Background: High-fidelity repair of DNA damage repair (DDR) (either single-strand- [SSBs] or double-strand breaks [DSBs]) is necessary for maintaining genomic integrity and cell survival. DDR alterations are commonly found in genitourinary malignancies involving either DSB repair by the homologous recombination (HR) repair (HRR) system (BRCA1/2 pathway) or the SSB repair through the poly (ADP-ribose) polymerase (PARP) pathway. PARP inhibitors (PARPi) exploit defects in the DNA repair pathway through synthetic lethality, DSBs being repaired only in HR-proficient cells but not in HR-deficient (HRD) cells. Summary: A growing body of evidence supports the need for identification of germinal and somatic DDR alterations in patients with genitourinary malignancies. PARPi have already shown significant survival benefits in patients harboring HRR mutations in advanced settings, paving the way for precision medicine. Key Messages: In advanced prostate cancer (PCa), somatic mutations in HRR pathway are observed in up to 27% of metastatic resistant-to-castration PCa (mCRPC), although occurring early in PCa development, and mainly involving BRCA2, ATM, CHEK2, and BRCA1. Overall, germinal alterations are present in roughly 30–50% of cases of HRR alterations, and relative risk of PCa in germinal BRCA2 alteration carriers is 4.65-fold higher compared to noncarriers. Determination of DDR gene status is recommended in metastatic patients, a fortiori in mCRPC setting, since it could be a putative biomarker of response to first line of treatment (androgen-receptor signaling inhibitors [ARSI] vs. taxane-based chemotherapy) and allows to assess eligibility for PARPi use. Thus, olaparib (combined with androgen deprivation therapy) recently improved overall survival in mCRPC HRD patients, after new hormonal therapy (NHT) and led to its approvement for patients with an alteration in 14 of 15 prespecified HRR genes. Moreover, since preclinical data suggested synergic action between PARPi and ARSI, the use of either olaparib or niraparib has also been proposed in combination with NHT, with a radiological progression-free survival improvement when used with abiraterone. In urothelial carcinoma, a DDR gene alteration is identified in 23–54% of patients mostly in muscle-invasive bladder cancer, with a strong association between DDR gene mutation and a higher tumor mutation burden and sensitivity to cisplatin-based chemotherapy and immunotherapy. Recent phase 2 trials supported the use of HRR status to select patients for PARPi treatment in advanced urothelial carcinoma. Finally, in renal cell carcinomas (RCCs), pathogenic germline variants in DDR genes were identified in 7.3% of the cases, and deleterious somatic alterations have also been described as recurrent genomic events in patients with advanced RCC.

Naturally occurring DNA damage arises continuously during cell life, leading to single-strand DNA breaks (SSBs) or DNA double-strand breaks (DSBs). High-fidelity and timely repair of SSBs and DSBs is therefore necessary for maintaining genomic integrity and cell survival by minimizing mutagenesis or improper apoptosis, which may otherwise lead to carcinogenesis [1]. Thus, DNA damage repair (DDR) alterations are commonly identified in various malignancies, including genitourinary cancers. We aimed to describe herein the known evidence of DDR alterations in prostate cancer (PCa), urothelial, and renal cell carcinoma (RCC) and its therapeutic implications.

Reparation System of DSBs and Homologous Recombination Repair

DSBs are a highly toxic form of DNA damage, arising from intrinsic and extrinsic sources [2]. Eukaryotes are equipped with several mechanisms to repair DSBs, including nonhomologous end-joining, microhomology-mediated end-joining, or homologous recombination (HR) repair (HRR), among others [3]. HR is a tightly regulated molecular mechanism essential for maintaining genomic stability by repairing DSBs or lesions that stall DNA replication forks. Its main pathway involves serine/threonine kinase ATM which activates several targets, including BRCA1, through CHK2 protein [4]. BRCA2 and RAD51 will form a complex with PALB2 after activation by BRCA1, subsequently leading to G2-M checkpoint arrest [5, 6]. The role of those proteins can be divided between sensors of the DSBs, such as ATM or CHK2, and effectors of the reparation, such as BRCA1/2 or PALB2.

In HR-proficient cells, broken DNA is accurately restored by the use of the respective sequence from second chromosome during the cell cycle phases of late S to G2. On the contrary, DSBs occurring in HR-deficient (HRD) cells lead to cell cycle arrest and cell death.

Reparation System of SSBs and Role of PARP and PARP Inhibitors

SSBs repair relies on base excision repair (BER) through poly (ADP-ribose) polymerase (PARP) enzyme, nucleic acid excision repair through xeroderma pigmentosum enzyme and polymerases, and mismatch repair (MMR) through MLH1 and MSH2. PARP also plays a role in transcriptional regulation of various genes, including androgen receptors (ARs) [7].

PARP inhibitors (PARPi) are targeted therapies inducing both a catalytic inhibition of PARP enzyme and a trapping of PARP1 and 2 at the same time, avoiding SSB reparation by preventing BER enzyme to access DNA. The trapping efficiency differs between the different PARPi, talazoparib having the greater stereospecific PARP-DNA trapping ability than other PARPi [8].

PARPi exploit defects in the DNA repair pathway through synthetic lethality. Indeed, if PARylation is inhibited and the BER pathway is impaired, SSBs persist and cause stalled replication forks and eventually DSBs. At this step, DSBs will be repaired with the HRR system in HR-proficient but not HRD cells [9].

Prevalence of Germinal and Somatic Mutations in Localized and Metastatic Settings

Castro et al. [10] and others have described and characterized BRCA2 as an important prognostic factor for aggressive PCa; however, the mutation frequency was low, and most estimates suggested that BRCA2 accounted for a very small fraction of PCa (1–2%), even when early onset family history-positive cases were examined [11, 12]. Of note, familial forms of PCa are observed in 20% of the cases. In a seminal paper, Robinson et al. [13] identified mutations in three DNA repair genes, BRCA1/2 and ATM, at a surprisingly high rate in men unselected for age at diagnosis or family history, but rather for aggressive disease.

More recently, Pritchard et al. [13, 14] demonstrated an elevated rate of mutations (11%) in a number of DNA repair genes in men with metastatic PCa (with 5.3% mutations of BRCA2), with more than 100 genetic pathogenic variants identified. In the most advanced setting of metastatic castration-resistant PCa (mCRPC), mutations in HRR pathway are observed in up to 27% of mCRPC, mainly involving BRCA2 (12–18%), ATM (3–6%), CHEK2 (2–5%), and BRCA1 (<2%). Overall, germinal alterations are present in roughly 30–50% of cases of HRR alterations [14, 15] with reported discrepancies due to the existence of bi-allelic somatic alterations, and the lack of constitutional testing in most studies.

Epidemiology of PCa in Carriers of DDR Mutations and Impact on Screening

Relative risk (RR) of PCa in germinal BRCA2 alteration carriers is 4.65-fold higher compared to noncarriers, with an RR of 2.5–8.6 [16]. These alterations are associated with higher stage and grade at diagnosis and poorer cancer-specific survival when BRCA2 mutation is located outside the ovarian cancer cluster region [17‒19]. Risk for PCa is also slightly higher in BRCA1 alteration carriers as compared with noncarriers (RR 1.35) but without adverse features.

These results led to recommend digital rectal examination along with PSA testing from the age of 40 years old for patients with BRCA2 mutation in the French Guidelines [20], based on the results of the IMPACT study [21]. Nevertheless, as shown by Shuai Li et al. [22], cumulative risk for PCa in BRCA2 mutation carriers is mainly driven by the overall incidence of PCa in elderly, suggesting that early screening at 40 years old is not supported by robust data. Of note, BRCA2 carriers were at increased risk of other cancers such as male breast (RR 44 vs. noncarriers), stomach (RR 6.39), and pancreatic (RR 3.34) cancers. In the NCCN guidelines updates, genetic counseling is recommended in patients with aggressive disease (≥T3a, intraductal, ≥Gleason 8, positive lymph nodes), Ashkenazi Jewish Ancestry, ≥2 relatives with cancer in the spectrum of hereditary breast-ovarian cancer or Lynch syndrome.

DDR Mutations in Localized PCa: Therapeutic Implications

Intensification of curative options, such as extended androgen deprivation therapy, in localized PCa with DDR mutations is not supported by any robust evidence to date, although BRCA mutation has been confirmed as an independent prognostic factor for metastasis-free survival (HR 2.36) and cancer-specific survival (HR 2.17) [23]. To our knowledge, no phase 3 study has assessed efficacy of PARPi in localized PCa in combination with surgery, radiation therapy, and androgen deprivation therapy.

If active surveillance is proposed to BRCA2 alteration carriers with low-risk PCa, the patient should be well informed that no clear guidelines of specific follow-up in this setting are available and that recent publications found a higher risk of grade reclassification in progressors [24]. These recent data suggest that the natural history of BRCA2-mutated PCa is probably accelerated even in the localized setting.

DDR Mutations in Advanced Settings (Metastatic Hormone-Sensitive PCa and mCRPC): Therapeutic Implications

Somatic HRR alterations seem to occur early in PCa development, with genetic alterations being stable over time [25, 26]. Therefore, HRR mutations may be detected in archived primary tumor biopsies or specimen without the need to repeat testing on metastatic tissue samples.

The ESMO provided guidance for precision medicine, and every patient with metastatic PCa should be at least tested for somatic alterations of BRCA1/2 [27]. The NCCN recommends searching for somatic alterations of BRCA1/2, ATM, PALB2, FANCA, RAD51D, CHEK2, and CDK12 for metastatic PCa and microsatellite instability for mCRPC (www.nccn.org). Mutations should be sought using NGS preferentially with large panel, since recent data suggest that not only BRCA1/2 mutations (i.e., PALB2 loss) can confer a PARPi sensitivity.

Determination of DDR gene status in mCRPC patients has been proposed to select first-line treatment options (AR pathway vs. taxane-based chemotherapy) with discrepant results. Thus, in PROREPAIR-B study, germinal BRCA2 mutations had a deleterious impact on mCRPC outcomes that may have been affected by the first line of treatment (with a shorter survival when using docetaxel as first-line therapy before AR pathway inhibitors) [17]. This benefit was also confirmed by Antonarakis et al. [28]. In the opposite, Annala et al. [29] reported shorter progression-free survival in 22 germline DDR carriers versus noncarriers (3.3 vs. 6.2 months, respectively) when treated with first-line AR pathway inhibitors. The multicenter BRCA2MEN study is currently planned to validate the role of BRCA2 as a predictive biomarker to select the first-line therapy in patients with mCRPC.

Another utility of HRR mutation testing is to assess eligibility for PARPi in mCRPC settings. Thus, PARPi recently improved overall survival (OS) in mCRPC HRD patients, after new hormonal therapy (NHT) [30], and four PARPi are currently in late development in PCa: olaparib, rucaparib, niraparib, and talazoparib (Tables 1, 2).

Table 1.

Trials using single-agent PARPi in mCRPC

 Trials using single-agent PARPi in mCRPC
 Trials using single-agent PARPi in mCRPC
Table 2.

Phase 3 trials using PARPi in combination with other drugs in mCRPC

 Phase 3 trials using PARPi in combination with other drugs in mCRPC
 Phase 3 trials using PARPi in combination with other drugs in mCRPC

Olaparib was the first one to show an OS improvement [30]. The first positive signals arose from the phase II TOPARP-A study which enrolled 50 pretreated mCRPC patients to receive olaparib [31]. Eighty-eight percent of the HR-altered patients (14/16 patients) had a composite response (objective radiological response, or a biochemical response, or a decrease in the circulating tumor cell), compared to only 6% for the HR-efficient patients. Based on these results, 98 HRD patients with mCRPC were enrolled in TOPARP-B [32]. A preplanned analysis subgroup showed an important objective radiological response for BRCA1/2 patients (11/21, 52.4%) and PALB2 patients (2/6, 33.3%), but not ATM patients (5.3%) [32].

In parallel, the study PROfound compared, in a phase 3 trial, the efficacy of olaparib to enzalutamide or abiraterone in patients with mCRPC previously treated with at least one NHT [30, 33] +/− taxanes (65% of the patients), with patients in the control arm receiving another NHT [30]. The study was positive in the primary endpoint (radiographic progression-free survival [rPFS]) and in OS, while 67% of the patients in the control arm crossed over (Table 1). Given these results, olaparib was approved by the Food and Drug Administration (FDA) for patients with an alteration in 14 of the 15 prespecified HRR genes. Indeed, preclinical data, generated after the PROfound design, suggest that PPP2R2A loss of function does not confer sensitivity to PARPi and was then withdrawn from the FDA list [33]. The European Medicines Agency was more conservative and gave its approval only for BRCA1/2, given concern for a lower benefit regarding ATM. Other studies using rucaparib, niraparib, and talazoparib are presented in Table 1 [34‒36].

Preclinical data suggest synergic action between olaparib and androgen pathway inhibitors: AR signaling regulates DNA repair, thus inducing HRR deficiency and conferring a BRCAness state, and PARPi may increase activity of NHT via AR-dependent transcription [37]. Thus, use of PARPi has also been proposed in combination with NHT. A phase 2 trial showed encouraging results in terms of radiographic progression-free survival (rPFS) (HR 0.65) with olaparib in combination with abiraterone versus abiraterone alone in mCRPC pretreated with docetaxel, irrespective of HRR mutation status [38]. Hence, this clinical benefit led to 2 phase 3 trials comparing abiraterone with either olaparib (PROpel) or niraparib (MAGNITUDE), with both studies having met their primary endpoint with an rPFS improvement in the combination arm (irrespective of HRR mutations in PROpel but without benefit in HRR-negative population in MAGNITUDE), and OS data still immature [39] (Table 2). Other combinations with checkpoint inhibitors are ongoing (NCT02861573, NCT03834519 KEYLINK-010 study, etc.) (Table 3), since PARPi could act as immunomodulatory agents, inducing PD-L1 expression through interferon after activation of the cGAS/STING pathway via accumulation of unrepaired DNA fragments [40].

Table 3.

Ongoing trials using PARPi in combination in mCRPC

 Ongoing trials using PARPi in combination in mCRPC
 Ongoing trials using PARPi in combination in mCRPC

Urothelial carcinoma (UC) displays a complex genomic landscape, including defective DNA damage response and repair at the somatic genomic level. A recent targeted exome sequencing (n = 237 genes) identified a DDR gene alteration in 54% of patients analyzed (not all being considered deleterious), with a higher frequency in muscle-invasive bladder cancer (BC) (MIBC) compared to low-grade non-MIBC and high-grade upper tract urothelial cancer (UTUC), the latter displaying a distinct mutation profile than BC [41]. In a recent study, compared to urothelial BC, UTUC had higher clonal and subclonal mutation numbers, TP53, PIK3CA, and FGFR3 mutations being the putative driver genes in UTUC, whereas for BC, the driver gene may be BRCA1. Conversely, there was no significant difference in the number of DDR mutations between UTUC and urothelial BC [42].

In high-grade non-MIBC and MIBC cohorts, there was a strong association between DDR gene mutation and higher tumor mutation burden. Using a more stringent definition of mutation in DDR genes, DDR gene alterations were observed in 23% of patients with ATM, ERCC2, PRKDC, and ATRX mutations being most common (in 4.3, 4.1, 2.4, and 2.2% of the cases, respectively). ERCC2 alterations seemed to be linked to smoking habits in patients in this study [41], and recent data suggested that PRKDC alterations may serve as potential biomarkers of response to immune checkpoint blockade [43].

In addition, recent studies have suggested that there is an association between DDR gene alterations and sensitivity to cisplatin-based regimens and to immunotherapy treatment for UC (PD-1/PD-L1 blockade) [44, 45]. Teo et al. [45] were the first to report the association between immunotherapy and defective DDR mechanisms beyond MMR deficiency (pembrolizumab already approved for MMR-deficient tumors, regardless of primary tumor site). Regarding platinum sensitivity, cell line studies reported that DDR-related mechanism could be of particular relevance in acquired cisplatin resistance mechanisms of individual UC cells and that targeting of CHK1 (encoded by the CHEK1 gene) might be useful to overcome acquired cisplatin resistance of certain subtypes of UC [46].

Recently, the JAVELIN Bladder 100 trial showed there was a survival advantage for the use of avelumab immunotherapy in maintenance setting in patients who had not progressed with 1st line chemotherapy for advanced/metastatic UC [47], with PD-L1 expression by tumor cells being a putative biomarker of response as well as tumor mutational burden [48].

Regarding germline mutations, recent work using targeted sequencing (i.e., targeted clinical germline testing) of known cancer susceptibility genes revealed that 14–24% of UC patients harbored pathogenic germline variants and 18.6% had clinically actionable variants. ATM and BRCA2 were highlighted in these studies as potential UC predisposition genes but account for only a small fraction of the genetic predisposition for the disease since other genes (i.e., MLH1, MSH2, etc.) are also validated as UC risk genes [49]. Individually rare but collectively common germline variants can explain a substantial fraction of the missing genetic predisposition to UC. A recent study on putative deleterious germline variants in DDR genes in UC opened the door to trials of PARPi in properly selected UC patients [50]. Data presented during the ASCO-GU 2022, supporting the use of HRR status to select patients for PARPi treatment in advanced UC, are summarized in Table 4.

Table 4.

PARPi recent results in UC (ASCO-GU 2022)

 PARPi recent results in UC (ASCO-GU 2022)
 PARPi recent results in UC (ASCO-GU 2022)

In other genitourinary malignancies, such as RCCs, the analysis of pathogenic/likely pathogenic germline variant frequency and spectrum has been determined on a cohort of patients with early onset of the disease [51]. These variants were identified in 17.7% of patients, of which 8.6% were in non-RCC-associated genes, including 7.3% in DDR genes such as BRCA1/2, ATM, BARD1, PALB2, and CHEK2. CHK2 kinase (encoded by the CHK2 gene) has a role in the mitotic phosphorylation of the tumor suppressor BRCA1 [4]. On the basis of its role during DDR, CHK2 loss in human tumors may be exploited to develop novel therapies based on synthetic lethal interactions, but the clinical utility of the strategy of identification of germline variants requires validation and further validation in RCC.

At the somatic level, deleterious DDR gene alterations have been described as recurrent genomic events in patients with advanced RCC (most frequently altered genes being CHEK2 and ATM) and were mostly clonal in a recent cohort [52]. Loss-of-function events in these genes may affect outcomes with immunotherapy in metastatic RCC (with independent correlation with OS), and these hypothesis-generating results deserve further study.

A growing body of evidence supports the need for identification of germinal and somatic DDR alterations in patients with genitourinary malignancies. PARPi have already shown significant survival benefits in patients harboring HHR mutations in advanced settings, paving the way for precision medicine. Further studies will address the benefit of testing at earlier stages of disease to improve outcome.

The authors have no conflicts of interest to declare.

The authors declare no funding source for this manuscript.

Charles Dariane: data collection and manuscript writing. Marc-Olivier Timsit: project development, manuscript writing, manuscript editing, critical revision, and supervision.

1.
Helleday
T
,
Eshtad
S
,
Nik-Zainal
S
.
Mechanisms underlying mutational signatures in human cancers
.
Nat Rev Genet
.
2014 Sep
;
15
(
9
):
585
98
.
2.
Ciccia
A
,
Elledge
SJ
.
The DNA damage response: making it safe to play with knives
.
Molecular Cell
.
2010 Oct
;
40
(
2
):
179
204
.
3.
Karl
LA
,
Peritore
M
,
Galanti
L
,
Pfander
B
.
DNA double strand break repair and its control by nucleosome remodeling
.
Front Genet
.
2021
;
12
:
821543
.
4.
Stolz
A
,
Ertych
N
,
Bastians
H
.
Tumor suppressor CHK2: regulator of DNA damage response and mediator of chromosomal stability
.
Clin Cancer Res
.
2011 Feb
;
17
(
3
):
401
5
.
5.
Zhang
J
,
Willers
H
,
Feng
Z
,
Ghosh
JC
,
Kim
S
,
Weaver
DT
,
.
Chk2 phosphorylation of BRCA1 regulates DNA double-strand break repair
.
Mol Cell Biol
.
2004 Jan
;
24
(
2
):
708
18
.
6.
Zhang
F
,
Fan
Q
,
Ren
K
,
Andreassen
PR
.
PALB2 functionally connects the breast cancer susceptibility proteins BRCA1 and BRCA2
.
Mol Cancer Res
.
2009 Jul
;
7
(
7
):
1110
8
.
7.
Schiewer
MJ
,
Goodwin
JF
,
Han
S
,
Brenner
JC
,
Augello
MA
,
Dean
JL
,
.
Dual roles of PARP-1 promote cancer growth and progression
.
Cancer Discov
.
2012 Dec
;
2
(
12
):
1134
49
.
8.
Boussios
S
,
Abson
C
,
Moschetta
M
,
Rassy
E
,
Karathanasi
A
,
Bhat
T
,
.
Poly (ADP-ribose) polymerase inhibitors: talazoparib in ovarian cancer and beyond
.
Drugs R D
.
2020 Jun
;
20
(
2
):
55
73
.
9.
Teyssonneau
D
,
Margot
H
,
Cabart
M
,
Anonnay
M
,
Sargos
P
,
Vuong
N-S
,
.
Prostate cancer and PARP inhibitors: progress and challenges
.
J Hematol Oncol
.
2021 Mar
;
14
(
1
):
51
.
10.
Castro
E
,
Goh
C
,
Olmos
D
,
Saunders
E
,
Leongamornlert
D
,
Tymrakiewicz
M
,
.
Germline BRCA mutations are associated with higher risk of nodal involvement, distant metastasis, and poor survival outcomes in prostate cancer
.
J Clin Oncol
.
2013 May
;
31
(
14
):
1748
57
.
11.
Kote-Jarai
Z
,
Leongamornlert
D
,
Saunders
E
,
Tymrakiewicz
M
,
Castro
E
,
Mahmud
N
,
.
BRCA2 is a moderate penetrance gene contributing to young-onset prostate cancer: implications for genetic testing in prostate cancer patients
.
Br J Cancer
.
2011 Oct
;
105
(
8
):
1230
4
.
12.
Sinclair
CS
,
Berry
R
,
Schaid
D
,
Thibodeau
SN
,
Couch
FJ
.
BRCA1 and BRCA2 have a limited role in familial prostate cancer
.
Cancer Res
.
2000 Mar
;
60
(
5
):
1371
5
.
13.
Robinson
D
,
Van Allen
EM
,
Wu
Y-M
,
Schultz
N
,
Lonigro
RJ
,
Mosquera
J-M
,
.
Integrative clinical genomics of advanced prostate cancer
.
Cell
.
2015 May
;
162
(
2
):
454
28
.
14.
Abida
W
,
Armenia
J
,
Gopalan
A
,
Brennan
R
,
Walsh
M
,
Barron
D
,
.
Prospective genomic profiling of prostate cancer across disease states reveals germline and somatic alterations that may affect clinical decision making
.
JCO Precis Oncol
.
2017 Jul
;
2017
(
1
):
1
16
.
15.
Pritchard
CC
,
Mateo
J
,
Walsh
MF
,
De Sarkar
N
,
Abida
W
,
Beltran
H
,
.
Inherited DNA-repair gene mutations in men with metastatic prostate cancer
.
N Engl J Med
.
2016 Aug
;
375
(
5
):
443
53
.
16.
The Breast Cancer Linkage Consortium
.
Cancer risks in BRCA2 mutation carriers
.
JNCI J Natl Cancer Inst
.
1999 Aug
;
91
(
15
):
1310
6
.
17.
Castro
E
,
Romero-Laorden
N
,
Del Pozo
A
,
Lozano
R
,
Medina
A
,
Puente
J
,
.
PROREPAIR-B: a prospective cohort study of the impact of germline DNA repair mutations on the outcomes of patients with metastatic castration-resistant prostate cancer
.
J Clin Oncol
.
2019 Feb
;
37
(
6
):
490
503
.
18.
Na
R
,
Zheng
SL
,
Han
M
,
Yu
H
,
Jiang
D
,
Shah
S
,
.
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
.
2017 May
;
71
(
5
):
740
7
.
19.
Nyberg
T
,
Frost
D
,
Barrowdale
D
,
Evans
DG
,
Bancroft
E
,
Adlard
J
,
.
Prostate cancer risks for male BRCA1 and BRCA2 mutation carriers: a prospective cohort study
.
Eur Urol
.
2020 Jan
;
77
(
1
):
24
35
.
20.
Rozet
F
,
Mongiat-Artus
P
,
Hennequin
C
,
Beauval
JB
,
Beuzeboc
P
,
Cormier
L
,
.
[French ccAFU guidelines: update 2020-2022: prostate cancer]
.
Prog Urol
.
2020 Nov
;
30
(
12S
):
S136
251
.
21.
Page
EC
,
Bancroft
EK
,
Brook
MN
,
Assel
M
,
Hassan Al Battat
M
,
Thomas
S
,
.
Interim results from the IMPACT study: evidence for prostate-specific antigen screening in BRCA2 mutation carriers
.
Eur Urol
.
2019 Dec
;
76
(
6
):
831
42
.
22.
Li
S
,
Silvestri
V
,
Leslie
G
,
Rebbeck
TR
,
Neuhausen
SL
,
Hopper
JL
,
.
Cancer risks associated with BRCA1 and BRCA2 pathogenic variants
.
J Clin Oncol
.
2022 Jan
:
JCO2102112
.
23.
Castro
E
,
Goh
C
,
Leongamornlert
D
,
Saunders
E
,
Tymrakiewicz
M
,
Dadaev
T
,
.
Effect of BRCA mutations on metastatic relapse and cause-specific survival after radical treatment for localised prostate cancer
.
Eur Urol
.
2015 Aug
;
68
(
2
):
186
93
.
24.
Carter
HB
,
Helfand
B
,
Mamawala
M
,
Wu
Y
,
Landis
P
,
Yu
H
,
.
Germline mutations in ATM and BRCA1/2 are associated with grade reclassification in men on active surveillance for prostate cancer
.
Eur Urol
.
2019 May
;
75
(
5
):
743
9
.
25.
Mateo
J
,
Seed
G
,
Bertan
C
,
Rescigno
P
,
Dolling
D
,
Figueiredo
I
,
.
Genomics of lethal prostate cancer at diagnosis and castration resistance
.
J Clin Invest
.
2020 Apr
;
130
(
4
):
1743
51
.
26.
Schweizer
MT
,
Sivakumar
S
,
Tukachinsky
H
,
Coleman
I
,
De Sarkar
N
,
Yu
EY
,
.
Concordance of DNA repair gene mutations in paired primary prostate cancer samples and metastatic tissue or cell-free DNA
.
JAMA Oncol
.
2021 Jun
;
7
(
9
):
1378
5
.
27.
Parker
C
,
Castro
E
,
Fizazi
K
,
Heidenreich
A
,
Ost
P
,
Procopio
G
,
.
Prostate cancer: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up
.
Ann Oncol
.
2020 Sep
;
31
(
9
):
1119
34
.
28.
Antonarakis
ES
,
Lu
C
,
Luber
B
,
Liang
C
,
Wang
H
,
Chen
Y
,
.
Germline DNA-repair gene mutations and outcomes in men with metastatic castration-resistant prostate cancer receiving first-line abiraterone and enzalutamide
.
Eur Urol
.
2018 Aug
;
74
(
2
):
218
25
.
29.
Annala
M
,
Struss
WJ
,
Warner
EW
,
Beja
K
,
Vandekerkhove
G
,
Wong
A
,
.
Treatment outcomes and tumor loss of heterozygosity in germline DNA repair-deficient prostate cancer
.
Eur Urol
.
2017 Jul
;
72
(
1
):
34
42
.
30.
Hussain
M
,
Mateo
J
,
Fizazi
K
,
Saad
F
,
Shore
N
,
Sandhu
S
,
.
Survival with olaparib in metastatic castration-resistant prostate cancer
.
N Engl J Med
.
2020 Dec
;
383
(
24
):
2345
57
.
31.
Mateo
J
,
Carreira
S
,
Sandhu
S
,
Miranda
S
,
Mossop
H
,
Perez-Lopez
R
,
.
DNA-repair defects and olaparib in metastatic prostate cancer
.
N Engl J Med
.
2015 Oct
;
373
(
18
):
1697
708
.
32.
Mateo
J
,
Porta
N
,
Bianchini
D
,
McGovern
U
,
Elliott
T
,
Jones
R
,
.
Olaparib in patients with metastatic castration-resistant prostate cancer with DNA repair gene aberrations (TOPARP-B): a multicentre, open-label, randomised, phase 2 trial
.
Lancet Oncol
.
2020 Jan
;
21
(
1
):
162
74
.
33.
de Bono
J
,
Mateo
J
,
Fizazi
K
,
Saad
F
,
Shore
N
,
Sandhu
S
,
.
Olaparib for metastatic castration-resistant prostate cancer
.
New Engl J Med
.
2020 May
;
382
(
22
):
2091
102
.
34.
Abida
W
,
Patnaik
A
,
Campbell
D
,
Shapiro
J
,
Bryce
AH
,
McDermott
R
,
.
Rucaparib in men with metastatic castration-resistant prostate cancer harboring a BRCA1 or BRCA2 gene alteration
.
J Clin Oncol
.
2020 Nov
;
38
(
32
):
3763
72
.
35.
Smith
MR
,
Scher
HI
,
Sandhu
S
,
Efstathiou
E
,
Lara
PN
,
Yu
EY
,
.
Niraparib in patients with metastatic castration-resistant prostate cancer and DNA repair gene defects (GALAHAD): a multicentre, open-label, phase 2 trial
.
Lancet Oncol
.
2022 Feb
;
23
(
3
):
362
73
.
36.
de Bono
JS
,
Mehra
N
,
Scagliotti
GV
,
Castro
E
,
Dorff
T
,
Stirling
A
,
.
Talazoparib monotherapy in metastatic castration-resistant prostate cancer with DNA repair alterations (TALAPRO-1): an open-label, phase 2 trial
.
Lancet Oncol
.
2021 Sep
;
22
(
9
):
1250
64
.
37.
Li
L
,
Karanika
S
,
Yang
G
,
Wang
J
,
Park
S
,
Broom
BM
,
.
Androgen receptor inhibitor-induced “BRCAness” and PARP inhibition are synthetically lethal for castration-resistant prostate cancer
.
Sci Signal
.
2017 May
;
10
(
480
):
eaam7479
.
38.
Clarke
N
,
Wiechno
P
,
Alekseev
B
,
Sala
N
,
Jones
R
,
Kocak
I
,
.
Olaparib combined with abiraterone in patients with metastatic castration-resistant prostate cancer: a randomised, double-blind, placebo-controlled, phase 2 trial
.
Lancet Oncol
.
2018 Jul
;
19
(
7
):
975
86
.
39.
Clarke
NW
,
Armstrong
AJ
,
Thiery-Vuillemin
A
,
Oya
M
,
Shore
N
,
Loredo
E
,
.
Abiraterone and olaparib for metastatic castration-resistant prostate cancer
.
NEJM Evid
.
2022 Jun 3
. https://doi.org/10.1056/evidoa2200043.
40.
Chabanon
RM
,
Muirhead
G
,
Krastev
DB
,
Adam
J
,
Morel
D
,
Garrido
M
,
.
PARP inhibition enhances tumor cell-intrinsic immunity in ERCC1-deficient non-small cell lung cancer
.
J Clin Invest
.
2019 Mar
;
129
(
3
):
1211
28
.
41.
Nassar
AH
,
Umeton
R
,
Kim
J
,
Lundgren
K
,
Harshman
L
,
Van Allen
EM
,
.
Mutational analysis of 472 urothelial carcinoma across grades and anatomic sites
.
Clin Cancer Res
.
2019 Apr
;
25
(
8
):
2458
70
.
42.
Yang
K
,
Yu
W
,
Liu
H
,
Ding
F
,
Zhang
Y
,
Zhang
Y
,
.
Comparison of genomic characterization in upper tract urothelial carcinoma and urothelial carcinoma of the bladder
.
Oncologist
.
2021 Aug
;
26
(
8
):
e1395
405
.
43.
Tan
KT
,
Yeh
C-N
,
Chang
Y-C
,
Cheng
J-H
,
Fang
W-L
,
Yeh
Y-C
,
.
PRKDC: new biomarker and drug target for checkpoint blockade immunotherapy
.
J Immunother Cancer
.
2020 Mar
;
8
(
1
):
e000485
.
44.
Teo
MY
,
Bambury
RM
,
Zabor
EC
,
Jordan
E
,
Al-Ahmadie
H
,
Boyd
ME
,
.
DNA damage response and repair gene alterations are associated with improved survival in patients with platinum-treated advanced urothelial carcinoma
.
Clin Cancer Res
.
2017 Jul
;
23
(
14
):
3610
8
.
45.
Teo
MY
,
Seier
K
,
Ostrovnaya
I
,
Regazzi
AM
,
Kania
BE
,
Moran
MM
,
.
Alterations in DNA damage response and repair genes as potential marker of clinical benefit from PD-1/PD-L1 blockade in advanced urothelial cancers
.
J Clin Oncol
.
2018 Jun
;
36
(
17
):
1685
94
.
46.
Höhn
A
,
Krüger
K
,
Skowron
MA
,
Bormann
S
,
Schumacher
L
,
Schulz
WA
,
.
Distinct mechanisms contribute to acquired cisplatin resistance of urothelial carcinoma cells
.
Oncotarget
.
2016 Jul
;
7
(
27
):
41320
35
.
47.
Powles
T
,
Park
SH
,
Voog
E
,
Caserta
C
,
Valderrama
BP
,
Gurney
H
,
.
Avelumab maintenance therapy for advanced or metastatic urothelial carcinoma
.
N Engl J Med
.
2020 Sep
;
383
(
13
):
1218
30
.
48.
Powles
T
,
Sridhar
SS
,
Loriot
Y
,
Bellmunt
J
,
Mu
XJ
,
Ching
KA
,
.
Avelumab maintenance in advanced urothelial carcinoma: biomarker analysis of the phase 3 JAVELIN Bladder 100 trial
.
Nat Med
.
2021 Dec
;
27
(
12
):
2200
11
.
49.
Nassar
AH
,
Abou Alaiwi
S
,
AlDubayan
SH
,
Moore
N
,
Mouw
KW
,
Kwiatkowski
DJ
,
.
Prevalence of pathogenic germline cancer risk variants in high-risk urothelial carcinoma
.
Genet Med
.
2020
;
22
(
4
):
709
18
.
50.
Vosoughi
A
,
Zhang
T
,
Shohdy
KS
,
Vlachostergios
PJ
,
Wilkes
DC
,
Bhinder
B
,
.
Common germline-somatic variant interactions in advanced urothelial cancer
.
Nat Commun
.
2020 Dec
;
11
(
1
):
6195
.
51.
Truong
H
,
Sheikh
R
,
Kotecha
R
,
Kemel
Y
,
Reisz
PA
,
Lenis
AT
,
.
Germline variants identified in patients with early-onset renal cell carcinoma referred for germline genetic testing
.
Eur Urol Oncol
.
2021 Dec
;
4
(
6
):
993
1000
.
52.
Ged
Y
,
Chaim
JL
,
DiNatale
RG
,
Knezevic
A
,
Kotecha
RR
,
Carlo
MI
,
.
DNA damage repair pathway alterations in metastatic clear cell renal cell carcinoma and implications on systemic therapy
.
J Immunother Cancer
.
2020 Jun
;
8
(
1
):
e000230
.