and associated clinical presentation of prostate cancer in South African men, Prostate, 76 (2016) 349–358. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [147].Lindberg J, Mills IG, Klevebring D, Liu W, Neiman M, Xu J, Wikstrom P, Wiklund P, Wiklund F, Egevad L, Gronberg H, The mitochondrial and autosomal mutation landscapes of prostate cancer, Eur Urol, 63 (2013) 702–708. [DOI] [PubMed] [Google Scholar]
- [148].Kalsbeek AMF, Chan EKF, Corcoran NM, Hovens CM, Hayes VM, Mitochondrial genome variation and prostate cancer: a review of the mutational landscape and application to clinical management, Oncotarget, 8 (2017) 71342–71357. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [149].Hopkins JF, Sabelnykova VY, Weischenfeldt J, Simon R, Aguiar JA, Alkallas R, Heisler LE, Zhang J, Watson JD, Chua MLK, Fraser M, Favero F, Lawerenz C, Plass C, Sauter G, McPherson JD, van der Kwast T, Korbel J, Schlomm T, Bristow RG, Boutros PC, Mitochondrial mutations drive prostate cancer aggression, Nat Commun, 8 (2017) 656. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [150].Lopez-Otin C, Blasco MA, Partridge L, Serrano M, Kroemer G, The hallmarks of aging, Cell, 153 (2013) 1194–1217. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [151].Schopf B, Weissensteiner H, Schafer G, Fazzini F, Charoentong P, Naschberger A, Rupp B, Fendt L, Bukur V, Giese I, Sorn P, Sant’Anna-Silva AC, Iglesias-Gonzalez J, Sahin U, Kronenberg F, Gnaiger E, Klocker H, OXPHOS remodeling in high-grade prostate cancer involves mtDNA mutations and increased succinate oxidation, Nat Commun, 11 (2020) 1487. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [152].Arnold RS, Sun Q, Sun CQ, Richards JC, O’Hearn S, Osunkoya AO, Wallace DC, Petros JA, An inherited heteroplasmic mutation in mitochondrial gene COI in a patient with prostate cancer alters reactive oxygen, reactive nitrogen and proliferation, Biomed Res Int, 2013 (2013) 239257. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [153].Roobol MJ, Steyerberg EW, Kranse R, Wolters T, van den Bergh RC, Bangma CH, Schroder FH, A risk-based strategy improves prostate-specific antigen-driven detection of prostate cancer, Eur Urol, 57 (2010) 79–85. [DOI] [PubMed] [Google Scholar]
- [154].Mohler JL, Antonarakis ES, Armstrong AJ, D’Amico AV, Davis BJ, Dorff T, Eastham JA, Enke CA, Farrington TA, Higano CS, Horwitz EM, Hurwitz M, Ippolito JE, Kane CJ, Kuettel MR, Lang JM, McKenney J, Netto G, Penson DF, Plimack ER, Pow-Sang JM, Pugh TJ, Richey S, Roach M, Rosenfeld S, Schaeffer E, Shabsigh A, Small EJ, Spratt DE, Srinivas S, Tward J, Shead DA, Freedman-Cass DA, Prostate Cancer, Version 2.2019, NCCN Clinical Practice Guidelines in Oncology, J Natl Compr Canc Netw, 17 (2019) 479–505. [DOI] [PubMed] [Google Scholar]
- [155].Giri VN, Knudsen KE, Kelly WK, Cheng HH, Cooney KA, Cookson MS, Dahut W, Weissman S, Soule HR, Petrylak DP, Dicker AP, AlDubayan SH, Toland AE, Pritchard CC, Pettaway CA, Daly MB, Mohler JL, Parsons JK, Carroll PR, Pilarski R, Blanco A, Woodson A, Rahm A, Taplin ME, Polascik TJ, Helfand BT, Hyatt C, Morgans AK, Feng F, Mullane M, Powers J, Concepcion R, Lin DW, Wender R, Mark JR, Costello A, Burnett AL, Sartor O, Isaacs WB, Xu J, Weitzel J, Andriole GL, Beltran H, Briganti A, Byrne L, Calvaresi A, Chandrasekar T, Chen DYT, Den RB, Dobi A, Crawford ED, Eastham J, Eggener S, Freedman ML, Garnick M, Gomella PT, Handley N, Hurwitz MD, Izes J, Karnes RJ, Lallas C, Languino L, Loeb S, Lopez AM, Loughlin KR, Lu-Yao G, Malkowicz SB, Mann M, Mille P, Miner MM, Morgan T, Moreno J, Mucci L, Myers RE, Nielsen SM, O’Neil B, Pinover W, Pinto P, Poage W, Raj GV, Rebbeck TR, Ryan C, Sandler H, Schiewer M, Scott EMD, Szymaniak B, Tester W, Trabulsi EJ, Vapiwala N, Yu EY, Zeigler-Johnson C, Gomella LG, Implementation of Germline Testing for Prostate Cancer: Philadelphia Prostate Cancer Consensus Conference 2019, J Clin Oncol, 38 (2020) 2798–2811. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [156].Isaacsson Velho P, Silberstein JL, Markowski MC, Luo J, Lotan TL, Isaacs WB, Antonarakis ES, Intraductal/ductal histology and lymphovascular invasion are associated with germline DNA-repair gene mutations in prostate cancer, Prostate, 78 (2018) 401–407. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [157].Brooks MA, Thomas L, Magi-Galluzzi C, Li J, Crager MR, Lu R, Abran J, Aboushwareb T, Klein EA, GPS Assay Association With Long-Term Cancer Outcomes: Twenty-Year Risk of Distant Metastasis and Prostate Cancer-Specific Mortality, JCO Precis Oncol, 5 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- [158].Herlemann A, Huang HC, Alam R, Tosoian JJ, Kim HL, Klein EA, Simko JP, Chan JM, Lane BR, Davis JW, Davicioni E, Feng FY, McCue P, Kim H, Den RB, Bismar TA, Carroll PR, Cooperberg MR, Decipher identifies men with otherwise clinically favorable-intermediate risk disease who may not be good candidates for active surveillance, Prostate Cancer Prostatic Dis, 23 (2020) 136–143. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [159].Kim HL, Li P, Huang HC, Deheshi S, Marti T, Knudsen B, Abou-Ouf H, Alam R, Lotan TL, Lam LLC, du Plessis M, Davicioni E, Fleshner N, Lane BR, Ross AE, Davis JW, Mohler JL, Trock BJ, Klein EA, Tosoian JJ, Hyndman ME, Bismar TA, Validation of the Decipher Test for predicting adverse pathology in candidates for prostate cancer active surveillance, Prostate Cancer Prostatic Dis, 22 (2019) 399–405. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [160].Klotz L, Active surveillance: patient selection, Curr Opin Urol, 23 (2013) 239–244. [DOI] [PubMed] [Google Scholar]
- [161].Musquera M, Fleshner NE, Finelli A, Zlotta AR, The REDUCE trial: chemoprevention in prostate cancer using a dual 5alpha-reductase inhibitor, dutasteride, Expert Rev Anticancer Ther, 8 (2008) 1073–1079. [DOI] [PubMed] [Google Scholar]
- [162].Thompson IM, Goodman PJ, Tangen CM, Lucia MS, Miller GJ, Ford LG, Lieber MM, Cespedes RD, Atkins JN, Lippman SM, Carlin SM, Ryan A, Szczepanek CM, Crowley JJ, Coltman CA Jr., The influence of finasteride on the development of prostate cancer, N Engl J Med, 349 (2003) 215–224. [DOI] [PubMed] [Google Scholar]
- [163].Thompson IM Jr., Goodman PJ, Tangen CM, Parnes HL, Minasian LM, Godley PA, Lucia MS, Ford LG, Long-term survival of participants in the prostate cancer prevention trial, N Engl J Med, 369 (2013) 603–610. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [164].Pircher A, Zieher M, Eigentler A, Pichler R, Schafer G, Fritz J, Puhr M, Steiner E, Horninger W, Klocker H, Heidegger I, Antidiabetic drugs influence molecular mechanisms in prostate cancer, Cancer Biol Ther, 19 (2018) 1153–1161. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [165].Patel T, Hruby G, Badani K, Abate-Shen C, McKiernan JM, Clinical outcomes after radical prostatectomy in diabetic patients treated with metformin, Urology, 76 (2010) 1240–1244. [DOI] [PubMed] [Google Scholar]
- [166].Algire C, Moiseeva O, Deschenes-Simard X, Amrein L, Petruccelli L, Birman E, Viollet B, Ferbeyre G, Pollak MN, Metformin reduces endogenous reactive oxygen species and associated DNA damage, Cancer Prev Res (Phila), 5 (2012) 536–543. [DOI] [PubMed] [Google Scholar]
- [167].Brajtbord JS, Leapman MS, Cooperberg MR, The CAPRA Score at 10 Years: Contemporary Perspectives and Analysis of Supporting Studies, Eur Urol, 71 (2017) 705–709. [DOI] [PubMed] [Google Scholar]
- [168].Pompe RS, Bandini M, Preisser F, Marchioni M, Zaffuto E, Tian Z, Salomon G, Schlomm T, Huland H, Graefen M, Tilki D, Shariat SF, Karakiewicz PI, Contemporary approach to predict early biochemical recurrence after radical prostatectomy: update of the Walz nomogram, Prostate Cancer Prostatic Dis, 21 (2018) 386–393. [DOI] [PubMed] [Google Scholar]
- [169].Cullen J, Rosner IL, Brand TC, Zhang N, Tsiatis AC, Moncur J, Ali A, Chen Y, Knezevic D, Maddala T, Lawrence HJ, Febbo PG, Srivastava S, Sesterhenn IA, McLeod DG, A Biopsy-based 17-gene Genomic Prostate Score Predicts Recurrence After Radical Prostatectomy and Adverse Surgical Pathology in a Racially Diverse Population of Men with Clinically Low- and Intermediate-risk Prostate Cancer, Eur Urol, 68 (2015) 123–131. [DOI] [PubMed] [Google Scholar]
- [170].de Bono J, Mateo J, Fizazi K, Saad F, Shore N, Sandhu S, Chi KN, Sartor O, Agarwal N, Olmos D, Thiery-Vuillemin A, Twardowski P, Mehra N, Goessl C, Kang J, Burgents J, Wu W, Kohlmann A, Adelman CA, Hussain M, Olaparib for Metastatic Castration-Resistant Prostate Cancer, N Engl J Med, 382 (2020) 2091–2102. [DOI] [PubMed] [Google Scholar]
