ABSTRACT
Alterations in the homologous recombination repair genes, such as BRCA1 and BRCA2, are prevalent in various cancers, presenting a unique opportunity to develop synthetic lethal strategies that target homologous recombination deficiency (HRD). Poly ADP‐ribose polymerase inhibitors (PARPis) have been developed to induce synthetic lethality in tumors with HRD by inhibiting the repair of single‐strand DNA breaks. Beyond the initial approach to target cancers associated with HRD, the utility of PARPis has expanded to combination therapy with immune checkpoint inhibitors, anti‐angiogenic drugs, or anti‐androgen drugs based on the molecular biological rationale. In the field of genitourinary (GU) cancer, PARPis, such as olaparib, rucaparib, and talazoparib, are approved by the Food and Drug Administration in metastatic prostate cancer patients with BRCA1/2 mutations, sometimes in combination with other agents (e.g., olaparib plus abiraterone acetate, or talazoparib plus enzalutamide). More recently, pivotal clinical trials have broadened the potential of PARPis to the other GU cancers, including urothelial carcinoma and renal cell carcinoma. In this review, we examine the biomarkers for the response to PARPis beyond mutations in BRCA1/2 and discuss the current state and future perspectives of PARPis in GU cancers.
Keywords: biomarkers, homologous recombination deficiency, PARP inhibitor, synthetic lethality, urogenital cancer
Abbreviations
- ACC
adrenocortical carcinoma
- BER
base excision repair
- BSC
best supportive care
- CAR‐T cell
chimeric antigen receptor T‐cell
- CI
confidence interval
- DCR
disease control rate
- DDR
dna damage repair
- DSBs
double‐strand breaks
- FAP
familial adenomatous polyposis
- GCT
germ cell tumor
- GU
genitourinary
- HR
homologous recombination
- HRD
homologous recombination deficiency
- HRR
homologous recombination repair
- ICI
immune checkpoint inhibitor
- KIRC
kidney renal clear cell carcinoma
- KPRC
kidney papillary renal cell carcinoma
- LOH
loss of heterozygosity
- MIBC
muscle‐invasive bladder cancer
- NHEJ
non‐homologous end joining
- ORR
objective response rate
- OS
overall survival
- PARP
poly ADP‐ribose polymerase
- PARPi
PARP inhibitor
- PBC
platinum‐based chemotherapy
- PC
prostate cancer
- PCC
pheochromocytoma
- PFS
progression‐free survival
- PGL
paraganglioma
- RCC
renal cell carcinoma
- SD
stable disease
- SSBs
single‐strand breaks
- TCGA
the cancer genome atlas
- TGCT
testicular germ cell tumor
- UC
urothelial carcinoma
- UTUC
upper tract urothelial carcinoma
- WES
whole‐exome sequencing
1. Introduction
Poly(ADP‐ribose) polymerases (PARPs) are enzymes involved in poly ADP‐ribosylation (PARylation), a post‐translational modification critical for repairing DNA single‐strand breaks (SSBs). Among the 17 PARP family members, PARP1 and PARP2 are the primary mediators of DNA repair and genomic stability, especially in response to SSBs [1, 2, 3]. Inhibition of these enzymes leads to SSB accumulation and replication‐associated double‐strand breaks (DSBs), ultimately triggering cancer cell death—a concept that led to the development of PARP inhibitors (PARPis) [4, 5, 6].
The clinical relevance of PARPis grew dramatically following the discovery of synthetic lethality in BRCA1/2‐mutated tumors, which lack homologous recombination (HR)‐mediated DSB repair [7, 8]. BRCA1 and BRCA2 genes are central to the HR repair (HRR) pathway, and their inactivation results in genomic instability and increased cancer susceptibility [9, 10, 11]. While synthetic lethality was initially attributed to the accumulation of DSBs caused by unrepaired SSBs, alternative mechanisms such as PARP trapping, replication fork collapse, and single‐strand gap accumulation have also been proposed [12, 13, 14, 15, 16, 17, 18].
BRCA1/2 mutations are found in approximately 10%–18% of patients with advanced prostate cancer (PC) [19, 20]. The U.S. Food and Drug Administration has approved olaparib, rucaparib, and talazoparib for BRCA1/2‐mutant metastatic PC [19, 20, 21, 22], and more recently, PARPi‐based combination therapies (e.g., with androgen receptor signaling inhibitors) have been approved [23, 24, 25, 26]. Compared to the BRCA1/2 mutant population in PC patients, BRCA1/2 mutation rates in other genitourinary (GU) cancers are generally low, which might delay the application of PARPis in patients with these cancers [27, 28]. However, with advances in understanding HR and expanded genome sequencing efforts, mutations in HR‐related genes beyond BRCA1/2 are now being identified in GU cancers, opening new avenues for PARPi‐based strategies [29, 30].
In light of these advancements, this article provides the potential biomarkers of PARPis beyond mutations in BRCA1 and BRCA2 and discusses the current state and future perspectives of PARPis in GU cancers.
2. Biomarkers of PARP Inhibitors Beyond BRCA1/2
Although BRCA1/2 mutations remain the most established predictors of PARPi sensitivity, accumulating evidence indicates that tumors with homologous recombination deficiency (HRD) can also respond to PARPis even in the absence of BRCA1/2 alterations [31, 32]. HRD denotes an impaired ability to repair DNA double strand breaks via the HRR pathway. Clinically, HRD is often inferred from genomic “scar” assays—most notably the HRD score—which combines three chromosomal instability metrics: loss of heterozygosity, telomeric allelic imbalance, and large‐scale state transitions [33, 34]. Higher HRD scores generally correlate with improved responses to PARPis and platinum based chemotherapy regardless of BRCA1/2 status [35]. Commercial assays such as Myriad myChoice CDx incorporate the HRD score and help guide PARPi therapy across several cancer types [36, 37].
In parallel, the concept of “BRCAness” was introduced to describe tumors that phenotypically resemble BRCA‐mutant cancers despite lacking BRCA1/2 mutations [19, 20]. These tumors often harbor alterations in other HRR‐related genes such as ATM, CHEK2, PALB2, RAD51, and FANCA, and may exhibit comparable therapeutic efficacy to BRCA‐mutated cancers. For example, favorable responses to PARPis have been reported in PALB2‐mutated breast and pancreatic cancers, as well as ATM‐mutated PCs [21, 22, 24]. These findings imply the possible and broader use of PARPi for patients with BRCAness, depending on the findings by means of cancer gene panel testing.
Beyond genes commonly included in cancer gene panels, certain genes play a critical role at the transcription levels in determining sensitivity to PARPis. For instance, Schlafen 11 (SLFN11), a putative DNA/RNA helicase, has emerged as a significant determinant of PARPi sensitivity [38, 39]. In previous research, high SLFN11 expression enhanced PARPi cytotoxicity by preventing stalled replication fork recovery, which was closely associated with improved outcomes following olaparib treatment [38, 40]. Notably, over 50% of clear cell renal cell carcinomas (RCCs) exhibit high SLFN11 expression [41, 42], suggesting that SLFN11 may serve as a predictive biomarker in GU cancers even in the absence of BRCA1/2 mutations.
Taken together, these markers support a paradigm in which PARPis may benefit selected patients based on molecular features beyond BRCA1/2 mutation status and expand the therapeutic application of PARPi in GU cancers.
3. PARP Inhibitors as Antitumor Immune Activators
Improvement of the antitumor immune microenvironment by PARPis has been reported in multiple studies [43]. For instance, talazoparib, a PARPi in PC treatment, promotes the accumulation of cytoplasmic DNA and the activation of the STING pathway in vitro [44]. Huang et al. also demonstrated that talazoparib promotes STING activation in murine ovarian cancer models, leading to increased infiltration of antitumor immune cells and enhanced functionality of CD8+ T and NK cells [45]. In addition, PARPis (olaparib and talazoparib) are associated with the upregulation of PD‐L1 expression on cancer cells in vitro and in vivo [46]. Given that GU cancers are often treated with immune checkpoint inhibitors (ICIs), these findings provide a strong rationale for combining PARPis with ICIs in GU cancers, regardless of BRCA mutation.
4. PARP Inhibition in Urothelial Carcinoma
Urothelial carcinoma (UC) has a complex genomic landscape that includes deficiencies in DNA damage repair (DDR) genes at both the germline and somatic levels. The DDR gene mutations were relatively common across all UC subtypes, with the most frequently mutated genes being ATM (4.3%), ERCC2 (4.1%), PRKDC (2.4%), and ATRX (2.2%), particularly in muscle‐invasive bladder cancer (MIBC) [47, 48, 49, 50]. These alterations generally increase tumor mutational burden and may contribute to the sensitivity to DNA‐damaging agents such as PARPis [51, 52]. Although PARPis have not yet been approved for UC, their efficacy has been investigated in multiple clinical trials as monotherapy, in combination with immune checkpoint inhibitors, and as maintenance or neoadjuvant therapy (Table 1). Key trials are summarized in Table 1.
TABLE 1.
Clinical trials on poly ADP‐ribose polymerase inhibitors for urothelial carcinoma.
Trial name | Year of enrollment | Phase | Study design | Patient population | HRD status | Treatment | Enrollment number | Control | Primary endpoint | Results |
---|---|---|---|---|---|---|---|---|---|---|
BISCAY [53] | 2016–2019 | Ib | Open‐label | Advanced/metastatic UC who had progressed on previous PBC treatment | Yes/No | Durvalumab + Olaparib | n = 35 | Durvalumab | The safety of treatment | RR was 35.7% mutations with HRD and 9.1% without HRD |
BAYOU [54] | 2016–2019 | II | Randomized, double‐blind | Untreated, platinum‐ineligible metastatic UC patients | Yes/No | Durvalumab + Olaparib | n = 76 | Durvalumab + placebo | PFS | 4.2 months (95% CI, 3.6–5.6) vs. 3.5 months (95% CI, 1.9–5.1) |
ATLAS [55] | 2018–2019 | II | Open‐label | Locally advanced/unresectable or metastatic UC patients post‐PBC and/or ICI treatment | Yes/No | Rucaparib | n = 97 | None | ORR | 0% (95% CI, 0–3.8) |
ATLANTIS [56, 57] | 2017–2021 | II | Randomized, umbrella‐design screening trial | Metastatic or locally advanced UC patients with response or stable disease post 1st‐line chemotherapy | Yes | Rucaparib | n = 19 | Placebo | PFS | 35.3 weeks (80% CI, 11.7–35.6) vs. 15.1 weeks (80% CI, 11.9–22.6) |
JAVELIN PARP Medley [58] | 2017–2019 | II | Open‐label, basket‐type | Patients with advanced solid tumors (including UC) | Yes/No | Talazoparib + Avelumab | n = 40 | None | ORR | 15.0% (95% CI, 5.7–29.8) |
Meet‐URO12 [59, 60] | 2019–2021 | II | Randomized, open‐label | Advanced UC patients post‐platinum‐based chemotherapy without disease progression | Yes/No | Niraparib + best supportive care (BSC) | n = 39 | BSC alone | PFS | 2.1 months (95% CI, 0.9–3.2) vs. 2.4 months (95% CI, 1.6–3.2) |
NEODURVARIB [61, 62] | 2018–2019 | II | Open‐label | MIBC patients scheduled for radical cystectomy | Yes/No | Durvalumab + Olaparib | n = 29 | None |
Impact of neoadjuvant treatment in the molecular profile of MIBC |
Genetic alterations remained unchanged |
Abbreviations: BSC, best supportive care; CI, confidence interval; HRD, homologous recombination deficiency; ICI, immune checkpoint inhibitor; MIBC, muscle‐invasive bladder cancer; ORR, objective response rate; PBC, platinum‐based chemotherapy; PFS, progression‐free survival; UC, urothelial carcinoma.
4.1. PARPi Monotherapy in UC
4.1.1. ATLAS Trial
The ATLAS trial (NCT03397394) was a phase II, single‐arm study evaluating rucaparib monotherapy in 97 patients with locally advanced or metastatic UC who had previously received platinum‐based chemotherapy (PBC) and/or ICIs [55]. Overall, 44/97 (45.4%) patients had received two prior therapies for advanced disease; 93/97 (95.9%) patients had received prior PBC, 71/97 (73.2%) patients had received prior ICIs, and 67/97 (69.1%) had received both a PBC and an ICI (separately or combined). Among the 97 enrolled patients, 20 (20.6%) were HRD‐positive, 30 (30.9%) were HRD‐negative, and 47 (48.5%) had an unknown HRD status. No confirmed objective responses were observed in either the overall cohort or the HRD‐positive subgroup (0%; 95% CI, 0%–3.8%). Although 20.6% of patients were HRD‐positive, none achieved an objective response. Median progression‐free survival was 1.8 months, and efficacy showed no correlation with HRD status. These findings indicate that PARPi monotherapy offers limited benefit in unselected or HRD‐positive urothelial carcinoma, possibly owing to insufficient biomarker specificity or the advanced disease stage.
4.2. PARPis in Combination With ICIs
4.2.1. BISCAY Trial
The BISCAY trial (NCT02546661) is a phase Ib study investigating the safety and improvement of clinical activity of durvalumab, a PD‐L1 inhibitor, in combination with various targeted therapies [53]. The trial included two non‐randomized cohorts combining olaparib with durvalumab immunotherapy, selected based on six biomarkers for second‐line or later treatment of metastatic UC. One cohort included an HRR‐selected group (n = 14) comprising patients with mutations in HRR genes, such as ATM and BRCA1/2, while the other was an HRR‐unselected group (n = 21). In the HRR‐selected cohort, the objective response rate (ORR) was 35.7%, compared with 9.1% in the HRR‐unselected group receiving the same combination. Nonetheless, the regimen did not meet the predefined efficacy threshold for further development, and survival outcomes were comparable between the two arms. These results highlight the need for robust biomarker strategies and larger randomized studies to clarify the value of PARPi–ICI combinations.
4.2.2. BAYOU Trial
The BAYOU trial (NCT03459846) is a phase II, double‐blind, randomized trial designed to evaluate the efficacy and safety of durvalumab combined with olaparib in untreated, platinum‐ineligible patients with metastatic UC [54]. The trial enrolled 154 patients with UC who were not selected based on their HRR status. Of these, 78 patients were assigned to the durvalumab plus olaparib group and 76 to the durvalumab plus placebo group. HRR mutations were found in 17 patients (21.8%) in the durvalumab plus olaparib group and 14 patients (18.4%) in the durvalumab plus placebo group. In the durvalumab plus olaparib group, the median PFS was 4.2 months (95% CI, 3.6–5.6), compared to 3.5 months (95% CI, 1.9–5.1) in the durvalumab plus placebo group (hazard ratio, 0.94; 95% CI, 0.64–1.39; stratified log‐rank p = 0.789). The study failed to meet its primary endpoint because the durvalumab–olaparib arm showed no PFS benefit in the overall population. In patients with HRR mutations, however, combination therapy significantly prolonged PFS (5.6 vs. 1.8 months; p < 0.001). These data suggest that PARPi–ICI synergy may be confined to biomarker‐selected populations.
4.2.3. JAVELIN PARP Medley
The JAVELIN PARP Medley trial (NCT03330405) is a non‐randomized, basket‐type phase Ib and phase II trial involving patients with advanced solid tumors, including UC [58]. This trial is assessing the safety and clinical efficacy of combination therapy involving talazoparib and the anti‐PD‐L1 antibody, avelumab. For the enrollment of UC cases, HRR biomarker selection was not applied, and out of the 211 total patients enrolled, 40 were UC cases. Among them, 18 patients (45%) were DDR‐positive. The ORR was comparable between patients who had and had not received prior platinum‐based therapy (14.3% vs. 16.7%). The median duration of response was not reached (range, 3.9 to ≥ 14.7 months). Although durable responses occurred in a minority of patients, the absence of stratification limits the interpretability and clinical relevance of these findings.
4.3. PARPis As Maintenance Therapy
4.3.1. | ATLANTIS Trial
The ATLANTIS trial (ISRCTN25859465) is a multi‐center, randomized phase II umbrella design screening trial exploring novel targeted therapies in biomarker‐defined subgroups [56, 57]. Eligible patients are those with metastatic or locally advanced UC who achieved objective response or SD after 4–8 cycles of first‐line chemotherapy. Multiple novel agents are being used in parallel, and patients are entered into ATLANTIS subgroup trials according to their tumor biomarker profiles. Each comparison control arm is matched with a placebo, and, where possible, comparisons are double‐blind. Of the 248 patients who underwent pre‐screening for biomarkers, 74 (29.8%) were DDR biomarker‐positive. Among these, 40 patients were randomly assigned to the rucaparib comparison arm (rucaparib: 20 patients, placebo: 20 patients). The median PFS was 35.3 weeks with rucaparib (80% CI, 11.7–35.6) versus 15.1 weeks with the placebo (80% CI, 11.9–22.6), with an adjusted hazard ratio of 0.53 (80% CI, 0.30–0.92; one‐sided p = 0.07). Median OS was not reached in the rucaparib arm, whereas it was 72.3 weeks in the placebo arm (adjusted hazard ratio 1.22; 80% CI 0.62–2.38; p = 0.35). Although exploratory, these results suggest a potential role for PARP inhibitors as maintenance therapy, particularly for patients with DDR alterations who respond to first‐line chemotherapy.
4.3.2. Meet‐URO12 Trial
The Meet‐URO 12 trial (NCT03945084) was a multi‐center, randomized, open‐label phase II trial that investigated the efficacy of niraparib as maintenance therapy for patients with advanced UC [60]. The study included 58 patients with advanced UC who had completed PBC and had no disease progression. The patients were randomized to receive either niraparib plus best supportive care (BSC) (n = 39) or BSC alone (n = 19). No significant PFS benefit was observed (2.1 vs. 2.4 months; hazard ratio, 0.92; 95% CI, 0.49–1.75; p = 0.81), and the trial was halted early, partly due to the availability of avelumab maintenance, which demonstrated a clear survival benefit in this setting [59].
4.4. Neoadjuvant Applications
4.4.1. NEODURVARIB Trial
The NEODURVARIB trial (NCT03534492) was a phase II clinical trial designed to evaluate the impact of neoadjuvant combination therapy with durvalumab and olaparib on the molecular profile of MIBC [61, 62]. A total of 29 patients with cT2‐T4a MIBC, scheduled for radical cystectomy, were enrolled. Imaging‐based evaluations following neoadjuvant therapy showed a partial response rate of 24.1% and stable disease in 51.7% of the cases, progressive disease in 10.3% of the cases, and non‐evaluable in 13.8% of the cases. Of the 26 cystectomies performed, the pathological complete response rate was 50%. Although no clear genomic predictors of response were identified, these early findings are promising and suggest that PARPi–ICI combinations may be feasible in the neoadjuvant setting, especially in patients ineligible for cisplatin‐based chemotherapy.
4.5. Clinical Implications and Future Perspectives
Taken together, these trials indicate that the efficacy of PARP inhibitors in UC depends strongly on patient selection. PARPi monotherapy offers limited benefit, whereas PARPi–ICI combinations or use in maintenance or neoadjuvant settings appear more promising, particularly for patients with DDR or HRR alterations. Future studies should adopt biomarker‐enriched designs, evaluate earlier lines of therapy, and explore combination strategies that enhance PARPi sensitivity and overcome resistance.
5. PARP Inhibition in Renal Cell Carcinoma
Although BRCA1/2 mutations are rare in RCC, recent genomic profiling has shown that a subset of patients harbor HRR or DDR gene alterations [63]. These molecular features provide a rationale for evaluating PARPis in RCC. Although no PARPi is currently approved for this indication, several early‐phase trials are assessing their efficacy as monotherapy, in combination with ICIs, or even in HRR wild‐type cases (Table 2). Key examples are summarized in Table 2.
TABLE 2.
Clinical trials on poly ADP‐ribose polymerase inhibitors for renal cell carcinoma.
Trial name | Year of enrollment | Phase | Study design | Patient population | Treatment | HRD status | Enrollment number | Control | Primary endpoint | Results |
---|---|---|---|---|---|---|---|---|---|---|
ORCHID [64] | 2019— | II | Non‐randomized, open‐label | Patients with mRCC having BAP1 or other DNA repair gene mutations | Olaparib | Yes | 11 (still recruiting up to 20 participants) | None | DCR | DCR was 22% |
WIRE [65] | 2020— | II | Non‐randomized, proof‐of‐mechanism | Patients after nephrectomy | Cediranib, Olaparib, Durvalumab (monotherapy or combination) | Yes/No | 76 (recruiting) | None | Changes in capillary permeability and changes to intra‐tumoral CD8+ T‐cell infiltration | Not reported yet |
NICARAGUA [66] | 2019–2014 | II | Non‐randomized, open‐label | Patients with RCC (including UC) | Niraparib, Cabozantinib | Yes/No | 5 RCC and 14 UC | None | PFS | Not reported yet. 14 (74%) patients had SD |
IMAGENE [67] | 2022–2023 | II | Non‐randomized, proof‐of‐concept | Patients with HRR gene mutations resistant to ICIs | Niraparib, PD‐1 inhibitor | Yes | Not reported yet | None | ORR | Not reported yet |
Abbreviations: DCR, disease control rate; HRD, homologous recombination deficiency; HRR, homologous recombination repair; ICI, immune checkpoint inhibitor; mRCC, metastatic renal cell carcinoma; ORR, objective response rate; RCC, renal cell carcinoma; SD, stable disease; UC, urothelial carcinoma.
5.1. Monotherapy Potential of PARPis in RCC
5.1.1. ORCHID Trial
The ORCHID trial (NCT03786796) is a phase II, single‐arm study evaluating olaparib monotherapy in patients with metastatic RCC and mutations in DDR genes (e.g., BAP1, ATM, PALB2, BRCA1/2) [64]. The trial targets patients with metastatic RCC who have previously undergone treatment with angiogenesis inhibitors or ICIs, with a planned enrollment of up to 20 participants. The trial is expected to be completed in 2025, and interim results from 11 patients, whose genetic abnormalities were identified in BAP1 (61.5%), ATM (15.4%), PALB2 (15.4%), BRCA1 (7.7%), and BRCA2 (7.7%), showed a disease control rate of 18% and tumor shrinkage in 27%, although the ORR was low (9%). These findings suggest limited but detectable activity of olaparib in molecularly selected RCC patients.
5.1.2. WIRE Trial (Monotherapy Arms)
The WIRE trial (NCT03741426) is a phase II, multi‐arm, multi‐center, non‐randomized, proof‐of‐mechanism study using a Bayesian adaptive design to evaluate monotherapy or combination therapy of investigational drugs during the “window‐of‐opportunity” period of at least 2 weeks prior to nephrectomy or partial nephrectomy [65]. Participants would undergo multiparametric MRI before and after treatment, and angiogenic and non‐angiogenic tissues would be collected at surgery. One of the five initial arms includes olaparib monotherapy in patients with surgically resectable ccRCC. While clinical efficacy endpoints such as PFS or OS are not the primary focus of this trial, the primary endpoint for the olaparib monotherapy arm is a > 30% reduction in tumor vascular permeability, measured by Ktrans on dynamic contrast‐enhanced MRI (DCE‐MRI).
This trial is expected to elucidate whether PARP inhibition alone can induce biologically meaningful effects in ccRCC, a disease not classically associated with HRD but potentially effective due to replication stress induced by VHL loss or SETD2 mutations, both common in ccRCC.
5.2. Combination Therapy With ICIs
5.2.1. WIRE Trial (Combination Arms)
The WIRE trial also investigates the immunomodulatory potential of PARP inhibitors through two additional arms: durvalumab monotherapy and durvalumab combined with olaparib. The primary endpoint in these arms is a ≥ 30% increase in CD8+ T cell infiltration, measured by immunohistochemistry comparing pre‐treatment biopsy and nephrectomy tissue. Importantly, preclinical data suggest that PARP inhibition may upregulate PD‐L1 expression and activate the STING pathway, potentially enhancing antitumor immune responses. By comparing monotherapy and combination arms, the WIRE trial will determine whether olaparib enhances the immunogenicity of RCC tumors and whether PARPi–ICI synergy exists in this tumor type, which has shown moderate sensitivity to immune checkpoint blockade in metastatic settings. Translational endpoints will assess changes in cytokine profiles, immune cell subsets, and potential predictive biomarkers of response or resistance.
5.2.2. IMAGENE Trial
The IMAGENE trial (jRCT2051210120), a multi‐center, phase II study, is currently being conducted to investigate niraparib plus anti‐PD‐1 antibody in patients with HRR‐mutant solid tumors after previous ICI treatment [67]. Patients with a variety of metastatic cancers, including UC and RCC, have been enrolled in this trial. This trial aims to confirm which patients would benefit from the targeted combination therapy for patients with HRR gene‐mutated tumors even after the failure of ICIs.
5.3. Application Beyond Classical HRR Mutations
5.3.1. Non‐BRCA/Non‐HRR Mutations
Several studies indicate that DDR gene mutations beyond BRCA1/2, such as ATM, CHEK2, and CDK12, occur in RCC and may predict response to PARPis [68, 69]. A case report documented niraparib efficacy in a patient with metastatic clear cell RCC harboring CDK12 and RAD51C mutations, further expanding the potential biomarker landscape [70].
5.3.2. DDX11 As a Novel Biomarker
In vitro studies have reported that a deficiency in DDX11, a DEAD/DEAH box helicase involved in sister chromatid cohesion, significantly increases sensitivity to olaparib, suggesting that DDX11 may determine PARPi sensitivity in RCC [71]. While clinical evidence is still limited, these findings raise the possibility of PARPi beyond canonical HRR contexts.
5.4. PARPi in Combination With Other Targeted Therapies
5.4.1. NICARAGUA Trial
The NICARAGUA trial (NCT03425201), a phase II trial, aimed to evaluate the efficacy of niraparib and cabozantinib, a tyrosine kinase inhibitor targeting c‐MET and TAM kinases, in patients with advanced UC or RCC [66]. Among 19 patients (predominantly urothelial), combination therapy, including niraparib and cabozantinib, resulted in partial remission in only three patients (16%) (all of them with mUC disease), while the remaining 14 cases (74%) showed SD. This suggests that the benefit of this combination therapy in RCC may be modest and highlights the need for biomarker‐guided patient selection.
5.5. Clinical Implications and Future Perspectives
Although current data remain preliminary, the presence of DDR gene alterations, early signs of PARPi activity, and immunomodulatory effects provide a rationale for continued investigation of PARPis in RCC. Future research should aim to deepen our understanding of predictive biomarkers by incorporating both HRR and non‐HRR DDR gene alterations into trial designs. It will also be important to explore the potential role of PARP inhibitors in patients who have developed resistance to ICI or who are ICI‐naïve. In addition, integrating functional assays and molecular imaging—such as those used in the WIRE trial—may help refine patient selection strategies. Collectively, these approaches will be essential for optimizing the clinical use of PARPis in RCC and advancing toward more personalized therapeutic paradigms.
6. PARP Inhibition in Testicular Germ Cell Tumors
Testicular germ cell tumors (TGCTs) are highly curable with cisplatin‐based chemotherapy; however, a small subset of patients develops refractory or relapsed disease, for which effective therapeutic options remain limited [72]. Recent molecular studies have identified features of HRD and epigenetic silencing of DNA repair genes in TGCTs, providing a biological rationale for evaluating PARP inhibitors (PARPis) in this context. Although clinical evidence is still limited, preclinical studies and early‐phase trials suggest the potential utility of PARPis, particularly in biomarker‐defined populations and in combination with other agents (Table 3).
TABLE 3.
Clinical trials on poly ADP‐ribose polymerase inhibitors for rere cancers.
Trial name | Cancer type | Year of enrollment | Phase | Study design | Patient population | Treatment | HRD status | Enrollment number | Control | Primary endpoint | Results |
---|---|---|---|---|---|---|---|---|---|---|---|
IGG‐02 [73][ | GCTs | 2015–2019 | II | Open‐label | Patients with relapsed or refractory metastatic GCTs | Olaparib | Yes/No | 18 | None | OS | The 12‐month OS was 27.8% (95% CI: 10.1–48.9) |
GCT‐SK‐004 [74] | GCTs | 2016–2020 | II | Non‐randomized, open‐label | Patients with relapsed or refractory GCTs | Veliparib + Gemcitabine + Carboplatin | Yes/No | 15 | None | 12‐month PFS | 12‐month PFS was achieved in 1 (6.7%) patient |
NCT04394858 [75] | PCC/PGL | 2021— | II | Randomized, open‐label | Patients with APP with radiographic evidence of disease progression | Olaparib + Temozolomide | Yes/No | 76 (recruiting) | Temozolomide | PFS | Not reported yet |
Abbreviations: GCTs, germ cell tumors; HRD, homologous recombination deficiency; OS, overall survival; PCC, pheochromocytoma; PFS, progression‐free survival; PGL, paraganglioma.
6.1. PARP Inhibitor Monotherapy in TGCTs
6.1.1. Biological Rationale and Preclinical Data
Immunohistochemical studies have shown that PARP is overexpressed in TGCTs compared to normal testicular tissue, suggesting that PARP activation may be an early event in tumorigenesis [76]. Additionally, functional studies in embryonal carcinoma cell lines have demonstrated that HR repair is compromised, contributing to increased sensitivity to olaparib [77]. In TGCT models, loss of the CCDC6 gene—a regulator of DNA damage response—also impairs homologous recombination and enhances PARPi sensitivity [78].
Genomic analyses, however, show that somatic mutation rates in TGCTs are relatively low, with few recurrent mutations in HRR‐related genes [79]. However, only a mutation in the XRCC2 gene, a member of the RecA/Rad51‐related protein family, was identified at a rate of approximately 2.4%. Importantly, epigenetic alterations such as promoter hypermethylation of BRCA1 and RAD51C have been consistently reported, suggesting that transcriptional silencing of HR genes may represent a mechanism of HRD in TGCTs [80, 81].
6.1.2. IGG‐02 Trial
The IGG‐02 trial (NCT02533765) is a phase II, open‐label, single‐arm study evaluating olaparib monotherapy in patients with refractory or relapsed metastatic GCTs following cisplatin‐based chemotherapy [73]. Among 18 heavily pretreated patients (most with ≥ 3 prior lines of therapy), no objective responses were observed. SD was achieved in five patients (27.8%), with a 12‐week PFS rate of 27.8%. One patient with a BRCA1 germline mutation had SD for 4 months. The study suggests that PARPi monotherapy may have limited activity in unselected, heavily pretreated TGCT patients, though further evaluation in biomarker‐enriched or less heavily treated populations is warranted.
6.2. Combination Therapy With PARPis in TGCTs
6.2.1. GCT‐SK‐004 Trial
The GCT‐SK‐004 trial (NCT02860819) evaluated a combination of veliparib, gemcitabine, and carboplatin in 15 patients with relapsed or refractory extragonadal GCTs [74]. Partial responses were observed in four patients (26.7%), and SD in five patients (33.3%), although the primary endpoint of 12‐month PFS was achieved in only 1 patient (6.7%). The regimen was well tolerated but did not demonstrate substantial benefit. The study hypothesized that PARP inhibition might potentiate DNA damage induced by cytotoxic agents, particularly in the context of PTEN inactivation or defects in nucleotide excision repair, but clinical efficacy remains unproven.
6.3. Clinical Implications and Future Perspectives
Current clinical evidence for PARP inhibitors in TGCTs is limited and inconclusive, particularly in unselected, heavily pretreated populations. Preclinical data suggest that HRD exists in a subset of TGCTs, driven more by epigenetic silencing than genetic mutations. Therefore, future trials should emphasize biomarker‐driven patient selection, potentially integrating methylation profiling or functional HRD assays. In addition, combination therapies, particularly with DNA‐damaging agents or ICIs, may enhance efficacy in otherwise resistant cases. Further investigation in earlier treatment settings or less heavily pretreated cohorts will be essential to determine whether PARPis can play a therapeutic role in TGCT management.
7. Emerging and Exploratory Applications of PARP Inhibitors
Recent preclinical and early genomic evidence suggests that PARPis may have potential applications in rare genitourinary and endocrine tumors, including adrenocortical carcinoma (ACC) and pheochromocytoma/paraganglioma (PCC/PGL).
In ACC, germline HRR gene mutations are uncommon; however, somatic alterations and epigenetic dysregulation of DNA repair pathways, including BRCA1/2 and CHEK2, have been identified in small cohorts [82, 83, 84, 85]. These findings suggest that a molecularly defined subset of ACC may harbor PARPi‐sensitive phenotypes, although no clinical trials have yet been conducted to confirm this hypothesis. On the other hand, in PCC/PGL, particularly in tumors with “Cluster 1 mutations” (e.g., SDHA, SDHB, SDHC, SDHD, SDHAF2, FH, and VHL), results in the activation of the DNA repair system mediated by PARP, which contributes to chemotherapy resistance (specifically to temozolomide) [86, 87]. Therefore, combination therapy of the standard drug temozolomide with olaparib presents a promising approach for treating metastatic tumors with “Cluster 1 mutations.” [88] Preclinical studies utilizing an allograft mouse model have shown that combining olaparib with temozolomide can sensitize PCC/PGL models and suppress metastasis in SDHB‐deficient tumors [89]. A phase II clinical trial (NCT04394858) is currently evaluating this combination in metastatic cases, and its results are awaited [75].
Overall, while these exploratory findings are scientifically compelling, the clinical applicability of PARPis in ACC and PCC/PGL remains unproven, and further research is warranted to define biomarkers of response and identify patient populations most likely to benefit from PARP‐targeted strategies.
8. Conclusion
PARPis are primarily effective in patients with cancer harboring GU cancers with BRCA1/2 mutations. However, recent evidence prompts us to reconsider using PARPis with or without the other types of therapy depending on biomarkers beyond BRCA1/2 mutations. Further research will be needed to explore the exact mechanism for potential biomarkers to apply PARPis into the routine clinical practice in GU cancers.
Author Contributions
Yohei Okuda: conceptualization, writing – original draft, data curation, methodology, software, investigation, validation, formal analysis, visualization. Taigo Kato: conceptualization, writing – review and editing, supervision, data curation, writing – original draft, funding acquisition. Yu Ishizuya: supervision, writing – review and editing. Takuji Hayashi: writing – review and editing. Yoshiyuki Yamamoto: writing – review and editing, supervision. Koji Hatano: writing – review and editing, supervision. Atsunari Kawashima: writing – review and editing, supervision. Junko Murai: writing – review and editing, supervision, writing – original draft, funding acquisition. Norio Nonomura: writing – review and editing, project administration, supervision, resources.
Conflicts of Interest
Taigo Kato received lecture fees from Takeda Pharmaceutical Company Limited and Pfizer Global Supply Japan Inc. Junko Murai received lecture fees from Takeda Pharmaceutical Company Limited, Pfizer Global Supply Japan Inc., and AstraZeneca plc. The other authors declare that they have no conflicts of interest or financial ties related to this study. Norio Nonomura is an Editorial Board member of International Journal of Urology and a co‐author of this article. To minimize bias, he was excluded from all editorial decision‐making related to the acceptance of this article for publication.
References
- 1. D'Amours D., Desnoyers S., D'Silva I., and Poirier G. G., “Poly(ADP‐Ribosyl)ation Reactions in the Regulation of Nuclear Functions,” Biochemical Journal 342, no. Pt 2 (1999): 249–268. [PMC free article] [PubMed] [Google Scholar]
- 2. Amé J.‐C., Spenlehauer C., and de Murcia G., “The PARP Superfamily,” BioEssays 26, no. 8 (2004): 882–893. [DOI] [PubMed] [Google Scholar]
- 3. Caldecott K. W., “Single‐Strand Break Repair and Genetic Disease,” Nature Reviews. Genetics 9, no. 8 (2008): 619–631. [DOI] [PubMed] [Google Scholar]
- 4. de Murcia J. M., Niedergang C., Trucco C., et al., “Requirement of Poly(ADP‐Ribose) Polymerase in Recovery From DNA Damage in Mice and in Cells,” Proceedings of the National Academy of Sciences of the United States of America 94, no. 14 (1997): 7303–7307. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5. Tentori L. and Graziani G., “Chemopotentiation by PARP Inhibitors in Cancer Therapy,” Pharmacological Research 52, no. 1 (2005): 25–33. [DOI] [PubMed] [Google Scholar]
- 6. Calabrese C. R., Almassy R., Barton S., et al., “Anticancer Chemosensitization and Radiosensitization by the Novel Poly(ADP‐Ribose) Polymerase‐1 Inhibitor AG14361,” Journal of the National Cancer Institute 96, no. 1 (2004): 56–67. [DOI] [PubMed] [Google Scholar]
- 7. Bryant H. E., Schultz N., Thomas H. D., et al., “Specific Killing of BRCA2‐Deficient Tumours With Inhibitors of Poly(ADP‐Ribose) Polymerase,” Nature 434, no. 7035 (2005): 913–917. [DOI] [PubMed] [Google Scholar]
- 8. Lord C. J. and Ashworth A., “PARP Inhibitors: Synthetic Lethality in the Clinic,” Science 355, no. 6330 (2017): 1152–1158. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9. Li X. and Heyer W.‐D., “Homologous Recombination in DNA Repair and DNA Damage Tolerance,” Cell Research 18, no. 1 (2008): 99–113. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10. O'Donovan P. J. and Livingston D. M., “BRCA1 and BRCA2: Breast/Ovarian Cancer Susceptibility Gene Products and Participants in DNA Double‐Strand Break Repair,” Carcinogenesis 31, no. 6 (2010): 961–967. [DOI] [PubMed] [Google Scholar]
- 11. Farmer H., McCabe N., Lord C. J., et al., “Targeting the DNA Repair Defect in BRCA Mutant Cells as a Therapeutic Strategy,” Nature 434, no. 7035 (2005): 917–921. [DOI] [PubMed] [Google Scholar]
- 12. Fong P. C., Boss D. S., Yap T. A., et al., “Inhibition of Poly(ADP‐Ribose) Polymerase in Tumors From BRCA Mutation Carriers,” New England Journal of Medicine 361, no. 2 (2009): 123–134, 10.1056/NEJMoa0900212. [DOI] [PubMed] [Google Scholar]
- 13. Murai J., Huang S. Y., Das B. B., et al., “Trapping of PARP1 and PARP2 by Clinical PARP Inhibitors,” Cancer Research 72, no. 21 (2012): 5588–5599, 10.1158/0008-5472.CAN-12-2753. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14. Murai J., Huang S.‐Y. N., Renaud A., et al., “Stereospecific PARP Trapping by BMN 673 and Comparison With Olaparib and Rucaparib,” Molecular Cancer Therapeutics 13, no. 2 (2014): 433–443. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15. Murai J. and Pommier Y., “PARP Trapping Beyond Homologous Recombination and Platinum Sensitivity in Cancers,” Annual Review of Cancer Biology 3, no. 1 (2019): 131–150. [Google Scholar]
- 16. Hanzlikova H., Kalasova I., Demin A. A., Pennicott L. E., Cihlarova Z., and Caldecott K. W., “The Importance of Poly(ADP‐Ribose) Polymerase as a Sensor of Unligated Okazaki Fragments During DNA Replication,” Molecular Cell 71, no. 2 (2018): 319–331.e3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17. Jackson L. M. and Moldovan G.‐L., “Mechanisms of PARP1 Inhibitor Resistance and Their Implications for Cancer Treatment,” NAR Cancer 4, no. 4 (2022): zcac042, 10.1093/narcan/zcac042. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18. Onji H. and Murai J., “Reconsidering the Mechanisms of Action of PARP Inhibitors Based on Clinical Outcomes,” Cancer Science 113, no. 9 (2022): 2943–2951. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19. de Bono J., Mateo J., Fizazi K., et al., “Olaparib for Metastatic Castration‐Resistant Prostate Cancer,” New England Journal of Medicine 382, no. 22 (2020): 2091–2102. [DOI] [PubMed] [Google Scholar]
- 20. Abida W., Armenia J., Gopalan A., et al., “Prospective Genomic Profiling of Prostate Cancer Across Disease States Reveals Germline and Somatic Alterations That May Affect Clinical Decision Making,” Journal of Clinical Oncology: Precision Oncology 2017 (2017): PO.17.00029. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21. Hussain M., Mateo J., Fizazi K., et al., “Survival With Olaparib in Metastatic Castration‐Resistant Prostate Cancer,” New England Journal of Medicine 383, no. 24 (2020): 2345–2357. [DOI] [PubMed] [Google Scholar]
- 22. Fizazi K., Piulats J. M., Reaume M. N., et al., “Rucaparib or Physician's Choice in Metastatic Prostate Cancer,” New England Journal of Medicine 388, no. 8 (2023): 719–732. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23. Taylor A. K., Kosoff D., Emamekhoo H., Lang J. M., and Kyriakopoulos C. E., “PARP Inhibitors in Metastatic Prostate Cancer,” Frontiers in Oncology 13 (2023): 1159557. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24. Clarke N. W., Armstrong A. J., Thiery‐Vuillemin A., et al., “Abiraterone and Olaparib for Metastatic Castration‐Resistant Prostate Cancer,” New England Journal of Medicine Evidence 1, no. 9 (2022): EVIDoa2200043. [DOI] [PubMed] [Google Scholar]
- 25. Agarwal N., Azad A. A., Carles J., et al., “Talazoparib Plus Enzalutamide in Men With First‐Line Metastatic Castration‐Resistant Prostate Cancer (TALAPRO‐2): A Randomised, Placebo‐Controlled, Phase 3 Trial,” Lancet 402, no. 10398 (2023): 291–303. [DOI] [PubMed] [Google Scholar]
- 26. Chi K. N., Sandhu S., Smith M. R., et al., “Niraparib Plus Abiraterone Acetate With Prednisone in Patients With Metastatic Castration‐Resistant Prostate Cancer and Homologous Recombination Repair Gene Alterations: Second Interim Analysis of the Randomized Phase III MAGNITUDE Trial,” Annals of Oncology 34, no. 9 (2023): 772–782. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27. Wu J., Wang H., Ricketts C. J., et al., “Germline Mutations of Renal Cancer Predisposition Genes and Clinical Relevance in Chinese Patients With Sporadic, Early‐Onset Disease,” Cancer 125, no. 7 (2019): 1060–1069. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28. Złowocka‐Perłowska E., Tołoczko‐Grabarek A., Narod S. A., and Lubiński J., “Germline BRCA1 and BRCA2 Mutations and the Risk of Bladder or Kidney Cancer in Poland,” Hereditary Cancer in Clinical Practice 20, no. 1 (2022): 13. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29. Schlacher K., Christ N., Siaud N., Egashira A., Wu H., and Jasin M., “Double‐Strand Break Repair‐Independent Role for BRCA2 in Blocking Stalled Replication Fork Degradation by MRE11,” Cell 145, no. 4 (2011): 529–542. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30. Guo M. and Wang S. M., “The BRCAness Landscape of Cancer,” Cells 11, no. 23 (2022): 3877, 10.3390/cells11233877. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31. Esteller M., Silva J. M., Dominguez G., et al., “Promoter Hypermethylation and BRCA1 Inactivation in Sporadic Breast and Ovarian Tumors,” Journal of the National Cancer Institute 92, no. 7 (2000): 564–569. [DOI] [PubMed] [Google Scholar]
- 32. McCabe N., Turner N. C., Lord C. J., et al., “Deficiency in the Repair of DNA Damage by Homologous Recombination and Sensitivity to Poly(ADP‐Ribose) Polymerase Inhibition,” Cancer Research 66, no. 16 (2006): 8109–8115. [DOI] [PubMed] [Google Scholar]
- 33. Tutt A., Robson M., Garber J. E., et al., “Oral Poly(ADP‐Ribose) Polymerase Inhibitor Olaparib in Patients With BRCA1 or BRCA2 Mutations and Advanced Breast Cancer: A Proof‐of‐Concept Trial,” Lancet 376, no. 9737 (2010): 235–244. [DOI] [PubMed] [Google Scholar]
- 34. Mateo J., Carreira S., Sandhu S., et al., “DNA‐Repair Defects and Olaparib in Metastatic Prostate Cancer,” New England Journal of Medicine 373, no. 18 (2015): 1697–1708. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35. Mateo J., Porta N., Bianchini D., et al., “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 Oncology 21, no. 1 (2020): 162–174. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36. Moore K., Colombo N., Scambia G., et al., “Maintenance Olaparib in Patients With Newly Diagnosed Advanced Ovarian Cancer,” New England Journal of Medicine 379, no. 26 (2018): 2495–2505. [DOI] [PubMed] [Google Scholar]
- 37. Gruber J. J., Afghahi A., Timms K., et al., “A Phase II Study of Talazoparib Monotherapy in Patients With Wild‐Type BRCA1 and BRCA2 With a Mutation in Other Homologous Recombination Genes,” Nature Cancer 3, no. 10 (2022): 1181–1191. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38. Willis S. E., Winkler C., Roudier M. P., et al., “Retrospective Analysis of Schlafen11 (SLFN11) to Predict the Outcomes to Therapies Affecting the DNA Damage Response,” British Journal of Cancer 125, no. 12 (2021): 1666–1676. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39. Murai J., Thomas A., Miettinen M., and Pommier Y., “Schlafen 11 (SLFN11), a Restriction Factor for Replicative Stress Induced by DNA‐Targeting Anti‐Cancer Therapies,” Pharmacology & Therapeutics 201 (2019): 94–102. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40. Murai J., Feng Y., Yu G. K., et al., “Resistance to PARP Inhibitors by SLFN11 Inactivation Can Be Overcome by ATR Inhibition,” Oncotarget 7, no. 47 (2016): 76534–76550. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41. Takashima T., Sakamoto N., Murai J., et al., “Immunohistochemical Analysis of SLFN11 Expression Uncovers Potential Non‐Responders to DNA‐Damaging Agents Overlooked by Tissue RNA‐Seq,” Virchows Archiv 478, no. 3 (2021): 569–579. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42. Kaczorowski M., Ylaya K., Chłopek M., et al., “Immunohistochemical Evaluation of Schlafen 11 (SLFN11) Expression in Cancer in the Search of Biomarker‐Informed Treatment Targets: A Study of 127 Entities Represented by 6658 Tumors,” American Journal of Surgical Pathology 48 (2024): 1512–1521. [DOI] [PubMed] [Google Scholar]
- 43. Stewart R. A., Pilié P. G., and Yap T. A., “Development of PARP and Immune‐Checkpoint Inhibitor Combinations,” Cancer Research 78, no. 24 (2018): 6717–6725. [DOI] [PubMed] [Google Scholar]
- 44. Shen J., Zhao W., Ju Z., et al., “PARPi Triggers the STING‐Dependent Immune Response and Enhances the Therapeutic Efficacy of Immune Checkpoint Blockade Independent of BRCAness,” Cancer Research 79, no. 2 (2019): 311–319. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45. Huang J., Wang L., Cong Z., et al., “The PARP1 Inhibitor BMN 673 Exhibits Immunoregulatory Effects in a BRCA1(−/−) Murine Model of Ovarian Cancer,” Biochemical and Biophysical Research Communications 463, no. 4 (2015): 551–556. [DOI] [PubMed] [Google Scholar]
- 46. Jiao S., Xia W., Yamaguchi H., et al., “PARP Inhibitor Upregulates PD‐L1 Expression and Enhances Cancer‐Associated Immunosuppression,” Clinical Cancer Research 23, no. 14 (2017): 3711–3720. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47. Nassar A. H., Umeton R., Kim J., et al., “Mutational Analysis of 472 Urothelial Carcinoma Across Grades and Anatomic Sites,” Clinical Cancer Research 25, no. 8 (2019): 2458–2470. [DOI] [PubMed] [Google Scholar]
- 48. Yang K., Yu W., Liu H., et al., “Comparison of Genomic Characterization in Upper Tract Urothelial Carcinoma and Urothelial Carcinoma of the Bladder,” Oncologist 26, no. 8 (2021): e1395‐405‐e405. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49. Abbosh P. H. and Plimack E. R., “Molecular and Clinical Insights Into the Role and Significance of Mutated DNA Repair Genes in Bladder Cancer,” Bladder Cancer 4, no. 1 (2018): 9–18. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50. Nassar A. H., Abou Alaiwi S., AlDubayan S. H., et al., “Prevalence of Pathogenic Germline Cancer Risk Variants in High‐Risk Urothelial Carcinoma,” Genetics in Medicine 22, no. 4 (2020): 709–718, 10.1038/s41436-019-0720-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51. Jian W., Xu H.‐G., Chen J., et al., “Activity of CEP‐9722, a Poly (ADP‐Ribose) Polymerase Inhibitor, in Urothelial Carcinoma Correlates Inversely With Homologous Recombination Repair Response to DNA Damage,” Anti‐Cancer Drugs 25, no. 8 (2014): 878–886. [DOI] [PubMed] [Google Scholar]
- 52. Teo M. Y., Seier K., Ostrovnaya I., et al., “Alterations in DNA Damage Response and Repair Genes as Potential Marker of Clinical Benefit From PD‐1/PD‐L1 Blockade in Advanced Urothelial Cancers,” Journal of Clinical Oncology 36, no. 17 (2018): 1685–1694. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53. Powles T., Carroll D., Chowdhury S., et al., “An Adaptive, Biomarker‐Directed Platform Study of Durvalumab in Combination With Targeted Therapies in Advanced Urothelial Cancer,” Nature Medicine 27, no. 5 (2021): 793–801. [DOI] [PubMed] [Google Scholar]
- 54. Rosenberg J. E., Park S. H., Kozlov V., et al., “Durvalumab Plus Olaparib in Previously Untreated, Platinum‐Ineligible Patients With Metastatic Urothelial Carcinoma: A Multicenter, Randomized, Phase II Trial (BAYOU),” Journal of Clinical Oncology 41, no. 1 (2023): 43–53. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55. Grivas P., Loriot Y., Morales‐Barrera R., et al., “Efficacy and Safety of Rucaparib in Previously Treated, Locally Advanced or Metastatic Urothelial Carcinoma From a Phase 2, Open‐Label Trial (ATLAS),” BMC Cancer 21, no. 1 (2021): 593. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56. Fulton B., Jones R., Powles T., et al., “Atlantis: A Randomised Multi‐Arm Phase II Biomarker‐Directed Umbrella Screening Trial of Maintenance Targeted Therapy After Chemotherapy in Patients With Advanced or Metastatic Urothelial Cancer,” Trials 21, no. 1 (2020): 344. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57. Crabb S. J., Hussain S., Soulis E., et al., “A Randomized, Double‐Blind, Biomarker‐Selected, Phase II Clinical Trial of Maintenance Poly ADP‐Ribose Polymerase Inhibition With Rucaparib Following Chemotherapy for Metastatic Urothelial Carcinoma,” Journal of Clinical Oncology 41, no. 1 (2023): 54–64. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58. Yap T. A., Bardia A., Dvorkin M., et al., “Avelumab Plus Talazoparib in Patients With Advanced Solid Tumors: The JAVELIN PARP Medley Nonrandomized Controlled Trial,” JAMA Oncology 9, no. 1 (2023): 40–50. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59. Powles T., Park S. H., Voog E., et al., “Avelumab Maintenance Therapy for Advanced or Metastatic Urothelial Carcinoma,” New England Journal of Medicine 383, no. 13 (2020): 1218–1230. [DOI] [PubMed] [Google Scholar]
- 60. Vignani F., Tambaro R., De Giorgi U., et al., “Addition of Niraparib to Best Supportive Care as Maintenance Treatment in Patients With Advanced Urothelial Carcinoma Whose Disease Did Not Progress After First‐Line Platinum‐Based Chemotherapy: The Meet‐URO12 Randomized Phase 2 Trial,” European Urology 83, no. 1 (2023): 82–89. [DOI] [PubMed] [Google Scholar]
- 61. Rodriguez‐Moreno J. F., Ruiz‐Llorente S., De Velasco G., et al., “Comprehensive Molecular Characterization of Muscle‐Invasive Bladder Cancer (MIBC) Treated With Durvalumab Plus Olaparib in the Neoadjuvant Setting: Neodurvarib Trial,” Journal of Clinical Oncology 40, no. S6 (2022): 546–555.34985966 [Google Scholar]
- 62. Rodriguez‐Moreno J. F., de Velasco G., Alvarez‐Fernandez C., et al., “761P Impact of the Combination of Durvalumab (MEDI4736) Plus Olaparib (AZD2281) Administered Prior to Surgery in the Molecular Profile of Resectable Urothelial Bladder Cancer. NEODURVARIB Trial,” Annals of Oncology 31, no. S589 (2020): S589, 10.1016/j.annonc.2020.08.833. [DOI] [Google Scholar]
- 63. Pletcher J. P., Bhattacharjee S., Doan J. P., et al., “The Emerging Role of Poly (ADP‐Ribose) Polymerase Inhibitors as Effective Therapeutic Agents in Renal Cell Carcinoma,” Frontiers in Oncology 11 (2021): 681441. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64. Ged G., Rifkind I., Tony L., et al., “Orchid: A Phase II Study of Olaparib in Metastatic Renal Cell Carcinoma Patients HarborIng a BAP1 or Other DNA Repair Gene Mutations,” Oncologist 28, no. suppl 1 (2023): S1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65. Ursprung S., Mossop H., Gallagher F. A., et al., “The WIRE Study a Phase II, Multi‐Arm, Multi‐Centre, Non‐Randomised Window‐of‐Opportunity Clinical Trial Platform Using a Bayesian Adaptive Design for Proof‐of‐Mechanism of Novel Treatment Strategies in Operable Renal Cell Cancer ‐ A Study Protocol,” BMC Cancer 21, no. 1 (2021): 1238. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66. Castellano D. E., Duran I., Mellado B., et al., “Phase I‐II Study to Evaluate Safety and Efficacy of Niraparib Plus Cabozantinib in Patients With Advanced Urothelial/Kidney Cancer (Nicaragua Trial): Preliminary Data of Phase I Study,” Journal of Clinical Oncology 40, no. 6_suppl (2022): 490, 10.1200/JCO.2022.40.6_suppl.490. [DOI] [Google Scholar]
- 67. Kato T., Matsubara N., Shiota M., et al., “IMAGENE Trial: Multicenter, Proof‐Of‐Concept, Phase II Study Evaluating the Efficacy and Safety of Combination Therapy of Niraparib With PD‐1 Inhibitor in Solid Cancer Patients With Homologous Recombination Repair Genes Mutation,” BMC Cancer 22, no. 1 (2022): 1292. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68. Truong H., Sheikh R., Kotecha R., et al., “Germline Variants Identified in Patients With Early‐Onset Renal Cell Carcinoma Referred for Germline Genetic Testing,” European Urology Oncology 4, no. 6 (2021): 993–1000. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69. Ged Y., Chaim J. L., DiNatale R. G., et al., “DNA Damage Repair Pathway Alterations in Metastatic Clear Cell Renal Cell Carcinoma and Implications on Systemic Therapy,” Journal for Immunotherapy of Cancer 8, no. 1 (2020): e000230. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70. Yue X., Yang C., Cao D., and Li Y., “Niraparib for the Treatment of Metastatic ccRCC in a Patient With CDK12 and RAD51C Mutations: A Case Report,” Frontiers in Pharmacology 15 (2024): 1396606. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71. Park J. S., Lee M. E., Jang W. S., et al., “The DEAD/DEAH Box Helicase, DDX11, Is Essential for the Survival of Advanced Clear Cell Renal Cell Carcinoma and Is a Determinant of PARP Inhibitor Sensitivity,” Cancers (Basel) 13, no. 11 (2021): 2574. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72. Pfister D., Oechsle K., Schmidt S., et al., “First‐Line Salvage Treatment Options for Germ Cell Tumor Patients Failing Stage‐Adapted Primary Treatment: A Comprehensive Review Compiled by the German Testicular Cancer Study Group,” World Journal of Urology 40, no. 12 (2022): 2853–2861. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73. De Giorgi U., Schepisi G., Gurioli G., et al., “Olaparib as Salvage Treatment for Advanced Germ Cell Tumors After Chemotherapy Failure: Results of the Open‐Label, Single‐Arm, IGG‐02 Phase II Trial,” Journal of Clinical Oncology 38, no. 15_suppl (2020): 5058. [Google Scholar]
- 74. Mego M., Svetlovska D., Reckova M., et al., “Gemcitabine, Carboplatin and Veliparib in Multiple Relapsed/Refractory Germ Cell Tumours: The GCT‐SK‐004 Phase II Trial,” Investigational New Drugs 39, no. 6 (2021): 1664–1670. [DOI] [PubMed] [Google Scholar]
- 75. Del Rivero J. D., Perez K., Geyer S. M., et al., “Alliance A021804: A Prospective, Multi‐Institutional Phase II Trial Evaluating Temozolomide Versus Temozolomide and Olaparib for Advanced Pheochromocytoma and Paraganglioma,” Journal of Clinical Oncology 41, no. 16_suppl (2023): TPS4191. [Google Scholar]
- 76. Mego M., Cierna Z., Svetlovska D., et al., “PARP Expression in Germ Cell Tumours,” Journal of Clinical Pathology 66, no. 7 (2013): 607–612. [DOI] [PubMed] [Google Scholar]
- 77. Cavallo F., Graziani G., Antinozzi C., et al., “Reduced Proficiency in Homologous Recombination Underlies the High Sensitivity of Embryonal Carcinoma Testicular Germ Cell Tumors to Cisplatin and Poly (ADP‐Ribose) Polymerase Inhibition,” PLoS One 7, no. 12 (2012): e51563. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78. Morra F., Merolla F., Zito Marino F., et al., “The Tumour Suppressor CCDC6 Is Involved in ROS Tolerance and Neoplastic Transformation by Evading Ferroptosis,” Heliyon 7, no. 11 (2021): e08399. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79. Litchfield K., Summersgill B., Yost S., et al., “Whole‐Exome Sequencing Reveals the Mutational Spectrum of Testicular Germ Cell Tumours,” Nature Communications 6 (2015): 5973. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80. Shen H., Shih J., Hollern D. P., et al., “Integrated Molecular Characterization of Testicular Germ Cell Tumors,” Cell Reports 23, no. 11 (2018): 3392–3406. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81. Lobo J., Constâncio V., Guimarães‐Teixeira C., et al., “Promoter Methylation of DNA Homologous Recombination Genes Is Predictive of the Responsiveness to PARP Inhibitor Treatment in Testicular Germ Cell Tumors,” Molecular Oncology 15, no. 4 (2021): 846–865. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82. Lavoie J.‐M., Csizmok V., Williamson L. M., et al., “Whole‐Genome and Transcriptome Analysis of Advanced Adrenocortical Cancer Highlights Multiple Alterations Affecting Epigenome and DNA Repair Pathways,” Cold Spring Harbor Molecular Case Studies 8, no. 3 (2022): a006148, 10.1101/mcs.a006148. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83. El Ghorayeb N., Grunenwald S., Nolet S., et al., “First Case Report of an Adrenocortical Carcinoma Caused by a BRCA2 Mutation,” Medicine 95, no. 36 (2016): e4756. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84. Xie C., Tanakchi S., Raygada M., Davis J. L., and Del Rivero J., “Case Report of an Adrenocortical Carcinoma Associated With Germline CHEK2 Mutation,” Journal of the Endocrine Society 3, no. 1 (2019): 284–290. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85. Lippert J., Appenzeller S., Liang R., et al., “Targeted Molecular Analysis in Adrenocortical Carcinomas: A Strategy Toward Improved Personalized Prognostication,” Journal of Clinical Endocrinology and Metabolism 103, no. 12 (2018): 4511–4523. [DOI] [PubMed] [Google Scholar]
- 86. Ben‐Hur E., Utsumi H., and Elkind M. M., “Inhibitors of Poly (ADP‐Ribose) Synthesis Enhance Radiation Response by Differentially Affecting Repair of Potentially Lethal Versus Sublethal Damage,” British Journal of Cancer. Supplement 6 (1984): 39–42. [PMC free article] [PubMed] [Google Scholar]
- 87. Schlicker A., Peschke P., Bürkle A., Hahn E. W., and Kim J. H., “4‐Amino‐1,8‐Naphthalimide: A Novel Inhibitor of Poly(ADP‐Ribose) Polymerase and Radiation Sensitizer,” International Journal of Radiation Biology 75, no. 1 (1999): 91–100. [DOI] [PubMed] [Google Scholar]
- 88. Nölting S., Grossman A., and Pacak K., “Metastatic Phaeochromocytoma: Spinning Towards More Promising Treatment Options,” Experimental and Clinical Endocrinology & Diabetes 127, no. 2–3 (2019): 117–128, 10.1055/a-0715-1888. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89. Pang Y., Lu Y., Caisova V., et al., “Targeting NAD(+)/PARP DNA Repair Pathway as a Novel Therapeutic Approach to SDHB‐Mutated Cluster I Pheochromocytoma and Paraganglioma,” Clinical Cancer Research 24, no. 14 (2018): 3423–3432. [DOI] [PMC free article] [PubMed] [Google Scholar]